MOBILE VIEW  | 

RIVASTIGMINE AND RELATED AGENTS

Classification   |    Detailed evidence-based information

Therapeutic Toxic Class

    A) Rivastigmine, an acetylcholinesterase inhibitor, is used for the treatment of dementia of the Alzheimer's type and mild to moderate dementia associated with Parkinson's disease. Tacrine was withdrawn from the market in 2013.

Specific Substances

    A) ECHOTHIOPHATE IODIDE
    1) (2-Diethoxyphosphinylthioethyl)trimethylammonium iodide
    2) Ecostigmine iodide
    3) Ecothiopate iodide
    4) MI-217
    5) Phospholine iodide
    6) Molecular formula: C9-H23-I-NO3-PS
    7) CAS 6736-03-4 (echothiophate)
    8) CAS 513-10-0 (echothiophate iodide)
    RIVASTIGMINE
    1) (-)-m[(S)-1(Dimethylamino)ethyl]phenyl ethylmethylcarbamate
    2) ENA-713
    3) SDZ-212-713
    4) SDZ-ENA-713
    5) CAS 123441-03-2 (rivastigmine)
    6) CAS 129101-54-8 (rivastigmine hydrogen tartrate)
    TACRINE
    1) 9-Acridinamine,1,-2,3,4-tetrahydro-,monohydrochloride
    2) 9-Amino-1,2,3,4-tetrahydroacridine monohydrochloride
    3) Tacrinum
    4) Tetrahydroaminoacridine
    5) Tetrahydroaminoacrine
    6) THA
    7) CAS 1684-40-8 (Tacrine Hydrochloride)
    8) CAS 321-64-2 (Tacrine)
    9) Molecular Formula: C13-H14-N2.HCl
    VELNACRINE
    1) HP-029
    2) CAS 104675-29-8 (velnacrine)
    3) CAS 118909-22-1 (velnacrine maleate)

Available Forms Sources

    A) FORMS
    1) Rivastigmine is available in the United States as 1.5 mg, 3 mg, 4.5 mg, or 6 mg capsules and as a 2 mg/mL oral solution (packaged as 120 mL) (Prod Info EXELON(R) oral capsules, oral solution, 2015) and available as a transdermal patches of 4.6 mg/24 hours, 9.5 mg/24 hours and 13.3 mg/24 hours (Prod Info EXELON(R) PATCH transdermal system, 2015).
    2) ECHOTHIOPHATE IODIDE: Used as a miotic, cholinesterase inhibitor eye drop. It is available as 1.5 mg (0.03%) ophthalmic powder for solution, and as 0.06%, 0.125%, and 0.25% ophthalmic solution (Prod Info Phospholine iodide(R), echothiophate, 1996).
    B) USES
    1) RIVASTIGMINE
    a) Rivastigmine, an acetylcholinesterase inhibitor, is used for the treatment of mild to moderate dementia of the Alzheimer's type and mild to moderate dementia associated with Parkinson's disease (Prod Info EXELON(R) oral capsules, oral solution, 2015). The transdermal patch is also used to treat mild, moderate and severe dementia of the Alzheimer's type (Prod Info EXELON(R) PATCH transdermal system, 2015).
    2) OTHER AGENTS/NO LONGER AVAILABLE
    a) TACRINE: Because of continuing concerns of safety, tacrine was withdrawn from the market in 2013.
    b) METRIFONATE: It had been investigated as a treatment of Alzheimer's Disease. However, clinical studies produced only modest benefits and research was stopped after reports of muscle weakness, occasionally requiring respiratory support.
    c) VELNACRINE: Worldwide research and development of velnacrine for Alzheimer's disease was halted by Hoechst-Roussel in 1994 (Anon, 1994).

Life Support

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

Clinical Effects

    0.2.1) SUMMARY OF EXPOSURE
    A) USES: Rivastigmine, an acetylcholinesterase inhibitor, is used for the treatment of dementia of the Alzheimer's type and mild to moderate dementia associated with Parkinson's disease.
    B) WITHDRAWN FROM MARKET: Because of ongoing concerns over the safety of TACRINE it was withdrawn from the market in 2013. Other agents, such as metrifonate and velnacrine are also no longer used in the treatment of Alzheimer's disease.
    C) PHARMACOLOGY: The precise mechanism of action of rivastigmine is unknown. However, it is thought to exert is effect by enhancing cholinergic function, by increasing the concentration of acetylcholine through reversible inhibition of its hydrolysis by cholinesterase.
    D) EPIDEMIOLOGY: Exposure has occurred. Severe clinical events or fatalities are uncommon.
    E) WITH THERAPEUTIC USE
    1) The most common adverse effects reported with rivastigmine therapy include: nausea, vomiting, anorexia, dyspepsia, and asthenia. Other clinical events include dizziness, headache, urinary tract infection, fatigue, insomnia, and confusion. Infrequent events may include depression, anxiety, hallucination, and aggressive reactions. Fluid depletion may occur through bronchorrhea, vomiting and diarrhea, polyuria, and sweating.
    2) CHOLINERGIC ACTIVITY: Other events that can develop due to increases in cholinergic activity include extrapyramidal symptoms, worsening of parkinsonian symptoms (ie, tremor), seizures, increases in gastric acid secretion, at risk to develop ulcers or gastrointestinal bleeding, bradycardia, and possible urinary obstruction. Drugs like rivastigmine the increase cholinergic activity should also be given with caution in patients with an underlying history of asthma or obstructive pulmonary disease.
    3) POSTMARKETING EXPERIENCE: Based on postmarketing experience with rivastigmine, hepatitis, aggression, Stevens-Johnson syndrome and disseminated allergic dermatitis have been reported.
    F) WITH POISONING/EXPOSURE
    1) Overdose information is limited. Cholinergic toxicity has been reported after overdose of rivastigmine, typical findings can include severe nausea, vomiting, salivation, sweating, bradycardia, hypotension, respiratory depression, collapse and convulsions. In severe cases, muscle weakness can develop and may result in death if respiratory muscles become involved. Other symptoms that have been associated with overdose of rivastigmine include: diarrhea, abdominal pain, dizziness, tremor, headache, somnolence, confusional state, hyperhidrosis, hypertension, hallucinations and malaise. Depression of blood cholinesterase may occur following overdoses.
    0.2.3) VITAL SIGNS
    A) HYPOTENSION: Blood pressure may be decreased following an overdose.
    B) BRADYCARDIA: Pulse rate may be decreased following an overdose.
    0.2.4) HEENT
    A) Miosis, salivation, and lacrimation are common systemic effects of cholinergic agonists and may occur with tacrine in therapeutic or overdose amounts.
    0.2.20) REPRODUCTIVE
    A) Rivastigmine is classified as US FDA Pregnancy Category B. Although animals studies involving rats and rabbits revealed no evidence of teratogenicity, reduced fetal/pup weights were observed in rats at maternally toxic doses of rivastigmine. It is unknown whether rivastigmine is excreted in human breast milk; however, the drug is excreted in rat milk. Echothiophate is classified as US FDA Pregnancy Category C.

Laboratory Monitoring

    A) Monitor vital signs and mental status.
    B) Monitor fluid status and electrolytes in patients that develop significant vomiting, diarrhea and sweating.
    C) Monitor red blood cell and plasma cholinesterase levels in patients with signs and symptoms of cholinergic poisoning.
    D) Signs and symptoms of cholinesterase inhibitor poisoning should be monitored. Cholinesterase activity may be depressed and should be monitored in plasma and whole blood.
    E) Monitor ECG following significant exposures. Overdoses have been associated with bradycardia.

Treatment Overview

    0.4.2) ORAL/PARENTERAL EXPOSURE
    A) MANAGEMENT OF MILD TO MODERATE TOXICITY
    1) Treatment is symptomatic and supportive. Monitor vital signs and mental status. Monitor fluid status and electrolyte balance if the patient develops significant fluid loss (ie, vomiting, diarrhea, diaphoresis). Treat nausea and vomiting with antiemetics and fluid replacement. Initially treat hypotension with IV fluids. Infuse 10 to 20 mL/kg of isotonic fluid. If hypotension persists, administer dopamine or norepinephrine. Atropine is the drug of choice to treat cholinergic effects such as bradycardia, bronchorrhea and bronchospasm.
    B) MANAGEMENT OF SEVERE TOXICITY
    1) Treatment is symptomatic and supportive. Treat seizure activity with IV benzodiazepines (diazepam or lorazepam). Consider phenobarbital or propofol if seizures recur. ANTIDOTES: Administer atropine for muscarinic manifestations (e.g. salivation, diarrhea, bronchorrhea); and pralidoxime for nicotinic manifestations (e.g. weakness, fasciculations) for severe toxicity. Assess respiratory function, airway management including intubation and mechanical ventilation as needed. Respiratory depression including muscle weakness can occur. Increasing muscle weakness is a possibility and may result in death if respiratory muscles are involved. Acute lung injury may develop with a significant exposure. Maintain adequate ventilation and oxygenation; serial ABGs and/or pulse oximetry as necessary; PEEP may be necessary.
    C) DECONTAMINATION
    1) PREHOSPITAL: Spontaneous vomiting can occur after a significant exposure. Excessive vomiting may increase the risk of esophageal rupture and induced emesis is probably not indicated.
    2) HOSPITAL: GI decontamination is unlikely to be necessary as spontaneous vomiting it likely to occur. Activated charcoal may be considered if coingestants are suspected, the ingestion is recent, and the airway can be protected.
    D) AIRWAY MANAGEMENT
    1) A significant exposure can result in cholinergic crisis include respiratory depression and severe respiratory muscle weakness. Monitor respiratory effort and ventilation. Administer oxygen. Intubation and mechanical ventilation may be necessary.
    E) ANTIDOTES
    1) Atropine is used to treat muscarinic effects (e.g. salivation, lacrimation, defecation, urination, bronchorrhea). Oximes are used to reverse neuromuscular blockade. Use of oximes is usually indicated for patients with moderate to severe toxicity.
    a) ATROPINE
    1) Atropine sulfate is the drug of choice to treat cholinergic effects such as bradycardia, bronchorrhea and bronchospasm. For significant muscarinic symptoms administer:
    a) ADULT: Initial dose of 1 to 2 mg IV repeat with subsequent doses based on clinical response.
    b) CHILD: Initial dose 0.05 mg/kg up to 4 mg (usual dose 1 mg), intramuscularly or intravenously every 10 to 30 minutes until muscarinic signs and symptoms subside, and repeat if they reappear.
    2) ALTERNATIVE THERAPY: Glycopyrrolate and methscopolamine bromide have been suggested as alternatives to atropine in treating the peripheral cholinergic symptoms.
    b) PRALIDOXIME
    1) Treat moderate to severe poisoning (fasciculations, muscle weakness, respiratory depression, coma, seizures) with pralidoxime in addition to atropine.
    F) SEIZURES
    1) IV benzodiazepines are indicated for seizures or agitation, diazepam 5 to 10 mg IV, lorazepam 2 to 4 mg IV; repeat as needed.
    G) BRONCHOSPASM
    1) ALBUTEROL: 0.25 to 0.5 mL in 2 to 4.5 mL of normal saline delivered every 4 to 6 hours per nebulizer. CHILD: 100 to 200 mcg 3 to 6 times a day.
    2) Epinephrine may assist in overcoming severe cardiovascular or bronchoconstrictor responses (DOSE: 0.1 to 1.0 mg SubQ).
    H) BRADYCARDIA
    1) ATROPINE: ADULT: Give 0.5 mg to 1 mg IV or intratracheally every 5 minutes up to a maximum total dose of 0.04 mg/kg. Doses less than 0.5 mg may cause paradoxical bradycardia in adults. PEDIATRIC: Give 0.02 mg/kg IV, intratracheally or intraosseously, repeating every 5 minutes up to a maximum of 1 mg.
    I) ENHANCED ELIMINATION
    1) Enhanced elimination procedures (ie, hemodialysis, peritoneal dialysis, or hemofiltration) are not anticipated to be clinically indicated due to the short-half life (1.5 hours) of rivastigmine. It also has a large volume of distribution in the range of 1.8 to 2.7 L/kg.
    J) PHARMACOKINETICS
    1) RIVASTIGMINE: Peak plasma concentrations are reached in approximately 1 hour. It is rapidly and completely absorbed with an absolute bioavailability of approximately 36% following ingestion of a 3-mg dose. Vd: 1.8 to 2.7 L/kg. It is rapidly and extensively metabolized in the liver, predominantly via cholinesterase-mediated hydrolysis. Rivastigmine is primarily eliminated by the kidneys (97%); and 0.4% is excreted in the feces. The elimination half-life of rivastigmine is approximately 1.5 hours.
    K) PATIENT DISPOSITION
    1) HOME CRITERIA: All children with ingestions should be sent to a healthcare facility for evaluation and treatment. Adults with a deliberate overdose should be sent to a healthcare facility for evaluation and treatment. Asymptomatic adults with an inadvertent ingestion of one or two pills can be monitored at home.
    2) OBSERVATION CRITERIA: Patients should be observed until all symptoms resolve. Patients that develop significant toxicity including respiratory depression should be admitted for a higher level of care.
    3) ADMISSION CRITERIA: Patients who develop cholinergic crisis including respiratory depression should be admitted. They should only be discharged when free of symptoms.
    4) CONSULT CRITERIA: Consult a medical toxicologist or a poison center for assistance with medical management in patients with severe overdose or in whom the diagnosis is unclear. Consult a pulmonologist or hospital intensivist if the patient develops evidence of respiratory compromise.

