MOBILE VIEW  | 

CALCIUM ANTAGONISTS

Classification   |    Detailed evidence-based information

Therapeutic Toxic Class

    A) Calcium antagonists (calcium channel blockers; slow channel blockers) include derivatives from four structural classes: the 1,4 dihydropyridines, the phenylalkylamines, the benzothiazepines, and the diarylaminopropylamine ethers. They block the influx of calcium into various cells, primarily vascular, cardiac, and smooth muscle tissue. They are used primarily for treatment of supraventricular tachycardia, angina, and hypertension.
    B) The following agents have their own specific managements; please refer to them as indicated:
    1) Amlodipine
    2) Diltiazem
    3) Felodipine
    4) Nifedipine
    5) Verapamil

Specific Substances

    A) GENERAL TERMS
    1) Antagonist, calcium
    2) Calcium antagonist drugs
    3) Calcium blockers
    4) Calcium channel blockers
    ARANIDIPINE (synonym)
    1) CAS 86780-90-7
    BARNIDIPINE (synonym)
    1) CAS 104713-75-9
    BENIDIPINE (synonym)
    1) Benidipine Hydrochloride
    2) CAS 105979-17-7 (benidipine)
    3) CAS 91599-74-5 (benidipine hydrochloride)
    CLEVIDIPINE (synonym)
    1) Clevidipine butyrate
    ISRADIPINE (synonym)
    1) Isrodipine
    2) PN-200-110
    3) Isopropyl methyl-4-(4-benzofurazanyl)-1,
    4) 4-dihydro-2,6-dimethyl 1-3,-5-
    5) pyridinedicarboxylate
    6) CAS 75695-93-1
    LACIDIPINE (synonym)
    1) Lacidipino
    2) GR-43659X
    3) GX-1048
    4) Molecular Formula: C(26)H(33)NO(6)
    5) CAS 103890-78-4
    LERCANIDIPINE (synonym)
    1) CAS 100427-26-7
    NICARDIPINE (synonym)
    1) RS 69216
    2) RS 69216-XX-07-0
    3) YC 93
    4) 2-(N-Methylbenzylamino)ethyl methyl 1,4-dihydro-2,
    5) 6-dimethyl-4-(3-nitrophenyl) pyridine-3, 5-dicarboxylate hydrochloride
    6) CAS 55985-32-5 (nicardipine)
    7) CAS 5427-84-3 (nicardipine hydrochloride)
    NILVADIPINE (synonym)
    1) CAS 75530-68-6
    NIMODIPINE (synonym)
    1) Bay-e-9736
    2) Isopropyl 2-methoxyethyl-1,4-dihydro-2,
    3) 6-dimethyl-4-(3-nitrophenyl)pyridine-3, 5-dicarboxylate
    4) CAS 66085-59-4
    NISOLDIPINE (synonym)
    1) 3,5-pyridinedicarboxylic acid, 1,4-dihydro-2,
    2) 6-dimethyl-4-(2-nitrophenyl)-methyl
    3) 2-methylpropyl ester
    NITRENDIPINE (synonym)
    1) CAS 39562-70-4
    PERHEXILINE (synonym)
    1) 2-(2,2-Dicyclohexylethyl)piperidine hydrogen maleate
    2) WSM-3978G
    3) CAS 6621-47-2 (perhexiline)
    4) CAS 6724-53-4 (perhexiline maleate)
    TIAPAMIL (synonym)
    1) CAS 57010-31-8

    1.2.1) MOLECULAR FORMULA
    1) CLEVIDIPINE: C21-H23-Cl2-N-O6 (Prod Info Cleviprex(R) intravenous injection, 2011)
    2) ISRADIPINE: C19-H21-N3-O5 (Prod Info isradipine oral capsules, 2011)
    3) NICARDIPINE HYDROCHLORIDE: C26-H29-N3-O6.HCl (Prod Info nicardipine HCl oral capsules, 2013)
    4) NIMODIPINE: C21-H26-N2-O7 (Prod Info nimodipine oral capsules, 2012)
    5) NISOLDIPINE: C20-H24-N2-O6 (Prod Info SULAR(R) extended release oral tablets, 2010)

Available Forms Sources

    A) FORMS
    1) IMMEDIATE-RELEASE PRODUCTS
    a) CLEVIDIPINE: 0.5 mg/mL as an injectable emulsion in 50 mL and 100 mL single-use vials (Prod Info Cleviprex(R) intravenous injection, 2011).
    b) ISRADIPINE: 2.5 mg and 5 mg capsules (Prod Info isradipine oral capsules, 2011).
    c) NICARDIPINE: 20 mg and 30 mg capsules (Prod Info nicardipine HCl oral capsules, 2013); 25 mg/10 mL (2.5 mg/mL) injection as single dose vials (Prod Info nicardipine HCl intravenous injection solution, 2012).
    d) NIMODIPINE: 30 mg capsules (Prod Info nimodipine oral capsules, 2012) and 60 mg/20 mL oral solution (Prod Info NYMALIZE(TM) oral solution, 2013).
    2) SUSTAINED-RELEASE PRODUCTS
    a) NICARDIPINE: 30 mg, 45 mg, and 60 mg sustained-release capsules (Prod Info CARDENE(R) SR oral sustained release capsules, 2010).
    b) NISOLDIPINE: 8.5 mg, 17 mg, 25.5 mg, and 34 mg extended-release tablets (Prod Info SULAR(R) extended release oral tablets, 2010).
    3) MIBEFRADIL DIHYDROCHLORIDE
    a) Mibefradil was removed from the market in June of 1998 because of the potential for numerous drug interactions.
    B) USES
    1) CLEVIDIPINE: Indicated for the management of hypertension (Prod Info Cleviprex(R) intravenous injection, 2011).
    2) ISRADIPINE: Indicated for the management of hypertension (Prod Info isradipine oral capsules, 2011).
    3) NICARDIPINE: Indicated for the treatment of stable angina and hypertension (Prod Info nicardipine HCl oral capsules, 2013).
    4) NIMODIPINE is used to improve neurological outcome after subarachnoid hemorrhage (Prod Info nimodipine oral capsules, 2012; Prod Info NYMALIZE(TM) oral solution, 2013).
    a) Nimodipine capsules should not be used by intravenous or other parenteral routes. Deaths and serious adverse events, including cardiac arrest, cardiovascular collapse, hypotension, and bradycardia have been reported following the intravenous administration of the contents of nimodipine capsules. If the patient can not swallow the capsule, the content of a capsule may be emptied by making a hole in both ends of a capsule with an 18-gauge needle. The contents should then be administered through the patient's naso-gastric tube and washed down the tube with 30 mL of normal saline solution (0.9%) (US Food and Drug Administration, 2006).
    5) NISOLDIPINE: Indicated for the treatment of hypertension (Prod Info SULAR(R) extended release oral tablets, 2010).

Life Support

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

Clinical Effects

    0.2.1) SUMMARY OF EXPOSURE
    A) USES: Class of agents used to primarily treat hypertension, dysrhythmias, and stable angina. The following agents have their own specific managements, please refer to them as indicated: amlodipine, diltiazem, felodipine, nifedipine, and verapamil.
    B) PHARMACOLOGY: Binds to and antagonizes L-type calcium channels located on all types of muscle cells resulting in relaxation of vascular smooth muscle and arterial vasodilation as well as decreased force of cardiac contraction and decreased heart rate and conduction.
    C) EPIDEMIOLOGY: Common overdose, which may result in significant morbidity and mortality.
    D) WITH THERAPEUTIC USE
    1) COMMON: Minor gastrointestinal effects, headache, and rash are commonly reported.
    E) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: Patients may have asymptomatic bradycardia or mild hypotension which may manifest as dizziness, fatigue, and/or lightheadedness.
    2) SEVERE TOXICITY: Can have profound bradycardia and dysrhythmias (including complete heart block) and hypotension resulting in cardiogenic shock and end-organ dysfunction including lethargy, syncope, altered mental status, seizures, cerebral ischemia, bowel ischemia, renal failure, metabolic acidosis, coma, and death. Hyperglycemia generally develops in patients with severe poisoning.
    0.2.3) VITAL SIGNS
    A) WITH POISONING/EXPOSURE
    1) Fever was reported following a benidipine ingestion.
    0.2.20) REPRODUCTIVE
    A) Clevidipine, isradipine, niCARdipine, nimodipine, and nisoldipine are classified as FDA pregnancy category C. In animal studies, teratogenicity has been reported with nimodipine.

Laboratory Monitoring

    A) Monitor vital signs frequently. Institute continuous cardiac monitoring and obtain serial ECGs.
    B) In patients with significant hypotension or bradycardia, monitor serum electrolytes, renal function, arterial or venous blood gas, and urine output.
    C) Obtain digoxin level in patients who also have access to digoxin.
    D) Monitor cardiac enzymes in patients with chest pain.
    E) Serum drug levels are not readily available and thus not immediately helpful.

Treatment Overview

    0.4.2) ORAL/PARENTERAL EXPOSURE
    A) MANAGEMENT OF MILD TO MODERATE TOXICITY
    1) Patients who have asymptomatic bradycardia can be admitted and observed with telemetry. Obtain peripheral intravenous access and monitor ECG. Mild hypotension may only require treatment with intravenous fluid administration.
    B) MANAGEMENT OF SEVERE TOXICITY
    1) Patients with bradycardia and hypotension require standard ACLS treatment. Place a central line and consider placement of an arterial line. Standard first line treatment includes atropine for bradycardia although in a serious poisoning it is rarely effective. High dose insulin and dextrose have been effective in animal studies and multiple case reports in patients with hypotension refractory to other modalities, and should be considered early in patients with significant hypotension. Use intravenous calcium in severe poisonings although in these cases, beneficial effects of calcium infusion (calcium chloride is preferred) may be very minimal or short-lived. Repeat bolus doses or a continuous intravenous infusion are often needed. Standard vasopressors should be administered to maintain blood pressure. Lipid emulsion has been successful in animal studies and several case reports of patients with hypotension refractory to other therapies. Intravenous glucagon has been used with variable success. In a patient whose hemodynamic status continues to be refractory despite the treatment described above, extracorporeal membrane oxygenation or cardiopulmonary bypass should be considered. Treat seizures with IV benzodiazepines; barbiturates or propofol may be needed if seizures persist or recur.
    C) DECONTAMINATION
    1) PREHOSPITAL: Not recommended because of the potential for abrupt deterioration.
    2) HOSPITAL: Because a calcium channel blocker overdose can be life-threatening, all significant ingestions should receive activated charcoal. Patients with altered mental status should be intubated prior to administration. Gastric lavage should be considered in patients with recent large ingestions if the airway is protected. Late gastric lavage may be effective following sustained-release products. Whole bowel irrigation should be considered early for patients who can protect their airway or who are intubated who have ingested sustained-release formulations; it can limit absorption from possible concretions. Whole bowel irrigation should NOT be performed in patients who are hemodynamically unstable.
    D) AIRWAY MANAGEMENT
    1) Intubate patients with coma, mental status depression or significant hemodynamic instability.
    E) HYPOTENSION
    1) Treat initially with fluids (insert a central venous or pulmonary artery catheter to guide fluid therapy if hypotension persists). Consider inserting an arterial line in patients with refractory hypotension. Intravenous calcium, vasopressors, high dose insulin/dextrose, intravenous lipid emulsion, and glucagon may all be useful for refractory hypotension. Pacemakers (external or internal), intraaortic balloon pump, and cardiopulmonary bypass have been used in patients refractory to other modalities.
    2) CALCIUM
    a) Intravenous calcium infusions have been shown to be helpful, although response is often short lived. Optimal dosing is not established; start with an initial IV infusion of about 13 to 25 mEq of calcium (10 to 20 mL of 10% calcium chloride or 30 to 60 mL of 10% calcium gluconate) followed by either repeat boluses every 15 to 20 minutes up to 3 to 4 doses or a continuous infusion starting with 0.5 mEq/kg/hr of calcium (0.2 to 0.4 mL/kg/hr of 10% calcium chloride or 0.6 to 1.2 mL of 10% calcium gluconate) and titrate as needed. Calcium dosing should be titrated to hemodynamic response rather than serum calcium concentration alone; central venous or pulmonary artery catheters may be useful to guide therapy. Monitor ECG and ionized calcium concentration. Patients with severe overdose have tolerated significant hypercalcemia (up to twice the upper limit of normal) without developing clinical or ECG evidence of hypercalcemia.
    3) INSULIN
    a) Administer a bolus of 1 unit/kg of insulin followed by an infusion of 0.1 to 1 unit/kg/hr, titrated to a systolic blood pressure of greater than 90 to 100 mmHg (bradycardia may or may not respond). Reassess every 30 minutes to titrate insulin infusion. Administer dextrose bolus to patients with an initial blood glucose of less than 250 mg/dL (adults 25 to 50 mL dextrose 50%, children 0.25 g/kg dextrose 25%). Begin a dextrose infusion of 0.5 g/kg/hr in all patients. Monitor blood glucose every 15 to 30 minutes until consistently 100 to 200 mg/dL for 4 hours, then monitor every hour. Titrate dextrose infusion to maintain blood glucose in the range of 100 to 200 mg/dL. As the patient improves, insulin resistance abates and dextrose requirements will increase. Supplemental dextrose will be needed for at least several hours after the insulin infusion is discontinued. Administer supplemental potassium initially if patient is hypokalemic (serum potassium less than 2.5 mEq/L). Monitor serum potassium every 4 hours and supplement as needed to maintain potassium of 2.5 to 2.8 mEq/L.
    4) VASOPRESSORS
    a) Anecdotal reports suggest that epinephrine, vasopressin, metaraminol, or phenylephrine may occasionally be effective in patients who do not respond to dopamine or norepinephrine.
    5) FAT EMULSION
    a) Lipid emulsion has been successful in animal studies and several case reports of patients with hypotension refractory to other therapies. 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.
    6) GLUCAGON
    a) DOSE: ADULT: Optimal dosing in calcium antagonist poisoning is not established. Initially, 3 to 5 mg IV, slowly over 1 to 2 minutes; may repeat treatment with a dose of 4 to 10 mg if there is no hemodynamic improvement within 5 minutes. CHILD: 50 mcg/kg; repeat doses may be used due to the short half-life of glucagon.
    7) L-CARNITINE
    a) L-carnitine may be useful to treat hypotension in the setting of calcium channel blocker overdose. It is not well studied but an animal study and one human case report suggest efficacy. The dose used in the human case report was 6 g IV followed by 1 g IV every 4 hours.
    8) PHOSPHODIESTERASE INHIBITORS
    a) There are case reports where a phosphodiesterase inhibitor (inamrinone, enoximone) appeared to improve blood pressure in patients unresponsive to other modalities.
    F) ENHANCED ELIMINATION
    1) Hemodialysis is likely not of value because of the high degree of protein binding and large volume of distribution.
    G) PATIENT DISPOSITION
    1) HOME CRITERIA: According to the AAPCC guidelines, healthy, asymptomatic patients with the following inadvertent single substance ingestions may be monitored at home (IR = immediate release, SR = sustained release):
    a) BEPRIDIL: ADULT: 300 mg or less; CHILD: no safe dose
    b) ISRADIPINE: ADULT: 20 mg or less; CHILD: 0.1 mg/kg or less
    c) NICARDIPINE: ADULT: 40 mg or less IR or chewed SR, or 60 mg or less SR; CHILD: less than 1.25 mg/kg
    d) NIMODIPINE: ADULT: 60 mg or less; CHILD: no safe dose
    e) NISOLDIPINE: ADULT: 30 mg or less; CHILD: no safe dose
    2) OBSERVATION CRITERIA: Symptomatic patients, those with underlying cardiovascular disease, those taking beta blockers or another cardiodepressant drug, and those with deliberate ingestions should be referred to a health care facility for treatment, evaluation and monitoring. Patients who have not developed signs or symptoms more than 6 hours after ingestion of an immediate-release product, 18 hours after ingestion of a sustained or extended-release product are unlikely to develop toxicity.
    a) According to the AAPCC guidelines, patients with the following inadvertent single substance ingestions should be referred to a healthcare facility (IR = immediate release, SR = sustained release):
    1) BEPRIDIL: ADULT: greater than 300 mg; CHILD: any amount
    2) ISRADIPINE: ADULT: greater than 20 mg; CHILD: greater than 0.1 mg/kg
    3) NICARDIPINE: ADULT: greater than 40 mg IR or chewed SR, or greater than 60 mg SR; CHILD: 1.25 mg/kg or more
    4) NIMODIPINE: ADULT: greater than 60 mg; CHILD: any amount
    5) NISOLDIPINE: ADULT: greater than 30 mg; CHILD: any amount
    3) ADMISSION CRITERIA: Patients who develop signs or symptoms of toxicity should be admitted to an intensive care setting.
    4) CONSULT CRITERIA: Consult a poison center or medical toxicologist in cases of severe poisonings or in cases where there is a history of a large exposure.
    5) For specific patient disposition information regarding AMLODIPINE, DILTIAZEM, NIFEDIPINE, and VERAPAMIL, refer to the individual document.
    H) PITFALLS
    1) Focus on antidote treatment should not be done in lieu of initially following standard ACLS protocols for treatment of bradycardia and hypotension. In severely poisoned patients, treatment should be aggressive, and the treatments and antidotes described above may need to be started simultaneously. Consider coingestants with other cardiopulmonary medications such as digoxin since these patients may be on multiple medications. Toxicity can be delayed and prolonged after overdose of modified release formulations.
    I) PHARMACOKINETICS
    1) Onset of action of the majority of agents is within 1 to 2 hours. Peak effects typically occur 6 to 12 hours after ingestion with controlled-release preparations. Most calcium channel blockers have large volumes of distribution and are highly protein bound.
    2) For specific pharmacokinetic information regarding AMLODIPINE, DILTIAZEM, NIFEDIPINE, and VERAPAMIL, refer to the individual document.
    J) TOXICOKINETICS
    1) Chewing or crushing modified release formulations may result in rapid absorption of the entire dose and subsequent toxicity. Hypotension and bradycardia generally develop within 6 hours after overdose of regular release products. Toxicity can be delayed, especially with overdoses of sustained release preparations and has occurred 15 to 22 hours after ingestion. In addition, duration of effect can be quite prolonged following overdose.
    2) For specific toxicokinetic information regarding AMLODIPINE, DILTIAZEM, NIFEDIPINE, and VERAPAMIL, refer to the individual document.
    K) DIFFERENTIAL DIAGNOSIS
    1) Ingestion of other cardioactive drugs (especially beta-blockers and digoxin) should be considered in a patient who is bradycardic and hypotensive.

