MOBILE VIEW  | 

CYANIDE

Classification   |    Detailed evidence-based information

Therapeutic Toxic Class

    A) Cyanides inhibit cytochrome oxidase, an enzyme necessary for cellular oxygen transport, thereby interfering with the oxygen uptake of cells. Cyanide is readily absorbed from all routes, including the skin and mucous membranes, and by inhalation, although alkali salts of cyanide are toxic only when ingested.

Specific Substances

    A) CYANIDE
    1) Carbon nitride ion
    2) CN
    3) Cyanide anion
    4) Cyanide ion
    5) Cyanure (French)
    6) Isocyanide
    7) CAS 57-12-5
    CALCIUM CYANIDE
    1) Calcium cyanide, solid
    2) Calcyanide
    3) Cyanogas
    4) Cyanure de Calcium (French)
    5) CAS 592-01-8
    CYANOGEN
    1) Carbon nitride
    2) Cyanogene (French)
    3) Cyanogen Gas
    4) Dicyan
    5) Dicyanogen
    6) Ethanedinitrile
    7) Nitriloacetonitrile
    8) Oxalic acid dinitrile
    9) Oxalonitrile
    10) Oxalyl Cyanide
    11) CAS 460-19-5
    CYANOGEN AZIDE
    1) CAS 764-05-6
    CYANOGEN BROMIDE
    1) Bromine cyanide
    2) Bromocyan
    3) Bromocyanide
    4) Bromocyanogen
    5) Bromure de Cyanogen (French)
    6) Campilit
    7) Cyanobromide
    8) Cyanogen Bromide
    9) Cyanogen Monobromide
    10) TL 822
    11) CAS 506-68-3
    CYANOGEN CHLORIDE
    1) Chlorcyan
    2) Chlorine cyanide
    3) Chlorocyan
    4) Chlorocyanide
    5) Chlorocyanogen
    6) Chlorure de Cyanogene (French)
    7) Cyanogen Chloride
    8) CAS 506-77-4
    CYANOGEN FLUORIDE
    1) CAS 1495-50-7
    CYANOGEN IODIDE
    1) CAS 506-78-5
    HYDROCYANIC ACID
    1) Acide Cyahydrique (French)
    2) Acido Cianidrico (Italian)
    3) Aero Liquid HCN
    4) Blausaeure (German)
    5) Blauwzuur (Dutch)
    6) Cyaanwaterstof (Dutch)
    7) Cyanwasserstoff (German)
    8) Cyclon
    9) Cyclone B
    10) Cyjanowodor (Polish)
    11) HCN
    12) Hydrocyanic acid, liquefied
    13) Hydrogen cyanide
    14) Prussic acid
    15) CAS 74-90-8
    POTASSIUM CYANIDE
    1) Cyanide of Potassium
    2) Cyanure de Potassium (French)
    3) Hydrocyanic acid, potassium salt
    4) Potassium cyanide, solution
    5) CAS 151-50-8
    SODIUM CYANIDE
    1) Cianuro di Sodio (Italian)
    2) Cyanide of Sodium
    3) Cyanure de Sodium (French)
    4) Cymag
    5) Hydrocyanic acid, sodium salt
    6) Kyanid sodny (Czech)
    7) Sodium cyanide, solid
    8) CAS 143-33-9
    ZINC CYANIDE
    1) CAS 557-21-1
    GENERAL TERMS
    1) CYANOGENE GAS
    2) CYANOGEN, LIQUEFIED
    3) CYANIDES, INORGANIC, SOLID, N.O.S.
    4) CYANIDES, INORGANIC, N.O.S.
    5) CYANIDE SOLUTIONS
    6) CYANIDE, POTASSIUM
    7) CYANIDE, CALCIUM
    8) CARBON HYDRIDE NITRIDE (CHN)
    9) FLUOHYDRIC ACID GAS
    10) KEMAMINE N-200
    11) HYDROXYANIC ACID, LIQUIFIED
    12) HYDROGEN CYANIDE, LIQUIFIED
    13) HYDROGEN CYANIDE GAS
    14) HYDROGEN CYANIDE, ANHYDROUS
    15) HYDROGEN CYANIDE, ABSORBED
    16) HYDROCYANIC ACID, UNSTABILIZED
    17) HYDROCYANIC ACID, SOLUTION
    18) HYDROCYANIC ACID GAS
    19) POTASSIUM CYANIDE (KCN)
    20) HYDROCYANIC ACID, AQUEOUS SOLUTION, WITH LESS THAN 5% HYDROGEN CYANIDE
    21) PRUSSIC ACID SOLUTION
    22) ACETONCIANHIDRINEI (ROMANIAN)
    23) HYDROCYANIC ACID, AQUEOUS SOLUTION, WITH NOT LESS THAN 5% HYDROCYANIC ACID
    24) OXYISOBUTYRIC NITRILE

    1.2.1) MOLECULAR FORMULA
    1) C-N

Available Forms Sources

    A) FORMS
    1) GOLD ORE EXTRACTION
    a) Mellor (1961) described the gold ore extraction process.
    b) AQUEOUS ALKALINE CYANIDE: The gold ore is treated with hot alkaline cyanide solution (potassium cyanide). The complexed gold is treated in wood or iron vats containing either zinc shavings or lead-coated zinc shavings and allowed to percolate upward through the shaving bed. Gold is precipitated as a fine powder.
    c) CYANIDE RECOVERY: The cyanide solution is recovered and reused. The vat sludge is used to wash the precipitated gold with water and then filtered to remove most water.
    d) FINAL STEP: The vat sludge is roasted at red heat and is treated with hydrochloric acid to remove zinc. Then it is washed, dried, mixed with a mixture of sodium carbonate, glass, borax, sand, and fluorspar (flux) and fused in a graphite crucible. The metallic gold is remelted and cast into ingots.
    2) DESCRIPTION
    a) Various cyanide compounds are listed below:
    b) POTASSIUM, SODIUM and CALCIUM CYANIDES are white, deliquescent, noncombustible solids with a faint odor of bitter almonds (ACGIH, 1991).
    c) HYDROGEN CYANIDE is a colorless liquid with a characteristic odor (Clayton & Clayton, 1982).
    d) CALCIUM CYANAMIDE is a white crystalline solid (Clayton & Clayton, 1982).
    e) CYANOGEN is a colorless gas (Clayton & Clayton, 1982).
    f) CYANOGEN CHLORIDE is a colorless liquid or gas (Clayton & Clayton, 1982).
    g) CYANOGEN BROMIDE is a colorless crystal (needles or cubes) (Clayton & Clayton, 1982).
    h) DIMETHYL CYANAMIDE is a colorless liquid (Clayton & Clayton, 1982).
    B) SOURCES
    1) Household uses of cyanide include fumigation, silver-polishing, and as fertilizers, rodenticides and insecticides.
    2) OTC TAMPERING: Episodes of deliberate tampering with OTC capsule medications in 1982, 1986, and 1991 have resulted in 11 deaths from cyanide poisoning (CDC, 1991; Wolnik et al, 1984; Varnell et al, 1987). Since 1982, OTC capsules have been required to have at least 2 tamper-resistant features. Medication with any alteration in these features should not be used, and should be provided to the FDA. Features may include:
    a) Sealing of the capsule with a band,
    b) Blister packs with foil backing,
    c) Sealing of the package with a safety tab,
    d) Identification of blister pack and box with identical code numbers.
    3) Imported metal cleaning solutions used by Hmong refugees, and sold in Hmong-American markets, to clean coins, jewelry, or polish silver, may contain cyanide salts (Garlich et al, 2012; Krieg & Saxena, 1987).
    a) Several cases of cyanide poisonings have been reported in the Hmong community within the United States following intentional ingestion of products used to clean metal, coins, or jewelry that contained sodium cyanide (Garlich et al, 2012).
    4) Aliphatic thiocyanates are commonly used as insecticides, and include Thanite (isobornyl thiocyanoacetate), Lethane 60, and Lethane 384. Liver enzymes liberate HCN from these insecticides, and antidotal measures for cyanide poisoning should be promptly instituted following significant exposure.
    5) Cyanide may also be liberated in the burning of plastics containing nitrogen, natural fabrics (wool or silk), polyurethane bedding or furniture, acrylic baths, nylon carpets, and melamine resin insulation, and thus should be suspected in smoke inhalations victims (Clark et al, 1981; Becker, 1985; Hall & Rumack, 1986; Silverman et al, 1988; Geller et al, 2006).
    6) "Coyote Gitter" shells contain potassium cyanide (Hall & Rumack, 1986).
    7) FRUIT CONTAMINATION: Following an episode of contamination of grapes with cyanide, the stability of potassium cyanide in spiked fruit was examined and found to be pH-dependent (Chadha et al, 1991).
    a) Fruits with a natural pH of less than 4 (apples, white grape juice, green grapes, strawberries) had no significant decrease in cyanide concentration after 4 hours.
    b) Fruits with a higher inherent pH (peaches, honeydew melon) had a rapid reduction in cyanide concentration, decreasing by 92% over 4 hours for the honeydew samples.
    8) PLANTS AND PLANT PARTS
    a) Amygdalin cyanogenic glycosides (found in apple, peach, apricot, plum, cherry, and almond seeds) may release hydrogen cyanide after ingestion (Klaassen et al, 1986; (Lasch & El Shawa, 1981; Geller et al, 2006).
    b) Laetrile, a once popular lay "cancer cure," also contains amygdalin and cyanide toxicity occurs with ingestion (Geller et al, 2006; Ellenhorn & Barceloux, 1988; Hall & Rumack, 1986).
    c) Cyanide content of Laetrile and Amygdalin (see Plant Cyanide management for average cyanide content of some plant products) (Davignon et al, 1978; Levi et al, 1965; Schmidt et al, 1978; Shragg et al, 1982; pp 1-7; Jee et al, 1978; Ames et al, 1978; Holzbecher et al, 1984):
    LaetrileHCN (mg/g)
    Tablets
    250 mg2.7
    500 mg29.0-51.5
    Parenteral
    10 mL, 3 gram vial23.2-57.9
    Amygdalin - Parenteral
    10 mL, 3 gram vial8.3-51.3

    d) Chronic consumption of unprocessed cassava or improperly prepared cassava has been associated with elevated thiocyanate levels, spastic paresis, sensory ataxia, and optic neuropathy (Casadei et al, 1990; Cliff et al, 1986; Freeman, 1988). The glycosides, contained within cassava, are hydrolyzed to glucose, hydrogen cyanide, and acetone by either intestinal beta-glucosidase or by the beta-glucosidase from the cassava plant (Geller et al, 2006).
    e) Other cyanogenic plants include the species of Linium and Prunus, and sorghum, lima beans, and bamboo sprouts (Ellenhorn & Barceloux, 1988).
    9) NITRILE COMPOUNDS: Cyanide can be released by hepatic metabolism from various nitrile compounds, such as malononitrile, succinonitrile, acetonitrile, and propionitrile following absorption into the body (Ballantyne, 1987; Boggild et al, 1990; Caravati & Litovitz, 1988).
    a) A deliberate poisoning with acetonitrile resulted in cyanide poisoning 11 hours later which was successfully treated by repeated boluses of nitrite and thiosulfate. The half-life of conversion of acetonitrile was 40 hours and harmful blood cyanide levels persisted for over 24 hours after ingestion. In cases of acetonitrile poisoning, monitoring in an intensive care unit should continue for 24-48 hours (Mueller & Borland, 1997).
    b) Cyanide toxicity has been reported to occur from an artificial nail remover containing acetonitrile (Caravati & Litovitz, 1988; Geller et al, 1991; Geller et al, 2006).
    C) USES
    1) ALKALINE CYANIDE SALTS
    a) The alkaline cyanide salts (potassium and sodium cyanide) are used for gold and silver ore extraction, recovery of metals from X-ray and photographic films, metal heat treating, electroplating, chelation, manufacture of dyes, pigments, and nylon, and as insecticides and fumigants (AAR, 1987; (ACGIH, 1986; Blanc et al, 1985; EPA, 1985) HSDB, 1990; OHM/TADS, 1990; (Proctor et al, 1988; Sax & Lewis, 1989).
    b) Individuals involved in the illegal manufacture of phencyclidine may be exposed to potassium cyanide and its intermediates (Hall & Rumack, 1986).
    c) Calcium cyanide is used mainly as a fumigant (ACGIH, 1986). Other uses may include stainless steel manufacture, cement stabilization, froth flotation of minerals, and as an insecticide, rodenticide, and miticide (HSDB, 1990).
    d) Magnesium cyanide (Cymag(R)) has been used for pest control, poisoning rabbits, and illegally for salmon poaching (the technique, known as "cymagging" involves adding the cyanide to water, which causes the fish to float to the surface) in Scotland (Fernando & Busuttil, 1991).
    2) HYDROGEN CYANIDE: Hydrogen cyanide is a gas that may be liquefied as HYDROCYANIC ACID and occurs during its manufacture as a chemical reagent (ACGIH, 1986).
    a) It is used in the production of chemical intermediates in synthetic fiber, plastics, nitrites, and cyanide salts manufacturing, and is used to fumigate ships, railroad cars, buildings, orchards, tobacco, and certain foods (ACGIH, 1986).
    b) Hydrogen cyanide may be produced during petroleum refining, electroplating, metallurgical, and photographic developing operations (ACGIH, 1986).
    3) CYANOGEN AND ITS HALIDES: Cyanogen and cyanogen halides (cyanogen bromide, cyanogen chloride, cyanogen iodide) release hydrogen cyanide and have been used as military chemical warfare agents (ACGIH, 1986; Barr, 1985).
    a) Cyanogen is used in organic synthesis, as a fumigant and pesticide, in gold extraction processes, as a fuel gas for cutting and welding, and as a rocket or missile propellant (AAR, 1987; (ACGIH, 1986).
    b) Cyanogen chloride is used in organic synthesis (ACGIH, 1986).
    4) METAL CYANIDES: Metal cyanides (particularly ferro- and ferri- cyanides) rarely produce cyanide poisoning due to the tight binding between the metal and the cyanyl group which prevents cyanide release (Jouglard et al, 1971).
    a) One case of death from exposure to potassium aurocyanide has been reported (Wright & Vesey, 1986).
    b) Another patient ingested a mercury oxycyanide antiseptic intended for topical use and developed both mild cyanide poisoning symptoms and mercury-induced acute renal failure (Jouglard et al, 1971).
    c) The metal cyanides are used in photography, metal tempering, electroplating, in the manufacture of pigments, in dyeing, and as chemical reagents (ILO, 1983).
    d) Cyanide-containing wastes, commonly found in soils at former manufactured gas plant sites, are often considered toxic. However, the blue-stained soils and rocks found at these sites contain the relatively non-toxic iron-complexed forms such as ferric ferrocyanide (Shifrin et al, 1996).
    5) NITROPRUSSIDE
    a) Cyanide is also a metabolite of nitroprusside and toxicity may result, especially with rapid infusion, prolonged use where tachyphylaxis requires high doses, or when renal failure is present (Ellenhorn & Barceloux, 1988; Gonzales & Sabatini, 1989; Ram et al, 1989; Geller et al, 2006).
    b) Infusions of sodium nitroprusside in rates above 2 mcg/kg/min may cause cyanide to accumulate to toxic concentrations in critically ill patients (Johanning et al, 1995).
    6) USES OF CYANIDE COMPOUNDS
    a) SODIUM and POTASSIUM CYANIDE are used in the extraction of gold and silver ores; electroplating; metal cleaning; as insecticides and fumigants; in heat treatment of metals; and as raw materials in the manufacture of dyes, pigments, nylon, and chelating agents. CALCIUM CYANIDE, the other cyanide of major commercial importance, is used chiefly as a fumigant since it readily releases hydrogen cyanide (HCN) when exposed to air (ACGIH, 1991).
    b) HYDROGEN CYANIDE: Chief uses are in fumigation of ships, buildings, orchards, and various foods; in electroplating; in mining; in the production of various resin monomers such as acrylates, methacrylates, and hexamethylenediamine; and in the production of other nitriles. It also has many uses as a chemical intermediate, and may be generated in such operations as blast furnaces, gas works, and coke ovens (Clayton & Clayton, 1982).
    c) CALCIUM CYANAMIDE: Major use is as a fertilizer. However, it has a number of other uses, such as an herbicide and a defoliant for cotton plants. Also used as a chemical intermediate (Clayton & Clayton, 1982).
    d) CYANOGEN is used as a fumigant, and may be encountered in situations in which there is heating of nitrogen-containing carbon bonds and in blast furnace gases, etc (Clayton & Clayton, 1982). Cyanogen and cyanogen halides (cyanogen bromide, cyanogen chloride, cyanogen iodide) release hydrogen cyanide and have been used as military chemical warfare agents (Barr, 1985).
    e) CYANOGEN CHLORIDE is used in organic synthesis and as a warning agent in fumigant gases (Clayton & Clayton, 1982).
    f) CYANOGEN BROMIDE is used in organic synthesis, as a fumigant and pesticide, and in gold-extraction processes. It has also been used in connection with cellulose technology (Clayton & Clayton, 1982).
    g) INTENTIONAL EXPOSURE
    1) Based on a 10-year study conducted in the US, cyanide is a rare form of intentional injury or death and an occupational association has been observed. Occupations that have easy access to cyanide include: chemists, jewelers, individuals involved in pest control, mineral refining, photography, electroplating, dyeing, printing, and salmon poaching. The study also found that males were more likely to use this method of suicide over females. In this study, the 14 of 17 fatal cyanide cases were males, and 8 of the fatalities were from the West Indies/Caribbean Islands or South America (Gill et al, 2004).

Life Support

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

Clinical Effects

    0.2.1) SUMMARY OF EXPOSURE
    A) SOURCES: Cyanide is primarily found as either a solid cyanide salt, a salt solution, or as hydrogen cyanide gas. It may be liberated as cyanide gas in house fires from the combustion of wool, silk, synthetic rubber, and polyurethane. It was previously found in artificial nail removers as acetonitrile.
    B) USES: Various industries including metal extraction and refining, electroplating, photography, manufacturing, and fumigation.
    C) PHARMACOLOGY: It is rapidly absorbed and has a very rapid onset of action.
    D) TOXICOLOGY: Cyanide is a general cellular poison resulting in impaired oxygen utilization and lactic acidosis. It interferes with oxidative phosphorylation by inhibiting cytochrome oxidase which prevents aerobic production of energy.
    E) EPIDEMIOLOGY: Rare exposure that can result in significant morbidity and mortality.
    F) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: Nausea, vomiting, headache, weakness, confusion, dizziness and shortness of breath.
    2) SEVERE TOXICITY: Coma, transient loss of consciousness with apnea ("knockdown"), hypotension, metabolic acidosis, seizures, and dysrhythmias.
    0.2.20) REPRODUCTIVE
    A) There are no reported cases of human teratogenicity due to cyanide itself; however, teratogenic effects have been observed in experimental animals exposed to cyanide and related compounds.

Laboratory Monitoring

    A) Monitor serum chemistry and lactate concentrations.
    B) Monitor arterial and venous blood gases.
    C) Cyanide levels can be measured to confirm the diagnosis, but are usually not available in a timely manner to be clinically useful.
    D) Institute continuous cardiac monitoring and obtain an ECG.
    E) Methemoglobin should also be monitored frequently in patients receiving intravenous sodium nitrite.
    F) Obtain a carboxyhemoglobin level in the setting of a fire.
    G) Consider a head CT for comatose patients.

Treatment Overview

    0.4.2) ORAL/PARENTERAL EXPOSURE
    A) MANAGEMENT OF MILD TO MODERATE TOXICITY
    1) Patients with even brief or small exposures should be monitored extremely closely for signs of deterioration. Supplemental oxygen should be administered immediately with continuous monitoring of vital signs. Establish intravenous access immediately. An antidote kit should available at the bedside. Worsening or severe acidosis, hypotension, seizures, dysrhythmias and coma indicate a more severe poisoning.
    B) MANAGEMENT OF SEVERE TOXICITY
    1) Elevated lactate, increased anion gap metabolic acidosis, and an elevated venous oxygen saturation all suggest a significant cyanide exposure. Manage airway early. Patients who are comatose or severely ill due to suspected cyanide poisoning should be administered a cyanide antidote kit. In addition, standard ACLS or PALS therapy should be provided to manage symptoms. Administer sodium bicarbonate for severe acidemia.
    C) DECONTAMINATION
    1) PREHOSPITAL: Prehospital activated charcoal can be considered for large ingestions in which there will be a delay in definitive healthcare; however, a poison center should initially be consulted.
    2) HOSPITAL: DERMAL EXPOSURE: Remove clothes and wash the skin with water. Healthcare providers should use personal protective equipment if there could be a dermal exposure. Activated charcoal binds poorly to cyanide salts; however, the lethal dose is so small that the use of activated charcoal should be considered in a patient that presents with one hour of an oral ingestion
    D) AIRWAY MANAGEMENT
    1) Patients who are comatose or with altered mental status need early endotracheal intubation and mechanical ventilation.
    E) CYANIDE ANTIDOTE
    1) SUMMARY: A cyanide antidote, either hydroxocobalamin or the sodium nitrite/sodium thiosulfate kit, should be administered to symptomatic patients. If cyanide toxicity develops concurrent with carbon monoxide poisoning (e.g., closed space fire), hydroxocobalamin is the preferred antidote. It that is not available, sodium thiosulfate may be used alone. Use of amyl nitrite or sodium nitrite will cause methemoglobinemia, further reducing oxygen carrying capacity.
    2) HYDROXOCOBALAMIN
    a) ADULT: Administer 5 g IV over 15 minutes. A second dose may be given (infused over 15 to 120 minutes) in patients with severe toxicity. PEDIATRIC: A dose of 70 mg/kg has been used. Hydroxocobalamin forms cyanocobalamin which is a nontoxic, water soluble metabolite that is eliminated in the urine. It is generally safer and easier to use than other antidotes (i.e., nitrite and thiosulfate kits). Sodium thiosulfate may also be administered with hydroxocobalamin, but it is not part of the kit. ADVERSE EFFECTS: Flushing is common. Hydroxocobalamin is bright red and causes discoloration of the skin, urine, and serum. It can also interfere with many colorimetric based tests.
    3) CYANIDE ANTIDOTE KIT
    a) An alternative, a sodium nitrite/sodium thiosulfate kit, is administered as follows: SODIUM NITRITE: ADULT: Administer 300 mg (10 mL of 3% solution) IV at a rate of 2.5 to 5 mL/min; PEDIATRIC (with normal hemoglobin concentration): 0.2 mL/kg of a 3% solution (6 mg/kg) IV at a rate of 2.5 to 5 mL/min, not to exceed 10 mL (300 mg). The dose may be lowered if the patient is severely anemic, but administration should not be delayed for laboratory results. Blood methemoglobin levels should be monitored for 30 to 60 minutes following the infusion to prevent severe toxicity. If methemoglobin concentration is greater than 30%, it should likely be reversed with methylene blue. Nitrites may also cause vasodilatory effects which may contribute to hypotension. A second dose, one-half of the first dose, may be administered 30 minutes later if there is inadequate clinical response. Use with caution if carbon monoxide poisoning is also suspected. SODIUM THIOSULFATE: Follow sodium nitrite with IV sodium thiosulfate. ADULT: Administer 50 mL (12.5 g) of a 25% solution IV; PEDIATRIC: 1 mL/kg of a 25% solution (250 mg/kg), not to exceed 50 mL (12.5 g) total dose. A second dose, one-half of the first dose, may be administered if signs of cyanide toxicity reappear. This agent enhances conversion of cyanide to thiocyanate which is eliminated in the urine. Patients with renal failure may need dialysis to eliminate thiocyanate. ALTERNATE ANTIDOTES: Kelocyanor(R) (dicobalt-EDTA) and 4-DMAP (4-dimethylaminophenol) are among the cyanide antidotes in clinical use outside the US.
    F) METHEMOGLOBINEMIA
    1) Initiate oxygen therapy. Treat with methylene blue if patient is symptomatic (usually at methemoglobin concentrations greater than 20% to 30% or at lower concentrations in patients with anemia, underlying pulmonary or cardiovascular disease). METHYLENE BLUE: INITIAL DOSE/ADULT OR CHILD: 1 mg/kg IV over 5 to 30 minutes; a repeat dose of up to 1 mg/kg may be given 1 hour after the first dose if methemoglobin levels remain greater than 30% or if signs and symptoms persist. NOTE: Methylene blue is available as follows: 50 mg/10 mL (5 mg/mL or 0.5% solution) single-dose ampules and 10 mg/1 mL (1% solution) vials. Additional doses may sometimes be required. Improvement is usually noted shortly after administration if diagnosis is correct. Consider other diagnoses or treatment options if no improvement has been observed after several doses. If intravenous access cannot be established, methylene blue may also be given by intraosseous infusion. Methylene blue should not be given by subcutaneous or intrathecal injection. NEONATES: DOSE: 0.3 to 1 mg/kg.
    G) ENHANCED ELIMINATION
    1) Antidotes increase elimination, however, the role of hemodialysis is uncertain.
    H) PATIENT DISPOSITION
    1) HOME CRITERIA: There is no role for home management of cyanide exposure.
    2) OBSERVATION CRITERIA: Any exposure to cyanide salts or cyanide gas should be referred to a healthcare facility. Patients who remain asymptomatic with normal laboratory studies can be discharged after 6 hours.
    3) ADMISSION CRITERIA: Any patient with symptomatic poisoning should be admitted to an intensive care unit.
    4) CONSULT CRITERIA: Consult a poison center or medical toxicologist for assistance in managing symptomatic patients.
    I) PITFALLS
    1) Treatment should not be delayed for laboratory results if a cyanide exposure is strongly suspected. If cyanide toxicity develops concurrent with carbon monoxide poisoning (e.g., closed space fire), hydroxocobalamin is the preferred antidote. It that is not available, sodium thiosulfate may be used alone. Use of amyl nitrite or sodium nitrite will cause methemoglobinemia, further reducing oxygen carrying capacity.
    J) PHARMACOKINETICS
    1) The volume of distribution is not completely known, but cyanide is widely distributed to organs and tissues. The half-life for conversion of a nonlethal dose of cyanide to thiocyanate is 20 to 60 minutes, but the elimination half-life is not completely known.
    K) TOXICOKINETICS
    1) The amount, duration of exposure, route of exposure and premorbid conditions of the patient will affect the time and severity of the poisoning. Symptoms typically begin within 5 minutes of a significant inhalational exposure and within 30 minutes of a significant oral exposure.
    L) DIFFERENTIAL DIAGNOSIS
    1) Carbon monoxide and hydrogen sulfide gas exposures may cause transient loss consciousness with apnea, coma, and acidosis. The differential of an elevated anion gap acidosis is otherwise extremely broad and consists of toxicologic and non-toxicologic causes.
    0.4.3) INHALATION EXPOSURE
    A) INHALATION: Move patient to fresh air. Monitor for respiratory distress. If cough or difficulty breathing develops, evaluate for respiratory tract irritation, bronchitis, or pneumonitis. Administer oxygen and assist ventilation as required. Treat bronchospasm with an inhaled beta2-adrenergic agonist. Consider systemic corticosteroids in patients with significant bronchospasm.
    B) Administer 100% oxygen, establish secure large bore IV.
    C) CYANIDE ANTIDOTE SUMMARY
    1) A cyanide antidote, either hydroxocobalamin or the sodium nitrite/sodium thiosulfate kit, should be administered to patients with symptomatic poisoning. If cyanide toxicity develops concurrent with carbon monoxide poisoning (e.g., closed space fire), hydroxocobalamin is the preferred antidote. It that is not available, sodium thiosulfate may be used alone. Use of amyl nitrite or sodium nitrite will cause methemoglobinemia, further reducing oxygen carrying capacity.
    D) HYDROXOCOBALAMIN
    1) ADULT: Administer 5 g IV over 15 minutes. A second dose may be given (infused over 15 to 120 minutes) in patients with severe toxicity. PEDIATRIC: A dose of 70 mg/kg has been used. Hydroxocobalamin forms cyanocobalamin which is a nontoxic, water soluble metabolite that is eliminated in the urine. It is generally safer and easier to use than other antidotes (i.e., nitrite and thiosulfate kits). Sodium thiosulfate may also be administered with hydroxocobalamin, but it is not part of the kit. ADVERSE EFFECTS: Flushing is common. Hydroxocobalamin is bright red and causes discoloration of the skin, urine, and serum. It can also interfere with many colorimetric based tests.
    E) CYANIDE ANTIDOTE KIT
    1) An alternative, a sodium nitrite/sodium thiosulfate kit, is administered as follows: SODIUM NITRITE: ADULT: Administer 300 mg (10 mL of 3% solution) IV at a rate of 2.5 to 5 mL/min; PEDIATRIC (with normal hemoglobin concentration): 0.2 mL/kg of a 3% solution (6 mg/kg) IV at a rate of 2.5 to 5 mL/min, not to exceed 10 mL (300 mg). The dose may be lowered if the patient is severely anemic, but administration should not be delayed for laboratory results. Blood methemoglobin levels should be monitored for 30 to 60 minutes following the infusion to prevent severe toxicity. If methemoglobin concentration is greater than 30%, it should likely be reversed with methylene blue. Nitrites may also cause vasodilatory effects which may contribute to hypotension. A second dose, one-half of the first dose, may be administered 30 minutes later if there is inadequate clinical response. Use with caution if carbon monoxide poisoning is also suspected. SODIUM THIOSULFATE: Follow sodium nitrite with IV sodium thiosulfate. ADULT: Administer 50 mL (12.5 g) of a 25% solution IV; PEDIATRIC: 1 mL/kg of a 25% solution (250 mg/kg), not to exceed 50 mL (12.5 g) total dose. A second dose, one-half of the first dose, may be administered if signs of cyanide toxicity reappear. This agent enhances conversion of cyanide to thiocyanate which is eliminated in the urine. Patients with renal failure may need dialysis to eliminate thiocyanate. ALTERNATE ANTIDOTES: Kelocyanor(R) (dicobalt-EDTA) and 4-DMAP (4-dimethylaminophenol) are among the cyanide antidotes in clinical use outside the US.
    F) METHEMOGLOBINEMIA
    1) Initiate oxygen therapy. Treat with methylene blue if patient is symptomatic (usually at methemoglobin concentrations greater than 20% to 30% or at lower concentrations in patients with anemia, underlying pulmonary or cardiovascular disease). METHYLENE BLUE: INITIAL DOSE/ADULT OR CHILD: 1 mg/kg IV over 5 to 30 minutes; a repeat dose of up to 1 mg/kg may be given 1 hour after the first dose if methemoglobin levels remain greater than 30% or if signs and symptoms persist. NOTE: Methylene blue is available as follows: 50 mg/10 mL (5 mg/mL or 0.5% solution) single-dose ampules and 10 mg/1 mL (1% solution) vials. Additional doses may sometimes be required. Improvement is usually noted shortly after administration if diagnosis is correct. Consider other diagnoses or treatment options if no improvement has been observed after several doses. If intravenous access cannot be established, methylene blue may also be given by intraosseous infusion. Methylene blue should not be given by subcutaneous or intrathecal injection. NEONATES: DOSE: 0.3 to 1 mg/kg.
    0.4.4) EYE EXPOSURE
    A) DECONTAMINATION: Remove contact lenses and irrigate exposed eyes with copious amounts of room temperature 0.9% saline or water for at least 15 minutes. If irritation, pain, swelling, lacrimation, or photophobia persist after 15 minutes of irrigation, the patient should be seen in a healthcare facility.
    0.4.5) DERMAL EXPOSURE
    A) OVERVIEW
    1) DECONTAMINATION: Remove contaminated clothing and jewelry and place them in plastic bags. Wash exposed areas with soap and water for 10 to 15 minutes with gentle sponging to avoid skin breakdown. A physician may need to examine the area if irritation or pain persists (Burgess et al, 1999).
    2) While cyanide can be absorbed through intact skin, most reported cases have involved whole-body immersion in cyanide solutions or large-area burns with molten cyanide solutions. Most nitrile compounds are well absorbed through intact skin, and may cause delayed onset of symptoms following exposure by this route.
    3) Administer 100% oxygen, establish secure large bore IV.
    4) CYANIDE ANTIDOTE SUMMARY
    a) A cyanide antidote, either hydroxocobalamin or the sodium nitrite/sodium thiosulfate kit, should be administered to patients with symptomatic poisoning. If cyanide toxicity develops concurrent with carbon monoxide poisoning (e.g., closed space fire), hydroxocobalamin is the preferred antidote. It that is not available, sodium thiosulfate may be used alone. Use of amyl nitrite or sodium nitrite will cause methemoglobinemia, further reducing oxygen carrying capacity.
    5) HYDROXOCOBALAMIN
    a) ADULT: Administer 5 g IV over 15 minutes. A second dose may be given (infused over 15 to 120 minutes) in patients with severe toxicity. PEDIATRIC: A dose of 70 mg/kg has been used. Hydroxocobalamin forms cyanocobalamin which is a nontoxic, water soluble metabolite that is eliminated in the urine. It is generally safer and easier to use than other antidotes (i.e., nitrite and thiosulfate kits). Sodium thiosulfate may also be administered with hydroxocobalamin, but it is not part of the kit. ADVERSE EFFECTS: Flushing is common. Hydroxocobalamin is bright red and causes discoloration of the skin, urine, and serum. It can also interfere with many colorimetric based tests.
    6) CYANIDE ANTIDOTE KIT
    a) An alternative, a sodium nitrite/sodium thiosulfate kit, is administered as follows: SODIUM NITRITE: ADULT: Administer 300 mg (10 mL of 3% solution) IV at a rate of 2.5 to 5 mL/min; PEDIATRIC (with normal hemoglobin concentration): 0.2 mL/kg of a 3% solution (6 mg/kg) IV at a rate of 2.5 to 5 mL/min, not to exceed 10 mL (300 mg). The dose may be lowered if the patient is severely anemic, but administration should not be delayed for laboratory results. Blood methemoglobin levels should be monitored for 30 to 60 minutes following the infusion to prevent severe toxicity. If methemoglobin concentration is greater than 30%, it should likely be reversed with methylene blue. Nitrites may also cause vasodilatory effects which may contribute to hypotension. A second dose, one-half of the first dose, may be administered 30 minutes later if there is inadequate clinical response. SODIUM THIOSULFATE: Follow sodium nitrite with IV sodium thiosulfate. ADULT: Administer 50 mL (12.5 g) of a 25% solution IV; PEDIATRIC: 1 mL/kg of a 25% solution (250 mg/kg), not to exceed 50 mL (12.5 g) total dose. A second dose, one-half of the first dose, may be administered if signs of cyanide toxicity reappear. This agent enhances conversion of cyanide to thiocyanate which is eliminated in the urine. Patients with renal failure may need dialysis to eliminate thiocyanate. ALTERNATE ANTIDOTES: Kelocyanor(R) (dicobalt-EDTA) and 4-DMAP (4-dimethylaminophenol) are among the cyanide antidotes in clinical use outside the US.
    7) METHEMOGLOBINEMIA
    a) Initiate oxygen therapy. Treat with methylene blue if patient is symptomatic (usually at methemoglobin concentrations greater than 20% to 30% or at lower concentrations in patients with anemia, underlying pulmonary or cardiovascular disease). METHYLENE BLUE: INITIAL DOSE/ADULT OR CHILD: 1 mg/kg IV over 5 to 30 minutes; a repeat dose of up to 1 mg/kg may be given 1 hour after the first dose if methemoglobin levels remain greater than 30% or if signs and symptoms persist. NOTE: Methylene blue is available as follows: 50 mg/10 mL (5 mg/mL or 0.5% solution) single-dose ampules and 10 mg/1 mL (1% solution) vials. Additional doses may sometimes be required. Improvement is usually noted shortly after administration if diagnosis is correct. Consider other diagnoses or treatment options if no improvement has been observed after several doses. If intravenous access cannot be established, methylene blue may also be given by intraosseous infusion. Methylene blue should not be given by subcutaneous or intrathecal injection. NEONATES: DOSE: 0.3 to 1 mg/kg.

