MOBILE VIEW  | 

LINDANE

Classification   |    Detailed evidence-based information

Therapeutic Toxic Class

    A) Lindane is an organochlorine insecticide in the hexachlorocyclohexane family. It is the most active, effective and toxic isomer of benzene hexachloride (BHC).

Specific Substances

    1) 1,2,3,4,5,6-hexachlorocyclohexane
    2) Benhexachlor
    3) Benzene Hexachloride, Gamma
    4) Gamma Benzene Hexachloride
    5) Gamma BHC
    6) Gamma HCH
    7) HCH
    8) Lindane
    9) CAS 58-89-9
    10) BHC (LINDANE)
    11) DRILL TOX-SPEZIAL AGLUKON
    12) GAMMA HCH (LINDANE)
    13) HCH (LINDANE)
    14) HEXACHLOROCYCLOHEXANE
    15) LINDAN
    1.2.1) MOLECULAR FORMULA
    1) C6-H6-Cl6

Available Forms Sources

    A) FORMS
    1) Lindane has been described as existing in the following forms: crystals, off-white powder, yellow to white crystalline powder, or as "needles from alcohol." Some sources report that the compound is odorless; others describe its odor as a slightly persistent musty or aromatic smell(HSDB, 2004; NIOSH , 2000; Budavari, 1996; Clayton & Clayton, 1994) .
    2) Humans use lindane primarily as a scabicide and pediculocide. When lindane is used for the treatment of scabies, it is usually in a 1% cream or ointment form, but may also be formulated as an emulsion, solution, aerosol, lotion, cream, or shampoo. It is also used to treat human head and body lice (ATSDR, 2003; Hayes & Laws, 1991).
    B) SOURCES
    1) Lindane can be manufactured by selectively crystallizing or using chromatographic adsorption on crude benzene hexachloride treated with methanol or acetic acid (HSDB, 2004).
    2) Methanol is the most common solvent used to extract lindane from HCH. Nitric acid is then used in the process for odor removal (HSDB, 2004).
    3) Lindane can be produced by combining benzene with chlorine and using photochlorination/isomer separation (Ashford, 1994).
    C) USES
    1) Lindane is used as an insecticide in a wide variety of applications, although its use in the US is restricted (Clayton & Clayton, 1994). Restrictions may be State dependent. For instance, a complete ban on the use or sale of lindane-containing products for the treatment of lice or scabies took effect in California in January of 2002 (Forrester et al, 2004).
    2) Some of the restricted applications include plant nurseries, livestock sprays, seed treatment, pet shampoos, household sprays, flea collars, and the treatment of hardwood logs. Treatment of food with lindane is limited to avocados and pecans (Clayton & Clayton, 1994).
    3) Lotions, shampoos, and creams containing lindane can be used on humans to treat lice and scabies (Clayton & Clayton, 1994).
    4) Lindane has been used on termites, lice, mosquitoes, and flies that can no longer be controlled by DDT (Clayton & Clayton, 1994).
    5) As per the EPA, lindane may no longer be registered for use in vaporizers, on many food crops, and in the dairy industry. It is no longer produced in the US (ACGIH, 1991) Extoxnet, 2000).
    6) Certain applications of lindane may only be conducted by certified applicators (ATSDR, 2003).
    7) Lindane is used as a therapeutic scabicide/pesticide in humans and animals (ACGIH, 1991; Budavari, 1996).

Life Support

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

Clinical Effects

    0.2.1) SUMMARY OF EXPOSURE
    A) USES: Lindane is an organochlorine insecticide most often used by topical application to treat body lice or scabies infestations, but has become second line treatment because safer, more effective agents have been developed.
    B) PHARMACOLOGY: Presumably it is a CNS stimulant in arthropods causing seizures and death.
    C) TOXICOLOGY: Lindane is believed to act as an indirect GABA antagonist, resulting in CNS stimulation and seizures.
    D) EPIDEMIOLOGY: Uncommon exposure that can result in significant morbidity and death, especially in children.
    E) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: Vomiting, headache, nausea, dizziness, diarrhea, and tremor.
    2) SEVERE TOXICITY: Seizures, CNS depression, agitation, ataxia, hypotension, respiratory depression, metabolic acidosis, and coma. Children appear to be more susceptible to the toxic effects of lindane and more likely to manifest severe effects.
    0.2.10) GENITOURINARY
    A) WITH POISONING/EXPOSURE
    1) Renal insufficiency and myoglobinuria secondary to rhabdomyolysis associated with prolonged seizures have been described.
    0.2.20) REPRODUCTIVE
    A) Lindane did not have reproductive effects in a 3-generation rat study. It has not been teratogenic in animals. It disturbed the estrus cycle and reduced fertility in female rats. It has been detected in human breast milk. It produced degenerative changes in the testes of rats.
    0.2.21) CARCINOGENICITY
    A) Several cases of acute myeloid leukemia have been reported with lindane exposure. Lindane has been carcinogenic in mice, inducing liver tumors.

Laboratory Monitoring

    A) Serum levels are not readily available or clinically useful for guiding management.
    B) Monitor serum electrolytes and blood glucose.
    C) Monitor creatinine kinase if seizures are prolonged.
    D) Obtain venous or arterial blood gas if acidosis is a concern.
    E) An EEG may be needed to rule out status epilepticus or guide aggressive anticonvulsive therapy.

Treatment Overview

    0.4.2) ORAL/PARENTERAL EXPOSURE
    A) MANAGEMENT OF MILD TO MODERATE TOXICITY
    1) Adequate decontamination of dermal exposures and supportive care are the mainstays of treatment for mild to moderate exposures. Seizures, respiratory depression, and CNS depression indicate a more severe poisoning.
    B) MANAGEMENT OF SEVERE TOXICITY
    1) Following adequate airway, respiratory, and circulatory support, treatment of seizures is standard anticonvulsant therapy with benzodiazepines as first line therapy followed by phenobarbital. Propofol infusions and/or neuromuscular blockade may be necessary in refractory cases. Phenytoin should not be used as it likely to be of minimal effectiveness. Severe metabolic acidosis can be treated with sodium bicarbonate. After stabilization remove contaminated clothing and decontaminate the skin with soap and water if there has been dermal exposure.
    C) DECONTAMINATION
    1) PREHOSPITAL: Prehospital oral decontamination should be avoided because of the risk of rapid onset CNS depression and seizures and subsequent aspiration.
    2) HOSPITAL: Generally, gastrointestinal decontamination should be avoided because of the potential for rapid onset CNS depression and seizures with subsequent aspiration.
    D) AIRWAY MANAGEMENT
    1) Patients who are comatose, seizing or with altered mental status may need tracheal intubation and mechanical respiratory support.
    E) ANTIDOTE
    1) None
    F) SEIZURES
    1) IV benzodiazepines, barbiturates.
    G) ENHANCED ELIMINATION
    1) Dialysis is likely to be of minimal benefit because of the large volume of distribution.
    H) PATIENT DISPOSITION
    1) HOME CRITERIA: Adults who intentionally ingest lindane or any child with an ingestion should be referred to a health care facility. Patients with prolonged or repeated dermal applications should be referred to a health care facility if they become symptomatic.
    2) OBSERVATION CRITERIA: After ingestion, patients who are asymptomatic after 6 hours can be discharged home.
    3) ADMISSION CRITERIA: Patients with persistently altered mental status, abnormal vital signs, or recurrent seizures should be admitted.
    4) CONSULT CRITERIA: Consult a poison center or medical toxicologist for assistance in managing severe poisonings.
    I) PITFALLS
    1) Phenytoin is likely to be of minimal effectiveness for seizure control.
    J) PHARMACOKINETICS
    1) Lindane can be absorbed via inhalation, ingestion, or by dermal exposure. Following ingestion, the half-life is several days, and after dermal application half life is 18 to 21 hours. The volume of distribution is not completely known but is considered to be large.
    K) TOXICOKINETICS
    1) Prolonged application, repeated applications, occlusive dressings, broken skin or chronic skin conditions will all increase absorption. Children absorb more after dermal application because of increased body surface area relative to mass and greater skin permeability. CNS toxicity generally develops within 15 to 120 minutes of ingestion.
    L) DIFFERENTIAL DIAGNOSIS
    1) The differential diagnosis is any condition that presents with seizures and hence is quite broad.
    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) 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).
    C) First responders should wear protective clothing to prevent secondary contamination. Be prepared to collect any vomitus or excreta in a manner to prevent further contamination; bag and treat as hazardous waste.
    D) Do not administer adrenergic amines, which may further increase myocardial irritability and produce refractory ventricular arrhythmias.
    E) Dialysis, exchange transfusion, and hemoperfusion are probably ineffective.
    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.
    B) 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).
    C) Carefully observe patient for development of clinical signs and symptoms, and administer treatment as described in DERMAL EXPOSURE where appropriate.
    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) 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).
    3) First responders should wear protective clothing to prevent secondary contamination. Be prepared to collect any vomitus or excreta in a manner to prevent further contamination; bag and treat as hazardous waste.
    4) ACUTE LUNG INJURY: Maintain ventilation and oxygenation and evaluate with frequent arterial blood gases and/or pulse oximetry monitoring. Early use of PEEP and mechanical ventilation may be needed.
    5) Do not give oils by mouth.
    6) Do not administer adrenergic amines, which may further increase myocardial irritability and produce refractory ventricular arrhythmias.
    7) CHOLESTYRAMINE - Oral administration may enhance the excretion of kepone and chlordane which are trapped in the enterohepatic circulation.
    8) Dialysis, exchange transfusion, and hemoperfusion are probably ineffective.

Range Of Toxicity

    A) TOXICITY: Most toxicity occurs after repeated or prolonged applications, but can also occur after ingestions and has also been reported after a single proper dermal application. Ingestion of as little as 5 mL of 1% lindane has caused seizures in a child. In an adult, ingestion of 45 mg caused seizures. An adult survived an ingestion of 1035 g.
    B) THERAPEUTIC DOSE: Typically applied as a 1% formulation to the hair or body; 30 mL is generally sufficient for an adult. The lotion is left on the skin for 8 to 12 hours and then washed off.

Summary Of Exposure

    A) USES: Lindane is an organochlorine insecticide most often used by topical application to treat body lice or scabies infestations, but has become second line treatment because safer, more effective agents have been developed.
    B) PHARMACOLOGY: Presumably it is a CNS stimulant in arthropods causing seizures and death.
    C) TOXICOLOGY: Lindane is believed to act as an indirect GABA antagonist, resulting in CNS stimulation and seizures.
    D) EPIDEMIOLOGY: Uncommon exposure that can result in significant morbidity and death, especially in children.
    E) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: Vomiting, headache, nausea, dizziness, diarrhea, and tremor.
    2) SEVERE TOXICITY: Seizures, CNS depression, agitation, ataxia, hypotension, respiratory depression, metabolic acidosis, and coma. Children appear to be more susceptible to the toxic effects of lindane and more likely to manifest severe effects.

Vital Signs

    3.3.2) RESPIRATIONS
    A) WITH POISONING/EXPOSURE
    1) Respiratory depression secondary to seizures is common with severe lindane intoxications (Proctor et al, 1988; Aks et al, 1995; Nordt & Chew, 2000).
    3.3.3) TEMPERATURE
    A) WITH POISONING/EXPOSURE
    1) FEVER occurred in a fatal case of endrin ingestion and may occur with lindane intoxications (Runhaar et al, 1985).
    3.3.4) BLOOD PRESSURE
    A) WITH POISONING/EXPOSURE
    1) Recurrent hypotension occurred in a fatal case of endrin ingestion (Runhaar et al, 1985).

Heent

    3.4.3) EYES
    A) WITH THERAPEUTIC USE
    1) Conjunctivitis has been caused by the use of ointments and lotions containing 0.1% to 1% lindane applied to eyelashes (Post & Juhlin, 1963). It is effective against crab lice in the eyelashes and does not cause excessive irritation when it is washed off within a few minutes after application (Grant & Schuman, 1993).
    2) Visual changes developed in 2 patients following therapeutic use of lindane (Nolan et al, 2012).
    B) WITH POISONING/EXPOSURE
    1) Optic atrophy and blindness have been described following systemic lindane toxicity in one patient (Danopoulos et al, 1953).
    2) AMBLYOPIA has been reported (Lee et al, 1976).
    3) Lindane can irritate the eyes (EPA, 1985).
    4) CASE REPORT: Papilledema and other components of pseudotumor cerebri were seen in a 45-year-old man with a 30-year history of using 20% solutions of lindane. A direct cause-effect relationship could not be proved (Verderber et al, 1991).
    3.4.5) NOSE
    A) WITH POISONING/EXPOSURE
    1) Lindane can irritate the nose (EPA, 1985).
    3.4.6) THROAT
    A) WITH POISONING/EXPOSURE
    1) Lindane can irritate the throat (EPA, 1985).

Cardiovascular

    3.5.2) CLINICAL EFFECTS
    A) CONDUCTION DISORDER OF THE HEART
    1) WITH POISONING/EXPOSURE
    a) High concentrations of organochlorine insecticides can cause cardiac dysrhythmias by increasing myocardial irritability (Morgan, 1993).
    b) Inhalation of lindane-hydrocarbon mixtures may result in ventricular dysrhythmias due to sensitization of the myocardium to circulating catecholamines; halogenated hydrocarbons have been most frequently implicated (Kulig & Rumack, 1981).
    c) CASE REPORT: A 56-year-old man vomited about 6 ounces (about 177 mL) of a white liquid about 30 minutes after ingesting about 12 ounces (about 355 mL) of an insecticide containing 20% lindane. Despite supportive therapy, including gastric lavage and activated charcoal, his mental status deteriorated, necessitating intubation about 75 minutes after ingestion. He experienced multiple seizures (a focal seizure followed by 3 brief generalized tonic-clonic seizures) from 3 to 14 hours postingestion and received diazepam, phenobarbital, and phenytoin. His arterial blood pH was 6.76 after the focal seizure and 7.05 after the first generalized seizure and he received sodium bicarbonate that corrected the pH to 7.34. He was extubated about 43 hours after ingestion, but he developed premature ventricular contractions and premature atrial contractions on day 3 and atrial fibrillation/flutter on day 5. Following supportive therapy, including IV lidocaine, digoxin, and IV verapamil, his condition resolved. On day 6, he developed ataxia, wide-based gait, slurred speech, paranoia, and depression with defects in higher mental functioning. On day 12, he still had neurological abnormalities (eg, poor memory, loss of task focus, inadequate mentation, and defects in fine motor coordination). At this time, he committed suicide by cutting his femoral artery. Toxicokinetics analysis revealed an estimated distribution half-life of 10.3 hours and a terminal half-life of 162.9 hours, which is much longer than the previously reported terminal half-life of 25 to 36 hours (Wiles et al, 2015).
    B) ELECTROCARDIOGRAM ABNORMAL
    1) WITH POISONING/EXPOSURE
    a) Based on reports of occupational exposure to lindane, ECG abnormalities were reported in 15% of 45 workers manufacturing technical-grade HCH (ATSDR, 2003).
    C) HYPOTENSIVE EPISODE
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: A 66-year-old man with scabies developed acute mental status changes, seizures, hypoxemia, severe respiratory acidosis, and hypotension (BP 73/48 mmHg; 11 hours after application) after a single application of a thin film of lindane (1%) to his skin from the neck to the toes. Prior to the lindane administration, fresh scratch marks were noted throughout the patient's body which might have increased dermal absorption of lindane significantly. In addition, the physician's order did not include instructions to wash lindane from the skin after its application. All workup for bacteremia, sepsis, and meningitis were unremarkable. An EEG showed severe generalized cerebral dysfunction. He experienced infectious complications and died 50 days after admission. During autopsy, he was diagnosed with hypoxic ischemic encephalopathy from lindane poisoning (Sudakin, 2007).
    D) TACHYCARDIA
    1) WITH THERAPEUTIC USE
    a) Tachycardia has been reported following lindane use (Nolan et al, 2012).

Respiratory

    3.6.2) CLINICAL EFFECTS
    A) DYSPNEA
    1) WITH POISONING/EXPOSURE
    a) Dyspnea has been reported from ingestion of lindane (Nolan et al, 2012; Lewis, 1996).
    B) HYPOXEMIA
    1) WITH THERAPEUTIC USE
    a) CASE REPORT - A 66-year-old man with scabies developed acute mental status changes, seizures, hypoxemia, severe respiratory acidosis, and hypotension approximately 8 hours after a single application of a thin film of lindane (1%) to his skin from the neck to the toes. Prior to the lindane administration, fresh scratch marks were noted throughout the patient's body which might have increased dermal absorption of lindane significantly. In addition, the physician's order did not include instructions to wash lindane from the skin after its application. All workup for bacteremia, sepsis, and meningitis were unremarkable. An EEG showed severe generalized cerebral dysfunction. He experienced infectious complications and died 50 days after admission. During autopsy, he was diagnosed with hypoxic ischemic encephalopathy from lindane poisoning (Sudakin, 2007).
    2) WITH POISONING/EXPOSURE
    a) Severe seizures can limit pulmonary gas exchange, and this may be an immediate cause of death (Morgan, 1993).
    C) HYPOVENTILATION
    1) WITH POISONING/EXPOSURE
    a) Hypoventilation necessitating mechanical ventilation has been reported in a 4-year-old child following oral ingestion of 5 mL. No overt tonic-clonic activity was present, although brief eye deviation was noted. Pulse oximetry was measured at 90 percent (Nordt & Chew, 2000).
    D) ACUTE LUNG INJURY
    1) WITH POISONING/EXPOSURE
    a) Pulmonary edema has occurred from lindane exposure, especially in children and where commercial formulations are in hydrocarbon solvents (Jaeger et al, 1984; EPA, 1985).
    E) PNEUMONITIS
    1) WITH POISONING/EXPOSURE
    a) Aspiration of petroleum distillate solvent is likely to cause a hydrocarbon pneumonitis, which is potentially fatal.