Range Of Toxicity

    A) TOXICITY: The toxic dose is variable depending on the particular agent. The following doses are considered to be potentially toxic (IR = immediate release, SR = sustained release): BEPRIDIL: ADULT: greater than 300 mg; CHILD: any amount. ISRADIPINE: ADULT: greater than 20 mg; CHILD: greater than 0.1 mg/kg. NICARDIPINE: ADULT: greater than 40 mg IR or chewed SR, or greater than 60 mg SR; CHILD: 1.25 mg/kg or more. NIMODIPINE: ADULT: greater than 60 mg; CHILD: any amount. NISOLDIPINE: ADULT: greater than 30 mg; CHILD: any amount. Single ingestions of therapeutic adult doses in children have resulted in death. Patients with underlying cardiovascular disease and the elderly tend to be more susceptible to the cardiac effects. In general, ingestions of phenylalkylamines (eg, verapamil) and benzothiazepines (eg, diltiazem) are more serious than ingestions of dihydropyridines (eg, niCARdipine).

Summary Of Exposure

    A) USES: Class of agents used to primarily treat hypertension, dysrhythmias, and stable angina. The following agents have their own specific managements, please refer to them as indicated: amlodipine, diltiazem, felodipine, nifedipine, and verapamil.
    B) PHARMACOLOGY: Binds to and antagonizes L-type calcium channels located on all types of muscle cells resulting in relaxation of vascular smooth muscle and arterial vasodilation as well as decreased force of cardiac contraction and decreased heart rate and conduction.
    C) EPIDEMIOLOGY: Common overdose, which may result in significant morbidity and mortality.
    D) WITH THERAPEUTIC USE
    1) COMMON: Minor gastrointestinal effects, headache, and rash are commonly reported.
    E) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: Patients may have asymptomatic bradycardia or mild hypotension which may manifest as dizziness, fatigue, and/or lightheadedness.
    2) SEVERE TOXICITY: Can have profound bradycardia and dysrhythmias (including complete heart block) and hypotension resulting in cardiogenic shock and end-organ dysfunction including lethargy, syncope, altered mental status, seizures, cerebral ischemia, bowel ischemia, renal failure, metabolic acidosis, coma, and death. Hyperglycemia generally develops in patients with severe poisoning.

Vital Signs

    3.3.1) SUMMARY
    A) WITH POISONING/EXPOSURE
    1) Fever was reported following a benidipine ingestion.
    3.3.3) TEMPERATURE
    A) WITH POISONING/EXPOSURE
    1) FEVER (38 degrees C) was reported in a 16-month-old child 2 hours after an inadvertent ingestion of two 4-mg benidipine tablets (0.8 mg/kg) (Akbayram et al, 2012).

Cardiovascular

    3.5.2) CLINICAL EFFECTS
    A) CARDIOVASCULAR FINDING
    1) WITH POISONING/EXPOSURE
    a) NIMODIPINE: Deaths and serious adverse events, including cardiac arrest, cardiovascular collapse, hypotension, and bradycardia have been reported following the intravenous administration of the contents of nimodipine capsules (US Food and Drug Administration, 2006).
    B) HYPOTENSIVE EPISODE
    1) WITH POISONING/EXPOSURE
    a) SIGNS/SYMPTOMS: Hypotension is common following significant overdose. Hypotension may be quite severe and unresponsive to pressor agents. Syncopal episodes secondary to impaired perfusion may occur (Devasahayam et al, 2012; Wood et al, 2005; Saravu & Balasubramanian, 2004; Gerloni & Copetti, 2004; Durward et al, 2003; SamiKarti et al, 2002; Schwab et al, 2002; Vadlamudi & Wijdicks, 2002; Herbert et al, 2001; Meyer et al, 2001; Cavagnaro et al, 2000; Ori et al, 2000; Luscher et al, 1994; Ramoska et al, 1993; Welch et al, 1992) .
    b) ONSET: Common within 1 to 5 hours postingestion, although delayed onset (more than 24 hours) and prolonged duration of symptoms can occur following sustained-release dosage forms (Tuka et al, 2009; Morimoto et al, 1999; Luscher et al, 1994; Buckley et al, 1993; Quezado et al, 1991; Spiller et al, 1991; Krick et al, 1990; de Faire & Lundman, 1978) .
    c) DURATION: In one series, 80% of cases were asymptomatic for all cardiovascular effects after 24 hours (Ramoska et al, 1993).
    d) LERCANIDIPINE/CASE REPORT: A 49-year-old man developed bradycardia and severe hypotension (77/40 mmHg), refractory to IV fluids, calcium and glucagon, after intentionally ingesting 560 mg of slow-release lercanidipine. Following hyperinsulinemic euglycemia therapy and norepinephrine administration, the patient's blood pressure normalized and he was discharged approximately 2 days post-ingestion (Hadjipavlou et al, 2011).
    C) CONDUCTION DISORDER OF THE HEART
    1) SUMMARY
    a) Electrophysiologic effects of calcium antagonists vary among verapamil, diltiazem, nifedipine, and amlodipine (Mitchell et al, 1982). In overdose, all agents can cause rhythm disturbances and conduction defects. Nifedipine, amlodipine, and other dihydropyridines lack the effects of other structural classes of calcium antagonists on AV nodal conduction. Therefore, these agents are more likely to result in reflex tachycardia secondary to diminished perfusion; bradycardia is twice as likely with verapamil and diltiazem. AV block, especially greater than first degree, is predominately a finding with verapamil.
    2) AV BLOCK
    a) FINDINGS: ECG manifestations may occur, including heart block, first-, second-, and third-degree AV block, junctional rhythm, QT interval prolongation, moderate S-T segment depression, low amplitude T-waves, prominent U-waves, and atrial fibrillation (Prod Info isradipine oral capsules, 2011; Henrikson & Chandra-Strobos, 2003; SamiKarti et al, 2002; Schwab et al, 2002; Ori et al, 2000; De Cicco et al, 1999; Howarth et al, 1994; Luscher et al, 1994; Quezado et al, 1991; McMillan, 1988; Moroni et al, 1980) .
    b) ONSET: AV block usually occurs 4 to 24 hours postexposure (Spiller et al, 1991; da Silva et al, 1979) .
    c) DURATION: Cardiac disturbances commonly persist for 9 to 48 hours, but have been reported to last as long as 7 days (Quezado et al, 1991). In one series, 80% of cases were asymptomatic for all cardiovascular effects after 24 hours (Ramoska et al, 1993).
    d) MIBEFRADIL/CASE REPORT: Sinus arrest occurred in a 78-year-old woman 4 days after beginning mibefradil therapy, 100 mg daily. The patient was initially managed with transvenous pacing until return of normal sinus rhythm approximately 36 hours after discontinuation of mibefradil (Sanders et al, 1998).
    3) BRADYCARDIA
    a) FINDINGS: Heart rates below 60 beats/min with accompanying hypotension at presentation are common (Prod Info nicardipine HCl oral capsules, 2013; Hadjipavlou et al, 2011; Durward et al, 2003; SamiKarti et al, 2002; Vadlamudi & Wijdicks, 2002; Schwab et al, 2002; Meyer et al, 2001; Snook et al, 2000; Howarth et al, 1994), but isolated bradycardia has also been noted (Krick et al, 1990).
    b) ONSET: Common within 1 to 5 hours postingestion, although delayed onset (more than 6 hours) can occur following sustained-release dosage forms (Morimoto et al, 1999). (Barrow et al, 1994; Rodgers et al, 1989)
    c) DURATION: Bradycardia for 36 to 52 hours after admission has been reported (Barrow et al, 1994; Howarth et al, 1994; Perkins, 1978). In one series, 80% of cases were asymptomatic for all cardiovascular effects after 24 hours (Ramoska et al, 1993). Ingestion of sustained-release products may result in prolonged or delayed effects (Barrow et al, 1994; Luscher et al, 1994).
    d) CASE REPORTS
    1) MIBEFRADIL: Numerous reports of decreased heart rate have occurred in patients while receiving therapeutic doses of mibefradil, especially in the elderly who may be at risk for slower heart rates or patients on concomitant beta-blocker therapy (Rogers & Prpic, 1998; Anon, 1997).
    4) TACHYCARDIA
    a) FINDINGS: A reflex tachycardia has primarily been reported with nifedipine overdose, but may occur with other dihydropyridine calcium antagonists (Prod Info isradipine oral capsules, 2011; Saravu & Balasubramanian, 2004; Lee et al, 2000; Stanek et al, 1997; Cosbey & Carson, 1997; Aya et al, 1996a; Castro JrG, Tsai PWC & Ozaki MM et al, 1994; Ferner et al, 1990; Whitebloom & Fitzharris, 1988).
    b) INCIDENCE: In one series of 28 nifedipine toxicities, tachycardia was present in 16 (57%) as compared with approximately 25% of cases involving either verapamil or diltiazem (Ramoska et al, 1993).
    c) ONSET: Common within 1 to 5 hours postingestion, although delayed onset (more than 6 hours) can occur following sustained-release dosage forms (Ramoska et al, 1993).
    d) DURATION: In one series, 80% of cases were asymptomatic for all cardiovascular effects after 24 hours (Ramoska et al, 1993).
    e) NICARDIPINE/CASE REPORT: Sinus tachycardia was reported in a 39-year-old woman who was 36 weeks pregnant and inadvertently received an overdose infusion of niCARdipine at 20 to 25 mg/hour over a 10-hour period, instead of the prescribed 2 mg/hour, to treat severe hypertension (Aya et al, 1996).
    f) BENIDIPINE/CASE REPORT: Tachycardia (151 beats/min), metabolic acidosis, and fever were reported in a 16-month-old child following an inadvertent ingestion of two 4-mg benidipine tablets (0.8 mg/kg). With supportive therapy, the patient recovered and was discharged approximately 2 days post-ingestion (Akbayram et al, 2012).
    D) CARDIAC ARREST
    1) WITH POISONING/EXPOSURE
    a) ISRADIPINE/CASE REPORT: A 5-year-old child, who was taking isradipine 2.5 mg twice daily for treatment of hypertension, presented with bradycardia and abdominal distention. Thirty minutes after hospital admission, the patient developed a ventricular escape rhythm (20 bpm), became apneic, then developed asystole. The patient responded to cardiac resuscitation and temporary pacing, with spontaneous cardiac activity returning approximately 12 hours postadmission (Romano et al, 2002). Serum isradipine concentration, obtained 1 hour postadmission, was 260.7 ng/mL. Twenty hours later, a second serum isradipine concentration was 27.4 ng/mL. The normal therapeutic concentration in adults receiving multiple daily doses is 2.2 to 8.21 ng/mL.
    E) MYOCARDIAL INFARCTION
    1) WITH POISONING/EXPOSURE
    a) BARNIDIPINE/CASE REPORT: A 25-year-old woman presented to the hospital with dizziness, syncope, lethargy, confusion, hypotension (60/30 mmHg), and tachycardia (130 bpm) after intentionally ingesting 500 mg of barnidipine. An ECG demonstrated ST-segment depression, and troponin I concentration was elevated, establishing a diagnosis of non-ST-segment elevation myocardial infarction. Coronary angiography was normal, so the myocardial ischemia was likely secondary to profound hypotension and tachycardia. With supportive care, the patient recovered and was discharged approximately 10 days postingestion (Guvenc et al, 2010).
    F) SHOCK
    1) WITH POISONING/EXPOSURE
    a) NISOLDIPINE/CASE REPORT: Severe shock, hypothermia, and renal insufficiency were reported in a 72-year-old woman following a suspected overdose ingestion of nisoldipine. With supportive therapy, including administration of IV glucagon and calcium chloride, the patient recovered without sequelae. A serum nisoldipine concentration of 1544 mcg/L was obtained at admission, although the exact time of ingestion was unknown (Louagie et al, 1998).

Respiratory

    3.6.2) CLINICAL EFFECTS
    A) ACUTE LUNG INJURY
    1) WITH POISONING/EXPOSURE
    a) Noncardiogenic pulmonary edema has been reported following calcium antagonist overdose (Durward et al, 2003; Humbert et al, 1991; Ori et al, 2000; Howarth et al, 1994; Leesar et al, 1994; Spurlock et al, 1991; Spiller et al, 2012; Ghosh & Sircar, 2008; Saravu & Balasubramanian, 2004).
    b) RISK FACTORS: Fluid loading to maintain blood pressure and oliguria secondary to hypotension may contribute (Herrington et al, 1986; Barrow et al, 1994).
    B) HYPERVENTILATION
    1) WITH POISONING/EXPOSURE
    a) BENIDIPINE/CASE REPORT: Kussmaul respiration was reported in a 16-month-old child following an inadvertent ingestion of two 4-mg benidipine tablets (0.8 mg/kg) (Akbayram et al, 2012).
    C) HYPOXEMIA
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Hypoxemia (partial pressure oxygen (PO2) of 60 mmHg with fractional inspired oxygen of 50%) was reported in an 81-year-old woman following a suspected overdose ingestion of nimodipine. Transthoracic echocardiography demonstrated normal left ventricular systolic function and chest X-ray was normal. Within 20 minutes after receiving IV calcium gluconate, the patient's oxygenation improved to a PO2 of 113 mmHg with a fractional inspired oxygen of 50% (Gerloni & Copetti, 2004).

Neurologic

    3.7.2) CLINICAL EFFECTS
    A) CENTRAL NERVOUS SYSTEM DEFICIT
    1) FINDINGS: Drowsiness, mental confusion, lethargy, and lightheadedness are common (Spiller et al, 2012; Prod Info isradipine oral capsules, 2011; Prod Info CARDENE(R) SR oral sustained release capsules, 2010; Verbrugge & vanWezel, 2007; Gerloni & Copetti, 2004; Lee et al, 2000; Herrington et al, 1986; Moroni et al, 1980; Candell et al, 1979; da Silva et al, 1979) ; circulatory collapse and/or coingestion complicate evaluation of mental status.
    B) CEREBRAL ARTERY OCCLUSION
    1) SUMMARY: Cerebral infarction may develop during supportive care following acute ingestion (Shah & Passalacqua, 1992; Wells et al, 1990; Samniah & Schlaeffer, 1988).
    C) SEIZURE
    1) WITH POISONING/EXPOSURE
    a) Seizure activity has been reported with calcium antagonist overdose and may result from acidosis, anoxia, or an existing predisposition (Hadjipavlou et al, 2011; Isbister, 2002; Wells et al, 1990; Malcolm et al, 1986; Passal & Crespin, 1984).
    D) HEADACHE
    1) WITH THERAPEUTIC USE
    a) Headaches are a common adverse event with calcium antagonist therapy (Prod Info NYMALIZE(TM) oral solution, 2013; Prod Info nicardipine HCl oral capsules, 2013; Prod Info isradipine oral capsules, 2011; Prod Info Cleviprex(R) intravenous injection, 2011; Prod Info SULAR(R) extended release oral tablets, 2010).
    2) WITH POISONING/EXPOSURE
    a) CASE REPORT: Headache, palpitations, and flushing were reported in a 39-year-old 36 weeks pregnant woman who inadvertently received an infusion of 20 to 25 mg/hour of niCARdipine over a 10-hour period, instead of the prescribed 2 mg/hour, to treat severe hypertension. With supportive therapy, the patient's symptoms resolved. A cesarean section was performed, with the newborn exhibiting normal hemodynamic parameters (Aya et al, 1996).

Gastrointestinal

    3.8.2) CLINICAL EFFECTS
    A) NAUSEA
    1) Nausea and vomiting are common (Spiller et al, 2012; Belson et al, 2000; Morimoto et al, 1999; Candell et al, 1979; de Faire & Lundman, 1977) .
    B) BEZOAR
    1) Tablet concretions ranging in size from golf to tennis ball size developing from sustained-release dosage forms have been found at autopsy. Gastroscopy may be required for confirmation if suspected since these masses have not been apparent on abdominal films (Sporer & Manning, 1993).
    2) Sustained-release nifedipine and verapamil have been associated with the development of bezoars (Wells et al, 2006; Niezabitowski et al, 2000; Taylor et al, 1998; Rankin & Edwards, 1990) and may occur with sustained release formulations of other calcium antagonists.
    C) VASCULAR INSUFFICIENCY OF INTESTINE
    1) SUMMARY
    a) Bowel necrosis and mesenteric ischemia may occur in the absence of prolonged hypotension (Donovan et al, 1999).
    D) GASTROINTESTINAL HEMORRHAGE
    1) WITH THERAPEUTIC USE
    a) A case control study, performed by Kaplan et al (2000), indicated that therapeutic use of calcium channel blockers may be associated with a 2-fold increased risk of gastrointestinal bleeding as compared with users of beta-blockers (Kaplan et al, 2000).

Genitourinary

    3.10.2) CLINICAL EFFECTS
    A) ACUTE RENAL FAILURE SYNDROME
    1) WITH POISONING/EXPOSURE
    a) Acute renal failure has been reported, usually in patients who develop prolonged hypotension and/or rhabdomyolysis after severe poisoning (Ghosh & Sircar, 2008; Gokel et al, 2000; Williamson & Dunham, 1996; Quezado et al, 1991; Pearigen & Benowitz, 1991; McMillan, 1988).
    b) CASE REPORT: Renal insufficiency (serum creatinine of 2.67 mg/dL) was reported in a 72-year-old woman following a suspected overdose ingestion of nisoldipine. A serum nisoldipine concentration of 1544 mcg/L was obtained at admission, although the exact time of ingestion was unknown (Louagie et al, 1998).
    B) OLIGURIA
    1) WITH POISONING/EXPOSURE
    a) Transient oliguria is associated with prolonged hypotension (Fauville et al, 1995; Barrow et al, 1994).
    b) CASE REPORT: Anuria, as well as hypotension and hypoxemia, was reported in an 81-year-old woman following a suspected overdose ingestion of nimodipine. Following calcium gluconate administration, the patient's anuria resolved with improvement in her blood pressure and oxygenation (Gerloni & Copetti, 2004).