Range Of Toxicity

    A) TOXIC DOSE: The threshold value limit for hydrogen cyanide is 10 parts per million (ppm) for an 8 hour day. A dose of potassium cyanide of 200 mg is likely lethal; however, an adult survived an ingestion of 1200 to 1500 mg potassium cyanide powder without visual or neurologic sequelae following cyanide antidotal therapy with hydroxocobalamin immediately followed by sodium thiosulfate. An immediate inhalational exposure of 270 ppm of hydrogen cyanide gas is likely lethal.

Summary Of Exposure

    A) SOURCES: Cyanide is primarily found as either a solid cyanide salt, a salt solution, or as hydrogen cyanide gas. It may be liberated as cyanide gas in house fires from the combustion of wool, silk, synthetic rubber, and polyurethane. It was previously found in artificial nail removers as acetonitrile.
    B) USES: Various industries including metal extraction and refining, electroplating, photography, manufacturing, and fumigation.
    C) PHARMACOLOGY: It is rapidly absorbed and has a very rapid onset of action.
    D) TOXICOLOGY: Cyanide is a general cellular poison resulting in impaired oxygen utilization and lactic acidosis. It interferes with oxidative phosphorylation by inhibiting cytochrome oxidase which prevents aerobic production of energy.
    E) EPIDEMIOLOGY: Rare exposure that can result in significant morbidity and mortality.
    F) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: Nausea, vomiting, headache, weakness, confusion, dizziness and shortness of breath.
    2) SEVERE TOXICITY: Coma, transient loss of consciousness with apnea ("knockdown"), hypotension, metabolic acidosis, seizures, and dysrhythmias.

Heent

    3.4.3) EYES
    A) WITH POISONING/EXPOSURE
    1) MYDRIASIS: Dilated pupils are common in severe poisoning (Zakharov et al, 2015; Vogel et al, 1981).
    2) CONJUNCTIVITIS and lid edema have been reported in men exposed to cyanogen chloride (ACGIH, 1986; Grant, 1986). Eye irritation has been described in workers chronically to cyanide (Proctor et al, 1988).
    3) CORNEAL EDEMA: One case of corneal edema from exposure to hydrocyanic acid vapors has been reported (Grant, 1986).
    4) FUNDUSCOPIC EXAMINATION: Retinal arteries and veins that appear equally red on funduscopic examination suggest the diagnosis (Buchanan et al, 1976). Central retinal edema has been reported in a fatal case of cyanide poisoning (Grant, 1986).
    5) OPTIC NEURITIS: Chronic exposure to low levels of cyanide (as in cigarette smoking) coupled with either a defective rhodanese enzyme or protein-calorie malnutrition with decreased sulfur-containing amino acids intake has been postulated to lead to retrobulbar optic neuritis in certain cases (Grant, 1986). Damaged optic nerves have been observed in experimental animals injected with lethal or near lethal doses of cyanide (Grant, 1986).
    6) BLINDNESS is possible with cyanide-induced damage to optic nerves and retina (Grant, 1986).
    7) TRANSIENT BLINDNESS has been reported rarely in instances of sublethal cyanide poisoning (Vogel et al, 1981; Grant, 1986).
    8) EYE EXPOSURE/SYSTEMIC ABSORPTION: Accidental eye contamination with industrial chemicals may produce systemic symptoms (Ballantyne, 1983).
    a) EYE EXPOSURE: When solid particles or concentrated solutions of NaCN, KCN or HCN were instilled into rabbit eyes, the product was rapidly absorbed producing systemic toxicity and death (Ballantyne, 1983).
    9) CASE REPORT: A 30-year-old man experienced bilateral vision loss after exposure to sodium cyanide gas for approximately 1 hour without wearing a gas mask while working in a gold mine. Despite receiving antidotal therapy and supportive care, the patient's vision showed only a gradual improvement with a visual acuity of 20/400 in both eyes that persisted 5 months after exposure. The slit-lamp microscope examination, the Farnsworth dichotomous color vision test, the retinal nerve fiber layer examination, and flash electroretinography were normal; however, T2-weighted MRI scans showed bilateral high signal intensities of the putamen, and the amplitudes of the visual evoked potential and the multifocal visual evoked potential were reduced, indicating damage to the visual pathway believed to be a result of acute cyanide poisoning (Chen et al, 2011).
    3.4.4) EARS
    A) CHRONIC EXPOSURE: Functional hearing changes were described in a group of workers chronically exposed to cyanide (Saia et al, 1970).
    B) HEARING LOSS: Loss of hearing and severe perceptive deafness have been suggested in chronic cyanide poisonings caused by insufficiently processed cassava (van Heijst et al, 1994).
    3.4.5) NOSE
    A) WITH POISONING/EXPOSURE
    1) EPISTAXIS: Frequent nosebleeds have been described in workers chronically exposed to cyanide (Proctor et al, 1988).
    2) SEPTAL PERFORATION: Nasal obstruction, bleeding, and sloughs have occurred among workers in electroplating industry (ILO, 1983).
    3) IRRITATION: Nasal irritation leading to obstruction, bleeding, sloughs, and in some cases perforation of the septum has been reported in workers in the electroplating industry (Finkel, 1983; HSDB , 1991).
    3.4.6) THROAT
    A) WITH POISONING/EXPOSURE
    1) BURNING SENSATION in the mouth and throat may occur (Vogel et al, 1981).
    2) HOARSENESS occurred with chronic exposure to cyanogen chloride (ACGIH, 1986; Grant, 1986).

Cardiovascular

    3.5.2) CLINICAL EFFECTS
    A) TACHYARRHYTHMIA
    1) WITH POISONING/EXPOSURE
    a) Tachycardia and hypertension may be seen in the initial phases of cyanide poisoning (Garlich et al, 2012; Vogel et al, 1981).
    B) BRADYCARDIA
    1) WITH POISONING/EXPOSURE
    a) Bradycardia and hypotension are seen in the late phases of cyanide poisoning (Garlich et al, 2012; Hall & Rumack, 1986; Stewart, 1974) .
    b) CASE REPORT: A 17-year-old boy developed severe metabolic acidosis, hypotension, and bradycardia after unintentionally ingesting a beverage containing 1.5 g potassium cyanide. Despite administration of antidote therapy and supportive care, the patient continued to show hemodynamic instability. An MRI revealed complete cerebellar infarction, and the patient was declared brain dead approximately 60 hours post-ingestion (Peddy et al, 2006).
    C) ELECTROCARDIOGRAM ABNORMAL
    1) WITH POISONING/EXPOSURE
    a) Erratic atrial and ventricular cardiac rhythms with varying degrees of atrioventricular block followed by asystole may be seen in severe cyanide poisoning (Hall & Rumack, 1986). ST-T segment elevation or depression may occur (Cope, 1961).
    b) CASE REPORT: A 24-year-old man presented to the emergency department unresponsive (Glasgow Coma Scale score of 8) and foaming at the mouth approximately 2 hours after reportedly ingesting potassium cyanide. Laboratory data revealed metabolic acidosis (pH 6.9, pCO2 24 mmHg, HCO3 4.8 mmol/L), a serum lactate level of 29 mmol/L, and a blood cyanide concentration of 4.02 mcg/mL. Urine drug screen was positive for acetaminophen and benzodiazepines. An ECG demonstrated atrial fibrillation with a QRS of 116 ms and a QTc of 572 msec, with a heart rate of 116 beats/min. Treatment included administration of IV fluids, IV sodium bicarbonate, and an IV infusion of hydroxocobalamin. Following hydroxocobalamin therapy, the patient's serum lactate normalized to 1.4 mmol/L, his acidosis resolved, and a repeat ECG revealed normal sinus rhythm, with narrowing of his QRS complex (92 msec), a shortening of his QTc interval (406 msec) and a decrease in his heart rate to 71 beats/min. The patient was discharged to an inpatient psychiatric unit within 48 hours of presentation (Johnson et al, 2015).
    c) CASE REPORT: First degree atrioventricular block and bradycardia (40 beats/min) were reported in a 33-year-old who ingested potassium cyanide in a suicide attempt (Hsiao et al, 2015).
    D) PALPITATIONS
    1) WITH POISONING/EXPOSURE
    a) Palpitations have been described in workers chronically exposed to cyanide (Colle, 1972).
    E) HYPOTENSIVE EPISODE
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 30-year-old woman developed severe hypotension, coma, and severe metabolic acidosis after injecting herself with cyanide subcutaneously (blood cyanide ion level 4.6 mcg/mL) . After 4 hours of hemodialysis and 48 hours of supportive care, she recovered and was discharged without further sequelae (Prieto et al, 2005).
    b) CASE REPORT: A 17-year-old boy developed severe metabolic acidosis, hypotension, and bradycardia after unintentionally ingesting a beverage containing 1.5 g potassium cyanide. Despite administration of antidote therapy and supportive care, the patient continued to show hemodynamic instability. An MRI revealed complete cerebellar infarction, and the patient was declared brain dead approximately 60 hours post-ingestion (Peddy et al, 2006).
    c) CASE REPORT: A 29-year-old man presented with coma (Glasgow Coma Scale score of 3), tachypnea (35 to 45 breaths/minute), tachycardia (95 beats/minute), and hypotension (80/60 mmHg). Whole blood cyanide concentration was 6.9 mg/L and blood alcohol concentration was 270 mg/dL. Further questioning of the patient determined that he had intentionally ingested approximately 50 grams potassium dicyanoaurate (containing cyanide 25 mg/g; total amount of cyanide ingested 1250 mg [a 5-fold lethal dose of cyanide]). Following antidotal therapy with sodium thiosulfate and supportive care, the patient recovered uneventfully and was discharged to a psychiatric unit 2 days postingestion (Kampe et al, 2000).

Respiratory

    3.6.2) CLINICAL EFFECTS
    A) HYPERVENTILATION
    1) WITH POISONING/EXPOSURE
    a) Part of the initial presentation of cyanide poisoning may be hyperpnea and tachypnea (Kampe et al, 2000; Hall & Rumack, 1986).
    b) CASE REPORT: A 29-year-old man presented with coma (Glasgow Coma Scale score of 3), tachypnea (35 to 45 breaths/minute), tachycardia (95 beats/minute), and hypotension (80/60 mmHg). Whole blood cyanide concentration was 6.9 mg/L and blood alcohol concentration 270 mg/dL. Further questioning of the patient determined that he had intentionally ingested approximately 50 grams potassium dicyanoaurate (containing cyanide 25 mg/g; total amount of cyanide ingested 1250 mg [a 5-fold lethal dose of cyanide]). Following antidotal therapy with sodium thiosulfate and supportive care, the patient recovered uneventfully and was discharged to a psychiatric unit 2 days postingestion (Kampe et al, 2000).
    B) APNEA
    1) WITH POISONING/EXPOSURE
    a) Hypoventilation progressing to apnea may be seen in the later phases of cyanide poisoning and is a major cause of death (Vogel et al, 1981).
    b) CASE REPORT: A 17-year-old boy presented with coma, apnea, and seizures. On arrival at the emergency department, his Glasgow Coma Scale score was 3 with nonreactive pupils and absent corneal, gag, and cough reflexes. Venous blood gas analysis showed metabolic acidosis, with elevated anion gap and lactate levels, and evidence of decreased tissue oxygen utilization with an arteriovenous saturation difference of zero. The patient also developed persistent hypotension and bradycardia. Suspecting cyanide poisoning, the patient was given a cyanide antidote kit, consisting of 600 mg of sodium nitrite followed by 25 g of sodium thiosulfate. His bradycardia resolved within minutes and his blood pressure improved; however, over the next several hours, his hypotension recurred. The cyanide antidote kit was readministered four separate times because of fluctuating arteriovenous saturation differences, but the sodium nitrite infusion was discontinued due to a methemoglobin level of 35%. Over the next 48 hours, the patient continued to be hemodynamically unstable. An MRI showed complete cerebellar infarction and severe edema in the brain stem and upper spinal cord. He was declared brain dead approximately 60 hours following presentation. A police investigation reported that the patient had been poisoned with 1.5 grams of potassium cyanide placed in his beverage (Peddy et al, 2006).
    C) ACUTE LUNG INJURY
    1) WITH POISONING/EXPOSURE
    a) Noncardiogenic pulmonary edema has been reported in cases of acute cyanide poisoning (Graham et al, 1977). Pulmonary edema can also occur with chronic cyanogen chloride exposure (Gosselin et al, 1984). Pulmonary edema may result from a direct effect on the myocardium leading to left ventricular failure and increased pulmonary venous pressure.
    D) CYANOSIS
    1) WITH POISONING/EXPOSURE
    a) Cyanosis is generally a late finding and usually does not occur until circulatory collapse and tachycardia are evident, particularly at the premorbid stage of cyanide toxicity (Hall & Rumack, 1986).
    E) RESPIRATORY FINDING
    1) WITH POISONING/EXPOSURE
    a) CHRONIC: Respiratory tract irritation, chest discomfort, effort dyspnea, and varying degrees of persistent rhinitis, nasal obstruction, and bleeding have been described in workers chronically exposed to cyanide (Proctor et al, 1988; Saia et al, 1970; Colle, 1972; Finkel, 1983).

Neurologic

    3.7.2) CLINICAL EFFECTS
    A) HEADACHE
    1) WITH POISONING/EXPOSURE
    a) Headache may be an early sign of cyanide poisoning (Vogel et al, 1981).
    B) CENTRAL STIMULANT ADVERSE REACTION
    1) WITH POISONING/EXPOSURE
    a) Central nervous stimulation with varied presentations from anxiety to agitation and combative behavior may be seen in the early stages of cyanide poisoning (Vogel et al, 1981).
    C) COMA
    1) WITH POISONING/EXPOSURE
    a) Coma is common in severe poisoning (Johnson et al, 2015; Lenart et al, 2010; Hall & Rumack, 1986; Vogel et al, 1981).
    b) CASE REPORT: A 30-year-old woman developed severe hypotension, coma, and severe metabolic acidosis after injecting herself with cyanide subcutaneously (blood cyanide ion level 4.6 mcg/mL). After 4 hours of hemodialysis and 48 hours of supportive care, she recovered and was discharged without further sequelae (Prieto et al, 2005).
    c) CASE REPORT: A 17-year-old boy presented with coma, apnea, and seizures. On arrival at the emergency department, his Glasgow Coma Scale score was 3 with nonreactive pupils and absent corneal, gag, and cough reflexes. Venous blood gas analysis showed metabolic acidosis, with elevated anion gap and lactate levels, and evidence of decreased tissue oxygen utilization with an arteriovenous saturation difference of zero. The patient also developed persistent hypotension and bradycardia. Suspecting cyanide poisoning, the patient was given a cyanide antidote kit, consisting of 600 mg of sodium nitrite followed by 25 g of sodium thiosulfate. His bradycardia resolved within minutes and his blood pressure improved; however, over the next several hours, his hypotension recurred. The cyanide antidote kit was readministered four separate times because of fluctuating arteriovenous saturation differences, but the sodium nitrite infusion was discontinued due to a methemoglobin level of 35%. Over the next 48 hours, the patient continued to be hemodynamically unstable. An MRI showed complete cerebellar infarction and severe edema in the brain stem and upper spinal cord. He was declared brain dead approximately 60 hours following presentation. A police investigation reported that the patient had been poisoned with 1.5 grams of potassium cyanide placed in his beverage (Peddy et al, 2006).
    d) CASE REPORT: A 29-year-old man presented with coma (Glasgow Coma Scale score of 3), tachypnea (35 to 45 breaths/minute), tachycardia (95 beats/minute), and hypotension (80/60 mmHg). Whole blood cyanide concentration was 6.9 mg/L and blood alcohol concentration was 270 mg/dL. Further questioning of the patient determined that he had intentionally ingested approximately 50 grams potassium dicyanoaurate (containing cyanide 25 mg/g; total amount of cyanide ingested 1250 mg [a 5-fold lethal dose of cyanide]). Following antidotal therapy with sodium thiosulfate and supportive care, the patient recovered uneventfully and was discharged to a psychiatric unit 2 days postingestion (Kampe et al, 2000).
    e) CASE REPORT: A 33-year-old woman presented to the emergency department comatose (Glasgow Coma Scale [GCS] score of E1M1V2). An ECG indicated first-degree atrioventricular block and bradycardia (40 bpm), and arterial blood gas analysis revealed high anion gap metabolic acidosis (pH 6.81, PaCO2 24.7 mmHg, HCO3 3.8 mmol/L, anion gap 36.2). With supportive therapy, including administration of IV sodium bicarbonate and IV fluids, the patient regained consciousness (GCS E4M6Vt) 4 hours post-admission, with gradual improvement of laboratory parameters. Interview of the patient revealed that she had ingested a "small" amount of potassium cyanide (serum cyanide level was 3.5 mg/L) in a suicide attempt. The patient was discharged 7 days later following an uneventful recovery. There was no evidence of neurological sequelae at 6 months follow-up (Hsiao et al, 2015).
    f) CASE REPORT: A 58-year-old man intentionally ingested 1200 to 1500 mg of pure potassium cyanide powder and subsequently lost consciousness for 1 to 1.5 minutes and had one episode of generalized seizures. At presentation to the hospital, the patient was severely somnolent with mydriasis and lactic acidosis. The patient required emergent intubation, analgosedation and ventilation. Cyanide antidotal treatment was initiated 1.5 hours post-ingestion with hydroxocobalamin followed by sodium thiosulfate. Following antidotal therapy, the patient's arterial blood gases normalized on the day of admission. On hospital day 3, he was extubated, and the next day was discharged without visual or neurologic sequelae, which continued at his 5-month follow-up appointment (Zakharov et al, 2015).
    D) SEIZURE
    1) WITH POISONING/EXPOSURE
    a) Seizures are common in severe cyanide poisoning (Zakharov et al, 2015; Hall & Rumack, 1986; Peddy et al, 2006).
    E) PARALYSIS
    1) WITH POISONING/EXPOSURE
    a) Opisthotonos, trismus, and paralysis were reported in one case of cyanide poisoning (De Busk & Seidl, 1969).
    F) EXTRAPYRAMIDAL DISEASE
    1) WITH POISONING/EXPOSURE
    a) An 18-year-old patient who ingested between 975 and 1,300 mg of potassium cyanide developed a parkinsonian syndrome with rigidity and akinesis (Uitti et al, 1985).
    b) Parkinsonism developed progressively over 3 weeks after acute ingestion of 1,500 mg of potassium cyanide. Slowed gait, masked facies, hypotonia, mild rigidity, and minimal tremor were noted. Damage was permanent, and although not evident on CT or MRI scan at 6 months, was found on MRI at 12 months postingestion. There was no improvement with levodopa (Rosenberg et al, 1989).
    c) A 46-year-old woman developed progressive parkinsonism over a five-year period after acute cyanide poisoning in a 46-year-old woman (Carella et al, 1988). Drooling and dysphagia developed over this time, and marked tongue and mouth dystonia developed. Apraxia of eyelid was also apparent. Some improvement occurred with trihexyphenidyl treatment.
    d) A 39-year-old man who had suffered form long term psychosis attempted suicide by swallowing an unknown amount of sodium cyanide. CT scan of the head was normal on admission. The patient developed parkinsonian syndrome with severe dystonia. A follow up CT scan of the head showed mild cortical atrophy and bilateral lucencies in the putamen and external globus pallidus (Grandas et al, 1989).
    e) A 28-year-old man developed severe parkinsonian symptoms, micrographia, hypersalivation, and bilateral basal ganglia abnormalities on MRI after ingestion of 800 mg of potassium cyanide (Feldman & Feldman, 1990). With intensive medical and psychotherapeutic intervention, he survived, although with persistent deficits.
    f) A severe dystonia syndrome, with slurred speech, involuntary athetoid movements of the neck, truzakharnk, and limbs, generalized dystonia, hyperextension of the lower limbs and trunk, and occasional opisthotonoid posturing, developed a few days after apparent full recovery from ingestion of 1 gram of potassium cyanide in a 16-year-old man (Valenzuela et al, 1992).
    1) He responded to levodopa 3600 mg/day, which was given for 38 days. Follow-up 21 years later showed mild dystonia, athetoid hand movements, and symmetrical putamen hypodensities on CT scan.
    g) CASE REPORT: After ingesting potassium cyanide in a suicide attempt, a 35-year-old woman survived following antidote therapy and supportive care. On day 5, she developed agitation, choreoathetotic involuntary stereotypical movements of the upper and lower extremities and the trunk for several days. It was followed by akinetic mutism and buccofacial and ideomotoric aphasia. She also experienced severe rigid-akinetic syndrome, dysarthria, dysphagia and generalized dystonia several weeks later. MRI showed lesions in the caudate and lentiform nuclei, precentral cortex and cerebellum with a cortical precentral perfusion defect consistent with pseudolaminar cortical necrosis. On two occasions, a progressive loss of dopamine transporter suggestive of nigral neuronal apoptosis was observed by SPECT with beta-CIT (SPECT by [I-123] 2 beta-carbomethoxy-3-beta-(4-iodophenyl)-Tropan). FDG-PET and HMPAO SPECT showed striatal and frontal hypometabolism and hypoperfusion (Zaknun et al, 2005).
    h) CASE REPORT: A parkinsonian syndrome, consisting of akinesia and rigidity, was reported in a 20-year-old man following a suicide attempt with cyanide. An MRI revealed hypoxic-ischemic changes as evidenced by decreased signals of the T1 image and increased signals on the T2 image of the lentiform and caudate nuclei, and a slightly increased T1 image signal in the cerebellar white matter. A CT scan showed hypodense areas in the putamen and the caudate nuclei. Following IV administration of a dye, fluorine 18-fluoro-2-deoxy-glucose (F-18 FDG), a PET/CT scan demonstrated diminished FDG uptake in the caudate nuclei and the cerebellum, and the absence of accumulation in bilateral putamen (Sarikaya et al, 2006).
    G) NEUROLOGICAL FINDING
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: After drinking a beverage laced with potassium cyanide prepared by another individual, a 17-year-old boy developed profound coma and died 4 days later. The neuropathological findings showed acute hypoxic-ischemic encephalopathy with edema, narrowing of the sulci and flattening of the gyri, autolysis of basal ganglia, and pseudolaminar cortical necrosis (Riudavets et al, 2005).
    b) CASE REPORT: A 32-year-old woman developed a gait disturbance progressing to writhing on the ground, decreased conscious state, Glasgow Coma Score fluctuating between 6 and 11, pulse between 120 and 130 bpm, hypotension, tachypnea, and fixed and dilated pupils after taking 6 amygdalin tablets. She was intubated and developed marked metabolic acidosis, nephrogenic diabetes insipidus and profound hypotension despite fluid administration. A blood sample revealed a thiocyanate level of 445 micromol/L. After 16 hours of supportive care, she was awake, orientated and extubated. She was discharged after 3 days, symptomatically well (O'Brien et al, 2005).
    c) CASE REPORT: A 27-year-old male soldier was found in his barracks unresponsive, pulseless, and in respiratory distress after ingesting tobacco contaminated with potassium cyanide and heroin. Following successful resuscitation and supportive care, the patient regained consciousness, but complained of pain in his left upper and left lower extremities, with limited range of motion of his arm. Over the course of several months, the patient continued to experience moderate to severe pain (pain scale of 7/10), with numbness of his left shoulder and upper extremity and extending down to his fourth and fifth digit, and paresthesias in his left foot. Following a 4-day inpatient treatment with IV ketamine and ropivacaine therapy, the patient had complete resolution of pain and improved range of motion. At a four-month follow-up, the patient's pain continued to be well-controlled (pain scale averaging 2/10) (Lenart et al, 2010).
    H) SEQUELA
    1) WITH POISONING/EXPOSURE
    a) Most victims of acute poisoning either die acutely or fully recover. However, cases of patients developing neurological sequelae such as personality changes, paranoid psychosis, memory deficits, and extrapyramidal syndromes have been reported (Feldman & Feldman, 1990; Grandas et al, 1989; Jouglard et al, 1971; Jouglard et al, 1974; Uitti et al, 1985; Borgohain et al, 1995; Rosenow et al, 1995) (Kales et al, 1997).
    b) In 2 patients, neurological sequelae of oral cyanide intoxication were evaluated clinically and neuropsychologically. The clinical syndrome was characterized by extrapyramidal motor and cerebral symptoms such as bradykinesia, hypomimea, slowed speech, anteropulsion, and marked retropulsion, but with little tremor (Rosenow, 1995).
    c) Delayed onset of generalized dystonia followed a suicide attempt with potassium cyanide by a 27 year old woman (Borgohain et al, 1995).
    I) CHRONIC POISONING
    1) WITH POISONING/EXPOSURE
    a) SUMMARY: Chronic exposure may produce headache, vertigo, tremors, weakness, fatigue, dizziness, confusion, functional changes in hearing, motor aphasia, optic neuropathy, seizures, paresis/hemiparesis, myelopathy, and permanent mental impairment. Demyelinating nervous effects have been indicated by epidemiological studies of populations ingesting naturally occurring plant glucosides, most notably inadequately processed cassava.
    b) CENTRAL NERVOUS SYSTEM: A case of progressive mental deterioration, hypoacusis, lower facial deficit, homolateral palpebral ptosis, and fronto-occipital headache has been reported following chronic exposure (Mentesana, 1961).
    c) SEIZURES have been reported from chronic occupational exposure (Finkel, 1983).
    d) HEADACHE: Frequent headaches, vertigo, fatigue, poor appetite, and sleeping disturbances have been described in workers chronically exposed to cyanide (Saia et al, 1970; Colle, 1972).
    e) NEUROPATHY: Study results are mixed. Optic neuropathy, sensory ataxia, and spastic paresis with myelopathy of the corticospinal tracts have been ascribed to the high cyanide content of unprocessed cassava (Casadei et al, 1990; Cliff et al, 1986; Freeman, 1988; van Heijst et al, 1994).
    1) In a more recent two-year long, cohort study, the incidence of endemic ataxic polyneuropathy was examined. The use of cassava foods (a regional food which contains cyanogenic compounds) was not associated with an increase in the incidence of ataxic polyneuropathy. The authors concluded that cyanide was not the cause of endemic ataxic polyneuropathy in this region (Oluwole et al, 2003).

Gastrointestinal

    3.8.2) CLINICAL EFFECTS
    A) VOMITING
    1) WITH POISONING/EXPOSURE
    a) Nausea, vomiting, and abdominal pain may occur, especially after ingestion of cyanide salts (Hall & Rumack, 1986; Singh et al, 1989; Vogel et al, 1981).
    b) The odor of bitter almonds in expired breath or gastric contents of patients may not be detected by a significant portion of the population (Hall & Rumack, 1986; HSDB , 1991).
    B) GASTRIC ULCER
    1) WITH POISONING/EXPOSURE
    a) IRRITATION: Ingestion of cyanide salts can cause irritation or corrosion of the esophageal or gastric mucosa (HSDB, 1990) (Jouglard et al, 1974).
    C) CHRONIC POISONING
    1) WITH POISONING/EXPOSURE
    a) Nausea with occasional vomiting has been described in workers chronically exposed to cyanide (Colle, 1972; Saia et al, 1970). Anemia has also been reported (Gettler & St George, 1934).
    D) SMELL OF BREATH - FINDING
    1) WITH POISONING/EXPOSURE
    a) ALMOND ODOR: Patients with cyanide poisoning characteristically have an odor of bitter almonds in gastric contents or expired breath, although up to 50% of the population cannot detect its presence (Bonnichsen & Maely, 1966).

Genitourinary

    3.10.2) CLINICAL EFFECTS
    A) DIABETES INSIPIDUS
    1) WITH POISONING/EXPOSURE
    a) CASE SERIES: Of 16 patients with cyanide poisoning in Taiwan, 4 died and all were preceded by polyuria. The authors suggested an association between the development of diabetes insipidus, a terminal sign of brain injury, and poor prognosis in cyanide poisonings (Deng & Yang, 1994).
    3.10.3) ANIMAL EFFECTS
    A) ANIMAL STUDIES
    1) TESTIS DISORDER
    a) TESTICULAR DEGENERATION: Subchronic dosing (12 mg of potassium cyanide injected subcutaneously for 28 doses during 6 weeks) caused degenerative changes in the testes of rats (Haguenoer et al, 1975).

Acid-Base

    3.11.2) CLINICAL EFFECTS
    A) ACIDOSIS
    1) WITH POISONING/EXPOSURE
    a) METABOLIC ACIDOSIS: Elevated anion gap metabolic acidosis and elevated serum lactate levels are frequently found in cyanide poisoning (Zakharov et al, 2015; Garlich et al, 2012; Singh et al, 1989; Hall & Rumack, 1986; Vogel et al, 1981) .
    b) CASE REPORT: A 30-year-old woman developed severe metabolic acidosis (pH 6.74; bicarbonate 5 mmol/L; anion gap 33 mmol/L) with high levels of lactic acid (19.3 mmol/L) after injecting herself with cyanide subcutaneously (blood cyanide ion level 4.6 mcg/mL) . After 4 hours of hemodialysis and 48 hours of supportive care, she recovered and was discharged without further sequelae (Prieto et al, 2005).
    c) CASE REPORT: A 32-year-old woman developed severe metabolic acidosis (pH 7.09; bicarbonate 6 mmol/L; PaCO2 18 mmHg) after taking 6 amygdalin tablets. A blood sample revealed a thiocyanate level of 445 micromol/L. After 16 hours of supportive care, she recovered and was discharged 3 days later, symptomatically well (O'Brien et al, 2005).
    d) CASE REPORT: A 17-year-old boy developed severe metabolic acidosis, hypotension, and bradycardia after unintentionally ingesting a beverage containing 1.5 g potassium cyanide. An initial blood gas analysis revealed a pH of 7.11, a bicarbonate level of 7 mEq/L, a lactic acid level of 20.3 mmol/L, and an anion gap of 38 mEq/L. Despite administration of antidote therapy and supportive care, the patient continued to show hemodynamic instability. An MRI revealed complete cerebellar infarction, and the patient was declared brain dead approximately 60 hours post-ingestion (Peddy et al, 2006).
    e) LACTIC ACIDOSIS: The degree of lactic acidosis correlates with the severity of cyanide poisoning (Naughton, 1974; Trapp, 1970).
    f) CASE REPORT: A 24-year-old man presented to the emergency department unresponsive (Glasgow Coma Scale score of 8) and foaming at the mouth approximately 2 hours after reportedly ingesting potassium cyanide. Laboratory data revealed metabolic acidosis (pH 6.9, pCO2 24 mmHg, HCO3 4.8 mmol/L), a serum lactate level of 29 mmol/L, and a blood cyanide concentration of 4.02 mcg/mL. Urine drug screen was positive for acetaminophen and benzodiazepines. An ECG demonstrated atrial fibrillation with a QRS of 116 ms and a QTc of 572 msec, with a heart rate of 116 beats/min. Treatment included administration of IV fluids, IV sodium bicarbonate, and an IV infusion of hydroxocobalamin. Following hydroxocobalamin therapy, the patient's serum lactate normalized to 1.4 mmol/L, his acidosis resolved (pH 7.49), and a repeat ECG revealed normal sinus rhythm, with narrowing of his QRS complex (92 msec), a shortening of his QTc interval (406 msec) and a decrease in his heart rate to 71 beats/min. The patient was discharged to an inpatient psychiatric unit within 48 hours of presentation (Johnson et al, 2015).
    g) CASE REPORT: A 33-year-old woman presented to the emergency department comatose (Glasgow Coma Scale [GCS] score of E1M1V2). An ECG indicated first-degree atrioventricular block and bradycardia (40 bpm), and arterial blood gas analysis revealed high anion gap metabolic acidosis (pH 6.81, PaCO2 24.7 mmHg, HCO3 3.8 mmol/L, anion gap 36.2). With supportive therapy, including administration of IV sodium bicarbonate and IV fluids, the patient regained consciousness (GCS E4M6Vt) 4 hours post-admission, with gradual improvement of laboratory parameters. Interview of the patient revealed that she had ingested a "small" amount of potassium cyanide (serum cyanide level was 3.5 mg/L) in a suicide attempt. The patient was discharged 7 days later following an uneventful recovery. There was no evidence of neurological sequelae at 6 months follow-up (Hsiao et al, 2015).

Dermatologic

    3.14.2) CLINICAL EFFECTS
    A) DISORDER OF SKIN
    1) WITH POISONING/EXPOSURE
    a) DERMAL ABSORPTION: Cyanide has been said to be absorbed through intact skin and carries a "skin designation" for workplace exposures (ACGIH, 1986) (ACGIH, 1990).
    1) However, most cases of toxicity from dermal exposure have been due to industrial accidents with immersion in vats of cyanide solutions (Bismuth et al, 1984; Dodds & McKnight, 1985) or severe, large total body area burns with molten cyanide salts (Bourrelier & Paulet, 1971). Cyanide is maintained in solution with alkali solutions, which may cause corrosive skin burns.
    B) DISCOLORATION OF SKIN
    1) WITH POISONING/EXPOSURE
    a) The skin may be a bright pink color due to high concentrations of oxyhemoglobin in the venous return (HSDB , 1991).
    C) CONTACT DERMATITIS
    1) WITH POISONING/EXPOSURE
    a) Dermal contact with the moist solid material can result in itching and skin irritation or ulceration, whereas liquid or solid sodium cyanide can cause pain and second-degree burns after a few minutes' contact (CHRIS, 1990) (Proctor et al, 1988).
    D) IRRITATION SYMPTOM
    1) WITH POISONING/EXPOSURE
    a) Chronic occupational cyanide exposure has been associated with irritation of skin and mucous membranes; such complaints occurred with exposure to highly alkaline aerosols or solutions of cyanide salts (Finkel, 1983; Hartung, 1982; Proctor et al, 1988). Papules, scarlet rash, eczema, and itching have been reported (HSDB, 1990).
    E) MACULOPAPULAR ERUPTION
    1) WITH POISONING/EXPOSURE
    a) Workers, such as electroplaters and picklers, who are daily exposed to cyanide solutions may develop a "cyanide" rash, characterized by itching, and by macular, papular, and vesicular eruptions (Lewis, 1992).
    b) CASE REPORT: A 30-year-old woman developed severe hypotension, coma, and severe metabolic acidosis after injecting herself with cyanide subcutaneously (blood cyanide ion level 4.6 mcg/mL). Six macular, erythematous lesions (1-3 cm in diameter) on her left arm and hemithorax were noted on physical examination. These lesions developed into blisters with epidermal necrosis. After 4 hours of hemodialysis and 48 hours of supportive care, she recovered and was discharged without further sequelae (Prieto et al, 2005).
    3.14.3) ANIMAL EFFECTS
    A) ANIMAL STUDIES
    1) SKIN DISORDER
    a) DERMAL ABSORPTION: Hydrogen cyanide gas was absorbed through the skin of dogs and guinea pigs; the outcome was fatal at high enough concentrations (Walton & Witherspoon, 1926).

Musculoskeletal

    3.15.2) CLINICAL EFFECTS
    A) SEQUELA
    1) WITH POISONING/EXPOSURE
    a) Permanent motor impairment may occur with chronic exposure (Baselt, 1988; Baselt & Cravey, 1989).

Endocrine

    3.16.2) CLINICAL EFFECTS
    A) HYPERGLYCEMIA
    1) WITH POISONING/EXPOSURE
    a) INSULIN RESISTANCE occurred in a severely cyanide poisoned patient (Singh et al, 1989).
    1) In tropical areas where traditionally processed cassava (containing cyanide) is consumed, diabetes is observed at an early age of onset and characterized by insulin resistance (Hugh-Jones, 1955).
    B) GOITER
    1) WITH POISONING/EXPOSURE
    a) THYROID ABNORMALITIES: Chronically cyanide-exposed workers have developed enlarged thyroid glands and decreased iodine uptake, presumably because of interference from the presence of the thiocyanate natural cyanide detoxification product (HSDB, 1990) (Finkel, 1983; Proctor et al, 1988).
    1) Abnormal thyroid function tests have been reported following chronic cyanide exposure in the occupational setting (Blanc et al, 1985; Banerjee et al, 1997).