Range Of Toxicity

    A) TOXIC DOSE: A toxic dose has not been established. RIVASTIGMINE: An adult developed vomiting, bradycardia, hypertension, salivation, diaphoresis and drowsiness after ingesting 90 mg. TACRINE: An estimated human lethal dose is reported as 30 mg/kg when unopposed by anticholinergics. METRIFONATE: Doses as high as 72 mg/kg have produced severe abdominal colic, vomiting, and muscle weakness.
    B) THERAPEUTIC DOSE: Dosing for Alzheimer's Disease is as follows: RIVASTIGMINE: 1.5 mg orally twice daily; if tolerated, increase dose every 2 weeks by 1.5 mg twice daily to a maximum dose of 6 mg twice daily. Other agents (ie, tacrine and metrifonate) have been withdrawn from the market.

Summary Of Exposure

    A) USES: Rivastigmine, an acetylcholinesterase inhibitor, is used for the treatment of dementia of the Alzheimer's type and mild to moderate dementia associated with Parkinson's disease.
    B) WITHDRAWN FROM MARKET: Because of ongoing concerns over the safety of TACRINE it was withdrawn from the market in 2013. Other agents, such as metrifonate and velnacrine are also no longer used in the treatment of Alzheimer's disease.
    C) PHARMACOLOGY: The precise mechanism of action of rivastigmine is unknown. However, it is thought to exert is effect by enhancing cholinergic function, by increasing the concentration of acetylcholine through reversible inhibition of its hydrolysis by cholinesterase.
    D) EPIDEMIOLOGY: Exposure has occurred. Severe clinical events or fatalities are uncommon.
    E) WITH THERAPEUTIC USE
    1) The most common adverse effects reported with rivastigmine therapy include: nausea, vomiting, anorexia, dyspepsia, and asthenia. Other clinical events include dizziness, headache, urinary tract infection, fatigue, insomnia, and confusion. Infrequent events may include depression, anxiety, hallucination, and aggressive reactions. Fluid depletion may occur through bronchorrhea, vomiting and diarrhea, polyuria, and sweating.
    2) CHOLINERGIC ACTIVITY: Other events that can develop due to increases in cholinergic activity include extrapyramidal symptoms, worsening of parkinsonian symptoms (ie, tremor), seizures, increases in gastric acid secretion, at risk to develop ulcers or gastrointestinal bleeding, bradycardia, and possible urinary obstruction. Drugs like rivastigmine the increase cholinergic activity should also be given with caution in patients with an underlying history of asthma or obstructive pulmonary disease.
    3) POSTMARKETING EXPERIENCE: Based on postmarketing experience with rivastigmine, hepatitis, aggression, Stevens-Johnson syndrome and disseminated allergic dermatitis have been reported.
    F) WITH POISONING/EXPOSURE
    1) Overdose information is limited. Cholinergic toxicity has been reported after overdose of rivastigmine, typical findings can include severe nausea, vomiting, salivation, sweating, bradycardia, hypotension, respiratory depression, collapse and convulsions. In severe cases, muscle weakness can develop and may result in death if respiratory muscles become involved. Other symptoms that have been associated with overdose of rivastigmine include: diarrhea, abdominal pain, dizziness, tremor, headache, somnolence, confusional state, hyperhidrosis, hypertension, hallucinations and malaise. Depression of blood cholinesterase may occur following overdoses.

Vital Signs

    3.3.1) SUMMARY
    A) HYPOTENSION: Blood pressure may be decreased following an overdose.
    B) BRADYCARDIA: Pulse rate may be decreased following an overdose.
    3.3.4) BLOOD PRESSURE
    A) HYPOTENSION may occur in an overdose due to muscarinic cholinergic effects of tacrine (Wilcock et al, 1988; Forsyth et al, 1989; Prod Info Cognex(R), tacrine hydrochloride, 2000).
    3.3.5) PULSE
    A) BRADYCARDIA: Decreased pulse rate may occur due to cholinergic effects of these drugs (Wilcock et al, 1988; Forsyth et al, 1989; Prod Info Cognex(R), tacrine hydrochloride, 2000; Brvar et al, 2005; Sener & Ozsarac, 2006).

Heent

    3.4.1) SUMMARY
    A) Miosis, salivation, and lacrimation are common systemic effects of cholinergic agonists and may occur with tacrine in therapeutic or overdose amounts.
    3.4.3) EYES
    A) RIVASTIGMINE: MIOSIS may occur with systemic poisoning as a result of cholinergic agonist effects of these agents (Raucci et al, 2014; Lai et al, 2005).
    B) CASE REPORT: Miosis and nystagmus were reported in a 59-year-old man who intentionally ingested 288 mg of rivastigmine and 280 mg of citalopram in a suicide attempt (Brvar et al, 2005).
    3.4.5) NOSE
    A) TACRINE: RHINITIS has been reported as an adverse effect following therapeutic doses of tacrine in 8% of treated patients (Prod Info Cognex(R), tacrine hydrochloride, 2000).
    3.4.6) THROAT
    A) SALIVATION may occur with systemic poisonings of these agents (Forsyth et al, 1989; Prod Info Cognex(R), tacrine hydrochloride, 2000; Brvar et al, 2005; Sener & Ozsarac, 2006; Raucci et al, 2014).

Cardiovascular

    3.5.2) CLINICAL EFFECTS
    A) HYPOTENSIVE EPISODE
    1) WITH THERAPEUTIC USE
    a) TACRINE: Hypotension, accompanied by reflex tachycardia, may initially occur as a sign of cholinergic crisis (Prod Info Cognex(R), tacrine hydrochloride, 2000). Therapeutic doses may also result in a hypotensive episode as a result of cholinergic effects (Wilcock et al, 1988). Hypotension may be severe, leading to cardiovascular collapse or shock (Prod Info Cognex(R), tacrine hydrochloride, 2000).
    B) BRADYCARDIA
    1) WITH THERAPEUTIC USE
    a) Hypotension and sinus bradycardia of 40 beats per minute were reported in an elderly patient with Alzheimer's disease approximately 1 hour following a 25 mg oral dose. The patient collapsed and was unconscious. Recovery was uneventful and subsequent serial cardiac enzymes and ECG were normal (Wilcock et al, 1988).
    2) WITH POISONING/EXPOSURE
    a) TACRINE: Overdose may cause a cholinergic crisis which may be characterized by bradycardia as well as hypotension (Prod Info Cognex(R), tacrine hydrochloride, 2000; Cummings, 1998).
    b) CASE REPORT (RIVASTIGMINE): A 59-year-old man presented with somnolence and vomiting approximately 5 hours after ingesting 288 mg of rivastigmine and 280 mg of citalopram. Prior to presentation, he was found comatose and had two seizures that spontaneously resolved. Three episodes of bradycardia (30 bpm) occurred approximately 5 to 13 hours post presentation. With supportive therapy, the patient recovered and was discharged 5 days postingestion (Brvar et al, 2005).
    c) CASE REPORT (RIVASTIGMINE): A brief episode of bradycardia (50 bpm) in conjunction with respiratory depression (10 breaths/minute) was reported in a 38-year-old man approximately 4 hours after intentionally ingesting 90 mg of rivastigmine (Sener & Ozsarac, 2006).
    C) VENTRICULAR PREMATURE BEATS
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT (RIVASTIGMINE): A 3-year-old child inadvertently ingested 1 to 2 4.5 mg rivastigmine tablets and rapidly developed confusion, drowsiness, hyporeactivity, sialorrhea, sweating, miosis, 2 episodes of diarrhea and evidence of respiratory failure requiring immediate intubation. A reduction of serum cholinesterase to 732 Units/L (normal, 3000 to 11,000 Units/L) was also noted shortly after exposure. An initial ECG showed sinus rhythm at 134 beats/min, normal AV conduction, normal QT interval and frequent ventricular and monormorphic ectopic beats (VEBs) with bigeminy and trigeminy. An echocardiogram was normal. Although the patient was extubated within 13 hours, she had persistent VEBs and was placed on a 24-holter monitor which showed multiple episodes of nonsustained ventricular tachycardia (maximum 3 beats) with no supraventricular tachycardia detected. The patient was discharged on day 4 with no cardiac symptoms and no further therapy was prescribed. Outpatient follow-up showed a normal ECG but she continued to have frequent isolated VEBs by Holter monitoring. Eventually, these episodes became more infrequent and the child remained asymptomatic and was developing normally. The authors suggested that exposure to rivastigmine caused overstimulation of the autonomic nervous system which uncovered a previously silent condition in this patient (Raucci et al, 2014).
    D) HEART BLOCK
    1) Various dysrhythmias, primarily heart block, may theoretically occur because of its cholinergic effects.
    E) HYPERTENSIVE EPISODE
    1) WITH POISONING/EXPOSURE
    a) RIVASTIGMINE: Hypertension has been reported following overdose ingestions of rivastigmine (Sener & Ozsarac, 2006; Brvar et al, 2005).

Respiratory

    3.6.2) CLINICAL EFFECTS
    A) ACUTE LUNG INJURY
    1) Dyspnea and bronchoconstriction may be noted and is more pronounced in asthmatic patients. Bronchospasm or pulmonary edema may be life-threatening. Cholinergic crisis may result in muscle weakness which may cause a respiratory paralysis leading to apnea (Prod Info Cognex(R), tacrine hydrochloride, 2000).
    2) ECHOTHIOPHATE IODIDE: A 76-year-old woman developed bronchospasm, dyspnea and ultimately respiratory failure secondary to cholinergic toxicity from echothiophate eye drops (Manoguerra et al, 1995).
    B) DECREASED RESPIRATORY FUNCTION
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT (RIVASTIGMINE): Respiratory depression (10 breaths/minute) was reported in a 38-year-old man approximately 4 hours after intentionally ingesting 90 mg of rivastigmine. His oxygen saturation (SpO2) was 91%. With supportive care, the patient completely recovered 16 hours later (Sener & Ozsarac, 2006).
    b) CASE REPORT (RIVASTIGMINE): A 3-year-old child inadvertently ingested 1 to 2 4.5 mg rivastigmine tablets and rapidly developed confusion, drowsiness, hyporeactivity, sialorrhea, sweating, miosis and 2 episodes of diarrhea. She also develop early signs of respiratory failure and required immediate intubation and sedation. A reduction in serum cholinesterase level to 732 Units/L (normal, 3000 to 11,000 Units/L) was also noted shortly after exposure. Thirteen hours after admission, the child was slowly weaned off the respirator and was successfully extubated. Neurologic function was normal (Raucci et al, 2014).
    C) TACHYPNEA
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT (RIVASTIGMINE): Rapid, shallow breathing (28 breaths/minute) with periods of apnea occurred in a 59-year-old man after intentionally ingesting 288 mg of rivastigmine and 280 mg of citalopram in a suicide attempt. Lung examination revealed diffuse crackles and his oxygen saturation (SpO2) was 76% on room air. He also developed excessive salivary and bronchial secretions. Following supportive therapy, including mechanical ventilation for approximately 24 hours, the patient recovered and was discharged 5 days postingestion (Brvar et al, 2005).
    D) SPUTUM ABNORMAL - AMOUNT
    1) WITH THERAPEUTIC USE
    a) TACRINE: Bronchial secretions are increased and bouts of coughing may occur (Prod Info Cognex(R), tacrine hydrochloride, 2000).

