MOBILE VIEW  | 

ANESTHETICS-LOCAL

Classification   |    Detailed evidence-based information

Therapeutic Toxic Class

    A) This management includes lidocaine and related drugs with local anesthetic properties, with the exception of benzocaine, benzonatate, and cocaine (see separate Managements).
    B) Local anesthetics are divided into three groups on the basis of the type of bond (ester, amide, or neither) between their aromatic and amino.

Specific Substances

    A) ESTERS
    1) Amylocaine (synonym)
    2) Butacaine (synonym)
    3) Butyl aminobenzoate (synonym)
    4) Chloroprocaine (synonym)
    5) Cyclomethycaine (synonym)
    6) Hexylacaine (synonym)
    7) Isobucaine (synonym)
    8) Meprylcaine (synonym)
    9) Oxybuprocaine (benoxinate) (synonym)
    10) Piperocaine (synonym)
    11) Procaine (synonym)
    12) Proparacaine (proxymetacaine) (synonym)
    13) Propoxycaine (synonym)
    14) Tetracaine (amethocaine) (synonym)
    AMIDES
    1) Articaine (synonym)
    2) Bupivacaine (synonym)
    3) Butanilicaine (synonym)
    4) Carticaine (synonym)
    5) Dibucaine (cinchocaine) (synonym)
    6) Etidocaine (synonym)
    7) Lidocaine (lignocaine) (synonym)
    8) Mepivacaine (synonym)
    9) Oxethazaine (synonym)
    10) Prilocaine (propitocaine) (synonym)
    11) Ropivacaine (synonym)
    OTHER
    1) Diperidon (synonym)
    2) Dyclonine (synonym)
    3) LOCAL ANESTHETICS
    4) Pramoxine (synonym)

    1.2.1) MOLECULAR FORMULA
    1) CHLOROPROCAINE HYDROCHLORIDE: C13H19ClN2O2-HCl
    2) LIDOCAINE: C14H22N2O
    3) LIDOCAINE HYDROCHLORIDE MONOHYDRATE: C14H22N2O-HCl-H20
    4) MEPIVACAINE HYDROCHLORIDE: C15H22N2O-HCl
    5) ROPIVACAINE HYDROCHLORIDE: C17H26N2O-HCl-H2O
    6) TETRACAINE: C15H24N2O2

Available Forms Sources

    A) FORMS
    1) In pure form, local anesthetics are white, crystalline powders. Commercial preparations for infiltration anesthesia are available as solutions of 0.25% to 2% concentration. Higher concentrations may be used topically.
    2) Lidocaine is available as a liquid, powder, spray and oral viscous solutions, (2% and 4%), and a ready-to-use lidocaine 10% oral spray, delivering 800 metered doses per 60 mL aerosol bottle (each metered dose delivers 10 mg lidocaine base).
    3) Dyclonine hydrochloride is available in lozenges containing 1.2 mg each.
    4) Local anesthetics may also contain vasopressors such as epinephrine.
    B) USES
    1) A mixture of lidocaine (500 to 1000 mg/L), epinephrine (0.25 to 1 mg/L) and sodium bicarbonate (12.5 mmol/L) in 0.9% saline is infused into the surgical site for local anesthesia and to minimize blood loss during tumescent liposuction. Doses of lidocaine during this procedure can reach 55 mg/kg. Deaths from precipitous bradycardia and hypotension during this procedure may be secondary to lidocaine toxicity (Rao et al, 1999).

Life Support

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

Clinical Effects

    0.2.1) SUMMARY OF EXPOSURE
    A) USES: Local anesthetics are used for analgesia. They are administered topically and by local injection, regional injection, and for spinal and epidural anesthesia.
    B) PHARMACOLOGY: These agents reversibly block sodium channels and block the action potentials responsible for nerve conduction.
    C) TOXICOLOGY: Toxicity is caused by sodium channel blockade leading to CNS and cardiac effects. Some of these agents and their metabolites can oxidize iron leading to methemoglobinemia.
    D) EPIDEMIOLOGY: Toxicity is uncommon and severe toxicity is rare.
    E) WITH THERAPEUTIC USE
    1) Allergic reactions to the ester or amide classes of local anesthetics are possible though extremely rare. The ester class (benzocaine, chloroprocaine, cocaine, cyclomethycaine, dimethocaine, larocaine, propoxycaine, procaine, novocaine, proparacaine, tetracaine, and amethocaine) accounts for the majority of true IgE-mediated allergic reactions due to metabolism of PABA, a known allergen. Reactions to the amide class (articaine, bupivacaine, carticaine, cinchocaine, dibucaine, etidocaine, lidocaine, mepivacaine, piperocaine, prilocaine, ropivacaine, and trimecaine) are often secondary to the methylparaben preservative in the multidose vials. Clinically insignificant methemoglobinemia may result from exposure to many local anesthetics. Patients may develop vasovagal reactions following administration of these agents. Visual loss has rarely been associated with intranasal bupivacaine use during nasal surgery. Visual symptoms may persist for years.
    F) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: Subjective effects in mild toxicity include drowsiness, impending doom, headache, dizziness, paresthesias, euphoria, numbness of the mouth, lightheadedness, tinnitus, anxiety, confusion, tremors, agitation, disorientation, hallucinations, and lethargy. Peripheral effects may result in temporary paralysis. Suppression of the gag reflex may occur with oropharyngeal exposure. Methemoglobinemia can develop in patients who have no other evidence of toxicity.
    2) SEVERE TOXICITY: Severe toxicity may cause cardiac, respiratory, and CNS toxicity, as well as methemoglobinemia. CNS effects precede significant cardiovascular toxicity, except following massive IV injection. While there is often a progression of symptoms after IM, subQ, or continuous IV infusion, seizures and coma may occur suddenly after rapid IV administration. Cardiac effects are due to sodium channel blockade and can be manifested by a range of toxic effects including hypotension, AV block, bradycardia, QRS prolongation, ventricular dysrhythmias, asystole, and cardiovascular collapse. Spinal, epidural, or inadvertent intrathecal administration may lead to respiratory failure. Hypoxia may be secondary to cardiac toxicity or direct motor paralysis of respiratory muscles leading to respiratory depression, apnea, or respiratory arrest. Methemoglobinemia is well described with usage of prilocaine and benzocaine, and rarely reported with lidocaine, tetracaine, Cetacaine, and propitocaine. It can develop after therapeutic doses and in patients who have no other evidence of toxicity.
    3) INHALATION EXPOSURE: Methemoglobinemia can rarely result from inhalation exposure to local anesthetics.
    4) DERMAL EXPOSURE: Topical preparations can lead to systemic toxicity ranging from mild to severe. Infants are at a higher risk for systemic effects from dermal exposure.
    0.2.20) REPRODUCTIVE
    A) Lidocaine and the combination of lidocaine/prilocaine and lidocaine/tetracaine are classified as FDA pregnancy category B. Articaine hydrochloride/epinephrine and bupivacaine hydrochloride are classified as FDA pregnancy category C. Local anesthetics readily cross the placenta. Fetal or neonatal poisoning may occur as a result of spinal or regional nerve blocks, systemic maternal poisoning, or inadvertent intracranial injection of local anesthetics into the fetus during labor and delivery.

Laboratory Monitoring

    A) Methemoglobin concentrations should be checked in cyanotic patients.
    B) Obtain arterial blood gas in cases of respiratory depression, hypotension, or persistent cardiac dysrhythmias.
    C) Monitor vital signs and electrolytes.
    D) Obtain an ECG and institute continuous cardiac monitoring in symptomatic patients.

Treatment Overview

    0.4.2) ORAL/PARENTERAL EXPOSURE
    A) MANAGEMENT OF MILD TO MODERATE TOXICITY
    1) Supportive care is the mainstay of treatment in mild to moderate toxicity. Paresthesias and paralysis resolve in virtually all cases. Anxiety can be managed with benzodiazepines. Patients with significant oropharyngeal anesthesia should be positioned upright with nothing by mouth (NPO) to avoid aspiration.
    B) MANAGEMENT OF SEVERE TOXICITY
    1) Patients with severe toxicity (eg, depressed mental status, seizures, hypotension, dysrhythmias, cardiac arrest) should receive standard supportive care (intubation and mechanical ventilation, intravenous fluids and pressors for hypotension, benzodiazepines for seizures). Consider intravenous lipid therapy early for patients with ventricular dysrhythmias or hypotension.
    C) DECONTAMINATION
    1) PREHOSPITAL: No prehospital gastrointestinal decontamination is appropriate. Symptomatic patients should be placed on oxygen and transported to a health care facility.
    2) HOSPITAL: Activated charcoal and gastric lavage are discouraged due to the risk of aspiration in cases when oropharyngeal anesthesia is present. There is no role for whole bowel irrigation.
    D) AIRWAY MANAGEMENT
    1) Endotracheal intubation and mechanical ventilation may be necessary in cases of high spinal, epidural, or intrathecal administration or cases with significant cardiac or neurologic toxicity. Patients should remain intubated until they are able to pass a spontaneous breathing trial with an adequate tidal volume.
    E) ANTIDOTE
    1) METHEMOGLOBINEMIA: Initiate oxygen therapy. Treat with methylene blue if patient is symptomatic (usually at methemoglobin concentrations greater than 20% to 30% or at lower concentrations in patients with anemia, underlying pulmonary or cardiovascular disease). METHYLENE BLUE: INITIAL DOSE/ADULT OR CHILD: 1 mg/kg IV over 5 to 30 minutes; a repeat dose of up to 1 mg/kg may be given 1 hour after the first dose if methemoglobin levels remain greater than 30% or if signs and symptoms persist. NOTE: Methylene blue is available as follows: 50 mg/10 mL (5 mg/mL or 0.5% solution) single-dose ampules and 10 mg/1 mL (1% solution) vials. Additional doses may sometimes be required. Improvement is usually noted shortly after administration if diagnosis is correct. Consider other diagnoses or treatment options if no improvement has been observed after several doses. If intravenous access cannot be established, methylene blue may also be given by intraosseous infusion. Methylene blue should not be given by subcutaneous or intrathecal injection. NEONATES: DOSE: 0.3 to 1 mg/kg.
    F) FAT EMULSION
    1) Patients who develop significant CNS (eg, agitation, confusion, seizures, mental status depression) or cardiovascular toxicity should be treated with intravenous lipids. Administer 1.5 mL/kg of 20% lipid emulsion over 2 to 3 minutes as an IV bolus, followed by an infusion of 0.25 mL/kg/min. Evaluate the patient's response after 3 minutes at this infusion rate. The infusion rate may be decreased to 0.025 mL/kg/min (ie, 1/10 the initial rate) in patients with a significant response. This recommendation has been proposed because of possible adverse effects from very high cumulative rates of lipid infusion. Monitor blood pressure, heart rate, and other hemodynamic parameters every 15 minutes during the infusion. If there is an initial response to the bolus followed by the re-emergence of hemodynamic instability during the lowest-dose infusion, the infusion rate may be increased back to 0.25 mL/kg/min or, in severe cases, the bolus could be repeated. A maximum dose of 10 mL/kg has been recommended by some sources. Where possible, lipid resuscitation therapy should be terminated after 1 hour or less, if the patient's clinical status permits. In cases where the patient's stability is dependent on continued lipid infusion, longer treatment may be appropriate.
    G) CONDUCTION DISORDER OF THE HEART
    1) QRS widening of more than 120 msec should be treated with intravenous lipids as above and sodium bicarbonate (1 to 2 mEq/kg) boluses, starting with one ampule (50 mEq) in adults until narrowing is achieved. Monitor acid/base status and keep pH below 7.55.
    H) CARDIAC ARREST
    1) Initiate CPR immediately and administer intravenous lipids as above. Good neurological outcomes have been reported after prolonged CPR. ACLS algorithms should be applied in conjunction with lipid emulsion administration with the following modifications: if epinephrine is used, small initial doses (10 mcg to 100 mcg boluses) are recommended. Avoid vasopressin, lidocaine, calcium channel blockers and beta blockers. Cardiac bypass should be considered if unresponsive to the above therapies.
    I) SEIZURE
    1) Seizures should be treated with benzodiazepines (in addition to intravenous lipids) as first line therapy followed by phenobarbital or propofol. Diazepam (ADULT: 5 to 10 mg, repeat every 10 to 15 min as needed; CHILD: 0.2 to 0.5 mg/kg, repeat every 5 min as needed) or lorazepam (ADULT: 2 to 4 mg, repeat every 10 to 15 min as needed; CHILD: 0.05 to 0.1 mg/kg, repeat every 5 min as needed) may be given.
    J) HYPOTENSIVE EPISODE
    1) Hypotension should be treated with fluids initially, intravenous lipids, and direct acting vasopressors, such as norepinephrine, phenylephrine, or epinephrine if necessary. Bradycardic patients should be given atropine (ADULT: 1 mg; CHILD: 0.02 mg/kg, minimum 0.1 mg, maximum 1 mg) or epinephrine.
    K) NAUSEA AND VOMITING
    1) Nausea and vomiting should be treated with antiemetics, either metoclopramide (ADULT: 10 to 20 mg every 6 hours as needed; CHILD: 0.1 to 0.2 mg/kg every 6 hours as needed) or ondansetron (ADULT: 4 to 8 mg every 6 hours as needed; CHILD: 0.15 mg/kg every 4 hours as needed) are reasonable choices.
    L) ENHANCED ELIMINATION
    1) No method of enhanced elimination has proven beneficial in local anesthetic toxicity.
    M) PATIENT DISPOSITION
    1) HOME CRITERIA: Lidocaine doses under 6 mg/kg are unlikely to cause serious adverse effects and can be managed at home. Patients with more than mild symptoms should be referred to a health care facility.
    2) OBSERVATION CRITERIA: Patients should be observed for 8 hours following methylene blue administration to rule out recurrence of methemoglobinemia. Patients with tachycardia only can be observed until this symptom resolves; other cardiac toxicities should be admitted.
    3) ADMISSION CRITERIA: Patients with persistent cardiac dysrhythmias, mental status changes, seizures, respiratory failure, or recurrence of methemoglobinemia, despite treatment with methylene blue, should be admitted. Patients with respiratory, CNS, or cardiac toxicity should be admitted to an ICU.
    4) CONSULT CRITERIA: A medical toxicologist or poison control center should be consulted in cases that involve cardiac or CNS toxicity or clinically significant methemoglobinemia.
    N) PITFALLS
    1) Failure to consider cardiac toxicity when CNS toxicity (eg, seizures, CNS depression) is present. Failure to inflate a blood pressure cuff proximal to an infiltration site when significant toxicity is present may lead to continued systemic exposure. Failure to consider excess local anesthetic exposure in cyanotic infants. Inadvertent intravascular administration of a dose intended for tissue infiltration or nerve block can cause systemic toxicity. Therapeutic use of topical anesthetic sprays at or near the normal dose can cause methemoglobinemia in susceptible individuals.
    O) PHARMACOKINETICS
    1) These agents are well absorbed across the skin and gastrointestinal mucosa. The volume of distributions are generally large, with a moderate to high degree of protein binding. They are primarily metabolized by plasma pseudocholinesterase; lidocaine is hepatically metabolized.
    P) TOXICOKINETICS
    1) Intravenous administration can lead to rapid systemic toxicity and the duration of effects are dependent upon the individual agent's duration of action (eg, bupivacaine lasting longer than lidocaine). Local administration rarely leads to severe cardiac or neurologic toxicity. Methemoglobinemia has been reported following injection, ingestion, and dermal application.
    Q) DIFFERENTIAL DIAGNOSIS
    1) Sodium channel blockers (ie, tricyclic antidepressants, cocaine, quinidine, and propranolol) should be considered in cases of wide QRS cardiac toxicity or seizures. Other agents that cause methemoglobinemia (ie, dapsone, phenazopyridine) should be considered.
    0.4.4) EYE EXPOSURE
    A) DECONTAMINATION: Remove contact lenses and irrigate exposed eyes with copious amounts of room temperature 0.9% saline or water for at least 15 minutes. If irritation, pain, swelling, lacrimation, or photophobia persist after 15 minutes of irrigation, the patient should be seen in a healthcare facility.
    0.4.5) DERMAL EXPOSURE
    A) OVERVIEW
    1) DECONTAMINATION
    a) Wash exposed skin with soap and water.
    0.4.6) PARENTERAL EXPOSURE
    A) INTRAVENOUS INJECTION
    1) Intravascular administration, either inadvertent or during regional anesthetic techniques, such as Bier blocks, can lead to systemic toxicity. Inflation of a blood pressure cuff proximal to the site of injection may limit the systemic effects. The cuff can intermittently be released for short durations when systemic toxicity resolves, limiting the patient from prolonged systemic exposure.
    2) INTRAVENOUS INFUSION: Stop drug infusion at first sign of toxicity.
    B) INTRATHECAL INJECTION
    1) Inadvertent intrathecal injection has been reported with local anesthetics. Endotracheal intubation and mechanical ventilation are likely to be necessary because of diaphragmatic paralysis. Drainage of cerebrospinal fluid may accelerate recovery. The following recommendations are based on experience with antineoplastic drugs. Keep the patient upright if possible. Immediately drain at least 20 mL CSF; drainage of up to 70 mL has been tolerated in adults. In severe overdoses, this may be followed with CSF exchange (remove serial 20 mL aliquots CSF and replace with equivalent volumes of warmed, preservative free normal saline or lactated ringers).

Range Of Toxicity

    A) Toxicity varies by agent. LIDOCAINE:ADULT: IV injection of 800 to 2000 mg in adults has caused seizures followed by cardiac arrest. PEDIATRIC: In children, ingestion of 5 to 25 mL 2% viscous lidocaine has caused seizures. BENZOCAINE: Therapeutic doses of topical spray may cause methemoglobinemia. DIBUCAINE: PEDIATRIC: Two toddlers developed generalized seizures and severe dysrhythmias, and subsequently died after ingesting 15 mg/kg to 19 mg/kg of 1% dibucaine ointment. An 18-month-old child became comatose and developed generalized tonic-clonic seizures, but survived, after ingesting 12 mg/kg of 1% dibucaine ointment. PRAMOXINE: Ingestions up to 150 mg in children (11 mg/kg) and 250 mg in adults caused minor toxicity.
    B) THERAPEUTIC DOSE: Varies by agent. LIDOCAINE: ADULT: A single dose limit is 2 mg/kg for IV doses. PEDIATRIC: For viscous lidocaine 2% in children 3 years and older, the recommended dose is approximately 3 to 5 mg/kg swish and spit (for the mouth) or swish and swallow (for the pharynx) every 3 hours as needed, up to a MAX of 8 doses/day. In infants and children less than 3 years of age, 1.25 mL is applied topically to the area every 3 hours as needed, up to a MAX of 8 doses/day. BENZOCAINE: For treatment of mouth ulcers, benzocaine may be applied topically to the affected areas up to 4 times daily. PRAMOXINE: For hemorrhoids, apply 1% ointment or cream rectally or topically to anorectal area up to 5 times daily. ESTERS: Maximum subQ doses are: chloroprocaine 10 mg/kg; procaine 10 mg/kg; tetracaine 3 mg/kg. AMIDES: Maximum subQ doses are: bupivacaine 2 mg/kg; etidocaine 4 mg/kg; lidocaine 5 mg/kg; mepivacaine 5 mg/kg; prilocaine 8 mg/kg; ropivacaine 3 mg/kg.

Summary Of Exposure

    A) USES: Local anesthetics are used for analgesia. They are administered topically and by local injection, regional injection, and for spinal and epidural anesthesia.
    B) PHARMACOLOGY: These agents reversibly block sodium channels and block the action potentials responsible for nerve conduction.
    C) TOXICOLOGY: Toxicity is caused by sodium channel blockade leading to CNS and cardiac effects. Some of these agents and their metabolites can oxidize iron leading to methemoglobinemia.
    D) EPIDEMIOLOGY: Toxicity is uncommon and severe toxicity is rare.
    E) WITH THERAPEUTIC USE
    1) Allergic reactions to the ester or amide classes of local anesthetics are possible though extremely rare. The ester class (benzocaine, chloroprocaine, cocaine, cyclomethycaine, dimethocaine, larocaine, propoxycaine, procaine, novocaine, proparacaine, tetracaine, and amethocaine) accounts for the majority of true IgE-mediated allergic reactions due to metabolism of PABA, a known allergen. Reactions to the amide class (articaine, bupivacaine, carticaine, cinchocaine, dibucaine, etidocaine, lidocaine, mepivacaine, piperocaine, prilocaine, ropivacaine, and trimecaine) are often secondary to the methylparaben preservative in the multidose vials. Clinically insignificant methemoglobinemia may result from exposure to many local anesthetics. Patients may develop vasovagal reactions following administration of these agents. Visual loss has rarely been associated with intranasal bupivacaine use during nasal surgery. Visual symptoms may persist for years.
    F) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: Subjective effects in mild toxicity include drowsiness, impending doom, headache, dizziness, paresthesias, euphoria, numbness of the mouth, lightheadedness, tinnitus, anxiety, confusion, tremors, agitation, disorientation, hallucinations, and lethargy. Peripheral effects may result in temporary paralysis. Suppression of the gag reflex may occur with oropharyngeal exposure. Methemoglobinemia can develop in patients who have no other evidence of toxicity.
    2) SEVERE TOXICITY: Severe toxicity may cause cardiac, respiratory, and CNS toxicity, as well as methemoglobinemia. CNS effects precede significant cardiovascular toxicity, except following massive IV injection. While there is often a progression of symptoms after IM, subQ, or continuous IV infusion, seizures and coma may occur suddenly after rapid IV administration. Cardiac effects are due to sodium channel blockade and can be manifested by a range of toxic effects including hypotension, AV block, bradycardia, QRS prolongation, ventricular dysrhythmias, asystole, and cardiovascular collapse. Spinal, epidural, or inadvertent intrathecal administration may lead to respiratory failure. Hypoxia may be secondary to cardiac toxicity or direct motor paralysis of respiratory muscles leading to respiratory depression, apnea, or respiratory arrest. Methemoglobinemia is well described with usage of prilocaine and benzocaine, and rarely reported with lidocaine, tetracaine, Cetacaine, and propitocaine. It can develop after therapeutic doses and in patients who have no other evidence of toxicity.
    3) INHALATION EXPOSURE: Methemoglobinemia can rarely result from inhalation exposure to local anesthetics.
    4) DERMAL EXPOSURE: Topical preparations can lead to systemic toxicity ranging from mild to severe. Infants are at a higher risk for systemic effects from dermal exposure.

Vital Signs

    3.3.2) RESPIRATIONS
    A) Respiratory depression and apnea may occur in patients with coma or seizures (Selden & Burke, 1988). It may also result from the cephalad spread of caudal or epidural anesthesia (Lumb & Carli, 1989). Tachypnea may occur in the early stages of poisoning.
    3.3.4) BLOOD PRESSURE
    A) Hypotension may occur in patients with bradycardia or cardiac arrest. It can also occur as a direct effect following intravenous injection (Yukioka et al, 1990) or as a result of sympathetic blockade following epidural or spinal anesthesia.
    3.3.5) PULSE
    A) The pulse is slow or absent with advanced toxicity. It may be increased early in the course of poisoning ("excitatory" phase).

Heent

    3.4.3) EYES
    A) The pupils are usually normal unless seizures or cardiac arrest have developed. Miosis, diplopia, and blurred vision have been reported.
    B) EPITHELIAL DAMAGE/TETRACAINE: Chronic repeated topical use of tetracaine ophthalmic ointment (0.5%) may result in toxic effects to the corneal epithelium (Duffin & Olson, 1984).
    1) Two cases of crystalline keratopathy occurred following abuse of topical ocular anesthetics (proparacaine hydrochloride) (Kintner et al, 1990).
    2) Ring keratitis corneal edema, Descemet's membrane folds, and a subtotal corneal epithelial defect have been described after abuse of topical ocular anesthetics (Webber et al, 1999).
    3) CASE SERIES: Toxic keratopathy was reported in 8 patients following direct application of topical anesthetics to the eye over a period of several days. Prior to the application of the anesthetic, 5 patients complained of a corneal foreign body, 1 patient was diagnosed with basal membrane dystrophy, 1 patient received a chemical burn to the eye, and 1 patient was exposed to ultraviolet radiation. The keratopathy occurred in the right eye of 2 patients, in the left eye of 4 patients, and bilaterally in 2 patients, with an initial best corrected visual acuity of "counting fingers" in the majority of patients. Visual acuity improved with discontinuation of the anesthetic and supportive treatment, although penetrating keratoplasties were performed in 2 patients, and 6 patients developed corneal leukomas (Yeniad et al, 2010).
    C) MARCAINE: VISUAL LOSS occurred in a 37-year-old woman after she was given intranasal marcaine and adrenaline prior to nasal surgery. She also received general anesthesia and cocaine pledgets in both nostrils. Three other similar cases are described; in all cases, a local anesthetic was injected intranasally. Most of these patients' visual abnormalities were still persisting years later (Savino et al, 1990).
    D) EMLA: CORNEAL ABRASIONS and conjunctivitis were reported in 2 patients following topical application of EMLA cream (consisting of lidocaine 25 mg/mL, prilocaine 25 mg/mL, and carbopol 934) to the face. The patients completely recovered within 4 days after topical administration of antibiotics and steroids (Eaglstein, 1999).
    E) LIDOCAINE: TEMPORARY BLINDNESS developed in a 21-year-old woman undergoing intravenous regional anesthesia (Bier Block) for hand surgery when 30 mL of 2% lidocaine (600 mg or 12 mg/kg body weight) was injected into the dorsum of the hand in error due to a misreading of the medication vial (Sawyer & vonSchroeder, 2002). The patient developed immediate tachycardia, seizurelike activity, and loss of consciousness after the tourniquet was inadvertently completely deflated. After regaining consciousness, the patient complained of loss of visual acuity lasting 5 to 10 minutes. Thereafter her visual acuity, visual fields, and extraocular eye movements were entirely normal.
    3.4.4) EARS
    A) Local anesthetics have been used to treat tinnitus (Bysis, 1985). At toxic doses, they cause tinnitus (Bazerbachi et al, 2014; Cooper & May, 2009; Lankin et al, 1983)
    3.4.6) THROAT
    A) PRAMOXINE: In a retrospective series of 177 cases of ingestion of pramoxine-containing products called to a poison center, mouth irritation developed in 5 patients and oral numbness in 3 (Spiller et al, 2006). Oral numbness and irritation were short lived.