Reproductive

    3.20.1) SUMMARY
    A) There are no reported cases of human teratogenicity due to cyanide itself; however, teratogenic effects have been observed in experimental animals exposed to cyanide and related compounds.
    3.20.2) TERATOGENICITY
    A) LACK OF INFORMATION
    1) At the time of this review, there were no reported cases of human teratogenicity due to cyanide itself.
    B) ANIMAL STUDIES
    1) CONGENITAL ANOMALY
    a) RELATED AGENTS
    1) POTASSIUM CYANIDE - Fetotoxicity has occurred in the offspring of rats and domestic animals administered potassium cyanide (RTECS, 1990). However, some rat studies have shown no teratogenicity (Schardein, 1985).
    2) SODIUM CYANIDE - Sodium cyanide solution delivered by constant infusion to pregnant Golden Hamsters at doses from 0.126 to 0.1295 mmol/kg/hour between days 6 and 9 of gestation caused a high incidence of both resorptions and malformations in the offspring (Doherty et al, 1982).
    a) Neural tube defects (exencephaly, encephalocele) were the most common malformations. Hydropericardium and crooked tails were also found.
    b) Concomitant infusion of sodium thiosulfate prevented both maternal signs of toxicity and the teratogenic effects of the sodium cyanide infusion.
    3) ACETONITRILE - Pregnant hamsters exposed by either inhalation of 5,000 to 8,000 ppm or given 100 to 400 mg/kg oral or intraperitoneal doses of acetonitrile delivered offspring with severe axial skeletal disorders (Willhite, 1983).
    a) Injections of sodium thiosulfate antagonized the teratogenic effects (Willhite, 1983).
    b) Elevated cyanide and thiocyanate levels were found in all tissues studied 2.5 hours after oral or intraperitoneal dosing, and suggested that the in vivo liberation of cyanide from acetonitrile was responsible for the observed teratogenic effects (Willhite, 1983).
    4) ACRYLONITRILE AND PROPIONITRILE - Intraperitoneal injections of acrylonitrile or propionitrile to hamsters on day 8 of gestation resulted in exencephaly, encephaloceles, and rib abnormalities in the offspring (Willhite et al, 1981).
    a) Sodium thiosulfate injections protected the fetus from these effects, except at larger nitrile doses where thiosulfate protected the dam against overt poisoning but did not protect the fetus against malformations (Willhite et al, 1981).
    b) The teratogenic effects of both nitriles may be related to the metabolic release of cyanide after absorption (Willhite et al, 1981).
    c) In a separate experiment, propionitrile given by gavage to rats produced no teratogenic effects (Johannsen et al, 1986).
    5) CYANOGENIC GLYCOSIDE - Rats fed cassava powder (containing high concentrations of a cyanogenic glycoside) as 50 to 80% of their diet during the first 5 days of pregnancy showed a low incidence of limb defects, open eye defects, microcephaly, and fetal growth retardation in fetuses collected on day 20 of pregnancy (Singh, 1981).
    6) LAETRILE - Laetrile given orally to pregnant hamsters produced skeletal malformations in the offspring and increased levels of tissue cyanide (Willhite, 1982).
    a) Intravenous administration of laetrile produced neither effect (Willhite, 1982).
    b) Sodium thiosulfate administration protected the fetus from teratogenic effects (Willhite, 1982). These data suggested that the teratogenic effects were due to cyanide released in vivo from oral laetrile dosing (Willhite, 1982).

Carcinogenicity

    3.21.1) IARC CATEGORY
    A) IARC Carcinogenicity Ratings for CAS57-12-5 (International Agency for Research on Cancer (IARC), 2016; International Agency for Research on Cancer, 2015; IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2010; IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2010a; IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2008; IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2007; IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2006; IARC, 2004):
    1) Not Listed
    3.21.3) HUMAN STUDIES
    A) PULMONARY CARCINOMA
    1) Acrylonitrile has carcinogenic properties in some species of experimental animals and has been suggested to be associated with a slight increase in deaths from lung cancer and other malignancies in humans (Buchter & Peter, 1984; Geiger et al, 1983).
    a) Whether the metabolic release of cyanide after absorption plays any role in carcinogenesis is unknown.
    b) In isolated cell preparations, the release of cyanide does not appear to play a role in cell death (Geiger et al, 1983).
    B) LACK OF EFFECT
    1) There are no reports of carcinogenicity in humans or experimental animals due to cyanide itself.

Monitoring Parameters Levels

    4.1.1) SUMMARY
    A) Monitor serum chemistry and lactate concentrations.
    B) Monitor arterial and venous blood gases.
    C) Cyanide levels can be measured to confirm the diagnosis, but are usually not available in a timely manner to be clinically useful.
    D) Institute continuous cardiac monitoring and obtain an ECG.
    E) Methemoglobin should also be monitored frequently in patients receiving intravenous sodium nitrite.
    F) Obtain a carboxyhemoglobin level in the setting of a fire.
    G) Consider a head CT for comatose patients.
    4.1.2) SERUM/BLOOD
    A) ACID/BASE
    1) Obtain blood for arterial blood gases, venous pO2 or measured venous %O2 saturation.
    B) BLOOD/SERUM CHEMISTRY
    1) Obtain blood for electrolytes, serum lactate, and whole blood cyanide levels.
    2) Blood cyanide levels and associated symptoms (Graham et al, 1977):
    a) Blood cyanide levels and associated symptoms (Graham et al, 1977):
    1) No symptoms: Less than 0.2 mg/L (mcg/mL) (0.02 mg%; SI = 7.7 mcmol/L)
    2) Flushing and tachycardia: 0.5-1.0 mg/L (mcg/mL) (0.05-0.1 mg%; SI = 19.2 to 38.5 mcmol/L)
    3) Obtundation: 1.0-2.5 mg/L (mcg/mL) (0.1-0.25 mg%; SI = 38.5 to 96.1 mcmol/L)
    4) Coma and respiratory depression: Greater than 2.5 mg/L (mcg/mL) (0.25 mg%; SI = 96.1 mcmol/L)
    5) Death: Greater than 3 mg/L (mcg/mL) (0.3 mg%; SI = 115.4 mcmol/L)
    C) LABORATORY INTERFERENCE
    1) Based on in vivo studies, hydroxocobalamin (an approved cyanide antidote), due to its red color, may interfere with the following spectrophotometric tests: serum AST, creatinine, bilirubin, magnesium and serum iron (Beckerman et al, 2009; Curry et al, 1992).
    2) In vitro studies, involving hydroxocobalamin samples, have reported artificially increased laboratory parameters of serum creatinine, bilirubin, triglycerides, cholesterol, total protein, glucose, albumin, and alkaline phosphatase concentrations. These studies have also reported artificially decreased parameters of serum ALT and amylase concentrations (Beckerman et al, 2009).
    3) CARBOXYHEMOGLOBIN: In vitro and in vivo studies, involving rabbits and using a co-oximeter as a measurement tool, have demonstrated hydroxocobalamin interference with carboxyhemoglobin measurements, with a dose-dependent 14.7% increase in carboxyhemoglobin concentration (Beckerman et al, 2009; Lee et al, 2007). It has been suggested that , this interference is not clinically significant, particularly in emergent situations involving suspected cases of cyanide poisoning (Borron et al, 2007). However, it is recommended that a carboxyhemoglobin measurement be obtained prior to hydroxocobalamin administration in order to avoid any possible interference .
    D) OTHER
    1) LABORATORY VALUES: Arterial blood gases and serum electrolytes are useful in the assessment of potential elevated anion gap metabolic acidosis in patients poisoned with cyanide (Brennan, 1988; Hall & Rumack, 1986; Vogel et al, 1981).
    2) A REDUCED ARTERIO-CENTRAL VENOUS MEASURED %O2 SATURATION DIFFERENCE may be seen due to cellular inability to extract oxygen (Graham et al, 1977; Paulet, 1955). Arterialization of venous blood gases (elevated venous pO2 or measured venous %O2 saturation) may serve as an early clue in the diagnosis of cyanide poisoning (Hall & Rumack, 1986; Johnson & Mellors, 1988).
    3) Serum lactate levels may be useful in monitoring the severity of poisoning and the efficacy of treatment (Vogel et al, 1981; Baud et al, 1994), as lactate is a marker of cellular hypoxia.
    4) Monitor methemoglobin levels during nitrite administration, especially if more than one dose is needed. Maintain methemoglobin levels below 30% (Hall & Rumack, 1986).
    a) It may be feasible to use a modified commercial six-wavelength hemoglobin photometer (Radiometer OSM3) for easy and rapid analysis of methemoglobin and methemoglobin cyanide in small samples of blood (Zijlstra & Buursma, 1993).
    5) CYANIDE LEVELS AFTER HYDROXOCOBALAMIN: A 51-year-old man received a hydroxocobalamin infusion as an antidote following an intentional ingestion of an unknown amount of potassium cyanide solution (3 g/L) and, one hour after the hydroxocobalamin infusion, the patient's cyanide plasma level was 42 mcg/dL. His plasma cyanide level before the hydroxocobalamin infusion was 29 mcg/dL. It is believed that hydroxocobalamin was extracting cyanide from the red blood cells (which typically contain a higher concentration of cyanide than in plasma) to form cyanocobalamin (vitamin B12 ) in plasma, which was subsequently detected as cyanide (Weng et al, 2004).
    6) BREATH HYDROGEN CYANIDE LEVEL: A recent study suggests that the use of breath hydrogen cyanide level may be a potential indicator of systemic intoxication. However, a commercial test is not available at this time (Stamyr et al, 2008).
    4.1.3) URINE
    A) URINARY LEVELS
    1) Cyanide and thiocyanate levels can also be measured in timed urine collections which may yield useful information on cyanide clearance. However, such testing is seldom done clinically; it is more a research tool.
    2) URINARY CYANIDE EXCRETION: 24-hour urinary excretion has been measured after chronic exposure to cyanide fumes and cyanide aerosols (Chandra et al, 1980):
    WORKERS (n=23)Cyanide mcg/100 mL
    RANGEMEAN
    Smokers (n=8)0 to 186.23
    Nonsmokers (n=15)0 to 14.675.4
    Controls (n=20) .
    Smokers (n=10)0 to 8.453.2
    Nonsmokers (n=10)0 to 4.362.15

Radiographic Studies

    A) CHEST RADIOGRAPH
    1) If respiratory tract irritation or pulmonary symptoms are present, a chest x-ray is recommended.
    B) MRI
    1) MRI studies may be useful in identifying the location and extent of injury in patients with cyanide-induced parkinsonian syndrome (Rosenberg et al, 1989; Carella et al, 1988; Feldman & Feldman, 1990; Grandas et al, 1989).

Methods

    A) MULTIPLE ANALYTICAL METHODS
    1) Cyanide can be measured chemically by several methods. Blood cyanide levels may be useful in confirming the diagnosis. However, it is not clinically useful unless the results are available within a reasonable time. Treatment should be initiated base on clinical judgement.
    2) SERUM CYANIDE LEVELS: Can be measured quickly using a Flame Thermionic Detector Gas-Chromatograph (Yoshida et al, 1989).
    3) SERUM CYANIDE LEVELS/COLORIMETRIC ASSAY: A rapid, methemoglobin-based assay has been developed that uses the color change associated with the cyanomethemoglobin complex to measure cyanide in blood with relatively good accuracy and sensitivity. The assay was validated against a polarographic method with correlation between methods of 0.983 (Tung et al, 1997).
    4) BIOLOGICAL SPECIMENS/SPECTROSCOPY: Cyanide can be liberated from biological specimens by acidification, followed by absorption in alkali and interaction with chromophoric reagents for quantification by absorbance spectroscopy (HSDB, 1990).
    5) BIOLOGICAL SPECIMENS/HEADSPACE GAS CHROMATOGRAPHY: Automated headspace gas chromatography has been described by Calafat and Stanfill (2002) as a rapid method for quantitatively measuring cyanide concentrations in whole blood from blood samples contaminated with potassium cyanide and from blood samples of victims with smoke inhalation injuries. This method was able to detect a wide range of concentrations (30 to 6000 mcg/L) in approximately 17 minutes, which included time of incubation, gas chromatography run time, and manual sample preparation. Limit of detection was calculated to be 13.8 mcg/L (Calafat & Stanfill, 2002).
    6) BIOLOGICAL SPECIMENS/GAS CHROMATOGRAPHY: Cyanide can also be measured in biological fluids by gas chromatography following conversion to cyanogen chloride by reaction with chloramine-T (HSDB, 1990). Zamecnik & Tam (1987) describe another gas chromatographic process which measures cyanide in blood with acetonitrile as an internal standard (Zamecnik & Tam, 1987).
    7) BIOLOGICAL SPECIMENS/GAS-LIQUID CHROMATOGRAPHY: Determining low-level hydrocyanic acid in solution using gas-liquid chromatography is reported by Schneider & Freund (1972) (Schneider & Freund, 1972).
    8) BIOLOGICAL SPECIMENS/PAPER CHROMATOGRAPHY: A qualitative method for cyanide detection using tartaric acid and paper chromatography and a quantitative method for detection in blood involving absorption by sulfuric acid have been described (Free & Free, 1970).
    9) BIOLOGICAL SPECIMENS/ION SPECIFIC ELECTRODE: An ion-specific electrode method has sometimes been used for measuring cyanide in biological specimens (Bismuth et al, 1984).
    10) BIOLOGICAL SPECIMENS/FLUORIMETRIC METHOD: A fluorometric diffusion method based on detection of fluorescing p-benzoquinone derivatives can be used to determine cyanide in biological fluids (HSDB, 1990).
    11) BIOLOGICAL SPECIMENS/MICRODISTILLATION: An automated microdistillation assay technique has been developed that can provide whole blood and plasma cyanide levels in less than one-half hour (Groff et al, 1985) but is not yet generally available.
    12) BIOLOGICAL SPECIMENS/PAPER STRIP: A semiquantitative paper strip method (CYANTESMO, available from Gallard-Schlesinger, Carle Place, NY) is sensitive to concentrations of 0.2 mg/L of cyanide in whole blood. A 5-hour incubation time is required to rule out false negative results, but concentrations of greater than 1 mg/L require only 30 to 60 minutes to be detected (Fligner et al, 1992).
    13) GASTRIC ASPIRATE EXAMINATION: Cyanide presence in gastric aspirate can be detected by adding a few crystals of FeSo4 to 5 to 10 mL of the aspirate (Graham et al, 1977).
    a) Add 4 to 5 drops of 20% NaOH, then boil and cool the solution (Graham et al, 1977).
    b) Adding 8 to 10 drops of 10% HCl will result in a greenish-blue precipitate if cyanide is present (Graham et al, 1977).
    c) Salicylates may interfere with this test resulting in an initial blue-green color that converts to a purple color (Graham et al, 1977).
    B) OTHER
    1) STABILITY DURING STORAGE: Up to 50% of cyanide was lost within 12 minutes after spiking blood in an unstoppered container. Loss of cyanide from forensic specimens may be minimized, but not eliminated by rapid analysis after death, collection of blood from a closed source (subclavian vein), storage in a fluoride-oxalate tube with minimal dead space and sealed immediately, and storage frozen at minus 20 degrees C (Bright et al, 1990).
    2) ENZYME-BASED DETECTION: Cyanide detection using a tyrosinase-coupled oxygen electrode has been studied. This system imitates cyanide's site of toxicity in the mitochondria. A terminal sequence of electron transfer in aerobic respiration is mimicked by a mediator coupling of tyrosinase catalysis to an electro-chemical system (Smit & Rechnitz, 1993).

Life Support

    A) Support respiratory and cardiovascular function.

Patient Disposition

    6.3.1) DISPOSITION/ORAL EXPOSURE
    6.3.1.1) ADMISSION CRITERIA/ORAL
    A) All symptomatic patients should be admitted to the hospital following a cyanide exposure. Whenever the cyanide antidote kit is used, the patient should be admitted to the intensive care unit.
    6.3.1.2) HOME CRITERIA/ORAL
    A) There is no role for home management of cyanide exposure.
    6.3.1.3) CONSULT CRITERIA/ORAL
    A) Consult a poison center or medical toxicologist for assistance in managing symptomatic patients.
    6.3.1.5) OBSERVATION CRITERIA/ORAL
    A) Asymptomatic patients with a history of significant cyanide exposure but who are asymptomatic should be observed closely in the hospital. Vascular access should be established, laboratory evaluations performed, and the cyanide antidote kit or hydroxocobalamin ready at the bedside. If laboratory evaluations are normal and the patient remains asymptomatic for at least 8 hours, they may be discharged from the hospital with appropriate follow-up instructions.

Monitoring

    A) Monitor serum chemistry and lactate concentrations.
    B) Monitor arterial and venous blood gases.
    C) Cyanide levels can be measured to confirm the diagnosis, but are usually not available in a timely manner to be clinically useful.
    D) Institute continuous cardiac monitoring and obtain an ECG.
    E) Methemoglobin should also be monitored frequently in patients receiving intravenous sodium nitrite.
    F) Obtain a carboxyhemoglobin level in the setting of a fire.
    G) Consider a head CT for comatose patients.