Acid-Base

    3.11.2) CLINICAL EFFECTS
    A) ACIDOSIS
    1) WITH POISONING/EXPOSURE
    a) BENIDIPINE/CASE REPORT: Metabolic acidosis (pH 7.10, pCO2 40 mmHg, pO2 29 mmHg, HCO3 15 mmol/L), tachycardia, Kussmaul respiration, and fever were reported in a 16-month-old child following an inadvertent ingestion of two 4-mg benidipine tablets (0.8 mg/kg). With supportive therapy, the patient recovered and was discharged approximately 2 days post-ingestion (Akbayram et al, 2012).
    2) Mild metabolic acidosis (pH 7.2 to 7.3) is common in patients with hypotension (Verbrugge & vanWezel, 2007; Fauville et al, 1995; McMillan, 1988; da Silva et al, 1979; de Faire & Lundman, 1977).

Dermatologic

    3.14.2) CLINICAL EFFECTS
    A) ERUPTION
    1) WITH THERAPEUTIC USE
    a) Minor skin rashes are reported for most calcium channel agents (Sun et al, 1994).
    B) FLUSHING
    1) WITH POISONING/EXPOSURE
    a) Flushing has been reported (Prod Info isradipine oral capsules, 2011; Prod Info CARDENE(R) SR oral sustained release capsules, 2010; Aya et al, 1996).

Musculoskeletal

    3.15.2) CLINICAL EFFECTS
    A) RHABDOMYOLYSIS
    1) WITH POISONING/EXPOSURE
    a) Rhabdomyolysis may occur in conjunction with acute renal failure following overdose ingestions of calcium antagonists (Gokel et al, 2000; Quezado et al, 1991).

Endocrine

    3.16.2) CLINICAL EFFECTS
    A) HYPERGLYCEMIA
    1) Serum glucose concentration correlates directly with the severity of calcium channel blocker intoxication; patients who develop hyperglycemia are likely to develop more severe cardiovascular manifestations of toxicity.
    2) A retrospective analysis, involving the measurement of hyperglycemia in adult patients following overdose ingestions of diltiazem or verapamil, showed an increase in the median peak serum glucose concentrations in those patients who met the composite endpoints measuring the severity of the overdose (ie, in-hospital mortality, necessity of a temporary pacemaker, or vasopressor therapy), as compared with those patients who did not meet the composite endpoints (364 mg/dl vs 145 mg/dl). The median increase in blood glucose was also greater in the patients who met the composite endpoints as compared to those patients who did not (71.2% vs 0%). Based on these results, there appears to be a direct correlation between serum glucose concentrations and the severity of calcium antagonist overdose, and may be useful as a predictor of the severity of the intoxication (Levine et al, 2007).
    3) CASE SERIES: In a series of 15 cases of calcium channel blocker overdose, 4 nondiabetic patients developed hyperglycemia (Howarth et al, 1994).

Reproductive

    3.20.1) SUMMARY
    A) Clevidipine, isradipine, niCARdipine, nimodipine, and nisoldipine are classified as FDA pregnancy category C. In animal studies, teratogenicity has been reported with nimodipine.
    3.20.2) TERATOGENICITY
    A) ANIMAL STUDIES
    1) NIMODIPINE
    a) RABBITS: In 2 animal studies, nimodipine has been shown to cause teratogenic malformations in rabbits such as stunted fetal growth unrelated to dose. Increased incidences of malformation and stunted fetuses were observed in 1 study with oral administration from day 6 through day 18 of pregnancy at doses of 1 and 10 mg/kg/day but not at 3 mg/kg/day. An increased incidence of stunted fetuses were reported in a second study at 1 mg/kg/day but not at higher doses of 3 and 10 mg/kg/day (Prod Info NYMALIZE(TM) oral solution, 2013; Prod Info nimodipine oral capsules, 2012).
    b) RATS: Resorption and stunted growth of fetuses were noted in rats administered oral nimodipine from day 6 through day 15 of pregnancy at doses of 100 mg/kg/day. Higher incidences of skeletal variation, stunted fetuses, and stillbirths were identified in 2 other rat studies using oral nimodipine from day 16 of gestation and continued until day 20 of pregnancy or day 21 postpartum at doses of 30 mg/kg/day, but no congenital malformations were evident at this dose (Prod Info NYMALIZE(TM) oral solution, 2013; Prod Info nimodipine oral capsules, 2012).
    3.20.3) EFFECTS IN PREGNANCY
    A) PREGNANCY CATEGORY
    1) Manufacturers have classified the following calcium antagonists as FDA pregnancy category C:
    1) CLEVIDIPINE (Prod Info Cleviprex(R) intravenous injection, 2011)
    2) ISRADIPINE (Prod Info isradipine oral capsules, 2011)
    3) NICARDIPINE (Prod Info nicardipine HCl oral capsules, 2013)
    4) NIMODIPINE (Prod Info NYMALIZE(TM) oral solution, 2013; Prod Info nimodipine oral capsules, 2012)
    5) NISOLDIPINE (Prod Info SULAR(R) extended release oral tablets, 2010)
    B) FETAL AND MATERNAL ADVERSE EFFECTS
    1) NICARDIPINE
    a) Pregnant, hypertensive women who were treated with IV niCARdipine experienced hypotension, reflex tachycardia, postpartum hemorrhage, tocolysis, headache, nausea, dizziness, and flushing. Fetal adverse effects ranged from none to transient fetal heart rate decelerations. In neonates, adverse events ranged from none to hypotension. During preterm labor, those treated with IV niCARdipine experienced pulmonary edema, dyspnea, hypoxia, hypotension, tachycardia, headache, and phlebitis at site of injection. Acidosis (pH less than 7.25) was reported in neonates (Prod Info nicardipine HCl intravenous injection solution, 2012).
    b) CASE REPORT: Headache, palpitations, and flushing were reported in a 39-year-old 36 weeks pregnant woman who inadvertently received an infusion of 20 to 25 mg/hour of niCARdipine over a 10-hour period, instead of the prescribed 2 mg/hour, to treat severe hypertension. With supportive therapy, the patient's symptoms resolved. A cesarean section was performed, with the newborn exhibiting normal hemodynamic parameters (Aya et al, 1996).
    C) PLACENTAL TRANSFER
    1) NICARDIPINE
    a) Low-level placental transfer of niCARdipine was observed in 10 of 40 hypertensive women, with no cord or serum accumulations evident in the fetus. Adverse fetal or neonatal outcomes in this study were not associated with niCARdipine. NiCARdipine is superior to nifedipine in that it acts more selectively on blood vessels than on myocardium, has a lesser negative inotropic effect, and produces less reflex tachycardia (Carbonne et al, 1993).
    D) LACK OF EFFECT
    1) NICARDIPINE
    a) A small-scale study (n=50) evaluating the use of niCARdipine for the treatment of hypertension in severe preeclampsia showed the drug to be safe and efficacious for both mother and fetus. NiCARdipine was administered for 7 days or less (n=10), 8 to 28 days (n=20), or 29 days or more (n=10). Long-term treatment appeared to be just as safe as short and medium-term protocols that were already established (Seki et al, 2002).
    2) NIMODIPINE
    a) In a study describing 10 pre-eclamptic women, nimodipine administration near term was associated with effective control of blood pressure. The patients were treated with 30 mg nimodipine orally every 4 hours from the time of admission through 24 hours following delivery. Doppler findings were consistent with vasodilatation in maternal and fetal cerebral vessels. Systolic and diastolic blood pressure were reduced significantly without apparent fetal distress. All infants were developing normally at 6 weeks of age (Belfort et al, 1994).
    E) ANIMAL STUDIES
    1) CLEVIDIPINE
    a) RATS, RABBITS: A decrease in fetal survival during organogenesis was observed in rats and rabbits at doses of 0.7 times (on a body surface area basis) the maximum recommended human dose (MRHD) in rats and 2 times the MRHD in rabbits (Prod Info Cleviprex(R) intravenous injection, 2011).
    b) RATS: Dose-related increases in maternal mortality, length of gestation, and prolonged parturition were observed during late gestation in pregnant rats at doses greater than or equal to one-sixth the maximum recommended human dose on a body surface area basis. Clevidipine crosses the placenta in rats (Prod Info Cleviprex(R) intravenous injection, 2011).
    2) ISRADIPINE
    a) RATS: Oral administration of isradipine in rats decreased maternal weight gain at the highest and maternally toxic dose of 60 mg/kg/day (150 times the maximum human therapeutic dose), but was not associated with embryotoxicity or adverse fetal or maternal outcome (Prod Info isradipine oral capsules, 2011).
    b) RABBITS: Reduction in maternal weight gain and increased fetal resorption were noted in rabbits administered isradipine 3 and 10 mg/kg/day (7.5 and 25 times, respectively, the maximum human therapeutic dose) (Prod Info isradipine oral capsules, 2011).
    3) NICARDIPINE
    a) RATS, RABBITS: Embryotoxicity was not reported in rats exposed to oral niCARdipine doses 8 times the maximum recommended human dose (MRHD) based on body surface area (mg/m(2)), However, dystocia, reduced birth weights and neonatal weight gain, and reduced neonatal survival were reported. There was evidence of embryotoxicity and maternal toxicity (maternal weight gain suppression) in rabbits exposed to oral doses 24 times the MRHD. There was also evidence of embryotoxicity when rats and rabbits were exposed to niCARdipine at 0.27 and 0.05 times the MRHD, respectively. Significant maternal mortality, but no fetal harm, was reported when rabbits were exposed to oral doses up to 16 times the MRHD (Prod Info nicardipine HCl intravenous injection solution, 2012).
    4) NISOLDIPINE
    a) RATS: Increased fetal resorptions was noted following nisoldipine administration to pregnant rats at doses of 100 mg/kg/day (approximately 5 times the maximum recommended human dose (MRHD) on a mg/m(2) basis). Decreased fetal weight was also observed at doses of 30 mg/kg/day (approximately 16 times the MRHD on a mg/m(2) basis) and 100 mg/kg/day (Prod Info SULAR(R) extended release oral tablets, 2010).
    b) RABBITS: Doses of 30 mg/kg/day (approximately 10 times the MRHD on a mg/m(2) basis) resulted in decreased fetal and placental weights when administered to pregnant rabbits (Prod Info SULAR(R) extended release oral tablets, 2010).
    3.20.4) EFFECTS DURING BREAST-FEEDING
    A) LACK OF INFORMATION
    1) GENERAL
    a) At the time of this review, no data were available to assess the potential effects of exposure to clevidipine, isradipine, or nisoldipine during lactation in humans (Prod Info Cleviprex(R) intravenous injection, 2011; Prod Info isradipine oral capsules, 2011; Prod Info SULAR(R) extended release oral tablets, 2010).
    B) BREAST MILK
    1) NICARDIPINE
    a) In a study of 11 nursing mothers who were treated with oral or IV niCARdipine 4 to 14 days postpartum (immediate-release, 40 to 80 mg/day (n=4); sustained-release, 100 to 150 mg/day (n=6); and IV 120 mg/day (n=1)), the peak milk concentration and mean milk concentration were 7.3 mcg/L (range, 1.9 to 18.8 mcg/L) and 4.4 mcg/L (range 1.3 to 13.8 mcg/L), respectively. Infants received an average of 0.073% and 0.14% of the weight-adjusted maternal oral and IV dose, respectively (Prod Info nicardipine HCl intravenous injection solution, 2012).
    b) In another study of 7 nursing mothers who were administered IV niCARdipine for an average of 1.9 days to treat preeclampsia, niCARdipine was not detectable (less than 5 mcg/L) in 82% of the 34 milk samples that were collected at unspecified times. There were detectable niCARdipine levels (range, 5.1 to 18.5 mcg/L) in 6 milk samples collected from 4 women who received 1 to 6.5 mg/hour of niCARdipine. In 1 woman who was treated with 5.5 mg/hour of niCARdipine, there was a niCARdipine concentration of 18.5 mcg/L. Less than 0.3 mcg/day (between 0.015% to 0.004% of the therapeutic dose in a 1-kg infant) was the estimated maximum niCARdipine dose in a nursing infant (Prod Info nicardipine HCl intravenous injection solution, 2012).
    2) NIMODIPINE
    a) In 1 study, breast milk levels of nimodipine were negligible with a milk/serum nimodipine concentration of 0.06 to 0.15 (Carcas et al, 1996). The authors estimated that a nursing infant would receive between 0.063 to 0.705 mcg/kg/day of nimodipine. This corresponds to between 0.008% and 0.092% of the weight-adjusted dose administered to the mother. In another study, breast milk levels paralleled serum levels, in a ratio of approximately 1:3 (Tonks, 1995).
    C) ANIMAL STUDIES
    1) CLEVIDIPINE
    a) RATS: A dose-related increase in maternal mortality, length of gestation, and prolonged parturition was observed during lactation in pregnant rats at doses greater than or equal to 1-sixth the maximum recommended human dose on a body surface area basis (Prod Info Cleviprex(R) intravenous injection, 2011).
    2) NIMODIPINE
    a) RATS: Nimodipine and/or its metabolites are excreted in the milk of lactating rats at concentrations much higher than in maternal plasma (Prod Info NYMALIZE(TM) oral solution, 2013; Prod Info nimodipine oral capsules, 2012).
    3.20.5) FERTILITY
    A) ANIMAL STUDIES
    1) CLEVIDIPINE
    a) RATS: No adverse effects on male or female fertility were observed in rats receiving up to 55 mg/kg/day (approximately equivalent to 504 mg/day maximum recommended human dose on a body surface area basis) (Prod Info Cleviprex(R) intravenous injection, 2011).
    2) ISRADIPINE
    a) RATS: No evidence of fertility impairment in male and female rats receiving doses up to 60 mg/kg/day (Prod Info isradipine oral capsules, 2011).
    3) NICARDIPINE
    a) RATS: There was no impairment of fertility in male or female rats exposed to oral niCARdipine at doses up to 8 times the maximum recommended human dose (Prod Info nicardipine HCl intravenous injection solution, 2012).
    4) NIMODIPINE
    a) RATS: There was no impairment of fertility in male or female Wistar rats exposed to oral nimodipine at doses up to 30 mg/kg/day (about 4 times the recommended clinical dose in humans) when administered daily for more than 10 weeks in males and 3 weeks in females prior to mating and continued until day 7 of pregnancy (Prod Info nimodipine oral capsules, 2012).
    5) NISOLDIPINE
    a) RATS: No evidence of fertility impairment in male and female rats receiving doses up to 30 mg/kg/day (approximately 5 times the recommended human dose on a mg/m(2) basis) (Prod Info SULAR(R) extended release oral tablets, 2010).

Carcinogenicity

    3.21.3) HUMAN STUDIES
    A) LACK OF EFFECT
    1) Several case-control analyses have shown that the use of calcium antagonists is NOT associated with a statistically significant increased risk of developing cancer as compared with non-users of calcium antagonists (Jick et al, 1997; Braun et al, 1998) Rosenberg et al, 1998; (Kanamasa et al, 1999; Cohen et al, 2000; Sorensen et al, 2000).

Monitoring Parameters Levels

    4.1.1) SUMMARY
    A) Monitor vital signs frequently. Institute continuous cardiac monitoring and obtain serial ECGs.
    B) In patients with significant hypotension or bradycardia, monitor serum electrolytes, renal function, arterial or venous blood gas, and urine output.
    C) Obtain digoxin level in patients who also have access to digoxin.
    D) Monitor cardiac enzymes in patients with chest pain.
    E) Serum drug levels are not readily available and thus not immediately helpful.
    4.1.2) SERUM/BLOOD
    A) BLOOD/SERUM CHEMISTRY
    1) Blood levels of calcium antagonists are not readily available. Blood levels generally do not predict toxicity or direct management, but may be of forensic or pharmacokinetic interest.
    2) Monitor serum electrolytes and renal function.
    3) Monitor serum glucose. Serum glucose concentration correlates directly with the severity of calcium channel blocker intoxication; patients who develop hyperglycemia are likely to develop more severe cardiovascular manifestations of toxicity (Levine et al, 2007).
    B) ACID/BASE
    1) Monitor respiratory function with arterial blood gases.
    4.1.4) OTHER
    A) OTHER
    1) MONITORING
    a) Monitor cardiovascular status to include blood pressure, ECG, and urinary output.
    b) Monitor respiratory function with blood gasses, pH, rate; pulmonary edema may occur.
    c) Monitor mental status; CNS depression due to direct drug effects, cardiovascular disruptions, and coingestion of CNS depressants is common. Seizures are rare but may occur.
    d) It has been suggested that continuous SvO2 monitoring using a fiber optic pulmonary artery catheter may be useful to monitor tissue oxygenation in cases of refractory hypotension secondary to calcium antagonist poisoning (Kamijo et al, 2006).

Methods

    A) GC/MS
    1) NICARDIPINE: Gas chromatography-mass spectrometry (GC/MS) was used for the post-mortem detection of niCARdipine in an 89-year-old man who died following inadvertent administration of 40 mL niCARdipine hydrochloride (containing 60 mg of niCARdipine) instead of 40 mL of 50% glucose. GC/MS detected 4.97 mcg/ml of niCARdipine and 5 mcg/mL of M-5 (niCARdipine's metabolite) from the patient's whole blood sample (Ikegaya et al, 2002).
    B) HPLC
    1) NISOLDIPINE: High performance liquid chromatography (HPLC) was used to measure the nisoldipine serum concentration in a 72-year-old woman who developed severe shock, hypothermia, and renal dysfunction following a suspected overdose ingestion of nisoldipine. HPLC determined that the nisoldipine serum concentration was 1544 mcg/L, obtained from a serum sample taken at admission; however the exact time of ingestion was unknown (Louagie et al, 1998).

Life Support

    A) Support respiratory and cardiovascular function.