Neurologic

    3.7.2) CLINICAL EFFECTS
    A) SEIZURE
    1) WITH THERAPEUTIC USE
    a) In a review article, 67 cases of severe adverse effects (AE), including 20 death (including 7 children and 4 patients aged 65 and older) following the labeled use (47 cases), excessive use (17 patients), and ingestion (3 cases) of lindane were identified. Seizures and respiratory problems developed in the majority of patients who developed severe AEs after using the labeled dose (defined as one 4-minute application of shampoo or one 8 to 12 hours of single total body application of lindane lotion 1%) (Nolan et al, 2012).
    b) CASE REPORT: A 38-year-old man with HIV, scabies, seborrheic dermatitis and tinea cruris developed status epilepticus 2 hours after 2 ounces of 1% lindane lotion was applied to his skin (Solomon et al, 1995). His HIV disease (potentially increased percutaneous absorption due to his skin disease) and coadministration of chlorpromazine were felt to have predisposed him to toxicity after therapeutic use.
    c) CASE REPORT: A 66-year-old man with scabies developed acute mental status changes, repeated seizures, hypoxemia, respiratory acidosis, and hypotension approximately 8 hours after a single application of a thin film of lindane (1%) to his skin from the neck to the toes. Prior to the lindane administration, fresh scratch marks were noted throughout the patient's body which might have increased dermal absorption of lindane significantly. In addition, the physician's order did not include instructions to wash lindane from the skin after its application. All workup for bacteremia, sepsis, and meningitis were unremarkable. An EEG showed severe generalized cerebral dysfunction. He experienced infectious complications and died 50 days after admission. During autopsy, he was diagnosed with hypoxic ischemic encephalopathy from lindane poisoning (Sudakin, 2007).
    2) WITH POISONING/EXPOSURE
    a) Lindane is a CNS excitant and convulsant. Accidental ingestion has caused fatalities. Both topical exposure and ingestion have resulted in seizures (Bhalla & Thami, 2004; Ryan et al, 1998; Zilker et al, 1999; Nordt & Chew, 2000). Effects in severe poisoning were repeated, violent, clonic seizures, sometimes superimposed on a continuous tonic spasm; onset can be rapid.
    b) Respiratory difficulty and cyanosis secondary to seizures may occur; coma and respiratory depression may ensue (Morgan, 1993).
    c) Most patients who survive recover completely over 1 to 3 days, usually within 24 hours (EPA, 1985); protracted illness is rare (Hathaway et el, 1996).
    d) Persons at highest risk for development of seizures include young children and elderly patients, who may rarely develop neurotoxicity following therapeutic topical doses (Tenenbein, 1991). Most reports of seizures have involved ingestions or inappropriate topical use of lindane.
    e) Grand mal seizures occurred in persons who had eaten benzene hexachloride-contaminated food (Nag et al, 1977; Nantel et al, 1977).
    f) CASE REPORT: A 56-year-old man vomited about 6 ounces (about 177 mL) of a white liquid about 30 minutes after ingesting about 12 ounces (about 355 mL) of an insecticide containing 20% lindane. Despite supportive therapy, including gastric lavage and activated charcoal, his mental status deteriorated, necessitating intubation about 75 minutes after ingestion. He experienced multiple seizures (a focal seizure followed by 3 brief generalized tonic-clonic seizures) from 3 to 14 hours postingestion and received diazepam, phenobarbital, and phenytoin. His arterial blood pH was 6.76 after the focal seizure and 7.05 after the first generalized seizure and he received sodium bicarbonate that corrected the pH to 7.34. He was extubated about 43 hours after ingestion, but he developed premature ventricular contractions and premature atrial contractions on day 3 and atrial fibrillation/flutter on day 5. Following supportive therapy, including IV lidocaine, digoxin, and IV verapamil, his condition resolved. On day 6, he developed ataxia, wide-based gait, slurred speech, paranoia, and depression with defects in higher mental functioning. On day 12, he still had neurological abnormalities (eg, poor memory, loss of task focus, inadequate mentation, and defects in fine motor coordination). At this time, he committed suicide by cutting his femoral artery. Toxicokinetics analysis revealed an estimated distribution half-life of 10.3 hours and a terminal half-life of 162.9 hours, which is much longer than the previously reported terminal half-life of 25 to 36 hours (Wiles et al, 2015).
    g) CASE REPORT: A 3-year-old boy developed generalized seizures in his sleep with transient loss of sensorium about 1 hour after ingesting 10 mL of lindane lotion. He presented with drowsiness, a GCS (Glasgow coma score) of 10/15, and papular skin lesions. Following supportive care, he recovered 24 hours later (Ramabhatta et al, 2014).
    h) CASE REPORT: A 6-year-old girl developed generalized seizures lasting 5 to 10 minutes after ingesting an unknown amount of lindane lotion. Following supportive care, he recovered 24 hours later (Ramabhatta et al, 2014).
    i) CASE REPORT: Starr & Clifford (1972) reported grand mal seizures and cyanosis in a 2-year-old female following ingestion of approximately 1.5 grams lindane from an insecticide pellet. Bilateral deep tendon reflexes were increased following seizure activity (Starr & Clifford, 1972).
    j) CASE REPORT: A 13-month-old boy sustained two generalized tonic clonic seizures after ingesting an unknown amount of lindane lotion (Aks et al, 1995).
    k) CASE REPORT: A 2-year-old boy developed what appeared to be petit mal seizures (staring blindly into space without responding to external stimuli) after ingesting one teaspoon of lindane lotion (Aks et al, 1995).
    l) CASE REPORT: A 9-month-old infant developed seizures following accidental exposure to a 20% lindane insecticide which had been applied a day previously to a wooden deck where she was playing (Ryan et al, 1998).
    m) CASE REPORT - A 3-year-old boy ingested approximately 1 teaspoon of 1% lindane shampoo. His mother induced vomiting in the boy twice. One hour after ingestion, he developed a tonic-clonic seizure lasting 4 to 5 minutes. He was discharged home from the ED after 3 hours of observation (Anon, 2005).
    n) CASE REPORT: A 3.5-year-old boy developed lethargy, irritability, tremors, nausea, vomiting, mild metabolic acidosis, and general clonic-tonic seizures lasting about 2 minutes after ingesting an unknown quantity of 35% lindane solution. An EEG showed a typical 3/sec spike and generalized wave discharge. Following supportive treatment, his symptoms improved gradually; he was discharged home 42 hours post-ingestion (Lifshitz & Gavrilov, 2002).
    B) PSYCHOMOTOR AGITATION
    1) WITH POISONING/EXPOSURE
    a) Agitation, tremors, restlessness, muscle spasms, and nervousness can occur with lindane intoxication and are common (Nolan et al, 2012; Lifshitz & Gavrilov, 2002; EPA, 1985; Fischer, 1994; Jaeger et al, 1984; Diaz, 1998; Zilker et al, 1999).
    C) DYSKINESIA
    1) WITH POISONING/EXPOSURE
    a) Cogwheel rigidity with no other signs of extrapyramidal disease, other than weakness, occurred in persons exhibiting hexachlorobenzene-induced porphyria (Peters et al, 1982). Myotonia may also be seen (Peters et al, 1982).
    D) ELECTROENCEPHALOGRAM ABNORMAL
    1) WITH POISONING/EXPOSURE
    a) Sensory disturbances, excitation with myoclonic jerking (Morgan, 1993), seizures, tremor, ataxia, agitation, nervousness, and amnesia may occur in association with EEG changes. EEG changes occurred in the absence of seizures in 15 of 17 persons occupationally exposed to lindane (Czegledi-Janko & Avar, 1970).
    b) ANIMAL STUDIES - In anaesthetized rats, lindane (20 mg/kg) elicited dose-dependent epileptic spikes in the hippocampus, along with rhythmic activity at 4 to 5 Hz (Nyitrai et al, 2002).
    E) BENIGN INTRACRANIAL HYPERTENSION
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT - A 45-year-old man with a 30-year history of using 20 percent solutions of lindane developed a CSF pressure of 400 mm and pseudotumor cerebri. A direct cause-effect relationship could not be proven. Papilledema was seen in this patient as a result of the pseudotumor cerebri (Verderber et al, 1991).
    F) SECONDARY PERIPHERAL NEUROPATHY
    1) WITH POISONING/EXPOSURE
    a) Early signs of organochlorine poisoning involve hyperesthesia and paresthesia of the face and extremities, headache, dizziness, and incoordination (Morgan, 1993). Peters et al (1982) report a 60 percent incidence of sensory neuropathy in the acute phase of oral hexachlorobenzene toxicity.
    b) Occasional reports have associated peripheral neuropathy with exposure to organochlorines. In one case, an agricultural worker developed polyneuritis of the sensory-motor type approximately two weeks after exposure to lindane in an unknown solvent (Tolot, 1969).
    G) HEADACHE
    1) WITH POISONING/EXPOSURE
    a) Severe headache may occur with exposure to lindane (Hathaway et al, 1996; Diaz, 1998).
    H) NEUROTOXICITY
    1) WITH POISONING/EXPOSURE
    a) Mild drowsiness, somnolence or lethargy may develop in patients with mild toxicity. More severe CNS depression may develop following seizures (Aks et al, 1995).
    b) CASE REPORT: A 56-year-old man vomited about 6 ounces (about 177 mL) of a white liquid about 30 minutes after ingesting about 12 ounces (about 355 mL) of an insecticide containing 20% lindane. Despite supportive therapy, including gastric lavage and activated charcoal, his mental status deteriorated, necessitating intubation about 75 minutes after ingestion. He experienced multiple seizures (a focal seizure followed by 3 brief generalized tonic-clonic seizures) from 3 to 14 hours postingestion and received diazepam, phenobarbital, and phenytoin. His arterial blood pH was 6.76 after the focal seizure and 7.05 after the first generalized seizure and he received sodium bicarbonate that corrected the pH to 7.34. He was extubated about 43 hours after ingestion, but he developed premature ventricular contractions and premature atrial contractions on day 3 and atrial fibrillation/flutter on day 5. Following supportive therapy, including IV lidocaine, digoxin, and IV verapamil, his condition resolved. On day 6, he developed ataxia, wide-based gait, slurred speech, paranoia, and depression with defects in higher mental functioning. On day 12, he still had neurological abnormalities (eg, poor memory, loss of task focus, inadequate mentation, and defects in fine motor coordination). At this time, he committed suicide by cutting his femoral artery. Toxicokinetics analysis revealed an estimated distribution half-life of 10.3 hours and a terminal half-life of 162.9 hours, which is much longer than the previously reported terminal half-life of 25 to 36 hours (Wiles et al, 2015).
    c) CASE REPORT: A 3-year-old boy developed generalized seizures in his sleep with transient loss of sensorium about 1 hour after ingesting 10 mL of lindane lotion. He presented with drowsiness, a GCS (Glasgow coma score) of 10/15, and papular skin lesions. Following supportive care, he recovered 24 hours later (Ramabhatta et al, 2014).
    d) In a review article, 67 cases of severe adverse effects (AE), including 20 death (including 7 children and 4 patients aged 65 and older) following the labeled use (47 cases), excessive use (17 patients), and ingestion (3 cases) of lindane were identified. Seizures and respiratory problems developed in the majority of patients who developed severe AEs after using the labeled dose (defined as one 4-minute application of shampoo or one 8 to 12 hours of single total body application of lindane lotion 1%). Three patients ingested lindane. The most common adverse effects were tremor, vertigo, paresis, and aphasia (Nolan et al, 2012).
    e) Lindane-induced neurotoxicity (seizures, coma) occurred in a 10-month-old boy after he received repeated topical applications of 1% lindane (twice daily for 10 days) for the treatment of scabies. Laboratory results also revealed anemia and hypoproteinemia. Following the discontinuation of lindane and supportive treatment, his symptoms improved without further sequelae. The authors suggested that malnutrition may be a risk factor for developing neurotoxicity with lindane application. In addition, patients with reduced body fat may have higher lindane levels (Bhalla & Thami, 2004).
    I) TOXIC ENCEPHALOPATHY
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: A 38-year-old man with HIV, scabies and seborrheic dermatitis and tinea cruris developed status epilepticus 2 hours after 2 ounces of 1 percent lindane lotion was applied to his skin (Solomon et al, 1995).
    1) Five days later he was demented and able to follow simple commands and communicate verbally but with minimal comprehension. He had a mild right arm tremor, hyperreflexia, positive Babinski's signs and frontal release signs. EEG revealed encephalopathy seven days after the seizure.
    b) CASE REPORT - A 66-year-old man with scabies developed acute mental status changes, seizures, hypoxemia, respiratory acidosis, and hypotension approximately 8 hours after a single application of a thin film of lindane (1%) to his skin from the neck to the toes. Prior to the lindane administration, fresh scratch marks were noted throughout the patient's body which might have increased dermal absorption of lindane significantly. In addition, the physician's order did not include instructions to wash lindane from the skin after its application. All workup for bacteremia, sepsis, and meningitis were unremarkable. An EEG showed severe generalized cerebral dysfunction. He experienced infectious complications and died 50 days after admission. During autopsy, he was diagnosed with hypoxic ischemic encephalopathy from lindane poisoning (Sudakin, 2007).
    J) SEQUELA
    1) WITH POISONING/EXPOSURE
    a) Following an acute poisoning after 3 applications of topical lindane, a 37-year-old female developed prolonged neurological and psychological symptoms for 20 months. Long-term effects included muscle spasm and extensor/flexor imbalance and myoclonic jerks, abdominal pain, urinary incontinence, and fecal incontinence. Long-term central effects included anxiety, depression, memory impairment, red/green visual changes, hallucinations, and incoordination (Hall & Hall, 1999).
    1) The authors suggest that the toxicity was severe due in part to repeated applications of lindane and the fact that this patient was protein-restricted (vegetarian).

Gastrointestinal

    3.8.2) CLINICAL EFFECTS
    A) NAUSEA, VOMITING AND DIARRHEA
    1) WITH POISONING/EXPOSURE
    a) When ingested, lindane has caused abdominal pain, nausea, vomiting and diarrhea, especially when contained in petroleum solvents (Lifshitz & Gavrilov, 2002; Jaeger et al, 1984). Vomiting is commonly reported after ingestion (Nordt & Chew, 2000).
    b) CASE SERIES: Based on a five-year retrospective review of lindane exposures reported to six poison centers in Texas, the most frequent symptoms associated with inadvertent ingestion were nausea, vomiting, and throat irritation (Forrester et al, 2004).
    c) Topical exposure to a 20% lindane insecticide has resulted in vomiting in an infant (Ryan et al, 1998).
    d) CASE REPORT: A 47-year-old man developed vomiting after ingesting 1 ounce of lindane (concentration unknown) instead of a cough syrup (Anon, 2005).
    B) PANCREATITIS
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Pancreatitis was reported 13 days after ingestion of 15 to 30 mL of lindane by a 35-year-old man (Munk & Nantel, 1977; Nantel et al, 1977).

Hepatic

    3.9.2) CLINICAL EFFECTS
    A) LIVER ENZYMES ABNORMAL
    1) WITH POISONING/EXPOSURE
    a) An important property of the chlorinated hydrocarbons, particularly lindane, is their capacity to induce the drug-metabolizing enzymes of the liver (Wells & Milhorn, 1983; Klaasen, 1985).
    b) CASE SERIES: Statistically significant increases in serum levels of lactate dehydrogenase (33%), and leucine aminopeptidase (45%), were reported in 19 individuals occupationally exposed (likely both inhalation and dermal exposure) to technical grade lindane for over 10 years in a lindane-formulating plant (ATSDR, 2003).
    c) CASE REPORT: A single case report documents abnormal liver function tests in a patient who used 1% lindane twice a day for 3 weeks and subsequently developed aplastic anemia. No other cause was apparent for the abnormal liver function tests (Rauch et al, 1990).
    d) Patients presenting with hexachlorobenzene-induced porphyria may develop elevated liver function tests (Peters et al, 1982).
    e) CASE REPORT: Elevated LDH levels were found in a 14-year-old male following at least eight topical applications of lindane over a 6-week period (Berry et al, 1987).
    B) LARGE LIVER
    1) WITH POISONING/EXPOSURE
    a) Chronic exposure may induce liver damage and hepatomegaly, as reviewed in Gosselin (1984) and reported by Peters et al (1982).
    3.9.3) ANIMAL EFFECTS
    A) ANIMAL STUDIES
    1) HEPATOCELLULAR DAMAGE
    a) Chronic exposure has caused hepatic damage in experimental animals (Budavari, 1996). Acute exposure has resulted in induction of liver microsomal drug-metabolizing enzymes (Kolmodin-Hedman et al, 1971).
    2) HEPATOMEGALY
    a) Lindane produced some hepatomegaly in rats at 800 ppm in the diet (Clayton & Clayton, 1994), and changes at the cellular level were seen in rats given lindane at 50 ppm in the diet (Ortega et al, 1957).

Genitourinary

    3.10.1) SUMMARY
    A) WITH POISONING/EXPOSURE
    1) Renal insufficiency and myoglobinuria secondary to rhabdomyolysis associated with prolonged seizures have been described.
    3.10.2) CLINICAL EFFECTS
    A) MYOGLOBINURIA
    1) WITH POISONING/EXPOSURE
    a) Myoglobinuria secondary to rhabdomyolysis has been described after ingestion of lindane (Jaeger et al, 1984; Rao et al, 1988; Hall & Hall, 1999).
    B) ABNORMAL RENAL FUNCTION
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 35-year-old man developed status epilepticus and myoglobinuria with subsequent acute renal insufficiency and generalized myolysis following ingestion of lindane-contaminated food (Nantel et al, 1977).
    3.10.3) ANIMAL EFFECTS
    A) ANIMAL STUDIES
    1) RENAL FUNCTION ABNORMAL
    a) Renal degeneration has been reported in rodents exposed to lindane (Proctor et al, 1988). Fat deposits in the proximal convoluted tubule has been reported in a review by Solomon et al (1977).
    b) Renal degeneration has been seen in rodents exposed chronically to lindane (Hathaway et al, 1996).

Acid-Base

    3.11.2) CLINICAL EFFECTS
    A) LACTIC ACIDOSIS
    1) WITH POISONING/EXPOSURE
    a) Lactic acidosis has been reported after ingestions of lindane (Jaeger et al, 1984).
    B) ACIDOSIS
    1) WITH POISONING/EXPOSURE
    a) Severe metabolic acidosis may be a consequence of severe convulsions (Morgan, 1993; Wiles et al, 2015), and the acidosis may be an immediate cause of death (Morgan, 1993).
    b) CASE REPORT: A 35-year-old male presented with status epilepticus and severe metabolic acidosis following ingestion of lindane-contaminated food (Nantel et al, 1977).
    c) CASE REPORT: A 3.5-year-old boy developed lethargy, irritability, tremors, nausea, vomiting, mild metabolic acidosis (pH 7.28, pCO2 41 mmHg, pO2 95 mmHg, HCO3 20 mmol/L), and general clonic-tonic seizures lasting about 2 minutes after ingesting an unknown quantity of 35% lindane solution. Following supportive treatment, his symptoms improved gradually; he was discharged home 42 hours postingestion (Lifshitz & Gavrilov, 2002).
    C) RESPIRATORY ACIDOSIS
    1) WITH THERAPEUTIC USE
    a) CASE REPORT - A 66-year-old man with scabies developed acute mental status changes, seizures, hypoxemia, severe respiratory acidosis (pH=6.94; pCO2=84; pO2=196), and hypotension approximately 8 hours after a single application of a thin film of lindane (1%) to his skin from the neck to the toes. Prior to the lindane administration, fresh scratch marks were noted throughout the patient's body which might have increased dermal absorption of lindane significantly. In addition, the physician's order did not include instructions to wash lindane from the skin after its application. All workup for bacteremia, sepsis, and meningitis were unremarkable. An EEG showed severe generalized cerebral dysfunction. He experienced infectious complications and died 50 days after admission. During autopsy, he was diagnosed with hypoxic ischemic encephalopathy from lindane poisoning (Sudakin, 2007).
    2) WITH POISONING/EXPOSURE
    a) Respiratory acidosis may develop in patients with significant CNS depression (Aks et al, 1995).