Oral Exposure

    6.5.1) PREVENTION OF ABSORPTION/PREHOSPITAL
    A) SUMMARY
    1) In symptomatic patients, institute emergency measures including basic life support and administration of cyanide antidote kit (if readily available) before performing the following steps.
    B) ACTIVATED CHARCOAL
    1) Absorption of cyanide is rapid and charcoal may only be beneficial if administered immediately after ingestion.
    2) Immediate administration of a large dose of superactivated charcoal (4 grams/kilogram) to rats given an oral lethal dose of potassium cyanide (35 to 40 milligrams/kilogram) prevented lethality. Eight of 26 treated animals died compared to 25 of 26 untreated animals.
    3) 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).
    4) 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) In symptomatic patients, institute emergency measures basic life support and administration of cyanide antidote kit (if readily available) before performing the following steps.
    B) ACTIVATED CHARCOAL
    1) The usefulness of activated charcoal may be questionable.
    a) 1 gram of activated charcoal may adsorb 35 milligrams of potassium cyanide (Anderson, 1946) but this is a low percentage.
    b) The absorption of cyanide is so rapid that charcoal may be of little use unless administered immediately after ingestion of cyanide.
    c) Immediate administration of a large dose of superactivated charcoal (4 grams/kilogram) to rats given an oral lethal dose of potassium cyanide (35 to 40 milligrams/kilogram) prevented lethality. Eight of 26 treated animals died compared to 25 of 26 untreated animals (Lambert et al, 1988).
    2) 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.
    3) 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).
    C) GASTRIC LAVAGE
    1) INDICATIONS: Consider gastric lavage with a large-bore orogastric tube (ADULT: 36 to 40 French or 30 English gauge tube {external diameter 12 to 13.3 mm}; CHILD: 24 to 28 French {diameter 7.8 to 9.3 mm}) after a potentially life threatening ingestion if it can be performed soon after ingestion (generally within 60 minutes).
    a) Consider lavage more than 60 minutes after ingestion of sustained-release formulations and substances known to form bezoars or concretions.
    2) PRECAUTIONS:
    a) SEIZURE CONTROL: Is mandatory prior to gastric lavage.
    b) AIRWAY PROTECTION: Place patients in the head down left lateral decubitus position, with suction available. Patients with depressed mental status should be intubated with a cuffed endotracheal tube prior to lavage.
    3) LAVAGE FLUID:
    a) Use small aliquots of liquid. Lavage with 200 to 300 milliliters warm tap water (preferably 38 degrees Celsius) or saline per wash (in older children or adults) and 10 milliliters/kilogram body weight of normal saline in young children(Vale et al, 2004) and repeat until lavage return is clear.
    b) The volume of lavage return should approximate amount of fluid given to avoid fluid-electrolyte imbalance.
    c) CAUTION: Water should be avoided in young children because of the risk of electrolyte imbalance and water intoxication. Warm fluids avoid the risk of hypothermia in very young children and the elderly.
    4) COMPLICATIONS:
    a) Complications of gastric lavage have included: aspiration pneumonia, hypoxia, hypercapnia, mechanical injury to the throat, esophagus, or stomach, fluid and electrolyte imbalance (Vale, 1997). Combative patients may be at greater risk for complications (Caravati et al, 2001).
    b) Gastric lavage can cause significant morbidity; it should NOT be performed routinely in all poisoned patients (Vale, 1997).
    5) CONTRAINDICATIONS:
    a) Loss of airway protective reflexes or decreased level of consciousness if patient is not intubated, following ingestion of corrosive substances, hydrocarbons (high aspiration potential), patients at risk of hemorrhage or gastrointestinal perforation, or trivial or non-toxic ingestion.
    6.5.3) TREATMENT
    A) OXYGEN
    1) Administer 100% humidified supplemental with assisted ventilation as required.
    a) Oxygen may reverse the cyanide-cytochrome oxidase complex and facilitate the conversion to thiocyanate following thiosulfate administration (Graham et al, 1977).
    b) Animal data suggests that supplemental oxygen as adjunct treatment of cyanide poisoning increases the overall antidotal efficacy of sodium nitrite and sodium thiosulfate (Burrows & Way, 1977) Way et al, 1966; (Sheehy & Way, 1968).
    B) FLUID/ELECTROLYTE BALANCE REGULATION
    1) Establish secure intravenous access; consider obtaining at least two intravenous lines. Administer crystalloids and vasopressors for hypotension. Administer sodium bicarbonate according to arterial blood gases and serum bicarbonate.
    C) CYANIDE ANTIDOTE
    1) A cyanide antidote, either hydroxocobalamin or the sodium nitrite/sodium thiosulfate kit, should be administered to patients with symptomatic poisoning. If cyanide toxicity develops concurrent with carbon monoxide poisoning (e.g., closed space fire), hydroxocobalamin is the preferred antidote. It that is not available, sodium thiosulfate may be used alone. Use of amyl nitrite or sodium nitrite will cause methemoglobinemia, further reducing oxygen carrying capacity.
    2) HYDROXOCOBALAMIN-CYANOKIT(R)
    a) ADULT DOSE: 5 g (two 2.5 g vials each reconstituted with 100 mL sterile 0.9% saline) administered as an intravenous infusion over 15 minutes. For severe poisoning, a second dose of 5 g may be infused intravenously over 15 minutes to 2 hours, depending on the patient's condition (Prod Info CYANOKIT(R) 2.5g IV injection, 2006).
    b) PEDIATRIC DOSE: A dose of 70 mg/kg has been used in pediatric patients, based on limited post-marketing experience outside the US (Prod Info CYANOKIT(R) 2.5g IV injection, 2006).
    c) INDICATIONS: Known or suspected cyanide poisoning.
    d) ADVERSE EFFECTS: Transient hypertension, allergic reactions (including anaphylaxis), nausea, headache, rash. Hydroxocobalamin's deep red color causes red-colored urine in all patients, and erythema of the skin in most (Prod Info CYANOKIT(R) 2.5g IV injection, 2006).
    e) LABORATORY INTERFERENCE: Because of its' color, hydroxocobalamin interferes with colorimetric determination of various laboratory parameters. It may artificially increase serum creatinine, bilirubin, triglycerides, cholesterol, total protein, glucose, albumin , alkaline phosphatase and hemoglobin. It may artificially decrease serum ALT and amylase. It may artificially increase urinary pH, glucose, protein, erythrocytes, leukocytes, ketones, bilirubin, urobilinogen, and nitrate (Prod Info CYANOKIT(R) 2.5g IV injection, 2006).
    f) HEMODIALYSIS INTERFERENCE: Dialysis machines have a spectrophotometric safety measure that can shut down after detecting blood leaking across the dialysis membrane. Hydroxocobalamin has a deep red color and can permeate the dialysis membrane, coloring the dialysate and causing the hemodialysis machine to shut down erroneously. In one case report, a patient with cyanide poisoning underwent dialysis after receiving 5 g of IV hydroxocobalamin because of refractory acidemia, reduced kidney function and hyperkalemia. A blood leak alarm caused the dialysis machine to shut down erroneously, delaying therapy, and resulting in the death of the patient (Stellpflug et al, 2013).
    g) Hydroxocobalamin, 5 to 20 grams administered intravenously, has been shown to be effective, as sole antidotal therapy, for acute cyanide poisoning following cyanide salt ingestion or inhalation (Borron et al, 2007a)
    h) In a controlled pilot study of male smokers, administration of 5 grams of intravenous 5% hydroxocobalamin significantly decreased whole blood cyanide levels (Forsyth et al, 1992).
    i) Hydroxocobalamin has been shown to be effective in treating cyanide poisoned animals and has the advantage of neither producing methemoglobinemia nor hypotension as does sodium nitrite (Forsyth et al, 1993).
    j) According to antidote stocking guidelines, hydroxocobalamin was preferred over the conventional cyanide antidote kit due to its wider indications, ease of use, and anticipated safety in widespread use. It can safely be used in patients with smoke inhalation (None Listed, 2008).
    k) In cases of cyanide ingestion, both the nitrite/thiosulfate combination and hydroxocobalamin seem to be effective antidotes. Hydroxocobalamin offers an improved safety profile for children and pregnant women, as well as patients suffering from cyanide poisoning in conjunction with smoke inhalation (Shepherd & Velez, 2008). Patients who are exposed to cyanide in house fires may have high carbon monoxide levels. Inducing methemoglobinemia with nitrites may further impair oxygen carrying capacity.
    l) Some authors advocate combined hydroxocobalamin-thiosulfate therapy because of possible synergy of complementary mechanisms (Kerns et al, 2008).
    1) CASE REPORT: A 58-year-old man intentionally ingested 1200 to 1500 mg of pure potassium cyanide powder and subsequently lost consciousness for 1 to 1.5 minutes and had one episode of generalized seizures. At presentation to the hospital, the patient was severely somnolent with mydriasis and lactic acidosis. The patient required emergent intubation, alalgosedation, and ventilation. Cyanide antidotal treatment was initiated 1.5 hours post-ingestion with hydroxocobalamin (2 subsequent IV infusions over 25 minutes for a total dose of 7.5 g) followed by a continuous IV infusion of 10% sodium thiosulfate at a dose of 1 mL/hour/kg (total dose 12 g). Following antidotal therapy, the patient's arterial blood gases normalized on the day of admission. On hospital day 3, he was extubated, and the next day was discharged without visual or neurologic sequelae, which continued at his 5-month follow-up appointment (Zakharov et al, 2015).
    m) CASE REPORT: A 24-year-old man developed metabolic acidosis with an elevated serum lactate level (29 mmol/L), and atrial fibrillation with a widened QRS complex (116 ms), a prolonged QTc interval (572 msec), and a heart rate of 116 beats/min after a reported intentional ingestion of potassium cyanide (blood cyanide concentration at ED presentation was 4.02 mcg/mL). Treatment included administration of IV fluids, IV sodium bicarbonate and an IV infusion of hydroxocobalamin 5 g. At 35 minutes, 2 hours, and 3 hours post-hydroxocobalamin administration, the patient's serum lactate level gradually decreased to 19, 11.4, and 6.5 mmol/L, respectively. Within 7 hours of presentation, the patient's serum lactate level normalized (1.4 mmol/L), his acidosis resolved, and a repeat ECG revealed a return to normal sinus rhythm with a heart rate of 71 beats/min, and a QRS and QTc of 92 msec and 406 msec, respectively. Within 48 hours of presentation, the patient was discharged to an inpatient psychiatric unit (Johnson et al, 2015).
    3) CYANIDE ANTIDOTE KIT
    a) OBTAIN AND PREPARE for administration a CYANIDE ANTIDOTE KIT, consisting of sodium nitrite and sodium thiosulfate.
    b) Antidotes should be administered in patients who are clinically symptomatic (i.e., unstable vital signs, acidosis, impaired consciousness, seizures, or coma).
    c) SODIUM NITRITE
    1) INDICATION
    a) Sodium nitrite should be given initially and administered as soon as vascular access is established.
    b) Further administration of sodium nitrite is dictated only by the clinical situation, provided no significant complications (hypotension, excessive methemoglobinemia) are present. Use with caution if carbon monoxide poisoning is also suspected.
    c) The goal of nitrite therapy is to achieve a methemoglobin level of 20% to 30%. This level is not based on clinical data, but represents the tolerated concentration without significant adverse symptoms from methemoglobin in an otherwise healthy individual. Clinical response has been reported to occur with methemoglobin levels in the range of 3.6% to 9.2% (DiNapoli et al, 1989; Johnson et al, 1989; Johnson & Mellors, 1988).
    2) ADULT DOSE
    a) 10 mL of a 3% solution (300 mg) administered intravenously at a rate of 2.5 to 5 mL/minute (Prod Info NITHIODOTE intravenous injection solution, 2011). Frequent blood pressure monitoring must accompany sodium nitrite injection and the rate slowed if hypotension occurs.
    b) If there is inadequate clinical response, an additional dose of sodium nitrite at half the amount of the initial dose may be administered 30 minutes following the first dose (Prod Info NITHIODOTE intravenous injection solution, 2011).
    3) PEDIATRIC DOSE
    a) The recommended pediatric sodium nitrite dose is 0.2 mL/kg of a 3% solution (6 mg/kg) administered intravenously at a rate of 2.5 to 5 mL/minute, not to exceed 10 mL (300 mg) (Prod Info NITHIODOTE intravenous injection solution, 2011).
    b) If there is inadequate clinical response, an additional dose of sodium nitrite at half the amount of the initial dose may be administered 30 minutes following the first dose (Prod Info NITHIODOTE intravenous injection solution, 2011; Berlin, 1970a).
    c) PRESENCE OF ANEMIA: If there is a reason to suspect the presence of anemia, the following initial sodium nitrite doses should be given, depending on the child's hemoglobin (sodium nitrite should not exceed the doses listed below; fatal methemoglobinemia may result) (Berlin, 1970a):
    1) Hemoglobin: 8 g/dL - Initial 3% sodium nitrite dose: 0.22 mL/kg (6.6 mg/kg)
    2) Hemoglobin: 10 g/dL - Initial 3% sodium nitrite dose: 0.27 mL/kg (8.7 mg/kg)
    3) Hemoglobin: 12 g/dL (average child) - Initial 3% sodium nitrite dose: 0.33 mL/kg (10 mg/kg)
    4) Hemoglobin: 14 g/dL - Initial 3% sodium nitrite dose: 0.39 mL/kg (11.6 mg/kg)
    4) It is highly recommended that total hemoglobin and methemoglobin concentrations be rapidly measured (30 minutes after dose), when possible, before repeating a dose of sodium nitrite to be sure that dangerous methemoglobinemia will not occur, especially in the pediatric patient.
    5) Monitor blood pressure frequently and treat hypotension by slowing infusion rate and giving crystalloids and vasopressors. Consider possible excessive methemoglobin formation if patient deteriorates during therapy.
    6) Excessive methemoglobinemia and hypotension are potential complications of nitrite therapy.
    7) In individuals with G6PD deficiency, therapy with methemoglobin-inducing agents is contraindicated because of the likelihood of serious hemolysis.
    d) SODIUM THIOSULFATE
    1) Sodium thiosulfate is the second component of the cyanide antidote kit. It is supplied as 50 mL of a 25% solution and it is administered intravenously. There are no adverse reactions to thiosulfate itself. The pediatric dose is adjusted for weight and not hemoglobin concentration.
    2) Sodium thiosulfate supplies sulfur for the rhodanese reaction, and is recommended after sodium nitrite, hydroxocobalamin, or 4-DMAP (4-dimethylaminophenol) administration (Marrs, 1988; Hall & Rumack, 1987).
    3) DOSE
    a) Follow sodium nitrite with IV sodium thiosulfate. ADULT: Administer 50 mL (12.5 g) of a 25% solution IV; PEDIATRIC: 1 mL/kg of a 25% solution (250 mg/kg), not to exceed 50 mL (12.5 g) total dose (Prod Info NITHIODOTE intravenous injection solution, 2011).
    b) A second dose, one-half of the first dose, may be administered if signs of cyanide toxicity reappear (Prod Info NITHIODOTE intravenous injection solution, 2011).
    c) Sodium thiosulfate is usually used in combination with sodium nitrite but may be used alone (Prod Info sodium thiosulfate IV injection, 2003).
    d) Sodium thiosulfate can be administered without sodium nitrite in patients at risk to develop further methemoglobinemia (ie excessive methemoglobinemia or hypotension after initial sodium nitrite administration or in the presence of methemoglobinemia or carboxyhemoglobin in patients with smoke inhalation due to fire). Sodium thiosulfate can also be used in combination with hydroxocobalamin to treat cyanide poisoning (Howland, 2011)
    e) CONTINUOUS INFUSION: It has been suggested that a continuous infusion of sodium thiosulfate be given after the initial bolus to maintain high thiosulfate levels. Low sodium intravenous fluids are required to avoid sodium overload. If large amounts of sodium thiosulfate are required, hemodialysis may be necessary to maintain a physiologic serum sodium level (Turchen et al, 1991).
    f) ADVERSE EVENTS: Sodium thiosulfate does not usually produce significant toxicity. Possible adverse events include hypotension, headache, nausea, vomiting, disorientation, and prolonged bleeding time (Prod Info NITHIODOTE intravenous injection solution, 2011).
    D) ACIDOSIS
    1) METABOLIC ACIDOSIS: Treat severe metabolic acidosis (pH less than 7.1) with sodium bicarbonate, 1 to 2 mEq/kg is a reasonable starting dose(Kraut & Madias, 2010). Monitor serum electrolytes and arterial or venous blood gases to guide further therapy.
    2) Acidosis may be difficult to correct prior to administration of antidotes in serious cyanide poisoning cases (Hall & Rumack, 1986).
    E) SEIZURE
    1) SUMMARY
    a) Attempt initial control with a benzodiazepine (eg, diazepam, lorazepam). If seizures persist or recur, administer phenobarbital or propofol.
    b) Monitor for respiratory depression, hypotension, and dysrhythmias. Endotracheal intubation should be performed in patients with persistent seizures.
    c) Evaluate for hypoxia, electrolyte disturbances, and hypoglycemia (or, if immediate bedside glucose testing is not available, treat with intravenous dextrose).
    2) DIAZEPAM
    a) ADULT DOSE: Initially 5 to 10 mg IV, OR 0.15 mg/kg IV up to 10 mg per dose up to a rate of 5 mg/minute; may be repeated every 5 to 20 minutes as needed (Brophy et al, 2012; Prod Info diazepam IM, IV injection, 2008; Manno, 2003).
    b) PEDIATRIC DOSE: 0.1 to 0.5 mg/kg IV over 2 to 5 minutes; up to a maximum of 10 mg/dose. May repeat dose every 5 to 10 minutes as needed (Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008).
    c) Monitor for hypotension, respiratory depression, and the need for endotracheal intubation. Consider a second agent if seizures persist or recur after repeated doses of diazepam .
    3) NO INTRAVENOUS ACCESS
    a) DIAZEPAM may be given rectally or intramuscularly (Manno, 2003). RECTAL DOSE: CHILD: Greater than 12 years: 0.2 mg/kg; 6 to 11 years: 0.3 mg/kg; 2 to 5 years: 0.5 mg/kg (Brophy et al, 2012).
    b) MIDAZOLAM has been used intramuscularly and intranasally, particularly in children when intravenous access has not been established. ADULT DOSE: 0.2 mg/kg IM, up to a maximum dose of 10 mg (Brophy et al, 2012). PEDIATRIC DOSE: INTRAMUSCULAR: 0.2 mg/kg IM, up to a maximum dose of 7 mg (Chamberlain et al, 1997) OR 10 mg IM (weight greater than 40 kg); 5 mg IM (weight 13 to 40 kg); INTRANASAL: 0.2 to 0.5 mg/kg up to a maximum of 10 mg/dose (Loddenkemper & Goodkin, 2011; Brophy et al, 2012). BUCCAL midazolam, 10 mg, has been used in adolescents and older children (5-years-old or more) to control seizures when intravenous access was not established (Scott et al, 1999).
    4) LORAZEPAM
    a) MAXIMUM RATE: The rate of intravenous administration of lorazepam should not exceed 2 mg/min (Brophy et al, 2012; Prod Info lorazepam IM, IV injection, 2008).
    b) ADULT DOSE: 2 to 4 mg IV initially; repeat every 5 to 10 minutes as needed, if seizures persist (Manno, 2003; Brophy et al, 2012).
    c) PEDIATRIC DOSE: 0.05 to 0.1 mg/kg IV over 2 to 5 minutes, up to a maximum of 4 mg/dose; may repeat in 5 to 15 minutes as needed, if seizures continue (Brophy et al, 2012; Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008; Sreenath et al, 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).
    7) PHENYTOIN/FOSPHENYTOIN
    a) Benzodiazepines and/or barbiturates are preferred to phenytoin or fosphenytoin in the treatment of drug or withdrawal induced seizures (Wallace, 2005).
    b) PHENYTOIN
    1) PHENYTOIN INTRAVENOUS PUSH VERSUS INTRAVENOUS INFUSION
    a) Administer phenytoin undiluted, by very slow intravenous push or dilute 50 mg/mL solution in 50 to 100 mL of 0.9% saline.
    b) ADULT DOSE: A loading dose of 20 mg/kg IV; may administer an additional 5 to 10 mg/kg dose 10 minutes after loading dose. Rate of administration should not exceed 50 mg/minute (Brophy et al, 2012).
    c) PEDIATRIC DOSE: A loading dose of 20 mg/kg, at a rate not exceeding 1 to 3 mg/kg/min or 50 mg/min, whichever is slower (Loddenkemper & Goodkin, 2011; Prod Info Dilantin(R) intravenous injection, intramuscular injection, 2013).
    d) CAUTIONS: Administer phenytoin while monitoring ECG. Stop or slow infusion if dysrhythmias or hypotension occur. Be careful not to extravasate. Follow each injection with injection of sterile saline through the same needle (Prod Info Dilantin(R) intravenous injection, intramuscular injection, 2013).
    e) SERUM CONCENTRATION MONITORING: Monitor serum concentrations over next 12 to 24 hours for maintenance of therapeutic concentrations. Therapeutic concentrations of 10 to 20 mcg/mL have been reported (Prod Info Dilantin(R) intravenous injection, intramuscular injection, 2013).
    c) FOSPHENYTOIN
    1) ADULT DOSE: A loading dose of 20 mg phenytoin equivalent/kg IV, at a rate not exceeding 150 mg phenytoin equivalent/minute; may give additional dose of 5 mg/kg 10 minutes after the loading infusion (Brophy et al, 2012).
    2) CHILD DOSE: 20 mg phenytoin equivalent/kg IV, at a rate of 3 mg phenytoin equivalent/kg/minute, up to a maximum of 150 mg phenytoin equivalent/minute (Loddenkemper & Goodkin, 2011).
    3) CAUTIONS: Perform continuous monitoring of ECG, respiratory function, and blood pressure throughout the period where maximal serum phenytoin concentrations occur (about 10 to 20 minutes after the end of fosphenytoin infusion) (Prod Info CEREBYX(R) intravenous injection, 2014).
    4) SERUM CONCENTRATION MONITORING: Monitor serum phenytoin concentrations over the next 12 to 24 hours; therapeutic levels 10 to 20 mcg/mL. Do not obtain serum phenytoin concentrations until at least 2 hours after infusion is complete to allow for conversion of fosphenytoin to phenytoin (Prod Info CEREBYX(R) intravenous injection, 2014).
    8) RECURRING SEIZURES
    a) If seizures are not controlled by the above measures, patients will require endotracheal intubation, mechanical ventilation, continuous EEG monitoring, a continuous infusion of an anticonvulsant, and may require neuromuscular paralysis and vasopressor support. Consider continuous infusions of the following agents:
    1) MIDAZOLAM: ADULT DOSE: An initial dose of 0.2 mg/kg slow bolus, at an infusion rate of 2 mg/minute; maintenance doses of 0.05 to 2 mg/kg/hour continuous infusion dosing, titrated to EEG (Brophy et al, 2012). PEDIATRIC DOSE: 0.1 to 0.3 mg/kg followed by a continuous infusion starting at 1 mcg/kg/minute, titrated upwards every 5 minutes as needed (Loddenkemper & Goodkin, 2011).
    2) PROPOFOL: ADULT DOSE: Start at 20 mcg/kg/min with 1 to 2 mg/kg loading dose; maintenance doses of 30 to 200 mcg/kg/minute continuous infusion dosing, titrated to EEG; caution with high doses greater than 80 mcg/kg/minute in adults for extended periods of time (ie, longer than 48 hours) (Brophy et al, 2012); PEDIATRIC DOSE: IV loading dose of up to 2 mg/kg; maintenance doses of 2 to 5 mg/kg/hour may be used in older adolescents; avoid doses of 5 mg/kg/hour over prolonged periods because of propofol infusion syndrome (Loddenkemper & Goodkin, 2011); caution with high doses greater than 65 mcg/kg/min in children for extended periods of time; contraindicated in small children (Brophy et al, 2012).
    3) PENTOBARBITAL: ADULT DOSE: A loading dose of 5 to 15 mg/kg at an infusion rate of 50 mg/minute or lower; may administer additional 5 to 10 mg/kg. Maintenance dose of 0.5 to 5 mg/kg/hour continuous infusion dosing, titrated to EEG (Brophy et al, 2012). PEDIATRIC DOSE: A loading dose of 3 to 15 mg/kg followed by a maintenance dose of 1 to 5 mg/kg/hour (Loddenkemper & Goodkin, 2011).
    4) THIOPENTAL: ADULT DOSE: 2 to 7 mg/kg, at an infusion rate of 50 mg/minute or lower. Maintenance dose of 0.5 to 5 mg/kg/hour continuous infusing dosing, titrated to EEG (Brophy et al, 2012)
    b) Endotracheal intubation, mechanical ventilation, and vasopressors will be required (Brophy et al, 2012) and consultation with a neurologist is strongly advised.
    c) Neuromuscular paralysis (eg, rocuronium bromide, a short-acting nondepolarizing agent) may be required to avoid hyperthermia, severe acidosis, and rhabdomyolysis. If rhabdomyolysis is possible, avoid succinylcholine chloride, because of the risk of hyperkalemic-induced cardiac dysrhythmias. Continuous EEG monitoring is mandatory if neuromuscular paralysis is used (Manno, 2003).
    F) METHEMOGLOBINEMIA
    1) CAUSE
    a) The goal of nitrite therapy has been to achieve a methemoglobin level of 20% to 30%. This level is not based on clinical data, but represents the tolerated concentration without significant adverse symptoms from methemoglobin in an otherwise healthy individual. Clinical response has been reported to occur with methemoglobin levels in the range of 3.6% to 9.2% (DiNapoli et al, 1989; Johnson et al, 1989; Johnson & Mellors, 1988).
    b) Clinically significant excessive methemoglobinemia has rarely occurred following sodium nitrite therapy for cyanide poisoning. It usually occurs in children receiving excessive nitrite doses. Intravenous sodium nitrite administered as a 3% solution over 15 to 20 minutes induces approximately 7% to 14% methemoglobin (Kirk et al, 1993).
    2) TREATMENT
    a) If excessive methemoglobinemia occurs, some authors have suggested that methylene blue should not be used because it could cause release of cyanide from the cyanmethemoglobin complex. Such authors have suggested that emergency exchange transfusion is the treatment of choice (Berlin, 1970). Hyperbaric oxygen therapy could be used to support the patient while preparations for exchange transfusion are being made.
    b) However, methylene or toluidine blue have been used successfully in this setting without worsening the course of the cyanide poisoning (van Heijst et al, 1987). There is some controversy over whether or not the induction of methemoglobinemia is the sodium nitrite mechanism of action in cyanide poisoning. As long as intensive care monitoring and further antidote doses (if required) are available, methylene blue can most likely be safely administered in this setting.
    c) SUMMARY
    1) Determine the methemoglobin concentration and evaluate the patient for clinical effects of methemoglobinemia (ie, dyspnea, headache, fatigue, CNS depression, tachycardia, metabolic acidosis). Treat patients with symptomatic methemoglobinemia with methylene blue (this usually occurs at methemoglobin concentrations above 20% to 30%, but may occur at lower methemoglobin concentrations in patients with anemia, or underlying pulmonary or cardiovascular disorders). Administer oxygen while preparing for methylene blue therapy.
    d) METHYLENE BLUE
    1) INITIAL DOSE/ADULT OR CHILD: 1 mg/kg IV over 5 to 30 minutes; a repeat dose of up to 1 mg/kg may be given 1 hour after the first dose if methemoglobin levels remain greater than 30% or if signs and symptoms persist. NOTE: Methylene blue is available as follows: 50 mg/10 mL (5 mg/mL or 0.5% solution) single-dose ampules (Prod Info PROVAYBLUE(TM) intravenous injection, 2016) and 10 mg/1 mL (1% solution) vials (Prod Info methylene blue 1% intravenous injection, 2011). REPEAT DOSES: Additional doses may be required, especially for substances with prolonged absorption, slow elimination, or those that form metabolites that produce methemoglobin. NOTE: Large doses of methylene blue may cause methemoglobinemia or hemolysis (Howland, 2006). Improvement is usually noted shortly after administration if diagnosis is correct. Consider other diagnoses or treatment options if no improvement has been observed after several doses. If intravenous access cannot be established, methylene blue may also be given by intraosseous infusion. Methylene blue should not be given by subcutaneous or intrathecal injection (Prod Info methylene blue 1% intravenous injection, 2011; Herman et al, 1999). NEONATES: DOSE: 0.3 to 1 mg/kg (Hjelt et al, 1995).
    2) CONTRAINDICATIONS: G-6-PD deficiency (methylene blue may cause hemolysis), known hypersensitivity to methylene blue, methemoglobin reductase deficiency (Shepherd & Keyes, 2004)
    3) FAILURE: Failure of methylene blue therapy suggests: inadequate dose of methylene blue, inadequate decontamination, NADPH dependent methemoglobin reductase deficiency, hemoglobin M disease, sulfhemoglobinemia, or G-6-PD deficiency. Methylene blue is reduced by methemoglobin reductase and nicotinamide adenosine dinucleotide phosphate (NADPH) to leukomethylene blue. This in turn reduces methemoglobin. Red blood cells of patients with G-6-PD deficiency do not produce enough NADPH to convert methylene blue to leukomethylene blue (do Nascimento et al, 2008).
    4) DRUG INTERACTION: Concomitant use of methylene blue with serotonergic drugs, including serotonin reuptake inhibitors (SRIs), selective serotonin reuptake inhibitors (SSRIs), serotonin and norepinephrine reuptake inhibitors (SNRIs), tricyclic antidepressants (TCAs), norepinephrine-dopamine reuptake inhibitors (NDRIs), triptans, and ergot alkaloids may increase the risk of potentially fatal serotonin syndrome (U.S. Food and Drug Administration, 2011; Stanford et al, 2010; Prod Info methylene blue 1% IV injection, 2011).
    e) TOLUIDINE BLUE OR TOLONIUM CHLORIDE (GERMANY)
    1) DOSE: 2 to 4 mg/kg intravenously over 5 minutes. Dose may be repeated in 30 minutes (Nemec, 2011; Lindenmann et al, 2006; Kiese et al, 1972).
    2) SIDE EFFECTS: Hypotension with rapid intravenous administration. Vomiting, diarrhea, excessive sweating, hypotension, dysrhythmias, hemolysis, agranulocytosis and acute renal insufficiency after overdose (Dunipace et al, 1992; Hix & Wilson, 1987; Winek et al, 1969; Teunis et al, 1970; Marquez & Todd, 1959).
    3) CONTRAINDICATIONS: G-6-PD deficiency; may cause hemolysis.
    G) HYPOTENSIVE EPISODE
    1) SUMMARY
    a) Infuse 10 to 20 milliliters/kilogram of isotonic fluid and keep the patient supine. If hypotension persists, administer dopamine or norepinephrine. Consider central venous pressure monitoring to guide further fluid therapy.
    2) DOPAMINE
    a) DOSE: Begin at 5 micrograms per kilogram per minute progressing in 5 micrograms per kilogram per minute increments as needed (Prod Info dopamine hcl, 5% dextrose IV injection, 2004). If hypotension persists, dopamine may need to be discontinued and a more potent vasoconstrictor (eg, norepinephrine) should be considered (Prod Info dopamine hcl, 5% dextrose IV injection, 2004).
    b) CAUTION: If ventricular dysrhythmias occur, decrease rate of administration (Prod Info dopamine hcl, 5% dextrose IV injection, 2004). Extravasation may cause local tissue necrosis, administration through a central venous catheter is preferred (Prod Info dopamine hcl, 5% dextrose IV injection, 2004).
    3) NOREPINEPHRINE
    a) PREPARATION: 4 milligrams (1 amp) added to 1000 milliliters of diluent provides a concentration of 4 micrograms/milliliter of norepinephrine base. Norepinephrine bitartrate should be mixed in dextrose solutions (dextrose 5% in water, dextrose 5% in saline) since dextrose-containing solutions protect against excessive oxidation and subsequent potency loss. Administration in saline alone is not recommended (Prod Info norepinephrine bitartrate injection, 2005).
    b) DOSE
    1) ADULT: Dose range: 0.1 to 0.5 microgram/kilogram/minute (eg, 70 kg adult 7 to 35 mcg/min); titrate to maintain adequate blood pressure (Peberdy et al, 2010).
    2) CHILD: Dose range: 0.1 to 2 micrograms/kilogram/minute; titrate to maintain adequate blood pressure (Kleinman et al, 2010).
    3) CAUTION: Extravasation may cause local tissue ischemia, administration by central venous catheter is advised (Peberdy et al, 2010).
    H) 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).
    I) DICOBALT EDETATE
    1) Dicobalt-EDTA (Kelocyanor(R)) is a highly effective cyanide chelating agent currently used clinically in Europe, Israel, and Australia (Davison, 1969; Hillman et al, 1974). It is not available in the United States.
    2) Kelocyanor(R) is supplied in 20 mL ampules containing 300 mg of dicobalt-EDTA and 4 g of dextrose in water for injection (Prod Info Kelocyanor(R), 1987).
    3) DOSE
    a) ADULT DOSE: One to two 20 mL ampules (300 to 600 mg) IV over about 1 to 5 minutes (Prod Info Kelocyanor(R), 1987) (Davison, 1969).
    1) If there is insufficient clinical improvement 5 minutes after giving the first 1 to 2 ampules (300 to 600 mg), an additional 20 milliliter ampule (300 mg) may be administered intravenously over about 1 to 5 minutes (Prod Info Kelocyanor(R), 1987) (Davison, 1969).
    2) Manufacturers recommend that the intravenous site where Kelocyanor(R) has been injected be flushed with 50 mL of 50% dextrose in water (Prod Info Kelocyanor(R), 1987).
    3) Kelocyanor(R) can be used with other standard cyanide antidotes (Prod Info Kelocyanor(R), 1978).
    b) PEDIATRIC DOSE: Pediatric doses have not been established by manufacturers. In Israel the recommended pediatric dose is 0.5 milliliter/kilogram, not to exceed 20 mL (Pers Comm, Uri Taitelman,MD, 1963).
    4) ADVERSE EFFECTS
    a) Serious adverse effects include hypotension, cardiac dysrhythmias, decrease cerebral blood flow, and angioedema (Dodds & McKnight, 1985; Wright & Vesey, 1986). These adverse reactions are magnified when a patient does not have cyanide intoxication. It may be prudent to reserve the use of Kelocyanor(R) to confirmed cyanide poisonings. The inherent problems in delaying treatment while waiting for confirmation is apparent and relegates this antidote to limited clinical usefulness; unless cyanide poisoning can be confirmed readily (Pronczuk de Garbino & Bismuth, 1981; Tyrer, 1981). Therefore, KELOCYANOR(R) SHOULD NOT BE USED IN CASES OF MILD CYANIDE POISONING OR DIAGNOSTIC UNCERTAINTY (Peden et al, 1986; Tyrer, 1981).
    b) ADVERSE EFFECTS may include nausea, vomiting, tachycardia, hypotension, hypertension, anaphylactic reactions, facial and neck edema, chest pain, diaphoresis, nervousness, tremulousness, gastrointestinal hemorrhages, seizures, cardiac arrhythmias, and rashes (Prod Info Kelocyanor(R), 1987 ) (Davison, 1969; Tyrer, 1981; Hillman et al, 1974; Dodds & McKnight, 1985; Wright & Vesey, 1986).
    J) 4-DIMETHYLAMINOPHENOL HYDROCHLORIDE
    1) 4-DMAP is a methemoglobin-inducing agent used in some European countries for the treatment of acute cyanide poisoning. A more rapid onset of methemoglobin production is observed following administration of 4-DMAP than following sodium nitrite. Methemoglobin peaks at 5 minutes after 4-DMAP versus 30 minutes after sodium nitrite (Kruszyna et al, 1982; Moore et al, 1987; Weger, 1990).
    2) The dose of 4-DMAP is 3 mg/kg and is coadministered with thiosulfate.
    3) ADVERSE EFFECTS: Excessive methemoglobinemia may be a major complication following the use of this agent (van Dijk et al, 1986). Hemolysis may occur with therapeutic doses (van Heijst et al, 1987).
    4) CASE REPORT: In Germany, 2 cases of life-threatening hydrocyanic acid (HCN) gas poisoning occurred via inhalation and percutaneous exposure. At the scene, a firefighter wearing a self-contained breathing apparatus, but no protective chemical suit was overcome and collapsed. Both victims were immediately given 4-DMAP (at approximately 50% the recommended dose). Cyanide levels were 5.3 and 6.75 mg/L, respectively upon arrival at the ED. Methemoglobin was 5.5 and 16.2%, respectively. Each patient also received 50 and 150 mg/kg N-acetylcysteine (NAC), respectively, along with 30 and 40 mL sodium thiosulfate 10%. Cyanide levels decreased (0.035 and 0.036 mg/L, respectively) significantly the following day, and both made a complete recovery. The authors concluded that NAC and sodium thiosulfate appeared to be efficient treatment of cyanide poisoning. Further study is recommended (Steffens et al, 2003).
    K) HYPERBARIC OXYGEN THERAPY
    1) The Undersea Medical Society has classified cyanide poisoning as a disorder for which hyperbaric oxygen therapy is mandatory (Category 1: approved for third party reimbursement and known effective as treatment) (Myers & Schnitzer, 1984). Category 1, a category intended for disorders in which the efficacy of hyperbaric oxygen has been established in extensive clinical trials. The placement of cyanide poisoning in Category 1 stands in contrast to the existing literature, which indicates that the role of hyperbaric oxygen as an adjunct to the chemical antidote treatment of the cyanide poisoned patient has not been clearly established.
    2) Animal data suggests that hyperbaric oxygen treatment may have direct effects in attenuating cyanide toxicity (Ivanov, 1959; Skene et al, 1966; Takano et al, 1980). However, not all animal studies have shown hyperbaric oxygen to improve outcome (Way et al, 1972).
    3) The reported results involving hyperbaric oxygen treatment in clinical cases of isolated cyanide poisonings refractory to standard antidote treatment (sodium nitrite and sodium thiosulfate) have been equivocal (Trapp, 1970; Litovitz et al, 1983; Trapp & Lepawsky, 1983; Hart et al, 1985; Davis & Ewer, 1988; Rosenberg et al, 1989; Feldman & Feldman, 1990; Scolnick et al, 1993; Goodhart, 1994). Further research in hyperbaric oxygen treatment and cyanide poisoning is necessary.
    4) The literature seems to indicate that the role of hyperbaric oxygen as an adjunct to the chemical antidote treatment of the cyanide poisoned patient has not been clearly established. Further research in this area is necessary. Because cyanide is among the most lethal poisons, and intoxication is rapid, "standard antidotal therapy" for isolated cyanide poisoning should be of primary importance. Hyperbaric oxygen may be an adjunct to be considered in patients who are not responding to supportive care and antidotal therapy, and for those patients poisoned by both cyanide and carbon monoxide (Hart et al, 1985).
    L) EXPERIMENTAL THERAPY
    1) STROMA-FREE METHEMOGLOBIN SOLUTION
    a) Stroma-free methemoglobin, oxidized hemoglobin to the ferric form in which the cell membrane has been removed, attenuates lethality and prevents hemodynamic changes in animal models (Breen et al, 1996; Ten Eyck et al, 1985) (Ten Eyck et al, 1986). The advantage to this treatment is that it provide exogenous methemoglobin without compromising oxygen-carrying capacity of native hemoglobin. Removal of the cell membrane eliminates the antigenicity problem (Marrs, 1988).
    b) AVAILABILITY: It has not been studied in human poisoning cases and is not available for human administration.
    2) ALPHA-KETOGLUTARIC ACID
    a) Alpha-ketoglutaric acid is also being tested as a replacement for sodium nitrite in combination with sodium thiosulfate in animal models where it has been efficacious in experimental cyanide poisoning (Bhattacharya & Vijayaraghavan, 2002; Moore et al, 1986).
    b) In vitro and animal studies show that alpha-ketaglutaric acid binds with cyanide and thereby antagonizes cyanide-induced inhibition of brain cytochrome oxidase (Norris et al, 1990).
    c) Alpha-ketoglutaric acid administered with sodium thiosulfate abolished the cyanide-induced decrease in brain gamma-aminobutyric acid in mice (Yamamoto, 1990).
    d) It has not been studied in human poisoning cases and is not available for human administration.
    3) 3-MERCAPTOPYRUVATE PRODRUGS
    a) A series of 3 prodrugs of 3-mercaptopyruvate (3-MP) have been developed as possible cyanide antidotes and have been successfully tested in animal models. When given intraperitoneally to mice 5 minutes post-cyanide administration, the protective index (P.I.; a ratio of the righting reflex recovery time of the cyanide-only controls divided by the righting reflex recovery times of the cyanide-plus-antidote-treated animals) ranged from 1.4 to 3.9 as compared with the cyanide-only controls with a P.I. of 1. The prodrugs also appeared to be effective when given to mice prior to cyanide administration. The P.I of the prodrugs ranged from 2.0 to 5.4 when given orally 30 to 60 minutes pre-cyanide administration, suggesting that the prodrugs may be useful as prophylactic agents by first responders; further investigation is warranted (Nagasawa et al, 2007).