Patient Disposition

    6.3.1) DISPOSITION/ORAL EXPOSURE
    6.3.1.1) ADMISSION CRITERIA/ORAL
    A) Patients who show the following signs of toxicity should be admitted to an intensive care setting (Pearigen & Benowitz, 1991):
    1) CARDIOVASCULAR: Hypotension or bradycardia (or tachycardia with nifedipine); heart block; A-V dissociation; asystole; congestive heart failure
    2) RESPIRATORY: Pulmonary edema
    3) GASTROINTESTINAL: Nausea or vomiting
    4) NEUROLOGICAL: Seizures; altered mental status
    6.3.1.2) HOME CRITERIA/ORAL
    A) Healthy, asymptomatic patients with the following inadvertent single substance ingestions may be monitored at home (IR = immediate release, SR = sustained release) (Olson et al, 2005):
    1) BEPRIDIL: ADULT: 300 mg or less; CHILD: no safe dose
    2) ISRADIPINE: ADULT: 20 mg or less; CHILD: 0.1 mg/kg or less
    3) NICARDIPINE: ADULT: 40 mg or less IR or chewed SR, or 60 mg or less SR; CHILD: less than 1.25 mg/kg
    4) NIMODIPINE: ADULT: 60 mg or less; CHILD: no safe dose
    5) NISOLDIPINE: ADULT: 30 mg or less; CHILD: no safe dose
    B) A retrospective study was conducted of 161 patients who inadvertently ingested double their usual dose of calcium channel blocker or more. Of the 104 patients who ingested double their usual dose, 9 (9%) developed cardiovascular signs or symptoms. Four of these symptomatic patients had ingested less than or equal to the maximum single dose for the drug, and another four had ingested a dose in between the maximum single dose and the maximum daily dose. Of the 57 patients who had ingested more than double their usual daily dose, 8 (14%) developed cardiovascular signs or symptoms. All of these patients had ingested an amount equal to or greater than the maximal daily dose. The toxicity of calcium antagonists following a therapeutic overdose can be highly variable; this could be due to the broad range of therapeutic doses and the pre-existing conditions in these patients. Because of this variability, home management may be difficult; poison centers should be conservative in their evaluation of these cases (Cantrell et al, 2005).
    C) For the specific home criteria regarding AMLODIPINE, DILTIAZEM, NIFEDIPINE, and VERAPAMIL, refer to the individual document.
    6.3.1.3) CONSULT CRITERIA/ORAL
    A) Consult a poison center or medical toxicologist in cases of severe poisonings or in cases where there is a history of a large exposure.
    6.3.1.5) OBSERVATION CRITERIA/ORAL
    A) Symptomatic patients, those with underlying cardiovascular disease, those taking beta blockers or another cardiodepressant drug, and those with deliberate ingestions should be referred to a health care facility for treatment, evaluation and monitoring. Patients who have not developed signs or symptoms more than 6 hours after ingestion of an immediate-release product, 18 hours after ingestion of a sustained or extended-release product are unlikely to develop toxicity.
    B) Patients with the following inadvertent single substance ingestions should be referred to a healthcare facility (IR = immediate release, SR = sustained release) (Olson et al, 2005):
    1) BEPRIDIL: ADULT: greater than 300 mg; CHILD: any amount
    2) ISRADIPINE: ADULT: greater than 20 mg; CHILD: greater than 0.1 mg/kg
    3) NICARDIPINE: ADULT: greater than 40 mg IR or chewed SR, or greater than 60 mg SR; CHILD: 1.25 mg/kg or more
    4) NIMODIPINE: ADULT: greater than 60 mg; CHILD: any amount
    5) NISOLDIPINE: ADULT: greater than 30 mg; CHILD: any amount
    C) Patients with a history of overdose with sustained release preparations should be observed and monitored in an intensive care setting up to 24 hours (Spiller et al, 1991; Clark & Hanna, 1993).
    D) One study determined that 81% of the poison centers surveyed (n=33) recommended prolonged observation (greater than 6 hours) in pediatric cases of single-tablet ingestions of sustained-release calcium antagonists (Brayer & Wax, 1998).
    E) CHILDREN LESS THAN 6 YEARS OF AGE: Based on 5 retrospective case reviews involving calcium channel blocker ingestions in children less than 6-years-old, patients should be observed in-hospital for 6 hours following ingestion of regular-release medications and for 12 to 24 hours following ingestion of sustained-release formulations (Ranniger & Roche, 2007).
    F) For the specific observation criteria regarding AMLODIPINE, DILTIAZEM, NIFEDIPINE, and VERAPAMIL, refer to the individual document.

Monitoring

    A) Monitor vital signs frequently. Institute continuous cardiac monitoring and obtain serial ECGs.
    B) In patients with significant hypotension or bradycardia, monitor serum electrolytes, renal function, arterial or venous blood gas, and urine output.
    C) Obtain digoxin level in patients who also have access to digoxin.
    D) Monitor cardiac enzymes in patients with chest pain.
    E) Serum drug levels are not readily available and thus not immediately helpful.