Cardiovascular

    3.5.2) CLINICAL EFFECTS
    A) CONDUCTION DISORDER OF THE HEART
    1) SUMMARY: A transient period of tachycardia and hypertension may precede bradycardia, hypotension, and asystole (Selden & Burke, 1988; Amitai et al, 1986; Applebaum & Halperin, 1986; Altman et al, 1985; Brown & Skiendzielowski, 1980; Marriott & Philips, 1974) . Myocardial contractility, vascular resistance, and cardiac output are depressed; stroke volume and ejection fraction may be reduced (Scott et al, 1989).
    2) COMPARATIVE TOXICITY: Local anesthetics vary in cardiotoxic potency. Bupivacaine depresses conduction and contractility at lower doses than ropivacaine in human volunteers (Scott et al, 1989), and was 2 to 4 times more potent than lidocaine in inducing adverse cardiovascular effects in an animal study (Thomas et al, 1986).
    3) RISK FACTORS: The fetus and neonate may be more susceptible. Heart block occurs more readily in patients with concomitant therapy (calcium channel blockers, beta blockers). Ventricular dysrhythmias occur particularly following seizures (Lee et al, 1999; Abouleish et al, 1998; Yan & Newman, 1998) and are most common in patients with preexisting cardiovascular disorders, such as myocardial infarction (Reynolds, 1987). Patients with underlying conduction abnormalities may develop asystole following therapeutic doses of lidocaine (Applebaum & Halperin, 1986; Grenadier et al, 1981).
    4) CASE REPORTS - PEDIATRIC
    a) BUPIVACAINE: Ventricular tachycardia was reported in a pediatric patient 10 hours after starting a continuous epidural infusion of 0.25% bupivacaine (2.5 mg/kg/hr). Serum bupivacaine level was 5.6 mcg/mL (McCloskey et al, 1992).
    b) BUPIVACAINE: Ventricular tachycardia occurred in an 11-month-old, 8 kg infant who received 8 mL (1 mL/kg) of 0.5% bupivacaine prior to undergoing a circumcision. Following administration of 20% intralipid at an IV bolus dose of 12 mL, followed by an infusion at a dose of 2 mL/min (0.25 mL/kg/min), the patient recovered uneventfully (Buck et al, 2014).
    c) PRAMOXINE: A 4-year-old child experienced frequent premature supraventricular complexes several hours after ingesting 100 mL of a lotion containing pramoxine (Stancavage et al, 1995).
    d) LIDOCAINE: Supraventricular tachycardia was reported in a 2-year-old after apparently aspirating and swallowing a small amount of 2% viscous lidocaine (Garrettson & McGee, 1992).
    e) DIBUCAINE: Two toddlers (aged 18 months and 21 months) developed generalized tonic-clonic seizures and severe dysrhythmias (bradycardia, ventricular tachycardia, ventricular fibrillation, asystole) following ingestions (15 mg/kg to 19 mg/kg) of 1% dibucaine ointment. Despite aggressive supportive measures, both children died within hours after ingestion (Dayan et al, 1996).
    5) CASE REPORTS - ADULT
    a) LIDOCAINE: Sinus tachycardia and hypotension occurred (with tourniquet deflation) in a 28-year-old pregnant woman following intravenous regional anesthesia administration of lignocaine for hand surgery. The patient recovered following supportive care (Coleman & Kelly, 1998). In a similar case, a 35-year-old pregnant woman, undergoing hand surgery using a tourniquet technique, also developed sinus tachycardia and hypotension following intravenous regional anesthesia administration of lidocaine and after deflation of the tourniquet (Cooper & May, 2009).
    b) LIDOCAINE: A 21-year-old man developed hypotension and severe bradycardia that rapidly progressed to asystole after gargling 20 mL of a 4% lidocaine solution (containing 800 mg of lidocaine) in preparation for an upper gastrointestinal endoscopy. Despite aggressive resuscitative measures, the patient subsequently died (Zuberi et al, 2000). Although the patient was advised not to swallow the solution, it is believed that some of the solution did enter the gastrointestinal tract, resulting in lidocaine toxicity.
    c) MEPIVACAINE: Severe bradycardia (10 to 15 bpm), requiring implantation of a pacemaker, was reported in a 30-year-old woman, with a history of AV block with a mild mitral prolapse, who received 400 mg of mepivacaine paracervically during an in-vitro fertilization procedure. The patient recovered following the pacemaker implantation (Ayestaran et al, 2000).
    d) LIDOCAINE: Cardiac arrest occurred after an adult woman was given a lidocaine block followed by a bupivacaine block in the axillary space to facilitate hand surgery (Long et al, 1989).
    e) LIDOCAINE: Toxicity was seen in a 22-year-old man during daily use of 240 mL viscous lidocaine for a severe tongue ulcer. His serum lidocaine concentration was 6.7 mcg/mL (Yamashita et al, 2002).
    f) ROPIVACAINE: A 25-year-old man developed isolated sinus tachycardia (130 beast per minute) and tonic-clonic seizures approximately 1 minute following unintentional intravascular administration of 10 mL (75 mg) of ropivacaine for a sciatic nerve block. The patient recovered following supportive therapy (Petitjeans et al, 2002). Plasma ropivacaine concentrations, obtained 15, 90, and 150 minutes after onset of symptoms, were 13.1, 11.6, and 3.6 mcmol/L, respectively.
    g) LIDOCAINE: A 57-year-old man with progressive heart failure developed ventricular tachycardia that recurred despite treatment with amiodarone and cardioversion. He was administered IV lidocaine bolus, as well as overdrive pacing, and cardioversion, followed by a continuous lidocaine infusion, at an initial dose of 1 mg/min increasing to 2 mg/min due to recurrent episodes of ventricular tachycardia. The patient developed mental status changes and tremor. A lidocaine level, obtained due to suspected lidocaine toxicity, was 7.6 mcg/mL (therapeutic range 2 to 5 mcg/mL), and his lidocaine infusion was discontinued. Over the next several hours, the patient became increasingly somnolent and confused, and an ECG showed normal sinus rhythm with progressive QRS widening, with subsequent cardiac arrest. Aggressive resuscitative measures were initiated without significant clinical improvement in the patient. Approximately 55 minutes after beginning resuscitation, 20% lipid emulsion therapy was started, with a return of normal sinus rhythm and narrowing of the QRS complex 5 minutes later. Repeat lidocaine levels, obtained 25 minutes and 55 minutes after the lipid infusion, were 3.5 mcg/mL and 3 mcg/mL, respectively (Dix et al, 2011).
    h) BUPIVACAINE: A 37-year-old man, undergoing shoulder surgery, developed bradycardia, seizures, and cardiac arrest immediately after receiving bupivacaine as a nerve block. Despite undergoing cardiopulmonary resuscitation for approximately 2 hours and being placed on cardiopulmonary bypass, the patient died approximately 7 hours post-injection. An autopsy revealed several cardiac abnormalities, including cardiomegaly, myocardial bridging, and lipomatous hypertrophy of the intra-atrial septum. Rash and laryngeal edema were also present. Post-mortem toxicology results revealed cardiac blood bupivacaine concentration of 1.1 mcg/mL and an elevated total serum tryptase concentration of 45.1 ng/mL in cardiac blood (reference range, less than 11.4 ng/mL). The abrupt onset of seizures and the elevated bupivacaine concentration suggest inadvertent intravascular injection instead of administration as a peripheral nerve block, although the elevated tryptase concentration and the presence of rash and laryngeal edema also suggest the possibility of bupivacaine hypersensitivity as a possible contributing factor (Dudley et al, 2011).
    B) ELECTROCARDIOGRAM ABNORMAL
    1) WITH POISONING/EXPOSURE
    a) DIBUCAINE: An 18-month-old infant was unresponsive and developed cyanosis and intermittent generalized tonic-clonic seizures approximately 30 minutes after ingesting approximately 150 mg (12.5 mg/kg) of a sunburn ointment containing 1% dibucaine. An ECG revealed a wide-complex rhythm with frequent paroxysmal ventricular contractions. A repeat ECG approximately 1 hour postingestion demonstrated AV blockade with a prolonged QRS interval (200 msec) and a QTc interval of 513 msec. With supportive care, including sodium bicarbonate IV boluses and IV magnesium sulfate, the patient's cardiac status normalized with narrowing of the QRS complex, and subsequently a complete recovery without sequelae (Nelsen et al, 2009).
    b) CASE REPORT/LIDOCAINE: A 15-year-old girl developed seizures after ingesting up to 2 grams (49 mg/kg) of viscous lidocaine. She was tachycardic (160 bpm) with a Glasgow Coma Scale of 3, and fixed and dilated pupils. An ECG revealed a QRS of 110 ms, that narrowed to 80 ms within 80 minutes after administration of IV fluids and IV sodium bicarbonate, and an arterial blood gas analysis indicated severe metabolic acidosis. A serum lidocaine concentration at 2 hours post-ingestion was 16.6 mcg/mL (reference range 1.2 to 5 mcg/mL) that decreased to 0.7 mcg/mL 6 hours post-ingestion. There was no evidence of epileptiform activity on an EEG, and a brain MRI showed hypoxic ischemic injury approximately 40 hours post-ingestion. The patient was apneic with no brainstem reflexes on hospital day 9, and brain death was subsequently declared (Drapkin et al, 2015).
    c) ROPIVACAINE: A 5-week-old preterm infant, born at gestational age of 36 weeks and scheduled for inguinal hernia repair, inadvertently received ropivacaine at a dose of 10 mg/kg, injected into the caudal space. The total ropivacaine dose received was 40 mg instead of the prescribed 8 mg. Within minutes, the patient developed bradycardia (50 bpm), widening of QRS complexes, elevated T waves, and hypotension (50/30 mmHg). With external chest compressions, administration of IV fluids and epinephrine, and controlled ventilation and sedation, the patient recovered and was discharged home 2 days later without neurologic sequelae (Hubler et al, 2010).
    2) ECG manifestations include sinus bradycardia, atrioventricular block, increased PR and QRS intervals, and asystole (Ruetsch et al, 1999; Jonville et al, 1990; Altman et al, 1985; Antonelli & Bloch, 1982; Gupta et al, 1976; Kunkel et al, 1974) .
    3) CASE REPORT/LIDOCAINE: An 87-year-old man suffered cardiac arrest after intraurethral administration of lidocaine for cystourethroscopy (Chang et al, 2005).
    4) CASE REPORT/ROPIVACAINE: A 57-year-old man, with liver dysfunction and end-stage renal disease, was admitted for a below-knee amputation and received a ropivacaine infusion (total dose 1,540 mg) postoperatively. He later developed lethargy, bradycardia, tinnitus, dysgeusia, and hallucinations. An ECG revealed widened QRS interval and prolonged QT interval without ST changes, and an echocardiogram showed an ejection fraction of 20% without wall motion abnormality. The patient's troponin-I-ES level peaked at 4.3. The patient minimally responded to administration of 2,480 mL of 20% intralipid and one session of plasmapheresis. He subsequently died following cardiac arrest (Bazerbachi et al, 2014).
    C) HYPOTENSIVE EPISODE
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT/ROPIVACAINE: Hypotension, refractory to IV ephedrine, occurred in a 26-year-old pregnant woman who inadvertently received epidural administration of ropivacaine, 111 mL/hour instead of 11 mL/hour (a 10-fold dosing error), during labor. The total dose of ropivacaine received during a 38-minute period was 140 mg. With supportive treatment including an infusion of phenylephrine, the patient recovered and, approximately 2 hours later, vaginally delivered a male infant with Apgar scores of 9 at 1 minute and at 5 minutes (Thyen et al, 2010).
    2) SUMMARY: Hypotension and cardiovascular collapse may occur secondary to sympathetic blockade during spinal or epidural use, depression of medullary vasomotor center, hypoxia and acidosis secondary to CNS toxicity, depressed cardiac contractility with low cardiac output, or vasodilation (Reynolds, 1987).
    3) CASE REPORTS - PEDIATRIC
    a) LIDOCAINE: Hypotension was reported in a 2-year-old approximately 24 hours after apparently aspirating and swallowing a small amount of 2% viscous lidocaine (Garrettson & McGee, 1992).
    4) CASE REPORTS - ADULT
    a) BUPIVACAINE: Following the accidental administration of 337.5 mg bupivacaine and 180 mcg sufentanil epidurally in less than 30 minutes, a 62-year-old patient suffered severe hypotension and moderate bradycardia. No dysrhythmias occurred and the patient recovered (Wolff et al, 1992).
    b) LIGNOCAINE: A pregnant 28-year-old patient received intravenous regional anesthesia (tourniquet technique) using lignocaine for hand surgery. Surgery was uneventful and, 45 minutes later, the tourniquet was deflated. Sixty seconds later, the patient developed hypotension and sinus tachycardia. Effects resolved following supportive care (Coleman & Kelly, 1998).
    c) LIDOCAINE: Hypotension was reported in a 21-year-old man after gargling with 20 mL of a 4% lidocaine solution (containing 800 mg lidocaine) in preparation for an upper gastrointestinal endoscopy (Zuberi et al, 2000).
    d) LIDOCAINE: A pregnant 35-year-old woman (at 25 weeks gestation) received intravenous regional anesthesia with lidocaine for hand surgery. Prior to lidocaine administration, a tourniquet was applied to the upper arm and inflated. Following lidocaine administration, the tourniquet was deflated in phases. Approximately 10 minutes following the final deflation, the patient developed hypotension, sinus tachycardia, tinnitus, and a metallic taste in her mouth. An ultrasound indicated that the fetal heart rate was normal. With supportive care, the patient recovered (Cooper & May, 2009).
    D) LEFT VENTRICULAR CARDIAC DYSFUNCTION
    1) CASE REPORT/BUPIVACAINE: A 22-year-old healthy woman developed hypotension (80/40 mmHg), chest pain, dyspnea, and dizziness approximately 15 minutes after receiving bupivacaine as a spinal anesthetic. An ECG demonstrated ST-segment depression on all leads, and rales were audible in both lungs. Vital signs, taken following transfer to the emergency department, showed continued hypotension (80/36 mmHg), tachycardia (119 beats per minute), and tachypnea (24 breaths per minute). Laboratory data revealed a creatinine kinase-MB level of 15.5 ng/mL (normal < 6.3 ng/mL) and a troponin I level of 2.33 ng/mL (normal < 0.2 ng/mL), a chest x-ray showed bilateral infiltrates consistent with pulmonary edema, and an echocardiogram revealed reduced myocardial contractility and severe hypokinesia of the left ventricle with a decreased ejection fraction of 27%. On hospital day 7, the patient spontaneously recovered with a normalization of her cardiac biomarkers. A coronary angiography revealed normal coronary arteries, and a repeat echocardiogram showed normal LV systolic function with an ejection fraction of 71% (Ryu et al, 2007).
    3.5.3) ANIMAL EFFECTS
    A) ANIMAL STUDIES
    1) HEART DISORDER
    a) BUPIVACAINE/LIDOCAINE: In a rat model, bupivacaine was 2 to 4 times more potent in inducing adverse cardiovascular effects than lidocaine (Thomas et al, 1986).

Respiratory

    3.6.2) CLINICAL EFFECTS
    A) ACUTE LUNG INJURY
    1) SUMMARY: Adult respiratory distress syndrome (ARDS) has been reported in patients with aspiration or during hypersensitivity reactions (Donohue, 1983; Promisloff & Dupont, 1983; Woelke & Tucker, 1983; Howard et al, 1982) .
    2) CASE REPORT - PEDIATRIC
    a) LIDOCAINE/INFANT: ARDS developed in a 2-year-old 2 days after apparently aspirating a small amount of 2% viscous lidocaine (Garrettson & McGee, 1992).
    3) CASE REPORTS - ADULT
    a) LIDOCAINE: Adult respiratory syndrome was reported following administration of less than 30 mL of 1% lidocaine solution for topical anesthesia prior to fiberoptic bronchoscopy (Promisloff & Dupont, 1983).
    b) LIDOCAINE: Respiratory distress was reported in a 21-year-old man after gargling with 20 mL of a 4% lidocaine solution (containing 800 mg of lidocaine) in preparation for an upper gastrointestinal endoscopy (Zuberi et al, 2000).
    B) HYPOVENTILATION
    1) NEONATAL TOXICITY: may include apnea and respiratory depression or arrest (Bozynski et al, 1987; Dodson, 1976; Jonville et al, 1990). Apnea may accompany coma, seizures, or asystole.
    2) BUPIVACAINE/CASE REPORT (ADULT): Following the accidental administration of 337.5 mg bupivacaine and 180 mcg sufentanil epidurally in less than 30 minutes, a 62-year-old patient suffered only mild respiratory depression (12 breaths/min) despite the high dose (Wolff et al, 1992).
    3) MEPIVACAINE/CASE REPORT (ADULT): Severe bradypnea and bradycardia was reported in a 30-year-old woman with a history of AV block who received mepivacaine paracervically during an in-vitro fertilization procedure. The patient recovered following implantation of a pacemaker (Ayestaran et al, 2000).
    C) HYPOREFLEXIA
    1) The cough and gag reflex may be impaired, even with parenteral administration (Smith & Kundahl, 1973), thereby facilitating development of aspiration and aggravating chronic obstructive pulmonary disease.
    D) BRONCHOSPASM
    1) LIDOCAINE/COHORT STUDY: Bronchoconstriction was induced in 5 of 20 (25%) confirmed asthmatic patients challenged with lidocaine (6 mL 4% Xylocaine topically). They could not relate this response to airway histamine responsiveness or to the preservative present in the Xylocaine (McAlpine & Thomson, 1989).
    E) APNEA
    1) BUPIVACAINE/CASE REPORT: A 28-year-old woman developed respiratory arrest, requiring intubation, within minutes of receiving an accidental spinal injection (intended for the paracervical area) of a bupivacaine-triamcinolone mixture. The patient gradually recovered and was extubated approximately 24 hours later (Wang & Chang, 1993).
    2) LIDOCAINE/CASE REPORT: A 47-year-old woman developed left upper and lower extremity weakness and respiratory depression after an inadvertent intrathecal injection of lidocaine during an attempted trigger point injection of the trapezius (Nelson & Hoffman, 1995).

Neurologic

    3.7.2) CLINICAL EFFECTS
    A) SEIZURE
    1) SUMMARY: These agents are anticonvulsants at low doses and convulsants at high doses (Schuster et al, 1987). Suppressed cortical patterns are found on the EEG. Myoclonic, grand mal, and tonic-clonic seizures have been reported. Marked CNS depression with coma may occur.
    2) ROUTE OF ADMINISTRATION SUMMARY: Seizures have been reported after intravenous, intramuscular (Malone et al, 1988), intercostal and epidural (Moore et al, 1982), subcutaneous (Alfano et al, 1984), topical application to burn patients or those with multiple erosive skin lesions (Lie et al, 1990; Wehner & Hamilton, 1984) , intraurethral (Sundaram, 1987), intraocular (Cydulka & Betzelos, 1990), intrapleural (Agarwal et al, 1992), intravesically (Clapp et al, 1999), stellate ganglion block (Wulf et al, 1991), and oral administration.
    3) ORAL ADMINISTRATION
    a) ORAL ADMINISTRATION: Oral ingestion of 5 to 30 mL of a 2% to 4% viscous Xylocaine (lidocaine) solution has resulted in seizures in children (Smith et al, 1992; Hess & Walson, 1988; Mofenson et al, 1983)Bowman et al, 1982; (Rothstein et al, 1982; Sakai & Lattin, 1980) . Seizures are most common following repeated ingestion (instead of mucous application) of recommended or excessive amounts of the viscous Xylocaine preparations (Smith et al, 1992; Rothstein et al, 1982) .
    1) Seizures have been reported following ingestion of Xylocaine(R) viscous when the solution was intended for topical use only (Giard et al, 1983; Mofenson et al, 1983; Fruncillo et al, 1982; Rothstein et al, 1982) .
    2) CASE REPORTS - PEDIATRICS
    a) LIDOCAINE: Seizures were reported in a 2-year-old within 10 to 15 seconds of apparently aspirating and swallowing a small amount of 2% viscous lidocaine (Garrettson & McGee, 1992).
    b) LIDOCAINE: A 15-year-old girl developed seizures after ingesting up to 2 grams (49 mg/kg) of viscous lidocaine. She was tachycardic (160 bpm) with a Glasgow Coma Scale of 3, and fixed and dilated pupils. An ECG revealed a QRS of 110 ms, that narrowed to 80 ms within 80 minutes after administration of IV fluids and IV sodium bicarbonate, and an arterial blood gas analysis indicated severe metabolic acidosis. A serum lidocaine concentration at 2 hours post-ingestion was 16.6 mcg/mL (reference range 1.2 to 5 mcg/mL) that decreased to 0.7 mcg/mL 6 hours post-ingestion. There was no evidence of epileptiform activity on an EEG, and a brain MRI showed hypoxic ischemic injury approximately 40 hours post-ingestion. The patient was apneic with no brainstem reflexes on hospital day 9, and brain death was subsequently declared (Drapkin et al, 2015).
    c) DIBUCAINE: Two toddlers (aged 18 months and 21 months) developed generalized tonic-clonic seizures and severe dysrhythmias following ingestions (15 mg/kg to 19 mg/kg) of 1% dibucaine ointment. Despite aggressive supportive measures, both children died within hours of ingestion (Dayan et al, 1996).
    1) A 2-year-old boy was lethargic, ataxic, experienced seizures, and subsequently developed respiratory and cardiac arrest after ingesting an unknown amount of 0.5% dibucaine cream. Despite resuscitative efforts, the patient's condition deteriorated and he died (Dayan et al, 1996).
    d) DIBUCAINE: Intermittent generalized tonic-clonic seizures occurred in an 18-month-old infant following ingestion of approximately 150 mg (12.5 mg/kg) of a sunburn ointment containing 1% dibucaine. The seizures resolved approximately 5 minutes following rectal administration of diazepam (Nelsen et al, 2009).
    3) CASE REPORTS - ADULTS
    a) LIDOCAINE: A 21-year-old man developed seizures, respiratory distress, hypotension, asystole, and subsequently died after gargling with 20 mL of 4% lidocaine solution (containing 800 mg of lidocaine) in preparation for an upper gastrointestinal endoscopy. Although the patient was advised not to swallow the lidocaine solution, it is believed that some of the solution did enter the gastrointestinal tract. The recommended maximum dose for local anesthesia is 200 mg (Zuberi et al, 2000).
    4) INTRAVENOUS ADMINISTRATION
    a) Seizures may occur following intravenous administration (Selden & Burke, 1988; Edgren et al, 1986; Brown & Skiendzielowski, 1980) .
    1) CASE REPORTS
    a) LIDOCAINE/INFANT: Intravenous injection of 12 mg/kg lidocaine in a one-month-old child (Jonville et al, 1990) resulted in seizures.
    b) LIDOCAINE/ADULT: A generalized tonic-clonic seizure occurred in a 54-year-old woman immediately after intravenous administration of lidocaine 200 mg during a cardiopulmonary bypass procedure. The patient recovered without any neurologic deficits (Lee et al, 1999).
    c) BUPIVACAINE/ADULT: A 37-year-old man, undergoing shoulder surgery, developed bradycardia, seizures, and cardiac arrest immediately after receiving bupivacaine as a nerve block. Despite undergoing cardiopulmonary resuscitation for approximately 2 hours and being placed on cardiopulmonary bypass, the patient died approximately 7 hours post-injection. An autopsy revealed several cardiac abnormalities, including cardiomegaly, myocardial bridging, and lipomatous hypertrophy of the intra-atrial septum. Rash and laryngeal edema were also present. Post-mortem toxicology results revealed cardiac blood bupivacaine concentration of 1.1 mcg/mL and an elevated total serum tryptase concentration of 45.1 ng/mL in cardiac blood (reference range, less than 11.4 ng/mL). The abrupt onset of seizures and the elevated bupivacaine concentration suggest inadvertent intravascular injection instead of administration as a peripheral nerve block, although the elevated tryptase concentration and the presence of rash and laryngeal edema also suggest the possibility of bupivacaine hypersensitivity as a possible contributing factor (Dudley et al, 2011).
    d) ROPIVACAINE/ADULT: A 74-year-old man experienced a tonic-clonic seizure approximately 90 seconds after inadvertent intravascular administration of ropivacaine. The seizure resolved following the intravenous administration of propofol (Ruetsch et al, 1999).
    5) INTRAMUSCULAR ADMINISTRATION
    a) Seizures may occur (Malone et al, 1988).
    6) INTERCOSTAL/EPIDURAL ADMINISTRATION
    a) BUPIVACAINE/CASE SERIES: Four cases of seizures with hypoxia and acidosis occurred following intercostal and epidural bupivacaine administration. Seizures occurred within 2 to 10 minutes of injection. The incidence of bupivacaine induced seizures following this procedure appears to be less than 0.2% (Moore et al, 1982a).
    b) BUPIVACAINE/CASE REPORTS (CHILDREN): There are several reports of tonic-clonic seizures occurring in pediatric patients 10 to 56 hours after starting a continuous epidural infusion. Serum bupivacaine levels ranged from 5.4 to 10.2 mcg/mL (McCloskey et al, 1992).
    c) ROPIVACAINE: Generalized seizures have occurred within minutes following accidental intravenous (during attempted epidural) ropivacaine administration in a child and in an adult. Both patients recovered without neurologic sequelae (Abouleish et al, 1998; Plowman et al, 1998).
    7) SUBCUTANEOUS ADMINISTRATION
    a) LIDOCAINE/INFANT: A 7-week-old developed seizures 20 to 30 minutes after receiving 3 ml of 1% lidocaine (6.7 mg/kg) in a dorsal penile nerve block for a circumcision (Donald & Derbyshire, 2004).
    b) LIDOCAINE/CHILD: Seizures have been reported following subcutaneous injection of 31 mg/kg of lidocaine in a 2-year-old child (Alfano et al, 1984).
    c) BUPIVACAINE/ADOLESCENT: A 13-year-old girl developed generalized tonic-clonic seizures followed by ventricular fibrillation approximately 30 minutes after subcutaneous administration of bupivacaine (total dose 30 mg) for wound debridement. The patient recovered following supportive measures (Yan & Newman, 1998).
    8) TOPICAL ADMINISTRATION
    a) Seizures have also occurred following topical application of local anesthetics to burn patients (Wehner & Hamilton, 1984) or patients with multiple erosive skin lesions (Lie et al, 1990).
    b) CASE REPORTS - PEDIATRIC
    1) EMLA: A 3-year-old girl developed seizures, coma, and a raise rash after topical application of EMLA cream under a plastic wrap in preparation for allergy skin testing (Parker et al, 2004). The child also developed methemoglobinemia to 17.7%. A serum lidocaine level, drawn 2.5 hours after EMLA application, was 3.0 mcg/mL.
    2) EMLA: A 21-month-old child developed generalized seizures 1 hour after topical administration of 75 grams of EMLA cream (containing lidocaine 2.5% and prilocaine 2.5%) to her chest and abdomen. The patient recovered after removal of the cream and administration of phenobarbital. Three hours after removal of the EMLA cream, serum lidocaine levels were 2.5 mcg/mL (Rincon et al, 2000).
    3) EMLA: A 4-month-old infant, with port wine stains on her torso and legs, developed generalized tonic-clonic seizures approximately 75 minutes following topical application of 60 grams of EMLA cream on both legs and occlusion with a plastic wrap. With intraosseous administration of lorazepam and fosphenytoin, the patient's seizures resolved and did not recur (Larson et al, 2013).
    9) INTRAURETHRAL ADMINISTRATION
    a) LIDOCAINE/CASE REPORT: An 80-year-old, previously seizure-free man experienced generalized tonic-clonic seizures after each of 2 intraurethral instillations of 20 mL of 2% lidocaine jelly (Sundaram, 1987).
    b) LIDOCAINE/CASE REPORT: An 87-year-old man suffered cardiac arrest after intraurethral administration of lidocaine for cystourethroscopy (Chang et al, 2005).
    10) STELLATE GANGLION BLOCK
    a) INCIDENCE: Wulf et al (1991), in a survey of German hospitals, reported that 0.76 out of every 1000 stellate ganglion blocks resulted in seizures.
    b) LIDOCAINE/CASE REPORT: Generalized seizures, cyanosis, and nystagmus occurred in 2 patients, a 28-year-old woman and a 51-year-old man while they were undergoing stellate ganglion blockade with 1% lidocaine injection (Mahli et al, 2002).
    11) INTRAOCULAR ADMINISTRATION
    a) PROPARACAINE/CASE REPORT: A 28-year-old woman with a corneal abrasion was given 2 drops of proparacaine hydrochloride in the conjunctival sac and experienced a tonic-clonic seizure. She was not on any other medication, nor did she have a history of seizures. This appears to have been an unusual adverse effect of this medication (Cydulka & Betzelos, 1990).
    12) INTRAPLEURAL
    a) BUPIVACAINE: Tonic-clonic seizures were reported in a pediatric patient 21 hours after starting a continuous intrapleural infusion (0.25 to 0.5 mg/kg/hr). Serum bupivacaine level was 5.6 mcg/mL (Agarwal et al, 1992).
    b) LIDOCAINE/CASE REPORT: A 40-year-old woman experienced 3 to 4 episodes of generalized tonic-clonic seizures within minutes after intraureteral administration of diluted lidocaine jelly for ureteral stone manipulation (Pantuck et al, 1997).
    13) ROPIVACAINE
    a) Seizures have occasionally been described after overdose (Eledjam, 2000; Mardirosoff & Dumont, 2000) or with therapeutic doses (Bisschop et al, 2001) of ropivacaine.
    b) CASE REPORT: A 25-year-old woman developed numbness of her lip, lost consciousness, and experienced a generalized seizure within 13 minutes of receiving a 300 mg dose injection (6.25 mg/kg) of ropivacaine as an axillary brachial plexus block for preparation of surgery on her finger. During the patient's seizure, her heart rate increased from 85 to 140 beats per minute, and her systolic blood pressure increased from 104 to 160 mmHg. The seizure spontaneously resolved after approximately 20 seconds and, during surgery, her heart rate and blood pressure normalized. Laboratory analysis of a blood sample, obtained 28 minutes postinjection, revealed a plasma ropivacaine concentration of 3.65 mcg/mL (Kimura et al, 2007).
    c) CASE REPORT: A 25-year-old man developed isolated sinus tachycardia (130 beats per minute) and tonic-clonic seizures approximately 1 minute following unintentional intravascular administration of 10 mL (75 mg) of ropivacaine for a sciatic nerve block. The patient recovered following supportive therapy (Petitjeans et al, 2002). Plasma ropivacaine concentrations, obtained 15, 90, and 150 minutes after onset of symptoms, were 13.1, 11.6, and 3.6 mcmol/L, respectively.
    B) CENTRAL NERVOUS SYSTEM DEFICIT
    1) Subjective effects include drowsiness, impending doom, headache, dizziness, paresthesias, euphoria, numbness of the mouth, lightheadedness, and tinnitus (Scott, 1986).
    a) LIDOCAINE: Signs may occur at total lidocaine serum concentrations of 3.5 to 5 mcg/mL.
    2) LIDOCAINE: Twitching, tremors, confusion, delirium, slurred speech, agitation, and seizures may occur at 5 to 11 mcg/mL. Tremors persisted for 48 hours following intravenous injection of 12 mg/kg lidocaine in an infant (Jonville et al, 1990).
    3) LIDOCAINE: CNS effects invariably precede significant cardiovascular toxicity in patients receiving long-term (12 to 72 hr) lidocaine infusions (Pieper et al, 1982).
    4) CASE REPORTS
    a) ADULTS
    1) DIBUCAINE: Severe intense burning phantom limb pain occurred following spinal anesthesia with 1.5 mL dibucaine 0.5% in 6% dextrose injected at 4,5 level in a 77-year-old man who had undergone a left above the knee amputation for osteosarcoma 30 years ago (Murphy & Anandaciva, 1984).
    2) LIGNOCAINE: A pregnant 28-year-old patient received intravenous regional anesthesia (tourniquet technique) using lignocaine for hand surgery. Surgery was uneventful and, 45 minutes later, the tourniquet was deflated. Sixty seconds later, the patient experienced anxiety, severe lightheadedness, dizziness, weakness, perioral paresthesias, and a feeling of "dissociation". The patient recovered following supportive care (Coleman & Kelly, 1998).
    3) LIDOCAINE: A 30-year-old woman received 5 grams of 40% lidocaine topically following laser surgery for treatment of stretch marks and, within 1 hour of lidocaine application, complained of lightheadedness, dizziness, and confusion. Seven hours after presentation to the emergency department, the patient's lidocaine blood level was 2.7 mg/mL (toxic levels > 5 mcg/mL). The patient recovered following supportive care (Goodwin & McMeekin, 1999).
    4) LEVOBUPIVACAINE: A 77-year-old woman who accidentally received 142.5 mg levobupivacaine intravenously developed disorientation, drowsiness, and slurred speech, and then became immediately agitated. With supportive care and immediate discontinuation of the injection, the agitation subsided and the patient's mental status returned to baseline. Serum levobupivacaine levels were 2.7 mcg/mL and 1.1 mcg/mL approximately 14 minutes and 120 minutes, respectively, after cessation of the injection (Kopacz & Allen, 1999).
    5) ROPIVACAINE: A 45-year-old woman developed perioral numbness and twitching and restlessness approximately 3 minutes after inducing a brachial plexus block via high-dose ropivacaine (6 mg/kg). The patient recovered with supportive care. Six weeks later, the patient again received high-dose ropivacaine (4.5 mg/kg) for induction of brachial plexus block. Twenty-five minutes after the injection, the patient experienced a strange feeling in her tongue followed by dysarthria and involuntary clonic movements of her head. Again, the patient recovered with no further sequelae (Ala-Kokko et al, 2000).
    6) EMLA: A 23-year-old man developed weakness, nausea and vomiting, confusion, disorientation, and lost consciousness briefly followed by development of a frontal headache approximately 45 minutes after topical application of EMLA cream to his chest, abdomen, and back for preparation of laser hair-removal treatment. The EMLA cream contained 2.5% lidocaine and 2.5% prilocaine. All laboratory tests, ECG, EEG, a head CT scan, and a lumbar puncture were normal; however, the patient's CSF subsequently showed a lidocaine level of approximately 1 mg/L. Two hours after onset of symptoms, the patient completely recovered without sequelae (Brosh-Nissimov et al, 2004).
    7) LIDOCAINE
    a) An 87-year-old woman, experiencing dyspnea and persistent ventricular tachycardia, received 2 IV bolus doses of lidocaine 50 mg and initiation of a lidocaine infusion of 2 mg/minute en route to the hospital via emergency medical services. At the emergency department, the patient's heart rate was 152 beats per minute and she received a third IV bolus dose of lidocaine 50 mg. An ECG demonstrated normal sinus rhythm with left ventricular hypertrophy, and her lidocaine infusion rate was increased to 4 mg/minute. The patient was transferred to another hospital, where it was noted that she was nonverbal, unresponsive to painful stimuli, with pupils 4 mm and nonreactive, suggesting a brainstem stroke. A CT scan of the head showed mild microvascular ischemic changes and a National Institute of Health (NIH) stroke score was 24, indicating severe neurological impairment. The patient's lidocaine infusion was discontinued, and 30 minutes later, a serum lidocaine concentration was obtained revealing a concentration of 7.9 mg/L (reference range 1.5 to 6 mg/L). After discontinuation of the infusion, the patient's mental status continually improved. Approximately 1 hour after cessation of her infusion, a repeat NIH score was 3, indicating minimal impairment. The total lidocaine dose that the patient had received was 811 mg over a period of 3 hours and 20 minutes (approximately 13 mg/kg, over 2 half-lives of the drug) (Bursell et al, 2009).
    b) HOIGNE SYNDROME: A 22-year-old woman, with a history of peptic ulcer disease and anxiety disorder, developed paresthesias of her right leg and ear, lightheadedness, severe anxiety, and restlessness, accompanied by visual hallucinations with a fear of impending death (Hoigne syndrome) after receiving lidocaine injections in preparation for a bone marrow biopsy. Total lidocaine dose was 360 mg (5 mg/kg). Initial vital signs demonstrated elevated blood pressure (144/61 mmHg), heart rate (133 beats/min), and respiratory rate (26 breaths per minute). Laboratory workup and a CT scan of the brain were normal. Symptoms improved following administration of IV lorazepam, and the patient was discharged following a medical toxicology consult (Thompson & Theobald, 2016).
    b) PEDIATRIC
    1) EMLA: A 4-year-old girl developed a headache, instability in walking, blurry vision, and perioral cyanosis within 90 minutes following topical application of 30 grams of EMLA cream for treatment of extensive molluscum contagiosum. Her oxygen saturation was 88% and her methemoglobin level was 19% via cooximetry. After washing off the EMLA cream and administering 1 mg/kg of methylene blue intravenously, the patient completely recovered without sequelae (Raso et al, 2006).
    C) COMA
    1) Coma has been reported following appropriate and inappropriate dosing of local anesthetics. Coma may occur suddenly after rapid IV or intraarterial injection.
    2) LIDOCAINE
    a) CASE REPORT: Two elderly, terminally ill cancer patients receiving lidocaine for palliative pain control developed severe somnolence following therapeutic doses (300 mg and 200 mg daily, respectively) with serum concentrations of more than 8.0 mcg/mL (therapeutic 1.5 to 5 mcg/mL). Both improved with drug withdrawal. Routine monitoring and modified dosing may be required in terminally ill patients (Tei et al, 2005).
    3) BUPIVACAINE
    a) CASE REPORT: A 28-year-old woman became unresponsive with flaccid extremities after receiving an accidental spinal injection (intended for the paracervical area) of a mixture containing Bupivacaine and triamcinolone. The patient gradually recovered without neurologic sequelae (Wang & Chang, 1993).
    4) BUPIVACAINE/EPINEPHRINE
    a) CASE REPORT: A 55-year-old man went almost immediately into a coma following inadvertent intraarterial injection of 26 mL 0.75% bupivacaine with epinephrine (5 mcg/mL) (Tuominen et al, 1991).
    5) DIBUCAINE
    a) CASE REPORT/INFANT: An 18-month-old infant became comatose with a Glasgow Coma Scale score of 3 following ingestion of approximately 150 mg (12.5 mg/kg) of a sunburn ointment containing 1% dibucaine. The patient also experienced generalized tonic-clonic seizures and ECG abnormalities, including AV nodal blockade with widening of the QRS complex. With supportive care, the patient recovered without sequelae (Nelsen et al, 2009).
    6) PRAMOXINE
    a) CASE REPORT: A 4-year-old boy ingested 100 mL of a lotion containing pramoxine and, several hours later, experienced vomiting, somnolence, dysrhythmias, tremulousness, hypertonia, and periods of unresponsiveness with tonic flexion of the extremities. The patient completely recovered 3 days after ingestion (Stancavage et al, 1995).
    D) ATAXIA
    1) WITH THERAPEUTIC USE
    a) LIDOCAINE: Ataxia, dysarthria, dysmetria, involuntary movements, loss of balance, and transient aphasia have been reported rarely after use of topical lidocaine for endoscopic procedures. Signs and symptoms resolved within 2 to 5 hours (Perney et al, 2003).