Hematologic

    3.13.2) CLINICAL EFFECTS
    A) APLASTIC ANEMIA
    1) WITH POISONING/EXPOSURE
    a) Large exposures to lindane have been associated with rare individual cases of aplastic anemia (Nolan et al, 2012; Morgan, 1993; Woodliff et al, 1966). At least eight cases have been reported for persons exposed to lindane from vaporizers (West, 1967; Loge, 1965; Baselt, 1997).
    b) The association of aplastic anemia with exposure to lindane vapors generated from solid pellets in the absence of solvent excludes the possible contribution of benzene, which has been used as a solvent in some commercial preparations of lindane.
    c) No evidence of pancytopenia with reticulopenia was found in persons occupationally exposed to lindane, however (Milby & Samuels, 1971).
    d) CASE REPORT: A case of severe, reversible hypoplastic anemia was reported in a 2-year-old child from suspected exposure to lindane from a vaporizer (Morgan, 1980).
    e) Acute leukemia has been reported from repeated topical exposures to related chlorinated hydrocarbon insecticides, namely chlordane and heptachlor, (Infante et al, 1978). Sidi et al (1983) report a case of a 66-year-old man with aplastic anemia following chronic exposure to lindane insecticide. He subsequently was diagnosed with acute myeloblastic leukemia and died soon thereafter from septicemia.
    f) CASE REPORT: A 21-year-old man using 1% lindane lotion (applied twice a day to the body for 3 weeks) developed aplastic anemia 3 weeks later (Rauch et al, 1990).
    B) LEUKOCYTOSIS
    1) WITH THERAPEUTIC USE
    a) Leukocytosis has been reported following the use of lindane (Nolan et al, 2012).
    C) PANCYTOPENIA
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Pancytopenia was reported in a 14-year-old boy who applied 1% lindane lotion on 8 occasions to open lesions, and left the lotion on for 48 hours after the final application (Berry et al, 1987).
    D) DISSEMINATED INTRAVASCULAR COAGULATION
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Disseminated intravascular coagulation (DIC) was described 8 hours postingestion in a 43-year-old woman who ingested 8 ounces of 20% lindane. The coagulopathy reversed following a decline in lindane serum levels (Rao et al, 1988).
    E) PORPHYRIA DUE TO TOXIC EFFECT OF SUBSTANCE
    1) WITH POISONING/EXPOSURE
    a) Mixed porphyria symptoms have been reported following an outbreak of accidental hexachlorobenzene ingestion from treated seed grain. Many patients had persistent abnormal porphyrin metabolism with significant neurologic, dermatologic, and orthopedic signs and symptoms (Peters et al, 1982).
    F) THROMBOCYTOPENIC PURPURA
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Idiopathic thrombocytopenic purpura developed in a 17-year-old woman who worked formulating wood preservatives (Hay & Singer, 1991). Her exposures included lindane, pentachlorophenol, tributyl-tin oxide, permethrin and petroleum based solvents.

Dermatologic

    3.14.2) CLINICAL EFFECTS
    A) CONTACT DERMATITIS
    1) WITH POISONING/EXPOSURE
    a) Extensive contact may result in dermatitis and local hypersensitivity reactions (Fiumara & Kahn, 1983; Budavari, 1996).
    B) EXANTHEMATOUS DISORDER
    1) WITH THERAPEUTIC USE
    a) Acute generalized exanthematous pustulosis has been reported following lindane use (Nolan et al, 2012).
    C) PURPURA
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: A 9-year-old boy developed Henoch-Schonlein (anaphylactoid) purpura 2 days following therapeutic topical application of 1% lindane shampoo (Fagan, 1981).
    D) SKIN ABSORPTION
    1) WITH THERAPEUTIC USE
    a) Percutaneous absorption may occur in patients with compromised epidermal barrier function (Friedman, 1987; Solomon et al, 1995).
    E) ERUPTION
    1) WITH POISONING/EXPOSURE
    a) Some skin irritation results from extensive contact with these agents or with petroleum distillates in which they are contained.
    b) CASE REPORT: Scattered small papules and wheal lesions with itching were reported on the extremities of a 24-year-old woman after daily applications of lindane for 24 consecutive days (Diaz, 1998). No areas of blistering, cracking, scaling, or excoriation were noted.
    F) SKIN LESION
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 3-year-old boy developed generalized seizures in his sleep with transient loss of sensorium about 1 hour after ingesting 10 mL of lindane lotion. She presented with drowsiness, a GCS (Glasgow coma score) of 10/15, and papular skin lesions. Following supportive care, he recovered 24 hours later (Ramabhatta et al, 2014).
    G) PORPHYRIA CUTANEA TARDA
    1) WITH POISONING/EXPOSURE
    a) There was a report of an outbreak of accidental hexachlorobenzene ingestions in persons consuming treated seed grain. Persistent abnormal porphyrin metabolism was seen 25 years later with significant porphyria cutanea tarda present in many patients. A syndrome called "pembe yara" or "pink sore", related to porphyria, was noted in the form of pigmented patches on the skin resembling a ringworm rash (Peters et al, 1982).
    1) Urine levels of uroporphyrin may be elevated.

Musculoskeletal

    3.15.2) CLINICAL EFFECTS
    A) RHABDOMYOLYSIS
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Rhabdomyolysis, muscle weakness and myoglobinuria were reported in a 35-year-old man who ingested 15 to 30 mL of lindane. Seizures were present for 2 hours (Munk & Nantel, 1977). Elevated creatine kinase with hemoglobinuria and myoglobinuria have been reported in patients exhibiting skeletal muscle damage from hexachlorobenzene toxicity (Peters et al, 1982).
    b) CASE SERIES: After a hexachlorobenzene ingestion epidemic, a reported 73% of patients developed profound muscle weakness in the acute phase (Peters et al, 1982).
    c) CASE REPORT: Rhabdomyolysis with increased creatine kinase levels developed 3 days after ingestion of a lindane-solvent mixture in one case (Jaeger et al, 1984).
    B) ARTHRITIS
    1) WITH POISONING/EXPOSURE
    a) Arthritis, with and without gout, has been reported as an adverse sequelae to acute lindane and chlorinated hydrocarbon ingestions. Uric acid levels may be elevated (Peters et al, 1982; Wells & Milhorn, 1983).

Endocrine

    3.16.2) CLINICAL EFFECTS
    A) GOITER
    1) WITH POISONING/EXPOSURE
    a) There was a report of a large number of patients presenting with palpable thyroid enlargement following ingestion of hexachlorobenzene-treated seed grain 25 years previously (Peters et al, 1982). No malignancies were seen.

Immunologic

    3.19.2) CLINICAL EFFECTS
    A) IMMUNE SYSTEM FINDING
    1) WITH POISONING/EXPOSURE
    a) One study evaluated immunological alterations in the blood of 20 human lindane poisoning cases. Lindane exposure was associated with higher levels of serum IL-2, IL-4, and TNF-alpha and lower levels of IFN-gamma. However, serum immunoglobulin (IgG, IgM, IgA, IgE) levels did not change (Seth et al, 2005).
    B) ANAPHYLAXIS
    1) WITH THERAPEUTIC USE
    a) Anaphylactic reaction has been reported following lindane use (Nolan et al, 2012).
    3.19.3) ANIMAL EFFECTS
    A) ANIMAL STUDIES
    1) LACK OF EFFECT
    a) RABBITS - Lindane given at 10 mg/kg/d for 90 days was not immunosuppressive in rabbits (Roszkowski, 1978).

Reproductive

    3.20.1) SUMMARY
    A) Lindane did not have reproductive effects in a 3-generation rat study. It has not been teratogenic in animals. It disturbed the estrus cycle and reduced fertility in female rats. It has been detected in human breast milk. It produced degenerative changes in the testes of rats.
    3.20.2) TERATOGENICITY
    A) FETOTOXICITY
    1) Fetotoxicity was observed in the rat, rabbit and hamster. Specific developmental abnormalities in the musculoskeletal system were observed in the rat and rabbit. Pre-implantation mortality and physical effects on the newborn were observed in rabbits. Stillbirth occurred in one dog study (RTECS , 2000).
    2) Lindane did not produce cleft palate or hydronephrosis in mice after a single oral dose of 30 or 45 mg/kg on day 12 of gestation; fetal and placental weights were decreased, and thymic weight was lower in exposed fetuses (Hassoun & Stohs, 1996).
    B) LACK OF EFFECT
    1) When given to rabbits and rats at 5, 10, or 20 mg/kg for 12 days during gestation, lindane was neither embryotoxic nor teratogenic (Clayton & Clayton, 1994).
    2) Lindane was not teratogenic in rats when given at doses up to 25 mg/kg/d from days 6 to 15 of gestation (Khera et al, 1979). Lindane was not teratogenic in mice, rats, hamsters, rabbits, cows, or pigs (Schardein, 1993).
    3) In experimental animals, lindane has typically not caused birth defects (Wolfe & Esher, 1980; Khera et al, 1979; Palmer, 1978; Dinerman, 1970; Dzierzawski, 1977; Earl, 1973; Pernov & Kyurkchiyev, 1974; McParland & McCracker, 1973; Palmer, 1978; Khera et al, 1979; Mametkuliev, 1976). Lindane did not produce cleft palate or hydronephrosis in mice after a single oral dose of 30 or 45 mg/kg on day 12 of gestation; fetal and placental weights were decreased, and thymic weight was lower in exposed fetuses (Hassoun & Stohs, 1996).
    3.20.3) EFFECTS IN PREGNANCY
    A) PREGNANCY CATEGORY
    LINDANEB
    Reference: Briggs et al, 1998
    B) HUMANS
    1) Lindane is transferred across the human placenta (Saxena, 1980) as early as the 22nd week of pregnancy (Fiserova-Bergerova, 1967), and is found in human fetal tissue (Yoshimura, 1979) and blood (Saxena, 1984) at levels comparable to maternal levels. Higher levels of lindane and other organochlorine pesticides have been found in maternal and fetal blood and placenta in spontaneous abortions and premature deliveries than in normal pregnancies (Saxena, 1981; Wasserman, 1982). Other studies found that lindane levels were not related to stillbirths (Curley, 1969) or other pathological conditions of pregnancy (Curley, 1969; Poradovsky, 1977).
    2) In women working at a pesticide plant exposed only to lindane, excessive blood loss after delivery and lower birth weights in offspring were noted (pp 88-91).
    C) STILLBIRTH
    1) A case was reported of intentional organochlorine insecticide ingestion in a 25-year-old female at 16 weeks of pregnancy. Death of twin fetuses was reported as well as vaginal hemorrhage (Konje et al, 1992).
    D) ANIMAL STUDIES
    1) STILLBIRTH
    a) Lindane increased postimplantation deaths in rats (Mametkuliev, 1976). It has also caused stillbirths in dogs (Earl, 1973) and rabbits (Khamidov, 1984).
    2) EMBRYONIC DEVELOPMENT
    a) Female CD-1 mice were given lindane orally (15 or 25 mg/kg) prior to mating or immediately after mating, and the results indicated that lindane exposure of developing oocytes in vivo led to an increase of irreversible damage (ie, lysis, fragmentation) in two-cell embryos. Based on the experimental doses used in this study (approximately 1000 to 10000 times the measured concentration of lindane in the general human population), the authors suggested that there appears to be an ample margin of safety for human embryonic development (Scascitelli & Pacchierotti, 2003).
    3.20.4) EFFECTS DURING BREAST-FEEDING
    A) BREAST MILK
    1) Concentrations of lindane may be found in human breast milk after maternal exposure and absorption, however, it is excreted rapidly in the urine and does not persist in the milk (Hayes & Laws, 1991).
    2) Highly lipophilic compounds which are stored in body fat such as dieldrin are likely to be transferred to breast milk (Morgan, 1993).
    3) Those chlorinated hydrocarbon insecticides which are more rapidly metabolized, such as lindane, are less likely to be detected in breast milk (Morgan, 1993).
    4) Levels of lindane found in human breast milk were 0.029 ppm in 1972 and 0.075 ppm in 1977 (Clayton & Clayton, 1994). Lindane has been found in human breast milk in Germany (Rappl & Waiblinger, 1975).
    5) Lindane has been found in ewe's milk at a level of 4 ppm after the animals were dipped with Entomoxan, which contains 0.5 percent BHC (Clayton & Clayton, 1994).
    6) The daily intake of total organochlorine pesticides residues calculated for the suckling infant was significantly higher when compared with the acceptable daily intake (ADI) as recommended by FAO/WHO (FAO/WHO, 1970).
    B) PORPHYRIA
    1) Twenty-five years following an outbreak of hexachlorobenzene toxicity, a significant degree of porphyria was found in persons who were breast-fed as infants by mothers exposed to lindane (Peters et al, 1982).
    3.20.5) FERTILITY
    A) TESTIS DISORDER
    1) RATS - Degenerative changes in the testes and lack of sperm occurred in rats fed relatively high levels of lindane (Hathaway et al, 1996) Srinivasan et al, 1988).
    B) FERTILITY DECREASED FEMALE
    1) RATS - Lindane given orally to female rats at 0.5 mg/kg for 4 months disturbed the estrus cycle, decreased conceptions and fertility, and was embryotoxic (HSDB , 2000).
    C) PREGNANCY DISORDER
    1) Lindane lengthened the gestation period by as much as two days in rats when given orally at 5, 10, or 15 mg/kg (HSDB , 2000).
    D) LACK OF EFFECT
    1) No reproductive abnormalities were reported in male workers with chronic occupational exposure to lindane (Tomczak et al, 1981).

Carcinogenicity

    3.21.1) IARC CATEGORY
    A) IARC Carcinogenicity Ratings for CAS58-89-9 (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
    3.21.2) SUMMARY/HUMAN
    A) Several cases of acute myeloid leukemia have been reported with lindane exposure. Lindane has been carcinogenic in mice, inducing liver tumors.
    3.21.3) HUMAN STUDIES
    A) CARCINOMA
    1) The International Agency for Research on Cancer (IARC) has listed some organochlorine agents (eg, DDT) as "probably carcinogenic to humans", although it also categorizes them as being inadequately assessed for human carcinogenic potential (IARC, 1982).
    2) Lindane is classified in Carcinogenicity Category A3 (confirmed animal carcinogen with unknown relevance to humans) by the ACGIH (ACGIH, 2000).
    B) LEUKEMIA
    1) At least three cases of acute myeloid leukemia have been reported with exposure to HCB or lindane (Sittig, 1985).
    2) Acute leukemia has been reported from repeated topical exposures to related chlorinated hydrocarbon insecticides, namely chlordane and heptachlor, (Infante et al, 1978). A case was reported of a 66-year-old male with aplastic anemia following chronic exposure to lindane insecticide. He was subsequently diagnosed with acute myeloblastic leukemia and died soon after from septicemia (Sidi et al, 1983).
    3) In vitro tests on human and rat hematopoietic progenitors proved the cytotoxicity of lindane. Granulocytic and macrophagic progenitors were destroyed by lindane. Cytotoxicity was observed with lindane concentrations similar to those measured in human blood in acute cases of intoxication and in fat tissues of exposed populations (Parent-Massin et al, 1994).
    3.21.4) ANIMAL STUDIES
    A) CARCINOMA
    1) Lindane was found to be carcinogenic and designated neoplastic by RTECS criteria with liver, lung, thorax or respiration tumors observed (Reuber, 1987; RTECS , 2000). Typical of other chlorinated hydrocarbon insecticides, lindane is a cancer promoter in laboratory studies (Clayton & Clayton, 1994).
    2) Liver tumors have been found in lindane-exposed rats (Hanada, 1976; Fitzhugh, 1950; Reuber, 1979) and mice (Hayes & Laws, 1991; Kashyap, 1979; Arai, 1972) following administration of relatively high oral doses (600 to 1000 ppm in the diet).
    B) LACK OF EFFECT
    1) Lindane given at 100 ppm in the diet totally protected rats from liver tumors induced by aflatoxin B1 (Angsubhakorn et al, 1989).
    2) Lindane was not carcinogenic in mice or rats in several feeding studies at lower doses (Didenko, 1973; Hiroshi, 1972; Weisse & Herbst, 1977; p 109). In mice, lindane did not cause tumors with dermal exposure at a dose which caused tumors when administered orally (Kashyap, 1979). It was not carcinogenic in rats or mice in an NCI bioassay (Clayton & Clayton, 1981).

Genotoxicity

    A) Lindane has been inactive in various short-term assays, but may be genotoxic at the chromosomal level. DNA damage was reported in human nasal and gastric mucosal cells after lindane exposure. Mutations have occurred in human lymphocytes.

Monitoring Parameters Levels

    4.1.1) SUMMARY
    A) Serum levels are not readily available or clinically useful for guiding management.
    B) Monitor serum electrolytes and blood glucose.
    C) Monitor creatinine kinase if seizures are prolonged.
    D) Obtain venous or arterial blood gas if acidosis is a concern.
    E) An EEG may be needed to rule out status epilepticus or guide aggressive anticonvulsive therapy.
    4.1.2) SERUM/BLOOD
    A) TOXICITY
    1) The toxic concentration of lindane in blood is 0.5 mcg/mL (HSDB , 2000).
    2) Serum levels are not readily available or clinically useful for guiding management.
    3) Monitor serum electrolytes and blood glucose.
    4) Monitor creatinine kinase if seizures are prolonged.
    5) Obtain venous or arterial blood gas if acidosis is a concern.
    4.1.3) URINE
    A) URINARY LEVELS
    1) A level of 4.95 mg BHC/100 mL of urine was found in a woman who had washed two calves with a solution of BHC. She developed severe seizures but survived (Clayton & Clayton, 1981).
    4.1.4) OTHER
    A) OTHER
    1) OTHER
    a) The average daily intake of lindane for persons living in the US was 5.6 mcg from 1965 to 1970. During this time, levels of lindane in body fat ranged from 0.2 to 0.6 ppm. Levels in Japanese, Formosans, and Argentinians have been higher (Clayton & Clayton, 1994).

Radiographic Studies

    A) An EEG may be needed to rule out status epilepticus or guide aggressive anticonvulsive therapy.

Methods

    A) CHROMATOGRAPHY
    1) Benzene hexachloride can be quantitated in serum, at levels above and below those likely to be associated with acute poisoning, by gas-liquid chromatographic methods. However, in most clinical circumstances, levels are not readily available on an emergency basis.
    2) Levels are useful in documenting lindane toxicity after the acute situation has resolved. The advice of professionals should be sought in evaluating benzene hexachloride concentrations.

Life Support

    A) Support respiratory and cardiovascular function.

Patient Disposition

    6.3.1) DISPOSITION/ORAL EXPOSURE
    6.3.1.1) ADMISSION CRITERIA/ORAL
    A) SUMMARY: Patients with persistently altered mental status, abnormal vital signs, or recurrent seizures should be admitted.
    B) Ingestion of as little as 5 mL of 1 percent lindane has caused respiratory depression and/or seizures in children aged 1 to 4 years (Nordt & Chew, 1999; (Aks et al, 1995) Jaeger et al, 1983). One 4-month-old developed increased muscle tone, tonic posturing, and poor orientation to visual stimuli after a total body application of 1 percent lindane was left on for 24 hours (Pramanik & Hansen, 1979). A 2-month old died after total body application of 1 percent lindane that was left on for 18 hours (Davies et al, 1983).
    1) Therefore, any toddler or child who ingests a product containing any amount of lindane should be referred to a medical facility for evaluation.
    2) Children who sustain a dermal exposure to lindane products should have the skin immediately washed well with soap and water. Children who develop signs of lethargy or CNS excitation, and young children/infants (less than 1 year old) with prolonged dermal exposure should be referred for medical evaluation and treatment.
    6.3.1.2) HOME CRITERIA/ORAL
    A) Adults who intentionally ingest lindane or any child with an ingestion should be referred to a health care facility. Patients with prolonged or repeated dermal applications should be referred to a health care facility if they become symptomatic.
    6.3.1.3) CONSULT CRITERIA/ORAL
    A) Consult a poison center or medical toxicologist for assistance in managing severe poisonings.
    6.3.1.5) OBSERVATION CRITERIA/ORAL
    A) After ingestion, patients who are asymptomatic after 6 hours can be discharged home.