Neurologic

    3.7.2) CLINICAL EFFECTS
    A) SEIZURE
    1) WITH POISONING/EXPOSURE
    a) TACRINE: Overdose may result in seizures, tremor, weakness, and vertigo from a cholinergic toxicity to the CNS (Summers et al, 1980; Prod Info Cognex(R), tacrine hydrochloride, 2000; Brvar et al, 2005).
    B) COMA
    1) WITH THERAPEUTIC USE
    a) TACRINE: Coma was noted in less than 1:1000 patients following therapeutic doses, but is an effect that may be seen on occasion with cholinergic drug overdoses (Prod Info Cognex(R), tacrine hydrochloride, 2000).
    2) WITH POISONING/EXPOSURE
    a) CASE REPORT (RIVASTIGMINE): A 59-year-old man presented with somnolence and vomiting approximately 5 hours after ingesting 288 mg of rivastigmine and 280 mg of citalopram. Prior to presentation, he was found comatose and had two seizures that spontaneously resolved. Three episodes of bradycardia (30 bpm) occurred approximately 5 to 13 hours post presentation. With supportive therapy, the patient recovered and was discharged 5 days postingestion (Brvar et al, 2005).
    C) PSYCHOMOTOR AGITATION
    1) WITH THERAPEUTIC USE
    a) RIVASTIGMINE: In postmarketing experience, aggression has been reported with therapeutic use of rivastigmine (Prod Info EXELON(R) oral capsules, oral solution, 2015).
    b) TACRINE: Agitation has been reported in 4% of patients treated with tacrine and is attributed to cholinergic toxicity (Wilcock et al, 1988). Agitation is commonly cited as a reason for drug withdrawal in clinical trials (Prod Info Cognex(R), tacrine hydrochloride, 2000).
    D) EXTRAPYRAMIDAL DISEASE
    1) WITH THERAPEUTIC USE
    a) RIVASTIGMINE: An exacerbation of extrapyramidal symptoms, in particular tremor, has been reported following therapeutic doses of rivastigmine in patients with dementia associated with Parkinson's disease (Prod Info EXELON(R) oral capsules, oral solution, 2015).
    b) CASE REPORT (TACRINE): A 67-year-old woman with a history of mild Parkinson's disease, not currently treated, and a 2 to 3 year history of progressive Alzheimer's disease who was started on tacrine 40 mg/day for 6 weeks was then increased to 80 mg/day. Two weeks after increasing the dose, the patient presented with severe tremor, nocturnal restlessness, limb stiffness, and stumbling gait. Because tacrine was improving her Alzheimer's symptoms, it was continued, and carbidopa-levodopa was added and extrapyramidal symptoms improved (Ott & Lannon, 1992).
    E) HEADACHE
    1) WITH THERAPEUTIC USE
    a) RIVASTIGMINE: Headache has been reported with therapeutic use of rivastigmine (Prod Info EXELON(R) oral capsules, oral solution, 2015).
    b) TACRINE: Frontal headaches have been described in patients on therapeutic tacrine doses. This is presumed to be due to a cholinergic effect (Forsyth et al, 1989; Prod Info Cognex(R), tacrine hydrochloride, 2000).
    F) CHOLINERGIC CRISIS
    1) WITH THERAPEUTIC USE
    a) CASE REPORT/ECHOTHIOPHATE IODIDE: A 76-year-old woman developed a severe cholinergic syndrome following the use of echothiophate iodide eye drops for glaucoma. The patient presented with profound muscle weakness and was given an initial diagnosis of myasthenia gravis. Laboratory tests revealed severely depressed red blood cell and serum cholinesterase levels (3 Units/mL and 550 Units/mL, respectively), and the eye drops were discontinued. Symptoms spontaneously resolved (Manoguerra et al, 1995).
    2) WITH POISONING/EXPOSURE
    a) RIVASTIGMINE
    1) CASE REPORT: An 11-month-old girl presented with rapid onset of generalized weakness, hypotonia, hyporeflexia, miosis and a weak cry after her mother had found her chewing on a capsule of rivastigmine. She did not develop diarrhea, excessive urination or lacrimation. After 24 hours she was sitting without assistance, and strength had returned to normal by 48 hours (Lai et al, 2005).
    2) CASE REPORT: A 3-year-old child inadvertently ingested 1 to 2 4.5 mg rivastigmine tablets and rapidly developed confusion, drowsiness, hyporeactivity, sialorrhea, sweating, miosis and 2 episodes of diarrhea. She also develop early signs of respiratory failure and required immediate intubation and sedation. A reduction in serum cholinesterase level to 732 Units/L (normal, 3000 to 11,000 Units/L) was noted shortly after exposure. Within a few hours serum cholinesterase levels gradually improved and returned to normal with supportive therapy (Raucci et al, 2014).

Gastrointestinal

    3.8.2) CLINICAL EFFECTS
    A) CHOLINERGIC CRISIS
    1) WITH THERAPEUTIC USE
    a) Patients receiving metrifonate (15 mg/kg) experienced adverse effects of nausea, vomiting and diarrhea, which were not seen at lower doses (Cummings, 1998). Gastroenteritis appears to be a dose-dependent effect (Prod Info Cognex(R), tacrine hydrochloride, 2000).
    b) The most common adverse effects, occurring in at least 5% of patients, reported with rivastigmine therapy include: nausea, vomiting, anorexia, dyspepsia, and asthenia. These events are likely due to the cholinergic effects of rivastigmine (Prod Info EXELON(R) oral capsules, oral solution, 2015).
    2) WITH POISONING/EXPOSURE
    a) Overdose may result in cholinergic signs and symptoms with these agents, including severe nausea and vomiting, abdominal pain, diarrhea, and salivation as a result of increased gastrointestinal motility (Prod Info EXELON(R) oral capsules, oral solution, 2015; Forsyth et al, 1989; Wilcock et al, 1988; Gauthier et al, 1989; Filip et al, 1991; Knapp et al, 1994; Davis et al, 1992; Prod Info Cognex(R), tacrine hydrochloride, 2000; Brvar et al, 2005; Sener & Ozsarac, 2006; Raucci et al, 2014).
    B) PEPTIC ULCER
    1) WITH THERAPEUTIC USE
    a) RIVASTIGMINE: An increase in gastric acid secretion due to an increase in cholinergic activity can develop with rivastigmine therapy. Patients may be at increased risk to develop peptic ulcers and/or gastrointestinal bleeding (Prod Info EXELON(R) oral capsules, oral solution, 2015).

Hepatic

    3.9.2) CLINICAL EFFECTS
    A) INFLAMMATORY DISEASE OF LIVER
    1) WITH THERAPEUTIC USE
    a) RIVASTIGMINE: In postmarketing experience, hepatitis has been reported with therapeutic use of rivastigmine (Prod Info EXELON(R) oral capsules, oral solution, 2015).
    B) LIVER DAMAGE
    1) WITH THERAPEUTIC USE
    a) TACRINE
    1) Significant dose-related elevations in liver enzymes, primarily SGPT (ALT), have been observed in 20% to 40% of Alzheimer's patients within 6 to 8 weeks after beginning oral tacrine. This appears to be a reversible effect (Watkins et al, 1994; Farlow et al, 1992; Summers et al, 1989; Gauthier et al, 1989; Forsyth et al, 1989; Davies et al, 1989; Marx, 1987; Byrne & Arie, 1989). Females are at higher risk than males (Prod Info Cognex(R), tacrine hydrochloride, 2000).
    2) Liver biopsies in several patients with elevated hepaticenzymes have demonstrated granulomatous hepatitis (Ames et al, 1988; Davies et al, 1989) and liver cell necrosis (Davies et al, 1989; Hammel et al, 1990). An immunologic mechanism has been suggested (Ames et al, 1988).
    3) In a clinical trial of 14 patients, four patients who received 150 to 200 mg/day tacrine were excluded from the trial due to increased plasma SGPT(Davies et al, 1989). Liver biopsies revealed liver cell necrosis in 3 patients and evidence of a granulomatous drug reaction in one. Liver enzymes returned to normal within several weeks of discontinuing the drug.
    4) Hepatic tissue was obtained from 4 cases who had abnormal liver enzymes in a clinical trial. All 4 showed focal mononuclear cell inflammation scattered with occasional eosinophils. No signs of non-caseating epithelial granulomatous reactions or cholestasis were evident (Summers et al, 1989).
    5) CASE REPORT (TACRINE): A 76-year-old woman, on 75 mg/day tacrine, presented to the ED 20 days after initiation of tacrine therapy with fever and jaundice. Her history was insignificant for previous liver disease, surgery, blood transfusions, or drug addiction. No other drugs were taken. Liver biopsy showed marked centrilobular hepatocyte necrosis associated with mild inflammatory infiltration. Liver function returned to normal within a few days of stopping tacrine (Hammel et al, 1990).

Genitourinary

    3.10.2) CLINICAL EFFECTS
    A) INCREASED FREQUENCY OF URINATION
    1) TACRINE: Urinary frequency, stimulation of ureters and urinary bladder may occur, with resultant involuntary urination as a result of cholinergic effects of tacrine, especially at higher doses or overdoses (Summers et al, 1986; Wilcock et al, 1988; Prod Info Cognex(R), tacrine hydrochloride, 2000).
    B) URINARY TRACT OBSTRUCTION
    1) WITH THERAPEUTIC USE
    a) RIVASTIGMINE: Drugs that increase cholinergic activity such as rivastigmine may increase the risk of urinary obstruction (Prod Info EXELON(R) oral capsules, oral solution, 2015).

Hematologic

    3.13.2) CLINICAL EFFECTS
    A) AGRANULOCYTOSIS
    1) WITH THERAPEUTIC USE
    a) TACRINE: One case of agranulocytosis has been reported out of a total patient population of 2568 tacrine treated patients (Anon, 1993; Prod Info Cognex(R), tacrine hydrochloride, 2000).
    b) TACRINE: Adverse hematologic effects reported in 0.1% to 1% of patients in clinical trials included anemia and lymphadenopathy, while leukopenia, thrombocytopenia, hemolysis, and pancytopenia were recorded less than 1:1000 patients (Prod Info Cognex(R), tacrine hydrochloride, 2000).
    B) DEFICIENCY OF CHOLINESTERASE
    1) WITH POISONING/EXPOSURE
    a) TACRINE: Depression of blood cholinesterase may occur following overdoses with these drugs. Decreases seen in plasma cholinesterase are immediate, while there is a gradual decline in erythrocyte cholinesterase levels (Prod Info Cognex(R), tacrine hydrochloride, 2000; Manoguerra et al, 1995; Raucci et al, 2014).
    b) CASE REPORT (RIVASTIGMINE): A 3-year-old child inadvertently ingested 1 to 2 4.5 mg rivastigmine tablets and developed a reduction in serum cholinesterase level to 732 Units/L (normal, 3000 to 11,000 Units/L) shortly after exposure. Within a few hours serum cholinesterase levels gradually improved and returned to normal with supportive therapy (Raucci et al, 2014).

Dermatologic

    3.14.2) CLINICAL EFFECTS
    A) EXCESSIVE SWEATING
    1) WITH THERAPEUTIC USE
    a) RIVASTIGMINE: Increased sweating has been reported with therapeutic use and following overdose of rivastigmine (Prod Info EXELON(R) oral capsules, oral solution, 2015).
    b) TACRINE: Diaphoresis occurred in 1% of treated patients (Prod Info Cognex(R), tacrine hydrochloride, 2000; Summers et al, 1986). This complication can usually be controlled by dose reduction or administration of a peripheral anticholinergic, such as glycopyrrolate (Summers et al, 1986).
    2) WITH POISONING/EXPOSURE
    a) RIVASTIGMINE: Overdosage and/or inadvertent exposure in a child may result in a cholinergic crisis which can cause increased sweating (Prod Info EXELON(R) oral capsules, oral solution, 2015; Sener & Ozsarac, 2006; Raucci et al, 2014).
    B) PURPURA
    1) WITH THERAPEUTIC USE
    a) TACRINE: Purpura has occurred in 2% of tacrine-treated patients in controlled clinical trials (Prod Info Cognex(R), tacrine hydrochloride, 2000).

Musculoskeletal

    3.15.2) CLINICAL EFFECTS
    A) MUSCLE PAIN
    1) WITH THERAPEUTIC USE
    a) TACRINE: Myalgia (9%) is among the more common adverse effects encountered during clinical trials with tacrine. Although not necessarily related to tacrine administration, fractures, arthralgia, arthritis, and hypertonias occurred in 1% of treated patients. Less frequently (0.1% to 1%), osteoporosis, tendonitis, bursitis, and gout were listed, with myopathy a rare (less than 1:1000 patients) event (Prod Info Cognex(R), tacrine hydrochloride, 2000).

Reproductive

    3.20.1) SUMMARY
    A) Rivastigmine is classified as US FDA Pregnancy Category B. Although animals studies involving rats and rabbits revealed no evidence of teratogenicity, reduced fetal/pup weights were observed in rats at maternally toxic doses of rivastigmine. It is unknown whether rivastigmine is excreted in human breast milk; however, the drug is excreted in rat milk. Echothiophate is classified as US FDA Pregnancy Category C.
    3.20.2) TERATOGENICITY
    A) LACK OF INFORMATION
    1) RIVASTIGMINE
    a) There are no adequate and well-controlled studies of rivastigmine in pregnant women (Prod Info EXELON(R) oral capsules, oral solution, 2013; Prod Info EXELON(R) PATCH transdermal system, 2012).
    B) ANIMAL STUDIES
    1) RIVASTIGMINE
    a) In animal studies, there was no evidence of teratogenicity following oral administration of rivastigmine to pregnant rats and rabbits. However, decreased fetal/pup weights were noted in rats at doses below the maximum recommended human dose but often resulted in some maternal toxicity (Prod Info EXELON(R) oral capsules, oral solution, 2013; Prod Info EXELON(R) PATCH transdermal system, 2012).
    3.20.3) EFFECTS IN PREGNANCY
    A) PREGNANCY CATEGORY
    1) Rivastigmine is classified as US FDA Pregnancy Category B (Prod Info EXELON(R) oral capsules, oral solution, 2013; Prod Info EXELON(R) PATCH transdermal system, 2012).
    2) Echothiophate is classified as US FDA Pregnancy Category C.
    3) Metrifonate is classified as US FDA Pregnancy Category B.
    3.20.4) EFFECTS DURING BREAST-FEEDING
    A) LACK OF EFFECT
    1) RIVASTIGMINE
    a) It is unknown whether rivastigmine is excreted in human breast milk (Prod Info EXELON(R) oral capsules, oral solution, 2013; Prod Info EXELON(R) PATCH transdermal system, 2012).
    B) ANIMAL STUDIES
    1) RIVASTIGMINE
    a) Concentrations of rivastigmine and its metabolites were approximately 2-fold higher in milk than in plasma when rats were administered rivastigmine orally (Prod Info EXELON(R) oral capsules, oral solution, 2013; Prod Info EXELON(R) PATCH transdermal system, 2012).