Oral Exposure

    6.5.1) PREVENTION OF ABSORPTION/PREHOSPITAL
    A) Prehospital gastrointestinal decontamination is not recommended because of the potential for abrupt deterioration.
    6.5.2) PREVENTION OF ABSORPTION
    A) SUMMARY
    1) Because a calcium channel blocker overdose can be life-threatening, all significant ingestions should receive activated charcoal. Patients with altered mental status should be intubated prior to administration. Gastric lavage should be considered in patients with recent large ingestions if the airway is protected. Late gastric lavage may be effective following sustained-release products. Whole bowel irrigation should be considered early for patients who can protect their airway or who are intubated who have ingested sustained-release formulations; it can limit absorption from possible concretions. Whole bowel irrigation should NOT be performed in patients who are hemodynamically unstable.
    2) If continued absorption is suspected in a symptomatic patient after these procedures, consider abdominal x-ray (if brand is radiopaque), ultrasound, or gastroscopy.
    B) ACTIVATED CHARCOAL
    1) Single doses of activated charcoal are recommended. While not proven effective, administration of a second dose should be considered especially in the setting of sustained-release dosage form ingestion or coingestion of drugs or chemicals which could delay gastric emptying or slow intestinal motility (Krenzelok, 1991; Pearigen & Benowitz, 1991).
    2) Multiple doses, beginning 7 hours post ingestion, did not significantly enhance elimination in a 38-year-old woman who had ingested 900 mg of diltiazem (Roberts et al, 1991).
    3) In a volunteer study, administration of activated charcoal 2 or 4 hours after ingestion of slow-release verapamil decreased absorption by 35% and 32% respectively (Laine et al, 1997a). Delayed administration of activated charcoal was not effective in reducing absorption of a conventional formulation of verapamil (Laine et al, 1997).
    4) In a randomized cross-over study, administration of activated charcoal 30 minutes after ingestion 80 mg verapamil reduced the peak plasma concentration of verapamil by 16% (Lapatto-Reiniluoto et al, 2000).
    5) CHARCOAL ADMINISTRATION
    a) Consider administration of activated charcoal after a potentially toxic ingestion (Chyka et al, 2005). Administer charcoal as an aqueous slurry; most effective when administered within one hour of ingestion.
    6) CHARCOAL DOSE
    a) Use a minimum of 240 milliliters of water per 30 grams charcoal (FDA, 1985). Optimum dose not established; usual dose is 25 to 100 grams in adults and adolescents; 25 to 50 grams in children aged 1 to 12 years (or 0.5 to 1 gram/kilogram body weight) ; and 0.5 to 1 gram/kilogram in infants up to 1 year old (Chyka et al, 2005).
    1) Routine use of a cathartic with activated charcoal is NOT recommended as there is no evidence that cathartics reduce drug absorption and cathartics are known to cause adverse effects such as nausea, vomiting, abdominal cramps, electrolyte imbalances and occasionally hypotension (None Listed, 2004).
    b) ADVERSE EFFECTS/CONTRAINDICATIONS
    1) Complications: emesis, aspiration (Chyka et al, 2005). Aspiration may be complicated by acute respiratory failure, ARDS, bronchiolitis obliterans or chronic lung disease (Golej et al, 2001; Graff et al, 2002; Pollack et al, 1981; Harris & Filandrinos, 1993; Elliot et al, 1989; Rau et al, 1988; Golej et al, 2001; Graff et al, 2002). Refer to the ACTIVATED CHARCOAL/TREATMENT management for further information.
    2) Contraindications: unprotected airway (increases risk/severity of aspiration) , nonfunctioning gastrointestinal tract, uncontrolled vomiting, and ingestion of most hydrocarbons (Chyka et al, 2005).
    C) GASTRIC LAVAGE
    1) Gastric lavage, followed by administration of activated charcoal, is recommended in patients with significant ingestions. A large bore orogastric tube is preferred due to the large size of some sustained-release dosage forms.
    2) Late gastric lavage may be effective following sustained-release products.
    3) In a volunteer study, gastric lavage administered 30 minutes after ingestion of 80 mg verapamil had no significant effect on reduction of peak plasma concentration of verapamil (Lapatto-Reiniluoto et al, 2000).
    4) INDICATIONS: Consider gastric lavage with a large-bore orogastric tube (ADULT: 36 to 40 French or 30 English gauge tube {external diameter 12 to 13.3 mm}; CHILD: 24 to 28 French {diameter 7.8 to 9.3 mm}) after a potentially life threatening ingestion if it can be performed soon after ingestion (generally within 60 minutes).
    a) Consider lavage more than 60 minutes after ingestion of sustained-release formulations and substances known to form bezoars or concretions.
    5) PRECAUTIONS:
    a) SEIZURE CONTROL: Is mandatory prior to gastric lavage.
    b) AIRWAY PROTECTION: Place patients in the head down left lateral decubitus position, with suction available. Patients with depressed mental status should be intubated with a cuffed endotracheal tube prior to lavage.
    6) LAVAGE FLUID:
    a) Use small aliquots of liquid. Lavage with 200 to 300 milliliters warm tap water (preferably 38 degrees Celsius) or saline per wash (in older children or adults) and 10 milliliters/kilogram body weight of normal saline in young children(Vale et al, 2004) and repeat until lavage return is clear.
    b) The volume of lavage return should approximate amount of fluid given to avoid fluid-electrolyte imbalance.
    c) CAUTION: Water should be avoided in young children because of the risk of electrolyte imbalance and water intoxication. Warm fluids avoid the risk of hypothermia in very young children and the elderly.
    7) COMPLICATIONS:
    a) Complications of gastric lavage have included: aspiration pneumonia, hypoxia, hypercapnia, mechanical injury to the throat, esophagus, or stomach, fluid and electrolyte imbalance (Vale, 1997). Combative patients may be at greater risk for complications (Caravati et al, 2001).
    b) Gastric lavage can cause significant morbidity; it should NOT be performed routinely in all poisoned patients (Vale, 1997).
    8) CONTRAINDICATIONS:
    a) Loss of airway protective reflexes or decreased level of consciousness if patient is not intubated, following ingestion of corrosive substances, hydrocarbons (high aspiration potential), patients at risk of hemorrhage or gastrointestinal perforation, or trivial or non-toxic ingestion.
    D) WHOLE BOWEL IRRIGATION
    1) Consider whole bowel irrigation following activated charcoal to limit absorption from possible concretions and sustained release products. It should only be performed in patients who can protect their airway or who are intubated. Whole bowel irrigation should not be performed in patients that are hemodynamically unstable. Sustained-release formulations have produced concretions composed of alginate hydrocolloid matrix despite initial lavage (Rankin & Edwards, 1990; Sporer & Manning, 1993; Buckley et al, 1993; Hendren et al, 1989).
    2) Repeat charcoal following whole bowel irrigation since the PEG/electrolyte solution may desorb drug from charcoal. If continued absorption is suspected in a symptomatic patient after these procedures, consider abdominal x-ray (if brand is radiopaque), ultrasound, or gastroscopy.
    3) CASE REPORT: One study reported the occurrence of delayed toxicity (up to 72 hours) from an overdose ingestion of sustained-release nifedipine despite treatment with whole bowel irrigation (Lawless et al, 1997).
    a) WHOLE BOWEL IRRIGATION/INDICATIONS: Whole bowel irrigation with a polyethylene glycol balanced electrolyte solution appears to be a safe means of gastrointestinal decontamination. It is particularly useful when sustained release or enteric coated formulations, substances not adsorbed by activated charcoal, or substances known to form concretions or bezoars are involved in the overdose.
    1) Volunteer studies have shown significant decreases in the bioavailability of ingested drugs after whole bowel irrigation (Tenenbein et al, 1987; Kirshenbaum et al, 1989; Smith et al, 1991). There are no controlled clinical trials evaluating the efficacy of whole bowel irrigation in overdose.
    b) CONTRAINDICATIONS: This procedure should not be used in patients who are currently or are at risk for rapidly becoming obtunded, comatose, or seizing until the airway is secured by endotracheal intubation. Whole bowel irrigation should not be used in patients with bowel obstruction, bowel perforation, megacolon, ileus, uncontrolled vomiting, significant gastrointestinal bleeding, hemodynamic instability or inability to protect the airway (Tenenbein et al, 1987).
    c) ADMINISTRATION: Polyethylene glycol balanced electrolyte solution (e.g. Colyte(R), Golytely(R)) is taken orally or by nasogastric tube. The patient should be seated and/or the head of the bed elevated to at least a 45 degree angle (Tenenbein et al, 1987). Optimum dose not established. ADULT: 2 liters initially followed by 1.5 to 2 liters per hour. CHILDREN 6 to 12 years: 1000 milliliters/hour. CHILDREN 9 months to 6 years: 500 milliliters/hour. Continue until rectal effluent is clear and there is no radiographic evidence of toxin in the gastrointestinal tract.
    d) ADVERSE EFFECTS: Include nausea, vomiting, abdominal cramping, and bloating. Fluid and electrolyte status should be monitored, although severe fluid and electrolyte abnormalities have not been reported, minor electrolyte abnormalities may develop. Prolonged periods of irrigation may produce a mild metabolic acidosis. Patients with compromised airway protection are at risk for aspiration.
    5) COMPLICATIONS: Complications were reported in two patients who were given whole bowel irrigation (WBI), and subsequently became hemodynamically unstable, after overdose ingestions of sustained-release formulations (Cumpston et al, 2010).
    a) The first patient, a 58-year-old man, received activated charcoal and whole bowel irrigation, at 2 L/hour, approximately 3 hours after reportedly ingesting 7.2 g of extended release diltiazem. Prior to WBI administration, the patient was hemodynamically stable. One hour later (4 hours post-ingestion), the patient developed bradycardia (50 bpm) and hypotension (70/40 mmHg), treated with atropine and IV fluids. After 7 hours of WBI (10 hours post-ingestion), the patient developed asystole, resolved with external pacing, and abdominal distension, resulting in cessation of WBI therapy. Despite aggressive supportive therapy, including vasopressors, calcium, and insulin, he continued to deteriorate and subsequently died. Autopsy was consistent with general hypoperfusion of the bowel without clear infarction.
    b) The second patient, a 40-year-old man, received activated charcoal and WBI after ingesting 90 tablets of verapamil 240 mg sustained release (21.6 g). Prior to WBI, the patient was tachycardic (111 bpm), but otherwise asymptomatic. The patient began vomiting 3.5 hours post-ingestion, and developed hypotension (80/43 mmHg) and a decrease in heart rate (73 bpm) at 4 hours post-ingestion. WBI was eventually stopped at 6 hours post-ingestion, due to persistent vomiting and hypotension and the development of pulmonary edema. The patient developed aspiration pneumonia believed to be due to aspiration of activated charcoal, polyethylene glycol electrolyte lavage solution, and gastric secretions. With supportive treatment of his pneumonia and severe hypotension, the patient gradually recovered and was discharged.
    E) ENDOSCOPY
    1) Following overdose of sustained release verapamil, concretions have occasionally been reported within the stomach or intestines. They are often not visible on plain x-ray films of the abdomen; gastrointestinal emptying methods to remove the concretions have not been proven. Endoscopy may be necessary following severe toxicity or prolonged symptoms (Prod Info CALAN(R) SR oral sustained release caplets, 2013).
    2) Tablet concretions ranging in size from golf to tennis ball size developing from sustained-release dosage forms have been found at autopsy. Gastroscopy and endoscopic removal may be required for confirmation if suspected since these masses have not been apparent on abdominal films (Sporer & Manning, 1993).
    3) As with other calcium channel blockers (eg, nifedipine), the formation of bezoars appear to be associated with acute overdose ingestion of extended-release tablets in a small number of cases. Treatment of the bezoar may be dependent on several factors, including the location, composition, and size of the bezoar, and patient symptoms. In the setting of acute overdose, endoscopic removal may be necessary. For symptomatic bezoars, surgery may be necessary if the bezoar cannot be removed endoscopically (Wells et al, 2006; Niezabitowski et al, 2000; Taylor et al, 1998).
    6.5.3) TREATMENT
    A) SUPPORT
    1) MANAGEMENT OF MILD TO MODERATE TOXICITY
    a) Patients who have asymptomatic bradycardia can be admitted and observed with telemetry. Obtain peripheral intravenous access and monitor ECG. Mild hypotension may only require treatment with intravenous fluid administration.
    2) MANAGEMENT OF SEVERE TOXICITY
    a) Patients with bradycardia and hypotension require standard ACLS treatment. Place a central line and consider placement of an arterial line. Standard first line treatment includes atropine for bradycardia although in a serious poisoning it is rarely effective. High dose insulin and dextrose have been effective in animal studies and multiple case reports in patients with hypotension refractory to other modalities, and should be considered early in patients with significant hypotension. Use intravenous calcium in severe poisonings although in these cases, beneficial effects of calcium infusion (calcium chloride is preferred) may be very minimal or short-lived. Repeat bolus doses or a continuous intravenous infusion are often needed. Standard vasopressors should be administered to maintain blood pressure. Lipid emulsion has been successful in animal studies and several case reports of patients with hypotension refractory to other therapies. Intravenous glucagon has been used with variable success. In a patient whose hemodynamic status continues to be refractory despite the treatment described above, extracorporeal membrane oxygenation or cardiopulmonary bypass should be considered. Treat seizures with IV benzodiazepines; barbiturates or propofol may be needed if seizures persist or recur.
    B) MONITORING OF PATIENT
    1) Serum concentrations are not readily available and not helpful to guide therapy.
    2) Monitor vital signs frequently.
    3) Institute continuous cardiac monitoring and obtain serial ECGs.
    4) Monitor serum electrolytes, blood glucose, and renal function. In patients with significant hypotension or bradycardia, monitor arterial or venous blood gas, and urine output.
    5) Obtain digoxin concentration in patients who also have access to digoxin.
    6) Monitor cardiac enzymes in patients with chest pain.
    7) It has been suggested that continuous SvO2 monitoring using a fiber optic pulmonary artery catheter may be useful to monitor tissue oxygenation in cases of refractory hypotension secondary to calcium antagonist poisoning (Kamijo et al, 2006).
    C) HYPOTENSIVE EPISODE
    1) FLUIDS
    a) INDICATION: Loss of systemic vascular resistance requires an increase in circulating volume. Complete response to fluids alone should not be expected, but volume replacement is a necessary component. Administer IV 0.9% NaCl at 10 to 20 mL/kg and place the patient in supine position. Consider central venous pressure monitoring to guide further fluid therapy.
    b) CAUTIONS: Monitor for signs of pulmonary edema (Pearigen & Benowitz, 1991).
    2) CALCIUM
    a) INDICATIONS: Calcium is used to reverse hypotension and improve cardiac conduction defects. Calcium administration has been most effective in overcoming mild toxicity from small overdoses or therapeutic use and is less useful in massive overdose cases since calcium channel blockade is non-competitive (DeRoos, 2011; Pearigen & Benowitz, 1991; Krenzelok, 1991; Clark & Hanna, 1993), but was successful in 11 of 30 cases in one series (Hofer et al, 1993).
    b) DRUG OF CHOICE: In some studies, calcium chloride is thought to produce more predictable increases in extracellular ionized calcium and a greater positive inotropic response (White et al, 1976; Haynes et al, 1985); however, other sources have found no differences in efficacy of calcium chloride and calcium gluconate. Calcium chloride provides 3 times more elemental calcium (13.4 mEq) than calcium gluconate (4.3 mEq) in the commercially available 1 gram ampules (DeRoos, 2011).
    c) ADULT DOSE: Optimal dosing is not established; begin with an initial IV infusion of about 13 to 25 mEq of calcium (10 to 20 mL of 10% calcium chloride or 30 to 60 mL of 10% calcium gluconate) followed by either repeat boluses every 15 to 20 minutes up to 3 to 4 doses or a continuous infusion of 0.5 mEq/kg/hr of calcium (0.2 to 0.4 mL/kg/hr of 10% calcium chloride or 0.6 to 1.2 mL of 10% calcium gluconate) (DeRoos, 2011). Some authors advocate administering 1 gram of calcium salts every 2 to 3 minutes until conduction block is reversed or clinical evidence of hypercalcemia develops (Howarth et al, 1994; Buckley et al, 1994). Calcium dosing should be titrated to hemodynamic response rather than serum calcium concentration alone; central venous or pulmonary artery catheters may be useful to guide therapy. Monitor ECG and ionized calcium concentration.
    1) NIMODIPINE/CASE REPORT: Hypotension, hypoxemia, and anuria were reported in an 81-year-old woman following a suspected overdose ingestion of nimodipine. Following IV administration of 20 mL calcium gluconate 10%, the patient's blood pressure normalized immediately, with improvement in her oxygenation and resolution of her anuria within 20 minutes (Gerloni & Copetti, 2004).
    2) CASE REPORT: A 25-year-old woman intentionally ingested 3.6 grams of sustained-release nifedipine and, 3 hours later, presented to the emergency department comatose with agonal respirations, no carotid pulse, and no recordable blood pressure. Following administration of 1 gram calcium chloride given intravenous push, the patient began to spontaneously breathe with the development of a carotid pulse and a BP of 82/40 mmHg. The patient's blood pressure continued to increase following further intravenous administration of calcium chloride over the next hour (Haddad, 1996).
    3) CASE REPORT/ HIGH DOSE ADMINISTRATION: A 40-year-old woman, with chronic renal insufficiency, developed hypotension and bradycardia after ingesting 30 mg of amlodipine. The patient's blood pressure and heart rate normalized following high-dose intravenous administration of calcium chloride, consisting of 4 grams given in the emergency department followed by 1.5 grams every 20 minutes (for a total of 13 grams given over 2 hours) (Hung & Olson, 2007).
    4) CASE SERIES: In one series, doses varied from 4.5 mEq to 95.2 mEq, with no evidence of a dose-response relation (Ramoska et al, 1993).
    5) ANIMAL DATA: A laboratory investigation using a canine model showed that administration of calcium chloride with digoxin, following verapamil- induced hypotension, resulted in a 15% to 21% increase in systolic blood pressure as compared with administration of calcium chloride alone, and did not result in ventricular dysrhythmias (Bania et al, 2000).
    d) HYPERCALCEMIA: Significant hypercalcemia may be necessary before severely intoxicated patients respond to aggressive calcium therapy, but the optimal calcium regimen has not been established. In patients who have received large doses of calcium for severe calcium channel blocker overdose (attaining serum calcium concentrations up to twice the upper limits of normal), hypercalcemia generally resolves within 48 hours without clinically apparent adverse effects (clinical or ECG) from hypercalcemia (Howarth et al, 1994; Buckley et al, 1994).
    1) CASE REPORT: Hypercalcemia with concentrations as high as 19.2 mg/dL (9.9 mg/dL upper limit of normal) secondary to treatment with calcium salts have been reported during aggressive therapy (Buckley et al, 1993) without adverse effects secondary to hypercalcemia. Such aggressive treatment with calcium salts may be necessary to reverse conduction defects (Howarth et al, 1994).
    2) CASE REPORT: Hypercalcemia (16.3 mg/dL) occurred following aggressive calcium chloride treatment in a 45-year-old woman following an overdose ingestion of sustained-release diltiazem. There were no ECG manifestations due to the hypercalcemia and the patient recovered uneventfully over the next 4 days (Hantsch et al, 1997).
    3) CASE REPORT: Hypercalcemia (peak calcium concentration 32.3 mg/dL) was reported in a 61-year-old woman who received a continuous calcium chloride infusion, initiated at 4 g/hour and decreased to 2 g/hour, following inadvertent ingestion of a second dose (360 mg) of her sustained release verapamil. At the same time, the patient, who had no previous history of renal impairment, developed a decrease in urine output that progressed to complete anuria. Analysis of the urine sediment showed coarse granular casts, indicating acute tubular necrosis. After initiating continuous hemodialysis, the patient's calcium level decreased to 8.3 mg/dL, however her condition deteriorated with persistent anuria and multi-organ failure resulting in her death 17 days post-admission. An autopsy revealed acute tubular necrosis, liver necrosis, multiple infarctions of the spleen, and dermal lesions on the trunk and extremities, secondary to calciphylaxis (Sim & Stevenson, 2008).
    e) PRECAUTIONS: Hypotension generally does not respond as well as conduction disturbances to calcium. If a patient does not respond after a doubling of the ionized calcium concentration, further calcium treatment may not be beneficial. If the patient has also ingested digoxin, avoid administering calcium until after digoxin-specific Fab is administered to prevent worsening digoxin toxicity (DeRoos, 2011).