Monitoring

    A) Serum levels are not readily available or clinically useful for guiding management.
    B) Monitor serum electrolytes and blood glucose.
    C) Monitor creatinine kinase if seizures are prolonged.
    D) Obtain venous or arterial blood gas if acidosis is a concern.
    E) An EEG may be needed to rule out status epilepticus or guide aggressive anticonvulsive therapy.

Oral Exposure

    6.5.1) PREVENTION OF ABSORPTION/PREHOSPITAL
    A) SUMMARY
    1) Prehospital oral decontamination should be avoided because of the risk of rapid onset CNS depression and seizures and subsequent aspiration.
    B) EMESIS/NOT RECOMMENDED
    1) Emesis is not recommended, since seizures commonly occur suddenly and without warning. Seizures have occurred as early as 15 minutes following ingestion (Jaeger et al, 1984). Death from ARDS and anoxia has occurred in an adult who seized during ipecac-induced vomiting (Kurt et al, 1986).
    6.5.2) PREVENTION OF ABSORPTION
    A) SUMMARY
    1) Generally, gastrointestinal decontamination should be avoided because of the potential for rapid onset CNS depression and seizures with subsequent aspiration.
    2) Activated charcoal did not reliably limit gastrointestinal absorption of lindane in animals. However, modest doses of charcoal and castor oil appeared to offer some beneficial effects (Morgan et al, 1977). Oral administration of cholestyramine reduced the incidence of seizures and death to a greater degree than activated charcoal in lindane-poisoned mice when used in equal doses (Kassner et al, 1993).
    3) Do NOT administer milk, fatty foods, or oils by mouth since these may enhance absorption of lindane.
    6.5.3) TREATMENT
    A) MONITORING OF PATIENT
    1) Serum levels are not readily available or clinically useful for guiding management.
    2) Monitor serum electrolytes and blood glucose.
    3) Monitor creatinine kinase if seizures are prolonged.
    4) Obtain venous or arterial blood gas if acidosis is a concern.
    5) EEG may be needed to rule out status epiliepticus or guide aggressive anticonvulsive therapy.
    B) SEIZURE
    1) SUMMARY
    a) Attempt initial control with a benzodiazepine (eg, diazepam, lorazepam). If seizures persist or recur, administer phenobarbital or propofol.
    b) Monitor for respiratory depression, hypotension, and dysrhythmias. Endotracheal intubation should be performed in patients with persistent seizures.
    c) Evaluate for hypoxia, electrolyte disturbances, and hypoglycemia (or, if immediate bedside glucose testing is not available, treat with intravenous dextrose).
    2) DIAZEPAM
    a) ADULT DOSE: Initially 5 to 10 mg IV, OR 0.15 mg/kg IV up to 10 mg per dose up to a rate of 5 mg/minute; may be repeated every 5 to 20 minutes as needed (Brophy et al, 2012; Prod Info diazepam IM, IV injection, 2008; Manno, 2003).
    b) PEDIATRIC DOSE: 0.1 to 0.5 mg/kg IV over 2 to 5 minutes; up to a maximum of 10 mg/dose. May repeat dose every 5 to 10 minutes as needed (Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008).
    c) Monitor for hypotension, respiratory depression, and the need for endotracheal intubation. Consider a second agent if seizures persist or recur after repeated doses of diazepam .
    3) NO INTRAVENOUS ACCESS
    a) DIAZEPAM may be given rectally or intramuscularly (Manno, 2003). RECTAL DOSE: CHILD: Greater than 12 years: 0.2 mg/kg; 6 to 11 years: 0.3 mg/kg; 2 to 5 years: 0.5 mg/kg (Brophy et al, 2012).
    b) MIDAZOLAM has been used intramuscularly and intranasally, particularly in children when intravenous access has not been established. ADULT DOSE: 0.2 mg/kg IM, up to a maximum dose of 10 mg (Brophy et al, 2012). PEDIATRIC DOSE: INTRAMUSCULAR: 0.2 mg/kg IM, up to a maximum dose of 7 mg (Chamberlain et al, 1997) OR 10 mg IM (weight greater than 40 kg); 5 mg IM (weight 13 to 40 kg); INTRANASAL: 0.2 to 0.5 mg/kg up to a maximum of 10 mg/dose (Loddenkemper & Goodkin, 2011; Brophy et al, 2012). BUCCAL midazolam, 10 mg, has been used in adolescents and older children (5-years-old or more) to control seizures when intravenous access was not established (Scott et al, 1999).
    4) LORAZEPAM
    a) MAXIMUM RATE: The rate of intravenous administration of lorazepam should not exceed 2 mg/min (Brophy et al, 2012; Prod Info lorazepam IM, IV injection, 2008).
    b) ADULT DOSE: 2 to 4 mg IV initially; repeat every 5 to 10 minutes as needed, if seizures persist (Manno, 2003; Brophy et al, 2012).
    c) PEDIATRIC DOSE: 0.05 to 0.1 mg/kg IV over 2 to 5 minutes, up to a maximum of 4 mg/dose; may repeat in 5 to 15 minutes as needed, if seizures continue (Brophy et al, 2012; Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008; Sreenath et al, 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) 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).
    8) In vitro studies suggest that lindane's neurotoxic effects are mediated via blockade of the GABA-receptor coupled sodium channel (Pomes et al, 1994). Therefore anticonvulsants which act by enhancing GABA activity such as benzodiazepines and barbiturates are preferred over phenytoin.
    a) ANIMALS - Pretreatment with phenytoin appeared to enhance the convulsive effects of lindane in rats (Tilson et al, 1985).
    C) 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).
    D) RHABDOMYOLYSIS
    1) SUMMARY: Early aggressive fluid replacement is the mainstay of therapy and may help prevent renal insufficiency. Diuretics such as mannitol or furosemide may be added if necessary to maintain urine output but only after volume status has been restored as hypovolemia will increase renal tubular damage. Urinary alkalinization is NOT routinely recommended.
    2) Initial treatment should be directed towards controlling acute metabolic disturbances such as hyperkalemia, hyperthermia, and hypovolemia. Control seizures, agitation, and muscle contractions (Erdman & Dart, 2004).
    3) FLUID REPLACEMENT: Early and aggressive fluid replacement is the mainstay of therapy to prevent renal failure. Vigorous fluid replacement with 0.9% saline (10 to 15 mL/kg/hour) is necessary even if there is no evidence of dehydration. Several liters of fluid may be needed within the first 24 hours (Walter & Catenacci, 2008; Camp, 2009; Huerta-Alardin et al, 2005; Criddle, 2003; Polderman, 2004). Hypovolemia, increased insensible losses, and third spacing of fluid commonly increase fluid requirements. Strive to maintain a urine output of at least 1 to 2 mL/kg/hour (or greater than 150 to 300 mL/hour) (Walter & Catenacci, 2008; Camp, 2009; Erdman & Dart, 2004; Criddle, 2003). To maintain a urine output this high, 500 to 1000 mL of fluid per hour may be required (Criddle, 2003). Monitor fluid input and urine output, plus insensible losses. Monitor for evidence of fluid overload and compartment syndrome; monitor serum electrolytes, CK, and renal function tests.
    4) DIURETICS: Diuretics (eg, mannitol or furosemide) may be needed to ensure adequate urine output and to prevent acute renal failure when used in combination with aggressive fluid therapy. Loop diuretics increase tubular flow and decrease deposition of myoglobin. These agents should be used only after volume status has been restored, as hypovolemia will increase renal tubular damage. If the patient is maintaining adequate urine output, loop diuretics are not necessary (Vanholder et al, 2000).
    5) URINARY ALKALINIZATION: Alkalinization of the urine is not routinely recommended, as it has never been documented to reduce nephrotoxicity, and may cause complications such as hypocalcemia and hypokalemia (Walter & Catenacci, 2008; Huerta-Alardin et al, 2005; Brown et al, 2004; Polderman, 2004). Retrospective studies have failed to demonstrate any clinical benefit from the use of urinary alkalinization (Brown et al, 2004; Polderman, 2004; Homsi et al, 1997).
    6) MANNITOL/INDICATIONS
    a) Osmotic diuretic used in the management of rhabdomyolysis and myoglobinuria (Zimmerman & Shen, 2013).
    7) RHABDOMYOLYSIS/MYOGLOBINURIA
    a) ADULT: TEST DOSE: (for patients with marked oliguria or those with inadequate renal function) 0.2 g/kg IV as a 15% to 25% solution infused over 3 to 5 minutes to produce a urine flow of at least 30 to 50 mL/hr; a second test dose may be given if urine flow does not increase within 2 to 3 hours. The patient should be reevaluated if there is inadequate response following the second test dose (Prod Info MANNITOL intravenous injection, 2009). TREATMENT DOSE: 50 to 100 g IV as a 15% to 25% solution may be used. The rate should be adjusted to maintain urinary output at 30 to 50 mL/hour (Prod Info mannitol IV injection, urologic irrigation, 2006) OR 300 to 400 mg/kg or up to 100 g IV administered as a single dose (Prod Info MANNITOL intravenous injection, 2009).
    b) PEDIATRIC: Dosing has not been established in patients less than 12 years of age(Prod Info Mannitol intravenous injection, 2009). TEST DOSE (for patients with marked oliguria or those with inadequate renal function): 0.2 g/kg or 6 g/m(2) body surface area IV as a 15% to 25% solution infused over 3 to 5 minutes to produce a urine flow of at least 30 to 50 mL/hr; a second test dose may be given if urine flow does not increase; TREATMENT DOSE: 0.25 to 2 g/kg or 60 g/m(2) body surface area IV as a 15% to 20% solution over 2 to 6 hours; do not repeat dose for persistent oliguria (Prod Info MANNITOL intravenous injection, 2009).
    8) ADVERSE EFFECTS
    a) Fluid and electrolyte imbalance, in particular sodium and potassium; expansion of the extracellular fluid volume leading to pulmonary edema or CHF exacerbations(Prod Info MANNITOL intravenous injection, 2009).
    9) PRECAUTION
    a) Contraindicated in well-established anuria or impaired renal function not responding to a test dose, pulmonary edema, CHF, severe dehydration; caution in progressive oliguria and azotemia; do not add to whole blood for transfusions(Prod Info Mannitol intravenous injection, 2009); enhanced neuromuscular blockade observed with tubocurarine(Miller et al, 1976).
    10) MONITORING PARAMETERS
    a) Renal function, urine output, fluid balance, serum potassium, serum sodium, and serum osmolality (Prod Info Mannitol intravenous injection, 2009).
    E) CONTRAINDICATED TREATMENT
    1) Do NOT administer adrenergic amines, which further increase myocardial irritability and produce refractory ventricular arrhythmias (Dreisbach, 1983; Bryson, 1986).
    F) CHOLESTYRAMINE
    1) Cholestyramine (4 grams every eight hours) has accelerated excretion of kepone and chlordane in excessively exposed workers, and probably would have a similar effect on other slowly excreted organochlorines which are trapped in the enterohepatic circulation (Cohn et al, 1978) Garrettson et al, 1984-85).
    2) The effects of cholestyramine and activated charcoal were compared in acutely poisoned mice, at doses of 2.25 g/kg each. It was found that cholestyramine was more effective than charcoal in preventing absorption of lindane. However, this dose of cholestyramine would be not be feasible for humans (Kassner et al, 1993).
    G) CHRONIC POISONING
    1) It was reported that symptoms of chronic exposure to lindane, including porphyria, improved following treatment with IV and oral edetic acid therapy (EDTA) or BAL (Peters et al, 1982).

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) 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. Rescue personnel and bystanders should avoid direct contact with contaminated skin, clothing, or other objects (Burgess et al, 1999). Since contaminated leather items cannot be decontaminated, they should be discarded (Simpson & Schuman, 2002).
    E) Medical personnel should wear protective clothing to prevent secondary contamination. Be prepared to collect any vomitus or excreta in a manner to prevent further contamination; bag and treat as hazardous waste.
    6.7.2) TREATMENT
    A) 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) 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).
    B) 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).
    C) 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) OBSERVATION REGIMES
    1) All symptomatic patients should be carefully observed and treated in a controlled setting until all acute clinical signs and symptoms have resolved.
    B) Treatment should include recommendations listed in the ORAL EXPOSURE section when appropriate.

Dermal Exposure

    6.9.1) DECONTAMINATION
    A) DERMAL DECONTAMINATION
    1) Lindane was more effectively removed from human stratum corneum with soap and water than with water alone in a study of radiolabelled lindane emulsion (Nitsche et al, 1984).
    2) 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).
    3) 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. Rescue personnel and bystanders should avoid direct contact with contaminated skin, clothing, or other objects (Burgess et al, 1999). Since contaminated leather items cannot be decontaminated, they should be discarded (Simpson & Schuman, 2002).
    B) PERSONNEL PROTECTION
    1) Medical personnel should wear protective clothing to prevent secondary contamination. Be prepared to collect any vomitus or excreta in a manner to prevent further contamination; bag and treat as hazardous waste.
    6.9.2) TREATMENT
    A) 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) 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).
    B) 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).
    C) CONTRAINDICATED TREATMENT
    1) Do NOT administer adrenergic amines, which further increase myocardial irritability and produce refractory ventricular arrhythmias (Dreisbach, 1983; Bryson, 1986).
    D) CHOLESTYRAMINE
    1) Cholestyramine (4 grams every eight hours) has accelerated excretion of kepone and chlordane in excessively exposed workers, and probably would have a similar effect on other slowly excreted organochlorines which are trapped in the enterohepatic circulation (Cohn et al, 1978) Garrettson et al, 1984-85).
    E) CHRONIC POISONING
    1) Peters et al (1982) report that symptoms of chronic exposure to hexachlorobenzene improved following treatment with IV and oral edetic acid therapy.
    F) DIALYSIS PROCEDURE
    1) HEMODIALYSIS - Has not proven effective.
    2) HEMOPERFUSION - Effectiveness not known due to limited experience.
    3) Exchange transfusion, extracorporeal and peritoneal dialysis have not proven effective in management of these poisonings. There has been little or no experience with charcoal hemoperfusion in organochlorine poisonings.
    G) Treatment should include recommendations listed in the ORAL EXPOSURE section when appropriate.

Enhanced Elimination

    A) HEMODIALYSIS
    1) Dialysis is probably ineffective due to large volume of distribution.
    B) EXCHANGE TRANSFUSION
    1) Exchange transfusion is probably ineffective.
    C) HEMOPERFUSION
    1) A case report cites a 16-year-old girl with an initial serum lindane concentration of 0.25 microgram/milliliter which decreased to 0.05 microgram/milliliter after XAD-4 amberlite hemoperfusion for 2 hours (Daerr et al, 1985).
    D) CHOLESTYRAMINE
    1) Cholestyramine (4 grams every eight hours) has accelerated excretion of kepone and chlordane in excessively exposed workers, and probably would have a similar effect on other slowly excreted organochlorines which are trapped in the enterohepatic circulation (Cohn et al, 1978) Garrettson et al, 1984-85).

Case Reports

    A) ROUTE OF EXPOSURE
    1) DERMAL
    a) Lee et al (1976) reported the following cases of total body application of 1 percent lindane:
    AGEDURATION OF EXPOSURESYMPTOMS
    4-year-old1 timeVomiting, Seizures, Coma
    7-year-old40 hoursSeizures
    8-year-old18 daysSeizures
    10-year-old1 timeLethargy
    37-year-old3 daysDizziness, Amblyopia

    2) A 4-month-old had one total body application of 1% lindane. Twenty-four hours later he was found to have increased muscle tone and posturing indicating a possible seizure. Serum lindane concentration 46 hours after the application was 0.1 mcg/mL (Pramanik & Hansen, 1979).
    3) An 18-month-old received two total body applications of 1 percent lindane. Twelve hours after second treatment a sudden seizure occurred. Serum lindane concentration was 0.45 mcg/mL (Telch & Jarvis, 1982).
    4) A 2-month-old was found dead. In retrospect, it was determined that a total body application of 1 percent lindane had been used once 24 hours earlier. Post mortem serum lindane concentrations were 0.033 mcg/mL. It is doubtful whether this case represents lindane toxicity (Davies et al, 1983).
    5) A 3-year-old boy developed nausea, vomiting, tonoclonic activity, followed by coma after applying 1 percent lindane cream shortly after bathing for the treatment of scabies. His blood lindane level at 72 hours after application was 54 ng/mL (Friedman, 1987).
    6) A 24-year-old woman applied lindane shampoo, 2 ounces followed by 1 ounce, over excoriated skin over the course of 1 hour for presumed lice. She presented to the ED 20 hours later with agitation, athetotic movements, involuntary total body movements, and visual hallucinations. No significant medical or psychiatric history was determined. All symptoms resolved within 48 hours of onset (Fischer, 1994).
    7) CASE SERIES: Based on a five-year retrospective review of lindane exposures (total = 528) reported to six poison centers in Texas, the most frequent symptoms associated with inadvertent dermal exposure were erythema, dermal irritation, or pain and nausea. Most cases resulted from inadvertent or misuse of the product used in the treatment of pediculosis. Of those cases (total = 328) with outcome information available, no health effects were reported for 199 (60.7%) of cases, only 3 cases developed major effects, and no deaths were reported (Forrester et al, 2004).
    B) ACUTE EFFECTS
    1) ORAL EXPOSURE: A 27-month-old ate 95 percent lindane pellets. Seizures occurred thirty minutes later followed by coma and respiratory depression (Nicholls, 1958).
    2) A family of five ate dessert that was sprinkled with lindane crystals. In a 23-year-old who was pregnant, a seizure occurred about one and a half hours later. Three of the other four also had seizures. All patients suffered gastrointestinal symptoms (Wilson, 1959).
    3) A 30-month-old ate 95 percent lindane pellets. Seizures occurred thirty minutes later. Serum lindane concentrations were 0.84 mcg/mL (Starr & Clifford, 1972).
    4) A 35-year-old ate lindane-contaminated broccoli. Thirty minutes later he had a seizure. Subsequently, acidosis, muscle weakness, acute renal failure, myoglobinuria, hypertension, and anemia developed. Serum lindane concentration was 0.60 mcg/mL (Munk & Nantel, 1977).
    5) A 16-year-old took an unknown amount of 1 percent lindane. Seizures, coma, and respiratory depression developed at an unknown time afterwards. The serum concentration of lindane was 0.206 mcg/mL (Davies et al, 1983).
    6) The following cases of oral exposure to a lindane-solvent mixture were reported (Jaeger et al, 1984):
    AGE (yrs)AMT OF MIXTURESYMPTOMSONSET
    21 100 gVomiting Refractory30 min
    Seizures, Rhabdomyolysis90 min
    42100 gSeizures, Coma, Pulmonary, edema30 min
    410 gVomiting, Seizures30 min
    3.510 gVomiting, Agitation, Seizures2 hr
    35 to 10 gVomiting, Seizures15 min