Gastrointestinal

    3.8.2) CLINICAL EFFECTS
    A) VOMITING
    1) Nausea and vomiting may occur with overdose ingestions of local anesthetics (Dayan et al, 1996; Stancavage et al, 1995).
    2) EMLA: Nausea and vomiting occurred in a 23-year-old man following topical application of EMLA cream (containing 2.5% of lidocaine and 2.5% prilocaine) to his chest, abdomen, and back (Brosh-Nissimov et al, 2004).
    3) PRAMOXINE: In a retrospective series of 177 poison center cases of ingestion of pramoxine-containing products, nausea developed in 2 patients and vomiting in 4. Nausea and vomiting persisted for up to 4 hours (Spiller et al, 2006).
    B) HYPOACTIVE GAG REFLEX
    1) CASE REPORT/LIDOCAINE: A depressed gag reflex and significant pharyngeal secretions were reported in a 5-month-old following the oral administration of 2% viscous lidocaine (Smith et al, 1992).
    C) DIARRHEA
    1) WITH POISONING/EXPOSURE
    a) PRAMOXINE: In a retrospective series of 177 poison center cases of ingestion of pramoxine-containing products, diarrhea developed in 2 patients and persisted for up to 4 hours (Spiller et al, 2006).

Acid-Base

    3.11.2) CLINICAL EFFECTS
    A) ACIDOSIS
    1) Acidosis may occur in patients with hypoxia, respiratory depression, hypertension, or seizures.
    2) CASE REPORT/LIDOCAINE: A 15-year-old girl developed seizures after ingesting up to 2 grams (49 mg/kg) of viscous lidocaine. She was tachycardic (160 bpm) with a Glasgow Coma Scale of 3, and fixed and dilated pupils. An ECG revealed a QRS of 110 ms, that narrowed to 80 ms within 80 minutes after administration of IV fluids and IV sodium bicarbonate, and an arterial blood gas analysis indicated severe metabolic acidosis (pH 6.6, pCO2 91, lactate 18 mmol/L). A serum lidocaine concentration at 2 hours post-ingestion was 16.6 mcg/mL (reference range 1.2 to 5 mcg/mL) that decreased to 0.7 mcg/mL 6 hours post-ingestion. There was no evidence of epileptiform activity on an EEG, and a brain MRI showed hypoxic ischemic injury approximately 40 hours post-ingestion. The patient was apneic with no brainstem reflexes on hospital day 9, and brain death was subsequently declared (Drapkin et al, 2015).

Hematologic

    3.13.2) CLINICAL EFFECTS
    A) METHEMOGLOBINEMIA
    1) SUMMARY: Methemoglobinemia has been reported in patients who have received certain local anesthetics topically, intravenously, or subcutaneously. Numerous cases have been attributed to prilocaine or benzocaine. Isolated reports to tetracaine, (Olson & McEvoy, 1981), Cetacaine (Byrne et al, 2004; Ferraro et al, 1988), and lidocaine (Kotler et al, 1989) (Rothrock & Green, 1992) have been described, often in combination with other methemoglobin-inducing agents.
    2) VARIOUS AGENTS
    a) BENZOCAINE
    1) INCIDENCE: From November 1997 to March 2002 there were 132 cases of definite (n=99) or probable (n=33) methemoglobinemia related to benzocaine use reported to the FDA. Of these, 107 were considered serious adverse events and 2 were lethal. In most cases (123; 93%) the product was a spray. In the 69 cases that specified a dose, 37 (53.6%) involved use of a single spray, which is approximately the recommended amount (Moore et al, 2004).
    2) EAR DROPS: Benzocaine toxicity may have resulted in the death of a 4-month-old infant. The infant received 3 times the prescribed dose of ear drops containing 0.25% w/v benzocaine and 5.4% w/v antipyrine. Postmortem methemoglobin was 36% (Logan & Gordon, 2005).
    3) TOPICAL: Severe methemoglobinemia was reported in a toddler following topical use of benzocaine for an endoscopic procedure (Hegedus & Herb, 2005).
    4) SWISH AND SWALLOW: Benzocaine used as a local agent ("swish and swallow") during an outpatient transesophageal echocardiography produced methemoglobinemia (Wurdeman et al, 2000).
    5) SPRAY: A patient developed severe methemoglobinemia after the use of benzocaine spray during an endoscopic procedure. Following treatment with methylene blue, critical rebound phenomenon occurred, which required further methylene blue (Fitzsimons et al, 2004) .
    a) In one case series, benzocaine-induced methemoglobinemia developed in 37 (53%) of 69 patients treated with a single spray (recommended dose) of benzocaine (Moore et al, 2004a).
    6) ADULTERATION: Severe methemoglobinemia (a methemoglobin level of 37%) was reported in an adult following a "large" ingestion of street cocaine that was adulterated with benzocaine used as a cutting substance (McKinney et al, 1992).
    b) LIDOCAINE
    1) Methemoglobinemia has been associated with lidocaine use (Hansen-Flaschen, 1990; Kotler et al, 1989a).
    a) Methemoglobinemia occurs rarely with therapeutic use, and is usually not clinically significant. Routine methemoglobin monitoring is not necessary. There are a few cases during therapeutic use when excessive doses may have been employed, and often in combination with other agents known to cause methemoglobinemia (Kotler et al, 1989; Bun & Doughty, 1974).
    1) CLINICAL TRIAL: Forty cardiac patients were given a 1 mg/kg bolus of lidocaine IV followed by a maintenance infusion of 2 mg/min and a second bolus of 0.5 mg/kg. Infusion rates were adjusted up to 4 mg/min and lidocaine and methemoglobin levels were measured at 1 and 6 hours post-bolus. The highest methemoglobin level was 1.2%, which was not clinically significant (Weiss et al, 1987).
    b) TOPICAL: Severe methemoglobinemia occurred after topical lidocaine was used in patients prior to transesophageal echocardiography (Karim et al, 2001).
    c) BUPIVACAINE/LIDOCAINE
    1) AXILLARY BLOCK/BUPIVACAINE/LIDOCAINE: A patient developed clinical methemoglobinemia following an axillary block with bupivacaine and an additional injection of lidocaine in the operative field (Schroeder et al, 1999).
    d) CETACAINE
    1) Cetacaine may cause methemoglobinemia (Ferraro et al, 1988a).
    2) Four cases of Cetacaine-induced (14% benzocaine, 2% butyl aminobenzoate, 2% tetracaine hydrochloride) methemoglobinemia during endoscopic retrograde cholangiopancreatography procedures have been reported. Methemoglobin levels ranged from 17% to 44.5% (normal range 0.4% to 1.5%). Two patients required treatment with methylene blue, and the remaining 2 patients recovered with the administration of supplemental oxygen only (Byrne et al, 2004).
    e) EMLA CREAM
    1) Application of 150 g EMLA cream, which contained prilocaine and lidocaine, caused methemoglobinemia; symptoms developed within 1 hour (Hahn et al, 1999). In another case, EMLA cream that was left in place for 5 hours produced a methemoglobin level of 16% (Sinisterra et al, 2002).
    2) CASE REPORT: A 4-year-old girl developed a headache, instability in walking, blurry vision, and perioral cyanosis within 90 minutes following topical application of 30 grams of EMLA cream for treatment of extensive molluscum contagiosum. Her O2 saturation was 88% and her methemoglobin level was 19% via cooximetry. After washing off the EMLA cream and administering 1 mg/kg of methylene blue intravenously, the patient completely recovered without sequelae (Raso et al, 2006).
    3) CASE REPORT: A 4-month-old infant, with port wine stains on her torso and legs, became cyanotic and developed tonic-clonic seizures approximately 75 minutes following topical application of 60 grams of EMLA cream to both legs and occlusion with a plastic wrap. Seizures resolved with intraosseous administration of lorazepam and fosphenytoin. Initial laboratory data indicated a methemoglobin level of 22.8%. Following administration of methylene blue, the patient's methemoglobin level initially dropped to 6% approximately 1 hour later, rebounding to 14% at 8 hours, and dropping to 3.4% at 18 hours without further administration of methylene blue. The patient remained stable and was discharged without neurologic sequelae 4 days post-admission (Larson et al, 2013).
    f) PRILOCAINE
    1) Prilocaine has produced methemoglobinemia (CDC, 1994; Bellamy et al, 1992; Lloyd, 1992a; Marks & Desgrand, 1991; Bardoczky et al, 1990; Mandel, 1989; Ludwig, 1981) .
    2) PRILOCAINE: Methemoglobinemia has occurred with SC or epidural doses of 6 to 24 mg/kg, universal in adults given 900 mg or more; onset usually 30 to 60 minutes, but has been 4 to 6 hours or more after multiple injections. Peak MetHb levels occurred in 1.5 to 4 hours (range 10% to 41.5%) and returned to normal within 24 to 36 hours (untreated) and 1.5 to 24 hours (treated). Low levels (mean 6%) resolved in 12 hours.
    a) Methemoglobinemia has also been reported after topical use of excessive doses 25 g of EMLA cream (2.5% prilocaine and 2.5% lidocaine) in a 3-year-old (Touma & Jackson, 2001).
    b) Methemoglobinemia (20%) developed in an adult who used 150 grams EMLA cream to her legs below the knees prior to a laser epilation procedure. She had evidence of first-degree burns from a previous epilation procedure that probably enhanced dermal absorption. She also had signs and symptoms of systemic toxicity, including lightheadedness, tongue numbness, muscular twitching, and formication (Hahn et al, 2003).
    c) The etiology is metabolism to o-toluidine and 4-hydroxy-2- methylaniline, both known methemoglobin inducers (Lloyd, 1992; Bardoczky et al, 1990; Frayling et al, 1990; Klos & Hays, 1985; Duncan & Kobrinsky, 1983; Kreutz & Kinni, 1983; Spoerel et al, 1967; Scott et al, 1964).
    3) CASE REPORT: A 2.5-month-old child developed central cyanosis after receiving 30 mg (6 mg/kg) of prilocaine subcutaneously during a circumcision. O2 saturation, via pulse oximetry, was 90%, and his blood methemoglobin level at admission was 39%. Due to unavailability of methylene blue, the patient was given a single 300 mg/kg dose of ascorbic acid intravenously, resulting in complete resolution of his cyanosis within 4 hours. Over the next 16 hours, the patient's methemoglobin level decreased from 39% to 0.6% (Akbayram et al, 2012).
    g) PROPITOCAINE
    1) Propitocaine may produce methemoglobinemia (Poppers et al, 1956).
    h) TETRACAINE
    1) CASE REPORT: A 71-year-old woman, taking sulfamethoxazole/trimethoprim for a chronic urinary tract infection, developed methemoglobinemia after receiving a nasal spray containing tetracaine 1% in combination with oxymetazoline 0.025% in preparation for a transnasal laryngoscopic procedure. The patient was asymptomatic; however, her pulse oximetry, recorded in the emergency department, was 91% on 2 liters nasal cannula and her methemoglobin concentration was 10.5%. Following an observation period and weaning to room air, the patient remained asymptomatic and was discharged home. It is believed that concomitant administration of sulfamethoxazole and tetracaine may have resulted in development of methemoglobinemia (Whited & Cohen, 2012).

Dermatologic

    3.14.2) CLINICAL EFFECTS
    A) CONTACT DERMATITIS
    1) Allergic or contact dermatitis may develop following topical use.
    2) PRAMOXINE: Two cases of contact eczema have been reported with topical use of pramoxine (Van Ketel, 1981).
    B) PARESTHESIA
    1) Generalized burning and numbness have been reported.
    C) URTICARIA
    1) Urticaria and angioedema may accompany systemic allergic reactions (eg, anaphylaxis).
    D) ITCHING OF SKIN
    1) DIBUCAINE: Pruritus was associated with spinal anesthesia induced by dibucaine (Cashman, 1959).
    E) VASODILATATION
    1) CASE REPORT/LIDOCAINE: Vasodilation and pink coloring of skin have occurred from accidental intravenous administration of 12 mg/kg lidocaine in an infant (Jonville et al, 1990).

Musculoskeletal

    3.15.2) CLINICAL EFFECTS
    A) SPASMODIC MOVEMENT
    1) Weakness, tremors, twitching and shivering have been reported.
    B) MUSCLE PAIN
    1) BUPIVACAINE: Persistent neck pain was reported in a 40-year-old woman after receiving interscalene injections of large doses of bupivacaine with epinephrine during a surgical procedure. A biopsy of the left sternocleidomastoid muscle showed degenerating and regenerating muscle fibers, myophagia, fiber splitting, and inflammatory infiltrates with eosinophils. The patient gradually recovered within 3 months postoperatively (Hogan et al, 1994).

Endocrine

    3.16.2) CLINICAL EFFECTS
    A) DIABETES INSIPIDUS
    1) CASE REPORT/TETRACAINE: A chemical meningitis resulting from an intrathecal injection of 15 mg 1% tetracaine and 10% dextrose was associated with long-term diabetes insipidus in a 21-year-old man. He had also received 5 mg morphine, 50 mg hydroxyzine, and atropine 400 mcg. Diabetes insipidus resolved after 3 months of self-medication with nasal vasopressin acetate (Garfield et al, 1986).
    B) ABNORMAL ANTI-DIURETIC HORMONE
    1) LIDOCAINE: Syndrome of inappropriate antidiuretic hormone secretion occurred in a 2-year-old after apparently aspirating and swallowing a small amount of 2% viscous lidocaine (Garrettson & McGee, 1992).

Immunologic

    3.19.2) CLINICAL EFFECTS
    A) ACUTE ALLERGIC REACTION
    1) True allergic reactions (Type 1 anaphylaxis and Type 4 delayed hypersensitivity) are rare (Doyle & Goepferd, 1989; Bryant et al, 1983).

Reproductive

    3.20.1) SUMMARY
    A) Lidocaine and the combination of lidocaine/prilocaine and lidocaine/tetracaine are classified as FDA pregnancy category B. Articaine hydrochloride/epinephrine and bupivacaine hydrochloride are classified as FDA pregnancy category C. Local anesthetics readily cross the placenta. Fetal or neonatal poisoning may occur as a result of spinal or regional nerve blocks, systemic maternal poisoning, or inadvertent intracranial injection of local anesthetics into the fetus during labor and delivery.
    3.20.2) TERATOGENICITY
    A) LACK OF EFFECT
    1) OXYMETAZOLINE HYDROCHLORIDE/TETRACAINE HYDROCHLORIDE
    a) There are no adequate or well-controlled trials for the use of the combination oxymetazoline hydrochloride/tetracaine hydrochloride or the single agent tetracaine in pregnant women. However, limited data from epidemiologic studies of oxymetazoline used intranasally as a decongestant during pregnancy in humans did not show any associated teratogenicity (Prod Info KOVANAZE(TM) nasal spray, 2016).
    B) ANIMAL STUDIES
    1) LIDOCAINE
    a) There were no teratogenic effects in animals given subQ doses up to 1200 times the single dermal administration of 0.5 mg lidocaine in a 60 kg person (Prod Info Zingo(TM) intradermal injection powder, 2014).
    2) OXYMETAZOLINE HYDROCHLORIDE/TETRACAINE HYDROCHLORIDE
    a) In animal studies, structural abnormalities, including short forelimb digits, fused and irregular shaped arches in thoracic vertebrae, fused ribs, irregular number of ribs, and unossified forelimb phalanx, and reduced fetal weights were observed following the subQ administration of oxymetazoline to rats during organogenesis at a maternally toxic dose approximately 7.6 times the exposure from oxymetazoline at the maximum recommended human dose (MRHD) of the combination (Prod Info KOVANAZE(TM) nasal spray, 2016).
    3.20.3) EFFECTS IN PREGNANCY
    A) LACK OF INFORMATION
    1) BUPIVACAINE
    a) There are no adequate and well-controlled studies of bupivacaine liposome use in pregnant women. Bupivacaine rapidly crosses the placenta by passive diffusion, and the degree of plasma protein binding, ionization, and lipid solubility determine the rate and extent of diffusion. The fetal/maternal ratio is inversely related to the degree of plasma protein binding. Bupivacaine has a high protein binding capacity of 95% and a low fetal/maternal ratio of 0.2 to 0.4. Being lipid soluble and nonionized, bupivacaine rapidly diffuses to fetal blood from the maternal circulation (Prod Info EXPAREL(R) injection suspension, 2015).
    2) OXYMETAZOLINE HYDROCHLORIDE/TETRACAINE HYDROCHLORIDE
    a) There are no adequate or well-controlled trials for the use of the combination oxymetazoline hydrochloride/tetracaine hydrochloride or the single agent tetracaine in pregnant women (Prod Info KOVANAZE(TM) nasal spray, 2016).
    B) APNEA
    1) MEPIVACAINE: Bradycardia, apnea, and death have been reported in fetuses and neonates whose mothers received caudal, paracervical, or pudendal mepivacaine at delivery (Dodson et al, 1975; Finster et al, 1965; Rosefsky & Petersiel, 1968; Hillman et al, 1979; Shnider et al, 1968; Sinclair et al, 1965).
    a) LIDOCAINE: Similar toxicity has occurred with lidocaine (Bozynski et al, 1987; Selden & Burke, 1988).
    C) BRADYCARDIA
    1) BUPIVACAINE: Bupivacaine is contraindicated in obstetrical paracervical block anesthesia. Use of bupivacaine in obstetrical paracervical block has resulted in fetal bradycardia and death (Prod Info EXPAREL(R) injection suspension, 2015).
    a) The US Food and Drug Administration has recommended that 0.75% bupivacaine solutions not be used for obstetrical epidural anesthesia because there have been reports of cardiac arrest and death occurring with this concentration. As a result of this recommendation, the manufacturer has withdrawn the obstetric indications for bupivacaine 0.75% solution (Prod Info Marcaine(R), bupivacaine, 1991). However, this decision has been heavily criticized (Writer et al, 1984; Scott, 1984). It is felt that with proper use of bupivacaine 0.75% in obstetrics, toxic effects can be avoided.
    D) LABOR AND DELIVERY
    1) BUPIVACAINE: Use of bupivacaine as obstetrical paracervical block anesthesia is contraindicated . Local anesthetics cross the placental barrier and may cause varying degrees of toxicity. Epidural, caudal, or pudendal anesthesia may alter uterine contractility and maternal expulsive efforts thereby prolonging labor. This may increase the need for forceps assistance. Avoid aortocaval compression by the gravid uterus during administration (Prod Info Sensorcaine(R)-MPF with Epinephrine parenteral injection, 2013; Prod Info Sensorcaine(R) with Epinephrine parenteral injection, 2013; Prod Info Sensorcaine(R)-MPF parenteral injection, 2013; Prod Info Sensorcaine(R) parenteral injection, 2013).
    2) BUPIVACAINE: Maternal, fetal, and neonatal toxicity may occur when bupivacaine is used for epidural, caudal, or pudendal block anesthesia (Prod Info EXPAREL(R) injection suspension, 2015).
    E) MEDICATION ERROR
    1) EPISIOTOMY: During the administration of lidocaine local anesthesia for episiotomy, the baby's scalp may be inadvertently injected. Levels of the local anesthetic will probably be negligible in the cord blood, but can be found in a venous blood sample taken after birth (De Praeter et al, 1991; Kim et al, 1979).
    a) Toxicity often appears around 15 minutes after birth, with the baby becoming suddenly apneic, hypotonic, and with fixed pupils and decreased reflexes.
    b) Toxicity may occur with venous lidocaine levels as low as 3 mcg/mL. The usual lidocaine dose given for episiotomy is around 200 mg; this may result in a blood level of around 14 mcg/mL in the neonate.
    c) Intravenous lidocaine overdose may result in immediate cardiorespiratory arrest (Brown & Skiendzielowski, 1980; Edgren et al, 1986; Selden & Burke, 1988). Complete maternal and fetal recovery has been reported despite prolonged cardiac arrest (Selden & Burke, 1988).
    F) PREGNANCY CATEGORY
    1) The manufacturer has classified ARTICAINE HYDROCHLORIDE/EPINEPHRINE as FDA pregnancy category C (Prod Info Septocaine(R) Solution for intraoral submucosal injection, 2009).
    2) The manufacturer has classified BUPIVACAINE HYDROCHLORIDE as FDA pregnancy category C (Prod Info Sensorcaine(R)-MPF with Epinephrine parenteral injection, 2013; Prod Info Sensorcaine(R) with Epinephrine parenteral injection, 2013; Prod Info Sensorcaine(R)-MPF parenteral injection, 2013; Prod Info Sensorcaine(R) parenteral injection, 2013).
    a) Bupivacaine crosses the placental barrier. Use during pregnancy only if the potential maternal benefit outweighs the potential fetal risk. Use of bupivacaine as obstetrical paracervical block anesthesia is contraindicated (Prod Info Sensorcaine(R)-MPF with Epinephrine parenteral injection, 2013; Prod Info Sensorcaine(R) with Epinephrine parenteral injection, 2013; Prod Info Sensorcaine(R)-MPF parenteral injection, 2013; Prod Info Sensorcaine(R) parenteral injection, 2013).
    3) BUPIVACAINE LIPOSOME: There are no adequate or well-controlled studies of bupivacaine liposome use in pregnant women. Advise pregnant women of the potential risks to the fetus (Prod Info EXPAREL(R) injection suspension, 2015).
    4) The manufacturer has classified LIDOCAINE as FDA pregnancy category B (Prod Info Zingo(TM) intradermal injection powder, 2014).
    5) The manufacturer has classified LIDOCAINE/PRILOCAINE as FDA pregnancy category B (Prod Info Oraqix(R) oral transmucosal gel, 2010).
    6) LIDOCAINE/TETRACAINE is classified as FDA pregnancy category B (Prod Info PLIAGLIS(R) topical cream, 2012; Prod Info Synera (TM) topical patch, 2005).
    7) OXYMETAZOLINE HYDROCHLORIDE/TETRACAINE HYDROCHLORIDE: Use caution when administering the oxymetazoline hydrochloride/tetracaine hydrochloride combination to a pregnant woman, as there are no adequate or well-controlled studies of the combination's use in pregnant women (Prod Info KOVANAZE(TM) nasal spray, 2016).
    G) SEIZURES
    1) MEPIVACAINE: Neonatal intoxication secondary to paracervical and pudendal mepivacaine blocks during labor has resulted in depression at birth, bradycardia, and seizures within 6 hours (Hillman et al, 1979; Asling et al, 1970; Bozynski et al, 1987).
    H) ANIMAL STUDIES
    1) BUPIVACAINE
    a) SubQ administration of bupivacaine hydrochloride in animals at doses comparable to the maximum recommended human dose (MRHD) resulted in an increase in embryofetal deaths. In postnatal development studies, decreased pup survival was reported at doses comparable to the MRHD (Prod Info Sensorcaine(R)-MPF with Epinephrine parenteral injection, 2013; Prod Info Sensorcaine(R) with Epinephrine parenteral injection, 2013; Prod Info Sensorcaine(R)-MPF parenteral injection, 2013; Prod Info Sensorcaine(R) parenteral injection, 2013).
    b) SubQ doses of bupivacaine up to 1.6 times the maximum recommended human dose (MRHD) resulted maternal lethality, in a decrease in pup survival, and an increase in embryo-fetal deaths (Prod Info EXPAREL(R) injection suspension, 2015).
    2) LIDOCAINE/EPINEPHRINE
    a) Lidocaine containing 1:100,000 epinephrine at a subQ dose of 120 times the single dermal administration caused temporary developmental delays in the neonate offspring of animals (Prod Info Zingo(TM) intradermal injection powder, 2014).
    3) LIDOCAINE/PRILOCAINE
    a) No evidence of altered postnatal development, viability, or reproductive capacity in any offspring was reported in rats administered up to 2.8 times the maximum recommended human dose of prilocaine on a mg/m(2) basis from gestation day 6 to weaning (Prod Info Oraqix(R) oral transmucosal gel, 2010).
    b) In studies in which animals were treated with a lidocaine and prilocaine 1:1 mixture based on weight, there was no evidence of fetal harm at subQ doses of up to approximately equivalent to the maximum recommended human dose of lidocaine. Intramuscular prilocaine doses of up to approximately 11 times the maximum recommended human dose did not impair fertility or result in fetal harm. Similarly, approximately 1.5 times the maximum recommended human dose each of lidocaine and prilocaine administered subQ produced no teratogenic, embryotoxic, or fetotoxic effects (Prod Info Oraqix(R) oral transmucosal gel, 2010; Prod Info EMLA(R) cream, 2005).
    c) In a study of animals treated with lidocaine 5 mg/kg subQ, fetal harm was not noted. When animals were treated with lidocaine 15 mg/kg, there was maternal toxicity and delayed fetal development, including a nonsignificant 7% decrease in fetal weight and an increase in minor skeletal anomalies (skull and sternebral defects, reduced ossification of the phalanges). In a study where animals were treated with lidocaine or prilocaine up to 1.4-fold the maximum recommended human dose for 8 months (3 mating periods), there was no abnormal postnatal development in any offspring. However, the average number of pups per litter surviving until weaning of offspring (from the first 2 mating periods) was significantly reduced with both doses of either drug (Prod Info Oraqix(R) oral transmucosal gel, 2010).
    4) LIDOCAINE/TETRACAINE
    a) Lidocaine and tetracaine administered both individually and in combination in doses less than one-fold higher than the single dermal administration were not teratogenic in animals (Prod Info PLIAGLIS(R) topical cream, 2012; Prod Info Synera (TM) topical patch, 2005).
    5) OXYMETAZOLINE HYDROCHLORIDE/TETRACAINE HYDROCHLORIDE
    a) In animal studies, reduced implantation sites and live litter sizes were noted following subQ administration of oxymetazoline at approximately 1.5 times the MRHD and increased pup mortality at 6 times the MRHD in a prenatal and postnatal development study. SubQ administration of tetracaine hydrochloride to rats and rabbits during organogenesis at 32 and 6 times, respectively, the exposure from tetracaine hydrochloride at the MRHD of the combination did not result in adverse developmental effects (Prod Info KOVANAZE(TM) nasal spray, 2016)
    3.20.4) EFFECTS DURING BREAST-FEEDING
    A) LACK OF INFORMATION
    1) BUPIVACAINE
    a) The effect of bupivacaine on breastfed infants or breast milk production has not been studied (Prod Info EXPAREL(R) injection suspension, 2015).
    2) OXYMETAZOLINE HYDROCHLORIDE/TETRACAINE HYDROCHLORIDE
    a) It is unknown whether oxymetazoline, tetracaine, or their metabolites are excreted into human breast milk (Prod Info KOVANAZE(TM) nasal spray, 2016).
    B) BREAST MILK
    1) BUPIVACAINE
    a) Bupivacaine is excreted into human breast milk. Discontinue bupivacaine or discontinue nursing taking into account the importance of the drug to the mother (Prod Info Sensorcaine(R)-MPF with Epinephrine parenteral injection, 2013; Prod Info Sensorcaine(R) with Epinephrine parenteral injection, 2013; Prod Info Sensorcaine(R)-MPF parenteral injection, 2013; Prod Info Sensorcaine(R) parenteral injection, 2013).
    b) Bupivacaine and its metabolite (pipecolylxylidide) are present in breast milk at low levels. When deciding whether to continue treatment in a breastfeeding patient, consider the benefits of breastfeeding, the mother's need for bupivacaine, and the potential adverse effects on the infant (Prod Info EXPAREL(R) injection suspension, 2015).
    2) OXYMETAZOLINE HYDROCHLORIDE/TETRACAINE HYDROCHLORIDE
    a) Consider the developmental and health benefits of breastfeeding, along with the mother's clinical need for the drug, and potential adverse effects on the nursing infant from the combination agent or from the mother's underlying condition when administering the oxymetazoline hydrochloride/tetracaine hydrochloride combination to a lactating woman (Prod Info KOVANAZE(TM) nasal spray, 2016).
    C) LACK OF EFFECT
    1) GENERAL
    a) Local anesthetics are present in breast milk, but concentrations (roughly 40% of serum levels) would not be expected to produce detectable effects (Giuliani et al, 2001; Briggs et al, 1998; Zeisler et al, 1986).
    2) BUPIVACAINE
    a) In one study assessing the magnitude of excretion of lidocaine, bupivacaine, and PPX (a metabolite of bupivacaine) in the breast milk of 27 patients undergoing cesarean delivery, the milk/serum ratios based on AUC values were 1.07 +/- 0.82, 0.34 +/- 0.24, and 1.37 +/- 0.61, respectively. The majority of the infants appeared to be in good health and there were no adverse effects to the infants related to the local anesthetic agents (Ortega et al, 1999).
    3) LIDOCAINE
    a) In one study assessing the magnitude of excretion of lidocaine, bupivacaine, and PPX (a metabolite of bupivacaine) in the breast milk of 27 patients undergoing cesarean delivery, the milk/serum ratios based on AUC values were 1.07 +/- 0.82, 0.34 +/- 0.24, and 1.37 +/- 0.61, respectively. The majority of the infants appeared to be in good health and there were no adverse effects to the infants related to the local anesthetic agents (Ortega et al, 1999).
    b) A 34-year-old woman received 20 mg of lidocaine injection for dental surgery while breastfeeding. Samples of milk and plasma were tested for lidocaine and monoethylglycinexylidide by high performance liquid chromatography. Milk concentrations for the lidocaine ranged from 44 to 66 mcg/L, giving a milk/plasma ratio of 1.1. Milk concentrations for the metabolite monoethylglycinexylidide ranged from 35 to 41 mcg/L giving a milk/plasma ratio of 1.8. The infant levels for the parent drug and metabolite were estimated to be less than 0.01 mg/kg/day; these levels were not considered to be pharmacologically significant (Lebedevs et al, 1993).
    c) Lidocaine is considered compatible with breastfeeding by the American Academy of Pediatrics. The majority of uses of lidocaine are acute, and although some lidocaine appears in breast milk after IV administration, the concentration is not considered to be pharmacologically significant. Any amount found in breast milk is further reduced by poor oral bioavailability to the breastfeeding infant (Gilman et al, 1990).
    4) LIDOCAINE/TETRACAINE
    a) Lidocaine is excreted into human milk with a milk:plasma ratio of 0.4. It has not been determined if tetracaine is excreted into human milk. Lidocaine milk-plasma ratios have ranged from 1.07 using AUC values when used as epidural anesthetic for cesarean section (n=27) to 1.1 when measured 5 to 6 hours after a 20 mg single dose injection for a dental procedure. This translates into an estimated maximum daily lidocaine exposure via breast milk of 36 mcg/kg, which is deemed unlikely to cause adverse effects in a nursing infant (Prod Info PLIAGLIS(R) topical cream, 2012; Prod Info Synera (TM) topical patch, 2005)
    D) ANIMAL STUDIES
    1) OXYMETAZOLINE HYDROCHLORIDE/TETRACAINE HYDROCHLORIDE
    a) Following subQ administration of the oxymetazoline hydrochloride/tetracaine hydrochloride combination during the period of organogenesis through parturition and subsequent pup weaning, detectable levels of oxymetazoline, tetracaine, and the major tetracaine metabolite, p-butylaminobenzoic acid (PBBA), were present in the milk of lactating rats. The concentrations of oxymetazoline were dose dependent, with 2.5, 7, and 33.8 ng/mL of oxymetazoline corresponding to doses of 0.6, 1.5, and 7.6 times, respectively, the oxymetazoline exposure at the maximum recommended human dose (MRHD) of the combination. The concentrations of tetracaine and PBBA were generally consistent across all tetracaine dosing groups receiving 12 times the exposure as measured by PBBA at the MRHD, regardless of the oxymetazoline dose, with tetracaine concentrations ranging from 54.2 to 72.9 ng/mL and PBBA concentrations 100.5 to 131.2 ng/mL. However, animal data does not accurately predict human results (Prod Info KOVANAZE(TM) nasal spray, 2016).
    3.20.5) FERTILITY
    A) LACK OF INFORMATION
    1) BUPIVACAINE
    a) The effect of bupivacaine on fertility has not been studied (Prod Info EXPAREL(R) injection suspension, 2015).
    B) ANIMAL STUDIES
    1) LIDOCAINE
    a) Lidocaine delivered via continuous subcutaneous infusion or subcutaneous injection did not affect the overall fertility of male or female rats at doses up to 2-fold and less than one-fold higher, respectively, than the single dermal administration (based on a 1 g single dermal administration applied to 10 cm(2) for 60 minutes to a 60 kg person). Male rats, however, experienced dose-related decreases in daily sperm production, spermatogenic efficiency, and decreased homogenization-resistant sperm head count (Prod Info PLIAGLIS(R) topical cream, 2012).
    2) OXYMETAZOLINE HYDROCHLORIDE/TETRACAINE HYDROCHLORIDE
    a) In animal fertility studies, reduced percentage of mobile sperm and sperm counts were observed in rats administered subQ oxymetazoline hydrochloride at doses 2 times the oxymetazoline exposure at the maximum recommended human dose (MRHD) of the combination. No effect on male mating behavior were noted at any dose tested in this study. A reduction in the number of viable embryos occurred when female rats were administered oxymetazoline hydrochloride, alone or in combination with tetracycline hydrochloride, at doses equivalent to or greater than 0.7 times the MRHD. With oxymetazoline hydrochloride administration as monotherapy or in combination with tetracaine hydrochloride at doses 7.5 times the MRHD, the numbers of corpora lutea and implantation sites were reduced; however, when tetracaine hydrochloride was given alone, these effects were not observed. In fact, no effects on male or female fertility were observed following tetracaine hydrochloride administration to rats at doses 28 and 33 times the exposure for males and females, respectively, as measured by the major tetracaine metabolite, p-butylaminobenzoic acid, at the MRHD of the combination (Prod Info KOVANAZE(TM) nasal spray, 2016).
    3) TETRACAINE
    a) Subcutaneous administration of tetracaine to male and female rats, at doses less than one-fold higher than the single dermal administration, did not affect fertility (Prod Info PLIAGLIS(R) topical cream, 2012).