    f) Calcium therapy should be tailored to the individual patient. In one case report, a woman with a massive overdose of sustained-release nifedipine and severe hypotension was successfully treated with continuous IV calcium gluconate infusion, using an individually tailored protocol for dose titration of IV calcium infusion. In this protocol, the optimal calcium infusion rate was determined by achieving the lowest possible serum calcium concentration associated with a maximal contractility (ie, cardiac power index [CPI] = CI x MAP x 0.0022) rather than maintaining a predefined serum calcium concentration or using cardiac output (CO) or stroke volume (SV), as an indicator of cardiac function (ie, cardiac work). This allowed for a lower total dose of calcium used and a lower peak serum calcium concentration (Zhou et al, 2013).
    1) CASE REPORT: A 25-year-old woman presented with hypotension 5 hours after ingesting 6 grams of sustained-release nifedipine. Laboratory results revealed a serum nifedipine concentration of 7100 mcg/L (therapeutic range, 25 to 100 mcg/L), a serum ionized calcium concentration of 0.7 mmol/L, and a serum glucose of 21.8 mmol/L. Despite supportive care, including gastric lavage, activated charcoal, IV fluid, glucagon, insulin infusion, and several doses of IV bolus injection of calcium gluconate (10 mL of 10% solution), she continued to be hypotensive. In the ICU, she was unconscious with a blood pressure of 70/30 mm Hg and a heart rate of 140 beats/min. Laboratory results revealed severe metabolic acidosis. She developed respiratory failure 6 hours later and was intubated. At this time, she received 12 grams (27 mmol) of calcium gluconate (including 9 g received within a 3-hour period) over a 24-hour period and her serum ionized calcium concentration increased to 1.15 mmol/L. She continued to have hyperlactatemia (4.8 mmol/L) and tachycardia (HR 150 beats/min) and received high doses of vasopressors at 24 hours postadmission. Because of insufficient improvements in contractility (Cardiac power index [CPI], 0.87 W/m(2)) and systemic vascular resistance, she received continuous IV infusion of calcium gluconate at a rate of 1 g/hr, with the dose titrated to optimize the CPI. About 4 hours later, the vasopressor doses were gradually decreased. Overall, calcium therapy increased stroke volume (SV) by 145%, CPI by 199%, and mean arterial pressure (MAP) by 50%, and decreased heart rate by 18%. Serum ionized calcium concentration increased from 0.7 mmol/L to 1.58 mmol/L. Her serum nifedipine concentration decreased to 2900 mcg/L at 24 hours postingestion and to 1100 mcg/L at 42 hours postingestion. About 52 hours postadmission, IV calcium gluconate was stopped and at 96 hours postadmission, she recovered completely. She was discharged after 17 days of hospitalization (Zhou et al, 2013).
    g) CASE REPORT: A 37-year-old woman presented with hypotension (BP 80/45 mm Hg) and sinus tachycardia (115 beats/min) after ingesting 210 nifedipine 20 mg tablets (total dose: 4200 mg). Despite supportive therapy, including 2 doses of IV bolus injection of calcium gluconate (10 mL of 10% solution), her condition did not improve and she developed shortness of breath and circulatory shock (BP, 70/30 mm Hg). A chest radiograph revealed acute pulmonary edema. Serum glucose and ionized calcium concentration were 7.8 mmol/L and 1.14 mmol/L (normal range, 1.12 to 1.23 mmol/L), respectively. An echocardiography revealed normal left ventricular systolic function with ejection fraction of 62%. Despite treatment with multiple doses of calcium gluconate injections (10 mL of 10% solution; 0.3 mg/hr was the average dose within 24 hours; serum ionized calcium concentration was 1.27 mmol/L), her condition did not improve. At this time, she received continuous IV infusion of calcium chloride (rate: 20 mcg/min) started 25 hours after overdose, and titrated to maintain the serum ionized calcium concentration around 2 mmol/L. Her blood pressure improved soon after the calcium chloride solution was administered. Her pulmonary edema also improved following continuous calcium chloride infusion and diuretic therapy. Approximately 60 hours after presentation, her calcium chloride infusion was discontinued. Prior to discharge, debridement and skin grafting were required because of calcium chloride extravasation and skin necrosis over the peripheral drip site (Lam et al, 2001).
    h) CASE REPORT: A 25-year-old woman intentionally ingested 3.6 grams of sustained-release nifedipine and, 3 hours later, presented to the emergency department comatose with agonal respirations, no carotid pulse, and no recordable blood pressure. Following administration of 1 g calcium chloride given intravenous push, the patient began to spontaneously breathe with the development of a carotid pulse and a BP of 82/40 mmHg. The patient's blood pressure continued to increase following further intravenous administration of calcium chloride over the next hour (Haddad, 1996).
    i) CASE REPORT: Hypercalcemia (16.3 mg/dL) occurred following aggressive calcium chloride treatment in a 45-year-old woman following an overdose ingestion of sustained-release diltiazem. There were no ECG manifestations due to the hypercalcemia and the patient recovered uneventfully over the next 4 days (Hantsch et al, 1997).
    j) ADVERSE EFFECTS: Calcium chloride can cause tissue injury following extravasation; administer calcium chloride via central venous catheter. Hypercalcemia or hypophosphatemia may also occur following repeat dosing or continuous infusion; monitor serum calcium and phosphate concentrations. Nausea, vomiting, flushing, constipation, confusion, and angina have also been reported in patients receiving calcium (DeRoos, 2011).
    3) INSULIN/DEXTROSE
    a) DOSE
    1) Intravenous insulin infusion with supplemental dextrose, and potassium as needed, is recommended in patients with severe or persistent hypotension after a calcium channel blocker overdose (DeWitt & Waksman, 2004).
    2) Administer a bolus of 1 unit/kg of insulin followed by an infusion of 0.1 to 1 units/kg/hr, titrated to a systolic blood pressure of greater than 90 to 100 mmHg (bradycardia may or may not respond). Reassess every 30 minutes to determine the need for higher rates of insulin infusion (Lheureux et al, 2006). In some refractory cases, more aggressive high-dose insulin protocols have been suggested, starting with a 1 unit/kg insulin bolus, followed by a 1 unit/kg/hour continuous infusion. If there is no clinical improvement in the patient, the infusion rate may be increased by 2 units/kg/hour every 10 minutes, up to a maximum of 10 units/kg/hour (Engebretsen et al, 2011).
    3) Administer dextrose bolus to patients with an initial blood glucose of less than 250 mg/dL (adults 25 to 50 mL dextrose 50%, children 0.25 g/kg dextrose 25%). Begin a dextrose infusion of 0.5 g/kg/hr in all patients. Monitor blood glucose every 15 to 30 minutes until consistently 100 to 200 mg/dL for 4 hours, then monitor every hour. Titrate dextrose infusion to maintain blood glucose in the range of 100 to 200 mg/dL. As the patient improves, insulin resistance abates and dextrose requirements will increase. Supplemental dextrose will be needed for at least several hours after the insulin infusion is discontinued.
    4) Administer supplemental potassium initially if patient is hypokalemic (serum potassium less than 2.5 mEq/L). Monitor serum potassium every 4 hours and supplement as needed to maintain potassium of 2.5 to 2.8 mEq/L.
    5) Monitor blood pressure, pulse, ECG, mental status, serum glucose and potassium, urine output, and if possible cardiac function by way of echocardiogram/ultrasound, right heart catheter.
    b) CASE REPORTS
    1) SUMMARY: Insulin/dextrose infusions were administered to 5 patients who experienced severe circulatory shock following intentional calcium channel blocker overdose ingestions and who were unresponsive to conventional treatment. Blood pressures normalized within hours after receiving the infusions and all 5 patients recovered without sequelae. In 3 of the 4 patients, insulin and dextrose were administered as bolus doses, 10 units and 25 grams, respectively, with the subsequent administration of insulin infusion, the dose ranging from 0.1 units/kg/hr to 1.0 units/kg/hr, and dextrose (50% w/v) infusion, the dose ranging from 5 grams/hour to 15 grams/hr, via a central venous catheter. Each patient also received other supportive measures and inotropic agents (Yuan et al, 1999).
    2) Insulin infusions, with or without dextrose given concurrently, were administered to several hemodynamically unstable patients following calcium antagonist intoxication who were refractory to conventional therapy. Blood pressures in all patients normalized within hours after receiving the insulin (Agarwal et al, 2012; Hadjipavlou et al, 2011; Azendour et al, 2010; Verbrugge & vanWezel, 2007; Greene et al, 2007; Boyer et al, 2002).
    3) One study reviewed 13 case reports where high-dose insulin (10 to 20 units in 6 patients; 1 patient received 1000 units inadvertently; infusion rates 0.1 to 1 unit/kg/hr; duration either a single bolus dose or infusions for 5 to 96 hours) with supplemental dextrose and potassium (HDIDK) were used to treat calcium channel blockers overdose. These patients failed to respond clinically to other therapies. Twelve patients survived. HDIDK therapy was beneficial in seriously intoxicated patients with CCB-induced hypotension, hyperglycemia, and metabolic acidosis. However, this therapy did not consistently reverse bradycardia, heart block and intraventricular conduction delay (Shepherd & Klein-Schwartz, 2005).
    4) CASE SERIES: A single center, retrospective case review was conducted of patients who received high-dose insulin (HDI) therapy following intentional overdoses (n=11) or for treatment of multifactorial shock (n=3). The patients had either taken beta-blockers as a single agent (n=9), calcium channel blockers as a single agent (n=2), or a combination of beta blockers and calcium channel blockers (n=3). The average age of patients was 54 years (range 24 to 87 years). Of the 14 patients, 11 received dual therapy with HDI and vasopressor/inotrope support, and 3 patients received HDI only. The mean maximum HDI infusion dose was 6 units/kg/hour (range 0.5 to 10 units/kg/hour) with a mean duration of 22 hours (range 1 to 75 hours), and the vasopressor/inotropes administered included norepinephrine (n=9; mean maximum dose (MMD) 0.32 mcg/kg/min), dopamine (n=6; MMD 15 mcg/kg/min), phenylephrine (n=2; MMD 2.3 mcg/kg/min), vasopressin (n=5; MMD 0.04 unit/min), epinephrine (n=2; MMD 0.2 mcg/kg/min), and methylene blue (n=1; MMD 0.75 mg/kg/hour). Adverse effects of therapy included hypoglycemia (n=7), hypokalemia (n=5), and pulmonary edema (n=3). Of the 14 patients, 11 patients survived and the 3 deaths were from elderly patients who had developed multifactorial shock after receiving beta blockers during hospitalization (Robinson et al, 2015).
    5) TISSUE PERFUSION MONITORING: A 51-year-old man presented to the emergency department after ingesting 40 25-mg metoprolol tablets and an unknown amount of 5 mg amlodipine tablets. The patient was obtunded with vital signs indicating hypotension (systolic blood pressure in the 50s) and bradycardia (heart rate in the 20s). The patient was intubated and treatment included IV epinephrine and calcium gluconate boluses, and high dose insulin started at 1 unit/kg/hour. Repeat vital sign measurements demonstrated a blood pressure of 79/49 and a heart rate of 39. Tissue perfusion monitoring was started with an initial reading of 69% (normal 75% to 85%). An epinephrine infusion was initiated at 0.1 mcg/kg/hour and his high dose insulin infusion was increased to 10 units/kg/hour, resulting in an increase in the tissue perfusion monitoring measurements to 73% to 75% in direct correlation with mean arterial pressure (MAP) measurements ranging from 56 to 64 mmHg. Following transfer to an intensive care setting, tissue perfusion monitoring was used over the next 2 days to guide resuscitative efforts, with monitor readings increasing to the high 70s resulting in discontinuation of epinephrine and high dose insulin therapy. It is suggested that tissue perfusion monitoring correlates well with MAP measurements, used as a surrogate measure of tissue perfusion, and may be helpful in guiding high dose insulin therapy in patients with beta blocker and calcium channel blocker overdoses (Paetow et al, 2015).
    c) ANIMAL DATA
    1) In a canine model of severe verapamil animals treated with infusions of insulin (4 units/minute), 20% dextrose (0.7 to 1.0 mL/minute to maintain blood glucose within 10 mg/dL of baseline) and potassium (2 to 4 mEq/ hr to maintain serum potassium within 1 mEq/l of baseline levels) had improved survival and coronary artery blood flow compared with control animals and those treated with epinephrine or glucagon (Kline et al, 1993).
    2) It has also been determined that insulin, administered to canines with severe verapamil toxicity, increased myocardial carbohydrate metabolism and myocardial contractility as compared with other agents recommended to treat verapamil poisoning including epinephrine, calcium chloride, and glucagon (Kline et al, 1997; Kline et al, 1995).
    4) VASOPRESSOR AGENTS
    a) INDICATION: Direct acting alpha-agonists mediate vasoconstriction by mechanisms independent of calcium influx and are preferred (Jaeger et al, 1989), although normal vascular response cannot be expected. In case review series, dopamine is the most commonly cited agent, followed by epinephrine, isoproterenol, and norepinephrine (Watling et al, 1992; Erickson et al, 1991).
    b) RETROSPECTIVE STUDY: A retrospective chart review of patients admitted to a single tertiary care center for the treatment of verapamil or diltiazem overdose, confirmed by drug or urine drug screening, from 1987 through 2012 was conducted to evaluate the role of vasopressor therapy in the management and outcome of nondihydropyridine calcium-channel blocker overdoses. A total of 48 patients (median age 45 years, range 15 to 76 years) were included and 24 (50%) were verapamil ingestions; coingestions were also likely to occur (n=38 (79%)). A second group consisted of 12 patients that met inclusion criteria except for the absence of drug or urine testing. Vasopressors were given to 33 of 48 (69%) patients and most patients received multiple vasopressors (eg, epinephrine, dopamine, dobutamine, isoproterenol, phenylephrine or norepinephrine). Of these patients, the median number of vasopressors needed to treat hypotension was 2 (range, 1 to 5); norepinephrine (n=25 (52%) and dopamine (n=19 (40%)) were the 2 most common vasopressors used. High doses of vasopressors were needed in many patients:
    1) Norepinephrine (n=25): 15 (8.4 to 24.5 mcg/min) mcg min; maximum infusion rate: 100 mcg/min
    2) Dopamine (n=19): 19 (12 to 20 mcg/kg/min; maximum infusion rate: 100 mcg/kg/min
    3) Epinephrine (n=13): 20 (10 to 26 mcg/min; maximum infusion rate: 150 mcg/min
    4) Isoproterenol (n=13): 11 (5 to 25) mcg/min; maximum infusion rate: 60 mcg/min
    5) Dobutamine (n=7): 10 (7 to 15) mcg/kg/min; maximum infusion rate 245 mcg/kg/min
    6) Phenylephrine (n=3): 100 (100 to 175 mcg/min; maximum infusion rate: 250 mcg/min
    1) Intravenous calcium (chloride or gluconate) was also administered to 38 (79%) patients with a median total dose of elemental calcium of 2 g. In addition, 26 patients received glucagon. Hyperinsulinemic euglycemia therapy was administered to 3 patients that also received multiple vasopressors. Of the 31 patients that developed bradycardia including various degrees of AV block, all rhythms responded to vasopressors or glucagon except one patient. Ischemic complications were noted in 5 (10%) patients, with most complications present prior to the initiation of vasopressor therapy. One death was reported and did not appear to be a direct result of a calcium channel blocker overdose. Seven patients did not develop significant bradycardia or hypotension and required no treatment (Levine et al, 2013).
    c) CASE REPORTS: Two patients developed severe hypotension following calcium antagonist poisoning. Despite administration of fluids and vasopressor agents, hypotension persisted. SvO2 was continuously monitored in both patients, using a fiberoptic pulmonary catheter, in order to detect tissue hypoxia. The SvO2 remained between 71% and 85%, indicating adequate tissue oxygenation, and metabolic acidosis did not occur. Gradually, the hypotension of both patients resolved without more aggressive administration of vasopressor therapy, suggesting that higher infusion rates of vasopressor agents may not be necessary in patients with refractory hypotension, provided that tissue hypoxia can be excluded after volume resuscitation. Continuous SvO2 monitoring, using a fiberoptic pulmonary artery catheter, may be a useful index of tissue oxygenation (Kamijo et al, 2006).
    5) 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).
    6) Case reports have described the use of higher than conventional doses of dopamine (up to 40 to 50 mcg/kg/min) in patients with refractory hypotension after calcium antagonist overdose (Evans & Oram, 1999). Consider high rates of infusion in patients with refractory hypotension.
    7) NOREPINEPHRINE
    a) NOREPINEPHRINE
    1) 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).
    2) DOSE
    a) 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).
    b) CHILD: Dose range: 0.1 to 2 micrograms/kilogram/minute; titrate to maintain adequate blood pressure (Kleinman et al, 2010).
    c) CAUTION: Extravasation may cause local tissue ischemia, administration by central venous catheter is advised (Peberdy et al, 2010).
    8) EPINEPHRINE
    a) EPINEPHRINE
    1) ADULT
    a) BOLUS DOSE: 1 mg intravenously/intraosseously every 3 to 5 minutes to treat cardiac arrest (Link et al, 2015).
    b) INFUSION: Prepare by adding 1 mg (1 mL of 1:1000 (1 mg/mL) solution) to 250 mL D5W, yielding a concentration of 4 mcg/mL, and infuse this solution IV at a rate of 1 mcg/min to 10 mcg/min (maximum rate) (Lieberman et al, 2010). Used primarily for severe hypotension (systolic blood pressure 70 mm Hg), or anaphylaxis associated with hemodynamic or respiratory compromise, may also be used for symptomatic bradycardia if atropine and transcutaneous pacing are unsuccessful or not immediately available (Peberdy et al, 2010).
    2) PEDIATRIC
    a) CARDIOPULMONARY RESUSCITATION: INTRAVENOUS/INTRAOSSEOUS: OLDER INFANTS/CHILDREN: 0.01 mg/kg (0.1 mL/kg of 1:10,000 (0.1 mg/mL) solution); maximum 1 mg/dose. May repeat dose every 3 to 5 minutes (Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008; Sorrentino, 2005). ENDOTRACHEAL: OLDER INFANTS/CHILDREN: 0.1 mg/kg (0.1 mL/kg of 1:1000 (1 mg/mL) solution). Maximum 2.5 mg/dose (maximum total dose: 10 mg). May repeat every 3 to 5 minutes (Kleinman et al, 2010; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008). Follow ET administration with saline flush or dilute in isotonic saline (1 to 5 mL) based on the child's size (Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008).
    b) INFUSION: Used for the treatment of refractory hypotension, bradycardia, severe anaphylaxis. DOSE: 0.1 to 1 mcg/kg/min, titrate dose; start at lowest dose needed to reach desired clinical effects. Doses as high as 5 mcg/kg/min may sometimes be necessary. High dose epinephrine infusion may be useful in the setting of beta blocker poisoning (Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008).
    3) CAUTION
    a) Extravasation may cause severe local tissue ischemia (Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008); infusion through a central venous catheter is advised.
    b) Epinephrine has been found to be useful in other cases of calcium channel blocker (ie, diltiazem, verapamil) overdose. However, multiple vasopressors are likely to be needed to maintain clinical improvement following a significant overdose (Levine et al, 2013).
    c) Large doses may be required (Levine et al, 2013).
    d) One-time bolus doses of 1 mg have been used in addition to bolus-infusion regimens (Erickson et al, 1991). Epinephrine 1 mg bolus followed by 0.2 to 0.6 mcg/kg/min improved both SBP and urine flow for 18 hours (Henderson et al, 1992). Infusion rates up to 100 mcg/min have been reported (Anthony et al, 1986).
    e) ANIMAL STUDY: Intravenous epinephrine administration (1 to 2 mcg/kg bolus dose, followed by a maintenance infusion of 8 to 12 mcg/min for 30 minutes) was shown to be effective in significantly improving the mean arterial pressure (MAP) and both the systolic and diastolic blood pressures, as well as improving heart rate and A-V conduction, of anesthetized dogs who were given diltiazem infusions in high concentrations (Dimich et al, 1988).
    9) ISOPROTERENOL
    a) ISOPROTERENOL INDICATIONS
    1) Used for temporary control of hemodynamically significant bradycardia in a patient with a pulse; generally other modalities (atropine, dopamine, epinephrine, dobutamine, pacing) should be used first because of the tendency to develop ischemia and dysrhythmias with isoproterenol (Neumar et al, 2010).
    2) ADULT DOSE: Infuse 2 micrograms per minute, gradually titrating to 10 micrograms per minute as needed to desired response (Neumar et al, 2010).
    3) CAUTION: Decrease infusion rate or discontinue infusion if ventricular dysrhythmias develop(Prod Info Isuprel(TM) intravenous injection, intramuscular injection, subcutaneous injection, intracardiac injection, 2013).
    4) PEDIATRIC DOSE: Not well studied. Initial infusion of 0.1 mcg/kg/min titrated as needed, usual range is 0.1 mcg/kg/min to 1 mcg/kg/min (Prod Info Isuprel(TM) intravenous injection, intramuscular injection, subcutaneous injection, intracardiac injection, 2013).
    10) CAUTIONS: Normal vascular response may not be seen. Doses of isoproterenol, while beneficial to cardiac conduction (beta-1 effect), may worsen peripheral vascular resistance (beta-2 effects) (Krenzelok, 1991).
    11) PHENYLEPHRINE
    a) PHENYLEPHRINE
    1) MILD OR MODERATE HYPOTENSION