Carcinogenicity

    3.21.3) HUMAN STUDIES
    A) TACRINE
    1) Tacrine may be carcinogenic since it belongs to the chemical class, acridines, of which some members are animal carcinogens (Prod Info Cognex(R), tacrine hydrochloride, 2000).

Genotoxicity

    A) TACRINE
    1) Tacrine mutagenic to bacteria in the Ames test. In vitro, unscheduled DNA synthesis was induced in rat and mouse hepatocytes. Chromosomal aberration studies results were equivocal. In the in vitro mammalian mutation test, tacrine was not mutagenic (Prod Info Cognex(R), tacrine hydrochloride, 2000).
    B) RIVASTIGMINE
    1) Rivastigmine was not mutagenic in an in vitro bacterial reverse mutation (Ames) assay, hypoxanthine guanine phosphorbosyltransferase (HGPRT) assay, and an in vivo mouse micronucleus tests. It was clastogenic in in vitro chromosomal aberration studies in mammalian cells only in the presence of metabolic activation (Prod Info EXELON(R) oral capsules, oral solution, 2013).

Monitoring Parameters Levels

    4.1.1) SUMMARY
    A) Monitor vital signs and mental status.
    B) Monitor fluid status and electrolytes in patients that develop significant vomiting, diarrhea and sweating.
    C) Monitor red blood cell and plasma cholinesterase levels in patients with signs and symptoms of cholinergic poisoning.
    D) Signs and symptoms of cholinesterase inhibitor poisoning should be monitored. Cholinesterase activity may be depressed and should be monitored in plasma and whole blood.
    E) Monitor ECG following significant exposures. Overdoses have been associated with bradycardia.
    4.1.2) SERUM/BLOOD
    A) BLOOD/SERUM CHEMISTRY
    1) Monitor serum electrolytes. Volume depletion may occur from overdoses.
    2) Monitor serum renal function tests in severe exposures.
    B) HEMATOLOGIC
    1) Hematologic effects, such as leukopenia, thrombocytopenia and anemia should be monitored.
    C) ACID/BASE
    1) Monitor blood gases in comatose patients and those in whom aspiration of vomitus is suspected.
    D) OTHER
    1) Monitor red blood cell and plasma cholinesterase levels in patients with signs and symptoms of cholinergic poisoning.
    4.1.4) OTHER
    A) OTHER
    1) MONITORING
    a) ECG should be monitored for signs of cholinesterase inhibitor toxicity, such as bradycardia.
    b) EEG should be monitored for signs of seizure activity.
    c) Blood pressure should be monitored for hypotensive signs of cholinergic toxicity.
    d) Pulmonary function testing and chest x-ray may be indicated in patients with dyspnea or bronchospasm.

Radiographic Studies

    A) CHEST RADIOGRAPH
    1) Obtain a chest x-ray in patients with severe respiratory or CNS depression.

Methods

    A) CHROMATOGRAPHY
    1) TACRINE: Tacrine can be measured in biologic fluids by high performance liquid chromatography (HPLC) utilizing fluorescence detection (Forsyth et al, 1988; Cutler et al, 1990; Hartvig et al, 1990).

Life Support

    A) Support respiratory and cardiovascular function.

Patient Disposition

    6.3.1) DISPOSITION/ORAL EXPOSURE
    6.3.1.1) ADMISSION CRITERIA/ORAL
    A) Patients who develop cholinergic crisis including respiratory depression should be admitted. They should only be discharged when free of symptoms.
    6.3.1.2) HOME CRITERIA/ORAL
    A) All children with ingestions should be sent to a healthcare facility for evaluation and treatment. Adults with a deliberate overdose should be sent to a healthcare facility for evaluation and treatment. Asymptomatic adults with an inadvertent ingestion of one or two pills can be monitored at home.
    6.3.1.3) CONSULT CRITERIA/ORAL
    A) Consult a medical toxicologist or a poison center for assistance with medical management in patients with severe overdose or in whom the diagnosis is unclear. Consult a pulmonologist or hospital intensivist if the patient develops evidence of respiratory compromise.
    6.3.1.5) OBSERVATION CRITERIA/ORAL
    A) Patients should be observed until all symptoms resolve. Patients that develop significant toxicity including respiratory depression should be admitted for a higher level of care.

Monitoring

    A) Monitor vital signs and mental status.
    B) Monitor fluid status and electrolytes in patients that develop significant vomiting, diarrhea and sweating.
    C) Monitor red blood cell and plasma cholinesterase levels in patients with signs and symptoms of cholinergic poisoning.
    D) Signs and symptoms of cholinesterase inhibitor poisoning should be monitored. Cholinesterase activity may be depressed and should be monitored in plasma and whole blood.
    E) Monitor ECG following significant exposures. Overdoses have been associated with bradycardia.