    7) A 16-year-old drank 150 mL of 2.5 percent (3.75 g) lindane. She developed lethargy and a resting tremor. Her peak serum concentration was 0.25 mg/mL which decreased to 0.05 mg/mL following a 2 hour hemoperfusion with XAD-4 amberlite (Daerr et al, 1985)
    8) A 43-year-old woman ingested 8 ounces of 20 percent lindane. Shortly after ingestion bradycardia, hypotension, seizures, coma, and metabolic acidosis developed. At 8 hours post-ingestion DIC was noted.
    a) Subsequently rhabdomyolysis, myoglobinuria, and acute renal failure developed, resulting in death 11 days after admission (Rao et al, 1988).
    9) The following cases of lindane exposure were reported (Kurt et al, 1986):
    a) A 41-year-old took two ounces of 1 percent (600 mg) lindane orally accidentally. He received ipecac but had a seizure when vomiting. He eventually died of adult respiratory distress syndrome and severe anoxia. His peak serum lindane concentration was 1.3 mcg/mL.
    b) A 39-year-old ingested three tablespoonfuls of 1 percent (450 mg) lindane over one day. Her symptoms were headache, dizziness and nausea.
    c) A 4-year-old was given 1 tablespoonful of 1 percent lindane three times (450 mg) in one day. His symptoms were initial lethargy followed by a seizure.
    d) An 8-year-old developed vomiting after one tablespoonful of 1 percent (150 mg) lindane.
    10) Morgan & Murray (1990) reported a large poison center based experience of lindane toxicity. They found 2 children had seizures after 100 mg (amount and concentration of product not specified) and 1 adult seized after 900 mg (amount and concentration of product not specified).
    11) A 32-year-old female, with a history of a seizure disorder controlled by phenytoin, accidently ingested 10 to 25 mL of a 1 percent lindane solution (Burton et al, 1991).
    a) The patient experienced spontaneous vomiting and a generalized seizure of 1 to 2 minutes duration. The patient had a serum lindane level of 0.13 mcg/mL and serum phenytoin level of 7 mg/dL.
    12) CASE SERIES: Based on a five-year retrospective review of lindane exposures (total = 528) reported to six poison centers in Texas, the most frequent symptoms associated with inadvertent ingestion of lindane included vomiting, nausea, and throat irritation. Most cases resulted from inadvertent or misuse of the product used in the treatment of pediculosis. Of those cases (total = 328) with outcome information available, no health effects were reported for 199 (60.7%) of cases, only 3 cases developed major effects, and no deaths were reported (Forrester et al, 2004).
    13) A CDC report identified 870 cases of unintentional lindane ingestion during 1998-2003. The Toxic Exposure Surveillance System (TESS) reported 857 symptomatic cases; 778 low severity cases (91%), 71 moderate severity cases (8%), and 8 high severity cases (1%) were observed. The following sign and symptoms were reported: vomiting (59%), nausea (18%), oral irritation (19%), abdominal cramping (4%), cough (4%), and seizure (3%). The Sentinel Event Notification System for Occupational Risks-Pesticides (SENSOR-Pesticides) reported 13 symptomatic cases; 4 cases (31%) were classified as definite lindane exposures, 2 (15%) as probable, 6 (46%) as possible, and one (8%) as suspicious. Eight low severity cases (62%), 3 moderate severity cases (23%), and 2 high severity cases (15%) were observed. The following sign and symptoms were reported: vomiting (69%), nausea (46%), headache (23%), seizure (23%), abdominal cramping (8%), and confusion (8%) (Anon, 2005).
    14) In a review article, 67 cases of severe adverse effects (AE), including 20 death (including 7 children and 4 patients aged 65 and older) following the labeled use (47 patients), excessive use (17 patients), and ingestion (3 cases) of lindane were identified. Seizures and respiratory problems developed in the majority of patients who developed severe AEs using the labeled dose (defined as one 4-minute application of shampoo or one 8 to 12 hours of single total body application of lindane lotion 1%). Three patients ingested lindane. The most common adverse effects were tremor, vertigo, paresis, and aphasia. The following adverse effects were also reported: shortness of breath (n=3), visual changes (n=2), anaphylactic reaction (n=1), aplastic anemia (n=3), leukocytosis (n=1), tachycardia (n=1), alopecia (n=1), acute generalized exanthematous pustulosis (n=1) (Nolan et al, 2012).
    C) INFANT
    1) ORAL AND DERMAL EXPOSURE: A 13-month-old received total body applications of 1 percent lindane daily for 2 weeks. She was given approximately 50 mg of lindane orally following which she had a seizure (Lee et al, 1976).
    2) A 1-year-old was given 14 daily total body applications of 1 percent lindane followed by an accidental oral administration of 5 mL of 1 percent lindane. This resulted in vomiting. A focal seizure occurred 12 hours later followed by coma and hypotonia (Wheeler, 1977).

Summary

    A) TOXICITY: Most toxicity occurs after repeated or prolonged applications, but can also occur after ingestions and has also been reported after a single proper dermal application. Ingestion of as little as 5 mL of 1% lindane has caused seizures in a child. In an adult, ingestion of 45 mg caused seizures. An adult survived an ingestion of 1035 g.
    B) THERAPEUTIC DOSE: Typically applied as a 1% formulation to the hair or body; 30 mL is generally sufficient for an adult. The lotion is left on the skin for 8 to 12 hours and then washed off.

Therapeutic Dose

    7.2.1) ADULT
    A) LOTION
    1) Apply approximately 1 ounce topically from the neck down and wash off in 8 to 12 hours; DO NOT retreat; MAX: 2 ounces (Prod Info lindane 1% topical lotion, 2011)
    B) SHAMPOO
    1) Apply about 1 to 2 ounces to dry hair and allow to sit for 4 minutes; add small quantities of water until a good lather forms and then immediately rinse out shampoo; AVOID contact with other body surfaces; DO NOT retreat; MAX: 2 ounces (Prod Info lindane 1% topical shampoo, 2011)
    7.2.2) PEDIATRIC
    A) LOTION
    1) Apply approximately 1 ounce topically from the neck down and wash off in 8 to 12 hours; DO NOT retreat; MAX: 2 ounces (Prod Info lindane 1% topical lotion, 2011)
    2) USE EXTREME CAUTION in patients who weigh less than 50 kg, especially in infants (Prod Info lindane 1% topical lotion, 2011).
    B) SHAMPOO
    1) Apply about 1 to 2 ounces to dry hair and allow to sit for 4 minutes; add small quantities of water until a good lather forms and then immediately rinse out shampoo; AVOID contact with other body surfaces; DO NOT retreat; MAX: 2 ounces (Prod Info lindane 1% topical shampoo, 2011)
    2) USE CAUTION in patients who weigh less than 50 kg, especially in infants (Prod Info lindane 1% topical shampoo, 2011).

Minimum Lethal Exposure

    A) ACUTE LETHAL STUDIES
    1) Ingestion of 8 ounces of 20% lindane resulted in death in adults (Rao et al, 1988) Woolf et al, 1987).
    2) The lethal dose of lindane is estimated to be 125 mg/kg (HSDB , 2000).
    3) It is believed that 28 grams is the mean fatal dose of technical grade lindane in humans (Baselt, 1997).
    4) An adult male died after ingesting 2 ounces of 1% lindane lotion (Baselt, 1997).
    5) A man with scabies developed progressive severe neurotoxicity after a single application of a thin film of lindane (1%) to his skin from the neck to the toes. Prior to the lindane administration, fresh scratch marks were noted throughout the patient's body which might have increased dermal absorption of lindane significantly. In addition, the physician's order did not include instructions to wash lindane from the skin after its application. He experienced infectious complications and died 50 days after admission (Sudakin, 2007).
    6) In a review article, 67 cases of severe adverse effects, including 20 death (including 7 children and 4 patients aged 65 and older) following the labeled use, excessive use, and ingestion of lindane were identified. Seizures and respiratory problems developed in the majority of patients using the labeled dose (defined as one 4-minute application of shampoo or one 8 to 12 hours of single total body application of lindane lotion 1%). Three patients ingested lindane. The most common adverse effects were tremor, vertigo, paresis, and aphasia. The following adverse effects were also reported: shortness of breath (n=3), visual changes (n=2), anaphylactic reaction (n=1), aplastic anemia (n=3), leukocytosis (n=1), tachycardia (n=1), alopecia (n=1), acute generalized exanthematous pustulosis (n=1) (Nolan et al, 2012).
    B) CHRONIC LETHAL STUDIES
    1) Lindane and other isomers of hexachlorocyclohexane may be reasonably anticipated to be carcinogens (ACGIH, 1991). The ACGIH places lindane in Category A3: confirmed animal carcinogen with unknown relevance to humans (ACGIH, 2000).
    2) "NTP anticipated human carcinogen" (NTP, 2000).
    3) Firm evidence does not exist linking lindane exposure in humans to development of aplastic anaemia or leukaemia (IARC , 1998). The IARC classifies Lindane in Group 2B: The agent (mixture) is possibly carcinogenic to humans.The exposure circumstance entails exposures that are possibly carcinogenic to humans (IARC, 1987).
    a) In one study, workers involved in manufacturing lindane showed no evidence of chromosomal aberrations (IARC , 1998).
    C) CHILDREN
    1) ACUTE LETHAL STUDIES
    a) Ingestion of 6 mg/kg in a child has resulted in death (Falk & Hinrichs, 1957).
    b) A 2-month old died after total body application of 1 percent lindane that was left on for 18 hours (Davies et al, 1983).
    c) Excessive application of a 1% lindane lotion resulted in death for a 2-month-old, 4.5 kg child. The lindane concentration in the brain was 110 ppb, which was 3 times the amount found in the blood (HSDB , 2000).
    d) In a review article, 67 cases of severe adverse effects, including 20 death (including 7 children and 4 patients aged 65 and older) following the labeled use, excessive use, and ingestion of lindane were identified. Seizures and respiratory problems developed in the majority of patients using the labeled dose (defined as one 4-minute application of shampoo or one 8 to 12 hours of single total body application of lindane lotion 1%). Three patients ingested lindane. The most common adverse effects were tremor, vertigo, paresis, and aphasia. The following adverse effects were also reported: shortness of breath (n=3), visual changes (n=2), anaphylactic reaction (n=1), aplastic anemia (n=3), leukocytosis (n=1), tachycardia (n=1), alopecia (n=1), acute generalized exanthematous pustulosis (n=1) (Nolan et al, 2012).

Maximum Tolerated Exposure

    A) Toxic doses and the effects of lindane and related isomers vary with route and rate of absorption. Lindane, the gamma isomer of hexachlorocyclohexane, is more acutely toxic, while the beta and alpha isomers are retained in the body tissues longer and pose a greater chronic risk (Clayton & Clayton, 1994).
    B) ACUTE NON-LETHAL STUDIES
    1) An oral dose of 45 mg (approximately 0.65 mg/kg) has caused seizures (Lewis, 1996).
    2) CASE REPORT: A 24-year-old man, found 20 minutes after ingesting 100 mL of a lindane hair shampoo (calculated amount of lindane 1035 grams), developed seizures. Following symptomatic therapy he recovered without sequelae (Zilker et al, 1999).
    3) CASE REPORT: A 56-year-old man developed vomiting, seizures, cardiac dysrhythmias, and neurological abnormalities after ingesting about 12 ounces (about 355 mL) of an insecticide containing 20% lindane. Although he recovered following supportive care, he committed suicide 12 days after the initial presentation (Wiles et al, 2015).
    4) Large ingestions of 1% lindane (unknown amount in Davies' (1983) case) and greater than 2 ounces in Kurt (1986) have resulted in significant toxicity.
    5) In a review article, 67 cases of severe adverse effects (AE), including 20 death (including 7 children and 4 patients aged 65 and older) following the labeled use (47 patients) , excessive use (17 cases), and ingestion (3 cases) of lindane were identified. Seizures and respiratory problems developed in the majority of patients who developed severe AE after using the labeled dose (defined as one 4-minute application of shampoo or one 8 to 12 hours of single total body application of lindane lotion 1%). Three patients ingested lindane. The most common adverse effects were tremor, vertigo, paresis, and aphasia. The following adverse effects were also reported: shortness of breath (n=3), visual changes (n=2), anaphylactic reaction (n=1), aplastic anemia (n=3), leukocytosis (n=1), tachycardia (n=1), alopecia (n=1), acute generalized exanthematous pustulosis (n=1) (Nolan et al, 2012).
    C) CHRONIC EXPOSURE STUDY
    1) Humans tolerated purified gamma-lindane with no ill effects at 40 mg/man/day for 14 days, but the same dosage of the technical grade caused vertigo, headache, and diarrhea. In another study, lindane at 45 mg given three times per day for three days induced convulsions in one patient (ACGIH, 1991).
    D) CHILDREN
    1) ACUTE NON-LETHAL STUDIES
    a) As little as two total body applications of 1% Kwell on successive days in an 18-month-old child have resulted in a seizure (Telch & Jarvis, 1982).
    b) A 10-year-old patient suffered coma after a general total body application of 1% lindane (Lee & Groth, 1977).
    c) CASE REPORT: A 3-year-old boy developed generalized seizures in his sleep with transient loss of sensorium about 1 hour after ingesting 10 mL of lindane lotion. He presented with drowsiness, a GCS (Glasgow coma score) of 10/15, and papular skin lesions. Following supportive care, he recovered 24 hours later (Ramabhatta et al, 2014).
    d) One 4-month-old developed increased muscle tone, tonic posturing, and poor orientation to visual stimuli after a total body application of 1% lindane was left on for 24 hours (Pramanik & Hansen, 1979).
    e) Ingestion of as little as 5 mL of 1% lindane has caused respiratory depression and/or seizures in children aged 1 to 4 years (Nordt & Chew, 1999; (Aks et al, 1995) Jaeger et al, 1983).
    f) Seizures were reported in a 1-year-old child who was given one teaspoonful of 1% lindane (50 mg) in addition to topical application of 100 mg (Wheeler, 1977).
    g) Oral doses of about 50 mg/kg have caused severe toxicity in young children who ingested lindane pellets (Stormont & Conley, 1955).
    E) ANIMAL DATA
    1) The maximally tolerated dose for rats fed lindane in the diet is 1500 ppm for 90 days (Proctor et al, 1988).
    2) Minimal effects were seen in several species of laboratory animals exposed to an average concentration of 0.7 mg/m(3) of lindane, 7 hours per day, 5 days per week, for approximately 1 year. Rats exposed to 0.19 mg/m(3) continuously for 655 days, 24 hours per day, exhibited no abnormal pathology (ACGIH, 1991).
    3) Oral doses of 60 mg/kg produced seizures in 17.6% of rats; 100 mg/kg in 69.4 percent, and 150 mg/kg in 95.5%. The lethality rate at the highest dose was 18 percent (Tusell et al, 1987).
    4) Lindane is more toxic than DDT or dieldrin in some domestic animals, including calves (Lewis, 1996).
    5) Lindane reduced the pregnancy rate in ewes through a diet of 1 mg/kg/day (Beard et al, 1999).

Serum Plasma Blood Concentrations

    7.5.2) TOXIC CONCENTRATIONS
    A) TOXIC CONCENTRATION LEVELS
    1) CONCENTRATION LEVEL
    a) CASE REPORTS
    1) The toxic concentration of lindane in blood is 0.5 mcg/mL (HSDB , 2000).
    2) Lindane serum levels appear to correlate with severity of symptoms in acute ingestions (Aks et al, 1995).
    3) ADULT: A 32-year-old woman, with a history of a seizure disorder controlled by phenytoin, experienced spontaneous vomiting and a generalized seizure. The patient had a measured lindane level of 0.13 mcg/mL following ingestion of 10 to 25 mL of a 1% lindane solution (Burton et al, 1991).
    4) ADULT: An admission lindane serum level was reported to be 42.69 mcg/L (normal less than 2 mcg/L) approximately 20 minutes after an ingestion of 100 mL Quellada hair shampoo (total lindane 1035 grams) in a 24-year-old man (Zilker et al, 1999).
    5) CHILDREN: Lindane in plasma was 0.29 mg/L 5 hours after ingestion by a young boy. A serum level of 0.45 mg/L was seen in an 18-month old child after a lindane-containing lotion was applied to the whole body. Serum lindane was 0.84 mg/L 2 hours after a young girl ingested 1.6 grams of lindane. All these patients developed seizures (Baselt, 1997).
    2) OCCUPATIONAL
    a) In a study of occupational exposure, whole blood lindane levels of greater than 0.02 part per million (20 nanograms/mL) correlated with clinical symptoms of toxicity and EEG changes (Czegledi-Janko & Avar, 1970).
    3) ANIMAL DATA
    a) The threshold-blood concentration for initiation of seizures in rats was 1.3 to 1.7 mcg/mL (Tusell et al, 1987).

Workplace Standards

    A) ACGIH TLV Values for CAS58-89-9 (American Conference of Governmental Industrial Hygienists, 2010):
    1) Editor's Note: The listed values are recommendations or guidelines developed by ACGIH(R) to assist in the control of health hazards. They should only be used, interpreted and applied by individuals trained in industrial hygiene. Before applying these values, it is imperative to read the introduction to each section in the current TLVs(R) and BEI(R) Book and become familiar with the constraints and limitations to their use. Always consult the Documentation of the TLVs(R) and BEIs(R) before applying these recommendations and guidelines.
    a) Adopted Value
    1) Lindane
    a) TLV:
    1) TLV-TWA: 0.5 mg/m(3)
    2) TLV-STEL:
    3) TLV-Ceiling:
    b) Notations and Endnotes:
    1) Carcinogenicity Category: A3
    2) Codes: Skin
    3) Definitions:
    a) A3: Confirmed Animal Carcinogen with Unknown Relevance to Humans: The agent is carcinogenic in experimental animals at a relatively high dose, by route(s) of administration, at site(s), of histologic type(s), or by mechanism(s) that may not be relevant to worker exposure. Available epidemiologic studies do not confirm an increased risk of cancer in exposed humans. Available evidence does not suggest that the agent is likely to cause cancer in humans except under uncommon or unlikely routes or levels of exposure.
    b) Skin: This refers to the potential significant contribution to the overall exposure by the cutaneous route, including mucous membranes and the eyes, either by contact with vapors or, of likely greater significance, by direct skin contact with the substance. It should be noted that although some materials are capable of causing irritation, dermatitis, and sensitization in workers, these properties are not considered relevant when assigning a skin notation. Rather, data from acute dermal studies and repeated dose dermal studies in animals or humans, along with the ability of the chemical to be absorbed, are integrated in the decision-making toward assignment of the skin designation. Use of the skin designation provides an alert that air sampling would not be sufficient by itself in quantifying exposure from the substance and that measures to prevent significant cutaneous absorption may be warranted. Please see "Definitions and Notations" (in TLV booklet) for full definition.
    c) TLV Basis - Critical Effect(s): Liver dam; CNS impair
    d) Molecular Weight: 290.85
    1) For gases and vapors, to convert the TLV from ppm to mg/m(3):
    a) [(TLV in ppm)(gram molecular weight of substance)]/24.45
    2) For gases and vapors, to convert the TLV from mg/m(3) to ppm:
    a) [(TLV in mg/m(3))(24.45)]/gram molecular weight of substance
    e) Additional information:

    B) NIOSH REL and IDLH Values for CAS58-89-9 (National Institute for Occupational Safety and Health, 2007):
    1) Listed as: Lindane
    2) REL:
    a) TWA: 0.5 mg/m(3)
    b) STEL:
    c) Ceiling:
    d) Carcinogen Listing: (Not Listed) Not Listed
    e) Skin Designation: [skin]
    1) Indicates the potential for dermal absorption; skin exposure should be prevented as necessary through the use of good work practices and gloves, coveralls, goggles, and other appropriate equipment.
    f) Note(s):
    3) IDLH:
    a) IDLH: 50 mg/m3
    b) Note(s): Not Listed