Monitoring Parameters Levels

    4.1.1) SUMMARY
    A) Methemoglobin concentrations should be checked in cyanotic patients.
    B) Obtain arterial blood gas in cases of respiratory depression, hypotension, or persistent cardiac dysrhythmias.
    C) Monitor vital signs and electrolytes.
    D) Obtain an ECG and institute continuous cardiac monitoring in symptomatic patients.
    4.1.2) SERUM/BLOOD
    A) Methemoglobin concentrations should be checked in cyanotic patients.
    B) Obtain arterial blood gas in cases of respiratory depression, hypotension, or persistent cardiac dysrhythmias.
    C) SPECIFIC AGENT
    1) Prolonged intravenous lidocaine infusion may be kept within the therapeutic serum range (2 to 4 mcg/mL) by use of a nomogram which utilizes a single 4 hour serum determination (Wong & Hurwitz, 1985).
    4.1.4) OTHER
    A) OTHER
    1) MONITORING
    a) Monitor vital signs and electrolytes.
    2) ECG
    a) Obtain an ECG and institute continuous cardiac monitoring in symptomatic patients.

Methods

    A) CHROMATOGRAPHY
    1) Local anesthetics can be qualitatively detected by thin layer chromatography.
    B) IMMUNOASSAY
    1) An EMIT(R) homogeneous enzyme immunoassay is available for quantitation of lidocaine in serum or plasma. The assay's range of quantitation is 1 to 12 mcg/mL lidocaine. Clinical studies show excellent correlation between this method and gas chromatography.
    2) Plasma bound and unbound lidocaine concentrations can be determined by Abbott TDx with a sensitivity of 0.1 mg/L and the error of the assay less than 5% (Jonville et al, 1990).
    C) SAMPLING
    1) Blood levels (plasma or serum) of lidocaine may be falsely reduced if the blood sample has been allowed to make contact with the stopper of the Vacutainer collection tube due to the plasma protein binding reduction effect of the plasticizer tris (2-butoxyethyl) phosphate (TBEP), a chemical leached from such stoppers, causing the drug to be redistributed into the erythrocytes (Stargel et al, 1979).
    D) OTHER
    1) Serum protein (alpha1-acid glycoprotein) concentrations must be considered when interpreting lidocaine levels since assays generally measure total drug concentration. Free drug levels may be relatively higher in patients with low protein concentrations (Denson et al, 1988).

Life Support

    A) Support respiratory and cardiovascular function.

Patient Disposition

    6.3.1) DISPOSITION/ORAL EXPOSURE
    6.3.1.1) ADMISSION CRITERIA/ORAL
    A) Patients with persistent cardiac dysrhythmias, mental status changes, seizures, respiratory failure, or recurrence of methemoglobinemia, despite treatment with methylene blue, should be admitted. Patients with respiratory, CNS, or cardiac toxicity should be admitted to an ICU.
    6.3.1.2) HOME CRITERIA/ORAL
    A) LIDOCAINE INGESTION: Of 28 cases of lidocaine (lignocaine) ingestion in pediatric patients (mean ingested dose was 2.7 mg/kg), only two patients developed adverse effects. The first patient was an 8-month-old boy who developed agitation, increased salivation, and difficulty with solid food after ingesting a total lidocaine dose of 4.1 mg/kg and 0.1 mg/kg of chlorhexidine. The other patient was a 7-month-old girl who vomited twice within 30 minutes after ingesting a total lidocaine dose of 3.3 mg/kg and 0.08 mg/kg of chlorhexidine. The highest lidocaine dose ingested, within the 28 cases, was 5.9 mg/kg, indicating that doses under 6 mg/kg are unlikely to cause serious adverse effects and can be managed at home (Balit et al, 2006).
    6.3.1.3) CONSULT CRITERIA/ORAL
    A) A medical toxicologist or poison control center should be consulted in cases that involve cardiac or CNS toxicity or clinically significant methemoglobinemia.
    6.3.1.5) OBSERVATION CRITERIA/ORAL
    A) Patients should be observed for 8 hours following methylene blue administration to rule out recurrence of methemoglobinemia. Patients with tachycardia only can be observed until this symptom resolves; other cardiac toxicities should be admitted.

Monitoring

    A) Methemoglobin concentrations should be checked in cyanotic patients.
    B) Obtain arterial blood gas in cases of respiratory depression, hypotension, or persistent cardiac dysrhythmias.
    C) Monitor vital signs and electrolytes.
    D) Obtain an ECG and institute continuous cardiac monitoring in symptomatic patients.

Oral Exposure

    6.5.1) PREVENTION OF ABSORPTION/PREHOSPITAL
    A) NOT RECOMMENDED
    1) No prehospital gastrointestinal decontamination is appropriate.
    6.5.2) PREVENTION OF ABSORPTION
    A) SUMMARY
    1) Activated charcoal and gastric lavage are discouraged due to the risk of aspiration in cases when oropharyngeal anesthesia is present. There is no role for whole bowel irrigation.
    6.5.3) TREATMENT
    A) MONITORING OF PATIENT
    1) Methemoglobin concentrations should be checked in cyanotic patients.
    2) Obtain arterial blood gas in cases of respiratory depression, hypotension, or persistent cardiac dysrhythmias.
    3) Monitor vital signs and electrolytes.
    4) Obtain an ECG and institute continuous cardiac monitoring in symptomatic patients.
    B) SEIZURE
    1) Attempt initial control with a benzodiazepine (diazepam or lorazepam). If seizures persist or recur administer phenobarbital. Phenytoin may worsen cardiac dysrhythmias and should be avoided.
    a) Monitor for respiratory depression, hypotension, dysrhythmias, and the need for endotracheal intubation.
    b) Evaluate for hypoglycemia (or treat with intravenous dextrose ADULT: 100 mg IV, CHILD: 2 mL/kg 25% dextrose), electrolyte disturbances, and hypoxia.
    2) DIAZEPAM
    a) ADULT DOSE: Initially 5 to 10 mg IV, OR 0.15 mg/kg IV up to 10 mg per dose up to a rate of 5 mg/minute; may be repeated every 5 to 20 minutes as needed (Brophy et al, 2012; Prod Info diazepam IM, IV injection, 2008; Manno, 2003).
    b) PEDIATRIC DOSE: 0.1 to 0.5 mg/kg IV over 2 to 5 minutes; up to a maximum of 10 mg/dose. May repeat dose every 5 to 10 minutes as needed (Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008).
    c) Monitor for hypotension, respiratory depression, and the need for endotracheal intubation. Consider a second agent if seizures persist or recur after repeated doses of diazepam .
    3) NO INTRAVENOUS ACCESS
    a) DIAZEPAM may be given rectally or intramuscularly (Manno, 2003). RECTAL DOSE: CHILD: Greater than 12 years: 0.2 mg/kg; 6 to 11 years: 0.3 mg/kg; 2 to 5 years: 0.5 mg/kg (Brophy et al, 2012).
    b) MIDAZOLAM has been used intramuscularly and intranasally, particularly in children when intravenous access has not been established. ADULT DOSE: 0.2 mg/kg IM, up to a maximum dose of 10 mg (Brophy et al, 2012). PEDIATRIC DOSE: INTRAMUSCULAR: 0.2 mg/kg IM, up to a maximum dose of 7 mg (Chamberlain et al, 1997) OR 10 mg IM (weight greater than 40 kg); 5 mg IM (weight 13 to 40 kg); INTRANASAL: 0.2 to 0.5 mg/kg up to a maximum of 10 mg/dose (Loddenkemper & Goodkin, 2011; Brophy et al, 2012). BUCCAL midazolam, 10 mg, has been used in adolescents and older children (5-years-old or more) to control seizures when intravenous access was not established (Scott et al, 1999).
    4) LORAZEPAM
    a) MAXIMUM RATE: The rate of intravenous administration of lorazepam should not exceed 2 mg/min (Brophy et al, 2012; Prod Info lorazepam IM, IV injection, 2008).
    b) ADULT DOSE: 2 to 4 mg IV initially; repeat every 5 to 10 minutes as needed, if seizures persist (Manno, 2003; Brophy et al, 2012).
    c) PEDIATRIC DOSE: 0.05 to 0.1 mg/kg IV over 2 to 5 minutes, up to a maximum of 4 mg/dose; may repeat in 5 to 15 minutes as needed, if seizures continue (Brophy et al, 2012; Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008; Sreenath et al, 2009; Chin et al, 2008).
    5) PHENOBARBITAL
    a) ADULT LOADING DOSE: 20 mg/kg IV at an infusion rate of 50 to 100 mg/minute IV. An additional 5 to 10 mg/kg dose may be given 10 minutes after loading infusion if seizures persist or recur (Brophy et al, 2012).
    b) Patients receiving high doses will require endotracheal intubation and may require vasopressor support (Brophy et al, 2012).
    c) PEDIATRIC LOADING DOSE: 20 mg/kg may be given as single or divided application (2 mg/kg/minute in children weighing less than 40 kg up to 100 mg/min in children weighing greater than 40 kg). A plasma concentration of about 20 mg/L will be achieved by this dose (Loddenkemper & Goodkin, 2011).
    d) REPEAT PEDIATRIC DOSE: Repeat doses of 5 to 20 mg/kg may be given every 15 to 20 minutes if seizures persist, with cardiorespiratory monitoring (Loddenkemper & Goodkin, 2011).
    e) MONITOR: For hypotension, respiratory depression, and the need for endotracheal intubation (Loddenkemper & Goodkin, 2011; Manno, 2003).
    f) SERUM CONCENTRATION MONITORING: Monitor serum concentrations over the next 12 to 24 hours. Therapeutic serum concentrations of phenobarbital range from 10 to 40 mcg/mL, although the optimal plasma concentration for some individuals may vary outside this range (Hvidberg & Dam, 1976; Choonara & Rane, 1990; AMA Department of Drugs, 1992).
    6) OTHER AGENTS
    a) If seizures persist after phenobarbital, propofol or pentobarbital infusion, or neuromuscular paralysis with general anesthesia (isoflurane) and continuous EEG monitoring should be considered (Manno, 2003). Other anticonvulsants can be considered (eg, valproate sodium, levetiracetam, lacosamide, topiramate) if seizures persist or recur; however, there is very little data regarding their use in toxin induced seizures, controlled trials are not available to define the optimal dosage ranges for these agents in status epilepticus (Brophy et al, 2012):
    1) VALPROATE SODIUM: ADULT DOSE: An initial dose of 20 to 40 mg/kg IV, at a rate of 3 to 6 mg/kg/minute; may give an additional dose of 20 mg/kg 10 minutes after loading infusion. PEDIATRIC DOSE: 1.5 to 3 mg/kg/minute (Brophy et al, 2012).
    2) LEVETIRACETAM: ADULT DOSE: 1000 to 3000 mg IV, at a rate of 2 to 5 mg/kg/min IV. PEDIATRIC DOSE: 20 to 60 mg/kg IV (Brophy et al, 2012; Loddenkemper & Goodkin, 2011).
    3) LACOSAMIDE: ADULT DOSE: 200 to 400 mg IV; 200 mg IV over 15 minutes (Brophy et al, 2012). PEDIATRIC DOSE: In one study, median starting doses of 1.3 mg/kg/day and maintenance doses of 4.7 mg/kg/day were used in children 8 years and older (Loddenkemper & Goodkin, 2011).
    4) TOPIRAMATE: ADULT DOSE: 200 to 400 mg nasogastric/orally OR 300 to 1600 mg/day orally divided in 2 to 4 times daily (Brophy et al, 2012).
    7) PHENYTOIN
    a) Phenytoin may worsen or precipitate cardiac arrhythmias from local anesthetics and should be avoided (Wood, 1971).
    C) FAT EMULSION
    1) Intravenous lipid emulsion (ILE) has been effective in reversing severe cardiovascular toxicity from local anesthetic overdose in animal studies and human case reports. Several animal studies and human case reports have also evaluated the use of ILE for patients following exposure to other drugs. Although the results of these studies are mixed, there is increasing evidence that it can rapidly reverse cardiovascular toxicity and improve mental function for a wide variety of lipid soluble drugs. It may be reasonable to consider ILE in patients with severe symptoms who are failing standard resuscitative measures (Lavonas et al, 2015).
    2) The American College of Medical Toxicology has issued the following guidelines for lipid resuscitation therapy (LRT) in the management of overdose in cases involving a highly lipid soluble xenobiotic where the patient is hemodynamically unstable, unresponsive to standard resuscitation measures (ie, fluid replacement, inotropes and pressors). The decision to use LRT is based on the judgement of the treating physician. When possible, it is recommended these therapies be administered with the consultation of a medical toxicologist (American College of Medical Toxicology, 2016; American College of Medical Toxicology, 2011):
    a) Initial intravenous bolus of 1.5 mL/kg 20% lipid emulsion (eg, Intralipid) over 2 to 3 minutes. Asystolic patients or patients with pulseless electrical activity may have a repeat dose, if there is no response to the initial bolus.
    b) Follow with an intravenous infusion of 0.25 mL/kg/min of 20% lipid emulsion (eg, Intralipid). Evaluate the patient's response after 3 minutes at this infusion rate. The infusion rate may be decreased to 0.025 mL/kg/min (ie, 1/10 the initial rate) in patients with a significant response. This recommendation has been proposed because of possible adverse effects from very high cumulative rates of lipid infusion. Monitor blood pressure, heart rate, and other hemodynamic parameters every 15 minutes during the infusion.
    c) If there is an initial response to the bolus followed by the re-emergence of hemodynamic instability during the lowest-dose infusion, the infusion rate may be increased back to 0.25 mL/kg/min or, in severe cases, the bolus could be repeated. A maximum dose of 10 mL/kg has been recommended by some sources.
    d) Where possible, LRT should be terminated after 1 hour or less, if the patient's clinical status permits. In cases where the patient's stability is dependent on continued lipid infusion, longer treatment may be appropriate.
    3) MECHANISM OF ACTION
    a) In vitro studies have suggested that administration of a lipid emulsion infusion to treat toxicity of highly lipophilic drugs may create a 'lipid sink' where the lipophilic drug is removed from the tissues by partitioning into a plasma lipid phase that was created by the infusion. This is evidenced by an in vitro study that compared the effects of 1% lipid emulsion with a standard buffer after inducing asystole into isolated rats hearts following administration of bupivacaine. The rat hearts that received the lipid emulsion returned to spontaneous contractions more rapidly (average time to first heart beat 44.6 +/- 3.5 seconds vs 63.8 +/- 4.3 seconds in controls), with complete cardiac function. The lipid-treated hearts also showed a more rapid decline in the myocardial bupivacaine content (Leskiw & Weinberg, 2009; Weinberg, 2006; Weinberg et al, 2006).
    b) However, lipid rescue performed in vivo, particularly with bupivacaine-induced cardiac toxicity, appears to occur more rapidly than could be explained with the in vitro method of extracting a highly lipophilic drug across several tissue diffusion barriers into equilibrium with a plasma phase, indicating that there may be other alternative mechanisms of action. One such mechanism involves reversal of mitochondrial fatty acid transport inhibition.
    1) It is believed that bupivacaine inhibits carnitine acylcarnitine translocase (CACT), an enzyme used in mitochondrial fatty acid metabolism and transport. Fatty acids are responsible for approximately 80% to 90% of cardiac ATP synthesis. Inhibition of CACT, and ultimately the rapid loss of ATP synthesis, could contribute to cardiac toxicity. It is suggested that lipid infusion may increase the intracellular fatty acid content sufficiently to reverse the bupivacaine-induced inhibition of CACT (Weinberg, 2006).
    4) EFFICACY
    a) In a systemic review of the effect of ILE therapy for local anesthetic toxicity, it was determined that ILE was effective in reversing cardiovascular or neurological features in some patients with local anesthetic overdose; however, the information was limited from very low quality studies (n=113; 76 human studies, including 2 randomized control trials and 74 case reports or case series; 38 animal studies, including 29 randomized control studies, 3 observational studies, and 6 case reports or case series) and the authors could not determine if ILE therapy was more effective than vasopressors (Hoegberg et al, 2016).
    5) CONTRAINDICATION
    a) Propofol (which contains 10% lipids) should not be used in local anesthetic-induced cardiac toxicity. The large doses necessary for lipid rescue would actually result in propofol overdoses. In addition, propofol can lower blood pressure and is considered a mitochondrial poison (Felice & Schumann, 2008; Weinberg, 2006).
    6) CASE REPORTS
    a) ELDERLY
    1) A 75-year-old woman received 20 mL of 0.5 percent levobupivacaine for a lumbar plexus block. Within seconds she became unresponsive, developed tonic clonic seizures and became hypotensive (60/40 mmHg) with low voltage widened QRS complexes on ECG. She was treated with intravenous metaraminol, endotracheal intubation and 100 mL of intravenous 20% intralipid. Hypotension rapidly resolved, the QRS morphology normalized and her surgery continued uneventfully (Foxall et al, 2007).
    2) An 84-year-old woman developed generalized seizures followed by asystolic cardiac arrest after an inadvertent injection of 40 mL of ropivacaine 1% as part of an axillary plexus nerve block. Following 10 minutes of unsuccessful cardiopulmonary resuscitation, a bolus dose of 100 mL of 20% lipid emulsion was administered followed by a continuous infusion of 10 mL/min. After a total dose of 200 mL had been administered, cardiac function was restored and the patient completely recovered (Litz et al, 2006).
    3) An 83-year-old man developed asystolic cardiac arrest after receiving 26 mL of bupivacaine 0.5% preoperatively as a sciatic nerve block for post-operative pain management following a total knee arthroplasty. After initially administering chest compressions, the patient received a 250 mL (3 mL/kg) bolus dose of 20% lipid emulsion, intravenously administered over a 2-minute period, followed by a continuous infusion of 0.2 mL/kg/minute. Within 10 minutes, the ECG indicated sinus tachycardia with palpable pulses, and within 90 minutes, the patient was awake and responsive (Smith et al, 2008).
    4) An 82-year-old woman received 30 mL of 0.5% ropivacaine with epinephrine for a femoral nerve block, followed by 30 ml of 0.5% bupivacaine with epinephrine for a sciatic nerve block. About 20 seconds after completion of the sciatic block, she had a tonic clonic seizure. She was treated with 3 mg midazolam and bag valve mask ventilation. She had a second seizure about 60 seconds later and then developed ventricular tachycardia at a rate of 200 beats/minute with palpable femoral and carotid pulses. She received 150 mg amiodarone followed by 100 mL of 20% Intralipid. She then became hypotensive and received a synchronized countershock at 120 joules. She converted to sinus rhythm, and an additional 400 mL 20% Intralipid was infused over 15 minutes. She remained stable thereafter, with a normal mental status by 2 hours after the event (McCutchen & Gerancher, 2008).
    5) An elderly woman received 20 mL of 0.5% bupivacaine for a sciatic nerve block, followed by 2.2 mL of 0.75% bupivacaine for spinal anesthesia, and then 20 mL of 0.5% bupivacaine with epinephrine for a lumbar plexus block. Three minutes after the lumbar plexus block, she became non-verbal and had a seizure. She received 1.5 mL/kg (100 mL) of Intralipid within 2 minutes and seizure activity stopped; she regained consciousness within 3 to 4 minutes (Whiteside, 2008).
    6) A 91-year-old man received 30 mL of 1% mepivacaine and 10 mL of 1% prilocaine pre-operatively. Five minutes later the patient experienced dizziness, nausea, and agitation, and became unresponsive. He also developed supraventricular extrasystoles with intermittent bigeminy. Following an intravenous bolus dose of 1 mL/kg of 20% lipid emulsion, repeated 3 minutes later for a total amount of 100 mL, followed by a continuous infusion of 0.25 mL/kg/min (14 mL/min) for a total dose of 200 mL, the patient became responsive, with resolution of his cardiac abnormalities (Litz et al, 2008).
    b) ADULT
    1) A 58-year-old man developed cardiac arrest immediately after completion of a brachial plexus block using 20 mL of bupivacaine 0.5% and 20 mL mepivacaine 1.5%. He was not stable after 20 minutes of ACLS (intubation, defibrillation, 3 mg epinephrine, 2 mg atropine, 300 mg amiodarone, 40 units vasopressin), with dysrhythmias predominantly pulseless ventricular tachycardia and asystole. At that time he received 100 mL of 20% Intralipid through a peripheral vein, followed by defibrillation, and 1 mg each epinephrine and atropine. Sinus rhythm returned within 15 seconds (90 beats/min). An infusion of lipid emulsion was given for 2 hours at 0.5 mL/kg/min. He was extubated 2.5 hours later with no neurologic sequelae. Cardiac catheterization revealed total occlusion of the right coronary artery with a left ventricular ejection fraction of 32% (Rosenblatt et al, 2006).
    2) A 60-year-old man received 30 mL of 1.5% mepivacaine with epinephrine and 10 mL of 0.5% bupivacaine for a brachial plexus block. Five minutes later he developed labored respirations, followed by apnea and unresponsiveness. He became pulseless, and received CPR, atropine 1 mg, epinephrine 3 mg total, vasopressin 40 units, 100 mL of 8.4% sodium bicarbonate, magnesium sulfate 6 g, and 11 successive defibrillations without return of sustained cardiac activity. He received 250 mL of 20% lipid emulsion over 30 minutes beginning 10 minutes after CPR was started. Hemodynamic stability was eventually achieved and he ultimately recovered. There was no evidence of myocardial infarction or pulmonary embolism (Warren et al, 2008).
    3) A 36-year-old man developed perioral paresthesia, headache, dizziness, light-headedness, diplopia, tachycardia (153 bpm) and hypertension (180/110 mmHg) approximately 60 seconds after injection of 10 mL of 1% lignocaine and 20 mL of 0.5% bupivacaine into the muscles of his lower right leg. An ECG also showed ST segment depression. Within 5 minutes, the patient received 2 100-mL 20% lipid emulsion IV boluses, approximately 1 to 3 minutes apart, with subsequent improvement in his hemodynamic status and complete resolution of his headache, diplopia, and light-headedness. The patient also received an additional 100 mL of 20% lipid emulsion, infused over an hour, with a repeat ECG indicating complete reversal of the ST segment pattern (Espinet & Emmerton, 2009).
    4) A 57-year-old man experienced mental status changes, tremor and increasing somnolence and confusion, and developed progressive QRS widening with subsequent cardiac arrest after receiving 3 60-mg IV lidocaine bolus doses followed by a continuous lidocaine infusion, initially 1 mg/min and increased to 2 mg/min, for treatment of recurring ventricular tachycardia. A lidocaine level, obtained due to suspected lidocaine toxicity, was 7.6 mcg/mL (therapeutic range 2 to 5 mcg/mL) Despite aggressive resuscitative measures, following the cardiac arrest, the patient showed no significant clinical improvement. Approximately 55 minutes after beginning resuscitation, 20% lipid emulsion was administered as a 1 mL/kg bolus for 1 minute followed by a continuous infusion at 0.25 mL/kg/min for 30 minutes. Approximately 5 minutes later, the patient returned to normal sinus rhythm with narrowing of the QRS complex. Repeat lidocaine levels, obtained 25 minutes and 55 minutes after the infusion, were 3.5 mcg/mL and 3 mcg/mL, respectively (Dix et al, 2011).
    5) A 24-year-old man, undergoing surgical repair of a fractured clavicle, received a brachial plexus block with 40 mL 0.5% ropivacaine. Following surgery, the patient was restless and incoherent with slurred speech. Twitching of his limbs was also noted. Suspecting ropivacaine toxicity, 100 mL 20% lipid emulsion was administered over a 10-minute period. Within minutes, the patient recovered without sequelae. Plasma concentrations of total and unbound ropivacaine, obtained 2 hours and 20 minutes after ropivacaine administration but prior to lipid administration, were 1.99 and 0.13 mcg/mL, respectively. Following lipid emulsion administration, the total and unbound plasma ropivacaine concentrations decreased to 1.72 and 0.05 mcg/mL, respectively (Mizutani et al, 2011).
    c) ADOLESCENT
    1) A 13-year-old girl received a lumbar plexus block with 20 mL of a solution that was equal volumes of 1% lidocaine with epinephrine and 0.75% ropivacaine. After 15 minutes she developed ventricular tachycardia with hypertension (120/92 mmHg) and oxygen saturation decreased to 92%. She was treated with 150 mL (3 mL/kg) of a 20% lipid emulsion and within 2 minutes QRS complexes normalized (with ST depression), hypertension resolved, and oxygen saturation increased to 97%. After 30 minutes ST depression resolved and surgery proceeded uneventfully (Ludot et al, 2008).
    2) A 19-year-old patient developed dysarthria and progressive myoclonus of the upper extremities after unintentionally receiving 50 mL of 2% mepivacaine (1000 mg) instead of the prescribed 50 mL of 1% mepivacaine (500 mg) preoperatively for wrist surgery. Following intravenous administration of 100 mL of 20% lipid emulsion, the patient's symptoms resolved and the surgery was performed uneventfully (Charbonneau et al, 2009).
    3) A 17-year-old adolescent experienced seizures and became pulseless after receiving 20 mL of 0.5% bupivacaine for postoperative analgesia. Within 1 to 2 minutes after seizure onset, intravenous midazolam 3 mg followed by 20% lipid emulsion was administered. He developed ventricular fibrillation, and was successfully cardioverted to a narrow complex sinus tachycardia. During the entire resuscitation period, a total of 500 mL 20% lipid emulsion (approximately 8 mL/kg) was administered. The patient's cardiac status stabilized and daily follow-up for 1 week indicated no evidence of sequelae (Markowitz & Neal, 2009).
    d) PEDIATRIC
    1) INFANT: A 40-day-old infant developed tachycardia (baseline of 140 bpm increased to 170 bpm), hypotension (baseline of 75/35 mmHg decreased to 31/19 mmHg), and ECG abnormalities, including ST segment elevation and inverted T waves, after receiving bupivacaine with epinephrine as a caudal epidural block, in preparation for a right inguinal hernia repair. Despite intravenous administration of epinephrine 2 mcg/kg and 20 mL of 5% albumin, the patient's ECG abnormalities persisted. He was then given 10 mL of 20% lipid emulsion (Intralipid(R)) intravenously over 1 to 2 minutes, resulting in hemodynamic stability and resolution of ECG changes. The patient then underwent surgery and recovered uneventfully without sequelae (Shah et al, 2009).
    2) INFANT: A 3-month-old child developed generalized tonic-clonic seizures and ventricular dysrhythmias within minutes after receiving subcutaneous administration of 5 mL of bupivacaine 0.5% (4 mg/kg) and 5 mL of lignocaine 2% following a surgical procedure. Suspecting bupivacaine toxicity, the patient was given a 9 mL IV bolus of 20% lipid emulsion, followed by an infusion of 0.25 mL/kg/min. Following transfer to the ICU, the patient continued to have seizures refractory to IV midazolam and phenytoin, but controlled with 2 doses of diazepam. Following the lipid infusion, 4 hours post-admission to the ICU, the patient's heart rate and rhythm normalized and his neurologic status stabilized, with no further seizures reported (Admani & Essajee, 2010).
    3) INFANT: Ventricular tachycardia occurred in an 11-month-old, 8 kg infant who received 8 mL (1 mL/kg) of 0.5% bupivacaine prior to undergoing a circumcision. Following administration of 20% intralipid at an IV bolus dose of 12 mL (1.5 mL/kg), followed by an infusion at a dose of 2 mL/min (0.25 mL/kg/min), the patient recovered uneventfully (Buck et al, 2014).
    e) PREGNANT
    1) An 18-year-old pregnant woman (38 weeks gestational age) received 10 mL of 0.5% bupivacaine for epidural anesthesia. Within 90 seconds she became restless and agitated, followed by twitching then unresponsiveness. Aspiration of the epidural catheter revealed venous blood. She received two 50 mL boluses of 20% Intralipid, and then 100 mL as an infusion. Within 30 seconds she regained consciousness. She recovered completely, and both mother and neonate were discharged on postoperative day 4 (Spence, 2007).
    7) ANIMAL STUDY
    a) In a dog model of severe bupivacaine overdose, cardiovascular collapse occurred following a single 10 mg/kg IV bolus of bupivacaine. Three of 9 dogs were treated immediately with infusion of a 20% lipid emulsion and cardiac massage, while in 6 the infusion of 20% lipid emulsion was begun 10 minutes after cardiovascular collapse. Normal hemodynamics returned in all 9 dogs, without electrical cardioversion. The authors' recommended dose to treat bupivacaine-associated cardiac arrest was 1 mL/kg intravenous injection of 20% lipid emulsion as a bolus, followed by an infusion of 0.25 mL/kg/min for 10 minutes, while continuing life support measures. Bolus may be repeated every 5 minutes, for two to three doses. Patients require monitoring to avoid volume overload (Weinberg et al, 2004).
    b) Using a rat model, a study was conducted to evaluate the use of epinephrine with lipid infusion on the recovery from bupivacaine overdose. Following receipt of an IV bolus dose of bupivacaine 20 mg/kg, rats received one of six treatments (n=5 for all groups): saline, saline bolus plus 30% lipid emulsion (control), or 30% lipid emulsion plus epinephrine at 1 mcg/kg, 2.5 mcg/kg, 10 mcg/kg, or 25 mcg/kg. Initially, the addition of epinephrine resulted in a rapid return of spontaneous circulation in all animals within 5 minutes at epinephrine doses of 2.5 mcg/kg and greater; however, only 3 of the 5 rats at 10 mcg/kg, and 1 of the 5 rats at 25 mcg/kg achieved a sustained return of spontaneous circulation by 15 minutes. In contrast, the lipid control group resulted in a slower but more sustained recovery, with all animals achieving sustained return of spontaneous circulation at 10 and 15 minutes. In addition to a poor recovery at 15 minutes as compared with the lipid control, epinephrine administration at doses of 10 mcg/kg or greater resulted in increased lactate levels and worsening acidosis (Hiller et al, 2009).
    D) ACIDOSIS
    1) METABOLIC ACIDOSIS: Treat severe metabolic acidosis (pH less than 7.1) with sodium bicarbonate, 1 to 2 mEq/kg is a reasonable starting dose(Kraut & Madias, 2010). Monitor serum electrolytes and arterial or venous blood gases to guide further therapy.
    2) Respiratory acidosis should be corrected by assisted ventilation.
    E) METHEMOGLOBINEMIA
    1) SUMMARY
    a) Determine the methemoglobin concentration and evaluate the patient for clinical effects of methemoglobinemia (ie, dyspnea, headache, fatigue, CNS depression, tachycardia, metabolic acidosis). Treat patients with symptomatic methemoglobinemia with methylene blue (this usually occurs at methemoglobin concentrations above 20% to 30%, but may occur at lower methemoglobin concentrations in patients with anemia, or underlying pulmonary or cardiovascular disorders). Administer oxygen while preparing for methylene blue therapy.
    2) METHYLENE BLUE
    a) INITIAL DOSE/ADULT OR CHILD: 1 mg/kg IV over 5 to 30 minutes; a repeat dose of up to 1 mg/kg may be given 1 hour after the first dose if methemoglobin levels remain greater than 30% or if signs and symptoms persist. NOTE: Methylene blue is available as follows: 50 mg/10 mL (5 mg/mL or 0.5% solution) single-dose ampules (Prod Info PROVAYBLUE(TM) intravenous injection, 2016) and 10 mg/1 mL (1% solution) vials (Prod Info methylene blue 1% intravenous injection, 2011). REPEAT DOSES: Additional doses may be required, especially for substances with prolonged absorption, slow elimination, or those that form metabolites that produce methemoglobin. NOTE: Large doses of methylene blue may cause methemoglobinemia or hemolysis (Howland, 2006). Improvement is usually noted shortly after administration if diagnosis is correct. Consider other diagnoses or treatment options if no improvement has been observed after several doses. If intravenous access cannot be established, methylene blue may also be given by intraosseous infusion. Methylene blue should not be given by subcutaneous or intrathecal injection (Prod Info methylene blue 1% intravenous injection, 2011; Herman et al, 1999). NEONATES: DOSE: 0.3 to 1 mg/kg (Hjelt et al, 1995).
    b) CONTRAINDICATIONS: G-6-PD deficiency (methylene blue may cause hemolysis), known hypersensitivity to methylene blue, methemoglobin reductase deficiency (Shepherd & Keyes, 2004)
    c) FAILURE: Failure of methylene blue therapy suggests: inadequate dose of methylene blue, inadequate decontamination, NADPH dependent methemoglobin reductase deficiency, hemoglobin M disease, sulfhemoglobinemia, or G-6-PD deficiency. Methylene blue is reduced by methemoglobin reductase and nicotinamide adenosine dinucleotide phosphate (NADPH) to leukomethylene blue. This in turn reduces methemoglobin. Red blood cells of patients with G-6-PD deficiency do not produce enough NADPH to convert methylene blue to leukomethylene blue (do Nascimento et al, 2008).
    d) DRUG INTERACTION: Concomitant use of methylene blue with serotonergic drugs, including serotonin reuptake inhibitors (SRIs), selective serotonin reuptake inhibitors (SSRIs), serotonin and norepinephrine reuptake inhibitors (SNRIs), tricyclic antidepressants (TCAs), norepinephrine-dopamine reuptake inhibitors (NDRIs), triptans, and ergot alkaloids may increase the risk of potentially fatal serotonin syndrome (U.S. Food and Drug Administration, 2011; Stanford et al, 2010; Prod Info methylene blue 1% IV injection, 2011).
    3) TOLUIDINE BLUE OR TOLONIUM CHLORIDE (GERMANY)
    a) DOSE: 2 to 4 mg/kg intravenously over 5 minutes. Dose may be repeated in 30 minutes (Nemec, 2011; Lindenmann et al, 2006; Kiese et al, 1972).
    b) SIDE EFFECTS: Hypotension with rapid intravenous administration. Vomiting, diarrhea, excessive sweating, hypotension, dysrhythmias, hemolysis, agranulocytosis and acute renal insufficiency after overdose (Dunipace et al, 1992; Hix & Wilson, 1987; Winek et al, 1969; Teunis et al, 1970; Marquez & Todd, 1959).
    c) CONTRAINDICATIONS: G-6-PD deficiency; may cause hemolysis.
    F) BRADYCARDIA
    1) ATROPINE
    a) If symptomatic and heart rate is less than 60, consider administration of atropine.
    b) ATROPINE/DOSE
    1) 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).
    2) 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.
    a) There is no minimum dose (de Caen et al, 2015).
    b) MAXIMUM TOTAL DOSE: Children: 1 milligram; adolescents: 2 milligrams (Kleinman et al, 2010).
    2) If unresponsive, give epinephrine.
    G) HYPOTENSIVE EPISODE
    1) SUMMARY
    a) Infuse 10 to 20 milliliters/kilogram of isotonic fluid and keep the patient supine. If hypotension persists, administer dopamine or norepinephrine. Consider central venous pressure monitoring to guide further fluid therapy.
    2) DOPAMINE
    a) DOSE: Begin at 5 micrograms per kilogram per minute progressing in 5 micrograms per kilogram per minute increments as needed (Prod Info dopamine hcl, 5% dextrose IV injection, 2004). If hypotension persists, dopamine may need to be discontinued and a more potent vasoconstrictor (eg, norepinephrine) should be considered (Prod Info dopamine hcl, 5% dextrose IV injection, 2004).
    b) CAUTION: If ventricular dysrhythmias occur, decrease rate of administration (Prod Info dopamine hcl, 5% dextrose IV injection, 2004). Extravasation may cause local tissue necrosis, administration through a central venous catheter is preferred (Prod Info dopamine hcl, 5% dextrose IV injection, 2004).
    3) NOREPINEPHRINE
    a) PREPARATION: 4 milligrams (1 amp) added to 1000 milliliters of diluent provides a concentration of 4 micrograms/milliliter of norepinephrine base. Norepinephrine bitartrate should be mixed in dextrose solutions (dextrose 5% in water, dextrose 5% in saline) since dextrose-containing solutions protect against excessive oxidation and subsequent potency loss. Administration in saline alone is not recommended (Prod Info norepinephrine bitartrate injection, 2005).
    b) DOSE
    1) ADULT: Dose range: 0.1 to 0.5 microgram/kilogram/minute (eg, 70 kg adult 7 to 35 mcg/min); titrate to maintain adequate blood pressure (Peberdy et al, 2010).
    2) CHILD: Dose range: 0.1 to 2 micrograms/kilogram/minute; titrate to maintain adequate blood pressure (Kleinman et al, 2010).
    3) CAUTION: Extravasation may cause local tissue ischemia, administration by central venous catheter is advised (Peberdy et al, 2010).
    H) CARDIAC ARREST
    1) Initiate CPR immediately and administer intravenous lipids as above. Good neurological outcomes have been reported after prolonged CPR. ACLS algorithms should be applied in conjunction with lipid emulsion administration with the following modifications: if epinephrine is used, small initial doses (10 mcg to 100 mcg boluses) are recommended. Avoid vasopressin, lidocaine, calcium channel blockers and beta blockers(Neal et al, 2010). Cardiac bypass should be considered if unresponsive to the above therapies.
    I) NAUSEA AND VOMITING
    1) Nausea and vomiting should be treated with antiemetics, either metoclopramide (ADULT: 10 to 20 mg every 6 hours as needed; CHILD: 0.1 to 0.2 mg/kg every 6 hours as needed) or ondansetron (ADULT: 4 to 8 mg every 6 hours as needed; CHILD: 0.15 mg/kg every 4 hours as needed) are reasonable choices.