    a) INTRAVENOUS: ADULT: Usual dose: 0.2 mg; range: 0.1 mg to 0.5 mg. Maximum initial dose is 0.5 mg. A 0.5 mg IV dose can elevate the blood pressure for approximately 15 min (Prod Info phenylephrine HCl subcutaneous injection, intramuscular injection, intravenous injection, 2011). PEDIATRIC: Usual bolus dose: 5 to 20 mcg/kg IV repeated every 10 to 15 min as needed (Taketomo et al, 1997).
    2) CONTINUOUS INFUSION
    a) PREPARATION: Add 10 mg (1 mL of a 1% solution) to 500 mL of normal saline or dextrose 5% in water to produce a final concentration of 0.2 mg/mL.
    b) ADULT DOSE: To raise blood pressure rapidly; start an initial infusion of 100 to 180 mcg/min until blood pressure stabilizes; then reduce infusion to 40 to 60 mcg/min titrated to desired effect. If necessary, additional doses in increments of 10 mg or more may be added to the infusion solution and the rate of flow titrated to the desired effect (Prod Info phenylephrine HCl subcutaneous injection, intramuscular injection, intravenous injection, 2011).
    c) PEDIATRIC DOSE: Intravenous infusion should begin at 0.1 to 0.5 mcg/kg/min; titrate to the desired effect (Taketomo et al, 1997).
    3) ADVERSE EFFECTS
    a) Headache, reflex bradycardia, excitability, restlessness and rarely dysrhythmias may develop (Prod Info phenylephrine HCl subcutaneous injection, intramuscular injection, intravenous injection, 2011).
    12) DOBUTAMINE
    a) DOBUTAMINE
    1) DOSE: ADULT: Infuse at 5 to 10 micrograms/kilogram/minute IV. PEDIATRIC: Infuse at 2 to 20 micrograms/kilogram/minute IV or intraosseous, titrated to desired effect (Peberdy et al, 2010; Kleinman et al, 2010).
    2) CAUTION: Decrease infusion rate if ventricular ectopy develops (Prod Info dobutamine HCl 5% dextrose intravenous injection, 2012).
    13) GLUCAGON
    a) INDICATIONS: Glucagon exerts chronotropic and inotropic effects and can help reverse hypotension but may not improve heart rate.
    b) DOSES: ADULT: Optimal dosing in calcium antagonist poisoning is not established. Initially, 3 to 5 mg IV, slowly over 1 to 2 minutes; may repeat treatment with a dose of 4 to 10 mg if there is no hemodynamic improvement within 5 minutes. CHILD: 50 mcg/kg; repeat doses may be used due to the short half-life of glucagon (DeRoos, 2011).
    c) Empiric dosing has ranged from single doses of 2 mg (Anthony et al, 1986) to 17 mg (Ramoska et al, 1993). Continuous infusion of up to 5 mg/hr have also been used (Doyon & Roberts, 1993; Takahashi et al, 1993; Mahr et al, 1997; Papadopoulos & O'Neil, 2000).
    d) In a canine model of severe verapamil overdose glucagon (2.5 mg bolus followed by an infusion of 2.5 mg over 1 hour) increased cardiac output and heart rate and restored sinus rhythm without affecting mean arterial pressure or total peripheral resistance (Stone et al, 1995).
    e) CASE REPORT (CHILD): A 13-year-old girl intentionally ingested an unknown amount of 30 mg nifedipine and 0.1 mg clonidine and subsequently developed severe hypotension unresponsive to treatment with fluids, calcium chloride, and dopamine. Glucagon was then intravenously administered (a single dose of 5 mg followed by a continuous infusion of 3 mg/hr) with successful normalization of her blood pressure (Fant et al, 1997).
    14) PHOSPHODIESTERASE INHIBITORS
    a) INAMRINONE
    1) INAMRINONE
    a) PREPARATION: Dilute solution for infusion to a final concentration of 1 to 3 milligrams per milliliter in normal saline (Prod Info inamrinone intravenous solution, 2002).
    b) ADULT DOSE: Loading dose of 0.75 milligram/kilogram over 2 to 3 minutes followed by a maintenance infusion of 5 to 10 micrograms/kilogram per minute titrate to hemodynamic response. Repeat initial loading dose after 30 minutes if necessary (Prod Info inamrinone intravenous solution, 2002).
    c) PEDIATRIC DOSE: The safety and effectiveness of inamrinone in the pediatric population has not been established (Prod Info inamrinone intravenous solution, 2002).
    d) CAUTIONS: May cause hypotension and dysrhythmias (Prod Info inamrinone intravenous solution, 2002).
    b) ENOXIMONE
    1) VERAPAMIL: After ingesting 2800 mg of atenolol and 1600 mg of verapamil, a 57-year-old man with a history of ischemic heart disease (two previous acute myocardial infarctions) developed hypotension (BP 80/50 mmHg), and bradycardia (40 beats/min). An ECG revealed a sinus bradycardia with a first-degree heart block, previously absent. Despite treatment with fluid resuscitation, calcium salts, and norepinephrine/epinephrine inotropic support, only a modest hemodynamic effect was observed. Following treatment with a bolus of enoximone 1 mg/kg and a continuous infusion at 0.5 mcg/kg/min for 5 days, his hemodynamic status improved. The authors suggested that enoximone, a phosphodiesterase III inhibitor, may be an alternative agent to glucagon as they have an inotropic effect which is not mediated by a beta receptor (Sandroni et al, 2004).
    15) L-CARNITINE
    a) L-carnitine may be useful to treat hypotension in the setting of calcium channel blocker overdose. It is not well studied but an animal study and one human case report suggest efficacy. The dose used in the human case report was 6 g IV followed by 1 g IV every 4 hours.
    b) MECHANISM: Carnitine is synthesized in several human organs, including liver and kidneys. It is produced from amino acids lysine and methionine in 2 optical isomeric forms with only L-carnitine being the biological active isomer. It transports fatty acids from the cellular cytoplasm into the mitochondrial matrix by the carnitine shuttle. The fatty acids undergo beta-oxidation to form acetyl-CoA in the mitochondrial matrix. It then enters the Krebs cycle to produce cellular energy. In one animal study, it was proposed that verapamil toxicity changed the cardiac metabolism from free fatty acids to carbohydrate. It has been suggested that L-carnitine in combination with high-dose insulin can decrease insulin resistance, promote intracellular glucose transport, increase the uptake and hepatic beta-oxidation of fatty acids, and increase calcium channel sensitivity in patients with calcium channel blocker toxicity (Perez et al, 2011; St-Onge et al, 2013).
    c) ANIMAL STUDY: In a controlled, blinded animal study, 16 male rats were anesthetized and received a constant infusion of 5 mg/kg/hr of verapamil to produce severe verapamil toxicity. All animals received a bolus of 50 mg/kg of either L-carnitine or normal saline 5 minutes later. Mean arterial pressures (MAP) and heart rates of animals were recorded at baseline and at 15 and 30 minutes after the start of the experiment. The survival time was evaluated using the length of time (in minutes) from the initiation of verapamil until either the MAP had reached 10% of the baseline values or 150 minutes had passed without a suitable reduction in blood pressure. Animals in the L-carnitine group survived a median time of 140.75 minutes (interquartile range [IQR] = 98.6 to 150 minutes) as compared with 49.19 minutes (IQR = 39.2 to 70.97 minutes; p=0.0001) in the normal saline group. All animals in the L-carnitine group and one in the saline group survived over 90 minutes. Four animals in the L-carnitine group also survived the 150-minute protocol. At 15 minutes, a higher mean MAP was observed in the carnitine group (63 mm Hg; SD +/- 19.4 mmHg) as compared with the saline group (42.6 mmHg; SD +/_ 22.9 mmHg; p=0.047; a difference of 20.4 mmHg; 95% CI = 0.25 to 40.65 mmHg) (Perez et al, 2011).
    d) AMLODIPINE/METFORMIN OVERDOSE: A 68-year-old man with a history of hypertension, type II diabetes, benign prostate hypertrophy, and chronic anemia, ingested 300 mg of amlodipine, 3500 mg of metformin, and ethanol in a suicide attempt. He presented pale, diaphoretic, and hemodynamically unstable (BP 56/42 mmHg, heart rate 77 beats/min). Laboratory results revealed a blood glucose concentration of 13 mmol/L, serum creatinine of 103 mcmol/L (normal, 50 to 100 mcmol/L), serum sodium of 126 mmol/L, and an increased CK of 871 mmol/L. The arterial blood gas analysis an hour later showed a pH of 7, pCO2 of 42, bicarbonate of 10 mmol/L, and lactate of 14.1 mmol/L (anion-gap of 22). Despite supportive therapy for 10 hours, including calcium, glucagon, vasopressors, high-dose insulin (HDI; even after more than 1 hour of infusion at 320 Units/hr), dextrose, lipid emulsion, and bicarbonate for metabolic acidosis, he remained in refractory shock (BP 110/50 mmHg, norepinephrine running at 80 mcg/min), hyperglycemic (33 mmol/L), oliguric, and acidotic. Continuous renal replacement therapy was not initiated because he was considered too unstable. About 11 hours after presentation, he received L-carnitine 6 g IV followed by 1 g IV every 4 hours. His condition began to improve about 30 minutes after L-carnitine loading dose and his norepinephrine requirement continued to decrease. Vasopressors were discontinued within 36 hours of initial presentation and he was extubated successfully within 4 days of presentation. His serum amlodipine concentration was 83 ng/mL (therapeutic: 3 to 11 ng/mL) on arrival and peaked at 160 ng/mL several hours later. His metformin concentration was 24 mcg/mL (therapeutic: 1 to 2 mcg/mL) at arrival (St-Onge et al, 2013).
    16) VASOPRESSIN
    a) VERAPAMIL/LACK OF IMPROVEMENT: A 41-year-old woman intentionally ingested 80 tablets of sustained release verapamil (total dose 19.2 g) and developed severe toxicity about 14 hours after exposure. She became hypotensive and bradycardic with a third degree AV. Initial treatment included calcium, fluids, dopamine and isoproterenol; norepinephrine, epinephrine and vasopressin were added with only temporary clinical improvement in hypotension. Other treatments included intubation and transvenous pacing and hyperglycemia-euglycemia insulin therapy and glucagon. Acute renal failure developed and CVVH was started. On hospital day 4, lipid therapy was begun. Within 3 hours the norepinephrine dose was reduced and within 48 hours vasopressin was stopped. By day 6, transvenous pacing was no longer needed. Despite cardiac improvement, she developed further complications necessitating an urgent colectomy (Liang et al, 2011).
    b) AMLODIPINE AND DILTIAZEM: Two patients were given vasopressin infusions for the treatment of refractory hypotension following intentional ingestions of 800 mg amlodipine and 4800 mg sustained-release diltiazem, respectively. The vasopressin infusion, in the first patient, was initiated at a rate of 2.4 International Units (IU)/hour and titrated to 4.8 IU/hour over two hours. The second patient received a 20 IU bolus of vasopressin followed by a 4 IU/hour infusion. The hypotension resolved in both patients and they were subsequently discharged to rehabilitation facilities (Kanagarajan et al, 2007).
    c) ANIMAL STUDY: In a porcine model, 18 anesthetized swine, each receiving a verapamil infusion of 1 mg/kg/hr until the mean arterial blood pressure (MAP) decreased to 70% of baseline, were divided into two groups: one group that received a vasopressin infusion of 0.01 units/kg/min (n=8) and the control group that received an equal volume of normal saline (n=10). MAP, heart rate, and cardiac output were then measured every 5 minutes until t=60 minutes. The results showed that there was no significant difference in MAP, heart rate, and cardiac output between the two groups. Four of 8 animals in the vasopressin group died as compared with 2 of 10 animals in the control group. Death appeared to be related to hypotension and low cardiac output. Based on the results of this study, the authors conclude that treatment with vasopressin actually decreased the survival of swine following verapamil intoxication as compared to the swine treated with normal saline alone (Barry et al, 2005)
    17) SODIUM BICARBONATE
    a) ANIMAL DATA: In a study involving swine to determine the efficacy of hypertonic sodium bicarbonate in treating hypotension associated with severe verapamil toxicity, it was determined that swine, treated with 4 mEq/kg of 8.4% sodium bicarbonate given intravenously over 4 minutes, experienced a significant increase in mean arterial pressure and cardiac output as compared with animals in the control group, who were given 0.6% sodium chloride in 10% mannitol (Tanen et al, 2000).
    18) METARAMINOL
    a) AMLODIPINE: A 43-year-old man developed hypotension (BP 65/40 mmHg) after ingesting 560 mg of amlodipine. Despite treatment with fluid resuscitation, calcium salts, glucagon and norepinephrine/epinephrine inotropic support, there was no hemodynamic response. Following treatment with metaraminol (a loading bolus of 2 mg [equivalent to 25 mcg/kg] and intravenous infusion of 1 mcg/kg/min [83 mcg/min] for 36 hours), there was improvement in his blood pressure, cardiac output and urine output (Wood et al, 2005).
    19) TERLIPRESSIN
    a) FELODIPINE: A 61-year-old man developed hypotension (75/50 mmHg on admission) after ingesting 140 mg of felodipine. Despite administration of epinephrine and norepinephrine, the patient's hypotension persisted (mean arterial pressure 47 mmHg). A continuous infusion of 0.05 mcg/kg/min of terlipressin, a vasopressor, was initiated, resulting in the mean arterial pressure increasing from 47 to 95 mmHg; systemic vascular resistance and SvO2 also increased (Leone et al, 2005).
    D) 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) CASE REPORTS
    a) VERAPAMIL: A 41-year-old woman intentionally ingested 80 tablets of sustained release verapamil (total dose 19.2 g). She was decontaminated with multiple doses of activated charcoal. Fourteen hours after exposure, the patient suddenly became lethargic with a decrease in oxygen saturation requiring a 100% non-rebreather. She became hypotensive and bradycardic; a third degree AV block was noted on ECG. Initial treatment included calcium, fluids, dopamine and isoproterenol; norepinephrine, epinephrine and vasopressin were added with only temporary clinical improvement in hypotension. Other treatments included intubation and transvenous pacing and hyperglycemia-euglycemia insulin therapy and glucagon. Acute renal failure developed and CVVH was started. On hospital day 4, lipid therapy (100 mL bolus of 20% intralipid followed by 0.5 mL/kg/h; total dose: 4200 mL over 7 days) was begun. Within 3 hours the norepinephrine dose was reduced and within 48 hours vasopressin was stopped. By day 6, transvenous pacing was no longer needed. Despite cardiac improvement, her clinical course was further complicated by the development of a pneumatosis intestinalis necessitating an urgent colectomy. On day 55, the patient was discharged to a skilled nursing facility (Liang et al, 2011).
    b) AMLODIPINE: A 71-year-old woman with a medical history of hypertension, emphysema, and depression presented 1.5 hours after ingesting 27 tablets of amlodipine 5 mg. She was alert, oriented, and in no distress, and her vital signs were: temperature 36.6 degrees C, blood pressure (BP 85/44 mm Hg), pulse (heart rate, 79 beats/min), respiratory rate (20/min). An initial laboratory result revealed hyponatremia and anemia; all other laboratory results were normal. She developed hypotension (systolic BP, 79 mmHg) 2 hours later. Despite symptomatic therapy, she remained hypotensive in the ICU, and her urine output decreased to 10 mL/hr. Her condition deteriorated and pulmonary edema was noted clinically and on chest x-ray. She was intubated and her peri-intubation arterial blood gas (ABG) revealed pH=7.17, pCO2=31, and pO2=80. Based on a written lipid emulsion therapy protocol (written protocol: Intralipid 20%: 1.5 mL/kg over 1 min; then an infusion of 0.25 mL/kg/min; max total dose, 8 mL/kg. In practice, for an adult weighing 70 kg: take a 500 mL bag of Intralipid 20% and a 50 mL syringe; draw up 50 mL and give stat IV, x 2; then attach the bag to an IV administration set (macrodrip) and run it IV over the next 25 minutes), a maximum of 387.5 mL of intralipid for a 50 kg patient was ordered and treatment was started 12.5 hours after presentation. However, she inadvertently received a total of 2000 mL of 20% intralipid which ran for 4.5 hours (greater than 5 times the maximum suggested dose). Because of severe lipemia, no metabolic panel and CBC analysis could be obtained. There were no other obvious clinical affects from the intravenous lipid overdose On hospital day 2, the treatment team felt that recovery from the amlodipine overdose was unlikely. After her family decided to withdraw care, she died within the next 24 hours (West et al, 2010).
    c) VERAPAMIL: A 32-year-old man intentionally ingested large quantities of multiple medications, one of which was 13.44 grams of verapamil. He was found 12 hours later to be poorly responsive with incoherent speech and hypotensive (69/26 mmHg) with a pulse of 55 beats/min. Fluids, pressors, calcium and glucagon were all administered, but he remained hypotensive with junctional bradycardia, metabolic acidosis and renal insufficiency. Then, upon transfer, 100 mL of 20% lipids was infused followed by a 0.5 mL/kg/hr infusion for 24 hours. Blood pressure improved within an hour of initiation of intravenous lipid emulsion, and he made a full recovery (Young et al, 2009).
    d) VERAPAMIL: A 39-year-old woman presented to the emergency department with dyspnea, chest tightness, lethargy, diaphoresis, and hypotension (76/41 mmHg) after intentionally ingesting 17 240-mg tablets of extended release verapamil (total dose ingested 4.08 g). Despite treatment with IV fluids and norepinephrine therapy, the patient's hypotension persisted. Approximately 17 hours post-presentation, the patient was switched to dopamine, without effect, and, approximately 15 hours later, the patient was given 100 mL of 20% lipids, administered intravenously over 20 minutes, followed by a continuous infusion of 0.5 mL/kg/hour for the next 8 hours. During this time period, the patient's blood pressure improved and dopamine therapy was gradually discontinued (Franxman et al, 2011).
    4) ANIMAL DATA
    a) In a dog model of severe verapamil poisoning, dogs treated with 7 mg/kg 20% intravenous fat emulsion (after atropine and three doses of calcium chloride) had improved mean arterial pressure and 120 minute survival (100% vs 14%) compared with dogs treated with the same doses of atropine and calcium chloride, and 7 mg/kg 0.9% saline (Bania et al, 2007).
    b) In a rat model of lethal verapamil overdose, intralipid treatment prolonged survival (44 +/-21 vs 24 +/- 9 minutes; p=0.003) and doubled median lethal dose (25.7 mg/kg [95% CI = 24.7 to 26.7] vs 13.6 mg/kg [95% CI = 12.2 to 15]). In addition, during verapamil infusion, a less marked decrease in heart rate was noted in the Intralipid-treated group (6.8 bpm [95% CI=8.3 to 5.2] for intralipid vs 10.7 bpm [95% CI = 12.6 to 8.9] for saline; p=0.001) (Tebbutt et al, 2006).
    E) BRADYCARDIA
    1) CALCIUM
    a) INDICATIONS: Calcium is used to reverse hypotension and improve cardiac conduction defects. Calcium administration has been most effective in overcoming mild toxicity from small overdoses or therapeutic use and is less useful in massive overdose cases since calcium channel blockade is non-competitive (DeRoos, 2011; Pearigen & Benowitz, 1991; Krenzelok, 1991; Clark & Hanna, 1993), but was successful in 11 of 30 cases in one series (Hofer et al, 1993).
    b) DRUG OF CHOICE: In some studies, calcium chloride is thought to produce more predictable increases in extracellular ionized calcium and a greater positive inotropic response (White et al, 1976; Haynes et al, 1985); however, other sources have found no differences in efficacy of calcium chloride and calcium gluconate. Calcium chloride provides 3 times more elemental calcium (13.4 mEq) than calcium gluconate (4.3 mEq) in the commercially available 1 gram ampules (DeRoos, 2011).
    c) ADULT DOSE: Optimal dosing is not established; begin with an initial IV infusion of about 13 to 25 mEq of calcium (10 to 20 mL of 10% calcium chloride or 30 to 60 mL of 10% calcium gluconate) followed by either repeat boluses every 15 to 20 minutes up to 3 to 4 doses or a continuous infusion of 0.5 mEq/kg/hr of calcium (0.2 to 0.4 mL/kg/hr of 10% calcium chloride or 0.6 to 1.2 mL of 10% calcium gluconate) (DeRoos, 2011). Some authors advocate administering 1 gram of calcium salts every 2 to 3 minutes until conduction block is reversed or clinical evidence of hypercalcemia develops (Howarth et al, 1994; Buckley et al, 1994). Calcium dosing should be titrated to hemodynamic response rather than serum calcium concentration alone; central venous or pulmonary artery catheters may be useful to guide therapy. Monitor ECG and ionized calcium concentration.
    1) CASE REPORT/HIGH DOSE ADMINISTRATION: A 40-year-old woman, with chronic renal insufficiency, developed hypotension and bradycardia after ingesting 30 mg of amlodipine. The patient's blood pressure and heart rate normalized following high-dose intravenous administration of calcium chloride, consisting of 4 grams given in the emergency department followed by 1.5 grams every 20 minutes (for a total of 13 grams given over 2 hours) (Hung & Olson, 2007).
    d) HYPERCALCEMIA: Some degree of hypercalcemia may be necessary before severely intoxicated patients respond to aggressive calcium therapy, but the optimal calcium regimen has not been established. In patients who have received large doses of calcium for severe calcium channel blocker overdose (attaining serum calcium concentrations up to twice the upper limits of normal), hypercalcemia generally resolves with in 48 hours without clinically apparent adverse effects from hypercalcemia (Howarth et al, 1994; Buckley et al, 1994).
    1) CASE REPORT: Hypercalcemia with levels as high as 19.2 mg/dL (9.9 mg/dL upper limit of normal) secondary to treatment with calcium salts are reported during aggressive therapy (Buckley et al, 1993). Such aggressive treatment with calcium salts may be necessary to reverse conduction defects (Howarth et al, 1994).
    2) CASE REPORT: Hypercalcemia (16.3 mg/dL) occurred following aggressive calcium chloride treatment in a 45-year-old woman following an overdose ingestion of sustained-release diltiazem. There were no ECG manifestations due to the hypercalcemia and the patient recovered uneventfully over the next 4 days (Hantsch et al, 1997).