    C) Carcinogenicity Ratings for CAS58-89-9 :
    1) ACGIH (American Conference of Governmental Industrial Hygienists, 2010): A3 ; Listed as: Lindane
    a) A3 :Confirmed Animal Carcinogen with Unknown Relevance to Humans: The agent is carcinogenic in experimental animals at a relatively high dose, by route(s) of administration, at site(s), of histologic type(s), or by mechanism(s) that may not be relevant to worker exposure. Available epidemiologic studies do not confirm an increased risk of cancer in exposed humans. Available evidence does not suggest that the agent is likely to cause cancer in humans except under uncommon or unlikely routes or levels of exposure.
    2) EPA (U.S. Environmental Protection Agency, 2011): Not Assessed under the IRIS program. ; Listed as: gamma-Hexachlorocyclohexane (gamma-HCH)
    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 ; Listed as: Lindane
    5) MAK (DFG, 2002): Category 4 ; Listed as: Lindane
    a) Category 4 : Substances with carcinogenic potential for which genotoxicity plays no or at most a minor part. No significant contribution to human cancer risk is expected provided the MAK value is observed. The classification is supported especially by evidence that increases in cellular proliferation or changes in cellular differentiation are important in the mode of action. To characterize the cancer risk, the manifold mechanisms contributing to carcinogenesis and their characteristic dose-time-response relationships are taken into consideration.
    6) NTP (U.S. Department of Health and Human Services, Public Health Service, National Toxicology Project ): Not Listed

    D) OSHA PEL Values for CAS58-89-9 (U.S. Occupational Safety, and Health Administration (OSHA), 2010):
    1) Listed as: Lindane
    2) Table Z-1 for Lindane:
    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: 0.5
    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) References: ACGIH, 1991 Budavari, 1996 CHRIS, 2000 Clayton & Clayton, 1994 ) (Extoxnet, 2000; Hayes & Laws, 1991 ITI, 1995 Lewis, 2000 RTECS, 2000 ) Tusell et al, 1987 Note: All values below are from RTECS (2000) unless otherwise specified.
    1) LD50- (INTRAPERITONEAL)MOUSE:
    a) 125 mg/kg
    2) LD50- (ORAL)MOUSE:
    a) 44 mg/kg
    b) 86 mg/kg (Hayes & Laws, 1991)
    c) 340-562 mg/kg (Hayes & Laws, 1991)
    d) 59-246 mg/kg (Extoxnet, 2000)
    3) LD50- (SKIN)MOUSE:
    a) 300 mg/kg (Hayes & Laws, 1991)
    4) LD50- (INTRAPERITONEAL)RAT:
    a) 35 mg/kg
    5) LD50- (ORAL)RAT:
    a) 76 mg/kg
    b) Male, 88 mg/kg (Budavari, 1996)
    c) 88 mg/kg (Hayes & Laws, 1991)
    d) Female, 91 mg/kg (Budavari, 1996)
    e) 80-125 mg/kg (Tusell et al, 1987)
    f) 88-200 mg/kg (ACGIH, 1991)
    g) 88-270 mg/kg (Extoxnet, 2000)
    h) 50-500 mg/kg (CHRIS, 2000)
    i) 125 mg/kg (Hayes & Laws, 1991)
    j) 190 mg/kg (Hayes & Laws, 1991)
    k) 200 mg/kg (Hayes & Laws, 1991)
    6) LD50- (SKIN)RAT:
    a) 414 mg/kg
    b) 900 mg/kg (Hayes & Laws, 1991)
    c) 1000 mg/kg (Hayes & Laws, 1991)
    d) 500 mg/kg (Hayes & Laws, 1991; Lewis, 2000)
    e) 500-1000 mg/kg (ACGIH, 1991)
    7) LD50- (SUBCUTANEOUS)RAT:
    a) 50 mg/kg (Hayes & Laws, 1991)
    8) TCLo- (INHALATION)MOUSE:
    a) 1 mg/m(3) for 80D intermittent -- bone marrow changes
    9) TCLo- (INHALATION)RAT:
    a) 1700 mcg/m(3) for 17W intermittent -- serum composition changes; phosphatases; dehydrogenases

Pharmacologic Mechanism

    A) Lindane is an ectoparasiticide and ovicide effective against Sarcoptes scabiei and their ova. It exerts its parasiticidal action by direct absorption into the parasites and their ova (Prod Info Kwell(R), lindane, 1994).

Toxicologic Mechanism

    A) The mechanism of action of the chlorinated hydrocarbon insecticides is not fully understood (Finkel, 1983), but many of their toxicologic effects are similar. Therefore, this review is based on the properties of chlorinated hydrocarbon insecticides in general. Effects attributed specifically to lindane are identified.
    B) The principal neurotoxic action of these compounds is that of "axon poison," affecting primarily the CNS nerve cells. Essentially, the organochlorines interfere with the normal flux of Na+ and K+ ions across the axon membrane as nerve impulses pass. This results in irritability, disturbance of mental processes, sensory aberrations, and seizures (Morgan, 1982).
    C) In vitro studies suggest that lindane's neurotoxic effects are mediated via blockade of the GABA-receptor coupled sodium channel (Pomes et al, 1994).
    D) The organochlorines do not depress cholinesterase enzymes.

Physical Characteristics

    A) Lindane is a colorless or white to yellow crystalline powder or monoclinic prism. CHRIS (2000) reports that the solid is "light to dark brown." It has a bitter taste and may have a slightly musty or aromatic odor. Pure lindane is odorless (HSDB, 2005; NIOSH, 2005; ATSDR, 1994) NTP, 2000). Lindane has more vapor activity than most organochlorine insecticides (HSDB , 2000).
    B) Lindane in its liquid form floats on water; the solid sinks (CHRIS , 2000).
    C) Lindane is the gamma isomer of 1,2,3,4,5,6-hexachlorocyclohexane (HCH). There are seven other isomers of HCH, but non-gamma isomers can no longer be made or used in the United States (ACGIH, 1991; ATSDR, 1994; Hayes & Laws, 1991).

Molecular Weight

    A) 290.85

Other

    A) ODOR THRESHOLD
    1) Lower: 0.33 ppm (OHM/TADS, 2005)
    2) Medium: 1.8 ppm (OHM/TADS, 2005)
    B) TASTE THRESHOLD
    1) Lower: 0.8 ppm (OHM/TADS, 2005)
    2) Medium: 0.2 ppm (OHM/TADS, 2005)

Clinical Effects

    11.1.3) CANINE/DOG
    A) A 9-year-old border collie developed vomiting, salivation, shaking, unsteadiness, champing movements of the jaw, constricted pupils, rapid nystagmus of low amplitude, aggressiveness, and seizure activity.
    1) This occurred following an ingestion of an unknown quantity of seed dressing containing phenylmercury acetate (2% mercury w/w) and gamma-hexachlorocyclohexane (30% w/w) (Shaw, 1988).

Treatment

    11.2.1) SUMMARY
    A) DOG
    1) Symptomatic treatment was effective in allowing recovery from acute gamma-hexachlorocyclohexane toxicity in a 9-year-old border collie (Shaw, 1988).
    B) GENERAL TREATMENT
    1) Copious washing of the exposed external surfaces may help prevent percutaneous absorption (Shaw, 1988).
    11.2.2) LIFE SUPPORT
    A) GENERAL
    1) MAINTAIN VITAL FUNCTIONS: Secure airway, supply oxygen, and begin supportive fluid therapy if necessary.
    11.2.4) DECONTAMINATION
    A) GASTRIC DECONTAMINATION
    1) GENERAL TREATMENT
    a) Gastric lavage and administration of activated charcoal were used to decontaminate the gut and prevent further absorption of a seed dressing containing lindane and phenylmercury acetate (Shaw, 1988).
    11.2.5) TREATMENT
    A) GENERAL TREATMENT
    1) The seizure activity was treated initially by administration of intravenous diazepam, followed by induction of anesthesia by thiopentone sodium intravenously and maintenance with halothane and oxygen mixture (Shaw, 1988).

Continuing Care

    11.4.1) SUMMARY
    11.4.1.2) DECONTAMINATION/TREATMENT
    A) DOG
    1) Symptomatic treatment was effective in allowing recovery from acute gamma-hexachlorocyclohexane toxicity in a 9-year-old border collie (Shaw, 1988).
    B) GENERAL TREATMENT
    1) Copious washing of the exposed external surfaces may help prevent percutaneous absorption (Shaw, 1988).
    11.4.2) DECONTAMINATION
    11.4.2.2) GASTRIC DECONTAMINATION
    A) GASTRIC DECONTAMINATION
    1) GENERAL TREATMENT
    a) Gastric lavage and administration of activated charcoal were used to decontaminate the gut and prevent further absorption of a seed dressing containing lindane and phenylmercury acetate (Shaw, 1988).