Eye Exposure

    6.8.1) DECONTAMINATION
    A) EYE IRRIGATION, ROUTINE: Remove contact lenses and irrigate exposed eyes with copious amounts of room temperature 0.9% saline or water for at least 15 minutes. If irritation, pain, swelling, lacrimation, or photophobia persist after 15 minutes of irrigation, an ophthalmologic examination should be performed (Peate, 2007; Naradzay & Barish, 2006).

Dermal Exposure

    6.9.1) DECONTAMINATION
    A) Wash exposed skin with soap and water.

Enhanced Elimination

    A) SUMMARY
    1) No method of enhanced elimination has proven beneficial in local anesthetic toxicity.
    B) EXCHANGE TRANSFUSION
    1) Appears to remove only minimal amounts of mepivacaine (Hillman et al, 1979).
    C) DIURESIS
    1) Forced diuresis is unlikely to be of benefit in older children and adults in whom less than 5% to 10% of a dose is excreted in the urine as unchanged drug. In infants forced diuresis was not shown to increase clearance of mepivacaine (Hillman et al, 1979).
    D) URINARY ACIDIFICATION/NOT RECOMMENDED
    1) Although acidification of the urine will enhance the excretion of local anesthetics (Meffin et al, 1973), the contribution to overall elimination is small and the risk of such therapy outweighs the benefit.
    E) HEMODIALYSIS
    1) Hemodialysis does not appear to effectively remove these agents from the plasma. Only 5.6% and 2.8% of a total therapeutic lidocaine dose administered to 2 patients was removed during 5 hours of hemodialysis. The mean dialysance was 28.6 and 26.3 mL/minute in both patients, respectively (Jacobi et al, 1983).
    F) HEMOFILTRATION
    1) In three cases, lidocaine elimination was negligible with AV hemofiltration. This method does not appear to be effective for removing lidocaine from the blood (Saima et al, 1990).

Case Reports

    A) SPECIFIC AGENT
    1) BUPIVACAINE: Long et al (1989) report that a 27-year-old woman with a traumatic injury to her hand was given 525 mg 1.5% lidocaine in an axillary block. During subsequent surgery, she was also given 75 mg 0.25% bupivacaine and within 3 minutes of injection had a generalized tonic-clonic seizure and went into ventricular fibrillation.
    a) Treatment included defibrillation, epinephrine, bretylium, methylprednisolone, and cardiac bypass. The patient recovered; no blood lidocaine or bupivacaine levels were reported (Long et al, 1989).
    2) Bupivacaine has been used in the Bier block procedure (consists of applying a tourniquet to a limb, exsanguinating it, and instilling local anesthetic intravenously in preparation for surgery). Usual bupivacaine dose is 1.5 mL/kg of 0.2% solution. Toxicity results when the tourniquet slips, allowing a bolus of local anesthetic access to the systemic vasculature.
    a) In the United Kingdom, 7 deaths due to bupivacaine toxicity following this procedure have been reported between 1979 and 1983 (Heath, 1983; Heath, 1982) .
    3) CINCHOCAINE (Dibucaine): A 12-month-old girl ingested 300 mg of cinchocaine (30 mg/kg) and 3 grams of boric acid accidentally. Seizures, vomiting, and coughing developed approximately 15 minutes postingestion. The patient was treated with forced diuresis and hemodialysis and recovered fully (Egfjord et al, 1988).
    4) CINCHOCAINE (Dibucaine): An overdose of dibucaine chloride in 2 children resulted in severe neurological and cardiovascular disturbances and adult respiratory distress syndrome (Sutter & Pfenninger, 1982). Both children recovered completely following cardiovascular resuscitation, invasive neurointensive care, and controlled ventilation with high PEEP.
    5) LIDOCAINE: A fatal overdose occurred in a 13-month-old boy following an oral lidocaine preparation. Upon admission, serum lidocaine levels were 19.5 mcg/mL, with serum MEGX levels being 6.5 mcg/mL. The patient was treated with mechanical ventilation and brain resuscitation but did not regain consciousness, and died approximately 50 days following overdose (Amitai et al, 1986).
    6) LIDOCAINE: Massive lidocaine overdosage has been successfully treated with cardiopulmonary bypass and cardiac pacing (Noble et al, 1984) as well as prolonged CPR (Selden & Burke, 1988).
    7) LIDOCAINE: A 72-year-old, 64.5 kg woman became progressively groggy and nauseated. She failed to respond to oral commands and became obtunded. Her arms then flexed and stiffened and her eyes rolled upward. These reactions occurred following the second series of subcutaneous infiltration with 20 mL of 2% lidocaine hydrochloride. The lidocaine concentration measured was 8.5 mg/L (Pelter et al, 1989).
    8) LIGNOCAINE: Two cases of successful suicide with lignocaine were described. Blood lignocaine levels postmortem were 40 and 53 mg/L (Dawling et al, 1989).
    9) PROCAINE: Hypertension, tachycardia, and mydriasis occurred in a patient given a 4000 mg IV dose of procaine while receiving succinylcholine (Wikinski et al, 1970).
    10) TAC (TETRACAINE/ADRENALINE/COCAINE): A 7.5-month-old girl died when TAC solution was applied to nasal mucous membranes resulting in excessive drug absorption. Her death was probably due to cocaine toxicity; postmortem cocaine level was 11.9 mg/L (Dailey, 1988).
    a) A 19-month-old girl developed tachycardia, agitation, and dilated pupils when TAC was applied to a wound that extended from 2 mm above the vermillion border of the upper lip to the nostril (Tripp et al, 1991).

Summary

    A) Toxicity varies by agent. LIDOCAINE:ADULT: IV injection of 800 to 2000 mg in adults has caused seizures followed by cardiac arrest. PEDIATRIC: In children, ingestion of 5 to 25 mL 2% viscous lidocaine has caused seizures. BENZOCAINE: Therapeutic doses of topical spray may cause methemoglobinemia. DIBUCAINE: PEDIATRIC: Two toddlers developed generalized seizures and severe dysrhythmias, and subsequently died after ingesting 15 mg/kg to 19 mg/kg of 1% dibucaine ointment. An 18-month-old child became comatose and developed generalized tonic-clonic seizures, but survived, after ingesting 12 mg/kg of 1% dibucaine ointment. PRAMOXINE: Ingestions up to 150 mg in children (11 mg/kg) and 250 mg in adults caused minor toxicity.
    B) THERAPEUTIC DOSE: Varies by agent. LIDOCAINE: ADULT: A single dose limit is 2 mg/kg for IV doses. PEDIATRIC: For viscous lidocaine 2% in children 3 years and older, the recommended dose is approximately 3 to 5 mg/kg swish and spit (for the mouth) or swish and swallow (for the pharynx) every 3 hours as needed, up to a MAX of 8 doses/day. In infants and children less than 3 years of age, 1.25 mL is applied topically to the area every 3 hours as needed, up to a MAX of 8 doses/day. BENZOCAINE: For treatment of mouth ulcers, benzocaine may be applied topically to the affected areas up to 4 times daily. PRAMOXINE: For hemorrhoids, apply 1% ointment or cream rectally or topically to anorectal area up to 5 times daily. ESTERS: Maximum subQ doses are: chloroprocaine 10 mg/kg; procaine 10 mg/kg; tetracaine 3 mg/kg. AMIDES: Maximum subQ doses are: bupivacaine 2 mg/kg; etidocaine 4 mg/kg; lidocaine 5 mg/kg; mepivacaine 5 mg/kg; prilocaine 8 mg/kg; ropivacaine 3 mg/kg.

Therapeutic Dose

    7.2.1) ADULT
    A) MAXIMUM ALLOWABLE SUBQ DOSE
    1) The maximum allowable subQ doses of the individual local anesthetics are as follows (Schwartz & Kaufman, 2006):
    a) Bupivacaine: 2 mg/kg
    b) Chloroprocaine: 10 mg/kg
    c) Etidocaine: 4 mg/kg
    d) Lidocaine: 5 mg/kg
    e) Mepivacaine: 5 mg/kg
    f) Prilocaine: 8 mg/kg
    g) Procaine: 10 mg/kg
    h) Ropivacaine: 3 mg/kg
    i) Tetracaine: 3 mg/kg
    B) SPECIFIC SUBSTANCE
    1) BUPIVACAINE
    a) MAXIMUM SINGLE DOSE
    1) LOCAL ANESTHESIA: 175 mg without epinephrine; 225 mg with epinephrine (AMA, 1983; Prod Info Marcaine(R), bupivacaine, 1987)
    2) EPIDURAL ANESTHESIA: Up to 150 mg in non-obstetrical patients; up to 100 mg/dose in obstetrics (Prod Info Marcaine(R), bupivacaine, 1987)
    3) DENTAL ANESTHESIA: 9 to 18 mg per site of the 0.5% solution with epinephrine. Total dose for all injection sites should not exceed 90 mg in healthy adults (Prod Info Marcaine(R), bupivacaine, 1987).
    b) CONTINUOUS EPIDURAL INFUSION
    1) OBSTETRICS: Total dose should be limited to 320 mg when used without epinephrine (AMA, 1983).
    c) MAXIMUM DAILY DOSE
    1) LOCAL ANESTHESIA: A total dose of 400 mg should not be exceeded in 24 hours (Prod Info Marcaine(R), bupivacaine, 1987).
    2) BUPIVACAINE LIPOSOME
    a) BUNIONECTOMY: The recommended dose is 106 mg (8 mL) injected slowly by infiltration into the tissues surrounding the osteotomy (7 mL) and into the subcutaneous tissue (1 mL). MAXIMUM DOSE: 266 mg (20 mL (13.3 mg/mL) undiluted suspension) (Prod Info EXPAREL(TM) injection suspension, 2011).
    b) HEMORRHOIDECTOMY: The recommended dose is 266 mg (20 mL) diluted with 10 mL saline (total 30 mL), divided into 6 5-mL aliquots, with each aliquot injected slowly by infiltration into the tissues. MAXIMUM DOSE: 266 mg (20 mL (13.3 mg/mL) undiluted suspension) (Prod Info EXPAREL(TM) injection suspension, 2011).
    c) NOTE: Bupivacaine liposome is NOT bioequivalent with other formulations of bupivacaine even if the mg dosage is the same. Dosing cannot be converted from one formulation to another and different formulations should not be substituted for one another (Prod Info EXPAREL(TM) injection suspension, 2011).
    3) CINCHOCAINE/DIBUCAINE
    a) Available as 0.5% and 1% ointment
    b) SPINAL ANESTHESIA: 2.5 to 10 mg
    c) MAXIMAL DAILY DOSE: 100 mg (Egfjord et al, 1988)
    4) DYCLONINE
    a) THERAPEUTIC DOSE (Adults and children over 2): 1 to 3 mg orally every 2 hours as needed; MOUTHRINSE: 0.5% solution as a rinse or swab for relief of discomfort associated with painful oral lesions or ulcerations or as a swallow (5 to 15 mL) for pain relief associated with esophageal lesions (Prod Info Sucrets(R) Maximum Strength Sore Throat Lozenges, 1996; Prod Info Dyclone(R), dyclonine topical solution 0.5%, 1997).
    b) MAXIMAL SINGLE DOSE: 200 mg (S Sweetman , 2001)
    5) ETIDOCAINE
    a) INFILTRATION/NERVE BLOCK: 300 mg without epinephrine; 400 mg with epinephrine (Prod Info Duranest(R) injection, etidocaine, 1997)
    b) CAUDAL EPIDURAL BLOCK: up to 300 mg (Prod Info Duranest(R) injection, etidocaine, 1997)
    6) LIDOCAINE
    a) MAXIMUM DOSE
    1) LOCAL ANESTHESIA: A maximum single dose 4.5 mg/kg (total dose of 300 mg) topically, 300 mg (4.5 mg/kg) without epinephrine by injection, and 500 mg (7 mg/kg) with epinephrine by injection has been recommended (Prod Info Xylocaine(R) Viscous topical solution, 2014; Prod Info Xylocaine(R) injection solution, 2010). Administer 0.5 mg as an intradermal injection to site 1 to 3 minutes before procedure. A second 0.5 mg dose may be administered at a new site if necessary; multiple doses to the same site are not recommended (Prod Info Zingo(TM) intradermal injection powder, 2014).
    2) ANTIARRHYTHMIC DOSE: The recommended intravenous maximum loading dose is 300 mg in one hour (AMA, 1983).
    3) ENDOTRACHEAL: Doses up to 7.7 mg/kg have resulted in therapeutic serum levels in most patients, but levels in the toxic range in others (Ward, 1983).
    4) INTRAVENOUS REGIONAL ANESTHESIA: Maximum dose is 4 mg/kg (Prod Info Xylocaine(R) injection solution, 2010).
    5) ADVANCED CARDIAC LIFE SUPPORT: The maximum recommended intravenous bolus dose is 3 mg/kg (Prod Info Xylocaine(R), lidocaine, 1986).
    6) The topical administration of lidocaine 4 mg/kg as a 4% solution in 96 children (aged 2 weeks to 12 years) resulted in a majority of lidocaine concentrations in the accepted safe range (4 to 5 mcg/mL) and occasionally lidocaine concentrations over 8 mcg/mL with no clinical evidence of toxicity.
    a) Arterial plasma levels and venous plasma levels differed only during the first 10 minutes after which the levels became more comparable (Eyres et al, 1983).
    7) CONCENTRATION: Following the administration of topical lidocaine spray anesthesia for diagnostic fiberoptic bronchoscopy, the mean peak lidocaine concentration observed was 3.6 mcg/mL at an average of 43 minutes after the start of the procedure (Labedzki et al, 1983).
    a) When epinephrine-free lidocaine is used for paracervical anesthesia the total amount injected should not exceed 4.5 mg/kg of body weight or 300 mg (Berger et al, 1974).
    8) ORAL: When normal adults received 15 mL of 2% lidocaine hydrochloride solution (300 mg) every 3 hours for 8 days washed into the oral cavity, then spit out, the peak lidocaine levels (L) were 0.08 mcg/mL, MEGX 0.05 mcg/mL, and GX 0 mcg/mL; when the same dose was washed then swallowed, peak levels were L 0.37, MEGX 0.50, and GX 0.04 after the first dose.
    a) When the same amount was swallowed, levels were L 0.47 to 0.80, MEGX 0.59 to 1.13, and GX 0.08 to 0.16. This dose did not produce toxic blood levels (Greenblatt et al, 1985).
    b) TOPICAL DOSE
    1) IRRITATION OR INFLAMMATION OF MUCOUS MEMBRANES OF MOUTH OR PHARYNX: The usual adult dose is 15 mL of 2% solution administered topically no more often than every 3 hours as needed; MAX, 8 doses/24 hours. For irritation of the mouth, the dose is swished in the mouth and spit out. For irritation of the pharynx, the dose is gargled and may be swallowed (Prod Info Xylocaine(R) Viscous topical solution, 2014).
    7) LIDOCAINE/TETRACAINE
    a) SUPERFICIAL DERMATOLOGICAL PROCEDURES: Apply sufficient amount of topical cream to cover treated area (1 to 53 g; 3 to 121 cm) to a thickness of 1 mm; based on procedure, wait 20 to 60 minutes and remove pliable peel from skin (Prod Info PLIAGLIS(R) topical cream, 2012).
    8) MEPIVACAINE
    a) MAXIMUM SINGLE DOSE
    1) The recommended single dose or total dose per procedure for healthy adults is 400 mg; however, doses of up to 7 mg/kg (550 mg) have been given without adverse reactions (Prod Info Carbocaine(R), mepivacaine HCL, 1987a).
    b) TOTAL DAILY DOSE
    1) The total dose per 24 hours should not exceed 1000 mg (Prod Info Carbocaine(R), mepivacaine HCL, 1987a).
    9) OXYBUPROCAINE
    a) A 0.4% solution is used for topical ophthalmic application (S Sweetman , 2001).
    10) PRAMOXINE
    a) Pramoxine cream for hemorrhoids should be applied up to 5 times daily, usually after bowel movements, morning and night. The cream may be instilled in the rectum via an applicator or externally applied to the affected area (Prod Info Anusol(R), 1999; Prod Info Tronolane(R), 1996). The aerosol foam may be rectally administered as 1 applicatorful 3 or 4 times daily and after bowel evacuation. Aerosol containers must not be inserted into the rectum (Prod Info Proctofoam(R), 1990).
    11) PRILOCAINE
    a) DENTAL INFILTRATION/NERVE BLOCK: Usual adult dose of prilocaine without felypressin (a vasoconstrictor) is 40 to 80 mg. The adult dose of prilocaine with felypressin (0.03 units/mL) is 30 to 150 mg (1 to 5 mL) as a 3% solution (S Sweetman , 2001).
    12) PROCAINE
    a) INFILTRATION ANESTHESIA: 0.25% to 0.5% solutions of procaine hydrochloride have been used in doses of 350 to 600 mg (S Sweetman , 2001).
    b) PERIPHERAL NERVE BLOCK: 500 mg is the usual dose, but doses up to 1 gram have been given (S Sweetman , 2001).
    13) PROPARACAINE
    a) Used as a 0.5% solution for topical anesthesia of the eye (S Sweetman , 2001).
    14) PROPOXYCAINE
    a) Used as a 0.4% solution in combination with 2% procaine (S Sweetman , 2001).
    15) TETRACAINE
    a) Available as 0.5% and 1% solutions and ointments for mucosal and ophthalmic use (S Sweetman , 2001).
    b) PROLONGED SPINAL ANESTHESIA
    1) The usual dose should not exceed 15 mg; in exceptional cases 20 mg may be used (Prod Info Pontocaine(R), tetracaine, 1987b).
    16) TETRACAINE HYDROCHLORIDE/OXYMETAZOLINE HYDROCHLORIDE
    a) INTRANASAL: 2 sprays ipsilateral (on the same side) to the maxillary tooth where the dental procedure will be performed 4 to 5 minutes apart; initiate dental procedure 10 minutes following second spray; if a test drill 10 minutes after administration of a second initial spray does not provide adequate anesthesia, may give 1 additional spray (Prod Info KOVANAZE(TM) nasal spray, 2016)
    7.2.2) PEDIATRIC
    A) SPECIFIC AGENT
    1) BUPIVACAINE
    a) CONTINUOUS EPIDURAL INFUSION: A bupivacaine dose of 0.5 mL/kg of 0.25% without epinephrine prior to surgery, followed in 30 minutes by an infusion of 0.08 mL/kg/hour was reported effective and safe for postoperative pain relief in children following major lower extremity and genitourinary surgery (Desparmet et al, 1987).
    b) CAUDAL ANALGESIA: A dose of 3 mg/kg of 0.25% bupivacaine was effective in producing caudal epidural analgesia in 45 children (Eyres et al, 1983).
    2) BUPIVACAINE LIPOSOME
    a) Safety and effectiveness in pediatric patients below 18 years of age have not been established (Prod Info EXPAREL(TM) injection suspension, 2011).
    3) DYCLONINE
    a) THERAPEUTIC DOSE (Adults and children over 2): 1 to 3 mg orally every 2 hours as needed (Prod Info Sucrets(R) Maximum Strength Sore Throat Lozenges, 1996).
    4) LIDOCAINE
    a) IV
    1) HEAD TRAUMA, ICP PROTECTION DURING ET INTUBATION OR AIRWAY MANIPULATION: 1 to 2 mg/kg IV as a single dose 30 seconds to 5 minutes prior to airway manipulation (Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008; Mower & Knopp, 2007; Zelicof-Paul et al, 2005; Butler & Jackson, 2002; Marvez-Valls et al, 2002; Nakayama et al, 1992; Bedford et al, 1980).
    2) STATUS EPILEPTICUS, REFRACTORY:
    a) LOADING DOSE: 1 to 2 mg/kg IV bolus; if initial loading dose is effective, follow immediately with continuous infusion (Hattori et al, 2008; Yildiz et al, 2008; Sugai, 2007; Hamano et al, 2006; Kobayashi et al, 1999; Mitchell, 1996).
    b) CONTINUOUS INFUSION: 2 to 4 mg/kg/hour IV (Hattori et al, 2008; Yildiz et al, 2008; Sugai, 2007; Hamano et al, 2006). Doses as high as 6 to 8 mg/kg/hour have been used (Lin et al, 2009; Mitchell, 1996).
    3) REGIONAL ANESTHESIA INDUCTION: The total dose should not exceed 3 mg/kg (Prod Info Xylocaine(R) injection solution, 2010).
    b) INTRADERMAL
    1) AGED 3 YEARS AND OLDER: For topical local anesthetic, administer 0.5 mg by intradermal injection to site 1 to 3 minutes before procedure; may repeat 1 dose at a new location following failed attempt at venous access; multiple doses to the same site are not recommended (Prod Info Zingo(TM) intradermal injection powder, 2014).
    c) ORAL
    1) TRACHEAL SPRAY: 4 mg/kg (Eyres et al, 1983)
    d) TOPICAL
    1) CHILDREN 3 YEARS OF AGE OR OLDER: Viscous lidocaine 2%: dosage should be based on age, body weight, and physical condition; eg, 5 years old 50 kg weight, MAX 75 to 100 mg (Prod Info Xylocaine(R) Viscous topical solution, 2014).
    2) CHILDREN YOUNGER THAN 3 YEARS OF AGE: Viscous lidocaine 2%: up to 1.2 mL applied topically to immediate area with cotton-tipped applicator no more often than every 3 hours as needed; maximum 4 doses/12-hr period. Risk of fatal overdose in patients under 3 years; ensure correct dose and frequency of administration (Prod Info Xylocaine(R) Viscous topical solution, 2014).
    5) EMLA (LIDOCAINE/PRILOCAINE:
    a) Less than 3 months of age or less than 5 kg: applied to maximum application area of 10 cm(2) for maximum application time of 1 hour; Maximum total dose of 1 g (Prod Info EMLA(R) topical cream, 2006)
    b) 3 months to less than 12 months of age and greater than 5 kg: applied to maximum application area of 20 cm(2) for maximum application time of 4 hours; Maximum total dose of 2 g (Prod Info EMLA(R) topical cream, 2006)
    c) 1 to 6 years of age and greater than 10 kg: applied to maximum application area of 100 cm(2) for maximum application time of 4 hours; Maximum total dose of 10 g (Prod Info EMLA(R) topical cream, 2006).
    d) 7 to 12 years of age and greater than 20 kg: applied to maximum application area of 200 cm(2) for maximum application time of 4 hours; Maximum total dose of 20 g (Prod Info EMLA(R) topical cream, 2006).
    6) LIDOCAINE/TETRACAINE
    a) Safety and effectiveness of the topical cream have not been established in pediatric patients (Prod Info PLIAGLIS(R) topical cream, 2012).
    7) MEPIVACAINE
    a) MAXIMUM SINGLE DOSE: Should not exceed 5 to 6 mg/kg in children. In children less than 3 years of age or 30 pounds, the concentration should be less than 2% (Prod Info Carbocaine(R), mepivacaine HCL, 1987a).
    8) TETRACAINE HYDROCHLORIDE/OXYMETAZOLINE HYDROCHLORIDE
    a) INTRANASAL (WEIGHING 40 KG OR MORE): 2 sprays ipsilateral (on the same side) to the maxillary tooth where the dental procedure will be performed 4 to 5 minutes apart; initiate dental procedure 10 minutes following second spray (Prod Info KOVANAZE(TM) nasal spray, 2016)