Oral Exposure

    6.5.1) PREVENTION OF ABSORPTION/PREHOSPITAL
    A) SUMMARY
    1) Spontaneous vomiting is likely to occur after a significant exposure. Activated charcoal is not indicated prehospital
    6.5.2) PREVENTION OF ABSORPTION
    A) ACTIVATED CHARCOAL
    1) CHARCOAL ADMINISTRATION
    a) Consider administration of activated charcoal after a potentially toxic ingestion (Chyka et al, 2005). Administer charcoal as an aqueous slurry; most effective when administered within one hour of ingestion.
    2) CHARCOAL DOSE
    a) Use a minimum of 240 milliliters of water per 30 grams charcoal (FDA, 1985). Optimum dose not established; usual dose is 25 to 100 grams in adults and adolescents; 25 to 50 grams in children aged 1 to 12 years (or 0.5 to 1 gram/kilogram body weight) ; and 0.5 to 1 gram/kilogram in infants up to 1 year old (Chyka et al, 2005).
    1) Routine use of a cathartic with activated charcoal is NOT recommended as there is no evidence that cathartics reduce drug absorption and cathartics are known to cause adverse effects such as nausea, vomiting, abdominal cramps, electrolyte imbalances and occasionally hypotension (None Listed, 2004).
    b) ADVERSE EFFECTS/CONTRAINDICATIONS
    1) Complications: emesis, aspiration (Chyka et al, 2005). Aspiration may be complicated by acute respiratory failure, ARDS, bronchiolitis obliterans or chronic lung disease (Golej et al, 2001; Graff et al, 2002; Pollack et al, 1981; Harris & Filandrinos, 1993; Elliot et al, 1989; Rau et al, 1988; Golej et al, 2001; Graff et al, 2002). Refer to the ACTIVATED CHARCOAL/TREATMENT management for further information.
    2) Contraindications: unprotected airway (increases risk/severity of aspiration) , nonfunctioning gastrointestinal tract, uncontrolled vomiting, and ingestion of most hydrocarbons (Chyka et al, 2005).
    6.5.3) TREATMENT
    A) SUPPORT
    1) Monitor and support respiratory and cardiovascular function.
    B) ATROPINE
    1) ATROPINE
    a) SUMMARY: Overdose with cholinesterase inhibitors such as these agents can cause a cholinergic crisis characterized by severe nausea, vomiting, salivation, sweating, bradycardia, hypotension, collapse, and convulsions (Prod Info COGNEX(R) oral capsules, 2002).
    b) ATROPINE SULFATE is the drug of choice and will reverse actions at muscarinic receptors and most nicotinic receptors.
    c) ADULT: DOSE: Initial dose of 1 to 2 mg IV repeat with subsequent doses based on clinical response (Prod Info COGNEX(R) oral capsules, 2002).
    d) CHILD: DOSE: Initial dose 0.05 mg/kg up to 4 mg (usual dose 1 mg), intramuscularly or intravenously every 10 to 30 minutes until muscarinic signs and symptoms subside, and repeat if they reappear (Prod Info COGNEX(R) oral capsules, 2002).
    2) ALTERNATIVE THERAPY
    a) ATROPINE ALTERNATIVE: Glycopyrrolate and Methscopolamine bromide have been suggested as alternatives to atropine in treating the peripheral cholinergic symptoms induced by cholinergic, muscarinic agonists (Granacher et al, 1976).
    1) These agents should not contribute to the initial anticholinergic delirium, as they do not easily cross the blood brain barrier.
    2) Controversy exists on the effectiveness of glycopyrrolate to reverse cholinergic effects of tacrine. Wilcock et al (1988) and Summers et al (1986) found glycopyrrolate effective in reversing mild side effects. Levy (1988) found glycopyrrolate to aggravate the adverse effects of tacrine. Summers et al (1980) suggest glycopyrrolate to be effective only where peripheral muscarine toxicity is noted. These agents should be used cautiously, as atypical increases in blood pressure and heart rate have been reported (Prod Info Cognex(R), 1995).
    C) BRONCHOSPASM
    1) BRONCHOSPASM SUMMARY
    a) Administer beta2 adrenergic agonists. Consider use of inhaled ipratropium and systemic corticosteroids. Monitor peak expiratory flow rate, monitor for hypoxia and respiratory failure, and administer oxygen as necessary.
    2) ALBUTEROL/ADULT DOSE
    a) 2.5 to 5 milligrams diluted with 4 milliliters of 0.9% saline by nebulizer every 20 minutes for three doses. If incomplete response, administer 2.5 to 10 milligrams every 1 to 4 hours as needed OR administer 10 to 15 milligrams every hour by continuous nebulizer as needed. Consider adding ipratropium to the nebulized albuterol; DOSE: 0.5 milligram by nebulizer every 30 minutes for three doses then every 2 to 4 hours as needed, NOT administered as a single agent (National Heart,Lung,and Blood Institute, 2007).
    3) ALBUTEROL/PEDIATRIC DOSE
    a) 0.15 milligram/kilogram (minimum 2.5 milligrams) diluted with 4 milliliters of 0.9% saline by nebulizer every 20 minutes for three doses. If incomplete response administer 0.15 to 0.3 milligram/kilogram (maximum 10 milligrams) every 1 to 4 hours as needed OR administer 0.5 mg/kg/hr by continuous nebulizer as needed. Consider adding ipratropium to the nebulized albuterol; DOSE: 0.25 to 0.5 milligram by nebulizer every 20 minutes for three doses then every 2 to 4 hours as needed, NOT administered as a single agent (National Heart,Lung,and Blood Institute, 2007).
    4) ALBUTEROL/CAUTIONS
    a) The incidence of adverse effects of beta2-agonists may be increased in older patients, particularly those with pre-existing ischemic heart disease (National Asthma Education and Prevention Program, 2007). Monitor for tachycardia, tremors.
    5) CORTICOSTEROIDS
    a) Consider systemic corticosteroids in patients with significant bronchospasm. PREDNISONE: ADULT: 40 to 80 milligrams/day in 1 or 2 divided doses. CHILD: 1 to 2 milligrams/kilogram/day (maximum 60 mg) in 1 or 2 divided doses (National Heart,Lung,and Blood Institute, 2007).
    D) 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).
    E) SEIZURE
    1) Overdose with cholinesterase inhibitors such as rivastigmine can result in cholinergic crisis and can include potentially severe adverse events including hypotension, bradycardia, respiratory depress, collapse, and seizures (Prod Info EXELON(R) oral capsules, oral solution, 2015).
    2) SUMMARY
    a) Attempt initial control with a benzodiazepine (eg, diazepam, lorazepam). If seizures persist or recur, administer phenobarbital or propofol.
    b) Monitor for respiratory depression, hypotension, and dysrhythmias. Endotracheal intubation should be performed in patients with persistent seizures.
    c) Evaluate for hypoxia, electrolyte disturbances, and hypoglycemia (or, if immediate bedside glucose testing is not available, treat with intravenous dextrose).
    3) DIAZEPAM
    a) ADULT DOSE: Initially 5 to 10 mg IV, OR 0.15 mg/kg IV up to 10 mg per dose up to a rate of 5 mg/minute; may be repeated every 5 to 20 minutes as needed (Brophy et al, 2012; Prod Info diazepam IM, IV injection, 2008; Manno, 2003).
    b) PEDIATRIC DOSE: 0.1 to 0.5 mg/kg IV over 2 to 5 minutes; up to a maximum of 10 mg/dose. May repeat dose every 5 to 10 minutes as needed (Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008).
    c) Monitor for hypotension, respiratory depression, and the need for endotracheal intubation. Consider a second agent if seizures persist or recur after repeated doses of diazepam .
    4) NO INTRAVENOUS ACCESS
    a) DIAZEPAM may be given rectally or intramuscularly (Manno, 2003). RECTAL DOSE: CHILD: Greater than 12 years: 0.2 mg/kg; 6 to 11 years: 0.3 mg/kg; 2 to 5 years: 0.5 mg/kg (Brophy et al, 2012).
    b) MIDAZOLAM has been used intramuscularly and intranasally, particularly in children when intravenous access has not been established. ADULT DOSE: 0.2 mg/kg IM, up to a maximum dose of 10 mg (Brophy et al, 2012). PEDIATRIC DOSE: INTRAMUSCULAR: 0.2 mg/kg IM, up to a maximum dose of 7 mg (Chamberlain et al, 1997) OR 10 mg IM (weight greater than 40 kg); 5 mg IM (weight 13 to 40 kg); INTRANASAL: 0.2 to 0.5 mg/kg up to a maximum of 10 mg/dose (Loddenkemper & Goodkin, 2011; Brophy et al, 2012). BUCCAL midazolam, 10 mg, has been used in adolescents and older children (5-years-old or more) to control seizures when intravenous access was not established (Scott et al, 1999).
    5) LORAZEPAM
    a) MAXIMUM RATE: The rate of intravenous administration of lorazepam should not exceed 2 mg/min (Brophy et al, 2012; Prod Info lorazepam IM, IV injection, 2008).
    b) ADULT DOSE: 2 to 4 mg IV initially; repeat every 5 to 10 minutes as needed, if seizures persist (Manno, 2003; Brophy et al, 2012).
    c) PEDIATRIC DOSE: 0.05 to 0.1 mg/kg IV over 2 to 5 minutes, up to a maximum of 4 mg/dose; may repeat in 5 to 15 minutes as needed, if seizures continue (Brophy et al, 2012; Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008; Sreenath et al, 2009; Chin et al, 2008).
    6) PHENOBARBITAL
    a) ADULT LOADING DOSE: 20 mg/kg IV at an infusion rate of 50 to 100 mg/minute IV. An additional 5 to 10 mg/kg dose may be given 10 minutes after loading infusion if seizures persist or recur (Brophy et al, 2012).
    b) Patients receiving high doses will require endotracheal intubation and may require vasopressor support (Brophy et al, 2012).
    c) PEDIATRIC LOADING DOSE: 20 mg/kg may be given as single or divided application (2 mg/kg/minute in children weighing less than 40 kg up to 100 mg/min in children weighing greater than 40 kg). A plasma concentration of about 20 mg/L will be achieved by this dose (Loddenkemper & Goodkin, 2011).
    d) REPEAT PEDIATRIC DOSE: Repeat doses of 5 to 20 mg/kg may be given every 15 to 20 minutes if seizures persist, with cardiorespiratory monitoring (Loddenkemper & Goodkin, 2011).
    e) MONITOR: For hypotension, respiratory depression, and the need for endotracheal intubation (Loddenkemper & Goodkin, 2011; Manno, 2003).
    f) SERUM CONCENTRATION MONITORING: Monitor serum concentrations over the next 12 to 24 hours. Therapeutic serum concentrations of phenobarbital range from 10 to 40 mcg/mL, although the optimal plasma concentration for some individuals may vary outside this range (Hvidberg & Dam, 1976; Choonara & Rane, 1990; AMA Department of Drugs, 1992).
    7) OTHER AGENTS
    a) If seizures persist after phenobarbital, propofol or pentobarbital infusion, or neuromuscular paralysis with general anesthesia (isoflurane) and continuous EEG monitoring should be considered (Manno, 2003). Other anticonvulsants can be considered (eg, valproate sodium, levetiracetam, lacosamide, topiramate) if seizures persist or recur; however, there is very little data regarding their use in toxin induced seizures, controlled trials are not available to define the optimal dosage ranges for these agents in status epilepticus (Brophy et al, 2012):
    1) VALPROATE SODIUM: ADULT DOSE: An initial dose of 20 to 40 mg/kg IV, at a rate of 3 to 6 mg/kg/minute; may give an additional dose of 20 mg/kg 10 minutes after loading infusion. PEDIATRIC DOSE: 1.5 to 3 mg/kg/minute (Brophy et al, 2012).
    2) LEVETIRACETAM: ADULT DOSE: 1000 to 3000 mg IV, at a rate of 2 to 5 mg/kg/min IV. PEDIATRIC DOSE: 20 to 60 mg/kg IV (Brophy et al, 2012; Loddenkemper & Goodkin, 2011).
    3) LACOSAMIDE: ADULT DOSE: 200 to 400 mg IV; 200 mg IV over 15 minutes (Brophy et al, 2012). PEDIATRIC DOSE: In one study, median starting doses of 1.3 mg/kg/day and maintenance doses of 4.7 mg/kg/day were used in children 8 years and older (Loddenkemper & Goodkin, 2011).
    4) TOPIRAMATE: ADULT DOSE: 200 to 400 mg nasogastric/orally OR 300 to 1600 mg/day orally divided in 2 to 4 times daily (Brophy et al, 2012).
    F) HYPOTENSIVE EPISODE
    1) Overdose with cholinesterase inhibitors such as rivastigmine can result in cholinergic crisis and can include potentially severe adverse events including hypotension, bradycardia, respiratory depression, collapse, and seizures (Prod Info EXELON(R) oral capsules, oral solution, 2015).
    2) SUMMARY
    a) Infuse 10 to 20 milliliters/kilogram of isotonic fluid and keep the patient supine. If hypotension persists, administer dopamine or norepinephrine. Consider central venous pressure monitoring to guide further fluid therapy.
    3) DOPAMINE
    a) DOSE: Begin at 5 micrograms per kilogram per minute progressing in 5 micrograms per kilogram per minute increments as needed (Prod Info dopamine hcl, 5% dextrose IV injection, 2004). If hypotension persists, dopamine may need to be discontinued and a more potent vasoconstrictor (eg, norepinephrine) should be considered (Prod Info dopamine hcl, 5% dextrose IV injection, 2004).
    b) CAUTION: If ventricular dysrhythmias occur, decrease rate of administration (Prod Info dopamine hcl, 5% dextrose IV injection, 2004). Extravasation may cause local tissue necrosis, administration through a central venous catheter is preferred (Prod Info dopamine hcl, 5% dextrose IV injection, 2004).
    4) NOREPINEPHRINE
    a) PREPARATION: 4 milligrams (1 amp) added to 1000 milliliters of diluent provides a concentration of 4 micrograms/milliliter of norepinephrine base. Norepinephrine bitartrate should be mixed in dextrose solutions (dextrose 5% in water, dextrose 5% in saline) since dextrose-containing solutions protect against excessive oxidation and subsequent potency loss. Administration in saline alone is not recommended (Prod Info norepinephrine bitartrate injection, 2005).
    b) DOSE
    1) ADULT: Dose range: 0.1 to 0.5 microgram/kilogram/minute (eg, 70 kg adult 7 to 35 mcg/min); titrate to maintain adequate blood pressure (Peberdy et al, 2010).
    2) CHILD: Dose range: 0.1 to 2 micrograms/kilogram/minute; titrate to maintain adequate blood pressure (Kleinman et al, 2010).
    3) CAUTION: Extravasation may cause local tissue ischemia, administration by central venous catheter is advised (Peberdy et al, 2010).
    G) BRADYCARDIA
    1) ATROPINE/DOSE
    a) ADULT BRADYCARDIA: BOLUS: Give 0.5 milligram IV, repeat every 3 to 5 minutes, if bradycardia persists. Maximum: 3 milligrams (0.04 milligram/kilogram) intravenously is a fully vagolytic dose in most adults. Doses less than 0.5 milligram may cause paradoxical bradycardia in adults (Neumar et al, 2010).
    b) PEDIATRIC DOSE: As premedication for emergency intubation in specific situations (eg, giving succinylchoine to facilitate intubation), give 0.02 milligram/kilogram intravenously or intraosseously (0.04 to 0.06 mg/kg via endotracheal tube followed by several positive pressure breaths) repeat once, if needed (de Caen et al, 2015; Kleinman et al, 2010). MAXIMUM SINGLE DOSE: Children: 0.5 milligram; adolescent: 1 mg.
    1) There is no minimum dose (de Caen et al, 2015).
    2) MAXIMUM TOTAL DOSE: Children: 1 milligram; adolescents: 2 milligrams (Kleinman et al, 2010).
    H) PRALIDOXIME
    1) Pralidoxime should be considered in patients with severe nicotinic effects after large, acute, recent exposures.
    2) PRALIDOXIME/INDICATIONS
    a) Severe organophosphate poisoning with nicotinic (muscle and diaphragmatic weakness, respiratory depression, fasciculations, muscle cramps, etc) and/or central (coma, seizures) manifestations should be treated with pralidoxime in addition to atropine(Prod Info PROTOPAM(R) Chloride injection, 2010).
    3) PRALIDOXIME/CONTROVERSY
    a) Human studies have not substantiated the benefit of oxime therapy in acute organophosphate poisoning (Eddleston et al, 2002; de Silva et al, 1992); however oxime dosing in these studies was not optimized and methodology was unclear. Most authors advocate the continued use of pralidoxime in the clinical setting of severe organophosphate poisoning (Singh et al, 2001; Singh et al, 1998).
    b) It has been difficult to assess the value of pralidoxime in case studies because most of the patients have also received atropine therapy, or the pralidoxime was given late in the treatment or at a suboptimal dose (Peter et al, 2006; Rahimi et al, 2006).
    c) More recent observational studies have indicated that acetylcholinesterase inhibited by various organophosphate (OP) pesticides varies in its responsiveness to oximes; diethyl OPs (eg, parathion, quinalphos) appear to be effectively reactivated by oximes, while dimethyl OPs (eg, monocrotophos or oxydemeton-methyl) appear to respond poorly. Profenofos, an OP that is AChE inhibited by a S-alkyl link, was also found to not reactivate at all to oximes (Eddleston et al, 2008).
    4) PRALIDOXIME DOSE
    a) ADULT: A loading dose of 30 mg/kg (maximum: 2 grams) over 30 minutes followed by a maintenance infusion of 8 to 10 mg/kg/hr (up to 650 mg/hr) (Howland, 2011). In vitro studies have recommended a target plasma concentration of close to 17 mcg/mL necessary for pralidoxime to be effective, which is higher than the previously suggested concentration of at least 4 mcg/mL (Howland, 2011; Eddleston et al, 2002). ALTERNATE ADULT: An alternate initial dose for adults is 1 to 2 grams diluted in 100 mL of 0.9% sodium chloride infused over 15 to 30 minutes. Repeat initial bolus dose in 1 hour and then every 3 to 8 hours if muscle weakness or fasciculations persist (continuous infusion preferred). In patients with serious cholinergic intoxication, a continuous infusion of 500 mg/hr should be considered. In patients with acute lung injury, a 5% solution may be administered by a slow IV injection over at least 5 minutes (Howland, 2006). Intravenous dosing is preferred; however, intramuscular administration may be considered using a 1-g vial of pralidoxime reconstituted with 3 mL of sterile water for injection or 0.9% sodium chloride for injection, producing a solution containing 300 mg/mL (Howland, 2011). An initial intramuscular pralidoxime dose of 1 gram or up to 2 grams in cases of very severe poisoning has also been recommended (Haddad, 1990; S Sweetman , 2002).
    b) CHILD: A loading dose of 20 to 40 mg/kg (maximum: 2 grams/dose) infused over 30 to 60 minutes in 0.9% sodium chloride (Howland, 2006; Schexnayder et al, 1998). Repeat initial bolus dose in 1 hour and then every 3 to 8 hours if muscle weakness or fasciculations persist (continuous infusion preferred). ALTERNATE CHILD: An alternate loading dose of 25 to 50 mg/kg (up to a maximum dose of 2 g), followed via continuous infusion of 10 to 20 mg/kg/hr. In patients with serious cholinergic intoxication, a continuous infusion of 10 to 20 mg/kg/hr up to 500 mg/hr should be considered (Howland, 2006).
    c) Presently, the ideal dose has NOT been established and dosing is likely based on several factors: type of OP agent (ie, diethyl OPs appear to respond more favorably to oximes, while dimethyl OPs seem to respond poorly) which may relate to a variation in the speed of ageing, time since exposure, body load, and pharmacogenetics (Eddleston et al, 2008)
    d) CONTINUOUS INFUSION
    1) A continuous infusion of pralidoxime is generally preferred to intermittent bolus dosing to maintain a target concentration with less variation (Howland, 2011; Eddleston et al, 2008; Roberts & Aaron, 2007; Gallagher et al, 1989; Thompson, 1987). In an open label, randomized study of moderately severe organophosphate poisoned patients treated with high dose continuous infusions required less atropine, were less likely to be intubated and had shorter duration of ventilatory support than patients treated with intermittent bolus doses. HIGH DOSE CONTINUOUS INFUSION: In this study, an initial 2 g bolus (pralidoxime chloride or iodide) was given, followed by 1 g over an hour every hour for 48 hours. Followed by 1 g every 4 hours until the patient could be weaned from mechanical ventilation. The response to therapy was beneficial in patients exposed to either a dimethyl or diethyl organophosphate pesticide (Pawar et al, 2006).
    2) Infusion over a period of several days may be necessary and is generally well tolerated (Namba et al, 1971).
    e) MAXIMUM DOSE
    1) The maximum recommended dose for pralidoxime is 12 grams in 24 hours for adults (S Sweetman , 2002); based on WHO, this dose may be exceeded in severely poisoned adults (Tang et al, 2013).
    f) DURATION OF INTRAVENOUS DOSING
    1) Dosing should be continued for at least 24 hours after cholinergic manifestations have resolved (Howland, 2006). Prolonged administration may be necessary in severe cases, especially in the case of poisoning by lipophilic organophosphates (Wadia & Amin, 1988). Observe patients carefully for recurrent cholinergic manifestations after pralidoxime is discontinued.
    5) PRALIDOXIME/ADMINISTRATION
    a) Pralidoxime is best administered as soon as possible after exposure; ideally, within 36 hours of exposure (Prod Info PROTOPAM(R) CHLORIDE injection, 2006). However, patients presenting late (2 to 6 days after exposure) may still benefit (Borowitz, 1988; De Kort et al, 1988; Namba et al, 1971; Amos & Hall, 1965) .
    b) Some mechanisms which may account for pralidoxime efficacy with delayed administration include:
    1) Poisoning with an agent such as parathion or quinalphos which produces "slow aging" of acetylcholinesterase (Eddleston et al, 2008).
    2) Slow absorption of the organophosphate compound from the lower bowel or exposure to large amounts (Prod Info PROTOPAM(R) CHLORIDE injection, 2006).
    3) Release of the organophosphate from fat stores (Borowitz, 1988).
    4) Other actions of pralidoxime.
    6) SUMMARY
    a) Minimal toxicity when administered as directed; adverse effects may include: pain at injection site; transient elevations of CPK, SGOT, SGPT; dizziness, blurred vision, diplopia, drowsiness, nausea, tachycardia, hyperventilation, and muscular weakness (Prod Info PROTOPAM(R) CHLORIDE injection, 2006). Rapid injection may produce laryngospasm, muscle rigidity and tachycardia (Prod Info PROTOPAM(R) CHLORIDE injection, 2006).
    7) MINIMAL TOXICITY
    a) When administered as directed, pralidoxime has minimal toxicity (Prod Info PROTOPAM(R) CHLORIDE injection, 2006). Up to 40.5 grams have been administered over seven days (26 grams in the first 54 hours) without ill effects (Namba et al, 1971).
    b) One child developed delirium, visual hallucinations, tachycardia, mydriasis, and dry mucous membranes (Farrar et al, 1990). The authors were uncertain if these effects were related to 2-PAM or organophosphate poisoning per se.
    8) NEUROMUSCULAR BLOCKADE
    a) High doses have been reported to cause neuromuscular blockade, but this would not be expected to occur with recommended doses (Grob & Johns, 1958).
    9) VISUAL DISTURBANCES
    a) Oximes have produced visual disturbances (eg, blurred vision, diplopia) (Prod Info PROTOPAM(R) CHLORIDE injection, 2006).
    b) Transient increases in intraocular pressure may occur (Ballantyne B, 1987).
    10) ASYSTOLE
    a) Pralidoxime administered intravenously at an infusion rate of 2 grams over 10 minutes was associated with asystole in a single reported case, which occurred about 2 minutes after initiation of the infusion (Scott, 1986). A cause and effect relationship was not established.
    11) WEAKNESS
    a) Mild weakness, blurred vision, dizziness, headache, nausea, and tachycardia may occur if the rate of pralidoxime infusion exceeds 500 milligrams/minute (Jager & Stagg, 1958).
    12) ATROPINE SIDE EFFECTS
    a) Concomitant administration of pralidoxime may enhance the side effects of atropine administration (Hiraki et al, 1958). The signs of atropinization may occur earlier than anticipated when the agents are used together (Prod Info PROTOPAM(R) CHLORIDE injection, 2006).
    13) CARDIOVASCULAR
    a) Transient dose-dependent increases in blood pressure have occurred in adults receiving 15 to 30 milligrams/kilogram of 2-PAM (Calesnick et al, 1967). Increases in systolic and diastolic blood pressure have been observed in healthy volunteers given parenteral doses of pralidoxime (Prod Info PROTOPAM(R) CHLORIDE injection, 2006).
    b) Electrocardiographic changes and marked hypertension were observed at doses of 45 milligrams/kilogram (Calesnick et al, 1967).