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, 6th ed, Am Conference of Govt Ind Hyg, Inc, Cincinnati, OH, 1991.
    14) AIHA: 2006 Emergency Response Planning Guidelines and Workplace Environmental Exposure Level Guides Handbook, American Industrial Hygiene Association, Fairfax, VA, 2006.
    15) AMA Department of DrugsAMA Department of Drugs: AMA Evaluations Subscription, American Medical Association, Chicago, IL, 1992.
    16) ATSDR: Profile for Alpha-, Beta-, Gamma-, and Delta-Hexachlorocyclohexane (Update), Agency for Toxic Substances and Disease Registry, US Dept of Health and Human Services, Atlanta, GA, 1994.
    17) ATSDR: Toxicological Profile For Alpha-, Beta-, Gamma-, and Delta-Hexachlorocyclohexane. Agency for Toxic Substances and Disease Registry (CD-ROM Version). CRC Press, Inc.. Boca Raton, FL. 1999.
    18) ATSDR: Toxicological profile for alpha-, beta-, gamma-, and delta-hexachlorocyclohexane. US Department of Health and Human Services. Washington, DC, USA. 2003.
    19) Aks S, Krantz A, & Hryhorczuk D: Acute accidental lindane ingestion in toddlers. Ann Emerg Med 1995; 26:647-651.
    20) Alm H, Tiemann U, & Torner H: Influence of organochlorine pesticides on development of mouse embryos in vitro. Reprod Toxicol 1996; 10:321-326.
    21) 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.
    22) Angerer J: Internat Arch Occup Environ Health 1983; 52:59-67.
    23) Angsubhakorn S, Bhamarapravati B, & Pradermwong A: Minimal dose and time protection by lindane (gamma-isomer) of hexachlorocyclohexane) against liver tumors induced by aflatoxin B1. Internat J Cancer 1989; 43:531-534.
    24) Anon: Centers for Disease Control and Prevention (CDC). Unintentional topical lindane ingestions -- United States, 1998-2003. MMWR Morb Mort Wkly Rep 2005; 54(21):533-535.
    25) Anon: NCI NIH DHEW Pub No (NIH) 77-814, 1977.
    26) Ansell-Edmont: SpecWare Chemical Application and Recommendation Guide. Ansell-Edmont. Coshocton, OH. 2001. Available from URL: http://www.ansellpro.com/specware. As accessed 10/31/2001.
    27) Arai M: Nippon Gangakkai Kiji 1972; 31:33.
    28) 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.
    29) Ashford R: Ashford's Dictionary of Industrial Chemicals, Wavelength Publications Ltd, London, England, 1994.
    30) Babu KA: Bull Environ Contam Toxicol 1981; 26:508-512.
    31) Bakhitova LM & Pashin YV: Gig Sanit 1984; 5:82-83.
    32) Barthel E: Zeitsch Erkt Atmungs 1974; 147:7-17.
    33) Baselt RC: Biological Monitoring Methods for Industrial Chemicals, 3rd ed, PSG Publishing Company, Littleton, MA, 1997, pp 221-223.
    34) Bata Shoe Company: Industrial Footwear Catalog, Bata Shoe Company, Belcamp, MD, 1995.
    35) Beard AP, Bartlewski PM, & Rawlings NC: Endocrine and reproductive function in ewes exposed to the organochlorine pesticides lindane or pento-chlorophenol. J Toxicol Environ Health 1999; 56:23-46.
    36) Benes V & Sram R: Ind Med 1969; 38:50-52.
    37) Berry DH, Brewster MA, & Watson R: Untoward effects associated with lindane abuse. Am J Dis Child 1987; 141:125-126.
    38) Best Manufacturing: ChemRest Chemical Resistance Guide. Best Manufacturing. Menlo, GA. 2002. Available from URL: http://www.chemrest.com. As accessed 10/8/2002.
    39) Best Manufacturing: Degradation and Permeation Data. Best Manufacturing. Menlo, GA. 2004. Available from URL: http://www.chemrest.com/DomesticPrep2/. As accessed 04/09/2004.
    40) Bezuglyy VI: Vrachebnoe Delo 1976; 3:142-145.
    41) Bhalla M & Thami GP: Reversible neurotoxicity after an overdose of topical lindane in an infant. Pediatr Derm 2004; 21(5):597-599.
    42) Bhardwaj AC & Tyagi N: Impact of organochlorine pesticide on the morphobehaviour of pantala flavescens fabr (Libellulidae, Odonata). J Environ Biol 1993; 14:89-94.
    43) Boffa MJ, Brough PA, & Ead RD: Lindane neurotoxicity. Br J Dermatol 1995; 133:1010.
    44) Boss Manufacturing Company: Work Gloves, Boss Manufacturing Company, Kewanee, IL, 1998.
    45) Brassow HL: Internat Arch Occup Environ Health 1981; 48:81-87.
    46) Breivik K & Wania F: Mass Budgets, Pathways, and Equilibrium States of Two Hexachlorocyclohexanes in the Baltic Sea Environment. Environ Sci Technol 2002; 36:1024-1032.
    47) Brophy GM, Bell R, Claassen J, et al: Guidelines for the evaluation and management of status epilepticus. Neurocrit Care 2012; 17(1):3-23.
    48) 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.
    49) Brown CV, Rhee P, Chan L, et al: Preventing renal failure in patients with rhabdomyolysis: do bicarbonate and mannitol make a difference?. J Trauma 2004; 56(6):1191-1196.
    50) Bryson PD: Organochlorines, in: Comprehensive Review in Toxicology, Aspen Publications, Rockville, MD, 1986, pp 271-273.
    51) Budavari S: The Merck Index, 12th ed, Merck & Co, Inc, Whitehouse Station, NJ, 1996.
    52) Burgess JL, Kirk M, Borron SW, et al: Emergency department hazardous materials protocol for contaminated patients. Ann Emerg Med 1999; 34(2):205-212.
    53) Burton BT, Will VK, & Brunett PH: Seizure following lindane ingestion in a patient pretreated with phenytoin (Abstract). Vet Human Toxicol 1991; 33:391.
    54) CHRIS : CHRIS Hazardous Chemical Data. US Department of Transportation, US Coast Guard. Washington, DC (Internet Version). Edition expires October/31/2000; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    55) CHRIS: CHRIS Hazardous Chemical Data. US Department of Transportation, US Coast Guard. Washington, DC (Internet Version). Edition expires 2004; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    56) Camp NE: Drug- and toxin-induced Rhabdomyolysis. J Emerg Nurs 2009; 35(5):481-482.
    57) 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.
    58) Cerey K: Dokl Soobshch-Mezhdunar Kongr Zashch Rast 8th 1975; 4:178-186.
    59) Ceron JJ, Barba A, & Camara MA: Lindane and methylparathion residue degradation in carnation byproduct (Dianthus caryophyllus, L.) under different environmental conditions. Bull Environ Contam Toxicol 1995; 54:150-157.
    60) 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.
    61) ChemFab Corporation: Chemical Permeation Guide Challenge Protective Clothing Fabrics, ChemFab Corporation, Merrimack, NH, 1993.
    62) 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.
    63) Choonara IA & Rane A: Therapeutic drug monitoring of anticonvulsants state of the art. Clin Pharmacokinet 1990; 18:318-328.
    64) Chowdhury AR: Bull Environ Contam Toxicol 1987; 38:154-156.
    65) Clayton GD & Clayton FE: Patty's Industrial Hygiene and Toxicology, Vol 2B, Toxicology, 3rd ed, John Wiley & Sons, New York, NY, 1981.
    66) Clayton GD & Clayton FE: Patty's Industrial Hygiene and Toxicology, Volume 2B, Toxicology, 4th ed, John Wiley & Sons, New York, NY, 1994.
    67) Cleemann M, Poulsen ME, & Hilbert G: Deposition of lindane in Denmark. Chemosphere 1995; 30:2039-2049.
    68) Cohn WJ, Boylan JJ, & Blanke RV: Treatment of chlordecone (kepone) toxicity with cholestyramine. N Engl J Med 1978; 298:243-248.
    69) Comasec Safety, Inc.: Chemical Resistance to Permeation Chart. Comasec Safety, Inc.. Enfield, CT. 2003. Available from URL: http://www.comasec.com/webcomasec/english/catalogue/mtabgb.html. As accessed 4/28/2003.
    70) Comasec Safety, Inc.: Product Literature, Comasec Safety, Inc., Enfield, CT, 2003a.
    71) Cranz C: Contracep Fertil Sex 1981; 9:421-423.
    72) Criddle LM: Rhabdomyolysis. Pathophysiology, recognition, and management. Crit Care Nurse 2003; 23(6):14-22, 24-26, 28.
    73) Curley A & Kimbrough R: Arch Environ Health 1969; 18:155-164.
    74) Curley A: Arch Environ Health 1969; 19:628-632.
    75) Czegledi-Janko G & Avar P: Occupational exposure to lindane: clinical and laboratory findings. Br J Ind Med 1970; 27:283-286.
    76) 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.
    77) Daerr W, Kaukel E, & Schmoldt A: Hamoperfusion-eine therapeutische alternative zue fruhbehandlung der akuten Lindan-intoxikation (German). Dtsch Med Wschr 1985; 110:1253-1255.
    78) Dalsenter PR, Faqi AS, & Webb J: Reproductive toxicity and tissue concentrations of lindane in adult male rats. Human Exp Toxicol 1996; 15:406-410.
    79) Dalsenter PR, Faqi AS, & Webb J: Reproductive toxicity and toxicokinetics of lindane in the male offspring of rats exposed during lactation. Human Exp Toxicol 1997; 16:146-153.
    80) Davies JE, Dedhia HV, & Morgade C: Lindane poisonings. Arch Dermatol 1983; 119:142-144.
    81) Diaz JE: Lindane toxicity is preventable. Acad Emerg Med 1998; 5:1126-1128.
    82) Didenko GG: Gig Sanit 1973; 38:98-99.
    83) Dikshith TSS & Datta KK: ACTA Pharmacol Toxicol 1972; 31:1-10.
    84) Dikshith TSS: Toxicology 1978; 10:55-66.
    85) Dinerman AA: Gig Sanit 1970; 35:39-42.
    86) Dreisbach RH: Handbook of Poisoning, Lange Medical Publications, Norwalk, CT, 1983, pp 88-89.
    87) DuPont: DuPont Suit Smart: Interactive Tool for the Selection of Protective Apparel. DuPont. Wilmington, DE. 2002. Available from URL: http://personalprotection.dupont.com/protectiveapparel/suitsmart/smartsuit2/na_english.asp. As accessed 10/31/2002.
    88) DuPont: Permeation Guide for DuPont Tychem Protective Fabrics. DuPont. Wilmington, DE. 2003. Available from URL: http://personalprotection.dupont.com/en/pdf/tyvektychem/pgcomplete20030128.pdf. As accessed 4/26/2004.
    89) DuPont: Permeation Test Results. DuPont. Wilmington, DE. 2002a. Available from URL: http://www.tyvekprotectiveapprl.com/databases/default.htm. As accessed 7/31/2002.
    90) Dubois M, Grosse Y, & Thome JP: Metabolic activation and DNA-adducts detection as biomarkers of chlorinated pesticide exposures. Biomarkers 1997; 2:17-24.
    91) Dzierzawski A: Bull Vet Inst Pulawy 1977; 21:85-93.
    92) EPA: EPA chemical profile on lindane, Environmental Protection Agency, Washington, DC, 1985.
    93) EPA: Pesticide Fact Handbook, Noyes Publications, Park Ridge, NJ, 1988.
    94) 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/.
    95) 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.
    96) Earl FL: in: Pesticides and the Environment: A Continuing Controversy. Inter-Am Conv Toxicol Occup Med, N Miami 1973; 253-266.
    97) Erdman AR & Dart RC: Rhabdomyolysis. In: Dart RC, Caravati EM, McGuigan MA, et al, eds. Medical Toxicology, 3rd ed. Lippincott Williams & Wilkins, Philadelphia, PA, 2004, pp 123-127.
    98) Erdman AR: Insecticides. In: Dart RC, Caravati EM, McGuigan MA, et al, eds. Medical Toxicology. 3rd ed, 3rd. Lippincott Williams & Wilkins, Philadelphia, PA, 2004.
    99) FAO/WHO: Pesticides residues in food. WHO Technical Report Series No 458, World Health Organization, Geneva, Switzerland, 1970.
    100) Fagan JE: Henoch-Schonlein purpura and benzene hexachloride (Letter). Pediatrics 1981; 67:310-311.
    101) Falk W & Hinrichs R: Jacutin poisoning in childhood. Med Klin Wschr 1957; 42:1837.
    102) Feldmann RJ & Maibach HI: Percutaneous penetration of some pesticides and herbicides in man. Toxicol Appl Pharmacol 1974; 28:126-132.
    103) Feldmann RJ & Maibach HI: Pesticide percutaneous penetration in man. J Invest Dermatol 1970; 54:435-436.
    104) Ferrando MD, Sancho E, & Andreumoliner E: Effects of lindane on Daphnia magna during chronic exposure. J Environ Sci Health B-Pestic 1995; 30:815-825.
    105) Finkel AJ: Hamilton and Hardy's Industrial Toxicology, 4th ed, John Wright, PSG Inc, Boston, MA, 1983, pp 295.
    106) Fischer TF: Lindane toxicity in a 24-year-old woman. Ann Emerg Med 1994; 24:972-974.
    107) Fitzhugh OG: J Pharmacol Exp Ther 1950; 100:59-66.
    108) Fiumara NJ & Kahn S: Contact dermatitis due to self-medication. Am Fam Physician 1983; 28:137.
    109) Forrester MB, Sievert JS, & Stanley SK: Epidemiology of lindane exposures for pediculosis reported to poison centers in texas, 1998-2002. J Toxicol - Clin Toxicol 2004; 42(1):55-60.
    110) Friedman SJ: Lindane neurotoxic reaction in nonbullous ichthyosiform erythroderma. Arch Dermatol 1987; 123:1056-1058.
    111) Ginsburg CM, Lowry W, & Reisch JS: Absorption of lindane (gamma benzene hexachloride) in infants and children. J Pediatr 1977; 91:998-1000.
    112) Gladenko I: Veterinariya (Moscow) 1970; 47:91-93.
    113) Gosselin RE, Smith RP, & Hodge HC: Clinical Toxicology of Commercial Products, 5th ed, Williams & Wilkins, Baltimore, MD, 1984.
    114) Grant WM & Schuman JS: Toxicology of the Eye, 4th ed, Charles C Thomas, Springfield, IL, 1993.
    115) Grasso C: Ig Mod 1973; 66:362-371.
    116) Guardian Manufacturing Group: Guardian Gloves Test Results. Guardian Manufacturing Group. Willard, OH. 2001. Available from URL: http://www.guardian-mfg.com/guardianmfg.html. As accessed 12/11/2001.
    117) HSDB : Hazardous Substances Data Bank. National Library of Medicine. Bethesda, MD (Internet Version). Edition expires 1996; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    118) HSDB : Hazardous Substances Data Bank. National Library of Medicine. Bethesda, MD (Internet Version). Edition expires 2000; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    119) HSDB : Hazardous Substances Data Bank. National Library of Medicine. Bethesda, MD (Internet Version). Edition expires 2004; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    120) HSDB : Hazardous Substances Data Bank. National Library of Medicine. Bethesda, MD (Internet Version). Edition expires 2005; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    121) Haas CF: Mechanical ventilation with lung protective strategies: what works?. Crit Care Clin 2011; 27(3):469-486.
    122) Hall RCW & Hall RCW: Long-term psychological and neurological complications of lindane poisoning. Psychosomatics 1999; 40:513-517.
    123) Hanada M: Gann 1976; 64:511-513.
    124) Harbison RM: Hamilton and Hardy's Industrial Toxicology, 5th ed, Mosby, St. Louis, MO, 1998.
    125) Hartley D & Kidd H: The Agrochemicals Handbook, 2nd ed, The Royal Society of Chemistry, Nottingham, England, 1987.
    126) Hassoun EA & Stohs SJ: Comparative teratological studies on TCDD, endrin and lindane in C57BL/6J and DBA/2J mice. Comp Biochem Physiol 1996; 113:393-398.
    127) Hathaway GJ, Proctor NH, & Hughes JP: Chemical Hazards of the Workplace, 4th ed, Van Nostrand Reinhold Company, New York, NY, 1996.
    128) Hay A & Singer CRJ: Wood preservatives, solvents, and thrombocytopenic purpura (Letter). Lancet 1991; 338:766.
    129) Hayes WJ & Laws ER Jr: Handbook of Pesticide Toxicology, Vol 1-3, Academic Press, Inc, San Diego, CA, 1991.
    130) Hegenbarth MA & American Academy of Pediatrics Committee on Drugs: Preparing for pediatric emergencies: drugs to consider. Pediatrics 2008; 121(2):433-443.
    131) Hiroshi N: Gann 1972; 63:393.
    132) Holland MG: Insecticides: Organic Chlorines, Pyrethrins/Pyrethroids, and DEET. In: Flomenabaum N, Goldfrank L, Hoffman, R, et al. eds. Goldfrank's Toxicologic Emergencies. 8th ed, 8th. McGraw-Hill Professional, Columbus, OH, 2006.
    133) Homsi E, Barreiro MF, Orlando JM, et al: Prophylaxis of acute renal failure in patients with rhabdomyolysis. Ren Fail 1997; 19(2):283-288.
    134) Hoshizaki H, Niki Y, & Tajima H: A case of leukemia following exposure to insecticide. Nippon Ketsueki Gakkai Zasshi 1969; 32:672-677.
    135) Hosler J: Topical application of lindane cream (Kwell(R)) and antipyrine metabolism. J Invest Dermatol 1980; 74:51-53.
    136) Howard PH, Boethling RS, & Jarvis WF: Handbook of Environmental Degradation Rates, Lewis Publishers, Chelsea, MI, 1991.
    137) Howard PH: Handbook of Environmental Fate and Exposure Data for Organic Chemicals, Volume III: Pesticides, Lewis Publishers, Chelsea, MI, 1991.
    138) Huang DJ & Chen HC: Effects of chlordane and lindane on testosterone and vitellogenin levels in green neon shrimp (Neocaridina denticulata)HUANG2004. Int J Toxicol 2004; 23(2):91-95.
    139) Huerta-Alardin AL, Varon J, & Marik PE: Bench-to-bedside review: Rhabdomyolysis -- an overview for clinicians. Crit Care 2005; 9(2):158-169.
    140) Hvidberg EF & Dam M: Clinical pharmacokinetics of anticonvulsants. Clin Pharmacokinet 1976; 1:161.
    141) IARC : Monographs on the Evaluation of Carcinogenic Risk of Chemicals to Humans. Lindane monograph. International Agency for Research on Cancer, World Health Organization. Geneva, Switzerland. 1998. Available from URL: http://193.51.164.11/htdocs/Monographs/ Vol05/Lindane.html.
    142) 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.
    143) 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.
    144) 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.
    145) 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.
    146) 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.
    147) IARC: Chemicals, industrial processes and industries associated with cancer in humans, IARC Monographs, Volumes 1 to 29, 4(Suppl), International Agency for Research on Cancer, World Health Organization, Geneva, Switzerland, 1982, pp 1-292.
    148) 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.
    149) IARC: Monographs on the Evaluation of Carcinogenic Risk of Chemicals to Humans, 20(Suppl. 7), International Agency for Research on Cancer, World Health Organization, Geneva, Switzerland, 1987, pp 220.
    150) ICAO: Technical Instructions for the Safe Transport of Dangerous Goods by Air, 2003-2004. International Civil Aviation Organization, Montreal, Quebec, Canada, 2002.
    151) ILC Dover, Inc.: Ready 1 The Chemturion Limited Use Chemical Protective Suit, ILC Dover, Inc., Frederica, DE, 1998.
    152) ITI: Toxic and Hazardous Industrial Chemicals Safety Manual, The International Technical Information Institute, Tokyo, Japan, 1988.
    153) ITI: Toxic and Hazardous Industrial Chemicals Safety Manual, The International Technical Information Institute, Tokyo, Japan, 1995.
    154) Ilina VI & Blekherman NA: Pediat Akush Ginekol 1974; 36:46-49.
    155) Infante PF, Epstein SS, & Newton WA Jr: Blood dyscrasias and childhood tumors and exposure to chlordane and heptachlor. Scand J Work Environ Health 1978; 4:137-150.
    156) 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.
    157) 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.
    158) Jaeger U, Podczeck A, & Haubenstock A: Acute oral poisoning with lindane-solvent mixtures. Vet Human Toxicol 1984; 26:11-14.
    159) Kappler, Inc.: Suit Smart. Kappler, Inc.. Guntersville, AL. 2001. Available from URL: http://www.kappler.com/suitsmart/smartsuit2/na_english.asp?select=1. As accessed 7/10/2001.
    160) Kashyap SK: J Environ Sci Health 1979; B14:305-319.
    161) Kassner JT, Maher TJ, & Hull KM: Cholestyramine as an adsorbent in acute lindane toxicity: a murine model. Ann Emerg Med 1993; 22:1392-1396.
    162) Khamidov MK: Med Zh Uzb 1984; 5:57-60.
    163) Khera KS, Whalen C, & Trivett G: Teratogenicity studies on pesticidal formulations of dimethoate, diuron and lindane in rats. Bull Environ Contam Toxicol 1979; 22:522-529.
    164) Kimberly-Clark, Inc.: Chemical Test Results. Kimberly-Clark, Inc.. Atlanta, GA. 2002. Available from URL: http://www.kc-safety.com/tech_cres.html. As accessed 10/4/2002.
    165) Kiraly J: Arch Environ Contam Toxicol 1979; 8:309-312.
    166) Klaasen CD: Nonmetallic environmental toxicants: air pollutants, solvents and vapors, and pesticides, in: Goodman A, Goodman LS, Rall TW et al (Eds). The Pharmacological Basis of Therapeutics, 7th ed, MacMillan Publishing Co, New York, NY, 1985, pp 1628-1650.
    167) Kollef MH & Schuster DP: The acute respiratory distress syndrome. N Engl J Med 1995; 332:27-37.
    168) Kolmodin-Hedman B, Alexanderson B, & Sjoqvist F: Effect of exposure to lindane on drug metabolism: decreased hexobarbital sleeping-times and increased antipyrine diasppearance rate in rats. Toxicol Appl Pharmacol 1971; 20:299-307.
    169) Konje JC, Otolorin EO, & Sotunmbi PT: Insecticide poisoning in pregnancy. J Reprod Med 1992; 37:992-994.
    170) Kulig K & Rumack B: Hydrocarbon ingestion. Curr Top Emerg Med 1981; 3:1-5.
    171) Kurt TL, Bost R, & Gilliland M: Accidental Kwell (lindane) ingestions. Vet Human Toxicol 1986; 28:552-569-571.
    172) LaCrosse-Rainfair: Safety Products, LaCrosse-Rainfair, Racine, WI, 1997.
    173) Lakkad BC: Mutat Res 1982; 101:315-320.
    174) Lee B & Groth P: Scabies: Transcutaneous poisoning during treatment. Pediatrics 1977; 59:643.
    175) Lee B, Groth P, & Turner W: Suspected reactions to gamma benzene hexachloride (Letter). JAMA 1976; 236:2846.
    176) Leibovich DL: Aktual Vopr Gig Naselennykh Mest, Mater Vses Konf Molodykh Uchenykh, 1973.
    177) Lewis RJ: Sax's Dangerous Properties of Industrial Materials, 10th ed, Van Nostrand Reinhold Company, New York, NY, 2000.
    178) Lewis RJ: Sax's Dangerous Properties of Industrial Materials, 9th ed, Van Nostrand Reinhold Company, New York, NY, 1996.
    179) Lifshitz M & Gavrilov V: Acute lindane poisoning in a child. Isr Med Assoc J 2002; 4(9):731-732.
    180) Loddenkemper T & Goodkin HP: Treatment of Pediatric Status Epilepticus. Curr Treat Options Neurol 2011; Epub:Epub.
    181) Loganovskii NG: Gig Tr Prof Zabol 1971; 15:46-48.
    182) Loge JP: Aplastic anemia following exposure to benzene hexachloride (lindane). JAMA 1965; 193:104-108.
    183) MAPA Professional: Chemical Resistance Guide. MAPA North America. Columbia, TN. 2003. Available from URL: http://www.mapaglove.com/pro/ChemicalSearch.asp. As accessed 4/21/2003.
    184) MAPA Professional: Chemical Resistance Guide. MAPA North America. Columbia, TN. 2004. Available from URL: http://www.mapaglove.com/ProductSearch.cfm?id=1. As accessed 6/10/2004.
    185) Malyshev LS: Med Zh Uzb 1972; 11:26-29.
    186) Mametkuliev C: Zdravookhr Turkm 1976; 20:28-31.
    187) Manno EM: New management strategies in the treatment of status epilepticus. Mayo Clin Proc 2003; 78(4):508-518.
    188) Mar-Mac Manufacturing, Inc: Product Literature, Protective Apparel, Mar-Mac Manufacturing, Inc., McBee, SC, 1995.
    189) Marcotrigiano GO & Storelli MM: Heavy metal, polychlorinated biphenyl and organochlorine pesticide residues in marine organisms: risk evaluation for consumers. Vet Res Commun 2003; 27 Suppl 1:183-195.
    190) Marigold Industrial: US Chemical Resistance Chart, on-line version. Marigold Industrial. Norcross, GA. 2003. Available from URL: www.marigoldindustrial.com/charts/uschart/uschart.html. As accessed 4/14/2003.
    191) McNutt TL & Harris C: Lindane embryotoxicity and differential alteration of cysteine and glutathione levels in rat embryos and visceral yolk sacs. Reprod Toxicol 1994; 8:351-362.
    192) McParland PJ & McCracker RM: Vet Rec 1973; 93:369-371.
    193) Meister R: Farm Chemicals Handbook, 82, Meister Publishing Co, Willoughby, OH, 1996.
    194) Memphis Glove Company: Permeation Guide. Memphis Glove Company. Memphis, TN. 2001. Available from URL: http://www.memphisglove.com/permeation.html. As accessed 7/2/2001.
    195) Mgbenka BO, Oluah NS, & Umeike I: Effect of Gammalin 20 (Lindane) on Differential White Blood Cell Counts of the African Catfish, Clarias albopunctatus. Bull Environ Contam Toxicol 2003; 71:248-254.
    196) Milby TH & Samuels AJ: Human exposure to lindane. J Occup Med 1971; 13:256-258.
    197) Milby TH: J Occup Med 1968; 10:584-587.
    198) Miller RD, Sohn YJ, & Matteo RS: Enhancement of d-tubocurarine neuromuscular blockade by diuretics in man. Anesthesiology 1976; 45:442-445.
    199) Montgomery Safety Products: Montgomery Safety Products Chemical Resistant Glove Guide, Montgomery Safety Products, Canton, OH, 1995.
    200) Morgan DP, Dotson TB, & Lin LI: Effectiveness of activated charcoal, mineral oil, and castor oil in limiting gastrointestinal absorption of a chlorinated hydrocarbon pesticide. Clin Toxicol 1977; 11:61-70.