Minimum Lethal Exposure

    A) ADULT
    1) HEXYLCAINE: The lowest fatal single dose in adults reported is 15 milliliters of a 5% solution.
    2) LIDOCAINE: There have been over 5 cases of lidocaine overdose in which a 800 to 2000 mg intravenous bolus was mistakenly injected when a lower dose was ordered. There was confusion over the two types of prepackaged syringes (100 mg vs 1000 or 2000 mg). In all 5 patients, seizures occurred prior to cardiopulmonary arrest. Three survived (Bryant et al, 1983). A 76-year-old man inadvertently received a 500 mg intravenous bolus of lidocaine to treat paroxysmal ventricular dysrhythmia. He sustained tonic-clonic seizures followed by cardiac arrest from which he was resuscitated, but died 8 days later from anoxic brain injury (Kudo et al, 2004).
    3) CASE REPORT/ROPIVACAINE: A 57-year-old man, with liver dysfunction and end-stage renal disease, was admitted for a below-knee amputation and received a ropivacaine infusion (total dose 1,540 mg) postoperatively. He later developed lethargy, bradycardia, tinnitus, dysgeusia, and hallucinations. An ECG revealed widened QRS interval and prolonged QT interval without ST changes, and an echocardiogram showed an ejection fraction of 20% without wall motion abnormality. The patient's troponin-I-ES level peaked at 4.3. The patient minimally responded to administration of 2,480 mL of 20% intralipid and one session of plasmapheresis. He subsequently died following cardiac arrest (Bazerbachi et al, 2014).
    4) PEDIATRIC
    a) DIBUCAINE: Two toddlers (ages 18 months and 21 months) developed generalized tonic-clonic seizures and severe dysrhythmias (bradycardia, ventricular tachycardia, ventricular fibrillation, asystole) following ingestions (15 mg/kg to 19 mg/kg) of 1% dibucaine ointment. Despite aggressive supportive measures, both children died within hours after ingestion (Dayan et al, 1996).
    b) LIDOCAINE: A 15-year-old girl developed seizures after ingesting up to 2 grams (49 mg/kg) of viscous lidocaine. She was tachycardic (160 bpm) with a Glasgow Coma Scale of 3, and fixed and dilated pupils. An ECG revealed a QRS of 110 ms, that narrowed to 80 ms within 80 minutes after administration of IV fluids and IV sodium bicarbonate, and an arterial blood gas analysis indicated severe metabolic acidosis. A serum lidocaine concentration at 2 hours post-ingestion was 16.6 mcg/mL (reference range 1.2 to 5 mcg/mL) that decreased to 0.7 mcg/mL 6 hours post-ingestion. There was no evidence of epileptiform activity on an EEG, and a brain MRI showed hypoxic ischemic injury approximately 40 hours post-ingestion. The patient was apneic with no brainstem reflexes on hospital day 9, and brain death was subsequently declared (Drapkin et al, 2015).

Maximum Tolerated Exposure

    A) SPECIFIC SUBSTANCE
    1) BENZOCAINE
    a) From November 1997 to March 2002 there were 132 cases of definite (99 cases) or probable (33 cases) methemoglobinemia related to benzocaine use reported to the FDA. Of these, 107 were considered serious adverse events and 2 were lethal. In most cases (123; 93%) the product was a spray. In the 69 cases that specified a dose, 37 (53.6%) involved use of a single spray, which is approximately the recommended amount(Moore et al, 2004).
    2) BUPIVACAINE
    a) CASE REPORT/ADULT: Following the accidental administration of 337.5 mg bupivacaine and 180 mcg sufentanil epidurally in less than 30 minutes, a 62-year-old patient became unconscious, suffered severe hypotension, moderate bradycardia, and extensive sensory and motor block. Despite the high dose only mild respiratory depression (12 breaths/min) occurred. No arrhythmias or signs of central nervous system toxicity occurred and the patient recovered (Wolff et al, 1992).
    b) CASE REPORT/INFANT: Ventricular tachycardia occurred in an 11-month-old, 8 kg infant who received 8 mL (1 mL/kg) of 0.5% bupivacaine prior to undergoing a circumcision. Following administration of 20% intralipid at an IV bolus dose of 12 mL, followed by an infusion at a dose of 2 mL/min (0.25 mL/kg/min), the patient recovered uneventfully (Buck et al, 2014).
    3) DIBUCAINE
    a) CASE REPORT/INFANT: An 18-month-old infant became comatose with a Glasgow Coma Scale score of 3 following ingestion of approximately 150 mg (12.5 mg/kg) of a sunburn ointment containing 1% dibucaine. The patient also experienced generalized tonic-clonic seizures and ECG abnormalities, including AV nodal blockade with widening of the QRS complex. With supportive care, the patient recovered without sequelae (Nelsen et al, 2009).
    4) DYCLONINE
    a) MAXIMUM TOLERATED EXPOSURE: Adults given 300 to 600 mg daily for up to 12 weeks developed no adverse effects (FDA, 1982).
    b) Systemic effects in dogs were first noticeable after 1 mg/kg intravenously (mild-lowered blood pressure: 10 millimeters of mercury), and progressed to respiratory failure at 5 mg/kg and death at 10 mg/kg (FDA, 1982).
    c) LOCAL EFFECTS: Mucosal irritation and sloughing has occurred with use of concentrated solutions (2% or more) (FDA, 1982).
    5) HEXYLCAINE
    a) The highest known single dose with survival in a child was observed in a 2 year, 8 month boy given a glottis spray of an adult dose of 3.5 mL 5% hexylcaine spray. In 5 minutes his pupils dilated, apnea developed, generalized seizures (response to intravenous thiopental) were seen, and erythematous areas appeared on the arms, legs, and chest (Goldberg & Goodman, 1957).
    b) Trochlear nerve palsy followed spinal anesthesia with hexylcaine hydrochloride 2 mL (50 mg) in 10% dextrose (Ledbetter & Elliott, 1963).
    c) Ingestion of 20 mL of hexylcaine led to apnea, stupor, and seizures. The patient responded to symptomatic treatment (Spellberg, 1959).
    d) High doses may produce methemoglobinemia and excitatory or depressant central nervous system effects responsive to symptomatic therapy (Windholz, 1983).
    6) LIDOCAINE
    a) INTRAVENOUS
    1) An accidental injection of a 1 gram bolus of lidocaine intravenously resulted in asystole apnea and multiple grand mal seizures which responded to intravenous epinephrine, isoproterenol, dopamine and diazepam (Finkelstein & Kreeft, 1979).
    2) There have been over 5 case reports of lidocaine overdose in which a 800 to 2000 mg intravenous bolus of lidocaine was mistakenly injected when a lower dose was ordered. There was confusion over the two types of prepackaged syringes (100 mg and 1000 to 2000 mg). In all five patients seizures occurred and cardiopulmonary arrest. Three survived (Bryant et al, 1983).
    3) Unresponsiveness, respiratory arrest, marked vasodilation, seizures, and hypotension occurred in an infant following accidental intravenous administration of 12 mg/kg of lidocaine. Recovery was complete with supportive care (Jonville et al, 1990).
    b) ORAL
    1) Ingestion of 5 to 25 milliliters of a 2% viscous Xylocaine (lidocaine) solution has resulted in seizures in children (Sakai & Lattin, 1980; Rothstein et al, 1982).
    2) Of 28 cases of lidocaine (lignocaine) ingestion in pediatric patients (mean ingested dose was 2.7 mg/kg), only two patients developed adverse effects. The first patient was an 8-month-old boy who developed agitation, increased salivation, and difficulty with solid food after ingesting a total lidocaine dose of 4.1 mg/kg and 0.1 mg/kg of chlorhexidine. The other patient was a 7-month-old girl who vomited twice within 30 minutes after ingesting a total lidocaine dose of 3.3 mg/kg and 0.08 mg/kg of chlorhexidine. The highest lidocaine dose ingested, within the 28 cases, was 5.9 mg/kg, indicating that doses under 6 mg/kg are unlikely to cause serious adverse effects and can be managed at home (Balit et al, 2006).
    c) SUBCUTANEOUS INFILTRATION
    1) Infants less than 1 square meter in body surface area were more likely to develop elevated serum lidocaine concentrations following subcutaneous infiltration prior to cardiac catheterization in a study of 10 children (Palmisano et al, 1991).
    2) CASE REPORT: A 37-year-old woman experienced agitation, aggressiveness, and a sensation of a near-death experience following infiltration of 0.1% lidocaine subcutaneously in her abdomen prior to liposuction. Total lidocaine dose was 1 g (13 mg/kg). Laboratory data revealed a lidocaine concentration of 9.7 mcg/mL (therapeutic, 1 to 5 mcg/mL). Following supportive therapy, the patient recovered and was discharged the next day without sequelae (Hines et al, 2015).
    d) TOPICAL
    1) Delayed hypotension and tachycardia with cardiovascular collapse and adult respiratory syndrome has been reported following the administration of less than 30 milliliters of 1% lidocaine solution for topical anesthesia prior to fiberoptic bronchoscopy (Promisloff & Dupont, 1983).
    2) An 11-month-old male infant developed seizures and a plasma lidocaine concentration of 10 micrograms/milliliter, following the application of 2% viscous lidocaine to his gums 5 to 6 times daily for a week (Mofenson et al, 1983).
    7) MEPIVACAINE
    a) Hypotension, seizures, and death have occurred following an intravenous or paracervical dose of 200 mg (Grimes & Cates, 1976) and after a subcutaneous dose of 3000 mg (Sunshine & Fike, 1964).
    8) PRAMOXINE
    a) In a retrospective poison center study of 177 patients who ingested topical pramoxine-containing products, only minor effects were reported. The largest amounts reported resulted only in vomiting; 250 mg in a 2-year-old (11 mg/kg) and 250 mg in an adult (Spiller et al, 2006).
    9) PRILOCAINE
    a) SUMMARY: Methemoglobinemia is dose-related (correlating best with dose/m(3) BSA), occurs universally in adults given 900 mg or more subcutaneously or epidurally. It is more common with doses of greater than 8 mg/kg, but idiosyncratic cases of 6 to 8 mg/kg have been reported (Lloyd, 1992; Kreutz & Kinni, 1983; Duncan & Kobrinsky, 1983; Scott et al, 1964; Spoerel et al, 1967).
    b) STUDY: The average methemoglobin level was 6.2% after 600 mg and 12.6% after 900 mg epidurally in 20 patients (Spoerel et al, 1967).
    c) CASE REPORTS: The dose and peak methemoglobin (MeHg) level in various reports was as follows:
    PRILOCAINE-INDUCED METHEMOGLOBINEMIA
    AGEDOSEMeHgONSETREF
    neonate6 mg/kg21%75 minLloyd, 1992
    6 weeks6.7 mg/kg35.7%60 minDuncan, 1983
    5 years52 mg32.2%50 minLudwig, 1981
    5 years24 mg/kg41.5%20-30 minKlos, 1985
    19 years7.85 mg/kg37.8%4-4.5 hrsKreutz, 1983
    19 years9.3 mg/kg9.9%90 minBardoczky, 1990

    10) ROPIVACAINE
    a) CASE REPORT: Hypotension (90 to 100/50 mmHg) occurred in a 26-year-old pregnant woman who inadvertently received epidural administration of ropivacaine, 111 mL/hour instead of 11 mL/hour (a 10-fold dosing error), during labor. The total dose of ropivacaine received during a 38-minute period was 140 mg (Thyen et al, 2010).
    b) CASE REPORT/INFANT: A 5-week-old preterm infant, born at gestational age of 36 weeks and scheduled for inguinal hernia repair, inadvertently received ropivacaine at a dose of 10 mg/kg, injected into the caudal space. The total ropivacaine dose received was 40 mg instead of the prescribed 8 mg. Within minutes, the patient developed bradycardia (50 bpm), widening of QRS complexes, elevated T waves, and hypotension (50/30 mmHg). With external chest compressions, administration of IV fluids and epinephrine, and controlled ventilation and sedation, the patient recovered and was discharged home 2 days later without neurologic sequelae (Hubler et al, 2010).

Serum Plasma Blood Concentrations

    7.5.2) TOXIC CONCENTRATIONS
    A) TOXIC CONCENTRATION LEVELS
    1) SPECIFIC SUBSTANCE
    a) BUPIVACAINE
    1) Therapeutic plasma concentrations are less than 3 micrograms/milliliter.
    2) Toxic effects have occurred with levels as low as 1.1 micrograms/milliliter (Hasselstrom & Morgensen, 1984).
    3) Bupivacaine is more toxic than lidocaine or etidocaine when given intravenously (Scott, 1975).
    4) Toxic effects (seizures) in PEDIATRIC patients have occurred at serum bupivacaine levels above 5.4 micrograms/milliliter (McCloskey et al, 1992; Agarwal et al, 1992).
    5) In general, serum bupivacaine levels above 4.0 micrograms/milliliter have been associated with toxic effects in ADULTS (Agarwal et al, 1992).
    6) Accidental intravenous administration of levobupivacaine (142.5 mg) resulted in disorientation, drowsiness, slurred speech, and agitation, with serum levobupivacaine levels of 2.7 mcg/mL and 1.1 mcg/mL drawn approximately 14 minutes and 120 minutes, respectively, after cessation of the injection (Kopacz & Allen, 1999).
    7) A 37-year-old man, undergoing shoulder surgery, developed bradycardia, seizures, and cardiac arrest immediately after receiving bupivacaine as a nerve block. Despite undergoing cardiopulmonary resuscitation for approximately 2 hours and being placed on cardiopulmonary bypass, the patient died approximately 7 hours post-injection. Post-mortem toxicology results revealed cardiac blood bupivacaine concentration of 1.1 mcg/mL (Dudley et al, 2011).
    b) DIBUCAINE
    1) A 12-month-old infant developed plasma dibucaine chloride concentration of 71 nanograms/milliliter at 6 hours following an ingestion of 300 milligrams of dibucaine chloride and 3 grams of boric acid (Egfjord et al, 1988).
    c) ETIDOCAINE
    1) Peak plasma concentrations following therapeutic doses range from 0.5 to 1.5 micrograms/milliliter.
    d) LIDOCAINE
    1) Therapeutic plasma concentrations range from 1.5 to 5 micrograms/milliliter.
    2) Mild/subjective toxicity may occur with therapeutic drug levels. Plasma concentrations of 5 to 10 micrograms/milliliter are associated with marked CNS toxicity (Fruncillo et al, 1982; Rothstein et al, 1982).
    3) Drug levels are usually above 15 micrograms/milliliter in fatal cases (Brown & Skiendzielowski, 1980; Mather & Cousins, 1979).
    4) Hepatic lidocaine concentrations in patients whose death is ascribed to lidocaine ranged from 15 to 71 milligrams/kilogram, whereas in those whose death was not attributed to lidocaine levels ranged from 0 to 14 milligrams/kilogram (Peat et al, 1985).
    5) The serum lidocaine concentration was 8.5 milligrams/liter following subcutaneous infiltration of 40 milliliters of 2 percent lidocaine at two sites in a 72-year-old woman (Pelter et al, 1989).
    6) Elevated serum lidocaine concentrations (7.2 and 6.8 micrograms/milliliter) developed in 2 infants following subcutaneous infiltration of lidocaine (47 and 25 milligrams/kilogram) prior to cardiac catheterization (Palmisano et al, 1991).
    7) Lidocaine plasma concentrations at 1.2 and 3.5 hours following intravenous administration of 12 milligrams/kilogram of lidocaine in an infant were 3.8 and 1.5 milligrams/liter, respectively (Jonville et al, 1990).
    8) Dizziness, light-headedness, and confusion were the only reported effects in a 30-year-old woman with a lidocaine blood level of 2.7 mg/mL following topical application of 5 grams of 40% lidocaine (Goodwin & McMeekin, 1999).
    9) A CSF lidocaine level of 1 mg/L, corresponding to a serum lidocaine level of 12.5 to 16 mg/L, was reported in a 23-year-old man following topical application of EMLA cream, containing 2.5% of lidocaine and 2.5% of prilocaine, to his chest, abdomen, and back (Brosh-Nissimov et al, 2004)
    10) Toxicity was observed in a 22-year-old man during daily use of 240 mL viscous lidocaine for a severe tongue ulcer. His serum lidocaine concentration was 6.7 mcg/mL (Yamashita et al, 2002).
    e) LIGNOCAINE
    1) Two cases of successful suicide with lignocaine were described by Dawling et al (1989). Blood lignocaine levels postmortem were 40 and 53 milligrams/liter.
    f) MEPIVACAINE
    1) Therapeutic plasma levels are less than 5 micrograms/milliliter (Meffin et al, 1973; Shnider et al, 1968).
    2) Toxicity may occur at levels of 4 micrograms/milliliter or less.
    3) Fatalities are usually associated with levels above 10 micrograms/milliliter (Dodson et al, 1975; Sinclair et al, 1965).
    g) PROCAINE
    1) Peak plasma concentrations after therapeutic doses range from 3 to 11 micrograms/milliliter.
    2) Levels above 20 micrograms/milliliter are associated with marked clinical toxicity (Usubiaga et al, 1966).
    h) ROPIVACAINE
    1) CASE REPORT: A 25-year-old woman developed numbness of her lip, lost consciousness, and experienced a generalized seizure within 13 minutes of receiving a 300-mg dose injection (6.25 mg/kg) of ropivacaine as an axillary brachial plexus block for preparation of surgery on her finger. Following supportive care, the patient recovered without sequelae. Laboratory analysis of a blood sample, obtained 28 minutes post-injection, revealed a plasma ropivacaine concentration of 3.65 mcg/mL (Kimura et al, 2007).
    2) CASE REPORT: A 25-year-old man developed isolated sinus tachycardia (130 bpm) and tonic-clonic seizures approximately 1 minute following unintentional intravascular administration of 10 mL (75 mg) of ropivacaine for a sciatic nerve block. Plasma ropivacaine concentrations, obtained 15, 90, and 150 minutes after onset of symptoms, were 13.1, 11.6, and 3.6 mcmol/L, respectively (Petitjeans et al, 2002).
    3) CASE REPORT/INFANT: A 5-week-old preterm infant, born at gestational age of 36 weeks and scheduled for inguinal hernia repair, inadvertently received ropivacaine at a dose of 10 mg/kg, injected into the caudal space. The total ropivacaine dose received was 40 mg instead of the prescribed 8 mg. Within minutes, the patient developed bradycardia (50 bpm), widening of QRS complexes, elevated T waves, and hypotension (50/30 mmHg). Serum ropivacaine concentration, obtained 20 minutes following administration, was 6 mcg/mL (Hubler et al, 2010).

Pharmacologic Mechanism

    A) Local anesthetics produce a conduction block at the cell membrane of the involved nerve. All nerves (sensory, motor, and autonomic) are affected but small diameter pain/temperature (AS fibers) and autonomic ones (B and C fibers) are most susceptible. This conduction block is eventually reversible and is produced by a transient increase in sodium permeability which would normally produce the action potential and permit its propagation. Anesthetics prevent the increase of sodium pump permeability which occurs during normal impulse transmission. They also interfere with permeability of potassium in the resting nerve. The permeability of both sodium and potassium are calcium dependent and it is believed that local anesthetics compete with calcium for membrane binding sites.
    B) Local anesthetics also have intrinsic vasodilator activity.
    C) Lidocaine depresses spontaneous depolarization (phase 4 of the action potential) and shortens the action potential and refractory period of cardiac muscle and Purkinje (conducting system) cells.
    D) Sympathetic blockade from spinal or epidural anesthesia causes decreased cardiac output and vasodilatation with subsequent hypotension. Their relative potency in this regard is bupivacaine>/=etidocaine>/=lidocaine>/=mepivacaine>/= prilocaine.
    E) Dyclonine has a different chemical structure than "caine" type anesthetics. Hypotension is due to reduced cardiac output and peripheral vasodilation. At high doses, respiratory stimulation, followed by respiratory depression occurs. Unlike other local anesthetics, dyclonine has anticonvulsant activity (FDA, 1982).
    F) LEVOBUPIVACAINE (the S(-) isomer of bupivacaine) and ROPIVACAINE (the S(-) isomer of the propyl homolog of bupivacaine) may be less toxic than bupivacaine itself (a 50:50 racemic mixture of the stereoisomers). Ropivacaine has been called the safest long-acting local anesthetic for clinical use, because of its higher tolerated dose and unbound plasma concentration (Graf, 2001). A double-blinded, randomized, crossover trial was performed by administering 0.5% levobupivacaine and 0.5% ropivacaine intravenously at 10 mg/minute (maximum 150 mg) on separate days after a washout period to 13 adult male volunteers. The agents did not differ from each other in the incidence of ECG effects; however, subjects receiving levobupivacine reported fewer CNS effects (Stewart et al, 2003).
    G) Local anesthetics are weak bases with pKa's ranging from 7.5 to 9. They are available as hydrochloride or sulfate salts and free bases. Solutions are acidic (pH 4-7) in order to increase their stability
    H) Local anesthetics, in order of water solubility and increasing potency and duration of action following infiltration are: procaine</=chloroprocaine</=prilocaine</=mepivacaine</= lidocaine</=etidocaine</=bupivacaine</=tetracaine</= chichocaine.
    I) Anesthetic potency increases as lipid solubility increases. Rapidity of action (onset of effects) increases with increasing lipid solubility and decreasing pKa.
    J) Increasing the pH of the diluent increases the amount of pharmacologically active anesthetic (free, unchanged amine).
    K) Duration of action increases as lipid solubility and protein binding increase and decreases as vasodilating activity increases.
    L) True allergy to local anesthetics, particularly the amide agents, is rare. The para-aminobenzoic acid metabolite of ester type anesthetics can act as a hapten.

Toxicologic Mechanism

    A) Toxicity is due to an exaggerated pharmacological activity, primarily involving the cardiovascular and central nervous systems.
    B) High drug levels result in depressed membrane function of all excitable tissues eventually resulting in membrane instability.
    C) CARDIOVASCULAR TOXICITY
    1) Cardiovascular effects include vasodilation, decreased myocardial contractility, cardiac conduction delays or blocks, and eventually vascular collapse and cardiac arrest. Although rare, membrane instability may sometimes precipitate tachyarrhythmias.
    2) Bupivacaine and etidocaine, in particular, tend to occupy the myocardial sodium channel for prolonged periods of time, possessing rate-dependent channel blockade pharmacokinetics, and causing a reduction in the rate of ventricular conduction by depression of the rapid depolarization phase (Vmax). A uni-directional blockade of myocardial-conducting pathways is then created which promotes the frequent occurrence of re-entrant dysrhythmias and which may result in ventricular fibrillation (Bacsik et al, 1995).
    3) Animal studies have shown that cardiotoxicity that follows local anesthetic- induced seizures may be due to increased uptake of the anesthetics into the brain because of increased blood-brain barrier permeability during the seizures. This increased uptake may lead to CNS-mediated inotropic and chronotropic suppression of the myocardium (Bacsik et al, 1995).
    D) CNS depression may cause paradoxical behavioral and neuromuscular excitation (eg, seizures) as a result of blockade of inhibitory synapses.
    E) The relative toxicity of local anesthetics parallels their pharmacological potency.

Physical Characteristics

    A) BUPIVACAINE HYDROCHLORIDE: White, crystalline powder that is freely soluble in 95% ethanol, soluble in water, and slightly soluble in chloroform or acetone (Prod Info Marcaine(TM) injection solution, 2009).
    B) BUPIVACAINE LIPOSOME: White to off-white preservative-free aqueous suspension of multivesicular liposomes containing bupivacaine (Prod Info EXPAREL(TM) injection suspension, 2011).
    C) LIDOCAINE HYDROCHLORIDE MONOHYDRATE: Freely soluble in water, soluble in alcohol and chloroform, and insoluble in ether with a melting point of about 74 to 79 degrees C (Prod Info Zingo(TM) intradermal injection powder, 2014).
    D) MEPIVACAINE HYDROCHLORIDE: White, crystalline, odorless powder that is soluble in water, but very resistant to both acid and alkaline hydrolysis (Prod Info Carbocaine(TM) solution for injection, 2010).
    E) ROPIVACAINE HYDROCHLORIDE: White crystalline powder with a solubility of 53.8 mg/mL in water at 25 degrees C (Prod Info NAROPIN(R) Injection solution, 2010).

Ph

    A) BUPIVACAINE HYDROCHLORIDE: 4 to 6.5 (Prod Info Marcaine(TM) injection solution, 2009)
    B) BUPIVACAINE LIPOSOME: 5.8 to 7.4 (Prod Info EXPAREL(TM) injection suspension, 2011)
    C) CHLOROPROCAINE HYDROCHLORIDE: 2.7 to 4 (Prod Info Nesacaine(R) Nesacaine(R)-MPF solution injection, 2010)
    D) LIDOCAINE HYDROCHLORIDE: approximately 6.5 (range, 5 to 7) (Prod Info Xylocaine(R) injection solution, 2010)
    E) MEPIVACAINE HYDROCHLORIDE: 4.5 to 6.8 (Prod Info Carbocaine(TM) solution for injection, 2010)

Molecular Weight

    A) BUPIVACAINE HYDROCHLORIDE: 342.91 (USAN, 1997)
    B) BUPIVACAINE LIPOSOME: 288.4 (Prod Info EXPAREL(TM) injection suspension, 2011)
    C) CHLOROPROCAINE HYDROCHLORIDE: 307.22 (Prod Info chloroprocaine hcl injection, 2005)
    D) LIDOCAINE: 234.34 (Prod Info lidocaine hcl 3%, hydrocortisone acetate 0.5% topical cream, 2006)
    E) LIDOCAINE HYDROCHLORIDE: 270.8 (Prod Info Xylocaine(R) injection solution, 2010)
    F) LIDOCAINE HYDROCHLORIDE MONOHYDRATE: 288.8 Daltons (Prod Info Zingo(TM) intradermal injection powder, 2014)
    G) MEPIVACAINE HYDROCHLORIDE: 282.81 (Prod Info POLOCAINE(R) DENTAL 3%, POLOCAINE(R) DENTAL 2% dental injection, 2004)
    H) ROPIVACAINE HYDROCHLORIDE: 328.89 (Prod Info NAROPIN(R) Injection solution, 2010)
    I) TETRACAINE: 264.4 (Prod Info PLIAGLIS(R) topical cream, 2012)

Clinical Effects

    11.1.9) OVINE/SHEEP
    A) LIDOCAINE - Six 18-month-old cross-bred pregnant ewes were given 20 mg/kg 2% subcutaneous lidocaine prior to surgery. Within 15 minutes, they developed muscle tremors, recumbency, opisthotonos, mydriasis, and seizures. All recovered with supportive treatment (Scarratt & Troutt, 1986).