Enhanced Elimination

    A) HEMODIALYSIS
    1) Due to the short half-life (1.5 hours) of rivastigmine, dialysis (hemodialysis, peritoneal dialysis, or hemofiltration) is not clinically indicated. Rivastigmine also has a large volume of distribution (range, 1.8 to 2.7 L/kg); therefore, hemodialysis is unlikely to be beneficial (Prod Info EXELON(R) oral capsules, oral solution, 2015).

Summary

    A) TOXIC DOSE: A toxic dose has not been established. RIVASTIGMINE: An adult developed vomiting, bradycardia, hypertension, salivation, diaphoresis and drowsiness after ingesting 90 mg. TACRINE: An estimated human lethal dose is reported as 30 mg/kg when unopposed by anticholinergics. METRIFONATE: Doses as high as 72 mg/kg have produced severe abdominal colic, vomiting, and muscle weakness.
    B) THERAPEUTIC DOSE: Dosing for Alzheimer's Disease is as follows: RIVASTIGMINE: 1.5 mg orally twice daily; if tolerated, increase dose every 2 weeks by 1.5 mg twice daily to a maximum dose of 6 mg twice daily. Other agents (ie, tacrine and metrifonate) have been withdrawn from the market.

Therapeutic Dose

    7.2.1) ADULT
    A) SPECIFIC SUBSTANCE
    1) RIVASTIGMINE
    a) ALZHEIMER-ASSOCIATED DEMENTIA
    1) ORAL: 3 to 6 mg orally twice daily. The recommended starting oral dose is 1.5 mg twice daily, which may be increased after 2 weeks based on tolerability. MAXIMUM DOSE: 12 mg/day. If dosing is interrupted for less than or equal to 3 days, resume treatment at same or lower dose. If interruption lasts for more than 3 days, restart treatment at 1.5 mg twice daily and titrate appropriately (Prod Info EXELON(R) oral capsules, oral solution, 2013).
    2) TRANSDERMAL: The recommended starting dose is 4.6 mg/24 hr patch applied to the skin once daily. After 4 weeks, the dose may be increased to 9.5 mg/24 hr patch applied to the skin once daily. The maintenance dose for mild to moderate Alzheimer's disease is 9.5 mg/24 hours patch once daily; after a minimum of 4 weeks and good tolerability, may titrate to a maximum of 13.3 mg/24 hours patch once daily when no longer receiving therapeutic benefit at lower dose. The maintenance dose for severe Alzheimer's disease is 13.3 mg/24 hours patch once daily after minimum 4 weeks and good tolerability at lower dose of 9.5 mg/24 hours. MAXIMUM DOSE: 13.3 mg/24 hr patch applied to the skin once daily (Prod Info EXELON(R) PATCH transdermal patch, 2013).
    b) PARKINSON-ASSOCIATED DEMENTIA
    1) ORAL: 1.5 to 6 mg orally twice daily. The recommended starting oral dose is 1.5 mg twice daily, which may be increased after 4 weeks based on tolerability. MAXIMUM DOSE: 12 mg/day. If dosing is interrupted for less than or equal to 3 days, resume treatment at same or lower dose. If interruption lasts for more than 3 days, restart treatment at 1.5 mg twice daily and titrate appropriately (Prod Info EXELON(R) oral capsules, oral solution, 2013).
    2) TRANSDERMAL: The recommended starting dose is 4.6 mg/24 hr patch applied to the skin once daily. After 4 weeks, the dose may be increased to 9.5 mg/24 hr patch applied to the skin once daily for as long as therapeutic effect is maintained; may increase to a maximum effective dose of 13.3 mg/24 hours once daily. MAXIMUM DOSE: 13.3 mg/24 hr patch applied to the skin once daily (Prod Info EXELON(R) PATCH transdermal patch, 2013).
    2) TACRINE
    a) NOTE: Tacrine was withdrawn from the market in 2013. This information is kept for historical purposes only.
    b) Dose titration is required, with initial doses starting at 10 mg 4 times daily. Increase in 40 mg/day increments at 6 week intervals, up to a maximum of 160 mg/day (Prod Info Cognex(R), tacrine hydrochloride, 2000).
    c) Dose-related hepatotoxic effects have been observed (Marx, 1987; Gauthier et al, 1989) and the drug should be used with caution or not at all in patients with a history of past or current liver disease. Although prompt withdrawal of tacrine with evidence of drug-induced elevations in liver enzymes generally leads to reversal, long-term consequences of liver function are unknown (Prod Info Cognex(R), tacrine hydrochloride, 2000).
    3) METRIFONATE
    a) Dose titration for treatment of Alzheimer's Disease is required. A loading dosage of 2 mg/kg/day for 2 weeks followed by 0.65 mg/kg/day has been effective in clinical trials. Lower dosage regimens did not appear to be as effective (Lamb & Faulds, 1997).
    4) ECHOTHIOPHATE
    a) For open-angle glaucoma give 1 drop of 0.03% solution in affected eye twice daily. Use nasolacrimal occlusion to prevent systemic absorption (Prod Info Phospholine iodide(R), echothiophate, 1996).
    7.2.2) PEDIATRIC
    A) RIVASTIGMINE
    1) Safety and efficacy have not been established in pediatric patients (Prod Info EXELON(R) PATCH transdermal patch, 2013).

Minimum Lethal Exposure

    A) SUMMARY
    1) TACRINE
    a) Summers et al (1980) estimated the human lethal dose to be approximately 30 mg/kg when unopposed by anticholinergic agents. This is based on LD50 studies in animals and prelethal toxicity.

Maximum Tolerated Exposure

    A) SUMMARY
    1) RIVASTIGMINE
    a) CASE REPORT: A 59-year-old man presented with somnolence and vomiting approximately 5 hours after ingesting 288 mg of rivastigmine and 280 mg of citalopram. Prior to presentation, he was found comatose and had 2 seizures that spontaneously resolved. Three episodes of bradycardia (30 bpm) occurred approximately 5 to 13 hours post presentation. With supportive therapy, the patient recovered and was discharged 5 days post ingestion (Brvar et al, 2005).
    b) CASE REPORT: Dizziness, nausea and vomiting, sweating, salivation, hypertension, bradycardia and respiratory depression (respiratory rate of 10 per minute, oxygen saturation greater than 90%, no intervention needed) occurred in a 38-year-old man after intentionally ingesting 90 mg of rivastigmine. With supportive therapy, the patient completely recovered 16 hours postingestion (Sener & Ozsarac, 2006).
    2) TACRINE
    a) Reversible hepatotoxicity has been seen with dosages of 80 mg/day (Watkins et al, 1994).
    3) METRIFONATE
    a) Oral doses as high as 72 mg/kg have produced severe abdominal colic, vomiting, and muscle weakness (Williams, 1999).