    201) Morgan DP: Anemia associated with exposure to lindane. Arch Environ Health 1980; 35:307-310.
    202) Morgan DP: Pesticide Toxicology, in: Anthony TU (Ed), Survey of Contemporary Toxicology, Vol 2, John Wiley & Sons, New York, NY, 1982, pp 1-36.
    203) Morgan DP: Recognition and Management of Pesticide Poisonings, 4th ed, US Environmental Protection Agency, EPA-540/9-88-0015, Government Printing Office, Washington, DC, 1993, pp 17-24.
    204) Munk ZM & Nantel A: Acute lindane poisoning with development of muscle necrosis. Canad Med Assoc J 1977; 117:1050-1054.
    205) NFPA: Fire Protection Guide to Hazardous Materials, 13th ed., National Fire Protection Association, Quincy, MA, 2002.
    206) 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.
    207) NIOSH : Pocket Guide to Chemical Hazards. National Institute for Occupational Safety and Health. Cincinnati, OH (Internet Version). Edition expires 2000; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    208) NIOSH: Pocket Guide to Chemical Hazards. National Institute for Occupational Safety and Health. Cincinnati, OH (Internet Version). Edition expires 2005; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    209) 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.
    210) 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.
    211) 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.
    212) 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.
    213) Nag D, Singh GC, & Senon S: Epilepsy epidemic due to benzahexachlorine. Trop Geogr Med 1977; 29:229-232.
    214) Nagy Z: ACTA Microbiol Acad Sci Hung 1975; 22:209-214.
    215) Nantel AJ, Ayotte L, & Benedetti JL: A group of adults acutely poisoned by food contaminated with lindane. ACTA Pharmacol Toxicol 1977; 41:250.
    216) Naradzay J & Barish RA: Approach to ophthalmologic emergencies. Med Clin North Am 2006; 90(2):305-328.
    217) Narayanan B, Suidan MT, & Gelderloos AB: Treatment of semivolatile compounds in high strength wastes using an anaerobic expanded-bed GAC reactor. Water Res 1993; 27:171-180.
    218) Nat-Wear: Protective Clothing, Hazards Chart. Nat-Wear. Miora, NY. 2001. Available from URL: http://www.natwear.com/hazchart1.htm. As accessed 7/12/2001.
    219) 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.
    220) 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.
    221) 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.
    222) 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.
    223) 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.
    224) 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.
    225) 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.
    226) 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.
    227) 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.
    228) 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.
    229) 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.
    230) 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.
    231) 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.
    232) 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.
    233) 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.
    234) 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.
    235) 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.
    236) 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.
    237) 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.
    238) 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.
    239) 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.
    240) 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.
    241) 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.
    242) 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.
    243) 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.
    244) 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.
    245) 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.
    246) 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.
    247) 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.
    248) 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.
    249) 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.
    250) 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.
    251) 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.
    252) 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.
    253) 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.
    254) 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.
    255) 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.
    256) 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.
    257) 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.
    258) 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.
    259) 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.
    260) 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.
    261) 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.
    262) 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.
    263) 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.
    264) 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.
    265) 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.
    266) 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.
    267) 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.
    268) 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.
    269) 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.
    270) 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.
    271) 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.
    272) 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.
    273) 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.
    274) 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.
    275) 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.
    276) 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.
    277) 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.
    278) 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.
    279) 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.
    280) 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.
    281) 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.
    282) 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.
    283) 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.
    284) 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.
    285) 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.
    286) 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.
    287) 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.
    288) 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.
    289) 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.
    290) 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.
    291) 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.
    292) 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.
    293) 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.
    294) 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.
    295) National Research Council : Acute exposure guideline levels for selected airborne chemicals, 5, National Academies Press, Washington, DC, 2007.
    296) National Research Council: Acute exposure guideline levels for selected airborne chemicals, 6, National Academies Press, Washington, DC, 2008.
    297) National Research Council: Acute exposure guideline levels for selected airborne chemicals, 7, National Academies Press, Washington, DC, 2009.
    298) National Research Council: Acute exposure guideline levels for selected airborne chemicals, 8, National Academies Press, Washington, DC, 2010.
    299) Neese Industries, Inc.: Fabric Properties Rating Chart. Neese Industries, Inc.. Gonzales, LA. 2003. Available from URL: http://www.neeseind.com/new/TechGroup.asp?Group=Fabric+Properties&Family=Technical. As accessed 4/15/2003.
    300) Nicholls RW: A case of acute poisoning by benzene hexachloride. Med J Aust 1958; 1:42-43.
    301) Nigam SK: Bull Environ Contam Toxicol 1979; 23:431-437.
    302) Nitsche K, Lange M, & Bauer E: Quantitative distribution of locally applied lindane in human skin and subcutaneous fat in vitro. Dependence of penetration on the applied concentration, skin state, duration of action and nature and time of washing. Derm Beruf Umwelt 1984; 32:161-165.
    303) Nolan K, Kamrath J, & Levitt J: Lindane toxicity: a comprehensive review of the medical literature. [Review]. Pediatr Dermatol 2012; 29(2):141-146.
    304) Nordt SP & Chew G: Acute lindane poisoning in three children . J Emerg Med 2000; 18(1):51-53.
    305) North: Chemical Resistance Comparison Chart - Protective Footwear . North Safety. Cranston, RI. 2002. Available from URL: http://www.linkpath.com/index2gisufrm.php?t=N-USA1. As accessed April 30, 2004.
    306) North: eZ Guide Interactive Software. North Safety. Cranston, RI. 2002a. Available from URL: http://www.northsafety.com/feature1.htm. As accessed 8/31/2002.
    307) Nyitrai G, Kekesi KA, Szilagyi N, et al: Neurotoxicity of lindane and picrotoxin: Neurochemical and electrophysiological correlates in the rat hippocampus in vivo. Neurochem Res 2002; 27:139-145.
    308) OHM/TADS : Oil and Hazardous Materials/Technical Assistance Data System. US Environmental Protection Agency. Washington, DC (Internet Version). Edition expires 2000; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    309) OHM/TADS: Oil and Hazardous Materials Technical Assistance Data System. US Environmental Protection Agency. Washington, D.C. (Internet Version). Edition expires 2004; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    310) OHM/TADS: Oil and Hazardous Materials Technical Assistance Data System. US Environmental Protection Agency. Washington, D.C. (Internet Version). Edition expires 2005; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    311) OSHA: Personal Protective Equipment for General Industry. 59 FR 16334-16364, 59, Department of Labor, Occupational Safety and Health Administration, Washington, DC, 2000, pp 16334-16364.
    312) Ortega P, Hayes WJ, & Durham WF: Pathologic changes in the liver of rats after feeding low levels of various insecticides. AMA Arch Pathol 1957; 64:614-622.
    313) Palmer AK: Toxicology 1978; 10:54-54.
    314) Parent-Massin D, Thouvenot D, & Rio B: Lindane haematotoxicity confirmed by in vitro tests on human and rat progenitors. Human Exp Toxicol 1994; 13:103-106.
    315) Peate WF: Work-related eye injuries and illnesses. Am Fam Physician 2007; 75(7):1017-1022.
    316) Peper M, Ertl M, & Gerhard I: Long-term exposure to wood preserving chemicals containing pentachlorophenol and lindane is related to neurobehavioral performance in women. Am J Ind Med 1998; 35:632-641.
    317) Pernov K & Kyurkchiyev S: Gig Tr Prof Zabol 1974; 12:46-47.
    318) Peters HH, Gocmen A, & Cripps DJ: Epidemiology of hexachlorobenzene-induced porphyria in Turkey -- clinical and laboratory follow-up after 25 years. Arch Neurol 1982; 39:744-749.
    319) Petrescu ST: Rev Med Chir Soc Med Mat Iasi 1974; 78:831-842.
    320) Playtex: Fits Tough Jobs Like a Glove, Playtex, Westport, CT, 1995.
    321) Pohanish RP & Greene SA: Rapid Guide to Chemical Incompatibilities, Van Nostrand Reinhold Company, New York, NY, 1997.
    322) Poissant L & Koprivnjak J-F: Fate and atmospheric concentrations of alpha- and gamma-hexachlorocyclohexane in Quebec, Canada. Environ Sci Technol 1996; 30:845-851.
    323) Polderman KH: Acute renal failure and rhabdomyolysis. Int J Artif Organs 2004; 27(12):1030-1033.
    324) Pomes A, Frandsen A, & Sunol C: Lindane cytotoxicity in cultured neurocortical neurons is ameliorated by GABA and flunitrazepam. J Neurosci Res 1994; 39:663-668.
    325) Pool-Zobel BL, Guigas C, & Klein R: Assessment of genotoxic effects by lindane. Food Chem Toxicol 1993; 31:271-283.
    326) Poradovsky K: Cesk Gynekol 1977; 42:405-410.
    327) Post CF & Juhlin E: Demodex folliculorum and blepharitis. Arch Dermatol 1963; 88:292-302.
    328) Pramanik AK & Hansen RC: Transcutaneous gamma benzene hexachloride absorption and toxicity in infants and children. Arch Dermatol 1979; 115:1224-1225.
    329) Prasad AK, Pant N, & Srivastava SC: Effect of dermal application of hexachlorocyclohexane (HCH) on male reproductive system of rat. Human Exp Toxicol 1995; 14:484-488.
    330) Proctor NH, Hughes JP, & Fischman ML: Chemical Hazards of the Workplace, 2nd ed, JB Lippincott Co, Philadelphia, PA, 1988.
    331) Product Information: Kwell(R), lindane. Reed & Carnick, Jersey City, NJ, 1994.
    332) Product Information: MANNITOL intravenous injection, mannitol intravenous injection. Hospira, Inc. (per DailyMed), Lake Forest, IL, 2009.
    333) Product Information: Mannitol intravenous injection, mannitol intravenous injection. American Regent, Inc. (per DailyMed), Shirley, NY, 2009.
    334) Product Information: diazepam IM, IV injection, diazepam IM, IV injection. Hospira, Inc (per Manufacturer), Lake Forest, IL, 2008.
    335) Product Information: lindane 1% topical lotion, lindane 1% topical lotion. VersaPharm Incorporated (per DailyMed), Marietta, GA, 2011.
    336) Product Information: lindane 1% topical shampoo, lindane 1% topical shampoo. VersaPharm Incorporated (per DailyMed), Marietta, GA, 2011.
    337) Product Information: lorazepam IM, IV injection, lorazepam IM, IV injection. Akorn, Inc, Lake Forest, IL, 2008.
    338) Product Information: mannitol IV injection, urologic irrigation, mannitol IV injection, urologic irrigation. Abraxis Pharmaceutical Products, Schaumburg, IL, 2006.
    339) RTECS : Registry of Toxic Effects of Chemical Substances. National Institute for Occupational Safety and Health. Cincinnati, OH (Internet Version). Edition expires 2000; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    340) Ramabhatta S, Sunilkumar GR, & Somashekhar C: Lindane toxicity following accidental oral ingestion. Indian J Dermatol Venereol Leprol 2014; 80(2):181-182.
    341) Rao CV, Shreenivas R, & Singh V: Disseminated intravascular coagulation in a case of fatal lindane poisoning. Vet Human Toxicol 1988; 30:132-134.
    342) Rappl A & Waiblinger W: Zur Kontamination von Muttermilch mit Ruckstanden chlorierter Kohlenwasserstoffe (German). Dtsch Med Wochenschr 1975; 100:228-235-238.
    343) Rauch AE, Kowalsky SF, & Lesar TS: Lindane (KWELL)-induced aplastic anemia. Arch Intern Med 1990; 150:2393-2395.
    344) Reuber MD: Carcinogenicity of heptachlor and heptachlor epoxide. J Environ Pathol Toxicol Oncol 1987; 7:85-114.
    345) Reuber MD: Environ Res 1979; 19:460-481.
    346) River City: Protective Wear Product Literature, River City, Memphis, TN, 1995.
    347) Rocchi P: Arch Toxicol 1980; 45:101-108.
    348) Roszkowski J: Immuno-morphological investigations on the effect of lindane, chlorfenvinphos, and carbaryl on immune reactions. I. Experiments on rabbits. Bull Vet Inst Pulawy 1978; 22:25-30.
    349) Runhaar EA, Sangster B, & Greve PA: A case of fatal Endrin poisoning. Human Toxicol 1985; 4:241-247.
    350) Rupa DS, Reddy PP, & Reddy OS: Genotoxic effect of benzene hexachloride in cultured human lymphocytes. Human Genet 1989; 83:271-273.
    351) Ryan ML, Arnold TC, & Dick RM: Lindane: A case of dangerous playtime (Abstract). J Toxicol - Clin Toxicol 1998; 36:467.
    352) Safety 4: North Safety Products: Chemical Protection Guide. North Safety. Cranston, RI. 2002. Available from URL: http://www.safety4.com/guide/set_guide.htm. As accessed 8/14/2002.
    353) Saxena MC: Arch Toxicol 1984; 48:127-134.
    354) Saxena MC: J Anal Toxicol 1981; 5:6-9.
    355) Saxena MC: Toxicology 1980; 17:323.
    356) Scascitelli M & Pacchierotti F: Effects of lindane on oocyte maturation and preimplantation embryonic development in the mouse. Reprod Toxicol 2003; 17:299-303.
    357) Schardein JL: Chemically Induced Birth Defects, 2nd ed, Marcel Dekker, Inc, New York, NY, 1993, pp 667-668-697.
    358) 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.
    359) Servus: Norcross Safety Products, Servus Rubber, Servus, Rock Island, IL, 1995.
    360) Seth V, Ahmad RS, Suke SG, et al: Lindane-induced immunological alterations in human poisoning cases. Clin Biochem 2005; 38(7):678-680.
    361) Shaw JM: Successful outcome of combined organochlorine and organomercury poisoning in a dog. Vet Rec 1988; 122:516.
    362) Shivanandappa T & Krishnakumari MK: ACTA Pharmacol Toxicol 1983; 52:12-17.
    363) Sidi Y, Kiltchevsky E, & Shaklai M: Acute myeloblastic leukemia and insecticide. State J Med 1983; 83:161.
    364) Simpson WM & Schuman SH: Recognition and management of acute pesticide poisoning. Am Fam Physician 2002; 65(8):1599-1604.
    365) Sinha RR & Sinha SP: Comp Physiol Ecol 1983; 8:87-89.
    366) Sittig M: Handbook of Toxic and Hazardous Chemicals and Carcinogens, 2nd ed, Noyes Publications, Park Ridge, NJ, 1985.
    367) Sittig M: Handbook of Toxic and Hazardous Chemicals and Carcinogens, 3rd ed, Noyes Publications, Park Ridge, NJ, 1991.
    368) Solomon BK, Haut SR, & Carr EM: Neurotoxic reaction to lindane in an HIV-seropositive patient. J Fam Pract 1995; 40:291-296.
    369) 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.
    370) Standard Safety Equipment: Product Literature, Standard Safety Equipment, McHenry, IL, 1995.
    371) Starr HG & Clifford NJ: Acute lindane intoxication. Arch Environ Health 1972; 25:374-375.
    372) Starr HG Jr & Clifford NJ: Absorption of pesticides in a chronic skin disease. Arch Environ Health 1971; 22:396-400.
    373) 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.
    374) Stormont RT & Conley BE: Abuse of insecticide fumigating devices. JAMA 1955; 156:607.
    375) Sudakin DL: Fatality after a single dermal application of lindane lotion. Arch Environ Occup Health 2007; 62(4):201-203.
    376) Sundqvist K, Wingfors H, Brorstom-Lunden E, et al: Air-Sea Gas Exchange of HCHs and PCBs and Enantiomers of alpha-HCH in the Kattegat Sea Region. Env Poll 2004; 128:73-83.
    377) Taylor EJ, Blockwell SJ, & Maund SJ: Effects of lindane on the life-cycle of a freshwater macroinvertebrate chironomus riparius meigen (insecta, diptera). Arch Environ Contam Toxicol 1993; 24:145-150.
    378) Telch J & Jarvis DA: Acute intoxication with lindane (gamma benzene hexachloride). Canad Med Assoc J 1982; 126:662-663.
    379) Tenenbein M: Seizures after lindane therapy. J Am Ger Soc 1991; 39:394-395.
    380) Tidou AS, Moreteau JC, & Ramade F: Effects of lindane and deltamethrin on zooplankton communities of experimental ponds. Hydrobiologia 1992; 232:157-168.
    381) Tiemann U, Schneider F, & Tuchscherer A: Effects of organochlorine pesticides on DNA synthesis of cultured oviductal and uterine cells and on estrogen receptor of uterine tissue from heifers. Arch Toxicol 1996; 70:490-496.
    382) Tilson HA, Hong JS, & Mactutus CF: Effects of 5,5-diphenylhydantoin (phenytoin) on neurobehavioral toxicity of organochlorine insecticides and permethrin. J Pharmacol Exp Ther 1985; 233:285-289.
    383) Tingley: Chemical Degradation for Footwear and Clothing. Tingley. South Plainfield, NJ. 2002. Available from URL: http://www.tingleyrubber.com/tingley/Guide_ChemDeg.pdf. As accessed 10/16/2002.
    384) Tolot F: Lindane-induced polyneuritis. J Med Lyon 1969; 50:747-754.
    385) Tomczak S, Baumann K, & Lehnert G: Occupational exposure to hexachlorocyclohexane. IV. Sex hormone alterations in HCH exposed workers. Internat Arch Occup Environ Health 1981; 48:281-287.
    386) Trelleborg-Viking, Inc.: Chemical and Biological Tests (database). Trelleborg-Viking, Inc.. Portsmouth, NH. 2002. Available from URL: http://www.trelleborg.com/protective/. As accessed 10/18/2002.
    387) Trelleborg-Viking, Inc.: Trellchem Chemical Protective Suits, Interactive manual & Chemical Database. Trelleborg-Viking, Inc.. Portsmouth, NH. 2001.
    388) Tsushimoto G: Arch Environ Contam Toxicol 1983; 12:721-729.
    389) Tusell JM, Sunol C, & Gelpi E: Relationship between lindane concentration in blood and brain and convulsant response in rats after oral or intraperitoneal administration. Arch Toxicol 1987; 60:432-437.
    390) 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.
    391) 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.
    392) 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-.
    393) 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.
    394) 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.
    395) 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.
    396) 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.
    397) 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-.
    398) U.S. Occupational Safety, and Health Administration (OSHA): Process safety management of highly hazardous chemicals. 29 CFR 2010 2010; 29(1910.119):348-.
    399) 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.
    400) Vanholder R, Sever MS, Erek E, et al: Rhabdomyolysis. J Am Soc Nephrol 2000; 11(8):1553-1561.
    401) Verderber L, Lavin P, & Wesley R: Pseudotumour cerebri and chronic benzene hexachloride (lindane) exposure. J Neurol Neurosurg Psychiatr 1991; 54:1123.
    402) Verschueren K: Handbook of Environmental Data on Organic Chemicals. 4th ed. CD-ROM version. Wiley-Interscience. Hoboken, NJ. 2001.
    403) Verzhanskii PS: Gumoraln Regul Rodovoi Deyat Lech Ee Narushenii, 1976.
    404) Vidal A: Developments in solar photocatalysis for water purification. Chemosphere 1998; 36:2593-2606.
    405) Waite DT, Gurprasad JF, Sproull DV, et al: Atmospheric Movement of Lindane (gamma-Hexachlorocyclohexane) from Canola Fields Planted with Treated Seed. J Environ Qual 2001; 30:768-775.
    406) Walter LA & Catenacci MH: Rhabdomyolysis. Hosp Physician 2008; 44(1):25-31.
    407) Wasserman M: Environ Res 1982; 28:106-112,2.
    408) Weisse I & Herbst M: Toxicology 1977; 7:233-238.
    409) Wells Lamont Industrial: Chemical Resistant Glove Application Chart. Wells Lamont Industrial. Morton Grove, IL. 2002. Available from URL: http://www.wellslamontindustry.com. As accessed 10/31/2002.
    410) Wells WL & Milhorn HT Jr: Suicide attempt by toxaphene ingestion: a case report. J Mississippi State Med Assoc 1983; 24:329-330.
    411) West I: Lindane and hematologic reactions. Arch Environ Health 1967; 15:97-101.
    412) Wheeler M: Gamma benzene hexachloride (Kwell) poisoning in a child. West J Med 1977; 6:518-521.
    413) Wiberg K, Brorstrom-Lunden E, Wnagberg I, et al: Concentrations and Fluxes of Hexachlorocyclohexanes and Chiral Composition of alpha-HCH in Environmental Samples from the Southern Baltic Sea. Environ Sci Technol 2001; 35:4739-4746.
    414) Wiles DA, Russell JL, Olson KR, et al: Massive lindane overdose with toxicokinetics analysis. J Med Toxicol 2015; 11(1):106-109.
    415) Willson DF, Truwit JD, Conaway MR, et al: The Adult Calfactant in Acute Respiratory Distress Syndrome (CARDS) Trial. Chest 2015; Epub:Epub.
    416) 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.
    417) Wilson JS: Lindane poisoning in a family. Med J Aust 1959; 2:684.
    418) Wolfe & Esher: Bull Environ Contam Toxicol 1980; 24:894.
    419) Woodliff HJ, Connor PM, & Scopa J: Aplastic anemia association with insecticides. Med J Aust 1966; 1:628-629.
    420) Workrite: Chemical Splash Protection Garments, Technical Data and Application Guide, W.L. Gore Material Chemical Resistance Guide, Workrite, Oxnard, CA, 1997.
    421) Yoshimura M: Kinki Daigaku Igaku Zasshi 1979; 4:209-218.
    422) Zilker T, Haberkorn M, & Angerer J: Severe poisoning by Quellada H, A gamma-HCH-containing shampoo in a human - Monitoring of lindane and its metabolites in serum and urine (abstract). J Toxicol-Clin Toxicol 1999; 37:364.
    423) Zimmerman JL & Shen MC: Rhabdomyolysis. Chest 2013; 144(3):1058-1065.