Inhalation Exposure

    6.7.1) DECONTAMINATION
    A) Move patient from the toxic environment to fresh air. Monitor for respiratory distress. If cough or difficulty in breathing develops, evaluate for hypoxia, respiratory tract irritation, bronchitis, or pneumonitis.
    B) OBSERVATION: Carefully observe patients with inhalation exposure for the development of any systemic signs or symptoms and administer symptomatic treatment as necessary.
    C) INITIAL TREATMENT: Administer 100% humidified supplemental oxygen, perform endotracheal intubation and provide assisted ventilation as required. Administer inhaled beta-2 adrenergic agonists, if bronchospasm develops. Consider systemic corticosteroids in patients with significant bronchospasm (National Heart,Lung,and Blood Institute, 2007). Exposed skin and eyes should be flushed with copious amounts of water.
    D) High concentrations of cyanide gas may cause a rapid loss of consciousness (Peden et al, 1986). Rescuers should wear self-contained positive pressure breathing apparatus to avoid contaminating themselves during rescue attempts (AAR, 1987) (NFPA, 1986).
    6.7.2) TREATMENT
    A) OXYGEN
    1) Administer 100% oxygen to maintain an elevated PO2.
    a) Oxygen may reverse the cyanide-cytochrome oxidase complex and facilitate the conversion to thiocyanate following thiosulfate administration (Graham et al, 1977).
    b) There is fairly good evidence that 100% oxygen, combined with traditional nitrite/thiosulfate therapy is better than thiosulfate alone (Litovitz et al, 1983; Way et al, 1972).
    B) HYPERBARIC OXYGEN THERAPY
    1) Hyperbaric oxygen (HBO) therapy is approved for cyanide poisoning as a category one condition (approved for third party reimbursement and known effective as treatment) by the Undersea Medical Society (Myers & Schnitzer, 1984).
    2) Hyperbaric oxygen has been suggested to improve clinical outcome, especially in those patients with CNS toxicity not responding to more traditional therapy.
    3) Animal studies have both confirmed and refuted this (Skene et al, 1966; Way et al, 1972; Takano et al, 1980).
    4) Case reports suggest that HBO may be of value (Carden, 1970; Trapp, 1970). However, one patient treated with supportive therapy, antidotes, and HBO did not survive a serious poisoning (Litovitz et al, 1983).
    5) Hyperbaric oxygen should be reserved for those patients with significant symptoms (ie, coma, seizures) who do not respond to normal supportive and antidotal therapy, and for those patients poisoned by both cyanide and carbon monoxide secondary to smoke inhalation (Hart et al, 1985).
    C) FLUID/ELECTROLYTE BALANCE REGULATION
    1) Establish secure large bore IV line.
    D) CYANIDE ANTIDOTE
    1) SUMMARY
    a) A cyanide antidote, either hydroxocobalamin or the sodium nitrite/sodium thiosulfate kit, should be administered to patients with symptomatic poisoning. If cyanide toxicity develops concurrent with carbon monoxide poisoning (e.g., closed space fire), hydroxocobalamin is the preferred antidote. It that is not available, sodium thiosulfate may be used alone. Use of amyl nitrite or sodium nitrite will cause methemoglobinemia, further reducing oxygen carrying capacity.
    2) HYDROXOCOBALAMIN
    a) ADULT DOSE: 5 g (two 2.5 g vials each reconstituted with 100 mL sterile 0.9% saline) administered as an intravenous infusion over 15 minutes. For severe poisoning, a second dose of 5 g may be infused intravenously over 15 minutes to 2 hours, depending on the patient's condition (Prod Info CYANOKIT(R) 2.5g IV injection, 2006).
    b) PEDIATRIC DOSE: A dose of 70 mg/kg has been used in pediatric patients, based on limited post-marketing experience outside the US (Prod Info CYANOKIT(R) 2.5g IV injection, 2006).
    c) INDICATIONS: Known or suspected cyanide poisoning.
    d) ADVERSE EFFECTS: Transient hypertension, allergic reactions (including anaphylaxis), nausea, headache, rash. Hydroxocobalamin's deep red color causes red-colored urine in all patients, and erythema of the skin in most (Prod Info CYANOKIT(R) 2.5g IV injection, 2006).
    e) LABORATORY INTERFERENCE: Because of its' color, hydroxocobalamin interferes with colorimetric determination of various laboratory parameters. It may artificially increase serum creatinine, bilirubin, triglycerides, cholesterol, total protein, glucose, albumin , alkaline phosphatase and hemoglobin. It may artificially decrease serum ALT and amylase. It may artificially increase urinary pH, glucose, protein, erythrocytes, leukocytes, ketones, bilirubin, urobilinogen, and nitrate (Prod Info CYANOKIT(R) 2.5g IV injection, 2006).
    f) HEMODIALYSIS INTERFERENCE: Dialysis machines have a spectrophotometric safety measure that can shut down after detecting blood leaking across the dialysis membrane. Hydroxocobalamin has a deep red color and can permeate the dialysis membrane, coloring the dialysate and causing the hemodialysis machine to shut down erroneously. In one case report, a patient with cyanide poisoning underwent dialysis after receiving 5 g of IV hydroxocobalamin because of refractory acidemia, reduced kidney function and hyperkalemia. A blood leak alarm caused the dialysis machine to shut down erroneously, delaying therapy, and resulting in the death of the patient (Stellpflug et al, 2013).
    g) Hydroxocobalamin, 5 to 20 grams administered intravenously, has been shown to be effective, as sole antidotal therapy, for acute cyanide poisoning following cyanide salt ingestion or inhalation, as well as cyanide poisoning following smoke inhalation (Borron et al, 2007b; Borron et al, 2007a).
    h) In a controlled pilot study of male smokers, administration of 5 grams of intravenous 5% hydroxocobalamin significantly decreased whole blood cyanide levels (Forsyth et al, 1992).
    3) CYANIDE ANTIDOTE KIT
    a) OBTAIN AND PREPARE for administration a CYANIDE ANTIDOTE KIT, consisting of sodium nitrite and sodium thiosulfate.
    1) Antidotes should be used only in significantly symptomatic patients (ie, impaired consciousness, convulsions, acidosis, or unstable vital signs).
    2) Even when patients are rendered comatose by the inhalation of hydrogen cyanide gas, antidotes may not be necessary if the exposure is rapidly terminated, the patient has regained consciousness on arrival at the medical facility, and there is no acidosis or abnormality of the vital signs (Peden et al, 1986).
    b) SODIUM NITRITE
    1) INDICATION
    a) Sodium nitrite should be given initially and administered as soon as vascular access is established.
    b) Further administration of sodium nitrite is dictated only by the clinical situation, provided no significant complications (hypotension, excessive methemoglobinemia) are present. Use with caution if carbon monoxide poisoning is also suspected.
    c) The goal of nitrite therapy is to achieve a methemoglobin level of 20% to 30%. This level is not based on clinical data, but represents the tolerated concentration without significant adverse symptoms from methemoglobin in an otherwise healthy individual. Clinical response has been reported to occur with methemoglobin levels in the range of 3.6% to 9.2% (DiNapoli et al, 1989; Johnson et al, 1989; Johnson & Mellors, 1988).
    2) ADULT DOSE
    a) 10 mL of a 3% solution (300 mg) administered intravenously at a rate of 2.5 to 5 mL/minute (Prod Info NITHIODOTE intravenous injection solution, 2011). Frequent blood pressure monitoring must accompany sodium nitrite injection and the rate slowed if hypotension occurs.
    b) If there is inadequate clinical response, an additional dose of sodium nitrite at half the amount of the initial dose may be administered 30 minutes following the first dose (Prod Info NITHIODOTE intravenous injection solution, 2011).
    3) PEDIATRIC DOSE
    a) The recommended pediatric sodium nitrite dose is 0.2 mL/kg of a 3% solution (6 mg/kg) administered intravenously at a rate of 2.5 to 5 mL/minute, not to exceed 10 mL (300 mg) (Prod Info NITHIODOTE intravenous injection solution, 2011).
    b) If there is inadequate clinical response, an additional dose of sodium nitrite at half the amount of the initial dose may be administered 30 minutes following the first dose (Prod Info NITHIODOTE intravenous injection solution, 2011; Berlin, 1970a).
    c) PRESENCE OF ANEMIA: If there is a reason to suspect the presence of anemia, the following initial sodium nitrite doses should be given, depending on the child's hemoglobin (sodium nitrite should not exceed the doses listed below; fatal methemoglobinemia may result) (Berlin, 1970a):
    1) Hemoglobin: 8 g/dL - Initial 3% sodium nitrite dose: 0.22 mL/kg (6.6 mg/kg)
    2) Hemoglobin: 10 g/dL - Initial 3% sodium nitrite dose: 0.27 mL/kg (8.7 mg/kg)
    3) Hemoglobin: 12 g/dL (average child) - Initial 3% sodium nitrite dose: 0.33 mL/kg (10 mg/kg)
    4) Hemoglobin: 14 g/dL - Initial 3% sodium nitrite dose: 0.39 mL/kg (11.6 mg/kg)
    4) It is highly recommended that total hemoglobin and methemoglobin concentrations be rapidly measured (30 minutes after dose), when possible, before repeating a dose of sodium nitrite to be sure that dangerous methemoglobinemia will not occur, especially in the pediatric patient.
    5) Monitor blood pressure frequently and treat hypotension by slowing infusion rate and giving crystalloids and vasopressors. Consider possible excessive methemoglobin formation if patient deteriorates during therapy.
    6) Excessive methemoglobinemia and hypotension are potential complications of nitrite therapy.
    7) In individuals with G6PD deficiency, therapy with methemoglobin-inducing agents is contraindicated because of the likelihood of serious hemolysis.
    c) SODIUM THIOSULFATE
    1) Sodium thiosulfate is the second component of the cyanide antidote kit. It is supplied as 50 mL of a 25% solution and it is administered intravenously. There are no adverse reactions to thiosulfate itself. The pediatric dose is adjusted for weight and not hemoglobin concentration.
    2) Sodium thiosulfate supplies sulfur for the rhodanese reaction, and is recommended after sodium nitrite, hydroxocobalamin, or 4-DMAP (4-dimethylaminophenol) administration (Marrs, 1988; Hall & Rumack, 1987).
    3) DOSE
    a) Follow sodium nitrite with IV sodium thiosulfate. ADULT: Administer 50 mL (12.5 g) of a 25% solution IV; PEDIATRIC: 1 mL/kg of a 25% solution (250 mg/kg), not to exceed 50 mL (12.5 g) total dose (Prod Info NITHIODOTE intravenous injection solution, 2011).
    b) A second dose, one-half of the first dose, may be administered if signs of cyanide toxicity reappear (Prod Info NITHIODOTE intravenous injection solution, 2011).
    c) Sodium thiosulfate is usually used in combination with sodium nitrite but may be used alone (Prod Info sodium thiosulfate IV injection, 2003).
    d) Sodium thiosulfate can be administered without sodium nitrite in patients at risk to develop further methemoglobinemia (ie excessive methemoglobinemia or hypotension after initial sodium nitrite administration or in the presence of methemoglobinemia or carboxyhemoglobin in patients with smoke inhalation due to fire). Sodium thiosulfate can also be used in combination with hydroxocobalamin to treat cyanide poisoning (Howland, 2011)
    e) CONTINUOUS INFUSION: It has been suggested that a continuous infusion of sodium thiosulfate be given after the initial bolus to maintain high thiosulfate levels. Low sodium intravenous fluids are required to avoid sodium overload. If large amounts of sodium thiosulfate are required, hemodialysis may be necessary to maintain a physiologic serum sodium level (Turchen et al, 1991).
    f) ADVERSE EVENTS: Sodium thiosulfate does not usually produce significant toxicity. Possible adverse events include hypotension, headache, nausea, vomiting, disorientation, and prolonged bleeding time (Prod Info NITHIODOTE intravenous injection solution, 2011).
    E) MONITORING OF PATIENT
    1) Obtain blood for arterial blood gases with venous pO2 or measured venous %O2 saturation, electrolytes, serum lactate, and whole blood cyanide levels.
    2) The following set of laboratory values suggest poisoning with an agent that inhibits oxidative phosphorylation (ie, cyanide, hydrogen sulfide) (Hall & Rumack, 1986).
    a) Cyanide and hydrogen sulfide poisoning are treated in essentially the same manner, (See Hydrogen Sulfide Management) with the exception of lack of efficacy of sodium thiosulfate in hydrogen sulfide poisoning. Administering sodium thiosulfate will most likely do no harm to a hydrogen sulfide poisoned patient.
    b) Arterial pO2: Usually normal until the stage of apnea. Usually remains normal until terminal stages of the poisoning if supplemental oxygen and assisted ventilation are provided.
    c) Serum electrolytes: Elevated anion gap (Na - (C1 + CO2)) (normals 12 to 16 milliequivalents/liter (12 to 16 millimoles/liter) or less) is present from the presence of unmeasured organic anions (usually lactate).
    d) Serum Lactate: Elevated (normals 0.6 to 1.8 milliequivalents/liter) (0.6 to 1.8 millimoles/liter) due to anaerobic metabolism with excessive production of lactic acid.
    e) Arterio-Central Venous Measured %O2 Saturation Difference: Due to cellular inability to extract and use oxygen, more is present on the venous side. The MEASURED values of arterial and central venous %O2 saturation approach each other with MEASURED central venous %O2 saturation greater than 70%.
    1) Arteriolization of venous blood gases (elevated venous pO2 or measured venous %O2 saturation) may serve as an early clue in the diagnosis of cyanide poisoning (Hall & Rumack, 1986; Johnson & Mellors, 1988).
    F) ACIDOSIS
    1) METABOLIC ACIDOSIS: Treat severe metabolic acidosis (pH less than 7.1) with sodium bicarbonate, 1 to 2 mEq/kg is a reasonable starting dose(Kraut & Madias, 2010). Monitor serum electrolytes and arterial or venous blood gases to guide further therapy.
    G) SEIZURE
    1) SUMMARY
    a) Attempt initial control with a benzodiazepine (eg, diazepam, lorazepam). If seizures persist or recur, administer phenobarbital or propofol.
    b) Monitor for respiratory depression, hypotension, and dysrhythmias. Endotracheal intubation should be performed in patients with persistent seizures.
    c) Evaluate for hypoxia, electrolyte disturbances, and hypoglycemia (or, if immediate bedside glucose testing is not available, treat with intravenous dextrose).
    2) DIAZEPAM
    a) ADULT DOSE: Initially 5 to 10 mg IV, OR 0.15 mg/kg IV up to 10 mg per dose up to a rate of 5 mg/minute; may be repeated every 5 to 20 minutes as needed (Brophy et al, 2012; Prod Info diazepam IM, IV injection, 2008; Manno, 2003).
    b) PEDIATRIC DOSE: 0.1 to 0.5 mg/kg IV over 2 to 5 minutes; up to a maximum of 10 mg/dose. May repeat dose every 5 to 10 minutes as needed (Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008).
    c) Monitor for hypotension, respiratory depression, and the need for endotracheal intubation. Consider a second agent if seizures persist or recur after repeated doses of diazepam .
    3) NO INTRAVENOUS ACCESS
    a) DIAZEPAM may be given rectally or intramuscularly (Manno, 2003). RECTAL DOSE: CHILD: Greater than 12 years: 0.2 mg/kg; 6 to 11 years: 0.3 mg/kg; 2 to 5 years: 0.5 mg/kg (Brophy et al, 2012).
    b) MIDAZOLAM has been used intramuscularly and intranasally, particularly in children when intravenous access has not been established. ADULT DOSE: 0.2 mg/kg IM, up to a maximum dose of 10 mg (Brophy et al, 2012). PEDIATRIC DOSE: INTRAMUSCULAR: 0.2 mg/kg IM, up to a maximum dose of 7 mg (Chamberlain et al, 1997) OR 10 mg IM (weight greater than 40 kg); 5 mg IM (weight 13 to 40 kg); INTRANASAL: 0.2 to 0.5 mg/kg up to a maximum of 10 mg/dose (Loddenkemper & Goodkin, 2011; Brophy et al, 2012). BUCCAL midazolam, 10 mg, has been used in adolescents and older children (5-years-old or more) to control seizures when intravenous access was not established (Scott et al, 1999).
    4) LORAZEPAM
    a) MAXIMUM RATE: The rate of intravenous administration of lorazepam should not exceed 2 mg/min (Brophy et al, 2012; Prod Info lorazepam IM, IV injection, 2008).
    b) ADULT DOSE: 2 to 4 mg IV initially; repeat every 5 to 10 minutes as needed, if seizures persist (Manno, 2003; Brophy et al, 2012).
    c) PEDIATRIC DOSE: 0.05 to 0.1 mg/kg IV over 2 to 5 minutes, up to a maximum of 4 mg/dose; may repeat in 5 to 15 minutes as needed, if seizures continue (Brophy et al, 2012; Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008; Sreenath et al, 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).
    7) PHENYTOIN/FOSPHENYTOIN
    a) Benzodiazepines and/or barbiturates are preferred to phenytoin or fosphenytoin in the treatment of drug or withdrawal induced seizures (Wallace, 2005).
    b) PHENYTOIN
    1) PHENYTOIN INTRAVENOUS PUSH VERSUS INTRAVENOUS INFUSION
    a) Administer phenytoin undiluted, by very slow intravenous push or dilute 50 mg/mL solution in 50 to 100 mL of 0.9% saline.
    b) ADULT DOSE: A loading dose of 20 mg/kg IV; may administer an additional 5 to 10 mg/kg dose 10 minutes after loading dose. Rate of administration should not exceed 50 mg/minute (Brophy et al, 2012).
    c) PEDIATRIC DOSE: A loading dose of 20 mg/kg, at a rate not exceeding 1 to 3 mg/kg/min or 50 mg/min, whichever is slower (Loddenkemper & Goodkin, 2011; Prod Info Dilantin(R) intravenous injection, intramuscular injection, 2013).
    d) CAUTIONS: Administer phenytoin while monitoring ECG. Stop or slow infusion if dysrhythmias or hypotension occur. Be careful not to extravasate. Follow each injection with injection of sterile saline through the same needle (Prod Info Dilantin(R) intravenous injection, intramuscular injection, 2013).
    e) SERUM CONCENTRATION MONITORING: Monitor serum concentrations over next 12 to 24 hours for maintenance of therapeutic concentrations. Therapeutic concentrations of 10 to 20 mcg/mL have been reported (Prod Info Dilantin(R) intravenous injection, intramuscular injection, 2013).
    c) FOSPHENYTOIN
    1) ADULT DOSE: A loading dose of 20 mg phenytoin equivalent/kg IV, at a rate not exceeding 150 mg phenytoin equivalent/minute; may give additional dose of 5 mg/kg 10 minutes after the loading infusion (Brophy et al, 2012).
    2) CHILD DOSE: 20 mg phenytoin equivalent/kg IV, at a rate of 3 mg phenytoin equivalent/kg/minute, up to a maximum of 150 mg phenytoin equivalent/minute (Loddenkemper & Goodkin, 2011).
    3) CAUTIONS: Perform continuous monitoring of ECG, respiratory function, and blood pressure throughout the period where maximal serum phenytoin concentrations occur (about 10 to 20 minutes after the end of fosphenytoin infusion) (Prod Info CEREBYX(R) intravenous injection, 2014).
    4) SERUM CONCENTRATION MONITORING: Monitor serum phenytoin concentrations over the next 12 to 24 hours; therapeutic levels 10 to 20 mcg/mL. Do not obtain serum phenytoin concentrations until at least 2 hours after infusion is complete to allow for conversion of fosphenytoin to phenytoin (Prod Info CEREBYX(R) intravenous injection, 2014).
    H) METHEMOGLOBINEMIA
    1) While clinically significant excessive methemoglobinemia has occurred following sodium nitrite therapy for cyanide poisoning, such instances are rare and usually occur in children receiving excessive nitrite doses.
    a) Inducing a "therapeutic methemoglobin level" of 25% is unnecessary to insure satisfactory clinical outcome (Johnson et al, 1989).
    2) If excessive methemoglobinemia occurs, some authors have suggested that methylene blue should not be used because it could cause release of cyanide from the cyanmethemoglobin complex. Such authors have suggested that emergency exchange transfusion is the treatment of choice (Berlin, 1970). Hyperbaric oxygen therapy could be used to support the patient while preparations for exchange transfusion are being made.
    3) However, methylene or toluidine blue have been used successfully in this setting without worsening the course of the cyanide poisoning (van Heijst et al, 1987). There is some controversy over whether or not the induction of methemoglobinemia is the sodium nitrite mechanism of action in cyanide poisoning. As long as intensive care monitoring and further antidote doses (if required) are available, methylene blue can most likely be safely administered in this setting.
    4) SUMMARY
    a) Determine the methemoglobin concentration and evaluate the patient for clinical effects of methemoglobinemia (ie, dyspnea, headache, fatigue, CNS depression, tachycardia, metabolic acidosis). Treat patients with symptomatic methemoglobinemia with methylene blue (this usually occurs at methemoglobin concentrations above 20% to 30%, but may occur at lower methemoglobin concentrations in patients with anemia, or underlying pulmonary or cardiovascular disorders). Administer oxygen while preparing for methylene blue therapy.
    5) METHYLENE BLUE
    a) INITIAL DOSE/ADULT OR CHILD: 1 mg/kg IV over 5 to 30 minutes; a repeat dose of up to 1 mg/kg may be given 1 hour after the first dose if methemoglobin levels remain greater than 30% or if signs and symptoms persist. NOTE: Methylene blue is available as follows: 50 mg/10 mL (5 mg/mL or 0.5% solution) single-dose ampules (Prod Info PROVAYBLUE(TM) intravenous injection, 2016) and 10 mg/1 mL (1% solution) vials (Prod Info methylene blue 1% intravenous injection, 2011). REPEAT DOSES: Additional doses may be required, especially for substances with prolonged absorption, slow elimination, or those that form metabolites that produce methemoglobin. NOTE: Large doses of methylene blue may cause methemoglobinemia or hemolysis (Howland, 2006). Improvement is usually noted shortly after administration if diagnosis is correct. Consider other diagnoses or treatment options if no improvement has been observed after several doses. If intravenous access cannot be established, methylene blue may also be given by intraosseous infusion. Methylene blue should not be given by subcutaneous or intrathecal injection (Prod Info methylene blue 1% intravenous injection, 2011; Herman et al, 1999). NEONATES: DOSE: 0.3 to 1 mg/kg (Hjelt et al, 1995).
    b) CONTRAINDICATIONS: G-6-PD deficiency (methylene blue may cause hemolysis), known hypersensitivity to methylene blue, methemoglobin reductase deficiency (Shepherd & Keyes, 2004)
    c) FAILURE: Failure of methylene blue therapy suggests: inadequate dose of methylene blue, inadequate decontamination, NADPH dependent methemoglobin reductase deficiency, hemoglobin M disease, sulfhemoglobinemia, or G-6-PD deficiency. Methylene blue is reduced by methemoglobin reductase and nicotinamide adenosine dinucleotide phosphate (NADPH) to leukomethylene blue. This in turn reduces methemoglobin. Red blood cells of patients with G-6-PD deficiency do not produce enough NADPH to convert methylene blue to leukomethylene blue (do Nascimento et al, 2008).
    d) DRUG INTERACTION: Concomitant use of methylene blue with serotonergic drugs, including serotonin reuptake inhibitors (SRIs), selective serotonin reuptake inhibitors (SSRIs), serotonin and norepinephrine reuptake inhibitors (SNRIs), tricyclic antidepressants (TCAs), norepinephrine-dopamine reuptake inhibitors (NDRIs), triptans, and ergot alkaloids may increase the risk of potentially fatal serotonin syndrome (U.S. Food and Drug Administration, 2011; Stanford et al, 2010; Prod Info methylene blue 1% IV injection, 2011).
    6) TOLUIDINE BLUE OR TOLONIUM CHLORIDE (GERMANY)
    a) DOSE: 2 to 4 mg/kg intravenously over 5 minutes. Dose may be repeated in 30 minutes (Nemec, 2011; Lindenmann et al, 2006; Kiese et al, 1972).
    b) SIDE EFFECTS: Hypotension with rapid intravenous administration. Vomiting, diarrhea, excessive sweating, hypotension, dysrhythmias, hemolysis, agranulocytosis and acute renal insufficiency after overdose (Dunipace et al, 1992; Hix & Wilson, 1987; Winek et al, 1969; Teunis et al, 1970; Marquez & Todd, 1959).
    c) CONTRAINDICATIONS: G-6-PD deficiency; may cause hemolysis.
    I) DICOBALT EDETATE
    1) Kelocyanor(R) (dicobalt-EDTA) is a highly effective cyanide chelating agent currently used clinically in Europe, Israel, and Australia (Davison, 1969; Hillman et al, 1974). It is not available in the US.
    2) PRECAUTIONS
    a) Significant toxicity from the antidote (severe hypertension or hypotension, cardiac ischemia or arrhythmias) may be seen in patients incorrectly diagnosed as being poisoned with cyanide and administered this antidote (Pronczuk de Garbino & Bismuth, 1981; Tyrer, 1981). Therefore, KELOCYANOR(R) SHOULD NOT BE USED in CASES of MILD CYANIDE POISONING or DIAGNOSTIC UNCERTAINTY (Peden et al, 1986; Tyrer, 1981).
    b) Severe anaphylactoid reactions with periorbital and massive facial edema and airway compromise may also occur (Dodds & McKnight, 1985; Wright & Vesey, 1986).
    c) ADVERSE EFFECTS can include nausea, vomiting, tachycardia, hypotension, hypertension, anaphylactic reactions, facial and neck edema, chest pain, diaphoresis, nervousness, tremulousness, gastrointestinal hemorrhages, convulsions, cardiac irregularities, and rashes (Prod Info, 1986; Prod Info, 1987) (Davison, 1969; Tyrer, 1981; Hillman et al, 1974).
    3) DOSE
    a) ADULTS: One to two 20 mL ampules (300 to 600 mg) injected intravenously over about 1 to 5 minutes (Prod Info, 1978) (Prod Info, 1986) (Prod Info, 1987) (Davison, 1969).
    1) A third 20 mL ampule (300 mg) can be injected intravenously over about 1 to 5 minutes, 5 minutes after the first 1 to 2 ampules if there is not sufficient clinical improvement (Prod Info, 1978) (Prod Info, 1986) (Prod Info, 1987) (Davison, 1969).
    2) Manufacturers recommend following the Kelocyanor(R) injection with intravenous injection of 50 mL of 50% dextrose in water (Prod Info, 1978) (Prod Info, 1986) (Prod Info, 1987).
    3) Kelocyanor(R) can be used with other standard cyanide antidotes (Prod Info, 1978).
    b) CHILDREN: Pediatric doses have not been established by manufacturers. A suggested dose used in Israel for children is 0.5 milliliter per kilogram (not to exceed 20 mL) (Personal Communication, Uri Taitelman, MD, 1963).
    4) Kelocyanor(R) is supplied in 20 mL ampules containing 300 mg of dicobalt-EDTA and 4 grams of dextrose in water for injection (Prod Info, 1978) (Prod Info, 1986) (Prod Info, 1987).
    J) 4-DIMETHYLAMINOPHENOL HYDROCHLORIDE
    1) 4-DMAP is a methemoglobin inducing agent used in some European countries for the treatment of acute cyanide poisoning. Excessive methemoglobinemia may be a major complication following the use of this agent (van Dijk et al, 1986).
    2) In individuals with G6PD deficiency, therapy with methemoglobin-inducing agents is contraindicated because of the likelihood of serious hemolysis.
    K) EXPERIMENTAL THERAPY
    1) STROMA-FREE METHEMOGLOBIN SOLUTION: Stroma-free methemoglobin solution prepared from outdated human red blood cells by oxidation of the ferrous iron of hemoglobin to the ferric form in vitro has been studied in experimental animals and shows promise as a cyanide antidote (Ten Eyck et al, 1985).
    a) By virtue of its in vitro production, stroma-free methemoglobin solution has the advantage of sparing the oxygen-carrying capacity of the blood (Marrs, 1988).
    b) It has not been studied in human poisoning cases and is not available for human administration.
    2) ALPHA-KETOGLUTARIC ACID: Alpha-ketoglutaric acid is also being tested as a replacement for sodium nitrite in combination with sodium thiosulfate in animal models where it has been efficacious in experimental cyanide poisoning (Bhattacharya & Vijayaraghavan, 2002; Moore et al, 1986).
    a) In vitro and animal studies show that alpha-ketoglutaric acid binds with cyanide and thereby antagonizes cyanide-induced inhibition of brain cytochrome oxidase (Norris et al, 1990).
    b) Alpha-ketoglutaric acid administered with sodium thiosulfate abolished the cyanide-induced decrease in brain gamma-aminobutyric acid in mice (Yamamoto, 1990).
    c) It has not been studied in human poisoning cases and is not available for human administration.
    3) CHLORPROMAZINE: Chlorpromazine has been studied in various animal models as a possible cyanide antidote. Conflicting reports of efficacy have been published (Pettersen & Cohen, 1986).
    a) Animal and in vitro studies show that chlorpromazine can decrease peroxidation of lipid membranes and prevent cyanide-induced calcium influx believed to be responsible for neurotoxicity (Johnson et al, 1986; Maduh et al, 1988).
    b) It has not been studied in human poisoning cases.
    4) OTHER INVESTIGATIONAL ANTIDOTES: Animal studies to identify alternate cyanide antidotes have tested phenoxybenzamine, centrophenoxine, naloxone hydrochloride, etomidate, para-aminopropiophenone, calcium-ion-channel blockers, and prodrugs of 3-mercaptopyruvate (Amery et al, 1981; Ashton et al, 1980; Bright & Marrs, 1987; Budavari, 1976; Dubinsky et al, 1984; Johnson et al, 1986; Leung et al, 1984; Bright & Marrs, 1987; Marrs, 1988) (Rump & Edelwijn, 1986) (Vick & Froehlich, 1985; Nagasawa et al, 2007).
    L) HYPOTENSIVE EPISODE
    1) SUMMARY
    a) Infuse 10 to 20 milliliters/kilogram of isotonic fluid and keep the patient supine. If hypotension persists, administer dopamine or norepinephrine. Consider central venous pressure monitoring to guide further fluid therapy.
    2) DOPAMINE
    a) DOSE: Begin at 5 micrograms per kilogram per minute progressing in 5 micrograms per kilogram per minute increments as needed (Prod Info dopamine hcl, 5% dextrose IV injection, 2004). If hypotension persists, dopamine may need to be discontinued and a more potent vasoconstrictor (eg, norepinephrine) should be considered (Prod Info dopamine hcl, 5% dextrose IV injection, 2004).
    b) CAUTION: If ventricular dysrhythmias occur, decrease rate of administration (Prod Info dopamine hcl, 5% dextrose IV injection, 2004). Extravasation may cause local tissue necrosis, administration through a central venous catheter is preferred (Prod Info dopamine hcl, 5% dextrose IV injection, 2004).
    3) NOREPINEPHRINE
    a) PREPARATION: 4 milligrams (1 amp) added to 1000 milliliters of diluent provides a concentration of 4 micrograms/milliliter of norepinephrine base. Norepinephrine bitartrate should be mixed in dextrose solutions (dextrose 5% in water, dextrose 5% in saline) since dextrose-containing solutions protect against excessive oxidation and subsequent potency loss. Administration in saline alone is not recommended (Prod Info norepinephrine bitartrate injection, 2005).
    b) DOSE
    1) ADULT: Dose range: 0.1 to 0.5 microgram/kilogram/minute (eg, 70 kg adult 7 to 35 mcg/min); titrate to maintain adequate blood pressure (Peberdy et al, 2010).
    2) CHILD: Dose range: 0.1 to 2 micrograms/kilogram/minute; titrate to maintain adequate blood pressure (Kleinman et al, 2010).
    3) CAUTION: Extravasation may cause local tissue ischemia, administration by central venous catheter is advised (Peberdy et al, 2010).
    M) 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).
    N) Treatment should include recommendations listed in the ORAL EXPOSURE section when appropriate.

Eye Exposure

    6.8.1) DECONTAMINATION
    A) EYE IRRIGATION, ROUTINE: Remove contact lenses and irrigate exposed eyes with copious amounts of room temperature 0.9% saline or water for at least 15 minutes. If irritation, pain, swelling, lacrimation, or photophobia persist after 15 minutes of irrigation, an ophthalmologic examination should be performed (Peate, 2007; Naradzay & Barish, 2006).
    6.8.2) TREATMENT
    A) OCULAR ABSORPTION
    1) There are no reports of systemic poisoning in humans exposed to cyanide by the ocular route; however, deaths in experimental animals have followed ocular exposure (Ballantyne, 1983).
    2) Patients exposed by this route should be observed for several hours in a controlled setting for the possible development of symptoms of systemic cyanide poisoning.
    B) SUPPORT
    1) Treatment should include recommendations listed in the INHALATION EXPOSURE section when appropriate.
    C) Treatment should include recommendations listed in the ORAL EXPOSURE section when appropriate.

Dermal Exposure

    6.9.1) DECONTAMINATION
    A) DECONTAMINATION: Remove contaminated clothing and wash exposed area thoroughly with soap and water for 10 to 15 minutes. A physician may need to examine the area if irritation or pain persists (Burgess et al, 1999).
    6.9.2) TREATMENT
    A) SKIN ABSORPTION
    1) Cyanide can be absorbed and cause systemic cyanide poisoning by the dermal route, usually only following severe burns from molten material (Bourrelier & Paulet, 1971) or total immersion in cyanide solution (Bismuth et al, 1984; Dodds & McKnight, 1985).
    B) OXYGEN
    1) Administer 100% oxygen to maintain an elevated PO2.
    a) Oxygen may reverse the cyanide-cytochrome oxidase complex and facilitate the conversion to thiocyanate following thiosulfate administration (Graham et al, 1977).
    b) There is fairly good evidence that 100% oxygen, combined with traditional nitrite/thiosulfate therapy is better than thiosulfate alone (Litovitz et al, 1983; Way et al, 1972).
    C) HYPERBARIC OXYGEN THERAPY
    1) Hyperbaric oxygen (HBO) therapy is approved for cyanide poisoning as a category one condition (approved for third party reimbursement and known effective as treatment) by the Undersea Medical Society (Myers & Schnitzer, 1984).
    2) Hyperbaric oxygen has been suggested to improve clinical outcome, especially in those patients with CNS toxicity not responding to more traditional therapy.
    3) Animal studies have both confirmed and refuted this (Skene et al, 1966; Way et al, 1972; Takano et al, 1980).
    4) Case reports suggest that HBO may be of value (Carden, 1970; Trapp, 1970). However, one patient treated with supportive therapy, antidotes, and HBO did not survive a serious poisoning (Litovitz et al, 1983).
    5) Hyperbaric oxygen should be reserved for those patients with significant symptoms (ie, coma, seizures) who do not respond to normal supportive and antidotal therapy, and for those patients poisoned by both cyanide and carbon monoxide secondary to smoke inhalation (Hart et al, 1985).
    D) FLUID/ELECTROLYTE BALANCE REGULATION
    1) Establish secure large bore IV line.
    E) CYANIDE ANTIDOTE
    1) SUMMARY
    a) A cyanide antidote, either hydroxocobalamin or the sodium nitrite/sodium thiosulfate kit, should be administered to patients with symptomatic poisoning. If cyanide toxicity develops concurrent with carbon monoxide poisoning (e.g., closed space fire), hydroxocobalamin is the preferred antidote. It that is not available, sodium thiosulfate may be used alone. Use of amyl nitrite or sodium nitrite will cause methemoglobinemia, further reducing oxygen carrying capacity.
    2) HYDROXOCOBALAMIN - CYANOKIT(R)
    a) ADULT DOSE: 5 g (two 2.5 g vials each reconstituted with 100 mL sterile 0.9% saline) administered as an intravenous infusion over 15 minutes. For severe poisoning, a second dose of 5 g may be infused intravenously over 15 minutes to 2 hours, depending on the patient's condition (Prod Info CYANOKIT(R) 2.5g IV injection, 2006).
    b) PEDIATRIC DOSE: A dose of 70 mg/kg has been used in pediatric patients, based on limited post-marketing experience outside the US (Prod Info CYANOKIT(R) 2.5g IV injection, 2006).
    c) INDICATIONS: Known or suspected cyanide poisoning.
    d) ADVERSE EFFECTS: Transient hypertension, allergic reactions (including anaphylaxis), nausea, headache, rash. Hydroxocobalamin's deep red color causes red-colored urine in all patients, and erythema of the skin in most (Prod Info CYANOKIT(R) 2.5g IV injection, 2006).
    e) LABORATORY INTERFERENCE: Because of its' color, hydroxocobalamin interferes with colorimetric determination of various laboratory parameters. It may artificially increase serum creatinine, bilirubin, triglycerides, cholesterol, total protein, glucose, albumin , alkaline phosphatase and hemoglobin. It may artificially decrease serum ALT and amylase. It may artificially increase urinary pH, glucose, protein, erythrocytes, leukocytes, ketones, bilirubin, urobilinogen, and nitrate (Prod Info CYANOKIT(R) 2.5g IV injection, 2006).
    f) HEMODIALYSIS INTERFERENCE: Dialysis machines have a spectrophotometric safety measure that can shut down after detecting blood leaking across the dialysis membrane. Hydroxocobalamin has a deep red color and can permeate the dialysis membrane, coloring the dialysate and causing the hemodialysis machine to shut down erroneously. In one case report, a patient with cyanide poisoning underwent dialysis after receiving 5 g of IV hydroxocobalamin because of refractory acidemia, reduced kidney function and hyperkalemia. A blood leak alarm caused the dialysis machine to shut down erroneously, delaying therapy, and resulting in the death of the patient (Stellpflug et al, 2013).
    g) In a controlled pilot study of male smokers, administration of 5 grams of intravenous 5% hydroxocobalamin significantly decreased whole blood cyanide levels (Forsyth et al, 1992).
    3) CYANIDE ANTIDOTE KIT
    a) OBTAIN AND PREPARE for administration a CYANIDE ANTIDOTE KIT, consisting of sodium nitrite and sodium thiosulfate.
    1) Antidotes should be used only in significantly symptomatic patients (ie, impaired consciousness, convulsions, acidosis, or unstable vital signs).
    2) Even when patients are rendered comatose by the inhalation of hydrogen cyanide gas, antidotes may not be necessary if the exposure is rapidly terminated, the patient has regained consciousness on arrival at the medical facility, and there is no acidosis or abnormality of the vital signs (Peden et al, 1986).
    b) SODIUM NITRITE
    1) INDICATION
    a) Sodium nitrite should be given initially and administered as soon as vascular access is established.
    b) Further administration of sodium nitrite is dictated only by the clinical situation, provided no significant complications (hypotension, excessive methemoglobinemia) are present. Use with caution if carbon monoxide poisoning is also suspected.
    c) The goal of nitrite therapy is to achieve a methemoglobin level of 20% to 30%. This level is not based on clinical data, but represents the tolerated concentration without significant adverse symptoms from methemoglobin in an otherwise healthy individual. Clinical response has been reported to occur with methemoglobin levels in the range of 3.6% to 9.2% (DiNapoli et al, 1989; Johnson et al, 1989; Johnson & Mellors, 1988).
    2) ADULT DOSE
    a) 10 mL of a 3% solution (300 mg) administered intravenously at a rate of 2.5 to 5 mL/minute (Prod Info NITHIODOTE intravenous injection solution, 2011). Frequent blood pressure monitoring must accompany sodium nitrite injection and the rate slowed if hypotension occurs.
    b) If there is inadequate clinical response, an additional dose of sodium nitrite at half the amount of the initial dose may be administered 30 minutes following the first dose (Prod Info NITHIODOTE intravenous injection solution, 2011).
    3) PEDIATRIC DOSE
    a) The recommended pediatric sodium nitrite dose is 0.2 mL/kg of a 3% solution (6 mg/kg) administered intravenously at a rate of 2.5 to 5 mL/minute, not to exceed 10 mL (300 mg) (Prod Info NITHIODOTE intravenous injection solution, 2011).
    b) If there is inadequate clinical response, an additional dose of sodium nitrite at half the amount of the initial dose may be administered 30 minutes following the first dose (Prod Info NITHIODOTE intravenous injection solution, 2011; Berlin, 1970a).
    c) PRESENCE OF ANEMIA: If there is a reason to suspect the presence of anemia, the following initial sodium nitrite doses should be given, depending on the child's hemoglobin (sodium nitrite should not exceed the doses listed below; fatal methemoglobinemia may result) (Berlin, 1970a):
    1) Hemoglobin: 8 g/dL - Initial 3% sodium nitrite dose: 0.22 mL/kg (6.6 mg/kg)
    2) Hemoglobin: 10 g/dL - Initial 3% sodium nitrite dose: 0.27 mL/kg (8.7 mg/kg)
    3) Hemoglobin: 12 g/dL (average child) - Initial 3% sodium nitrite dose: 0.33 mL/kg (10 mg/kg)
    4) Hemoglobin: 14 g/dL - Initial 3% sodium nitrite dose: 0.39 mL/kg (11.6 mg/kg)
    4) It is highly recommended that total hemoglobin and methemoglobin concentrations be rapidly measured (30 minutes after dose), when possible, before repeating a dose of sodium nitrite to be sure that dangerous methemoglobinemia will not occur, especially in the pediatric patient.
    5) Monitor blood pressure frequently and treat hypotension by slowing infusion rate and giving crystalloids and vasopressors. Consider possible excessive methemoglobin formation if patient deteriorates during therapy.
    6) Excessive methemoglobinemia and hypotension are potential complications of nitrite therapy.
    7) In individuals with G6PD deficiency, therapy with methemoglobin-inducing agents is contraindicated because of the likelihood of serious hemolysis.
    c) SODIUM THIOSULFATE
    1) Sodium thiosulfate is the second component of the cyanide antidote kit. It is supplied as 50 mL of a 25% solution and it is administered intravenously. There are no adverse reactions to thiosulfate itself. The pediatric dose is adjusted for weight and not hemoglobin concentration.
    2) Sodium thiosulfate supplies sulfur for the rhodanese reaction, and is recommended after sodium nitrite, hydroxocobalamin, or 4-DMAP (4-dimethylaminophenol) administration (Marrs, 1988; Hall & Rumack, 1987).
    3) DOSE
    a) Follow sodium nitrite with IV sodium thiosulfate. ADULT: Administer 50 mL (12.5 g) of a 25% solution IV; PEDIATRIC: 1 mL/kg of a 25% solution (250 mg/kg), not to exceed 50 mL (12.5 g) total dose (Prod Info NITHIODOTE intravenous injection solution, 2011).
    b) A second dose, one-half of the first dose, may be administered if signs of cyanide toxicity reappear (Prod Info NITHIODOTE intravenous injection solution, 2011).
    c) Sodium thiosulfate is usually used in combination with sodium nitrite but may be used alone (Prod Info sodium thiosulfate IV injection, 2003).
    d) Sodium thiosulfate can be administered without sodium nitrite in patients at risk to develop further methemoglobinemia (ie excessive methemoglobinemia or hypotension after initial sodium nitrite administration or in the presence of methemoglobinemia or carboxyhemoglobin in patients with smoke inhalation due to fire). Sodium thiosulfate can also be used in combination with hydroxocobalamin to treat cyanide poisoning (Howland, 2011)
    e) CONTINUOUS INFUSION: It has been suggested that a continuous infusion of sodium thiosulfate be given after the initial bolus to maintain high thiosulfate levels. Low sodium intravenous fluids are required to avoid sodium overload. If large amounts of sodium thiosulfate are required, hemodialysis may be necessary to maintain a physiologic serum sodium level (Turchen et al, 1991).
    f) ADVERSE EVENTS: Sodium thiosulfate does not usually produce significant toxicity. Possible adverse events include hypotension, headache, nausea, vomiting, disorientation, and prolonged bleeding time (Prod Info NITHIODOTE intravenous injection solution, 2011).
    F) MONITORING OF PATIENT
    1) Obtain blood for arterial blood gases with venous pO2 or measured venous %O2 saturation, electrolytes, serum lactate, and whole blood cyanide levels.
    2) The following set of laboratory values suggest poisoning with an agent that inhibits oxidative phosphorylation (ie, cyanide, hydrogen sulfide) (Hall & Rumack, 1986).
    a) Cyanide and hydrogen sulfide poisoning are treated in essentially the same manner, (See Hydrogen Sulfide Management) with the exception of lack of efficacy of sodium thiosulfate in hydrogen sulfide poisoning. Administering sodium thiosulfate will most likely do no harm to a hydrogen sulfide poisoned patient.
    b) Arterial pO2: Usually normal until the stage of apnea. Usually remains normal until terminal stages of the poisoning if supplemental oxygen and assisted ventilation are provided.
    c) Serum electrolytes: Elevated anion gap (Na - (C1 + CO2)) (normals 12 to 16 milliequivalents/liter (12 to 16 millimoles/liter) or less) is present from the presence of unmeasured organic anions (usually lactate).
    d) Serum Lactate: Elevated (normals 0.6 to 1.8 milliequivalents/liter) (0.6 to 1.8 millimoles/liter) due to anaerobic metabolism with excessive production of lactic acid.
    e) Arterio-Central Venous Measured %O2 Saturation Difference: Due to cellular inability to extract and use oxygen, more is present on the venous side. The MEASURED values of arterial and central venous %O2 saturation approach each other with MEASURED central venous %O2 saturation greater than 70%.
    1) Arteriolization of venous blood gases (elevated venous pO2 or measured venous %O2 saturation) may serve as an early clue in the diagnosis of cyanide poisoning (Hall & Rumack, 1986; Johnson & Mellors, 1988).
    G) ACIDOSIS
    1) METABOLIC ACIDOSIS: Treat severe metabolic acidosis (pH less than 7.1) with sodium bicarbonate, 1 to 2 mEq/kg is a reasonable starting dose(Kraut & Madias, 2010). Monitor serum electrolytes and arterial or venous blood gases to guide further therapy.
    H) SEIZURE
    1) SUMMARY
    a) Attempt initial control with a benzodiazepine (eg, diazepam, lorazepam). If seizures persist or recur, administer phenobarbital or propofol.
    b) Monitor for respiratory depression, hypotension, and dysrhythmias. Endotracheal intubation should be performed in patients with persistent seizures.
    c) Evaluate for hypoxia, electrolyte disturbances, and hypoglycemia (or, if immediate bedside glucose testing is not available, treat with intravenous dextrose).
    2) DIAZEPAM
    a) ADULT DOSE: Initially 5 to 10 mg IV, OR 0.15 mg/kg IV up to 10 mg per dose up to a rate of 5 mg/minute; may be repeated every 5 to 20 minutes as needed (Brophy et al, 2012; Prod Info diazepam IM, IV injection, 2008; Manno, 2003).
    b) PEDIATRIC DOSE: 0.1 to 0.5 mg/kg IV over 2 to 5 minutes; up to a maximum of 10 mg/dose. May repeat dose every 5 to 10 minutes as needed (Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008).
    c) Monitor for hypotension, respiratory depression, and the need for endotracheal intubation. Consider a second agent if seizures persist or recur after repeated doses of diazepam .
    3) NO INTRAVENOUS ACCESS
    a) DIAZEPAM may be given rectally or intramuscularly (Manno, 2003). RECTAL DOSE: CHILD: Greater than 12 years: 0.2 mg/kg; 6 to 11 years: 0.3 mg/kg; 2 to 5 years: 0.5 mg/kg (Brophy et al, 2012).
    b) MIDAZOLAM has been used intramuscularly and intranasally, particularly in children when intravenous access has not been established. ADULT DOSE: 0.2 mg/kg IM, up to a maximum dose of 10 mg (Brophy et al, 2012). PEDIATRIC DOSE: INTRAMUSCULAR: 0.2 mg/kg IM, up to a maximum dose of 7 mg (Chamberlain et al, 1997) OR 10 mg IM (weight greater than 40 kg); 5 mg IM (weight 13 to 40 kg); INTRANASAL: 0.2 to 0.5 mg/kg up to a maximum of 10 mg/dose (Loddenkemper & Goodkin, 2011; Brophy et al, 2012). BUCCAL midazolam, 10 mg, has been used in adolescents and older children (5-years-old or more) to control seizures when intravenous access was not established (Scott et al, 1999).
    4) LORAZEPAM
    a) MAXIMUM RATE: The rate of intravenous administration of lorazepam should not exceed 2 mg/min (Brophy et al, 2012; Prod Info lorazepam IM, IV injection, 2008).
    b) ADULT DOSE: 2 to 4 mg IV initially; repeat every 5 to 10 minutes as needed, if seizures persist (Manno, 2003; Brophy et al, 2012).
    c) PEDIATRIC DOSE: 0.05 to 0.1 mg/kg IV over 2 to 5 minutes, up to a maximum of 4 mg/dose; may repeat in 5 to 15 minutes as needed, if seizures continue (Brophy et al, 2012; Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008; Sreenath et al, 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).
    I) METHEMOGLOBINEMIA
    1) While clinically significant excessive methemoglobinemia has occurred following sodium nitrite therapy for cyanide poisoning, such instances are rare and usually occur in children receiving excessive nitrite doses.
    a) Inducing a "therapeutic methemoglobin level" of 25% is unnecessary to insure satisfactory clinical outcome (Johnson et al, 1989).
    2) If excessive methemoglobinemia occurs, some authors have suggested that methylene blue should not be used because it could cause release of cyanide from the cyanmethemoglobin complex. Such authors have suggested that emergency exchange transfusion is the treatment of choice (Berlin, 1970). Hyperbaric oxygen therapy could be used to support the patient while preparations for exchange transfusion are being made.
    3) However, methylene or toluidine blue have been used successfully in this setting without worsening the course of the cyanide poisoning (van Heijst et al, 1987). There is some controversy over whether or not the induction of methemoglobinemia is the sodium nitrite mechanism of action in cyanide poisoning. As long as intensive care monitoring and further antidote doses (if required) are available, methylene blue can most likely be safely administered in this setting.
    4) SUMMARY
    a) Determine the methemoglobin concentration and evaluate the patient for clinical effects of methemoglobinemia (ie, dyspnea, headache, fatigue, CNS depression, tachycardia, metabolic acidosis). Treat patients with symptomatic methemoglobinemia with methylene blue (this usually occurs at methemoglobin concentrations above 20% to 30%, but may occur at lower methemoglobin concentrations in patients with anemia, or underlying pulmonary or cardiovascular disorders). Administer oxygen while preparing for methylene blue therapy.
    5) METHYLENE BLUE
    a) INITIAL DOSE/ADULT OR CHILD: 1 mg/kg IV over 5 to 30 minutes; a repeat dose of up to 1 mg/kg may be given 1 hour after the first dose if methemoglobin levels remain greater than 30% or if signs and symptoms persist. NOTE: Methylene blue is available as follows: 50 mg/10 mL (5 mg/mL or 0.5% solution) single-dose ampules (Prod Info PROVAYBLUE(TM) intravenous injection, 2016) and 10 mg/1 mL (1% solution) vials (Prod Info methylene blue 1% intravenous injection, 2011). REPEAT DOSES: Additional doses may be required, especially for substances with prolonged absorption, slow elimination, or those that form metabolites that produce methemoglobin. NOTE: Large doses of methylene blue may cause methemoglobinemia or hemolysis (Howland, 2006). Improvement is usually noted shortly after administration if diagnosis is correct. Consider other diagnoses or treatment options if no improvement has been observed after several doses. If intravenous access cannot be established, methylene blue may also be given by intraosseous infusion. Methylene blue should not be given by subcutaneous or intrathecal injection (Prod Info methylene blue 1% intravenous injection, 2011; Herman et al, 1999). NEONATES: DOSE: 0.3 to 1 mg/kg (Hjelt et al, 1995).
    b) CONTRAINDICATIONS: G-6-PD deficiency (methylene blue may cause hemolysis), known hypersensitivity to methylene blue, methemoglobin reductase deficiency (Shepherd & Keyes, 2004)
    c) FAILURE: Failure of methylene blue therapy suggests: inadequate dose of methylene blue, inadequate decontamination, NADPH dependent methemoglobin reductase deficiency, hemoglobin M disease, sulfhemoglobinemia, or G-6-PD deficiency. Methylene blue is reduced by methemoglobin reductase and nicotinamide adenosine dinucleotide phosphate (NADPH) to leukomethylene blue. This in turn reduces methemoglobin. Red blood cells of patients with G-6-PD deficiency do not produce enough NADPH to convert methylene blue to leukomethylene blue (do Nascimento et al, 2008).
    d) DRUG INTERACTION: Concomitant use of methylene blue with serotonergic drugs, including serotonin reuptake inhibitors (SRIs), selective serotonin reuptake inhibitors (SSRIs), serotonin and norepinephrine reuptake inhibitors (SNRIs), tricyclic antidepressants (TCAs), norepinephrine-dopamine reuptake inhibitors (NDRIs), triptans, and ergot alkaloids may increase the risk of potentially fatal serotonin syndrome (U.S. Food and Drug Administration, 2011; Stanford et al, 2010; Prod Info methylene blue 1% IV injection, 2011).
    6) TOLUIDINE BLUE OR TOLONIUM CHLORIDE (GERMANY)
    a) DOSE: 2 to 4 mg/kg intravenously over 5 minutes. Dose may be repeated in 30 minutes (Nemec, 2011; Lindenmann et al, 2006; Kiese et al, 1972).
    b) SIDE EFFECTS: Hypotension with rapid intravenous administration. Vomiting, diarrhea, excessive sweating, hypotension, dysrhythmias, hemolysis, agranulocytosis and acute renal insufficiency after overdose (Dunipace et al, 1992; Hix & Wilson, 1987; Winek et al, 1969; Teunis et al, 1970; Marquez & Todd, 1959).
    c) CONTRAINDICATIONS: G-6-PD deficiency; may cause hemolysis.
    J) DICOBALT EDETATE
    1) Kelocyanor(R) (dicobalt-EDTA) is a highly effective cyanide chelating agent currently used clinically in Europe, Israel, and Australia (Davison, 1969; Hillman et al, 1974). It is not available in the US.
    2) PRECAUTIONS
    a) Significant toxicity from the antidote (severe hypertension or hypotension, cardiac ischemia or arrhythmias) may be seen in patients incorrectly diagnosed as being poisoned with cyanide and administered this antidote (Pronczuk de Garbino & Bismuth, 1981; Tyrer, 1981). Therefore, KELOCYANOR(R) SHOULD NOT BE USED in CASES of MILD CYANIDE POISONING or DIAGNOSTIC UNCERTAINTY (Peden et al, 1986; Tyrer, 1981).
    b) Severe anaphylactoid reactions with periorbital and massive facial edema and airway compromise may also occur (Dodds & McKnight, 1985; Wright & Vesey, 1986).
    c) ADVERSE EFFECTS can include nausea, vomiting, tachycardia, hypotension, hypertension, anaphylactic reactions, facial and neck edema, chest pain, diaphoresis, nervousness, tremulousness, gastrointestinal hemorrhages, convulsions, cardiac irregularities, and rashes (Prod Info, 1986) (Prod Info, 1987) (Davison, 1969; Tyrer, 1981; Hillman et al, 1974).
    3) DOSE
    a) ADULTS: One to two 20 mL ampules (300 to 600 mg) injected intravenously over about 1 to 5 minutes (Prod Info, 1978) (Prod Info, 1986) (Prod Info, 1987) (Davison, 1969).
    1) A third 20 mL ampule (300 mg) can be injected intravenously over about 1 to 5 minutes, 5 minutes after the first 1 to 2 ampules if there is not sufficient clinical improvement (Prod Info, 1978) (Prod Info, 1986) (Prod Info, 1987) (Davison, 1969).
    2) Manufacturers recommend following the Kelocyanor(R) injection with intravenous injection of 50 milliliters of 50% dextrose in water (Prod Info, 1978) (Prod Info, 1986) (Prod Info, 1987).
    3) Kelocyanor(R) can be used with other standard cyanide antidotes (Prod Info, 1978).
    b) CHILDREN: Pediatric doses have not been established by manufacturers. A suggested dose used in Israel for children is 0.5 milliliter per kilogram (not to exceed 20 mL) (Personal Communication, Uri Taitelman, MD, 1963).
    4) Kelocyanor(R) is supplied in 20 mL ampules containing 300 mg of dicobalt-EDTA and 4 grams of dextrose in water for injection (Prod Info, 1978) (Prod Info, 1986) (Prod Info, 1987).
    K) 4-DIMETHYLAMINOPHENOL HYDROCHLORIDE
    1) 4-DMAP is a methemoglobin inducing agent used in some European countries for the treatment of acute cyanide poisoning. Excessive methemoglobinemia may be a major complication following the use of this agent (van Dijk et al, 1986).
    L) EXPERIMENTAL THERAPY
    1) STROMA-FREE METHEMOGLOBIN SOLUTION: Stroma-free methemoglobin solution prepared from outdated human red blood cells by oxidation of the ferrous iron of hemoglobin to the ferric form in vitro has been studied in experimental animals and shows promise as a cyanide antidote (Ten Eyck et al, 1985).
    a) By virtue of its in vitro production, stroma-free methemoglobin solution has the advantage of sparing the oxygen-carrying capacity of the blood (Marrs, 1988).
    b) It has not been studied in human poisoning cases and is not available for human administration.
    2) ALPHA-KETOGLUTARIC ACID
    a) Alpha-ketoglutaric acid is also being tested as a replacement for sodium nitrite in combination with sodium thiosulfate in animal models where it has been efficacious in experimental cyanide poisoning (Bhattacharya & Vijayaraghavan, 2002; Moore et al, 1986).
    b) In vitro and animal studies show that alpha-ketaglutaric acid binds with cyanide and thereby antagonizes cyanide-induced inhibition of brain cytochrome oxidase (Norris et al, 1990).
    c) Alpha-ketoglutaric acid administered with sodium thiosulfate abolished the cyanide-induced decrease in brain gamma-aminobutyric acid in mice (Yamamoto, 1990).
    d) It has not been studied in human poisoning cases and is not available for human administration.
    3) CHLORPROMAZINE
    a) Chlorpromazine has been studied in various animal models as a possible cyanide antidote. Conflicting reports of efficacy have been published (Pettersen & Cohen, 1986).
    b) Animal and in vitro studies show that chlorpromazine can decrease peroxidation of lipid membranes and prevent cyanide-induced calcium influx believed to be responsible for neurotoxicity (Johnson et al, 1986; Maduh et al, 1988).
    c) It has not been studied in human poisoning cases.
    4) OTHER INVESTIGATIONAL ANTIDOTES
    a) Animal studies to identify alternate cyanide antidotes have tested phenoxybenzamine, centrophenoxine, naloxone hydrochloride, etomidate, para-aminopropiophenone, calcium-ion-channel blockers, and prodrugs of 3-mercaptopyruvate (Amery et al, 1981; Ashton et al, 1980; Bright & Marrs, 1987; Budavari, 1976; Dubinsky et al, 1984; Johnson et al, 1986; Leung et al, 1984; Bright & Marrs, 1987; Marrs, 1988) (Rump & Edelwijn, 1986) (Vick & Froehlich, 1985; Nagasawa et al, 2007).
    M) HYPOTENSIVE EPISODE
    1) SUMMARY
    a) Infuse 10 to 20 milliliters/kilogram of isotonic fluid and keep the patient supine. If hypotension persists, administer dopamine or norepinephrine. Consider central venous pressure monitoring to guide further fluid therapy.
    N) 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).
    O) Treatment should include recommendations listed in the ORAL EXPOSURE section when appropriate.