Toxicity Information

    7.7.1) TOXICITY VALUES
    A) ECHOTHIOPHATE IODIDE
    1) LD50- (ORAL)MOUSE:
    a) 5100 mcg/kg (RTECS, 2002)
    2) LD50- (SUBCUTANEOUS)MOUSE:
    a) 130 mcg/kg (RTECS, 2002)
    3) LD50- (ORAL)RAT:
    a) 174 mcg/kg (RTECS, 2002)
    4) LD50- (SKIN)RAT:
    a) >100 mg/kg (RTECS, 2002)
    5) LD50- (SUBCUTANEOUS)RAT:
    a) 174 mcg/kg (RTECS, 2002)
    B) METRIFONATE
    1) LD50- (ORAL)MOUSE:
    a) 300 mg/kg (RTECS, 2002)
    2) LD50- (SKIN)MOUSE:
    a) 1710 mg/kg (RTECS, 2002)
    3) LD50- (SUBCUTANEOUS)MOUSE:
    a) 267 mg/kg (RTECS, 2002)
    4) LD50- (INTRAMUSCULAR)RAT:
    a) 395 mg/kg (RTECS, 2002)
    5) LD50- (ORAL)RAT:
    a) 450 mg/kg (RTECS, 2002)
    6) LD50- (SKIN)RAT:
    a) 2 gm/kg (RTECS, 2002)
    C) TACRINE
    1) LD50- (INTRAMUSCULAR)MOUSE:
    a) 29 mg/kg (RTECS, 2002)
    2) LD50- (INTRAPERITONEAL)MOUSE:
    a) 33 mg/kg (Gershon & Olaria, 1960)
    3) LD50- (ORAL)MOUSE:
    a) 39800 mcg/kg (RTECS, 2002)
    4) LD50- (INTRAMUSCULAR)RAT:
    a) 34 mg/kg (RTECS, 2002)
    5) LD50- (INTRAPERITONEAL)RAT:
    a) 35-40 mg/kg (Gershon & Olaria, 1960)
    6) LD50- (ORAL)RAT:
    a) 40 mg/kg (Prod Info Cognex(R), 2000)
    b) 70 mg/kg (RTECS, 2002)

Pharmacologic Mechanism

    A) RIVASTIGMINE
    1) The exact mechanism of rivastigmine is unknown but it is thought to exert its effect by enhancing cholinergic function. This appears to occur by increasing the concentration of acetylcholine through reversible inhibition of its hydrolysis by cholinesterase (Prod Info EXELON(R) oral capsules, oral solution, 2015).
    B) TACRINE
    1) SUMMARY: In vitro and in vivo studies have demonstrated that these agents are potent inhibitors of acetylcholinesterase as well as butyrylcholinesterase. They are also partial agonists at muscarinic receptors and block reuptake of dopamine, serotonin and norepinephrine, and inhibit monoamine oxidase activity (Shutske et al, 1989; Summers et al, 1980; Mesulam et al, 1987; Ho & Freeman, 1965; Heilbronn, 1961).
    2) The rationale for the use of these agents in Alzheimer's disease is based on well-established evidence of a severe loss of cholinergic neurons in this degenerative disease (Davies & Maloney, 1988) (Byrne & Arie, 1989) (Coyle et al, 1983) and the association between cholinergic pathways and memory disorder (Smith & Swash, 1978; Anon, 1987; Drachman & Leavitt, 1974). Deficits in the cholinergic system have correlated with the severity of dementia, as well as senile plaques and neurofibrillary tangles (Perry et al, 1978; Byrne & Arie, 1989).
    3) Tacrine has also been demonstrated to potently inhibit cholinesterases within plaques and tangles (Mesulam et al, 1987). In Alzheimer's patients, increases in cerebrospinal fluid acetylcholine have been observed with tacrine treatment (Sattin et al, 1989), and the drug has produced modest but significant clinical improvement (Gauthier et al, 1989; Kaye et al, 1982). However, tacrine does not alter the progressive nature of the disease and with continual loss of functioning cholinergic neurons any benefit of tacrine will be lost eventually (Prod Info Cognex(R), tacrine hydrochloride, 2000).
    4) In addition to potent anticholinesterase activity, tacrine has also been reported to produce other effects which may contribute to benefits in Alzheimer's patients. The drug has been demonstrated to produce selective blockade of potassium channels in the central nervous system, an effect attributed to its structural similarity to 4-aminopyridine (Soni & Kam, 1982; Sattin et al, 1989). A complex inhibition of sodium inactivation, similar to that produced by gallamine, has also been reported (Schauf & Sattin, 1987).
    a) These effects could facilitate release of acetylcholine and prolongation of the action potential of presynaptic cholinergic neurons (Soni & Kam, 1982; Sattin et al, 1989; Schauf & Sattin, 1987). Other actions of tacrine have included alterations in phosphorylation and down regulation of M1 receptors (Byrne & Arie, 1989).
    b) It has been suggested that a hydroxylated metabolite of tacrine may chelate aluminum, which has been speculated to be a possible environmental pathogen in Alzheimer's disease (Sattin et al, 1989). However, there are no scientific data to support these properties of tacrine.
    C) ECHOTHIOPHATE
    1) Echothiophate is an irreversible cholinesterase inhibitor. Its actions are similar to those of neostigmine, but more prolonged. Miotic action begins within 10 to 30 minutes and persists for 1 to 4 weeks, causing a reduction in intra-ocular pressure (Sweetman, 2002).
    D) METRIFONATE
    1) Metrifonate inhibits the cholinesterases by covalent attachment, with resulting dimethyl phosphoryl-enzyme which hydrolyses only slowly, giving rise to a long lasting enzyme inhibition. The ratio of central to peripheral inhibition of cholinesterase is enhanced due to the property of this drug to readily penetrate the blood-brain barrier (Taylor, 1997).
    2) Enzyme inhibition following oral metrifonate has reached levels of 40% to 60% and 85% to 90% for acetyl- and butyrylcholinesterase, respectively (Giacobini, 1997).

Toxicologic Mechanism

    A) RIVASTIGMINE: As a cholinesterase inhibitor it can result in cholinergic crisis following overdose that can be characterized by severe nausea, vomiting, salivation, sweating, bradycardia, hypotension, respiratory depression, collapse and convulsions (Prod Info EXELON(R) oral capsules, oral solution, 2015).
    B) TACRINE: Nonpredictable elevations of serum liver transaminases associated with tacrine therapy in vivo are dependent upon bioactivation of tacrine, mediated by hepatic CYP1A2, to form a toxic compound (Madden et al, 1995).