Treatment

    11.2.2) LIFE SUPPORT
    A) GENERAL
    1) MAINTAIN VITAL FUNCTIONS: Secure airway, supply oxygen, and begin supportive fluid therapy if necessary.
    11.2.5) TREATMENT
    A) GENERAL TREATMENT
    1) SEIZURES/CNS EXCITATION -
    a) SEIZURES/LARGE ANIMALS: May be controlled with diazepam.
    1) HORSES/DIAZEPAM: Neonates: 0.05 to 0.4 milligrams/kilogram; Adults: 25 to 50 milligrams. Give slowly intravenously to effect; repeat in 30 minutes if necessary.
    2) CATTLE, SHEEP AND SWINE/DIAZEPAM: 0.5 to 1.5 milligrams/kilogram intravenously to effect.
    b) SEIZURES/DOGS & CATS:
    1) DIAZEPAM: 0.5 to 2 milligrams/kilogram intravenous bolus; may repeat dose every ten minutes for four total doses. Give slowly over 1 to 2 minutes to effect.
    2) PHENOBARBITAL: 5 to 30 milligrams/kilogram over 5 to 10 minutes intravenously to effect.
    3) REFRACTORY SEIZURES: Consider anaesthesia or heavy sedation. Administer pentobarbital to DOGS & CATS at a dose of 3 to 15 milligrams/kilogram intravenously slowly to effect. May need to repeat in 4 to 8 hours. Be sure to protect the airway.
    2) GENERAL INFORMATION -
    a) SUMMARY
    1) Begin treatment immediately.
    2) Keep animal warm and do not handle unnecessarily.
    3) Remove the patient and other animals from the source of contamination or remove dietary sources.
    b) Treatment should always be done on the advice and with the consultation of a veterinarian.
    c) Additional information regarding treatment of poisoned animals may be obtained from a Veterinary Toxicologist or the National Animal Poison Control Center.
    d) ASPCA ANIMAL POISON CONTROL CENTER
    1) ASPCA Animal Poison Control Center, 1717 S Philo Road, Suite 36 Urbana, IL 61802
    2) It is an emergency telephone service which provides toxicology information to veterinarians, animal owners, universities, extension personnel and poison center staff for a fee. A veterinary toxicologist is available for consultation.
    3) Contact information: (888) 426-4435 (hotline) or www.aspca.org (A fee may apply. Please inquire with the poison center). The agency will make follow-up calls as needed in critical cases at no extra charge.