Enhanced Elimination

    A) HEMODIALYSIS
    1) Hemodialysis as adjunct treatment to supportive care, intravenous sodium nitrite, and sodium thiosulfate has been reported in the management of a patient with cyanide toxicity (Wesson et al, 1985).
    2) It has been suggested that hemodialysis may be helpful in the following ways:
    1) Removal of the small extracellular reservoir of cyanide, particularly if cyanide is still being absorbed from the gastrointestinal tract.
    2) Correction of severe metabolic acidosis caused by cyanide poisoning.
    3) Removal of thiocyanate which should decrease both tissue and plasma cyanide levels.
    3) Limited animal studies have shown some potential effectiveness of hemodialysis when combined with thiosulfate infusion (Wesson et al, 1985; Gonzales & Sabatini, 1989).
    4) Hemodialysis may be considered after supportive care and pharmacologic therapy have been maximized.
    5) HEMODIALYSIS INTERFERENCE: Dialysis machines have a spectrophotometric safety measure that can shut down after detecting blood leaking across the dialysis membrane. Hydroxocobalamin has a deep red color and can permeate the dialysis membrane, coloring the dialysate and causing the hemodialysis machine to shut down erroneously. In one case report, a patient with cyanide poisoning underwent dialysis after receiving 5 g of IV hydroxocobalamin because of refractory acidemia, reduced kidney function and hyperkalemia. A blood leak alarm caused the dialysis machine to shut down erroneously, delaying therapy, and resulting in the death of the patient (Stellpflug et al, 2013).
    B) HEMOPERFUSION
    1) Charcoal hemoperfusion as adjunct treatment to supportive care, intravenous sodium nitrite, and sodium thiosulfate has been reported in the management of a patient with cyanide toxicity (Kreig & Saxena, 1987).
    2) The outcome in this case was no different from that of other patients treated similarly without hemoperfusion.
    3) Hemoperfusion cannot be considered routine or standard adjunct therapy for cyanide poisoning at this time.

Case Reports

    A) ROUTE OF EXPOSURE
    1) A 23-year-old woman was admitted to the hospital after ingesting a cyanide-contaminated Sudafed 12 Hour capsule. She experienced sudden onset of seizures, became comatose, and was intubated. Physical exam revealed Doll's eyes. She developed metabolic acidosis, hyperglycemia, and severe hypotension. Treatment consisted of IV fluids, dopamine, Levophed, insulin, and 750 mEq sodium bicarbonate. Ten hours later she regained consciousness. Blood cyanide level at 75 minutes was 6.14 mg/L. No antidotal therapy was administered, but the patient survived with supportive therapy, arterial blood gas monitoring, and sodium bicarbonate administration (Woo & Pouw, 1993).
    B) SPECIFIC AGENT
    1) CASE SERIES: Two workers, aged 28 years and 34 years, developed cyanide toxicity after inhalation exposure to propionitrile. Measured cyanide level in the first patient was 5.0 mcg/mL; the patient became comatose, needed cardiopulmonary resuscitation, and developed seizures. The other patient had a blood cyanide level of 3.5 mcg/mL, and developed nausea, headache, dizziness, and confusion. Both patients recovered following supportive therapy, treatment with cyanide antidote kit, and in the first patient, treatment with hyperbaric oxygen (Scolnick et al, 1993).

Summary

    A) TOXIC DOSE: The threshold value limit for hydrogen cyanide is 10 parts per million (ppm) for an 8 hour day. A dose of potassium cyanide of 200 mg is likely lethal; however, an adult survived an ingestion of 1200 to 1500 mg potassium cyanide powder without visual or neurologic sequelae following cyanide antidotal therapy with hydroxocobalamin immediately followed by sodium thiosulfate. An immediate inhalational exposure of 270 ppm of hydrogen cyanide gas is likely lethal.

Minimum Lethal Exposure

    A) ROUTE OF EXPOSURE
    1) The fatal dose of cyanide salts is estimated at 200 to 300 mg for an adult, and 50 to 100 mg of hydrocyanic acid (Bonnichsen & Maehly, 1966) (Ansell & Lewis, 1970) (Baselt & Cravey, 1989). Specifically for potassium or sodium cyanide, the minimum lethal dose has been estimated to be about 3 milligrams/kilogram (van Heijst et al, 1987).
    2) Air concentrations of 0.2 to 0.3 milligram/cubic meter (200 to 300 parts per million) are rapidly fatal (ACGIH, 1986).
    3) Human responses to hydrogen cyanide were identified by ACGIH (1986):
    EffectAirborne Concentration (ppm)
    Immediately fatal270
    Fatal after 10 minutes181
    Fatal after 30 minutes135
    Fatal after 0.5 to 1 hour later, or dangerous to life110-135
    Tolerated for 0.5 to 1 hour without immediate or late effects45-54
    Slight symptoms after several hours18-36

    4) SPECIFIC AGENT
    a) POTASSIUM CYANIDE: A 17-year-old boy developed severe metabolic acidosis, hypotension, and bradycardia after unintentionally ingesting a beverage containing 1.5 g potassium cyanide. Despite administration of antidote therapy and supportive care, the patient continued to show hemodynamic instability. A MRI revealed complete cerebellar infarction, and the patient was declared brain dead approximately 60 hours post-ingestion (Peddy et al, 2006).
    5) LDLo/LCLo:
    a) CYANOGEN
    1) LDLo (UNK) DOG: 15 mg/kg (Sax & Lewis, 1989)
    2) LDLo (Subcutaneous) RABBIT: 13 mg/kg (Sax & Lewis, 1989)
    b) CYANOGEN BROMIDE
    1) LCLo (INHL) HUMAN: 92 ppm/10 minutes (Sax & Lewis, 1989)
    2) LCLo (INHL) MOUSE: 500 mg/m(3)/10 minutes (Sax & Lewis, 1989)
    c) CYANOGEN CHLORIDE
    1) LDLo (Subcutaneous) MOUSE: 39 mg/kg (Sax & Lewis, 1989)
    2) LDLo (Subcutaneous) DOG: 5 mg/kg (Sax & Lewis, 1989)
    3) LCLo (INHL) DOG: 79 ppm/8 hours (ITI, 1988)
    4) LDLo (Subcutaneous) RABBIT: 20 mg/kg (Sax & Lewis, 1989)
    5) LCLo (INHL) RABBIT: 207 ppm/30 minutes (Sax & Lewis, 1989)
    d) CYANOGEN IODIDE
    1) LDLo (ORAL) CAT: 18 mg/kg (Sax & Lewis, 1989)
    2) LDLo (Subcutaneous) CAT: 20 mg/kg (Sax & Lewis, 1989)
    3) LDLo (Subcutaneous) DOG: 19 mg/kg (Sax & Lewis, 1989)
    4) LDLo (Subcutaneous) RAT: 44 mg/kg (Sax & Lewis, 1989)
    e) HYDROGEN CYANIDE
    1) LDLo (ORAL) HUMAN: 5.7 mg/kg (Sax & Lewis, 1989)
    2) LCLo (INHL) HUMAN: 200 ppm/5 minutes (Sax & Lewis, 1989)
    3) LCLo (INHL) HUMAN: 120 mg/m(3)/1 hour (Sax & Lewis, 1989)
    4) LCLo (INHL) HUMAN: 200 mg/m(3)/10 minutes (Sax & Lewis, 1989)
    5) LCLo (INHL) MAN: 400 mg/m(3)/2 minutes (Sax & Lewis, 1989)
    6) LCLo (INHL) HUMAN: 5,000 mg/m(3) (ITI, 1988)
    7) LDLo (Subcutaneous) HUMAN: 1 mg/kg (Sax & Lewis, 1989)
    8) LDLo (UNK) MAN: 14.71 mg/kg (Sax & Lewis, 1989)
    9) LDLo (ORAL) DOG: 4 mg/kg (Sax & Lewis, 1989)
    10) LDLo (Subcutaneous) CAT: 11 mg/kg (Sax & Lewis, 1989)
    11) LDLo (ORAL) RABBIT: 4 mg/kg (Sax & Lewis, 1989)
    f) POTASSIUM CYANIDE
    1) LDLo (ORAL) HUMAN: 2,857 mcg/kg (RTECS, 1990)
    2) LDLo (IM) RAT: 8 mg/kg (RTECS, 1990)
    3) LDLo (IP) GUINEA PIG: 8 mg/kg (RTECS, 1990)
    4) LDLo (Subcutaneous) GUINEA PIG: 8 mg/kg (RTECS, 1990)
    5) LDLo (IV) GUINEA PIG: 5 mg/kg (RTECS, 1990)
    6) LDLo (INTRAARTERIAL) GUINEA PIG: 5 mg/kg (RTECS, 1990)
    7) LDLo (IV) DOG: 5 mg/kg (RTECS, 1990)
    8) LDLo (Subcutaneous) FROG: 60 mg/kg (RTECS, 1990)
    g) SODIUM CYANIDE
    1) LDLo (ORAL) MAN: 6,557 mcg/kg (Sax & Lewis, 1989)
    2) LDLo (ORAL) HUMAN: 2,857 mcg/kg (Sax & Lewis, 1989)
    3) LDLo (UNK) MAN: 2,206 mcg/kg (Sax & Lewis, 1989)
    4) LDLo (Subcutaneous) DOG: 6 mg/kg (Sax & Lewis, 1989)
    5) LDLo (Subcutaneous) RABBIT: 2,200 mcg/kg (Sax & Lewis, 1989)
    6) ORGAN TRANSPLANTATION: Transplantations of corneas, kidneys, and a pancreas have been performed successfully from cyanide-poisoned donors without evidence of adverse effects at follow-up (Lindquist et al, 1988; Hantson et al, 1991; Hantson et al, 1999; Ravishankar et al, 1998).

Maximum Tolerated Exposure

    A) INHALATION
    1) Patients have survived exposure to air concentrations of 500 milligrams/cubic meter of HYDROGEN CYANIDE (Bonsall, 1984), ingestions of one gram of POTASSIUM CYANIDE or more (Yacoub et al, 1974; Hall & Rumack, 1987; Valenzuela et al, 1992), and complete immersion in solutions of CYANIDE SALTS (Bismuth et al, 1984; Dodds & McKnight, 1985).
    2) 0.05 to 0.06 milligram/liter or 45 to 54 parts per million of HYDROGEN CYANIDE have been tolerated for 0.5 to 1 hour without immediate or late effects (ACGIH, 1986). However, slight symptoms develop after several hours of exposure to 0.02 to 0.04 milligram/liter or 18 to 36 parts per million (ACGIH, 1986).
    3) The lowest irritant concentration of CYANOGEN CHLORIDE that could be tolerated by man for 10 minutes is 1 part per million (0.0025 milligram/liter) (ACGIH, 1986). Two parts per million is intolerable for 10 minutes and 48 parts per million is fatal in 30 minutes (ACGIH, 1986).
    B) INGESTION
    1) CASE REPORT/POTASSIUM DICYANOAURATE: A 29-year-old man presented with coma (Glasgow Coma Scale score of 3), tachypnea (35 to 45 breaths/minute), tachycardia (95 bpm), and hypotension (80/60 mmHg). Whole blood cyanide concentration was 6.9 mg/L and blood alcohol concentration was 270 mg/dL. Further questioning of the patient determined that he had intentionally ingested approximately 50 grams potassium dicyanoaurate (containing cyanide 25 mg/g; total amount of cyanide ingested 1250 mg [a 5-fold lethal dose of cyanide]). Following antidotal therapy with sodium thiosulfate and supportive care, the patient recovered uneventfully and was discharged to a psychiatric unit 2 days post-ingestion (Kampe et al, 2000). The authors postulate that self-detoxification may have played a role in the patient's recovery. Concomitant ingestion of alcohol may have neutralized the hydrochloric acid in the stomach, resulting in decreased uptake of cyanide by the stomach.
    2) CASE REPORT/POTASSIUM CYANIDE: A 58-year-old man intentionally ingested 1200 to 1500 mg of pure potassium cyanide powder and subsequently lost consciousness for 1 to 1.5 minutes and had one episode of generalized seizures. At presentation to the hospital, the patient was severely somnolent with mydriasis and lactic acidosis. The patient required emergent intubation, analgosedation and ventilation. Cyanide antidotal treatment was initiated 1.5 hours post-ingestion with hydroxocobalamin followed sodium thiosulfate. Following antidotal therapy, the patient's arterial blood gases normalized on the day of admission. On hospital day 3, he was extubated, and the next day was discharged without visual or neurologic sequelae, which continued at his 5-month follow-up appointment (Zakharov et al, 2015).
    3) CASE REPORT/POTASSIUM HEXACYANOFERATE: A 38-year-old woman attempted suicide by ingesting one coffee-spoonful of potassium hexacyanoferate. Hydroxocobalamin 5 g was infused as antidotal therapy. The patient's clinical course was uneventful. The patient developed a mild methemoglobinemia but serum cyanide concentrations never rose above toxic levels (Hantson et al, 1994).

Serum Plasma Blood Concentrations

    7.5.2) TOXIC CONCENTRATIONS
    A) TOXIC CONCENTRATION LEVELS
    1) CONCENTRATION LEVEL
    a) Patients treated with only supportive measures have survived severe poisoning with whole blood cyanide levels of 2.3 to 4.65 micrograms/milliliter (Vogel et al, 1981; Saincher et al, 1992).
    b) Patients treated with specific antidotes have survived severe poisoning with whole blood cyanide levels of 3.85 to 40 micrograms/milliliter (1,538 micromoles/liter) (Litovitz et al, 1983; Hall & Rumack, 1986; Hall & Rumack, 1986) 1987; (Feihl et al, 1982).
    c) Significant symptoms of poisoning generally occur with whole blood cyanide levels of 1 microgram/milliliter (38.5 micromoles/liter) or greater (Hall & Rumack, 1986).
    d) Blood cyanide levels and associated symptoms (Graham et al, 1977):
    1) No symptoms: Less than 0.2 mg/L (mcg/mL) (0.02 mg%; SI = 7.7 mcmol/L)
    2) Flushing and tachycardia: 0.5-1.0 mg/L (mcg/mL) (0.05-0.1 mg%; SI = 19.2 to 38.5 mcmol/L)
    3) Obtundation: 1.0-2.5 mg/L (mcg/mL) (0.1-0.25 mg%; SI = 38.5 to 96.1 mcmol/L)
    4) Coma and respiratory depression: Greater than 2.5 mg/L (mcg/mL) (0.25 mg%; SI = 96.1 mcmol/L)
    5) Death: Greater than 3 mg/L (mcg/mL) (0.3 mg%; SI = 115.4 mcmol/L)
    e) Blood cyanide levels after chronic exposure to cyanide fumes and cyanide aerosols (Chandra et al, 1980):
    WORKERS (n=23)CYANIDE mcg/100 mL
    RANGEMEAN
    Smokers (n=8)10 to 22056
    Nonsmokers (n=15)2 to 3618.3
    CONTROLS (n=20) .
    Smokers (n=10)0 TO 9.44.8
    Nonsmokers (n=10)0 TO 8.63.2

    1) Blood cyanide levels associated with smoking (Clark et al, 1981):
    a) Smokers: Up to 0.5 mg/L (mcg/mL)
    Smokers:Up to 0.5 mg/L (mcg/mL)
    .(SI = 19.2 mcmol/L)

    f) Post mortem blood cyanide levels after ingestion -
    1) Gill et al (2004) reported 17 cases -
    AVERAGE LEVELRANGE
    37.6 mg/L (mcg/mL)0.6 to 185.2 mg/L

    a) (Gill et al, 2004)
    2) Ballantyne et al (1974) reported 34 cases -
    AVERAGE LEVELRANGE
    12.4 mg/L (mcg/mL)1.1 to 53.1 mg/L (mcg/mL)
    (1.2 mg%)(0.1 to 5.3 mg%)
    (SI = 476.9 mcmol/L)(SI = 42.3 to 2042 mcmol/L)

    3) Rehlung (1967) reported 32 cases -
    AVERAGE LEVELRANGE
    36.5 mg/L (mcg/mL)0.4 to 230 mg/L (mcg/mL)
    (3.7 mg%)(0.04 to 23 mg%)
    (SI = 1403.8 mcmol/L)(SI = 15.4 to 8846 mcmol/L)

    g) Blood cyanide levels in smoke inhalation victims:
    a. (Hart et al, 1985)
    Patient 13.9 mcg/mL(expired)
    Patient 21.8 mcg/mL(survived)
    Patient 30.35 mcg/mL(survived)
    Patient 40.42 mcg/mL(survived)
    Patient 51.65 mcg/mL(survived)
    b. (Jones et al, 1987)
    Case 11.8 mcg/mL(expired)
    Case 22.4 mcg/mL(expired)
    Case 31.4 mcg/mL(expired)
    Case 41.5 mcg/mL(expired)

    1) In an analysis of 364 cases of cyanide poisoning in fire related fatalities, postmortem blood cyanide levels averaged 1.0 mg/L, with 31 victims having cyanide concentrations greater than 3.0 mg/L (Barillo et al, 1994).
    2) NOTE: Smoke inhalation exposes people to many chemicals. Cyanide is only one of the dangerous chemicals present in smoke.
    h) CASE REPORT - A blood cyanide level of 6.9 mg/L was reported in a 29-year-old man who intentionally ingested approximately 1250 mg of cyanide in the form of potassium dicyanoaurate (Kampe et al, 2000).

Workplace Standards

    A) ACGIH TLV Values for CAS57-12-5 (American Conference of Governmental Industrial Hygienists, 2010):
    1) Not Listed

    B) NIOSH REL and IDLH Values for CAS57-12-5 (National Institute for Occupational Safety and Health, 2007):
    1) Not Listed

    C) Carcinogenicity Ratings for CAS57-12-5 :
    1) ACGIH (American Conference of Governmental Industrial Hygienists, 2010): Not Listed
    2) EPA (U.S. Environmental Protection Agency, 2011): D ; Listed as: Cyanide, free
    a) D : Not classifiable as to human carcinogenicity.
    3) IARC (International Agency for Research on Cancer (IARC), 2016; International Agency for Research on Cancer, 2015; IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2010; IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2010a; IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2008; IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2007; IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2006; IARC, 2004): Not Listed
    4) NIOSH (National Institute for Occupational Safety and Health, 2007): Not Listed
    5) MAK (DFG, 2002): Not Listed
    6) NTP (U.S. Department of Health and Human Services, Public Health Service, National Toxicology Project ): Not Listed

    D) OSHA PEL Values for CAS57-12-5 (U.S. Occupational Safety, and Health Administration (OSHA), 2010):
    1) Listed as: Cyanides (as CN)
    2) Table Z-1 for Cyanides (as CN):
    a) 8-hour TWA:
    1) ppm:
    a) Parts of vapor or gas per million parts of contaminated air by volume at 25 degrees C and 760 torr.
    2) mg/m3: 5
    a) Milligrams of substances per cubic meter of air. When entry is in this column only, the value is exact; when listed with a ppm entry, it is approximate.
    3) Ceiling Value:
    4) Skin Designation: Yes
    5) Notation(s): Not Listed

Toxicity Information

    7.7.1) TOXICITY VALUES
    A) CALCIUM CYANIDE (References: ITI, 1988 Proctor et al, 1988 Sax & Lewis, 1989 RTECS, 1993)
    1) LD50- (ORAL)RAT:
    a) 39 mg/kg
    B) CYANIDE ION (References: ITI, 1988 Proctor et al, 1988 Sax & Lewis, 1989 RTECS, 1993)
    1) LD50- (INTRAPERITONEAL)MOUSE:
    a) 3 mg/kg
    C) CYANOGEN (References: ITI, 1988 Proctor et al, 1988 Sax & Lewis, 1989 RTECS, 1993)
    D) CYANOGEN BROMIDE (References: ITI, 1988; Proctor et al, 1988; Sax & Lewis, 1989; RTECS, 1993)
    E) CYANOGEN CHLORIDE (References: (ITI, 1988; Proctor et al, 1988; Sax & Lewis, 1989; RTECS, 1993)
    F) CYANOGEN IODIDE (References: ITI, 1988 Proctor et al, 1988 Sax & Lewis, 1989 RTECS, 1993)
    G) HYDROGEN CYANIDE (References: ITI, 1988 Proctor et al, 1988 Sax & Lewis, 1989 RTECS, 1993)
    1) LD50- (ORAL)MOUSE:
    a) 3.7 mg/kg
    H) POTASSIUM CYANIDE (References: ITI, 1988 Proctor et al, 1988 Sax & Lewis, 1989 RTECS, 1993)
    1) LD50- (INTRAPERITONEAL)MOUSE:
    a) 5991 mcg/kg
    2) LD50- (ORAL)MOUSE:
    a) 8500 mcg/kg
    3) LD50- (SUBCUTANEOUS)MOUSE:
    a) 6500 mcg/kg
    4) LD50- (INTRAPERITONEAL)RAT:
    a) 4 mg/kg
    5) LD50- (ORAL)RAT:
    a) 5 mg/kg
    b) 10 mg/kg
    6) LD50- (SUBCUTANEOUS)RAT:
    a) 9 mg/kg
    I) SODIUM CYANIDE (References: ITI, 1988 Proctor et al, 1988 Sax & Lewis, 1989 RTECS, 1993)
    1) LD50- (SUBCUTANEOUS)MOUSE:
    a) 3660 mcg/kg
    2) LD50- (ORAL)RAT:
    a) 6440 mcg/kg

Toxicologic Mechanism

    A) Cyanide (CN)- forms a stable complex with ferric iron (Fe3+) in the cytochrome oxidase enzymes, thereby inhibiting (=poisoning) cellular respiration. Therapy for cyanide poisoning is based on the following reactions:
    1.  Sodium nitrite
              +       ------->  Methemoglobin
          Hemoglogin            (ferric iron)
        (ferrous iron)
        NaNO2 + Hb Fe(2+) -----> Met Hb Fe(3+)
    2.  Methemoglobin
         + cyanide -------> Cyanmethemoglobin
        Met Hb Fe(3+)
         + CN-  ----------> (CN) Met Hb Fe(3+)
    3.  Cyanmethemoglobin ----->   Cyanide
            (slow dissociation)       +
                                 Methemoglobin
                                 (ferric iron)
        (CN)MET Hb Fe(3+) -----> CN- + MetHb Fe(3+)
                         (slow)
    4.  Cyanide                   Excreted
           +   ---> Thiocyanate --> in
        Thiosulfate                Urine
          CN-                     Excreted
           +  ---> CN S(2)O(3)3- --> in
        S(2)O(3)2-                 Urine
    

    1) In reaction (1), sodium nitrite reacts with hemoglobin in the red blood cells forming methemoglobin, which in turn can react with free cyanide ion forming cyanmethemoglobin, reaction (2), thereby binding free cyanide, and preventing its reaction with cytochrome oxidase enzymes in the cells.
    2) Cyanmethemoglobin dissociates slowly into free cyanide plus methemoglobin, reaction (3).
    3) The cyanide released by dissociation of cyanmethemoglobin then reacts with thiosulfate ion, reaction (4), forming thiocyanate, a relatively nontoxic compound that is excreted in the urine.
    B) In minimal lethal doses, cyanide affects primarily the CNS producing early stimulation followed by depression. It initially stimulates the peripheral chemoreceptors (causing increased respiration) and the carotid bodies (thereby slowing the heart) (Ellenhorn & Barceloux, 1988).
    C) Early CNS, respiratory, and myocardial depression result in decreased oxygenation of the blood and decreased cardiac output (Hall & Rumack, 1986). These effects produce both stagnation and hypoxemic hypoxia in addition to cytotoxic hypoxia from inhibition of mitochondrial cytochrome oxidase.
    D) Single doses of cyanide decrease oxidative metabolism, increase glycolysis, and inhibit brain glutamic acid decarboxylase, thereby decreasing GABA (Hartung, 1982; Gosselin et al, 1984). Besides the corpus callosum, the hippocampus, corpora striata, and substantia nigra are commonly damaged (Feldman & Feldman, 1990; Grandas et al, 1989) Klaassen et al, 1986; (Uitti et al, 1985).
    E) In vitro studies with guinea pig hippocampal slices suggest that cyanide has a direct effect on neurons not mediated by its inhibition of metabolism (Aitken & Braitman, 1989). This contradicts rat brain studies which showed KCN-induced abolition of EEG activity and reduction of cytochrome oxidase activity, ATP, and glycogen (MacMillan, 1989).
    F) KCN-induced membrane lipid peroxidation compromised antioxidant defenses in mouse brain and caused reductions in brain catalase, glutathione peroxidase, and glutathione reductase (Ardelt et al, 1989).
    G) Ardelt et al (1994) further demonstrated evidence of lipid peroxidation by measurement of elevated levels of conjugated dienes in mouse brain at 15 and 30 minutes after cyanide exposure but not after 60 minutes compared to controls. Cyanide exposure also produced elevated diene levels in the kidney, but not in the liver or heart (Ardelt et al, 1994).
    H) In vitro studies with rat hippocampal cell cultures suggest that KCN-mediated neurotoxicity is at least partly mediated via endogenous glutamate receptor activation (Sturm et al, 1993).