General Bibliography

    1) AMA Department of DrugsAMA Department of Drugs: AMA Evaluations Subscription, American Medical Association, Chicago, IL, 1992.
    2) Ames DJ, Bhathal PS, & Davies BM: Hepatotoxicity of tetrahydroacridine (letter). Lancet 1988; 1:887.
    3) Amos WC Jr & Hall A: Malathion poisoning treated with protopam. Ann Intern Med 1965; 62:1013-1016.
    4) Anon: Alzheimer drug dropped. Pharmaceutical J 1994; 253:186.
    5) Anon: Cholinergic treatment in Alzheimer's disease: encouraging results. Lancet 1987; 1:139-141.
    6) Anon: Tacrine for Alzheimer's disease. Med Lett Drugs Ther 1993; 35(905):87-8.
    7) 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.
    8) Ballantyne B: Oximes and visual disturbances: A review and experimental findings (Abstract). Presented at the AACT/AAPCC/ABMT/CAPCC Annual Scientific Meeting, Vancouver, BC (Sept 27-Oct 2), 1987.
    9) Borowitz SM: Prolonged organophosphate toxicity in a twenty-six-month-old child. J Pediatr 1988; 112:302-304.
    10) Brophy GM, Bell R, Claassen J, et al: Guidelines for the evaluation and management of status epilepticus. Neurocrit Care 2012; 17(1):3-23.
    11) 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.
    12) Brvar M, Mozina M, & Bunc M: Poisoning with rivastigmine. Clin Toxicol (Phila) 2005; 43(7):891-892.
    13) Byrne J & Arie T: Tetrahydroaminoacridine (THA) in Alzheimer's disease. Not ready for routine use. Br Med J 1989; 298:845-846.
    14) Calesnick B, Christensen JA, & Richter M: Human toxicity of various oximes. Arch Environ Health 1967; 15:599-608.
    15) 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.
    16) 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.
    17) 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.
    18) Choonara IA & Rane A: Therapeutic drug monitoring of anticonvulsants state of the art. Clin Pharmacokinet 1990; 18:318-328.
    19) Chyka PA, Seger D, Krenzelok EP, et al: Position paper: Single-dose activated charcoal. Clin Toxicol (Phila) 2005; 43(2):61-87.
    20) Cummings JL: Metrifonate: overview of safety and efficacy. Pharmacother 1998; 18:43S-46S.
    21) Cutler NR, Sedman AJ, & Prior P: Steady-state pharmacokinetics of tacrine in patients with Alzheimer's disease. Psychopharmacol Bull 1990; 26:231-234.
    22) Davies B, Andrews D, & Stargatt R: Tacrine in Alzheimer's disease (letter). Lancet 1989; 2:163-164.
    23) Davis KL, Thal LJ, & Gamzu ER: A double-blind, placebo-controlled multicenter study of tacrine for Alzheimer's disease. New Engl J Med 1992; 327:1253-1259.
    24) De Kort WL, Kiestra SH, & Sangster B: The use of atropine and oximes in organophosphate intoxications: a modified approach. Clin Toxicol 1988; 26:199-208.
    25) Drachman DA & Leavitt J: Human memory and the cholinergic system: a relationship to aging. Arch Neurol 1974; 30:113-121.
    26) Eddleston M, Buckley NA, Eyer P, et al: Management of acute organophosphorus pesticide poisoning. Lancet 2008; 371(9612):597-607.
    27) Eddleston M, Szinicz L, & Eyer P: Oximes in acute organophosphorus pesticide poisoning: a systemiatic review. Q J Med 2002; 95:275-283.
    28) Elliot CG, Colby TV, & Kelly TM: Charcoal lung. Bronchiolitis obliterans after aspiration of activated charcoal. Chest 1989; 96:672-674.
    29) FDA: Poison treatment drug product for over-the-counter human use; tentative final monograph. FDA: Fed Register 1985; 50:2244-2262.
    30) Farlow M, Gracon SI, & Hershey LA: A controlled trial of tacrine in Alzheimer's disease. JAMA 1992; 268:2523-2529.
    31) Farrar HC, Wells TG, & Kearns GL: Use of continuous infusion of pralidoxime for treatment of organophosphate poisoning in children. J Pediatr 1990; 116:658-661.
    32) Filip V, Vachek J, & Albrecht V: Pharmacokinetics and tolerance of 7-methoxytacrine following the single dose administration in healthy volunteers. Int J Clin Pharm 1991; 29:431-436.
    33) Forsyth DR, Ford JM, & Truman CA: Determination of tacrine hydrochloride in human serum by chloroform extraction, reversed-phase high-performance liquid chromatography and fluorimetric detection. J Chromatogr 1988; 433:352-358.
    34) Forsyth DR, Surmon DJ, & Morgan RA: Clinical experience with and side- effects of tacrine hydrochloride in Alzheimer's disease: a pilot study. Age Aging 1989; 18:223-229.
    35) Fredj G, Dietlin F, & Schwarzenberg L: Tetrahydroaminoacridine in HIV infections. Int J Clin Pharmacol Ther Toxicol 1989; 27:408-410.
    36) Gallagher K, Kearney T, & Mangione A: A case report of organophosphate (OP) poisoning supporting the use of pralidoxime (2-PAM) by continuous IV infusion (abstract 102). Vet Hum Toxicol 1989; 31:355.
    37) Gauthier S, Bouchard R, & Bacher Y: Progress report on the Canadian multicentre trial of tetrahydroaminoacridine with lecithin in Alzheimer's disease. Can J Neurol Sci 1989; 16:543-546.
    38) Giacobini E: Metrifonate - A viewpoint by Ezio Giacobini. Drugs & Aging 1997; 11(6):497.
    39) Golej J, Boigner H, Burda G, et al: Severe respiratory failure following charcoal application in a toddler. Resuscitation 2001; 49:315-318.
    40) Graff GR, Stark J, & Berkenbosch JW: Chronic lung disease after activated charcoal aspiration. Pediatrics 2002; 109:959-961.
    41) Granacher RP, Baldessarini RJ, & Messner E: Physostigmine treatment of delirium induced by anticholinergics. Am Family Pract 1976; 13:99-103.
    42) Grob D & Johns RJ: Use of oximes in the treatment of intoxication by anticholinesterase compounds in normal subjects. Am J Med 1958; 24:497.
    43) Haas CF: Mechanical ventilation with lung protective strategies: what works?. Crit Care Clin 2011; 27(3):469-486.
    44) Haddad LM: Organophosphates and other insecticides In: Haddad LM: Haddad LM, Winchester JF. Clinical management of poisoning and drug overdose, 2nd. W.B. Saunders Company, Philadelphia, 1990, pp 1076-87.
    45) Hammel P, Larrey D, & Bernuau J: Acute hepatitis after tetrahydroaminoacridine administration for Alzheimer's disease. J Clin Gastroenterol 1990; 12:329-331.
    46) Harris CR & Filandrinos D: Accidental administration of activated charcoal into the lung: aspiration by proxy. Ann Emerg Med 1993; 22:1470-1473.
    47) Hartvig P, Askmark H, & Aquilonius SM: Clinical pharmacokinetics of intravenous and oral 9-amino-1,2,3,4-tetrahydroacridine, tacrine. Eur J Clin Pharmacol 1990; 38:259-263.
    48) Hegenbarth MA & American Academy of Pediatrics Committee on Drugs: Preparing for pediatric emergencies: drugs to consider. Pediatrics 2008; 121(2):433-443.
    49) Heilbronn E: Inhibition of cholinesterases by tetrahydroaminoacridine. Acta Chem Scand 1961; 15:1386.
    50) Hiraki K, Namba Y, & Taniguchi Y: Effect of 2-pyridine aldoxime methiodide (PAM) against parathion (Folidol) poisoning. Analysis of 39 cases. Naika Ryoiki 1958; 6:84.
    51) Ho AK & Freeman SE: Anticholinesterase activity by tetrahydroaminoacrine and succinylcholine hydrosis. Nature 1965; 205:1118.
    52) Howland MA: Pralidoxime. In: Goldfrank LR, Flomenbaum N, Hoffman RS, et al, eds. Goldfrank's Toxicologic Emergencies, 9th ed. McGraw-Hill, New York, NY, 2011.
    53) Howland MA: Pralidoxime. In: Goldfrank LR, Flomenbaum N, Hoffman RS, et al, eds. Goldfrank's Toxicologic Emergencies. 8th ed., 8th ed. McGraw-Hill, New York, NY, 2006, pp -.
    54) Hvidberg EF & Dam M: Clinical pharmacokinetics of anticonvulsants. Clin Pharmacokinet 1976; 1:161.
    55) Jager BV & Stagg GN: Toxicity of diacetyl monoxime and of pyridine-2-aldoxime methiodide in man. Bull John Hopkins Hosp 1958; 102:203.
    56) Kaye WH, Sitaram N, & Weingartner H: Modest facilitation of memory in dementia with combined lecithin and anticholinesterase treatment. Biol Psychiatry 1982; 17:275-280.
    57) Kleinman ME, Chameides L, Schexnayder SM, et al: 2010 American Heart Association guidelines for cardiopulmonary resuscitation and emergency cardiovascular care. Part 14: pediatric advanced life support. Circulation 2010; 122(18 Suppl.3):S876-S908.
    58) Knapp MJ, Knopman DS, & Solomon PR: A 30-week randomized controlled trial of high-dose tacrine in patients with Alzheimer's disease. JAMA 1994; 271:985-991.
    59) Kollef MH & Schuster DP: The acute respiratory distress syndrome. N Engl J Med 1995; 332:27-37.
    60) Lai MW, Moen M, & Ewald MB: Pesticide-like poisoning from a prescription drug. N Eng J Med 2005; 353(3):317-318.
    61) Lamb HM & Faulds D: Metrifonate (review). Drugs & Aging 1997; 11(6):490-496.
    62) Loddenkemper T & Goodkin HP: Treatment of Pediatric Status Epilepticus. Curr Treat Options Neurol 2011; Epub:Epub.
    63) Madden S, Spaldin V, & Park BK: Clinical pharmacokinetics of tacrine. Clin Pharmacokinet 1995; 28(6):449-457.
    64) Manno EM: New management strategies in the treatment of status epilepticus. Mayo Clin Proc 2003; 78(4):508-518.
    65) Manoguerra A, Whitney C, & Clark RF: Cholinergic toxicity resulting from ocular instillation of echothiophate iodide eye drops. Clin Toxicol 1995; 33:463-465.
    66) Marx JL: Alzheimer's drug trial put on hold. Science 1987; 238:1041-1042.
    67) Mesulam MM, Geula C, & Moran MA: Anatomy of cholinesterase inhibition in Alzheimer's disease: effect of physostigmine and tetrahydroaminoacridine on plaques and tangles. Ann Neurol 1987; 22:683-691.
    68) 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.
    69) Namba T, Nolte CT, & Jackrel J: Poisoning due to organophosphate insecticides. Acute and chronic manifestations. Am J Med 1971; 50:475-492.
    70) National Asthma Education and Prevention Program: Expert Panel Report 3 (EPR-3): Guidelines for the Diagnosis and Management of Asthma-Summary Report 2007. J Allergy Clin Immunol 2007; 120(5 Suppl):S94-S138.
    71) 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.
    72) Neumar RW , Otto CW , Link MS , et al: Part 8: adult advanced cardiovascular life support: 2010 American Heart Association Guidelines for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2010; 122(18 Suppl 3):S729-S767.
    73) None Listed: Position paper: cathartics. J Toxicol Clin Toxicol 2004; 42(3):243-253.
    74) Ott BR & Lannon MC: Exacerbation of Parkinsonism by tacrine.. Clin Neuropharmacol 1992; 15(4):322-5.
    75) Pawar KS, Bhoite RR, Pillay CP, et al: Continuous pralidoxime infusion versus repeated bolus injection to treat organophosphorus pesticide poisoning: a randomised controlled trial. Lancet 2006; 368(9553):2136-2141.
    76) Peberdy MA , Callaway CW , Neumar RW , et al: 2010 American Heart Association guidelines for cardiopulmonary resuscitation and emergency cardiovascular care science. Part 9: post–cardiac arrest care. Circulation 2010; 122(18 Suppl 3):S768-S786.
    77) Perry EK, Tomlinson BE, & Blessed G: Correlation of cholinergic abnormalities with senile plaques and mental test scores in senile dementia. Br Med J 1978; 2:1457-1459.
    78) Peter JV, Moran JL, & Graham P: Oxime therapy and outcomes in human organophosphate poisoning: an evaluation using meta-analytic techniques. Crit Care Med 2006; 34(2):502-510.
    79) Pollack MM, Dunbar BS, & Holbrook PR: Aspiration of activated charcoal and gastric contents. Ann Emerg Med 1981; 10:528-529.
    80) Product Information: COGNEX(R) oral capsules, tacrine hydrochloride oral capsules. First Horizon Pharmaceutical Corp., Roswell, GA, 2002.
    81) Product Information: Cognex(R), tacrine hydrochloride. Parke-Davis, Morris Plains, NJ, 2000.
    82) Product Information: EXELON(R) PATCH transdermal patch, rivastigmine transdermal patch. Novartis Pharmaceuticals Corporation (per manufacturer), East Hanover, NJ, 2013.
    83) Product Information: EXELON(R) PATCH transdermal system, rivastigmine transdermal system. Novartis Pharmaceuticals Corporation (per FDA), East Hanover, NJ, 2015.
    84) Product Information: EXELON(R) PATCH transdermal system, rivastigmine transdermal system. Novartis Pharmaceuticals Corporation (per manufacturer), East Hanover, NJ, 2012.
    85) Product Information: EXELON(R) oral capsules, oral solution, rivastigmine tartrate oral capsules, oral solution. Novartis Pharmaceuticals Corporation (per FDA), East Hanover, NJ, 2013.
    86) Product Information: EXELON(R) oral capsules, oral solution, rivastigmine tartrate oral capsules, oral solution. Novartis Pharmaceuticals Corporation (per FDA), East Hanover, NJ, 2015.
    87) Product Information: PROTOPAM(R) CHLORIDE injection, pralidoxime chloride injection. Baxter Healthcare Corporation, Deerfield, IL, 2006.
    88) Product Information: PROTOPAM(R) Chloride injection, pralidoxime chloride injection. Baxter Healthcare Corporation, Deerfield, IL, 2010.
    89) Product Information: Phospholine iodide(R), echothiophate. Wyeth-Ayerst Laboratories, Philadelphia, PA, 1996.
    90) Product Information: diazepam IM, IV injection, diazepam IM, IV injection. Hospira, Inc (per Manufacturer), Lake Forest, IL, 2008.
    91) Product Information: dopamine hcl, 5% dextrose IV injection, dopamine hcl, 5% dextrose IV injection. Hospira,Inc, Lake Forest, IL, 2004.
    92) Product Information: lorazepam IM, IV injection, lorazepam IM, IV injection. Akorn, Inc, Lake Forest, IL, 2008.
    93) Product Information: norepinephrine bitartrate injection, norepinephrine bitartrate injection. Sicor Pharmaceuticals,Inc, Irvine, CA, 2005.
    94) Rahimi R, Nikfar S, & Abdollahi M: Increased morbidity and mortality in acute human organophosphate-poisoned patients treated by oximes: a meta-analysis of clinical trials. Hum Exp Toxicol 2006; 25(3):157-162.
    95) Rau NR, Nagaraj MV, Prakash PS, et al: Fatal pulmonary aspiration of oral activated charcoal. Br Med J 1988; 297:918-919.
    96) Raucci U, Vanacore N, Cecchetti C, et al: Transient cardiac effects in a child with acute cholinergic syndrome due to rivastigmine poisoning. J Emerg Med 2014; 47(1):21-25.
    97) Roberts DM & Aaron CK: Management of acute organophosphorus pesticide poisoning. BMJ 2007; 334(7594):629-634.
    98) S Sweetman : Martindale: The Complete Drug Reference. Pharmaceutical Press. London, England (Internet Version). Edition expires 2002; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    99) S Sweetman : Martindale: The Complete Drug Reference. Pharmaceutical Press. London, UK (Internet Version). Edition expires 2002; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    100) Sattin A, Muhoberac BB, & Aprison MH: Tetrahydroaminoacridine (THA) as a pharmacologic probe in Alzheimer's disease (AD) and other neurodegenerative disorders. Med Hypoth 1989; 29:155-159.
    101) Schauf CL & Sattin A: Tetrahydroaminoacridine blocks potsssium channels and inhibits sodium inactivation in Myxicola. J Pharmacol Exper Ther 1987; 243:609-613.
    102) Schexnayder S, Pames LP, & Kearns GL: The pharmacokinetics of Continuous infusion of pralidoxime in children with organophosphate poisoning. J Toxicol Clin Toxicol 1998; 36:549-555.
    103) 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.
    104) Scott RJ: Repeated asystole following PAM in organophosphate self-poisoning. Anaesth Intensive Care 1986; 14:458-468.
    105) Sener S & Ozsarac M: Case of the month: rivastigmine (Exelon) toxicity with evidence of respiratory depression. Emerg Med J 2006; 23(1):82-85.
    106) Shutske GM, Pierrat FA, & Kapples KJ: 9-Amino-1,2,3,4-tetrahydroacridin-1-ols: synthesis and evaluation as potential Alzheimer's disease therapeutics. J Med Chem 1989; 32:1805-1813.
    107) Singh G, Avasthi G, & Khurana D: Neurophysiological monitoring of pharmacological manipulation in acute organophosphate (OP) poisoning. The effects of pralidoxime, magnesium sulfate and pancuronium. Electroencephalol Clin Neurophysiol 1998; 107:140-148.
    108) Singh S, Chaudhry D, & Behera D: Aggressive atropinisation and continuous pralidoxime (2-PAM) infusion in patients with severe organophosphae poisoning: experience of a northwest Indian hospital. Human Exp Toxicol 2001; 20:15-18.
    109) Smith CM & Swash M: Hypothesis - possible biochemical basis of memory disorder in Alzheimer's disease. Ann Neurol 1978; 3:471-473.
    110) Soni N & Kam P: 4-Aminopyridine - a review. Anaesth Int Care 1982; 10:120-126.
    111) 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.
    112) 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.
    113) Summers WK, Kaufman KR, & Altman F: THA - a review of the literature and its use in treatment of five overdose patients. Clin Toxicol 1980; 16:269-281.
    114) Summers WK, Koehler AL, & Marsh GM: Long-term hepatotoxicity of tacrine (letter). Lancet 1989; 1:729.
    115) Summers WK, Majovski LV, & Marsh GM: Oral tetrahydroaminoacridine in long-term treatment of senile dementia, Alzheimer type. N Engl J Med 1986; 315:1241-1245.
    116) Tang X, Wang R, Xie H, et al: Repeated pulse intramuscular injection of pralidoxime chloride in severe acute organophosphorus pesticide poisoning. Am J Emerg Med 2013; 31(6):946-949.
    117) Taylor P: Metrifonate - A viewpoint by Palmer Taylor. Drugs & Aging 1997; 11(6):497.
    118) Thompson DF: Pralidoxime chloride continuous infusions. Ann Emerg Med 1987; 16:831-832.
    119) Wadia RS & Amin RB: Fenthion poisoning (letter). J Pediatr 1988; 113:950.
    120) Watkins PB, Zimmerman HJ, & Knapp MJ: Hepatotoxic effects of tacrine administration in patients with Alzheimer's disease. JAMA 1994; 271(13):992-998.
    121) Wilcock GK, Surmon D, & Forsyth D: Cholinergic side-effects of tetrahydroaminoacridine (letter). Lancet 1988; 2:1305.
    122) Willson DF, Truwit JD, Conaway MR, et al: The adult calfactant in acute respiratory distress syndrome (CARDS) trial. Chest 2015; 148(2):356-364.
    123) 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.
    124) de Caen AR, Berg MD, Chameides L, et al: Part 12: Pediatric Advanced Life Support: 2015 American Heart Association Guidelines Update for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2015; 132(18 Suppl 2):S526-S542.
    125) de Silva HJ, Wijewickrema R, & Senanayake N: Does pralidoxime affect outcome of management in acute organophosphorus poisoning?. Lancet 1992; 339(8802):1136-1138.