General Bibliography

    1) AMA Department of DrugsAMA Department of Drugs: AMA Evaluations Subscription, American Medical Association, Chicago, IL, 1992.
    2) AMA: AMA Drug Evaluations, 5th ed, American Medical Association, Chicago, IL, 1983.
    3) Abouleish EI, Elias M, & Nelson C: Ropivacaine-induced seizure after extradural anaesthesia. Br J Anaesth 1998; 80:843-844.
    4) Addiego JE, Ridgway D, & Bleyer WA: The acute management of intrathecal methotrexate overdose: pharmacologic rationale and guidelines. J Pediatr 1981; 98(5):825-828.
    5) Admani B & Essajee F: Successful resuscitation of a three month old child with intralipid infusion, presumed to have bupivacaine induced seizures and cardiovascular complications: case report. East Afr Med J 2010; 87(8):354-356.
    6) Agarwal R, Gutlove DP, & Lockhart CH: Seizures occurring in pediatric patients receiving continuous infusion of bupivacaine. Anesth Analg 1992; 75:284-286.
    7) Akbayram S, Akgun C, Dogan M, et al: Acquired methemoglobinemia due to application of prilocaine during circumcision. J Emerg Med 2012; 43(1):120-121.
    8) Ala-Kokko TI, Lopponen A, & Alahuhta S: Two instances of central nervous system toxicity in the same patient following repeated ropivacaine-induced brachial plexus block. Acta Anaesthesiol Scand 2000; 44:623-626.
    9) Alfano SN, Leicht MJ, & Skiendzielewski JJ: Lidocaine toxicity following subcutaneous administration. Ann Emerg Med 1984; 13:465-467.
    10) Altman RS, Smith-Coggins R, & Ampel LL: Local anesthetics. Ann Emerg Med 1985; 14:1209-1217.
    11) American College of Medical Toxicology : ACMT Position Statement: Interim Guidance for the Use of Lipid Resuscitation Therapy. J Med Toxicol 2011; 7(1):81-82.
    12) American College of Medical Toxicology: ACMT position statement: guidance for the use of intravenous lipid emulsion. J Med Toxicol 2016; Epub:Epub-.
    13) Amitai Y, Whitesell L, & Lovejoy FH: Death following accidental lidocaine overdose in a child. N Engl J Med 1986; 314:181-182.
    14) Antonelli D & Bloch L: Sinus standill following lidocaine administration. JAMA 1982; 248:827-828.
    15) Applebaum D & Halperin E: Asystole following a conventional therapeutic dose of lidocaine. Am J Emerg Med 1986; 4:143-145.
    16) Asling JH, Siinider SM, & Margolis AJ: Paracervical block anesthesia in obstetrics: etiology of fetal bradycardia following paracervical block anesthesia. Am J Obstet Gynecol 1970; 107:626-634.
    17) Ayestaran C, Matorras R, & Gomez S: Severe bradycardia and bradypnea following vaginal oocyte retrieval: a possible toxic effect of paracervical mepivacaine. Eur J Obstet Gynecol Reprod Biol 2000; 91:71-73.
    18) Bacsik CJ, Swift JQ, & Hargreaves KM: Toxic systemic reactions of bupivacaine and etidocaine. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 1995; 79:18-23.
    19) Balit CR, Lynch A, Gilmore SP, et al: Lignocaine and chlorhexidine toxicity in children resulting from mouth paint ingestion: a bottling problem. J Paediatr Child Health 2006; 42(6):350-353.
    20) Bardoczky GI, Wathieu M, & D'Hollander A: Prilocaine-induced methemoglobinemia evidenced by pulse oximetry. Acta Anaesthesiol Scand 1990; 34:162-164.
    21) Bazerbachi F, Rank K, & Chan A: Intravenous lipid rescue and ropivacaine systemic toxicity. J Anesth 2014; 28(1):139.
    22) Bedford RF, Persing JA, Pobereskin L, et al: Lidocaine or thiopental for rapid control of intracranial hypertension?. Anesth Analg 1980; 59:435-437.
    23) Bellamy MC, Hopkins PM, & Halsall PJ: A study into the incidence of methaemoglobinaemia after "three-in-one" block with prilocaine. Anaesthesia 1992; 47:1084-1085.
    24) Berger GS, Tyler CW, & Harrod EK: Maternal deaths associated with paracervical block anesthesia. Am J Obstet Gynecol 1974; 118:1142-1143.
    25) Berkow R: The Merck Manual, 16th ed, Merck & Co, Inc, Whitehouse Station, NJ, 1992.
    26) Bisschop DY, Alardo JP, & Razgallah B: Seizure induced by ropivacaine. Ann Pharmacotherapy 2001; 35:311-313.
    27) Blaney SM, Poplack DG, Godwin K, et al: Effect of body position on ventricular CSF methotrexate concentration following intralumbar administration. J Clin Oncol 1995; 13(1):177-179.
    28) Boyes RN, Scott DB, & Jebson PJ: Pharmacokinetics of lidocaine in man. Clin Pharmacol Ther 1971; 12:105-116.
    29) Bozynski MEA, Rubarth LB, & Patel JA: Lidocaine toxicity after maternal pudendal anesthesia in a term infant with fetal distress. Am J Perinatol 1987; 4:164-166.
    30) Briggs GG, Freeman RK, & Yaffe SJ: Drugs in Pregnancy and Lactation. 5th ed, Williams and Wilkins, Baltimore, MD, 1998.
    31) Brophy GM, Bell R, Claassen J, et al: Guidelines for the evaluation and management of status epilepticus. Neurocrit Care 2012; 17(1):3-23.
    32) Brosh-Nissimov T, Ingbir M, Weintal I, et al: Central nervous system toxicity following topical skin application of lidocaine. Eur J Clin Pharmacol 2004; 60:683-684.
    33) Brown DL & Skiendzielowski: Lidocaine toxicity. Ann Emerg Med 1980; 9:627-629.
    34) Bryant CA, Hoffman JR, & Nichter LS: Pitfalls and perils of intravenous lidocaine. West J Med 1983; 139:528-530.
    35) Buck D, Kreeger R, & Spaeth J: Case discussion and root cause analysis: bupivacaine overdose in an infant leading to ventricular tachycardia. Anesth Analg 2014; 119(1):137-140.
    36) Bun D & Doughty A: Methaemoglobinemia follow lignocaine. Lancet 1974; 2:971.
    37) Bursell B, Ratzan RM, & Smally AJ: Lidocaine toxicity misinterpreted as a stroke. West J Emerg Med 2009; 10(4):292-294.
    38) Butler J & Jackson R: Towards evidence based emergency medicine: best BETs from Manchester Royal Infirmary. Lignocaine premedication before rapid sequence induction in head injuries. Emerg Med J 2002; 19(6):554-554.
    39) Byrne MF, Mitchell RM, Gerke H, et al: The need for caution with topical anesthesia during endoscopic procedures, as liberal use may result in methemoglobinemia. J Clin Gastroenterol 2004; 38:225-229.
    40) Bysis SN: Tinnitus and lidocaine therapy. JAMA 1985; 253:2119.
    41) CDC: Prilocaine-induced methemoglobinemia - Wisconsin, 1993. CDC: MMWR 1994; 43:655-657.
    42) Cashman NN: Anaesthesia 1984; 39:248-250. (CYCLAINE(R)) as topical anesthetic in gastroscopy and esophagoscopy. Gastroenterology 1959; 36:120-121.
    43) Chamberlain JM, Altieri MA, & Futterman C: A prospective, randomized study comparing intramuscular midazolam with intravenous diazepam for the treatment of seizures in children. Ped Emerg Care 1997; 13:92-94.
    44) Chang YY, Ho CM, & Tsai SK: Cardiac arrest after intraurethral administration of lidocaine. J Formos Med Assoc 2005; 104(8):605-606.
    45) Charbonneau H, Marcou TA, Mazoit JX, et al: Early use of lipid emulsion to treat incipient mepivacaine intoxication. Reg Anesth Pain Med 2009; 34(3):277-278.
    46) 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.
    47) Choonara IA & Rane A: Therapeutic drug monitoring of anticonvulsants state of the art. Clin Pharmacokinet 1990; 18:318-328.
    48) Clapp CR, Poss WB, & Cilento BG: Lidocaine toxicity secondary to postoperative bladder instillation in a pediatric patient (abstract). Urol 1999; 53:1228.
    49) Coleman M & Kelly DJ: Local anaesthetic toxicity in a pregnant patient undergoing lignocaine-induced intravenous regional anaesthesia. Acta Anaesthesiol Scand 1998; 42:267-269.
    50) Cooper JS & May JW : Intravenous regional anesthesia toxicity in pregnancy. Plast Reconstr Surg 2009; 124(1):182e-183e.
    51) Cusson J, Nattel S, & Matthews C: Age-dependent lidocaine disposition in patients with acute myocardial infarction. Clin Pharmacol Ther 1985; 37:381-386.
    52) Cydulka RK & Betzelos S: Seizures following the use of proparacaine hydrochloride eye drops. J Emerg Med 1990; 8:131-133.
    53) Dailey RH: Fatality secondary to misuse of TAC solution. Ann Emerg Med 1988; 17:159-160.
    54) Dawling S, Flanagan RJ, & Widdop B: Fatal lignocaine poisoning: report of two cases and review of the literature. Human Toxicol 1989; 8:389-392.
    55) Dayan PS, Litovitz TL, & Crouch BI: Fatal accidental dibucaine poisoning in children. Ann Emerg Med 1996; 28:442-445.
    56) De Praeter C, Vanhaesebrouck P, & DePraeter N: Episiotomy and neonatal lidocaine intoxication (letter). Eur J Pediatr 1991; 150:685-686.
    57) Denson DD, Toltzis RJ, & Ernest TF: Rapid estimation of unbound lidocaine clearance in cardiac patients: implications for reducing toxicity. J Clin Pharm 1988; 28:995-1000.
    58) Desparmet J, Meistelman C, & Barre J: Continuous epidural infusion of bupivacaine for postoperative pain relief in children. Anesthesiology 1987; 67:108-110.
    59) Dix SK, Rosner GF, Nayar M, et al: Intractable cardiac arrest due to lidocaine toxicity successfully resuscitated with lipid emulsion. Crit Care Med 2011; 39(4):872-874.
    60) Dodson WE, Hillman RE, & Hillman LS: Brain tissue levels in a fatal case of neonatal mepicaine (Carbocaine) poisoning. J Pediatr 1975; 86:624-627.
    61) Dodson WE: Neonatal drug intoxication. Pediatr Clin North Am 1976; 3:399-409.
    62) Donald MJ & Derbyshire S: Lignocaine toxicity; a complication of local anaesthesia administered in the community. Emerg Med J 2004; 21:249-150.
    63) Donohue JF: Death from ARDS and cardiovascular collapse following lidocaine administration. Chest 1983; 3:585.
    64) Doyle KA & Goepferd SJ: An allergy to local anesthetics? The consequences of a misdiagnosis. J Dent Child 1989; 56:103-106.
    65) Drapkin Z, Caravati EM, Plumb J, et al: Intentional oral viscous lidocaine overdose resulting in seizure and death in an adolescent. Clin Toxicol (Phila) 2015; 53(7):719-720.
    66) Dudley MH, Fleming SW, Garg U, et al: Fatality involving complications of bupivacaine toxicity and hypersensitivity reaction. J Forensic Sci 2011; 56(5):1376-1379.
    67) Duffin RM & Olson RJ: Tetracaine toxicity. Ann Ophthalmol 1984; 16:836-837.
    68) Duncan PG & Kobrinsky N: Prilocaine-induced methemoglobinemia in a newborn infant. Anesthesiology 1983; 59:75-76.
    69) Dunipace AJ, Beaven R, Noblitt T, et al: Mutagenic potential of toluidine blue evaluated in the Ames test. Mutat Res 1992; 279(4):255-259.
    70) Eaglstein NF: Chemical injury to the eye from EMLA cream during erbium laser resurfacing. Dermatol Surg 1999; 25:590-591.
    71) Edgren B, Tilelli J, & Gehrz R: Intravenous lidocaine overdosage in a child. Clin Toxicol 1986; 24:51-58.
    72) Egfjord M, Jansen JA, & Flachs H: Combined boric acid and cinchocaine xchloride poisoning in a 12-month-old infant: evaluation of haemodialysis. Human Toxicol 1988; 17:175-178.
    73) Eledjam JJ: Ropivacaine overdose and systemic toxicity. Anaesthesia Int Care 2000; 28:705-707.
    74) Espinet AJ & Emmerton MT : The successful use of intralipid for treatment of local anesthetic-induced central nervous system toxicity: Some considerations for administration of intralipid in an emergency. Clin J Pain 2009; 25(9):808-809.
    75) Eyres RL, Bishop W, & Oppenheim RC: Plasma bupivacaine concentrations in children during caudal epidural analgesia. Anaesth Intens Care 1983; 11:20-22.
    76) FDA: Oral health care drug products for over-the-counter human use: establishment of a monograph. FDA: Fed Register 1982; 47:22760-22812.
    77) Feely J, Wade D, & McAllister CB: Effect of hypotension on liver blood flow and lidocaine disposition. N Engl J Med 1982; 307:866-869.
    78) Felice K & Schumann H: Intravenous lipid emulsion for local anesthetic toxicity: a review of the literature. J Med Toxicol 2008; 4(3):184-191.
    79) Ferraro L, Zeichner S, & Greenblatt G: Cetacaine-induced acute methemoglobinemia. Anesthesiology 1988; 69:614-615.
    80) Ferraro L, Zeichner S, & Greenblott G: Cetacaine-induced methemoglobinemia. Anesthesiology 1988a; 69:614-615.
    81) Finkelstein F & Kreeft J: Massive lidocaine poisoning. N Engl J Med 1979; 301:50.
    82) Finster M, Poppers PJ, & Sinclair JC: Accidental intoxication of the fetus with local anesthetic drug during caudal anesthesia. Am J Obstet Gynecol 1965; 92:922-924.
    83) Fisher AA: Allergic reactions to topical (surface) anesthetics with reference to the safety of tronothane (pramoxine hydrochloride). Cutis 1980; 25:584-591.
    84) Fitzsimons MG, Gaudette RR, & Hurford WE: Critical rebound methemoglobinemia after methylene blue treatment: case report. Pharmacotherapy 2004; 24(4):538-540.
    85) Foxall G, McCahon R, Lamb J, et al: Levobupivacaine-induced seizures and cardiovascular collapse treated with Intralipid. Anaesthesia 2007; 62(5):516-518.
    86) Frayling IM, Addison GM, & Chattergee K: Methaemoglobinaemia in children treated with prilocaine-lignocaine cream. Br Med J 1990; 301:153-154.
    87) Freedman MD, Gal J, & Freed CR: Extracorporeal pump assistance - novel treatment for acute lidocaine poisoning. Eur J Clin Pharmacol 1982; 22:129-135.
    88) Fruncillo RJ, Gibbons W, & Bowman SM: CNS toxicity after ingestion of topical lidocaine (letter). N Engl J Med 1982; 306:426-427.
    89) Garfield JM, Andriole GL, & Vetto JT: Prolonged diabetes insipidus subsequent to an episode of chemical meningitis. Anesthesiology 1986; 64:253-254.
    90) Garrettson LK & McGee EB: Rapid onset of seizures following aspiration of viscous lidocaine. Clin Toxicol 1992; 30(3):413-422.
    91) Giard MJ, Uden DL, & Whiltock DJ: Convulsions induced by oral viscous lidocaine (letter). Clin Pharm 1983; 2:110.
    92) Gilman AG, Rall TW, Nies AS, et alGilman AG, Rall TW, Nies AS, et al (Eds): Goodman and Gilman's The Pharmacological Basis of Therapeutics, 8th. Pergamon Press, New York, NY, 1990.
    93) Giuliani M, Grossi GB, & Pileri M: Could local anesthesia while breast-feeding be harmful to infants?. J Ped Gastroenterol Nutr 2001; 32:142-144.
    94) Goldberg G & Goodman DH: Case report of hexylcaine reaction. Anesthesiology 1957; 18:652-653.
    95) Goodwin DP & McMeekin TO: A case of lidocaine absorption from topical administration of 40% lidocaine cream (letter). J Am Acad Dermatol 1999; 41:280-281.
    96) Gosselin S & Isbister GK: Re: Treatment of accidental intrathecal methotrexate overdose. J Natl Cancer Inst 2005; 97(8):609-610.
    97) Graf BM: The cardiotoxicity of local anesthetics: the place of ropivacaine. Curr Top Med Chem 2001; 1(3):207-214.
    98) Greenblatt DJ, Benjamin DM, & Willis CR: Lidocaine plasma concentrations following administration of intraoral lidocaine solution. Arch Otolayngol 1985; 111:298-300.
    99) Grenadier E, Alpan G, & Keider S: Respiratory and cardiac arrest after the administration of lidocaine into the central venous system. Eur Heart J 1981; 2:235-237.
    100) Grimes DA & Cates W: Deaths from paracervical anesthesia used for first-trimester abortion, 1972-1975. N Engl J Med 1976; 295:1397-1399.
    101) Gupta P, Lichstein E, & Claade K: Lidocaine-induced heart block in patients with bundle branch block. Am J Cardiol 1976; 33:487-492.
    102) Hahn I, Hoffman RS, & Nelson LS: EMLA-induced methemoglobinemia (MetHB) and lidocaine toxicity (abstract). Clin Toxicol 1999; 37:621.
    103) Hahn IH, Hoffman RS, & Nelson LS: EMLA(R)-induced methemoglobinemia and systemic topical anesthetic toxicity. J Emerg Med 2003; 26(1):85-88.
    104) Hamano S , Sugiyama N , Yamashita S , et al: Intravenous lidocaine for status epilepticus during childhood. Dev Med Child Neurol 2006; 48(3):220-222.
    105) Hansen-Flaschen J: Methemoglobinemia after lidocaine administration (letter). Chest 1990; 98:519-520.
    106) Hart D: Psychotic reaction to procaine. J Emerg Nursing 1984; 10:9-10.
    107) Hasselstrom LJ & Morgensen T: Toxic reaction of bupivacaine at low plasma concentrations. Anesthesiology 1984; 61:99-100.
    108) Hattori H , Yamano T , Hayashi K , et al: Effectiveness of lidocaine infusion for status epilepticus in childhood: a retrospective multi-institutional study in Japan. Brain Dev 2008; 30(8):504-512.
    109) Heath ML: Bupivicaine toxicity and bier blocks (letter). Anesthesiology 1983; 59:481.
    110) Heath ML: Deaths after intravenous regional anaesthesia. Br Med J 1982; 285:913-914.
    111) Hegedus F & Herb K: Benzocaine-induced methemoglobinemia. Anesth Prog 2005; 52(4):136-139.
    112) Hegenbarth MA & American Academy of Pediatrics Committee on Drugs: Preparing for pediatric emergencies: drugs to consider. Pediatrics 2008; 121(2):433-443.
    113) Herman MI, Chyka PA, & Butlse AY: Methylene blue by intraosseous infusion for methemoglobinemia. Ann Emerg Med 1999; 33:111-113.
    114) Hess GP & Walson PD: Seizures secondary to oral viscous lidocaine. Ann Emerg Med 1988; 17:725-727.
    115) Hiller DB, Gregorio GD, Ripper R, et al: Epinephrine impairs lipid resuscitation from bupivacaine overdose: a threshold effect. Anesthesiology 2009; 111(3):498-505.
    116) Hillman LS, Hillman RE, & Dodson WE: Diagnosis, treatment and follow-up of neonatal mepivacaine intoxication secondary to paracervical and pudendal blocks during labor. J Pediatr 1979; 95:472-477.
    117) Hines EQ, Fernandez D, & Lugassy DM: Systemic lidocaine toxicity associated with tumescent liposuction (abstracts). Clin Toxicol (Phila) 2015; 53(7):680.
    118) Hix WR & Wilson WR: Toluidine blue staining of the esophagus: a useful adjunct in the panendoscopic evaluation of patients with squamous cell carcinoma of the head and neck. Arch Otolaryngol Head Neck Surg 1987; 113(8):864-865.
    119) Hjelt K, Lund JT, Scherling B, et al: Methaemoglobinaemia among neonates in a neonatal intensive care unit. Acta Paediatr 1995; 84(4):365-370.
    120) Hoegberg LC, Bania TC, Lavergne V, et al: Systematic review of the effect of intravenous lipid emulsion therapy for local anesthetic toxicity. Clin Toxicol (Phila) 2016; 54(3):167-193.
    121) Hogan Q, Dotson R, & Erickson S: Local anesthetic myotoxicity: a case and review. Anesthesiol 1994; 80:942-947.
    122) Howard JJ, Mohsenifar Z, & Simons SM: Adult respiratory distress syndrome following administration of lidocaine. Chest 1982; 81:644-645.
    123) Howland MA: Antidotes in Depth. In: Goldfrank LR, Flomenbaum N, Hoffman RS, et al, eds. Goldfrank's Toxicologic Emergencies. 8th ed., 8th ed. McGraw-Hill, New York, NY, 2006, pp 826-828.
    124) Hubler M , Gabler R , Ehm B , et al: Successful resuscitation following ropivacaine-induced systemic toxicity in a neonate. Anaesthesia 2010; 65(11):1137-1140.
    125) Hvidberg EF & Dam M: Clinical pharmacokinetics of anticonvulsants. Clin Pharmacokinet 1976; 1:161.
    126) Isohanni MH, Ahonen J, Neuvonen PJ, et al: Effect of ciprofloxin on the pharmacokinetics of intravenous lidocaine. Eur J Anaesthesiol 2005; 22(10):795-799.
    127) Jacobi J, McGory RS, McCoy H, et al: Hemodialysis clearance of total and unbound lidocaine. Clin Pharm 1983; 2:54-57.
    128) Jonville AP, Barbier P, & Blond MH: Accidental lidocaine overdosage in an infant. J Toxicol Clin Toxicol 1990; 28:101-106.
    129) Karim A, Ahmed S, Siddiqui R, et al: Methemoglobinemia complicating topical lidocaine used during endoscopic procedures. Am J Med 2001; 111(2):150-153.
    130) Kiese M , Lorcher W , Weger N , et al: Comparative studies on the effects of toluidine blue and methylene blue on the reduction of ferrihaemoglobin in man and dog. Eur J Clin Pharmacol 1972; 4(2):115-118.
    131) Kim JT, Jung CW, & Lee KH: The effect of insulin on the resuscitation of bupivacaine-induced severe cardiovascular toxicity in dogs. Anesth Analg 2004; 99:728-33.
    132) Kim WY, Pomerance JJ, & Miller AA: Lidocaine intoxication in a newborn following local anesthesia for episiotomy. Pediatrics 1979; 64:643-645.
    133) Kimura Y, Kamada Y, Kimura A, et al: Ropivacaine-induced toxicity with overdose suspected after axillary brachial plexus block. J Anesth 2007; 21(3):413-416.
    134) Kintner JC, Grossniklaus HE, & Lass JH: Infectious crystalline keratopathy associated with topical anesthetic abuse. Cornea 1990; 9:77-80.
    135) 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.
    136) Klos CP & Hays GL: Prilocaine-induced methemoglobinemia in a child with Shwachman syndrome. J Oral Maxillofac Surg 1985; 43:621-623.
    137) Kobayashi K , Ito M , Miyajima T , et al: Successful management of intractable epilepsy with intravenous lidocain and lidocain tapes. Pediatr Neurol 1999; 21(1):476-480.
    138) Kopacz DJ & Allen HW: Accidental intravenous levobupivacaine. Anesth Analg 1999; 89:1027-1029.
    139) Kotler RL, Hansen-Flaschen J, & Casey MP: Severe methaemoglinaemia after flexible fiberoptic bronchoscopy. Thorax 1989; 44:234-235.
    140) Kotler RL, Hansen-Flaschen J, & Casey MP: Severe methaemoglobinaemia after flexible fiberoptic bronchoscopy. Thorax 1989a; 44:234-235.
    141) Kraut JA & Madias NE: Metabolic acidosis: pathophysiology, diagnosis and management. Nat Rev Nephrol 2010; 6(5):274-285.
    142) Kreutz RW & Kinni ME: Life-threatening toxic methemoglobinemia induced by prilocaine. Oral Surg 1983; 56:480-482.
    143) Kudo K, Nishida N, Kiyoshima A, et al: A fatal case of poisoning by lidocaine overdoseage - analysis of lidocaine in formalin-fixed tissues: a case report. Med Sci Law 2004; 44(3):266-271.
    144) Kunkel F, Rowland M, & Scherman M: The electrophysiologic effects of lidocaine in patients with intraventricular conduction defects. Circulation 1974; 49:894-899.
    145) Kuster CG & Udin RD: Frequency of accidental intravascular injection of local anesthetics in children. J Dentistry for Children 1985; 52:183-187.
    146) Labedzki L, Ocks HR, & Abernethy DR: Potentially toxic serum lidocaine concentrations following spray anesthesia for bronchoscopy. Klin Wochenschr 1983; 61:379-380.
    147) Lankin DL, Jewell GM, & Grinvalsky HT: Psychotic-like reaction to procaine penicillin G. Ann Emerg Med 1983; 12:507-509.
    148) Larson A, Stidham T, Banerji S, et al: Seizures and methemoglobinemia in an infant after excessive EMLA application. Pediatr Emerg Care 2013; 29(3):377-379.
    149) Lavonas EJ, Drennan IR, Gabrielli A, et al: Part 10: Special Circumstances of Resuscitation: 2015 American Heart Association Guidelines Update for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2015; 132(18 Suppl 2):S501-S518.
    150) Lebedevs TH, Wojnar-Horton RE, Yapp P, et al: Excretion of lignocaine and its metabolite monoethylglycinexylidide in breast milk following its use in a dental procedure. A case study. J Clin Periodontol 1993; 20(8):606-8.
    151) Ledbetter AA Jr & Elliott JH: Trochlear nerve palsy following spinal anesthesia. A case report. J Okla State Med Assoc 1963; 56:152-155.
    152) Lee DL, Ayoub C, & Shaw RK: Grand mal seizure during cardiopulmonary bypass: probable lidocaine toxicity. J Cardiothoracic Vasc Anesth 1999; 13:200-202.
    153) Leskiw U & Weinberg GL: Lipid resuscitation for local anesthetic toxicity: is it really lifesaving?. Curr Opin Anaesthesiol 2009; 22(5):667-671.
    154) Lie RL, Vermeer BJ, & Edelbroek PM: Severe lidocaine intoxication by cutaneous absorption. J Am Acad Dermatol 1990; 23:1026-1028.
    155) Lin JJ , Lin KL , Wang HS , et al: Effect of topiramate, in combination with lidocaine, and phenobarbital, in acute encephalitis with refractory repetitive partial seizures. Brain Dev 2009; 31(8):605-611.
    156) Lindenmann J, Matzi V, Kaufmann P, et al: Hyperbaric oxygenation in the treatment of life-threatening isobutyl nitrite-induced methemoglobinemia--a case report. Inhal Toxicol 2006; 18(13):1047-1049.
    157) Litz RJ, Popp M, Stehr SN, et al: Successful resuscitation of a patient with ropivacaine-induced asystole after axillary plexus block using lipid infusion. Anaesthesia 2006; 61(8):800-801.
    158) Litz RJ, Roessel T, Heller AR, et al: Reversal of central nervous system and cardiac toxicity after local anesthetic intoxication by lipid emulsion injection. Anesth Analg 2008; 106(5):1575-7, table.
    159) Lloyd CJ: Chemically induced methaemoglobinaemia in a neonate. Br J Oral Maxill Surg 1992; 30:63-65.
    160) Lloyd CJ: Chemically induced methaemoglobinaemia in a neonate. Br J Oral Maxillofacial Surg 1992a; 30:63-65.
    161) Loddenkemper T & Goodkin HP: Treatment of Pediatric Status Epilepticus. Curr Treat Options Neurol 2011; Epub:Epub.
    162) Logan BK & Gordon AM: Death of an infant involving benzocaine. J Forensic Sci 2005; 50(6):1486-1488.
    163) Long WB, Rosenblum S, & Grady IP: Successful resuscitation of bupivacaine-induced cardiac arrest using cardiopulmonary bypass. Anesth Anal 1989; 69:403-406.
    164) Ludot H, Tharin JY, Belouadah M, et al: Successful resuscitation after ropivacaine and lidocaine-induced ventricular arrhythmia following posterior lumbar plexus block in a child. Anesth Analg 2008; 106(5):1572-4, table.
    165) Ludwig SC: Acute toxic methemoglobinemia following dental analgesia. Ann Emerg Med 1981; 10:265-266.
    166) Lumb AB & Carli F: Respiratory arrest after a caudal injection of bupivacaine. Anaesthesia 1989; 44:324-325.
    167) Mahli A, Coskun D, & Akcali DT: Aetiology of convulsions due to stellate ganglion block: a review and report of two cases. Eur J Anaesthesiol 2002; 19(5):376-380.
    168) Malone JD, Lebar RD, & Hilder R: Procaine-induced seizures after intramuscular procaine penicillin G. Milit Med 1988; 153:191-192.
    169) Mandel S: Methemoglobinemia following neonatal circumcision (letter). JAMA 1989; 261:702.
    170) Manno EM: New management strategies in the treatment of status epilepticus. Mayo Clin Proc 2003; 78(4):508-518.
    171) Mardirosoff C & Dumont L: Convulsions after the administration of high dose ropivacaine following an interscalenic block (letter). Can J Anaesth 2000; 47:1263.
    172) Markowitz S & Neal JM: Immediate lipid emulsion therapy in the successful treatment of bupivacaine systemic toxicity. Reg Anesth Pain Med 2009; 34(3):276-.
    173) Marks LF & Desgrand D: Prilocaine associated methaemoglobinaemia and the pulse oximeter. Anaesthesia 1991; 46:703.
    174) Marquez A & Todd M: Acute hemolytic anemia and agranulocytosis following intravenous administration of toluidine blue. Am Pract 1959; 10:1548-1550.
    175) Marriott JL & Philips K: Profound hypotension and bradycardia after a single bolus of lidocaine. J Electrocardiol 1974; 7:79-87.
    176) Marsch SCU, Schaefer HG, & Castelli I: Unusual psychological manifestation of systemic local anesthetic toxicity. Anesthesiol 1998; 88:531-533.
    177) Marvez-Valls E, Houry D, Ernst AA, et al: Protocol for rapid sequence intubation in pediatric patients -- a four-year study. Med Sci Monit 2002; 8(4):229-234.
    178) Mather LE & Cousins MJ: Local amesthetics and their current clinical use. Drug 1979; 18:185-205.
    179) Mather LE, Long GJ, & Thomas J: The intravenous toxicity and clearance of bupivacaine in man. Clin Pharm Ther 1971; 12:935-943.
    180) McAlpine LG & Thomson NC: Lidocaine-induced bronchoconstriction in asthmatic patients: relation to histamine airway responsiveness and effect of preservative. Chest 1989; 96:1012-1015.
    181) McCloskey JJ, Haun SE, & Deshpande JK: Bupivacaine toxicity secondary to continuous caudal epidural infusion in children. Anesth Analg 1992; 75:287-290.
    182) McCutchen T & Gerancher JC: Early intralipid therapy may have prevented bupivacaine-associated cardiac arrest. Reg Anesth Pain Med 2008; 33(2):178-180.
    183) McKinney CD, Postiglion KF, & Herold DA: Benzocaine-adultered street cocaine in association with methemoglobinemia. Clin Chem 1992; 38:596-597.
    184) Meffin P, Long GJ, & Thomas J: Clearance and metabolism of mepivacaine in the human neonate. Clin Pharmacol Ther 1973; 14:218-225.
    185) Mitchell WG: Status epilepticus and acute repetitive seizures in children, adolescents, and young adults: etiology, outcome, and treatment. Epilepsia 1996; 37(S1):S74-S80.
    186) Mizutani K, Oda Y, & Sato H: Successful treatment of ropivacaine-induced central nervous system toxicity by use of lipid emulsion: effect on total and unbound plasma fractions. J Anesth 2011; 25(3):442-445.
    187) Mofenson HC, Caraccio TR, & Miller H: Lidocaine toxicity from topical mucousal application. Clin Pediatr 1983; 22:190-192.
    188) Moore DC, Thompson GE, & Crawford RD: Long-acting local anesthetic drugs and convulsions with hypoxia and acidosis. Anesthesiology 1982a; 56:230-231.
    189) Moore DC, Thompson GE, & Crawford RD: Long-acting local anesthetic drugs and seizures with hypoxia and acidosis. Anesthesiology 1982; 56:230.
    190) Moore TJ, Walsh CS, & Cohen MR: Reported adverse event cases of methemoglobinemia associated with benzocaine products. Arch Intern Med 2004; 164:1192-1196.
    191) Moore TJ, Walsh CS, & Cohen MR: Reported adverse event cases of methemoglobinemia associated with benzocaine products. Arch Intern Med 2004a; 164(11):1192-1196.
    192) Mower WR III & Knopp RK: Clinical controversies: Lidocaine administration before rapid sequence intubation in patients with traumatic brain injuries. Ann Emerg Med 2007; 49(1):84-85.
    193) Murphy JP & Anandaciva S: Phantom limb pain and spinal anaesthesia (letter). Anaesthesia 1984; 39:188.
    194) Nakayama DK, Waggoner T, Venkataraman ST, et al: The use of drugs in emergency airway management in pediatric trauma. Ann Surg 1992; 216(2):205-211.
    195) Naradzay J & Barish RA: Approach to ophthalmologic emergencies. Med Clin North Am 2006; 90(2):305-328.
    196) Neal JM , Bernards CM , Butterworth JF , et al: ASRA Practice Advisory on Local Anesthetic Systemic Toxicity. Reg Anesth Pain Med 2010; 35(2):152-161.
    197) Nelsen J, Holland M, Dougherty M, et al: Severe Central Nervous System and Cardiovascular Toxicity in a Pediatric Patient After Ingestion of an Over-the-Counter Local Anesthetic. Pediatr Emerg Care 2009; 25(10):670-673.
    198) Nelson LS & Hoffman RS: Intrathecal lidocaine administration during trigger point injection (abstract). J Toxicol Clin Toxicol 1995; 33:547.
    199) Nemec K: Antidotes in acute poisoning. Eur J Hosp Pharm Sci Pract 2011; 17(4):53-55.
    200) 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.
    201) Noble J, Kennedy DJ, & Latimer RD: Massive lignocaine overdose during cardiopulmonary bypass. Br J Anaesth 1984; 56:1439-1441.
    202) Olkkola KT, Isohanni MH, Hamunen K, et al: The effect of erythromycin and fluvoxamine on the pharmacokinetics of intravenous lidocaine. Anesth Analg 2005; 100(5):1352-1356.
    203) Olson ML & McEvoy GK: Methemoglobinemia induced by local anesthetics. Am J Hosp Pharm 1981; 38:89-93.
    204) Ortega D, Vivivand X, Lorec AM, et al: Excretion of lidocaine and bupivacaine in breast milk following epidural anesthesia for cesarean delivery. Acta Anaesthesiologica Scandinavica 1999; 43(4):394-397.
    205) Palmisano JM, Meliones JN, & Crowley DC: Lidocaine toxicity after subcutaneous infiltration in children undergoing cardiac catheterization. Am J Cardiol 1991; 67:647-648.
    206) Pantuck AJ, Goldsmith JW, & Kuriyan JB: Seizures after ureteral stone manipulation with lidocaine. J Urol 1997; 157:2248.
    207) Parker JF, Vats A, & Bauer G: EMLA toxicity after application for allergy skin testing. Pediatrics 2004; 113(2):410-411.
    208) Peat MA, Deyman MA, & Crouch DJ: Concentrations of lidocaine and monoethylglycylxylidide (MEGX) in lidocaine associated deaths. J Forensic Sci 1985; 30:1048-1057.
    209) Peate WF: Work-related eye injuries and illnesses. Am Fam Physician 2007; 75(7):1017-1022.
    210) 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.
    211) Pelter MA, Vollmer TA, & Blum RL: Seizure-like reaction associated with subcutaneous lidocaine injection (letter). Clin Pharm 1989; 8:767-768.
    212) Perney P, Blanc F, Mourad G, et al: Transitory ataxia related to topically administered lidocaine. Ann Pharmacother 2003; 38:828-30.
    213) Petitjeans F, Mion G, Puidupin M, et al: Tachycardia and convulsions induced by accidental intravascular ropivacaine injection during sciatic block. Acta Anaesthesiol Scand 2002; 46(5):616-617.
    214) Pieper JA, Slaughter RL, & Anderson GD: Lidocaine clinical pharmacokinetics. Drug Intell Clin Pharm 1982; 16:291-294.
    215) Plowman AN, Bolsin S, & Mather LE: Central nervous system toxicity attributable to epidural ropivacaine hydrochloride. Anaesth Intens Care 1998; 26:204-206.
    216) Poklis A, Mackell MA, & Tucher EF: Tissue distribution of lidocaine after fatal accidental injections. J Forensic Sci 1984; 29:1229-1236.
    217) Poppers PJ, Vosburgh GJ, & Finster M: Methemoglobinemia following epidural analgesia during labor. Am J Obstet Gynecol 1956; 95:630-635.
    218) Product Information: Anusol(R), pramoxine ointment, suppositories. Physicians' Desk Reference (electronic version), MICROMEDEX, Inc, Englewood, CO, 1999.
    219) Product Information: Carbocaine(R), mepivacaine HCL. Winthrop-Breon Laboratories, New York, NY, 1987a.
    220) Product Information: Carbocaine(TM) solution for injection, Mepivacaine Hydrochloride solution for injection. Hospira Inc, Lake Forest, IL, 2010.
    221) Product Information: Duranest(R) injection, etidocaine. AstraZeneca LP, Wilmington, DE, 1997.
    222) Product Information: Dyclone(R), dyclonine topical solution 0.5%. Astra Pharmaceuticals, Westboro, MA, 1997.
    223) Product Information: EMLA(R) cream, lidocaine prilocaine cream. AstraZeneca Pharmaceuticals, Wilmington, DE, 2005.
    224) Product Information: EMLA(R) topical cream, lidocaine 2.5% and prilocaine 2.5% topical cream . Abraxis Pharmaceutical Products (per manufacturer), Schaumburg, IL, 2006.
    225) Product Information: EXPAREL(R) injection suspension, bupivacaine liposome injection suspension. Pacira Pharmaceuticals, Inc. (per FDA), San Diego, CA, 2015.
    226) Product Information: EXPAREL(TM) injection suspension, bupivacaine liposome injection suspension. Pacira Pharmaceuticals, Inc. (per Manufacturer), San Diego, CA, 2011.
    227) Product Information: KOVANAZE(TM) nasal spray, tetracaine HCl oxymetazoline HCl nasal spray. St. Renatus, LLC (per FDA), Fort Collins, CO, 2016.
    228) Product Information: Marcaine(R), bupivacaine. Winthrop Laboratories, New York, NY, 1991.
    229) Product Information: Marcaine(R), bupivacaine. Winthrop-Breon Laboratories, New York, NY, 1987.
    230) Product Information: Marcaine(TM) injection solution, Bupivacaine Hydrochloride, Bupivacaine Hydrochloride and Epinephrine injection solution. Hospira, Inc, Lake Forest, IL, 2009.
    231) Product Information: NAROPIN(R) Injection solution, ropivacaine HCl Injection solution. APP Pharmaceuticals LLC, Schaumburg, IL, 2010.
    232) Product Information: Nesacaine(R) Nesacaine(R)-MPF solution injection, chloroprocaine HCl solution injection. APP Pharmaceuticals, LLC, Schaumburg, IL, 2010.
    233) Product Information: Oraqix(R) oral transmucosal gel, lidocaine prilocaine periodental gel 2.5%/2.5% oral transmucosal gel. Dentsply Pharmaceutical, York, PA, 2010.
    234) Product Information: PLIAGLIS(R) topical cream, lidocaine 7% tetracaine 7% topical cream. Galderma Laboratories, L.P. (per Manufacturer), Fort Worth, TX, 2012.
    235) Product Information: POLOCAINE(R) DENTAL 3%, POLOCAINE(R) DENTAL 2% dental injection, mepivacaine hcl 3%, mepivacaine hcl 2%, levonordefrin dental injection. DENTSPLY Pharmaceutical, York, PA, 2004.
    236) Product Information: PROVAYBLUE(TM) intravenous injection, methylene blue intravenous injection. American Regent (per FDA), Shirley, NY, 2016.
    237) Product Information: Pontocaine(R), tetracaine. Winthrop-Breon Laboratories, New York, NY, 1987b.
    238) Product Information: Proctofoam(R), pramoxine. Reed & Carnrick, Piscataway, NJ, 1990.
    239) Product Information: Sensorcaine(R) parenteral injection, bupivacaine HCl parenteral injection. Fresenius Kabi USA, LLC (per FDA), Lake Zurich, IL, 2013.
    240) Product Information: Sensorcaine(R) with Epinephrine parenteral injection, bupivacaine HCl epinephrine parenteral injection. Fresenius Kabi USA, LLC (per FDA), Lake Zurich, IL, 2013.
    241) Product Information: Sensorcaine(R)-MPF parenteral injection, bupivacaine HCl parenteral injection. Fresenius Kabi USA, LLC (per FDA), Lake Zurich, IL, 2013.
    242) Product Information: Sensorcaine(R)-MPF with Epinephrine parenteral injection, bupivacaine HCl epinephrine parenteral injection. Fresenius Kabi USA, LLC (per FDA), Lake Zurich, IL, 2013.
    243) Product Information: Septocaine(R) Solution for intraoral submucosal injection, articaine hydrochloride 4% and epinephrine 1:200,000 articaine hydrochloride 4% and epinephrine 1:100,000 Solution for intraoral submucosal injection. Septodont, Lousiville, CO, 2009.
    244) Product Information: Sucrets(R) Maximum Strength Sore Throat Lozenges. Beecham Products USA, Pittsburgh, PA, 1996.
    245) Product Information: Synera (TM) topical patch, lidocaine/tetracaine topical patch. Ferndale Laboratories, Inc., Ferndale, MI, 2005.
    246) Product Information: Tronolane(R), pramoxine. Ross Labs, Consumer Products, Columbus, OH, 1996.
    247) Product Information: Xylocaine(R) Viscous topical solution, lidocaine HCl 2% topical solution. Fresenius Kabi USA, LLC (per FDA), Lake Zurich, IL, 2014.
    248) Product Information: Xylocaine(R) injection solution, lidocaine HCl, lidocaine HCl and epinephrine injection solution. APP Pharmaceuticals, LLC, Schaumburg, IL, 2010.
    249) Product Information: Xylocaine(R), lidocaine. Astra, Wilmington, DE, 1986.
    250) Product Information: Zingo(TM) intradermal injection powder, lidocaine HCl monohydrate intradermal injection powder. Medline Industries, Inc. (per DailyMed), Mundelein, IL, 2014.
    251) Product Information: chloroprocaine hcl injection, chloroprocaine hcl injection. Bedford Laboratories, Bedford, OH, 2005.
    252) Product Information: diazepam IM, IV injection, diazepam IM, IV injection. Hospira, Inc (per Manufacturer), Lake Forest, IL, 2008.
    253) Product Information: dopamine hcl, 5% dextrose IV injection, dopamine hcl, 5% dextrose IV injection. Hospira,Inc, Lake Forest, IL, 2004.
    254) Product Information: lidocaine hcl 3%, hydrocortisone acetate 0.5% topical cream, lidocaine hcl 3%, hydrocortisone acetate 0.5% topical cream. AAarons,Inc, Fairfield, NJ, 2006.
    255) Product Information: lorazepam IM, IV injection, lorazepam IM, IV injection. Akorn, Inc, Lake Forest, IL, 2008.
    256) Product Information: methylene blue 1% IV injection, methylene blue 1% IV injection. American Regent, Inc (per manufacturer), Shirley, NY, 2011.
    257) Product Information: methylene blue 1% intravenous injection, methylene blue 1% intravenous injection. Akorn, Inc. (per manufacturer), Lake Forest, IL, 2011.
    258) Product Information: norepinephrine bitartrate injection, norepinephrine bitartrate injection. Sicor Pharmaceuticals,Inc, Irvine, CA, 2005.
    259) Promisloff RA & Dupont DC: Death from ARDS and cardiovascular collapse following lidocaine administration (letter). Chest 1983; 3:585.
    260) Rao RB, Ely SF, & Hoffman RS: Deaths related to loposuction. N Eng J Med 1999; 340:1471-1475.
    261) Raso SM, Fernandez JB, Beobide EA, et al: Methemoglobinemia and CNS toxicity after topical application of EMLA to a 4-year-old girl with molluscum contagiosum. Pediatr Dermatol 2006; 23(6):592-593.
    262) Reynolds F: Adverse effects of local anesthetics. Br J Anaesth 1987; 59:78-95.
    263) Rincon E, Baker RL, & Iglesias AJ: CNS toxicity after topical application of EMLA cream on a toddler with molluscum contagiosum. Pediatr Emerg Care 2000; 16:252-254.
    264) Rosefsky JB & Petersiel ME: Perinatal deaths associated with mepivacaine paracervical-block anesthesia in labor. N Engl J Med 1968; 278:530-533.
    265) Rosenblatt MA, Abel M, Fischer GW, et al: Successful use of a 20% lipid emulsion to resuscitate a patient after a presumed bupivacaine-related cardiac arrest. Anesthesiology 2006; 105(1):217-218.
    266) Rothstein P, Dornbusch J, & Shaywitz BA: Prolonged seizures associated with the use of viscous lidocaine. J Pediatr 1982; 101:461-463.
    267) Ruetsch YA, Fattinger KE, & Borgeat A: Ropivicaine-induced convulsions and severe cardiac dysrhythmia after sciatic block. Anesthesiol 1999; 90:1784-1786.
    268) Ryu HY, Kim JY, Lim HK, et al: Bupivacaine induced cardiac toxicity mimicking an acute non-ST segment elevation myocardial infarction. Yonsei Med J 2007; 48(2):331-336.
    269) S Sweetman : Martindale: The Complete Drug Reference (electronic version). The Pharmaceutical Press. London, UK (Internet Version). Edition expires 2001; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    270) Saima S, Echizen H, & Yoshimoto K: Negligible removal of lidocaine during arteriovenous hemofiltration. Ther Drug Monit 1990; 12:154-156.
    271) Sakai R & Lattin J: Lidocaine ingestion. Am J Dis Child 1980; 134:323.
    272) Saravay SM: "Doom anxiety" and delirium in lidocaine toxicity. Am J Psych 1987; 144:159-163.
    273) Savino PJ, Burde RM, & Mills RP: Visual loss following intranasal anesthetic injection. J Clin Neuro-ophthalmol 1990; 10:140-144.
    274) Sawyer RJ & vonSchroeder H: Temporary bilateral blindness after acute lidocaine toxicity. Anesth Analg 2002; 95(1):224-226.
    275) Scarratt WK & Troutt HF: Iatrogenic lidocaine toxicosis in ewes. JAVMA 1986; 188:184-185.
    276) Schmidt JL, Blockus LE, & Richards RK: The pharmacology of pramoxine hydrochloride: a new topical anesthetic. Anesth Analg 1953; 32:418.
    277) Schroeder TH, Dieterich HJ, & Muhlbauer B: Methemoglobinemia after axillary block with bupivacaine and additional injection of lidocaine in the operative field. Acta Anaesthesiol Scand 1999; 43(4):480-482.
    278) Schuster MR, Paris PM, & Kaplan RM: Effect on the seizure threshold in dogs of tocainide/lidocaine administration. Ann Emerg Med 1987; 16:749-751.
    279) Schwartz DR & Kaufman B: Local anesthestics. In: Flomenbaum NE, Goldfrank LR, Hoffman RS, et al. eds. Goldfrank's Toxicologic Emergencies, McGraw-Hill, New York, NY, 2006, pp 1004-1015.
    280) Scott DB, Lee A, & Fagan D: Acute toxicity of ropivacaine compared with that of bupivacaine. Anesth Analg 1989; 69:563-569.
    281) Scott DB, Owen JA, & Richmond J: Methaemoglobinaemia due to prilocaine. Lancet 1964; 2:728-729.
    282) Scott DB: Evaluation of the toxicity of local anesthetic agents in man. Br J Anaesth 1975; 47:56-61.
    283) Scott DB: Toxic effects of local anaesthetic agents on the central nervous system. Br J Anaesth 1986; 58:732-735.
    284) Scott DB: Toxicity caused by local anaesthetic agents. Br J Anaesth 1984; 56:435-436.
    285) 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.
    286) Selden BS & Burke TJ: Complete maternal and fetal recovery after prolonged cardiac arrest. Ann Emerg Med 1988; 17:346-349.
    287) Shah S, Gopalakrishnan S, Apuya J, et al: Use of Intralipid in an infant with impending cardiovascular collapse due to local anesthetic toxicity. J Anesth 2009; 23(3):439-441.
    288) Shepherd G & Keyes DC: Methylene blue. In: Dart,RC, ed. Medical Toxicology, 3rd ed. 3rd ed, Philadelphia, PA, 2004, pp -.
    289) Shnider SM, Asling JH, & Margolis AJ: High fetal blood levels of mepivacaine and fetal bradycardia. N Engl J Med 1968; 279:947-948.
    290) Silber TJ & D'Angelo L: Psychosis and seizures following the injection of penicillin G procaine: Hoigne's syndrome. Am J Dis Child 1985; 139:335-337.
    291) Sinclair JC, Fox HA, & Lentz JF: Intoxication of the fetus by a local anesthetic. N Engl J Med 1965; 273:1172-1177.
    292) Sinisterra S, Miravet E, Alfonso I, et al: Methemoglobinemia in an infant receiving nitric oxide after the use of eutectic mixture of local anesthetic. J Pediatr 2002; 141(2):285-286.
    293) Smith FR & Kundahl PC: Intravenously administered lidocaine as cough depressant during general anesthesia for bronchography. Chest 1973; 63:427-429.
    294) Smith HM, Jacob AK, Segura LG, et al: Simulation education in anesthesia training: a case report of successful resuscitation of bupivacaine-induced cardiac arrest linked to recent simulation training. Anesth Analg 2008; 106(5):1581-1584.
    295) Smith M, Wolfram W, & Rose R: Toxicity--seizures in an infant caused by (or related to) oral viscous lidocaine use. J Emerg Med 1992; 10:587-590.
    296) Spellberg MA: Hexylcaine (CYCLAINE(R)) as topical anesthetic in gastroscopy and esophagoscopy. Gastroenterology 1959; 36:120-121.
    297) Spence AG: Lipid reversal of central nervous system symptoms of bupivacaine toxicity. Anesthesiology 2007; 107(3):516-517.
    298) Spiller HA, Baker SD, Smith GW, et al: Evaluation of pramoxine ingestion as reported to poison centers. Ann Pharmacother 2006; 40(5):996-997.
    299) Spoerel WE, Adamson DH, & Eberhard RS: The significance of methaemoglobinaemia induced by prilocaine (Citanest). Can Anaes Soc J 1967; 14:1-10.
    300) 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.
    301) Stancavage M, Osterhoudt KC, & Henretig FM: Caladryl ingestion: a new poisoning from a common household product (abstract). J Tox Clin Tox 1995; 33:520.
    302) Stanford SC , Stanford BJ , & Gillman PK : Risk of severe serotonin toxicity following co-administration of methylene blue and serotonin reuptake inhibitors: an update on a case report of post-operative delirium. J Psychopharmacol 2010; 24(10):1433-1438.
    303) Stargel WW, Rue CR, & Routledge PA: Importance of blood-collection tubes in plasma lidocaine determinations. Clin Chem 1979; 25:617-619.
    304) Stewart J, Kellett N, & Castro D: The central nervous system and cardiovascular effects of levobupivacaine and ropivacaine in healthy volunteers. Anesth Analg 2003; 97(2):412-416.
    305) Stymne B & Lillieborg S: Plasma concentrations of lignocaine and prilocaine after a 24-h application of analgesic cream (EMLA(R)) to leg ulcers. Br J Dermatol 2001; 145:530-534.
    306) Sugai K : Treatment of convulsive status epilepticus in infants and young children in Japan. Acta Neurol Scand Suppl 2007; 186:62-70.
    307) Sundaram MBM: Seizures after intraurethral instillation of lidocaine. CMAJ 1987; 137:219-220.
    308) Sunshine I & Fike WW: Value of thin-layer chromatography in two fatal cases of intoxication due to lidocaine and mepivacaine. N Engl J Med 1964; 271:487-490.
    309) Sutter M & Pfenninger J: Adult respiratory distress syndrome (ARDS) nach intoxikation durch lokalanasthetika. Schweiz Med Wschr 1982; 112:1710-1713.
    310) Tei V, Morita T, Shishido H, et al: Lidocaine intoxication at very small doses in terminally ill cancer patients. J Pain Symptom Mgt 2005; (30)1:6-7.
    311) Teunis BS, Leftwich EI, & Pierce LE: Acute methemoglobinemia and hemolytic anemia due to toluidine blue. Arch Surg 1970; 101:527-531.
    312) Thomas RD, Behbehani MM, & Coyle DE: Cardiovascular toxicity of local anesthetics: an alternative hypothesis. Anesth Analg 1986; 65:444-450.
    313) Thompson PO, Melmon KL, & Richardson JA: Lidocaine pharmacokinetics in advanced heart failure, liver disease, and renal failure in humans. Ann Intern Med 1973; 78:499-508.
    314) Thompson TM & Theobald JL : Hoigne syndrome: a little-known adverse effect of lidocaine. Am J Emerg Med 2016; 34(3):679-4.
    315) Thyen AB, McAllister RK, & Councilman LM: Epidural pump programming error leading to inadvertent 10-fold dosing error during epidural labor analgesia with ropivacaine. J Patient Saf 2010; 6(4):244-246.
    316) Touma S & Jackson JB: Lidocaine and prilocaine toxicity in a patient receiving treatment for mollusca contagiosa. J Am Acad Dermatol 2001; 44:399-400.
    317) Tsui BC, Malherbe S, Koller J, et al: Reversal of an unintentional spinal anesthetic by cerebrospinal lavage. Anesth Analg 2004; 98(2):434-6, table.
    318) Tuominen MK, Pere P, & Rosenberg PH: Unintentional arterial catheterization and bupivacaine toxicity associated with continuous interscalene brachial plexus block. Anesthesiology 1991; 75:356-358.
    319) U.S. Food and Drug Administration: FDA Drug Safety Communication: Serious CNS reactions possible when methylene blue is given to patients taking certain psychiatric medications. U.S. Food and Drug Administration. Silver Spring, MD. 2011. Available from URL: http://www.fda.gov/Drugs/DrugSafety/ucm263190.htm. As accessed 2011-07-26.
    320) USAN, 1997
    321) Usubiaga JE, Wikinski J, & Ferrero R: Local anesthetic-induced convulsions in man. Anesth Analg 1966; 45:611-620.
    322) Van Ketel WG: Allergy to pramoxine (pramocaine). Contact Dermatitis 1981; 7:49.
    323) Wang RY & Chang WK: Inadvertent spinal anesthesia following paracervical blockade (abstract). Vet Hum Toxicol 1993; 35:347.
    324) Ward JT: Endotracheal drug therapy. Am J Emerg Med 1983; 1:71-82.
    325) Warren JA, Thoma RB, Georgescu A, et al: Intravenous lipid infusion in the successful resuscitation of local anesthetic-induced cardiovascular collapse after supraclavicular brachial plexus block. Anesth Analg 2008; 106(5):1578-80, table.
    326) Webber SK, Sutton GL, & Lawless MA: Ring keratitis from topical anesthetic misuse. Aust NZ J Ophthalmol 1999; 27:440-442.
    327) Wehner D & Hamilton GC: Seizures following topical application of local anesthetics to burn patients. Ann Emerg Med 1984; 13:456-458.
    328) Weinberg G, Hertz P, & Newman J: Lipid, not propofol, treats bupivacaine overdose. Anesth and Analgesia 2004; 99(6):1875-1876.
    329) Weinberg G: Lipid rescue resuscitation from local anaesthetic cardiac toxicity. Toxicol Rev 2006; 25(3):139-145.
    330) Weinberg GL, Ripper R, Murphy P, et al: Lipid infusion accelerates removal of bupivacaine and recovery from bupivacaine toxicity in the isolated rat heart. Reg Anesth Pain Med 2006; 31(4):296-303.
    331) Weiss LD, Generalovich T, Heller MB, et al: Methemoglobin levels following intravenous lidocaine administration. Ann Emerg Med 1987; 16:323-325.
    332) Whited C & Cohen SM: Nasal tetracaine spray-induced methemoglobinemia. Otolaryngol Head Neck Surg 2012; 146(4):678-679.
    333) Whiteside J: Reversal of local anaesthetic induced CNS toxicity with lipid emulsion. Anaesthesia 2008; 63(2):203-204.
    334) Wikinski A, Usubiaga JE, & Wikinski RW: Cardiovascular and neurological effects of 4000 mg of procaine. JAMA 1970; 213:621-623.
    335) Windholz M: The Merck Index, 10th ed, Merck & Co, Inc, Rahway, NJ, 1983.
    336) Winek CL, Collom WD, & Martineau P: Toluidine blue intoxication. Clin Toxicol 1969; 2:1-3.
    337) Woelke BJ & Tucker RA: ARDS after local lidocaine administration. Chest 1983; 3:933-934.
    338) Wolff AP, Hasenbos MA, & Liem TH: Accidental overdose of epidural bupivacaine and sufentanil. Reg Anesth 1992; 17:237-238.
    339) Wong BYS & Hurwitz A: Simple method for maintaining serum lidocaine levels in the therapeutic range. Arch Intern Med 1985; 145:1588-1591.
    340) Wood RA: Sinoatrial arrest: an interaction between phenytoin and lignocaine. Br Med J 1971; 20:645.
    341) Writer WDR, Davies JM, & Strunin L: Trial by media: the bupivacaine story. Can Anesth Soc J 1984; 31:1-4.
    342) Wurdeman RL, Mohiuddin SM, & Holmberg MJ: Benzocaine-induced methemoglobinemia during an outpatient procedure. Pharmacother 2000; 20:735-738.
    343) Yamashita S, Sato S, Kakiuchi Y, et al: Lidocaine toxicity during frequent viscous lidocaine use for painful tongue ulcer. J Pain Symptom Manage 2002; 24(5):543-545.
    344) Yan AC & Newman RD: Bupivacaine-induced seizures and ventricular fibrillation in a 13-year-old girl undergoing wound debridement. Ped Emerg Care 1998; 14:354-355.
    345) Yeniad B, Canturk S, Esin Ozdemir F, et al: Toxic keratopathy due to abuse of topical anesthetic drugs. Cutan Ocul Toxicol 2010; 29(2):105-109.
    346) Yildiz B , Citak A , Ucsel R , et al: Lidocaine treatment in pediatric convulsive status epilepticus. Pediatr Int 2008; 50(1):35-39.
    347) Yukioka H, Hayashi M, & Fujimori M: Lidocaine intoxication during general anesthesia (letter). Anesth Analg 1990; 71:200-212.
    348) Zeisler JA, Gaarder TD, & De Mesquita SA: Lidocaine excretion in breast milk. Drug Intell Clin Pharm 1986; 20:691-693.
    349) Zelicof-Paul A, Smith-Lockridge A, Schnadower D, et al: Controversies in rapid sequence intubation in children. Curr Opin Pediatr 2005; 17(3):355-362.
    350) Zuberi BF, Shaikh MR, & Jatoi NUN: Lidocaine toxicity in a student undergoing upper gastrointestinal endoscopy. Gut 2000; 46:435.
    351) 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.
    352) do Nascimento TS, Pereira RO, de Mello HL, et al: Methemoglobinemia: from diagnosis to treatment. Rev Bras Anestesiol 2008; 58(6):651-664.