Physical Characteristics

    A) ODOR
    1) Clinically, cyanide poisoning is reported to produce a bitter, almond odor on the breath of the patient. However, only a small proportion of the population is genetically able to discern this characteristic odor (Lewis, 1992).
    2) HYDROGEN CYANIDE: Characteristic odor of bitter almonds (Clayton & Clayton, 1982; Patnaik, 1992)
    3) SODIUM CYANIDE
    a) The solid may have a light odor of hydrogen cyanide, especially is moisture is present (Clayton & Clayton, 1982).
    b) Faint odor of bitter almonds (ACGIH, 1991)
    4) POTASSIUM CYANIDE
    a) Similar to hydrogen cyanide (Clayton & Clayton, 1982)
    b) Faint odor of bitter almonds (ACGIH, 1991)
    5) CALCIUM CYANIDE
    a) Solid may have a rather definite odor of hydrogen cyanide (Clayton & Clayton, 1982)
    b) Faint odor of bitter almonds (ACGIH, 1991)
    6) CYANOGEN: Pungent odor (Clayton & Clayton, 1982)
    7) CYANOGEN CHLORIDE: Pungent odor (Clayton & Clayton, 1982)
    8) CYANOGEN BROMIDE: Penetrating odor (Clayton & Clayton, 1982)
    B) TASTE
    1) Typically, cyanide has a bitter, burning taste (Lewis, 1992).
    C) COLOR
    1) POTASSIUM, SODIUM and CALCIUM CYANIDES: White solid (ACGIH, 1991).
    2) HYDROGEN CYANIDE: Colorless or pale blue liquid or gas (Clayton & Clayton, 1982).
    3) CALCIUM CYANAMIDE: White crystalline solid (Clayton & Clayton, 1982).
    4) CYANOGEN: Colorless gas (Clayton & Clayton, 1982).
    5) CYANOGEN CHLORIDE: Colorless liquid or gas (Clayton & Clayton, 1982).
    6) CYANOGEN BROMIDE: Colorless crystal (needles or cubes) (Clayton & Clayton, 1982).
    7) DIMETHYL CYANAMIDE: Colorless liquid (Clayton & Clayton, 1982).

Ph

    A) Hydrogen cyanide is weakly acidic, but does not redden litmus (Budavari, 1996). The dissociation constant is relatively weak and is similar to that of amino acids (Patnaik, 1992).

Molecular Weight

    A) CYANIDE ION: 26.02
    B) POTASSIUM CYANIDE: 65.11
    C) SODIUM CYANIDE: 49.02
    D) CALCIUM CYANIDE: 92.12
    E) HYDROGEN CYANIDE: 27.03
    F) CALCIUM CYANAMIDE: 80.11
    G) CYANOGEN: 52.04
    H) CYANOGEN CHLORIDE: 61.48
    I) CYANOGEN BROMIDE: 105.93
    J) DIMETHYL CYANAMIDE: 70.10

General Bibliography

    1) 40 CFR 372.28: Environmental Protection Agency - Toxic Chemical Release Reporting, Community Right-To-Know, Lower thresholds for chemicals of special concern. National Archives and Records Administration (NARA) and the Government Printing Office (GPO). Washington, DC. Final rules current as of Apr 3, 2006.
    2) 40 CFR 372.65: Environmental Protection Agency - Toxic Chemical Release Reporting, Community Right-To-Know, Chemicals and Chemical Categories to which this part applies. National Archives and Records Association (NARA) and the Government Printing Office (GPO), Washington, DC. Final rules current as of Apr 3, 2006.
    3) 49 CFR 172.101 - App. B: Department of Transportation - Table of Hazardous Materials, Appendix B: List of Marine Pollutants. National Archives and Records Administration (NARA) and the Government Printing Office (GPO), Washington, DC. Final rules current as of Aug 29, 2005.
    4) 49 CFR 172.101: Department of Transportation - Table of Hazardous Materials. National Archives and Records Administration (NARA) and the Government Printing Office (GPO), Washington, DC. Final rules current as of Aug 11, 2005.
    5) 62 FR 58840: Notice of the National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances - Proposed AEGL Values, Environmental Protection Agency, NAC/AEGL Committee. National Archives and Records Administration (NARA) and the Government Publishing Office (GPO), Washington, DC, 1997.
    6) 65 FR 14186: Notice of the National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances - Proposed AEGL Values, Environmental Protection Agency, NAC/AEGL Committee. National Archives and Records Administration (NARA) and the Government Publishing Office (GPO), Washington, DC, 2000.
    7) 65 FR 39264: Notice of the National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances - Proposed AEGL Values, Environmental Protection Agency, NAC/AEGL Committee. National Archives and Records Administration (NARA) and the Government Publishing Office (GPO), Washington, DC, 2000.
    8) 65 FR 77866: Notice of the National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances - Proposed AEGL Values, Environmental Protection Agency, NAC/AEGL Committee. National Archives and Records Administration (NARA) and the Government Publishing Office (GPO), Washington, DC, 2000.
    9) 66 FR 21940: Notice of the National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances - Proposed AEGL Values, Environmental Protection Agency, NAC/AEGL Committee. National Archives and Records Administration (NARA) and the Government Publishing Office (GPO), Washington, DC, 2001.
    10) 67 FR 7164: Notice of the National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances - Proposed AEGL Values, Environmental Protection Agency, NAC/AEGL Committee. National Archives and Records Administration (NARA) and the Government Publishing Office (GPO), Washington, DC, 2002.
    11) 68 FR 42710: Notice of the National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances - Proposed AEGL Values, Environmental Protection Agency, NAC/AEGL Committee. National Archives and Records Administration (NARA) and the Government Publishing Office (GPO), Washington, DC, 2003.
    12) 69 FR 54144: Notice of the National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances - Proposed AEGL Values, Environmental Protection Agency, NAC/AEGL Committee. National Archives and Records Administration (NARA) and the Government Publishing Office (GPO), Washington, DC, 2004.
    13) ACGIH: Documentation of the Threshold Limit Values and Biological Exposure Indices, Vol 1, 6th ed, Am Conference of Govt Ind Hyg, Inc, Cincinnati, OH, 1991.
    14) ACGIH: Documentation of the Threshold Limit Values, 5th ed, Am Conference of Govt Ind Hyg, Inc, Cincinnati, OH, 1986.
    15) AIHA: 2006 Emergency Response Planning Guidelines and Workplace Environmental Exposure Level Guides Handbook, American Industrial Hygiene Association, Fairfax, VA, 2006.
    16) AMA Department of DrugsAMA Department of Drugs: AMA Evaluations Subscription, American Medical Association, Chicago, IL, 1992.
    17) Aitken PG & Braitman DJ: The effects of cyanide on neural and synaptic function in hippocampal slices. Neurotoxicol 1989; 10:239-248.
    18) 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.
    19) American Conference of Governmental Industrial Hygienists : ACGIH 2010 Threshold Limit Values (TLVs(R)) for Chemical Substances and Physical Agents and Biological Exposure Indices (BEIs(R)), American Conference of Governmental Industrial Hygienists, Cincinnati, OH, 2010.
    20) Amery WK, Wauquier A, & van Neuten JM: The anti-migrainous pharmacology of flunarizine (R14950), a calcium antagonist. Drugs Exper Clin Res 1981; 7:1-10.
    21) Ames MM, Kovach JS, & Flora KP: Initial pharmacologic studies of amygdalin (laetrile) in man. Res Commun Chem Pathol Pharmacol 1978; 22:175-185.
    22) Anderson AH: Experimental studies on the pharmacology of activated charcoal. Acta pharmacol 1946; 2:69-78.
    23) Ardelt BK, Borowitz JL, & Isom GE: Brain lipid peroxidation and antioxidant protectant mechanisms following acute cyanide intoxication. Toxicology 1989; 56:147-154.
    24) Ardelt BK, Borowitz JL, & Maduh EU: Cyanide-induced lipid peroxidation in different organs: subcellular distribution and hydroperoxide generation in neuronal cells. Toxicol 1994; 89:127-137.
    25) 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.
    26) Ashton D, van Reempts J, & Wauquier A: Behavioral, electroencephalographic and histological study of the protective effect of etomidate against histotoxic dysoxia produced by cyanide. Arch Int Pharmacodyn Ther 1980; 254:196-213.
    27) Ballantyne B: Acute systemic toxicity of cyanides by topical application to the eye. J Toxicol Cut Ocular Toxicol 1983; 2:119-129.
    28) Ballantyne B: Quantitative comparative cyanogenesis of malononitrile (MN) and succinonitrile (SN) (abstract). Vet Hum Toxicol 1987; 29:489.
    29) Banerjee KK, Bishayee A, & Marimuthu P: Evaluation of cyanide exposure and its effect on thyroid function of workers in cable industry. JOEM 1997; 39:258-260.
    30) Barillo DJ, Goode R, & Esch V: Cyanide poisoning in victims of fire: analysis of 364 cases and review of the literature. J Burn Care Rehabil 1994; 15:46-57.
    31) Barr SJ: Chemical warfare agents. Topics Emerg Med 1985; 7:62-70.
    32) Baselt RC & Cravey RH: Disposition of Toxic Drugs and Chemicals in Man, 3rd ed, Year Book Medical Publishers, Chicago, IL, 1989.
    33) Baselt RC: Biological Monitoring Methods for Industrial Chemicals, 2nd ed, PSG Publishing Company, Littleton, MA, 1988.
    34) Baud FJ, Imbert M, & Vicaut E: Time-course of lactic acidosis in acute cyanide poisonings treated with oxygen and high doses of hydroxocobalamin (abstract), EAPCCT XVI International Congress, Vienna, Austria, 1994.
    35) Becker CE: The role of cyanide in fires. Vet Hum Toxicol 1985; 27:487-490.
    36) Beckerman N, Leikin SM, Aitchinson R, et al: Laboratory interferences with the newer cyanide antidote: hydroxocobalamin. Semin Diagn Pathol 2009; 26(1):49-52.
    37) Berlin CM Jr: Treatment of cyanide poisoning in children. Pediatrics 1970; 46:793-796.
    38) Berlin: Treatment of cyanide poisoning in children. Pediatr 1970a; 46:793-796.
    39) Bhattacharya R & Vijayaraghavan R: Promising role of alpha-ketoglutarate in protecting against the lethal effects of cyanide. Hum Exp Toxicol 2002; 21(6):297-303.
    40) Bismuth C, Cantineau J-P, & Pontal P: Priorite de l'oxygenation dans l'intoxication cyanhydrique: A propos de 25 cas. J Toxicol Med 1984; 4:107-121.
    41) Blanc P, Hogan M, & Malin K: Cyanide intoxication among silver-reclaiming workers. JAMA 1985; 253:367-371.
    42) Boggild MD, Peck RW, & Tomson CRV: Acetonitrile ingestion: delayed onset of cyanide poisoning to concurrent ingestion of acetone. Postgrad Med J 1990; 66:40-41.
    43) Bonnichsen R & Maely AC: Poisoning by volatile compounds. J Forensic Sci 1966; 11:516-527.
    44) Bonsall JL: Survival without sequelae following exposure to 500 mg/m3 of hydrogen cyanide. Human Toxicol 1984; 3:57-60.
    45) Borgohain R, Singh AK, & Radhakrishna H: Delayed onset generalized dystonia after cyanide poisoning. Clin Neurol Neurosurg 1995; 97:213-215.
    46) Borowitz JL, Rathinavelu A, & Kanthasamy A: Accumulation of labeled cyanide in neuronal tissue. Toxicol Applied Pharmacol 1994; 129:80-85.
    47) Borron SW , Uhl W , Nolting A , et al: Interference of the cyanide antidote hydroxocobalamin with carboxyhemoglobin measurements should not limit clinical use in suspected or confirmed cyanide poisoning. Ann Emerg Med 2007; 50(5):624-625.
    48) Borron SW, Baud FJ, Barriot P, et al: Prospective study of hydroxocobalamin for acute cyanide poisoning in smoke inhalation. Ann Emerg Med 2007b; 49(6):794-801, 801.
    49) Borron SW, Baud FJ, Megarbane B, et al: Hydroxocobalamin for severe acute cyanide poisoning by ingestion or inhalation. Am J Emerg Med 2007a; 25(5):551-558.
    50) Bourrelier J & Paulet G: Intoxication cyanhydrique consecutive a des brulures graves par cyanure de sodium fondu. Sur trois cas traites par EDTA cobaltique. Presse Med 1971; 22:1013-1014.
    51) Breen PH, Isserles SA, & Tabac E: Protective effect of stroma-free methemoglobin during cyanide poisoning in dogs. Anesthesiology 1996; 85:558-564.
    52) Brennan DF: Cyanide poisoning and the anion gap (letter). Ann Emerg Med 1988; 17:298.
    53) Bright JE & Marrs TC: Effects of p-aminopropiophenone (PAPP), a cyanide antidote, on cyanide given by intravenous infusion. Hum Toxicol 1987; 6:133-137.
    54) Bright JE, Inns RH, & Tuckwell NJ: The effect of storage upon cyanide in blood samples. Hum Exp Toxicol 1990; 9:125-129.
    55) Brophy GM, Bell R, Claassen J, et al: Guidelines for the evaluation and management of status epilepticus. Neurocrit Care 2012; 17(1):3-23.
    56) 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.
    57) Buchter A & Peter H: Clinical toxicology of acrylonitrile. G Ital Med Lav 1984; 6:83-86.
    58) Budavari S: The Merck Index, 11th ed. Merck & Co, Inc, Rahway, NJ, 1989. E.SK 1 Burrows G & Way GL: Antagonism of cyanide toxicity by phenoxybenzamine. Fed Proc 1976; 35:533.
    59) Budavari S: The Merck Index, 12th ed, Merck & Co, Inc, Rahway, NJ, 1996.
    60) Burgess JL, Kirk M, Borron SW, et al: Emergency department hazardous materials protocol for contaminated patients. Ann Emerg Med 1999; 34(2):205-212.
    61) Burrows GE & Way JL: Cyanide intoxication in sheeps: Therapeutic value of oxygen or colbalt. Am J Vet Res 1977; 38:223-227.
    62) CDC: Cyanide poisonings associated with over-the-counter medication - Washington State, 1991. CDC: MMWR 1991; 40:161-168.
    63) Calafat AM & Stanfill SB: Rapid quantitation of cyanide in whole blood by automated headspace gas chromatography. Journal of Chromatography B 2002; 772(1):131-137.
    64) Caravati EM & Litovitz TL: Pediatric cyanide intoxication from an acetonitrile-containing cosmetic. JAMA 1988; 260:3470-3473.
    65) Caravati EM, Knight HH, & Linscott MS: Esophageal laceration and charcoal mediastinum complicating gastric lavage. J Emerg Med 2001; 20:273-276.
    66) Carden E: Hyperbaric oxygen in cyanide poisoning. Anaesthesia 1970; 25:442-443.
    67) Carella F, Grassi MP, & Savoiardo M: Dystonic-Parkinsonian syndrome after cyanide poisoning: clinical and MRI findings. J Neurol Neurosurg Psychiatr 1988; 51:1345-1348.
    68) Casadei E, Cliff J, & Neves J: Surveillance of urinary thiocyanate concentration after epidemic spastic paraparesis in Mozambique. J Trop Med Hyg 1990; 93:257-261.
    69) 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.
    70) Chadha RK, Bryce F, & Lawrence JF: Toxicity of potassium cyanide added to fresh fruit and juice. Food Chem Toxicol 1991; 29:681-684.
    71) 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.
    72) Chandra H, Gupta BN, & Bhargava SK: Chronic cyanide exposure - a biochemical and industrial hygiene study. J Anal Toxicol 1980; 4:161-165.
    73) Chen F, Jiang L, & Yang B: Visual loss caused by acute cyanide poisoning: A case report. Clin Toxicol (Phila) 2011; 49(2):121-123.
    74) 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.
    75) Choonara IA & Rane A: Therapeutic drug monitoring of anticonvulsants state of the art. Clin Pharmacokinet 1990; 18:318-328.
    76) Chyka PA, Seger D, Krenzelok EP, et al: Position paper: Single-dose activated charcoal. Clin Toxicol (Phila) 2005; 43(2):61-87.
    77) Clark CJ, Cambell D, & Reid WH: Blood carboxyhemoglobin and cyanide levels in fire survivors. Lancet 1981; 1:1332-1335.
    78) Clayton GD & Clayton FE: Patty's Industrial Hygiene and Toxicology, Vol 2C. Toxicology, 3rd ed, John Wiley & Sons, New York, NY, 1982, pp 4845-4863.
    79) Cliff J, Essers S, & Rosling H: Ankle clonus correlating with cyanide intake from cassava in rural children from Mozambique. J Trop Pediatr 1986; 32:186-189.
    80) Colle R: L'intoxication cyanhydrique chronique. Maroc Medicale 1972; 50:750-757.
    81) Cope C: The importance of oxygen in the treatment of cyanide poisoning. JAMA 1961; 175:1061.
    82) Curry SC, Connor DA, & Raschke RA: Effect of hydroxocobalamin on commonly ordered laboratory chemistries. Vet Hum Toxicol 1992; 34:345.
    83) DFG: List of MAK and BAT Values 2002, Report No. 38, Deutsche Forschungsgemeinschaft, Commission for the Investigation of Health Hazards of Chemical Compounds in the Work Area, Wiley-VCH, Weinheim, Federal Republic of Germany, 2002.
    84) 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.
    85) Davignon JP, Trissel LA, & Kleinman LM: Pharmaceutical assessment of amygdalin (laetrile) products. Cancer Treat Rep 1978; 62:99-104.
    86) Davis FM & Ewer T: Acute cyanide poisoning: case report of the use of hyperbaric oxygen. J Hyperb Med 1988; 3:103-106.
    87) Davison V: Cyanide poisoning: Kelocyanor -- a new treatment. Occup Health 1969; 21:306-308.
    88) De Busk RF & Seidl LG: Attempted suicide by cyanide. A report of two cases. Calif Med 1969; 110:394-396.
    89) Deng JF & Yang CC: Diabetes insipidus (DI) in cyanide poisoning - a case report, EAPCCT XVI International Congress, Vienna, Austria, 1994.
    90) DiNapoli J, Hall AH, & Drake R: Cyanide and arsenic poisoning by intravenous injection. Ann Emerg Med 1989; 18:308-311.
    91) Dodds C & McKnight C: Cyanide toxicity after immersion and the hazards of dicobalt edetate. Br Med J 1985; 291:785-786.
    92) Doherty PA, Ferm VH, & Smith RP: Congenital malformations induced by infusion of sodium cyanide in the Golden Hamster. Toxicol Appl Pharmacol 1982; 64:456-464.
    93) Dubinsky B, Sierchio JN, & Temple DE: Flunarizine and verapamil: effects on central nervous system and peripheral consequences of cytotoxic hypoxia in rats. Life Sci 1984; 34:1299-1306.
    94) Dunipace AJ, Beaven R, Noblitt T, et al: Mutagenic potential of toluidine blue evaluated in the Ames test. Mutat Res 1992; 279(4):255-259.
    95) EPA: EPA chemical profile on potassium cyanide, Environmental Protection Agency, Washington, DC, 1985.
    96) EPA: Search results for Toxic Substances Control Act (TSCA) Inventory Chemicals. US Environmental Protection Agency, Substance Registry System, U.S. EPA's Office of Pollution Prevention and Toxics. Washington, DC. 2005. Available from URL: http://www.epa.gov/srs/.
    97) ERG: Emergency Response Guidebook. A Guidebook for First Responders During the Initial Phase of a Dangerous Goods/Hazardous Materials Incident, U.S. Department of Transportation, Research and Special Programs Administration, Washington, DC, 2004.
    98) Ellenhorn MJ & Barceloux DG: Medical Toxicology: Diagnosis and Treatment of Human Poisoning, Elsevier, New York, NY, 1988.
    99) Elliot CG, Colby TV, & Kelly TM: Charcoal lung. Bronchiolitis obliterans after aspiration of activated charcoal. Chest 1989; 96:672-674.
    100) FDA: Poison treatment drug product for over-the-counter human use; tentative final monograph. FDA: Fed Register 1985; 50:2244-2262.
    101) Feihl F, Domenighetti D, & Perret CI: Intoxication massive au cyanure avec evolution favorable. Schweiz Med Wschr 1982; 112:1280-1282.
    102) Feldman JM & Feldman MD: Sequelae of attempted suicide by cyanide ingestion: a case report. Int J Psychiatry Med 1990; 20:173-179.
    103) Feldstein M & Klendshoj NC: J Lab Chin Med 1954; 44:166-170. As cited in NIOSH: Criteria Document: Hydrogen Cyanide and Cyanide Salts. National Institutes of Occupational Safety and Health: DHEW Pub NIOSH 77-108, 1976.
    104) Fernando GCA & Busuttil A: Cyanide ingestion: case studies of four suicides. Am J Forensic Med Pathol 1991; 12:241-246.
    105) Finkel AJ: Hamilton and Hardy's Industrial Toxicology, 4th ed, John Wright, PSG Inc, Boston, MA, 1983.
    106) Fligner CL, Luthi R, & Linkaityte-Weiss E: Paper strip screening method for detection of cyanide in blood using CYANTESMO test paper. Am J Forensic Med Pathol 1992; 13:81-84.
    107) Forsyth JC, Becker CE, & Osterloh J: Hydroxocobalamin as a cyanide antidote: safety, efficacy, and pharmacokinetics in heavy smokers. Vet Hum Toxicol 1992; 34:338.
    108) Forsyth JC, Mueller PD, & Becker CE: Hydroxocobalamin as a cyanide antidote: safety, efficacy and pharmacokinetics in heavily smoking normal volunteers. Clin Toxicol 1993; 31:277-294.
    109) Fortin JL, Ruttimann M, Capellier G, et al: Successful organ transplantation after treatment of fatal cyanide poisoning with hydroxocobalamin. Clin Toxicol (Phila) 2007; 45(5):468-471.
    110) Free AH & Free HM: Laboratory detection of cyanide poisoning. In: Sunderman FW & Sunderman FW Jr (Eds): Laboratory Diagnosis of Diseases caused by Toxic Agents, Warren H Green, Inc, St Louis, MO, 1970, pp 286-291.
    111) Freeman AG: Optic neuropathy and chronic cyanide intoxication: a review. J R Soc Med 1988; 81:103-106.
    112) Freeman HM: Standard Handbook of Hazardous Waste Treatment and Disposal, McGraw-Hill Book Company, New York, NY, 1989.
    113) Garlich FM, Alsop JA, Anderson DL, et al: Poisoning and suicide by cyanide jewelry cleaner in the US Hmong community: a case series. Clin Toxicol (Phila) 2012; 50(2):136-140.
    114) Geiger LE, Hogy LL, & Guengerich FP: Metabolism of acrylonitrile by isolated rat hepatocytes. Cancer Res 1983; 43:3080-3087.
    115) Geller RJ, Barthold C, Saiers JA, et al: Pediatric cyanide poisoning: causes, manifestations, management, and unmet needs. Pediatrics 2006; 118(5):2146-2158.
    116) Geller RJ, Eskins BR, & Iknoian RC: Cyanide Toxicity from acetonitrile-containing false nail remover. Am J Emerg Med 1991; 9:268-270.
    117) Gettler AD & St George V: Cyanide poisoning. Am J Clin Pathol 1934; 4:429-437.
    118) Gill JR, Marker E, & Stajic M: Suicide by cyanide: 17 deaths. J Forensic Sci 2004; 49(4):826-828.
    119) Golej J, Boigner H, Burda G, et al: Severe respiratory failure following charcoal application in a toddler. Resuscitation 2001; 49:315-318.
    120) Gonzales J & Sabatini S: Cyanide poisoning: pathophysiology and current approaches to therapy. Int J Artif Organs 1989; 12:347-355.
    121) Goodhart GL: Patient treated with antidote kit and hyperbaric oxygen survives cyanide poisoning. Southern Med J 1994; 87:814-816.
    122) Gosselin RE, Smith RP, & Hodge HC: Clinical Toxicology of Commercial Products, 5th ed, Williams & Wilkins, Baltimore, MD, 1984.
    123) Graff GR, Stark J, & Berkenbosch JW: Chronic lung disease after activated charcoal aspiration. Pediatrics 2002; 109:959-961.
    124) Graham DL, Laman D, & Theodore J: Acute cyanide poisoning complicated by lactic acidosis and pulmonary edema. Arch Intern Med 1977; 137:1051-1055.
    125) Grandas F, Artieda J, & Obeso JA: Clinical and CT scan findings in a case of cyanide intoxication. Mov Disord 1989; 4:188-193.
    126) Grant WM: Toxicology of the Eye, 3rd ed, Charles C. Thomas, Springfield, IL, 1986, pp 287-290.
    127) Groff WA, Stemler FW, & Kaminskis A: Plasma free cyanide and blood total cyanide: A rapid completely automated microdistillation assay. Clin Toxicol 1985; 23:133-163.
    128) 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.
    129) HSDB : Hazardous Substances Data Bank. National Library of Medicine. Bethesda, MD (Internet Version). Edition expires 1991; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    130) HSDB : Hazardous Substances Data Bank. National Library of Medicine. Bethesda, MD (Internet Version). Edition expires 1993; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    131) Haas CF: Mechanical ventilation with lung protective strategies: what works?. Crit Care Clin 2011; 27(3):469-486.
    132) Haguenoer JM, Dequidt J, & Levy JC: Intoxications experimentales par l'acetonitrile. 4e note: Influence de l'hydroxocobalamine sur l'intoxication a moyen terme. Eur J Toxicol 1975; 8:113-121.
    133) Hall AH & Rumack BH: Clinical toxicology of cyanide. Ann Emerg Med 1986; 15:1067-1074.
    134) Hall AH & Rumack BH: Hydroxycobalamin/sodium thiosulfate as a cyanide antidote. J Emerg Med 1987; 5:115-121.
    135) Hantson P, N'Geye P, & Laforge M: Suicide attempt due to the ingestion of potassium hexacyanoferate (abstract), EAPCCT XVI International Congress, Vienna, Austria, 1994.
    136) Hantson P, Squifflet JP, & Vanormelingen P: Organ transplantation after fatal cyanide poisoning. Clin Transplantation 1999; 13:72-73.
    137) Hantson P, Squifflet JP, & Vekemans MC: Successful pancreas and renal transplantations from a donor who died of cyanide poisoning. Clin Transplantation 1991; 5:419-421.
    138) Harris CR & Filandrinos D: Accidental administration of activated charcoal into the lung: aspiration by proxy. Ann Emerg Med 1993; 22:1470-1473.
    139) Hart GB, Strauss MB, & Lennon PA: Treatment of smoke inhalation by hyperbaric oxygen. J Emerg Med 1985; 3:211-215.
    140) Hartung R: Potassium cyanide, in Clayton GD & Clayton FE (eds): Patty's Industrial Hygiene and Toxicology, Vol 2C, Toxicology, 3rd ed, John Wiley & Sons, New York, NY, 1982.
    141) Hegenbarth MA & American Academy of Pediatrics Committee on Drugs: Preparing for pediatric emergencies: drugs to consider. Pediatrics 2008; 121(2):433-443.
    142) Herman MI, Chyka PA, & Butlse AY: Methylene blue by intraosseous infusion for methemoglobinemia. Ann Emerg Med 1999; 33:111-113.
    143) Hillman B, Bardhan KD, & Bain JTB: The use of dicobalt edetate (Kelocyanor) in cyanide poisoning. Postgrad Med J 1974; 50:171-174.
    144) Hix WR & Wilson WR: Toluidine blue staining of the esophagus: a useful adjunct in the panendoscopic evaluation of patients with squamous cell carcinoma of the head and neck. Arch Otolaryngol Head Neck Surg 1987; 113(8):864-865.
    145) Hjelt K, Lund JT, Scherling B, et al: Methaemoglobinaemia among neonates in a neonatal intensive care unit. Acta Paediatr 1995; 84(4):365-370.
    146) Holzbecher MD, Moss MA, & Ellenberger HA: The cyanide content of Laetrile preparation, apricot, peach and apple seeds. Clin Toxicol 1984; 22:341-347.
    147) 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, 2006, pp 826-828.
    148) Howland MA: Sodium Thiosulfate. In: Nelson LS, Lewin NA, Howland MA, et al, eds. Goldfrank's Toxicologic Emergencies, 9th ed. McGraw Hill Medical, New York, NY, 2011, pp 1692-1694.
    149) Hsiao PJ, Chang CF, Chiu CC, et al: High Anion Gap Metabolic Acidosis after a Suicide Attempt with Cyanide: The Rebirth of Cyanide Poisoning. Intern Med 2015; 54(15):1901-1904.
    150) Hugh-Jones P: Diabetes in Jamaica. Lancet 1955; 2:891-897.
    151) Hvidberg EF & Dam M: Clinical pharmacokinetics of anticonvulsants. Clin Pharmacokinet 1976; 1:161.
    152) IARC Working Group on the Evaluation of Carcinogenic Risks to Humans : IARC Monographs on the Evaluation of Carcinogenic Risks to Humans: 1,3-Butadiene, Ethylene Oxide and Vinyl Halides (Vinyl Fluoride, Vinyl Chloride and Vinyl Bromide), 97, International Agency for Research on Cancer, Lyon, France, 2008.
    153) IARC Working Group on the Evaluation of Carcinogenic Risks to Humans : IARC Monographs on the Evaluation of Carcinogenic Risks to Humans: Formaldehyde, 2-Butoxyethanol and 1-tert-Butoxypropan-2-ol, 88, International Agency for Research on Cancer, Lyon, France, 2006.
    154) IARC Working Group on the Evaluation of Carcinogenic Risks to Humans : IARC Monographs on the Evaluation of Carcinogenic Risks to Humans: Household Use of Solid Fuels and High-temperature Frying, 95, International Agency for Research on Cancer, Lyon, France, 2010a.
    155) IARC Working Group on the Evaluation of Carcinogenic Risks to Humans : IARC Monographs on the Evaluation of Carcinogenic Risks to Humans: Smokeless Tobacco and Some Tobacco-specific N-Nitrosamines, 89, International Agency for Research on Cancer, Lyon, France, 2007.
    156) IARC Working Group on the Evaluation of Carcinogenic Risks to Humans : IARC Monographs on the Evaluation of Carcinogenic Risks to Humans: Some Non-heterocyclic Polycyclic Aromatic Hydrocarbons and Some Related Exposures, 92, International Agency for Research on Cancer, Lyon, France, 2010.
    157) IARC: List of all agents, mixtures and exposures evaluated to date - IARC Monographs: Overall Evaluations of Carcinogenicity to Humans, Volumes 1-88, 1972-PRESENT. World Health Organization, International Agency for Research on Cancer. Lyon, FranceAvailable from URL: http://monographs.iarc.fr/monoeval/crthall.html. As accessed Oct 07, 2004.
    158) ICAO: Technical Instructions for the Safe Transport of Dangerous Goods by Air, 2003-2004. International Civil Aviation Organization, Montreal, Quebec, Canada, 2002.
    159) ILO: ILO Encyclopaedia of Occupational Health and Safety, 3rd ed, Vol I. Parmeggiani L (Ed), International Labour Organization, Geneva, Switzerland, 1983.
    160) International Agency for Research on Cancer (IARC): IARC monographs on the evaluation of carcinogenic risks to humans: list of classifications, volumes 1-116. International Agency for Research on Cancer (IARC). Lyon, France. 2016. Available from URL: http://monographs.iarc.fr/ENG/Classification/latest_classif.php. As accessed 2016-08-24.
    161) International Agency for Research on Cancer: IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. World Health Organization. Geneva, Switzerland. 2015. Available from URL: http://monographs.iarc.fr/ENG/Classification/. As accessed 2015-08-06.
    162) Ivanov KP: The effect of elevated oxygen pressure on animals poisoned with potassium cyanide. Pharmacol Toxicol 1959; 22:476-479.
    163) Jee J, Yoshikawa F, & Pont L: Assay of amygdalin dosage forms from Mexico. J Pharm Sci 1978; 67:438-440.
    164) Johanning RJ, Zaska DE, & Tschida SJ: A retrospective study of sodium nitroprusside use and assessment of the potential risk of cyanide poisoning. Pharmacotherapy 1995; 15:773-777.
    165) Johannsen FR, Levinskas GJ, & Berteau PE: Evaluation of the teratogenic potential of three aliphatic nitriles in the rat. Fund Appl Toxicol 1986; 7:33-40.
    166) Johnson AR, Chauv S, Bair JD, et al: Rapid resolution of severe lactic acidosis following hydroxocobalamin administration in an intentional potassium cyanide overdose (abstracts). Clin Toxicol (Phila) 2015; 53(7):700.
    167) Johnson JD, Meisenheimer TL, & Isom GE: Cyanide-induced neurotoxicity: role of neuronal calcium. Toxicol Appl Pharmacol 1986; 84:464-469.
    168) Johnson RP & Mellors JW: Arteriolization of venous blood gases: a clue to the diagnosis of cyanide poisoning. J Emerg Med 1988; 6:401-404.
    169) Johnson WS, Hall AH, & Rumack BH: Cyanide poisoning successfully treated without 'therapeutic methemoglobin levels'. Am J Emerg Med 1989; 7:437-440.
    170) Jouglard J, Fagot G, & Deguigne B: L'intoxication cyanhydrique aigue et son traitement d'urgence. Marseille Med 1971; 9:571-575.
    171) Jouglard J, Nava G, & Botta A: A propos d'une intoxication aigue par le cyanure de potassium traitee par l'hydroxocobalamine. Marseille Med 1974; 12:617-624.
    172) Kampe S, Iffland R, Korenkov M, et al: Survival from a lethal blood concentration of cyanide with associated alcohol intoxication. Anaesthesia 2000; 55(12):1189-1191.
    173) Kerns W, Beuhler M, & Tomaszewski C: Hydroxocobalamin versus thiosulfate for cyanide poisoning. Ann Emerg Med 2008; 51(3):338-339.
    174) Kiese M , Lorcher W , Weger N , et al: Comparative studies on the effects of toluidine blue and methylene blue on the reduction of ferrihaemoglobin in man and dog. Eur J Clin Pharmacol 1972; 4(2):115-118.
    175) Kirk MA, Gerace R, & Kulig KW: Cyanide and methemoglobin kinetics in smoke inhalation victims treated with the cyanide antidote kit. Ann Emerg Med 1993; 22:1413-1418.
    176) 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.
    177) Kollef MH & Schuster DP: The acute respiratory distress syndrome. N Engl J Med 1995; 332:27-37.
    178) Kraut JA & Madias NE: Metabolic acidosis: pathophysiology, diagnosis and management. Nat Rev Nephrol 2010; 6(5):274-285.
    179) Krieg A & Saxena K: Cyanide poisoning from metal cleaning solutions. Ann Emerg Med 1987; 16:582-584.
    180) Kruszyna R, Kruszyna H, & Smith RP: Comaprison of hydroxylamine, 4-dimethylaminophenol and nitrite protection against cyanide poisoning in mice. Arch Toxicol 1982; 49:191-202.
    181) Lambert RJ, Kindler BL, & Schaeffer DJ: The efficacy of superactivated charcoal in treating rats exposed to a lethal oral dose of potassium cyanide. Ann Emerg Med 1988; 17:595-598.
    182) Lasch EE & El Shawa R: Multiple cases of cyanide poisoning by apricot kernels in children from Gaza. Pediatrics 1981; 68:5-7.
    183) Lee J , Mukai D , Kreuter K , et al: Potential interference by hydroxocobalamin on cooximetry hemoglobin measurements during cyanide and smoke inhalation treatments. Ann Emerg Med 2007; 49(6):802-805.
    184) Lenart M, Buckenmaier CC 3rd, Kim MJ, et al: Development of a complicated pain syndrome following cyanide poisoning in a U.S. soldier. Mil Med 2010; 175(4):292-294.
    185) Leung P, Sylvester DM, & Chiou F: Stereospecific effect of naloxone hydrochloride on cyanide intoxication. Toxicol Appl Pharmacol 1984; 83:525-530.
    186) Levi L, French WN, & Bickis IJ: Laetrile: a study of its physiochemical and biochemical properties. Can Med Assoc J 1965; 92:1057-1061.
    187) Lewis RJ: Sax's Dangerous Properties of Industrial Materials, 8th ed, Van Nostrand Reinhold Company, New York, NY, 1992, pp 974-975.
    188) Lindenmann J, Matzi V, Kaufmann P, et al: Hyperbaric oxygenation in the treatment of life-threatening isobutyl nitrite-induced methemoglobinemia--a case report. Inhal Toxicol 2006; 18(13):1047-1049.
    189) Lindquist TD, Oiland D, & Weber K: Cyanide poisoning victims as corneal transplant donors. Am J Ophthalmol 1988; 106:354-355.
    190) Litovitz TL, Larkin RF, & Myers RAM: Cyanide poisoning treated with hyperbaric oxygen. Am J Emerg Med 1983; 1:94-101.
    191) Loddenkemper T & Goodkin HP: Treatment of Pediatric Status Epilepticus. Curr Treat Options Neurol 2011; Epub:Epub.
    192) MacMillan VH: Cerebral energy metabolism in cyanide encephalopathy. J Cereb Blood Flow Metab 1989; 9:156-162.
    193) Maduh EU, Johnson JD, & Ardelt BK: Cyanide-induced neurotoxicity: mechanisms of attenuation by chlorpromazine. Toxicol Appl Pharmacol 1988; 96:60-67.
    194) Manno EM: New management strategies in the treatment of status epilepticus. Mayo Clin Proc 2003; 78(4):508-518.
    195) Marquez A & Todd M: Acute hemolytic anemia and agranulocytosis following intravenous administration of toluidine blue. Am Pract 1959; 10:1548-1550.
    196) Marrs TC: Antidotal treatment of acute cyanide poisoning. Adverse Drug React Acute Poisoning Rev 1988; 4:179-206.
    197) Moore SJ, Norris JC, & Ho IK: The efficacy of alphaketoglutaric acid in the antagonism of cyanide intoxication. Toxicol Appl Pharmacol 1986; 82:40-44.
    198) Moore SJ, Norris JC, & Walsh DA: Antidotal use of methemoglobin forming cyanide antagonists in concurrent carbon monoxide/cyanide intoxication. J Pharmacol Exp Ther 1987; 242:70-73.
    199) Mueller M & Borland C: Delayed cyanide poisoning following acetonitrile ingestion. Postgrad Med J 1997; 73:299-300.
    200) Myers RAM & Schnitzer BM: Hyperbaric oxygen use: Update 1984. Postgrad Med 1984; 76:83-95.
    201) NFPA: Fire Protection Guide on Hazardous Materials, 9th ed, National Fire Protection Association, Quincy, MA, 1986.
    202) NFPA: Fire Protection Guide to Hazardous Materials, 13th ed., National Fire Protection Association, Quincy, MA, 2002.
    203) 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.
    204) NRC: Acute Exposure Guideline Levels for Selected Airborne Chemicals - Volume 1, Subcommittee on Acute Exposure Guideline Levels, Committee on Toxicology, Board on Environmental Studies and Toxicology, Commission of Life Sciences, National Research Council. National Academy Press, Washington, DC, 2001.
    205) NRC: Acute Exposure Guideline Levels for Selected Airborne Chemicals - Volume 2, Subcommittee on Acute Exposure Guideline Levels, Committee on Toxicology, Board on Environmental Studies and Toxicology, Commission of Life Sciences, National Research Council. National Academy Press, Washington, DC, 2002.
    206) NRC: Acute Exposure Guideline Levels for Selected Airborne Chemicals - Volume 3, Subcommittee on Acute Exposure Guideline Levels, Committee on Toxicology, Board on Environmental Studies and Toxicology, Commission of Life Sciences, National Research Council. National Academy Press, Washington, DC, 2003.
    207) NRC: Acute Exposure Guideline Levels for Selected Airborne Chemicals - Volume 4, Subcommittee on Acute Exposure Guideline Levels, Committee on Toxicology, Board on Environmental Studies and Toxicology, Commission of Life Sciences, National Research Council. National Academy Press, Washington, DC, 2004.
    208) Nagasawa HT, Goon DJ, Crankshaw DL, et al: Novel, orally effective cyanide antidotes. J Med Chem 2007; 50(26):6462-6464.
    209) Naradzay J & Barish RA: Approach to ophthalmologic emergencies. Med Clin North Am 2006; 90(2):305-328.
    210) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for 1,2,3-Trimethylbenzene (Proposed). United States Environmental Protection Agency. Washington, DC. 2006k. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020d68a&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    211) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for 1,2,4-Trimethylbenzene (Proposed). United States Environmental Protection Agency. Washington, DC. 2006m. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020d68a&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    212) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for 1,2-Butylene Oxide (Proposed). United States Environmental Protection Agency. Washington, DC. 2008d. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648083cdbb&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    213) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for 1,2-Dibromoethane (Proposed). United States Environmental Protection Agency. Washington, DC. 2007g. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064802796db&disposition=attachment&contentType=pdf. As accessed 2010-08-18.
    214) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for 1,3,5-Trimethylbenzene (Proposed). United States Environmental Protection Agency. Washington, DC. 2006l. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020d68a&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    215) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for 2-Ethylhexyl Chloroformate (Proposed). United States Environmental Protection Agency. Washington, DC. 2007b. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648037904e&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    216) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Acrylonitrile (Proposed). United States Environmental Protection Agency. Washington, DC. 2007c. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648028e6a3&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    217) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Adamsite (Proposed). United States Environmental Protection Agency. Washington, DC. 2007h. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020fd29&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    218) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Agent BZ (3-quinuclidinyl benzilate) (Proposed). United States Environmental Protection Agency. Washington, DC. 2007f. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064803ad507&disposition=attachment&contentType=pdf. As accessed 2010-08-18.
    219) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Allyl Chloride (Proposed). United States Environmental Protection Agency. Washington, DC. 2008. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648039d9ee&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    220) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Aluminum Phosphide (Proposed). United States Environmental Protection Agency. Washington, DC. 2005b. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020c5ed&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    221) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Arsenic Trioxide (Proposed). United States Environmental Protection Agency. Washington, DC. 2007m. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480220305&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    222) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Automotive Gasoline Unleaded (Proposed). United States Environmental Protection Agency. Washington, DC. 2009a. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480a7cc17&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    223) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Biphenyl (Proposed). United States Environmental Protection Agency. Washington, DC. 2005j. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064801ea1b7&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    224) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Bis-Chloromethyl Ether (BCME) (Proposed). United States Environmental Protection Agency. Washington, DC. 2006n. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648022db11&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    225) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Boron Tribromide (Proposed). United States Environmental Protection Agency. Washington, DC. 2008a. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064803ae1d3&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    226) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Bromine Chloride (Proposed). United States Environmental Protection Agency. Washington, DC. 2007d. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648039732a&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    227) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Bromoacetone (Proposed). United States Environmental Protection Agency. Washington, DC. 2008e. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064809187bf&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    228) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Calcium Phosphide (Proposed). United States Environmental Protection Agency. Washington, DC. 2005d. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020c5ed&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    229) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Carbonyl Fluoride (Proposed). United States Environmental Protection Agency. Washington, DC. 2008b. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064803ae328&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    230) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Carbonyl Sulfide (Proposed). United States Environmental Protection Agency. Washington, DC. 2007e. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648037ff26&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    231) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Chlorobenzene (Proposed). United States Environmental Protection Agency. Washington, DC. 2008c. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064803a52bb&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    232) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Cyanogen (Proposed). United States Environmental Protection Agency. Washington, DC. 2008f. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064809187fe&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    233) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Dimethyl Phosphite (Proposed). United States Environmental Protection Agency. Washington, DC. 2009. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480a7cbf3&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    234) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Diphenylchloroarsine (Proposed). United States Environmental Protection Agency. Washington, DC. 2007l. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020fd29&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    235) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Ethyl Isocyanate (Proposed). United States Environmental Protection Agency. Washington, DC. 2008h. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648091884e&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    236) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Ethyl Phosphorodichloridate (Proposed). United States Environmental Protection Agency. Washington, DC. 2008i. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480920347&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    237) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Ethylbenzene (Proposed). United States Environmental Protection Agency. Washington, DC. 2008g. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064809203e7&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    238) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Ethyldichloroarsine (Proposed). United States Environmental Protection Agency. Washington, DC. 2007j. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020fd29&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    239) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Germane (Proposed). United States Environmental Protection Agency. Washington, DC. 2008j. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480963906&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    240) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Hexafluoropropylene (Proposed). United States Environmental Protection Agency. Washington, DC. 2006. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064801ea1f5&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    241) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Ketene (Proposed). United States Environmental Protection Agency. Washington, DC. 2007. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020ee7c&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    242) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Magnesium Aluminum Phosphide (Proposed). United States Environmental Protection Agency. Washington, DC. 2005h. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020c5ed&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    243) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Magnesium Phosphide (Proposed). United States Environmental Protection Agency. Washington, DC. 2005g. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020c5ed&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    244) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Malathion (Proposed). United States Environmental Protection Agency. Washington, DC. 2009k. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064809639df&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    245) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Mercury Vapor (Proposed). United States Environmental Protection Agency. Washington, DC. 2009b. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480a8a087&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    246) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Methyl Isothiocyanate (Proposed). United States Environmental Protection Agency. Washington, DC. 2008k. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480963a03&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    247) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Methyl Parathion (Proposed). United States Environmental Protection Agency. Washington, DC. 2008l. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480963a57&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    248) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Methyl tertiary-butyl ether (Proposed). United States Environmental Protection Agency. Washington, DC. 2007a. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064802a4985&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    249) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Methylchlorosilane (Proposed). United States Environmental Protection Agency. Washington, DC. 2005. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020c5f4&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    250) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Methyldichloroarsine (Proposed). United States Environmental Protection Agency. Washington, DC. 2007i. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020fd29&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    251) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Methyldichlorosilane (Proposed). United States Environmental Protection Agency. Washington, DC. 2005a. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020c646&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    252) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Nitrogen Mustard (HN1 CAS Reg. No. 538-07-8) (Proposed). United States Environmental Protection Agency. Washington, DC. 2006a. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020d6cb&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    253) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Nitrogen Mustard (HN2 CAS Reg. No. 51-75-2) (Proposed). United States Environmental Protection Agency. Washington, DC. 2006b. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020d6cb&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    254) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Nitrogen Mustard (HN3 CAS Reg. No. 555-77-1) (Proposed). United States Environmental Protection Agency. Washington, DC. 2006c. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020d6cb&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    255) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Nitrogen Tetroxide (Proposed). United States Environmental Protection Agency. Washington, DC. 2008n. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648091855b&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    256) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Nitrogen Trifluoride (Proposed). United States Environmental Protection Agency. Washington, DC. 2009l. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480963e0c&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    257) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Parathion (Proposed). United States Environmental Protection Agency. Washington, DC. 2008o. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480963e32&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    258) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Perchloryl Fluoride (Proposed). United States Environmental Protection Agency. Washington, DC. 2009c. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480a7e268&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    259) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Perfluoroisobutylene (Proposed). United States Environmental Protection Agency. Washington, DC. 2009d. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480a7e26a&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    260) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Phenyl Isocyanate (Proposed). United States Environmental Protection Agency. Washington, DC. 2008p. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648096dd58&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    261) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Phenyl Mercaptan (Proposed). United States Environmental Protection Agency. Washington, DC. 2006d. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020cc0c&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    262) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Phenyldichloroarsine (Proposed). United States Environmental Protection Agency. Washington, DC. 2007k. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020fd29&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    263) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Phorate (Proposed). United States Environmental Protection Agency. Washington, DC. 2008q. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648096dcc8&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    264) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Phosgene (Draft-Revised). United States Environmental Protection Agency. Washington, DC. 2009e. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480a8a08a&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    265) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Phosgene Oxime (Proposed). United States Environmental Protection Agency. Washington, DC. 2009f. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480a7e26d&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    266) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Potassium Cyanide (Proposed). United States Environmental Protection Agency. Washington, DC. 2009g. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480a7cbb9&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    267) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Potassium Phosphide (Proposed). United States Environmental Protection Agency. Washington, DC. 2005c. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020c5ed&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    268) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Propargyl Alcohol (Proposed). United States Environmental Protection Agency. Washington, DC. 2006e. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020ec91&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    269) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Selenium Hexafluoride (Proposed). United States Environmental Protection Agency. Washington, DC. 2006f. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020ec55&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    270) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Silane (Proposed). United States Environmental Protection Agency. Washington, DC. 2006g. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020d523&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    271) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Sodium Cyanide (Proposed). United States Environmental Protection Agency. Washington, DC. 2009h. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480a7cbb9&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    272) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Sodium Phosphide (Proposed). United States Environmental Protection Agency. Washington, DC. 2005i. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020c5ed&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    273) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Strontium Phosphide (Proposed). United States Environmental Protection Agency. Washington, DC. 2005f. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020c5ed&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    274) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Sulfuryl Chloride (Proposed). United States Environmental Protection Agency. Washington, DC. 2006h. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020ec7a&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    275) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Tear Gas (Proposed). United States Environmental Protection Agency. Washington, DC. 2008s. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648096e551&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    276) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Tellurium Hexafluoride (Proposed). United States Environmental Protection Agency. Washington, DC. 2009i. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480a7e2a1&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    277) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Tert-Octyl Mercaptan (Proposed). United States Environmental Protection Agency. Washington, DC. 2008r. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648096e5c7&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    278) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Tetramethoxysilane (Proposed). United States Environmental Protection Agency. Washington, DC. 2006j. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020d632&disposition=attachment&contentType=pdf. As accessed 2010-08-17.
    279) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Trimethoxysilane (Proposed). United States Environmental Protection Agency. Washington, DC. 2006i. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020d632&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    280) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Trimethyl Phosphite (Proposed). United States Environmental Protection Agency. Washington, DC. 2009j. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480a7d608&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    281) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Trimethylacetyl Chloride (Proposed). United States Environmental Protection Agency. Washington, DC. 2008t. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648096e5cc&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    282) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Zinc Phosphide (Proposed). United States Environmental Protection Agency. Washington, DC. 2005e. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020c5ed&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    283) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for n-Butyl Isocyanate (Proposed). United States Environmental Protection Agency. Washington, DC. 2008m. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064808f9591&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    284) National Heart,Lung,and Blood Institute: Expert panel report 3: guidelines for the diagnosis and management of asthma. National Heart,Lung,and Blood Institute. Bethesda, MD. 2007. Available from URL: http://www.nhlbi.nih.gov/guidelines/asthma/asthgdln.pdf.
    285) National Institute for Occupational Safety and Health: NIOSH Pocket Guide to Chemical Hazards, U.S. Department of Health and Human Services, Centers for Disease Control and Prevention, Cincinnati, OH, 2007.
    286) National Research Council : Acute exposure guideline levels for selected airborne chemicals, 5, National Academies Press, Washington, DC, 2007.
    287) National Research Council: Acute exposure guideline levels for selected airborne chemicals, 6, National Academies Press, Washington, DC, 2008.
    288) National Research Council: Acute exposure guideline levels for selected airborne chemicals, 7, National Academies Press, Washington, DC, 2009.
    289) National Research Council: Acute exposure guideline levels for selected airborne chemicals, 8, National Academies Press, Washington, DC, 2010.
    290) Naughton M: Acute cyanide poisoning. Anaesth Intensive Care 1974; 4:351-356.
    291) Nemec K: Antidotes in acute poisoning. Eur J Hosp Pharm Sci Pract 2011; 17(4):53-55.
    292) None Listed: Abstracts of the XXVIII International Congress of the European Association of Poison Centres and Clinical Toxicologists. May 6-9, 2008. Seville, Spain. Clin Toxicol (Phila) 2008; 46(5):351-421.
    293) None Listed: Position paper: cathartics. J Toxicol Clin Toxicol 2004; 42(3):243-253.
    294) Norris JC, Utley WA, & Hume AS: Mechanism of antagonizing cyanide-induced lethality by alpha-ketoglutaric acid. Toxicol 1990; 62:275-283.
    295) O'Brien B, Quigg C, & Leong T: Severe cyanide toxicity from 'vitamin supplements'. Eur J Emerg Med 2005; 12(5):257-258.
    296) Oluwole OSA, Onabolu AO, Cotgreave IA, et al: Incidence of endemic ataxic polyneuropathy and its relation to exposure to cyanide in a Nigerian community. J Neurol Neurosurg Psych 2003; 74:1417-1422.
    297) Patnaik P: A comprehensive guide to the hazardous properties of chemical substances, Van Nostrand Reinhold, New York, NY, 1992.
    298) Paulet G: Valeur et mecanisme d'action de l'oxygenotherapie dans le traitement de l'intoxication cyanhydrique. Arch Internat de Physiologie et de Biochimie 1955; 63:340-360.
    299) Peate WF: Work-related eye injuries and illnesses. Am Fam Physician 2007; 75(7):1017-1022.
    300) 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.
    301) Peddy SB, Rigby MR, & Shaffner DH: Acute cyanide poisoning. Pediatr Crit Care Med 2006; 7(1):79-82.
    302) Peden NR, Taha A, & McSorley PD: Industrial exposure to hydrogen cyanide: implications for treatment. Br Med J 1986; 293:538.
    303) Pollack MM, Dunbar BS, & Holbrook PR: Aspiration of activated charcoal and gastric contents. Ann Emerg Med 1981; 10:528-529.
    304) Prieto I, Pujol I, Santiuste C, et al: Acute cyanide poisoning by subcutaneous injection. Emerg Med J 2005; 22:389-390.
    305) Proctor NH, Hughes JP, & Fischman ML: Chemical Hazards of the Workplace, 2nd ed, JB Lippincott Co, Philadelphia, PA, 1988.
    306) Product Information: CEREBYX(R) intravenous injection, fosphenytoin sodium intravenous injection. Pfizer Labs (per FDA), New York, NY, 2014.
    307) Product Information: CYANOKIT(R) 2.5g IV injection, hydroxocobalamin IV injection. Merck Lipha Sante, Lyon, France, 2006.
    308) Product Information: Dilantin(R) intravenous injection, intramuscular injection, phenytoin sodium intravenous injection, intramuscular injection. Parke-Davis (per FDA), New York, NY, 2013.
    309) Product Information: NITHIODOTE intravenous injection solution, sodium nitrite intravenous injection solution and sodium thiosulfate intravenous injection solution. Hope Pharmaceuticals (per manufacturer), Scottsdale, AZ, 2011.
    310) Product Information: PROVAYBLUE(TM) intravenous injection, methylene blue intravenous injection. American Regent (per FDA), Shirley, NY, 2016.
    311) Product Information: diazepam IM, IV injection, diazepam IM, IV injection. Hospira, Inc (per Manufacturer), Lake Forest, IL, 2008.
    312) Product Information: dopamine hcl, 5% dextrose IV injection, dopamine hcl, 5% dextrose IV injection. Hospira,Inc, Lake Forest, IL, 2004.
    313) Product Information: lorazepam IM, IV injection, lorazepam IM, IV injection. Akorn, Inc, Lake Forest, IL, 2008.
    314) Product Information: methylene blue 1% IV injection, methylene blue 1% IV injection. American Regent, Inc (per manufacturer), Shirley, NY, 2011.
    315) Product Information: methylene blue 1% intravenous injection, methylene blue 1% intravenous injection. Akorn, Inc. (per manufacturer), Lake Forest, IL, 2011.
    316) Product Information: norepinephrine bitartrate injection, norepinephrine bitartrate injection. Sicor Pharmaceuticals,Inc, Irvine, CA, 2005.
    317) Product Information: sodium thiosulfate IV injection, sodium thiosulfate IV injection. American Regent Inc, Shirley, NY, 2003.
    318) Pronczuk de Garbino JP & Bismuth C: Propositions therapeutiques actuelles en cas d'intoxication par les cyanures. Toxicol Eur Res 1981; 3:69-76.
    319) RTECS : Registry of Toxic Effects of Chemical Substances. National Institute for Occupational Safety and Health. Cincinnati, OH (Internet Version). Edition expires 1991; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    320) RTECS : Registry of Toxic Effects of Chemical Substances. National Institute for Occupational Safety and Health. Cincinnati, OH (Internet Version). Edition expires 1993; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    321) Ram Z, Spiegelman R, & Findler G: Delayed postoperative neurological deterioration from prolonged sodium nitroprusside administration. J Neurosurg 1989; 71:605-607.
    322) Rau NR, Nagaraj MV, Prakash PS, et al: Fatal pulmonary aspiration of oral activated charcoal. Br Med J 1988; 297:918-919.
    323) Ravishankar DK, Kashi SH, & Lam FT: Organ transplantation from donor who died of cyanide poisoning: a case report. Clin Transplantation 1998; 12:142-143.
    324) Richmond J: HEW News, 1977.
    325) Riudavets MA, Aronica-Pollak P, & Troncoso JC: Pseudolaminar necrosis in cyanide intoxication. A neuropathology case report. Am J Forensic Med Pathol 2005; 26:189-191.
    326) Rosenberg NL, Myers JA, & Martin WRW: Cyanide-induced parkinsonism: clinical, MRI, and 6-fluorodopa PET studies. Neurology 1989; 39:142-144.
    327) Rosenow F, Herholz K, & Lanfermann H: Neurological sequelae of cyanide intoxication - the patterns of clinical, magnetic resonance imaging, and positron emission tomography findings. Ann Neurol 1995; 38:825-828.
    328) Saia B, DeRosa E, & Galzigna L: Considerations on chronic cyanide poisoning. Med Lav 1970; 62:580-586.
    329) Saincher A, Swirsky N, & Tenenbein: Survival after cyanide overdose with fatal levels treated only with supportive care (abstract). Vet Hum Toxicol 1992; 34:338.
    330) Sarikaya I, Apaydin H, Topal U, et al: Cyanide-induced parkinsonism and F-18 FDG PET/CT findings. Clin Nucl Med 2006; 31(6):363-364.
    331) Sax NI & Lewis RJ: Dangerous Properties of Industrial Materials, 7th ed, Van Nostrand Reinhold Company, New York, NY, 1989.
    332) Schardein JL: Chemically Induced Birth Defects, Marcel Dekker Inc, Inc, New York, 1985.
    333) Schmidt ES, Newton GW, & Sanders SM: Laetrile toxicity studies in dogs. JAMA 1978; 239:943-947.
    334) Schneider CR & Freund H: Determination of low level hydrocyanic acid in solution using gas-liquid chromatography. Anal Chem 1972; 34:69-74.
    335) Scolnick B, Hamel D, & Woolf AD: Successful treatment of life-threatening propionitrile exposure with sodium nitrite/sodium thiosulfate followed by hyperbaric oxygen. J Occup Environ Med 1993; 35:577-580.
    336) 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.
    337) Sheehy M & Way JL: Effect of oxygen on cyanide intoxication. III. Mithridate. J Pharmacol Exp Ther 1968; 161:163-168.
    338) Shepherd G & Keyes DC: Methylene blue. In: Dart,RC, ed. Medical Toxicology, 3rd ed. 3rd ed, Philadelphia, PA, 2004, pp -.
    339) Shepherd G & Velez LI: Role of hydroxocobalamin in acute cyanide poisoning. Ann Pharmacother 2008; 42(5):661-669.
    340) Shifrin NS, Beck BD, & Gauthier TD: Chemistry, toxicology and human health risk of cyanide compounds in soils at former manufactured gas plant sites. Regul Toxicol Pharmacol 1996; 23:106-118.
    341) Shragg TA, Albertson TE, & Fisher CJ: Cyanide poisoning after bitter almond ingestion. West J Med 1982; 136:65-69.
    342) Silverman SH, Purdue GF, & Hunt JL: Cyanide toxicity in burned patients. J Trauma 1988; 28:171-176.
    343) Singh BM, Coles N, & Lewis RA: The metabolic effects of fatal cyanide poisoning. Postgrad Med J 1989; 65:923-925.
    344) Singh JD: The teratogenic effects of dietary cassava on the pregnant albino rat: A preliminary report. Teratology 1981; 24:289-291.
    345) Skene WG, Norman JN, & Smith G: Effect of hyperbaric oxygen in cyanide poisoning. In Brown I, Cox B (Ed). Proceedings of the Third International Congress on hyperbaric oxygen, National Academy of Science, NRC, Washington, DC, 1966, pp 705-710.
    346) Smit MH & Rechnitz GA: Toxin detection using a tyrosinase-coupled oxygen electrode. Anal Chem 1993; 65:380-385.
    347) Spiller HA & Rogers GC: Evaluation of administration of activated charcoal in the home. Pediatrics 2002; 108:E100.
    348) 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.
    349) Stamyr K, Nord P, & Johanson G: Washout kinetics of inhaled hydrogen cyanide in breath. Toxicol Lett 2008; 179(1):59-62.
    350) Stanford SC , Stanford BJ , & Gillman PK : Risk of severe serotonin toxicity following co-administration of methylene blue and serotonin reuptake inhibitors: an update on a case report of post-operative delirium. J Psychopharmacol 2010; 24(10):1433-1438.
    351) Steffens W, Leng G, Bayer AG, et al: Percutaneous hydrocyanic acid poisoning (abstract). J Toxicol - Clin Toxicol 2003; 41(4):483.
    352) Stellpflug SJ, Gardner RL, Leroy JM, et al: Hydroxocobalamin hinders hemodialysis. Am J Kidney Dis 2013; 62(2):395-395.
    353) Stewart R: Cyanide poisoning. Clin Toxicol 1974; 5:561-564.
    354) 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.
    355) Sturm CD, Frisella WA, & Yoon KW: Attenuation of potassium cyanide-mediated neuronal cell death by adenosine. J Neurosurg 1993; 79:111-115.
    356) Takano T, Miyazaki Y, & Nashimoto I: Effect of hyperbaric oxygen on cyanide intoxication: in situ changes in intracellular oxidation reduction. Undersea Biomed Res 1980; 7:191-197.
    357) Ten Eyck RP, Schaerdel AD, & Ottinger WE: Stroma-free methemoglobin solution: an effective antidote for acute cyanide poisoning. Am J Emerg Med 1985; 3:519-523.
    358) Teunis BS, Leftwich EI, & Pierce LE: Acute methemoglobinemia and hemolytic anemia due to toluidine blue. Arch Surg 1970; 101:527-531.
    359) Thakore S & Murphy N: The potential role of prehospital administration of activated charcoal. Emerg Med J 2002; 19:63-65.
    360) Trapp W: Massive cyanide poisoning with recovery: a boxing day story. Can Med Assoc J 1970; 102:517.
    361) Trapp WG & Lepawsky M: 100% survival in five life-threatening acute cyanide poisoning victims treated by therapeutic spectrumm including hyperbaric oxygen, In: First European Conference on Hyperbaric Medicine, Amsterdam, The Netherlands, 1983.
    362) Tung A, Lynch J, & McDade WA: A new biological assay for measuring cyanide in blood. Anesth Analg 1997; 85:1045-1051.
    363) Turchen SG, Manoguerra AS, & Whitney C: Severe cyanide poisoning following suicidal ingestion of an acetonitrile-containing cosmetic. Am J Emerg Med 1991; 9:264-267.
    364) Tylleskar T, Legue FD, & Peterson S: Konzo in the Central African Republic. Neurology 1994; 44:959-961.
    365) Tyrer FH: Treatment of cyanide poisoning. J Soc Occup Med 1981; 31:65-66.
    366) U.S. Department of Energy, Office of Emergency Management: Protective Action Criteria (PAC) with AEGLs, ERPGs, & TEELs: Rev. 26 for chemicals of concern. U.S. Department of Energy, Office of Emergency Management. Washington, DC. 2010. Available from URL: http://www.hss.doe.gov/HealthSafety/WSHP/Chem_Safety/teel.html. As accessed 2011-06-27.
    367) U.S. Department of Health and Human Services, Public Health Service, National Toxicology Project : 11th Report on Carcinogens. U.S. Department of Health and Human Services, Public Health Service, National Toxicology Program. Washington, DC. 2005. Available from URL: http://ntp.niehs.nih.gov/INDEXA5E1.HTM?objectid=32BA9724-F1F6-975E-7FCE50709CB4C932. As accessed 2011-06-27.
    368) U.S. Environmental Protection Agency: Discarded commercial chemical products, off-specification species, container residues, and spill residues thereof. Environmental Protection Agency's (EPA) Resource Conservation and Recovery Act (RCRA); List of hazardous substances and reportable quantities 2010b; 40CFR(261.33, e-f):77-.
    369) U.S. Environmental Protection Agency: Integrated Risk Information System (IRIS). U.S. Environmental Protection Agency. Washington, DC. 2011. Available from URL: http://cfpub.epa.gov/ncea/iris/index.cfm?fuseaction=iris.showSubstanceList&list_type=date. As accessed 2011-06-21.
    370) U.S. Environmental Protection Agency: List of Radionuclides. U.S. Environmental Protection Agency. Washington, DC. 2010a. Available from URL: http://www.gpo.gov/fdsys/pkg/CFR-2010-title40-vol27/pdf/CFR-2010-title40-vol27-sec302-4.pdf. As accessed 2011-06-17.
    371) U.S. Environmental Protection Agency: List of hazardous substances and reportable quantities. U.S. Environmental Protection Agency. Washington, DC. 2010. Available from URL: http://www.gpo.gov/fdsys/pkg/CFR-2010-title40-vol27/pdf/CFR-2010-title40-vol27-sec302-4.pdf. As accessed 2011-06-17.
    372) U.S. Environmental Protection Agency: The list of extremely hazardous substances and their threshold planning quantities (CAS Number Order). U.S. Environmental Protection Agency. Washington, DC. 2010c. Available from URL: http://www.gpo.gov/fdsys/pkg/CFR-2010-title40-vol27/pdf/CFR-2010-title40-vol27-part355.pdf. As accessed 2011-06-17.
    373) U.S. Food and Drug Administration: FDA Drug Safety Communication: Serious CNS reactions possible when methylene blue is given to patients taking certain psychiatric medications. U.S. Food and Drug Administration. Silver Spring, MD. 2011. Available from URL: http://www.fda.gov/Drugs/DrugSafety/ucm263190.htm. As accessed 2011-07-26.
    374) U.S. Occupational Safety and Health Administration: Part 1910 - Occupational safety and health standards (continued) Occupational Safety, and Health Administration's (OSHA) list of highly hazardous chemicals, toxics and reactives. Subpart Z - toxic and hazardous substances. CFR 2010 2010; Vol6(SEC1910):7-.
    375) U.S. Occupational Safety, and Health Administration (OSHA): Process safety management of highly hazardous chemicals. 29 CFR 2010 2010; 29(1910.119):348-.
    376) Uitti RJ, Rajput AH, & Ashenhurst EM: Cyanide-induced parkinsonism: a clinicopathologic report. Neurology 1985; 35:921-925.
    377) United States Environmental Protection Agency Office of Pollution Prevention and Toxics: Acute Exposure Guideline Levels (AEGLs) for Vinyl Acetate (Proposed). United States Environmental Protection Agency. Washington, DC. 2006. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020d6af&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    378) Vale JA, Kulig K, American Academy of Clinical Toxicology, et al: Position paper: Gastric lavage. J Toxicol Clin Toxicol 2004; 42:933-943.
    379) Vale JA: Position Statement: gastric lavage. American Academy of Clinical Toxicology; European Association of Poisons Centres and Clinical Toxicologists. J Toxicol Clin Toxicol 1997; 35:711-719.
    380) Valenzuela R, Court J, & Godoy J: Delayed cyanide induced dystonia. J Neurol Neurosurg Psychiatr 1992; 55:198-199.
    381) Varnell RM, Stimac GK, & Fligner CL: CT diagnosis of toxic brain injury in cyanide poisoning:considerations for forensic medicine. AJNR Am J Neuroradiol 1987; 8(6):1063-1066.
    382) Vick JA & Froehlich HL: Studies of cyanide poisoning. Arch Int Pharmacodyn 1985; 273:314-322.
    383) Vogel SN, Sultan TR, & Ten Eyck RP: Cyanide poisoning. Clin Toxicol 1981; 18:367-383.
    384) Wallace KL: Toxin-Induced Seizures. In: Brent J, Wallace KL, Burkhart KK, et al, eds. Critical Care Toxicology, Elsevier Mosby, Philadelphia, PA, 2005.
    385) Walton DC & Witherspoon MG: Skin absorption of certain gases. J Pharmacol Exp Ther 1926; 26:315-324.
    386) Way JL, End E, & Sheehy MH: Effect of oxygen on cyanide intoxication. IV. Hyperbaric oxygen. Toxicol Appl Pharmacol 1972; 22:415-421.
    387) Weger NP: Treatment of cyanide poisoning with 4-dimethylaminophenol (DMAP): Experimental and clinical overview. Middle East J Anesth 1990; 10:389-412.
    388) Weng TI, Fang CC, & Chen WJ: Elevated plasma cyanide level after hydroxocobalamin infusion for cyanide poisoning. Am J Emerg Med 2004; 22:492-493.
    389) Wesson DE, Foley R, & Sabatini S: Treatment of acute cyanide intoxication with hemodialysis. Am J Nephrol 1985; 5:121-126.
    390) Willhite CC, Ferm VH, & Smith RP: Teratogenic effects of aliphatic nitriles. Teratology 1981; 23:317-323.
    391) Willhite CC: Congenital malformations induced by laetrile. Science 1982; 215:1513-1515.
    392) Willhite CC: Developmental toxicology of acetonitrile in the Syrian golden hamster. Teratology 1983; 27:313-325.
    393) Williams CL: An unusual case of cyanide poisoning during fumigation. Publ Health Rep 1938; 53:2094-2095.
    394) Willson DF, Truwit JD, Conaway MR, et al: The adult calfactant in acute respiratory distress syndrome (CARDS) trial. Chest 2015; 148(2):356-364.
    395) 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.
    396) Winek CL, Collom WD, & Martineau P: Toluidine blue intoxication. Clin Toxicol 1969; 2:1-3.
    397) Wolnik KA, Fricke FL, & Bonnin E: The Tylenol tampering incident-tracing the source. Anal Chem 1984; 56:466-468-470&474.
    398) Woo OF & Pouw TH: Cyanide tampered Sudafed 12 Hour capsules: survival with lethal level treated with supportive care. Vet Hum Toxicol 1993; 35:346.
    399) Wright IH & Vesey CJ: Acute poisoning with gold cyanide. Anaesthesia 1986; 41:936-939.
    400) Yacoub M, Faure J, & Morena H: L'intoxication cyanhydrique aigue: Donnees actuelles sur le metabolisme du cyanure et le traitement par l'hydroxocobalamine. J Eur Toxicol 1974; 7:22-29.
    401) Yamamoto HA: Protection against cyanide-induced convulsions with alpha-ketoglutarate. Toxicol 1990; 61:221-228.
    402) Yoshida M, Watabiki T, & Ishida N: The quantitative determination of cyanide by FTD-GC. Nippon Hoigaku Zasshi 1989; 40:179-185.
    403) Zakharov S, Vaneckova M, Seidl Z, et al: Successful Use of Hydroxocobalamin and Sodium Thiosulfate in Acute Cyanide Poisoning: A Case Report with Follow-up. Basic Clin Pharmacol Toxicol 2015; 117(3):209-212.
    404) Zaknun JJ, Stieglbauer K, Trenkler J, et al: Cyanide-induced akinetic rigid syndrome: clinical, MRI, FDG-PET, beta-CIT and HMPAO SPECT findings. Parkinsonism Relat Disord 2005; 11:125-129.
    405) Zamecnik J & Tam J: Cyanide in blood by gas chromatography with NP detector and acetonitrile as internal standard: application on air accident fire victims. J Anal Toxicol 1987; 11:47-48.
    406) Zijlstra WG & Buursma A: Rapid multicomponent analysis of hemoglobin derivatives for controlled antidotal use of methemoglobin-forming agents in cyanide poisoning. Clin Chem 1993; 39:1685-1689.
    407) do Nascimento TS, Pereira RO, de Mello HL, et al: Methemoglobinemia: from diagnosis to treatment. Rev Bras Anestesiol 2008; 58(6):651-664.
    408) van Dijk A, Douze JMC, & van Heijst ANP: Clinical evaluation of the cyanide antagonist 4-DMAP. (Abstract), II World Congress of the World Federation of Associations of Clinical Toxicology and Poison Control Centers, Brussels, Belgium, 1986.
    409) van Heijst ANP, Douze JMC, & van Kasteren RG: Therapeutic problems in cyanide poisoning. Clin Toxicol 1987; 25:383-398.
    410) van Heijst ANP, Maas RAA, & Mtanda AT: Chronic cyanide poisoning in relation to blindness and tropical neuropathy. J Toxicol Clin Toxicol 1994; 32:549-556.