    3) CASE REPORT: Hypercalcemia (peak calcium concentration 32.3 mg/dL) was reported in a 61-year-old woman who received a continuous calcium chloride infusion, initiated at 4 g/hour and decreased to 2 g/hour, following inadvertent ingestion of a second dose (360 mg) of her sustained release verapamil. At the same time, the patient, who had no previous history of renal impairment, developed a decrease in urine output that progressed to complete anuria. Analysis of the urine sediment showed coarse granular casts, indicating acute tubular necrosis. After initiating continuous hemodialysis, the patient's calcium level decreased to 8.3 mg/dL, however her condition deteriorated with persistent anuria and multi-organ failure resulting in her death 17 days post-admission. An autopsy revealed acute tubular necrosis, liver necrosis, multiple infarctions of the spleen, and dermal lesions on the trunk and extremities, secondary to calciphylaxis (Sim & Stevenson, 2008).
    4) CASE SERIES: In one series, doses varied from 4.5 mEq to 95.2 mEq, with no evidence of a dose-response relation (Ramoska et al, 1993).
    e) PRECAUTIONS: Hypotension generally does not respond as well as conduction disturbances to calcium. Significant hypercalcemia may be necessary before severely intoxicated patients respond to aggressive calcium therapy, but the optimal calcium regimen has not been established. If a patient does not respond after a doubling of the ionized calcium concentration, further calcium treatment may not be beneficial. If the patient has also ingested digoxin, avoid administering calcium until after digoxin-specific Fab is administered to prevent worsening digoxin toxicity (DeRoos, 2011).
    2) ATROPINE
    a) INDICATION: Bradydysrhythmia contributing to hypotension. May be more effective after calcium administration (Howarth et al, 1994).
    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).
    c) Up to 2 mg has been administered without effect (Ramoska et al, 1993).
    d) PRECAUTIONS: Atropine primarily blocks vagal effects at the SA node while calcium antagonists affect AV conduction; marked improvements in rate may not be seen.
    3) GLUCAGON
    a) INDICATIONS: Glucagon exerts chronotropic and inotropic effects and can help reverse hypotension but may not improve heart rate in calcium antagonist intoxication.
    b) DOSES: ADULT: Optimal dosing in calcium antagonist poisoning is not established. Initially, 3 to 5 mg IV, slowly over 1 to 2 minutes; may repeat treatment with a dose of 4 to 10 mg if there is no hemodynamic improvement within 5 minutes. CHILD: 50 mcg/kg; repeat doses may be used due to the short half-life of glucagon (DeRoos, 2011).
    c) Empiric dosing has ranged from single doses of 2 mg (Anthony et al, 1986) to 17 mg (Ramoska et al, 1993). Continuous infusion of up to 5 mg/hr have also been used (Doyon & Roberts, 1993; Takahashi et al, 1993; Mahr et al, 1997; Papadopoulos & O'Neil, 2000).
    4) ISOPROTERENOL
    a) INDICATION: Predominant beta-1 effects stimulate discharge rate at SA node, increase heart rate, and improve contractility (Krenzelok, 1991).
    b) ISOPROTERENOL INDICATIONS
    1) Used for temporary control of hemodynamically significant bradycardia in a patient with a pulse; generally other modalities (atropine, dopamine, epinephrine, dobutamine, pacing) should be used first because of the tendency to develop ischemia and dysrhythmias with isoproterenol (Neumar et al, 2010).
    2) ADULT DOSE: Infuse 2 micrograms per minute, gradually titrating to 10 micrograms per minute as needed to desired response (Neumar et al, 2010).
    3) CAUTION: Decrease infusion rate or discontinue infusion if ventricular dysrhythmias develop(Prod Info Isuprel(TM) intravenous injection, intramuscular injection, subcutaneous injection, intracardiac injection, 2013).
    4) PEDIATRIC DOSE: Not well studied. Initial infusion of 0.1 mcg/kg/min titrated as needed, usual range is 0.1 mcg/kg/min to 1 mcg/kg/min (Prod Info Isuprel(TM) intravenous injection, intramuscular injection, subcutaneous injection, intracardiac injection, 2013).
    5) PACEMAKER
    a) INDICATION: Consider using a pacemaker device in severely symptomatic patients (Rodgers et al, 1989; Quezado et al, 1991; MacDonald & Alguire, 1992; Bizovi et al, 1998; Sanders et al, 1998).
    6) INTRA-AORTIC BALLOON PUMP
    a) CASE REPORT: Intra-aortic balloon counterpulsation was required in addition to pacing in a 17-year-old girl who had taken an unknown quantity of acebutolol 400 mg capsules in addition to 480 mg sustained-release verapamil (Welch et al, 1992). Other successful applications have been reported (Melanson et al, 1993; Williamson & Dunham, 1996).
    7) CARDIOPULMONARY BYPASS
    a) CASE REPORT: Cardiopulmonary bypass was used in a 25-month-old child after verapamil overdose. Serum verapamil concentrations fell during the procedure, allowing successful pacing, but rose again after discontinuation of the procedure, and he subsequently died (Hendren et al, 1989).
    b) CASE REPORT: A 41-year-old man ingested 4800 to 6400 mg of verapamil in a suicide attempt and developed cardiac arrest. Despite cardiopulmonary resuscitation, percutaneous cardiopulmonary bypass was required 2.5 hours after cardiac arrest. Complete recovery was reported 6 months after exposure (Holzer et al, 1999).
    c) CASE REPORT: A 47-year-old man died after ingesting 10.8 grams of diltiazem despite aggressive treatment with calcium, glucagon, epinephrine, pressors and cardiopulmonary bypass (Martin et al, 1994a).
    F) EXTRACORPOREAL MEMBRANE OXYGENATION
    1) Extracorporeal membrane oxygenation has been used successfully in a limited number of reports of severe calcium channel blocker toxicity.
    2) CASE REPORT: A 16-year-old girl experienced multiple asystolic cardiac arrests with atrial standstill, refractory to therapy with high-dose epinephrine and transcutaneous cardiac pacing, after intentionally ingesting 12 grams of sustained-release diltiazem tablets. The patient's hemodynamic status significantly improved, with a return of brainstem reflexes, following a 48-hour period of extracorporeal membrane oxygenation (ECMO) (Durward et al, 2003).
    3) CASE REPORT: A 36-year-old man presented to the emergency department with decreased level of consciousness, dyspnea, hypoxemia (O2 sat 91%), and hypotension (80/40 mmHg) approximately 2 hours after intentionally ingesting 10 g atenolol and an unknown amount of nifedipine, lacidipine, fluoxetine, and sertraline. An ECG indicated prolonged QT interval and QRS widening. The patient rapidly deteriorated hemodynamically, developed cardiac arrest (successfully resuscitated), and persistent metabolic acidosis and shock with multiple organ failure despite aggressive decontamination and supportive therapies. ECMO was initiated 2 hours post-admission along with high-volume continuous veno-venous hemofiltration (HV-CVVH). Over the next 48 hours, the patient became hemodynamically stable and was weaned from ECMO; however, the patient's clinical course was complicated by the development of progressive neurologic impairment, resulting in a persistent reduction in motor skills, impaired coordination, gait ataxia, and mild aphasia (Rona et al, 2011).
    G) SEIZURE
    1) SUMMARY
    a) Attempt initial control with a benzodiazepine (eg, diazepam, lorazepam). If seizures persist or recur, administer phenobarbital or propofol.
    b) Monitor for respiratory depression, hypotension, and dysrhythmias. Endotracheal intubation should be performed in patients with persistent seizures.
    c) Evaluate for hypoxia, electrolyte disturbances, and hypoglycemia (or, if immediate bedside glucose testing is not available, treat with intravenous dextrose).
    2) DIAZEPAM
    a) ADULT DOSE: Initially 5 to 10 mg IV, OR 0.15 mg/kg IV up to 10 mg per dose up to a rate of 5 mg/minute; may be repeated every 5 to 20 minutes as needed (Brophy et al, 2012; Prod Info diazepam IM, IV injection, 2008; Manno, 2003).
    b) PEDIATRIC DOSE: 0.1 to 0.5 mg/kg IV over 2 to 5 minutes; up to a maximum of 10 mg/dose. May repeat dose every 5 to 10 minutes as needed (Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008).
    c) Monitor for hypotension, respiratory depression, and the need for endotracheal intubation. Consider a second agent if seizures persist or recur after repeated doses of diazepam .
    3) NO INTRAVENOUS ACCESS
    a) DIAZEPAM may be given rectally or intramuscularly (Manno, 2003). RECTAL DOSE: CHILD: Greater than 12 years: 0.2 mg/kg; 6 to 11 years: 0.3 mg/kg; 2 to 5 years: 0.5 mg/kg (Brophy et al, 2012).
    b) MIDAZOLAM has been used intramuscularly and intranasally, particularly in children when intravenous access has not been established. ADULT DOSE: 0.2 mg/kg IM, up to a maximum dose of 10 mg (Brophy et al, 2012). PEDIATRIC DOSE: INTRAMUSCULAR: 0.2 mg/kg IM, up to a maximum dose of 7 mg (Chamberlain et al, 1997) OR 10 mg IM (weight greater than 40 kg); 5 mg IM (weight 13 to 40 kg); INTRANASAL: 0.2 to 0.5 mg/kg up to a maximum of 10 mg/dose (Loddenkemper & Goodkin, 2011; Brophy et al, 2012). BUCCAL midazolam, 10 mg, has been used in adolescents and older children (5-years-old or more) to control seizures when intravenous access was not established (Scott et al, 1999).
    4) LORAZEPAM
    a) MAXIMUM RATE: The rate of intravenous administration of lorazepam should not exceed 2 mg/min (Brophy et al, 2012; Prod Info lorazepam IM, IV injection, 2008).
    b) ADULT DOSE: 2 to 4 mg IV initially; repeat every 5 to 10 minutes as needed, if seizures persist (Manno, 2003; Brophy et al, 2012).
    c) PEDIATRIC DOSE: 0.05 to 0.1 mg/kg IV over 2 to 5 minutes, up to a maximum of 4 mg/dose; may repeat in 5 to 15 minutes as needed, if seizures continue (Brophy et al, 2012; Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008; Sreenath et al, 2009; Chin et al, 2008).
    5) PHENOBARBITAL
    a) ADULT LOADING DOSE: 20 mg/kg IV at an infusion rate of 50 to 100 mg/minute IV. An additional 5 to 10 mg/kg dose may be given 10 minutes after loading infusion if seizures persist or recur (Brophy et al, 2012).
    b) Patients receiving high doses will require endotracheal intubation and may require vasopressor support (Brophy et al, 2012).
    c) PEDIATRIC LOADING DOSE: 20 mg/kg may be given as single or divided application (2 mg/kg/minute in children weighing less than 40 kg up to 100 mg/min in children weighing greater than 40 kg). A plasma concentration of about 20 mg/L will be achieved by this dose (Loddenkemper & Goodkin, 2011).
    d) REPEAT PEDIATRIC DOSE: Repeat doses of 5 to 20 mg/kg may be given every 15 to 20 minutes if seizures persist, with cardiorespiratory monitoring (Loddenkemper & Goodkin, 2011).
    e) MONITOR: For hypotension, respiratory depression, and the need for endotracheal intubation (Loddenkemper & Goodkin, 2011; Manno, 2003).
    f) SERUM CONCENTRATION MONITORING: Monitor serum concentrations over the next 12 to 24 hours. Therapeutic serum concentrations of phenobarbital range from 10 to 40 mcg/mL, although the optimal plasma concentration for some individuals may vary outside this range (Hvidberg & Dam, 1976; Choonara & Rane, 1990; AMA Department of Drugs, 1992).
    6) OTHER AGENTS
    a) If seizures persist after phenobarbital, propofol or pentobarbital infusion, or neuromuscular paralysis with general anesthesia (isoflurane) and continuous EEG monitoring should be considered (Manno, 2003). Other anticonvulsants can be considered (eg, valproate sodium, levetiracetam, lacosamide, topiramate) if seizures persist or recur; however, there is very little data regarding their use in toxin induced seizures, controlled trials are not available to define the optimal dosage ranges for these agents in status epilepticus (Brophy et al, 2012):
    1) VALPROATE SODIUM: ADULT DOSE: An initial dose of 20 to 40 mg/kg IV, at a rate of 3 to 6 mg/kg/minute; may give an additional dose of 20 mg/kg 10 minutes after loading infusion. PEDIATRIC DOSE: 1.5 to 3 mg/kg/minute (Brophy et al, 2012).
    2) LEVETIRACETAM: ADULT DOSE: 1000 to 3000 mg IV, at a rate of 2 to 5 mg/kg/min IV. PEDIATRIC DOSE: 20 to 60 mg/kg IV (Brophy et al, 2012; Loddenkemper & Goodkin, 2011).
    3) LACOSAMIDE: ADULT DOSE: 200 to 400 mg IV; 200 mg IV over 15 minutes (Brophy et al, 2012). PEDIATRIC DOSE: In one study, median starting doses of 1.3 mg/kg/day and maintenance doses of 4.7 mg/kg/day were used in children 8 years and older (Loddenkemper & Goodkin, 2011).
    4) TOPIRAMATE: ADULT DOSE: 200 to 400 mg nasogastric/orally OR 300 to 1600 mg/day orally divided in 2 to 4 times daily (Brophy et al, 2012).
    H) ACUTE LUNG INJURY
    1) ONSET: Onset of acute lung injury after toxic exposure may be delayed up to 24 to 72 hours after exposure in some cases.
    2) NON-PHARMACOLOGIC TREATMENT: The treatment of acute lung injury is primarily supportive (Cataletto, 2012). Maintain adequate ventilation and oxygenation with frequent monitoring of arterial blood gases and/or pulse oximetry. If a high FIO2 is required to maintain adequate oxygenation, mechanical ventilation and positive-end-expiratory pressure (PEEP) may be required; ventilation with small tidal volumes (6 mL/kg) is preferred if ARDS develops (Haas, 2011; Stolbach & Hoffman, 2011).
    a) To minimize barotrauma and other complications, use the lowest amount of PEEP possible while maintaining adequate oxygenation. Use of smaller tidal volumes (6 mL/kg) and lower plateau pressures (30 cm water or less) has been associated with decreased mortality and more rapid weaning from mechanical ventilation in patients with ARDS (Brower et al, 2000). More treatment information may be obtained from ARDS Clinical Network website, NIH NHLBI ARDS Clinical Network Mechanical Ventilation Protocol Summary, http://www.ardsnet.org/node/77791 (NHLBI ARDS Network, 2008)
    3) FLUIDS: Crystalloid solutions must be administered judiciously. Pulmonary artery monitoring may help. In general the pulmonary artery wedge pressure should be kept relatively low while still maintaining adequate cardiac output, blood pressure and urine output (Stolbach & Hoffman, 2011).
    4) ANTIBIOTICS: Indicated only when there is evidence of infection (Artigas et al, 1998).
    5) EXPERIMENTAL THERAPY: Partial liquid ventilation has shown promise in preliminary studies (Kollef & Schuster, 1995).
    6) CALFACTANT: In a multicenter, randomized, blinded trial, endotracheal instillation of 2 doses of 80 mL/m(2) calfactant (35 mg/mL of phospholipid suspension in saline) in infants, children, and adolescents with acute lung injury resulted in acute improvement in oxygenation and lower mortality; however, no significant decrease in the course of respiratory failure measured by duration of ventilator therapy, intensive care unit, or hospital stay was noted. Adverse effects (transient hypoxia and hypotension) were more frequent in calfactant patients, but these effects were mild and did not require withdrawal from the study (Wilson et al, 2005).
    7) However, in a multicenter, randomized, controlled, and masked trial, endotracheal instillation of up to 3 doses of calfactant (30 mg) in adults only with acute lung injury/ARDS due to direct lung injury was not associated with improved oxygenation and longer term benefits compared to the placebo group. It was also associated with significant increases in hypoxia and hypotension (Willson et al, 2015).
    8) PARTIAL LIQUID VENTILATION
    a) CASE REPORT: A 27-year-old man, who ingested approximately 24 grams of sustained-release verapamil and subsequently developed hypotension, bradycardia, and respiratory distress requiring mechanical ventilation, was enrolled in a phase II clinical trial and was given partial liquid ventilation (PLV) with Perflubron(R), a fluorocarbon, administered intratracheally every 2 hours for 4 days. The patient's pulmonary function significantly improved within hours of PLV administration (Szekely et al, 1999).
    1) Theoretically, the dense fluorocarbon improves ventilation: perfusion by redistribution of blood to the anterior portions of the lungs, eases pulmonary toilet (aids in the removal of exudate), and reduces potential further lung injury secondary to lower ventilator settings.
    I) BEZOAR
    1) Following overdose of sustained release verapamil, concretions have occasionally been reported within the stomach or intestines. They are often not visible on plain x-ray films of the abdomen; gastrointestinal emptying methods to remove the concretions have not been proven. Endoscopy may be necessary following severe toxicity or prolonged symptoms (Prod Info CALAN(R) SR oral sustained release caplets, 2013).
    2) Tablet concretions ranging in size from golf to tennis ball size developing from sustained-release dosage forms have been found at autopsy. Gastroscopy may be required for confirmation if suspected since these masses have not been apparent on abdominal films (Sporer & Manning, 1993).
    3) As with other calcium channel blockers (eg, nifedipine), the formation of bezoars appear to be associated with acute overdose ingestion of extended-release tablets in a small number of cases. Treatment of the bezoar may be dependent on several factors, including the location, composition, and size of the bezoar, and patient symptoms. In the setting of acute overdose, endoscopic removal may be necessary. For symptomatic bezoars, surgery may be necessary if the bezoar cannot be removed endoscopically (Wells et al, 2006; Niezabitowski et al, 2000; Taylor et al, 1998).
    a) CASE REPORT: A 61-year-old woman developed a bezoar after intentionally ingesting 167 60-mg extended-release nifedipine tablets (total dose of approximately 10 grams). Despite removal of the bezoar via an esophagogastroduodenoscopy, the patient died from multisystem failure 3 days post-operatively (Wells et al, 2006).
    J) EXPERIMENTAL THERAPY
    1) 4-AMINOPYRIDINE
    a) SUMMARY: Preliminary investigation suggests that 4-aminopyridine (a potassium channel inhibitor) may antagonize the effects of calcium channel blockers by facilitating inward calcium movement (Pearigen & Benowitz, 1991).
    1) Additional clinical studies are needed to demonstrate the safety and efficacy of 4-aminopyridine for treatment of calcium antagonist overdose.
    2) HUMAN/CASE REPORTS
    a) Equivocal benefits were reported following 10 mg of 4-aminopyridine in a 67-year-old man poisoned with verapamil (ter Wee et al, 1985).
    b) A 47-year-old woman intentionally ingested 20 1-mg tablets of lorazepam and 40 5-mg tablets of amlodipine and presented to the emergency department 7 hours later with a blood pressure of 124/65 mmHg and a heart rate of 75 bpm. Despite activated charcoal administration, the patient's blood pressure and heart rate continued to decrease to 90/38 mmHg and 58 bpm, respectively. Following intravenous administration of 4-aminopyridine at a dosage regimen of 50 mcg/kg/hour over a 3-hour period, her blood pressure increased to 110/50 mmHg and her heart rate increased to 68 bpm (Wilffert et al, 2007).
    3) ANIMAL DATA
    a) The effectiveness of 4-aminopyridine (4-AP) and Bay K 8644 (calcium channel activator) were evaluated in verapamil-poisoned Wistar rats. Both agents were able to increase survival time with 70% of the animals surviving in the 4-AP-treated group and 50% surviving in the Bay K 8644-treated group. Average survival in the control groups was 20% for the calcium group and 40% for the adrenaline group, respectively.
    b) MECHANISM: 4-AP has an indirect effect on calcium channels and is able to block potassium K(1) channels on the cytoplasm side leading to depolarization and opening of voltage-dependent calcium channels resulting in improved blood pressure and heart rate. Bay K 644 evokes the release of calcium from endoplasmic reticulum and activation of the calcium ion influx to improve the contraction of the vascular smooth muscle and myocardium and electrical conduction in the heart.
    c) The authors concluded that despite its dose-dependent epileptogenic action (easily controlled in this study with antiepileptic therapy), 4-AP was the most effective therapy (Magdalan, 2003).
    d) 4-Aminopyridine reversed verapamil toxicity in artificially ventilated cats (Agoston et al, 1984).
    2) LEVOSIMENDAN
    a) CASE REPORTS: Two patients who overdosed on calcium antagonists and developed cardiovascular collapse, unresponsive to conventional therapies, improved and then recovered without sequelae following levosimendan infusions (Varpula et al, 2009).
    1) The first patient, a 47-year-old woman, became severely hypotensive and developed asystole, that required ventricular pacing, after ingesting 16 grams of verapamil. Despite calcium, glucagon, epinephrine boluses, norepinephrine, continuous vasopressin infusion, insulin and glucose infusion, intra aortic balloon pump, her mean arterial pressure (MAP) could not be maintained above 50 mmHg. Levosimendan was then initiated at an initial loading dose of 2 mcg/kg followed by an infusion of 0.2 mcg/kg/hour that was continued for 30 hours. Within 2 hours of initiating levosimendan, MAP could be maintained at 60 mmHg without epinephrine boluses, and she eventually recovered with normal neurologic function.
    2) The second patient, a 38-year-old man, became comatose and acidotic, with an unmeasurable mean arterial pressure (MAP), after ingesting 630 mg amlodipine, 300 mg of zopiclone, and an unknown amount of citalopram and acetaminophen. Despite conventional treatment with glucagon, calcium, vasopressors, and insulin and glucose, the patient's condition deteriorated, with persistent hypotension, worsening acidosis, right ventricular dilation, and a left ventricle ejection fraction (LEVF) of 38%. Levosimendan was then initiated at an infusion rate of 0.1 to 0.2 mcg/kg/min. Ninety minutes after initiation of therapy, the patient's LVEF improved to 72%. The patient gradually recovered and was discharged approximately 11 days post-ingestion.
    b) ANIMAL DATA: The efficacy of levosimendan, an inotropic agent, on cardiac output, blood pressure, and heart rate was assessed in rats following intoxication with verapamil, infused at 6 mg/kg/hr until mean arterial blood pressure decreased to 50% of baseline, then the infusion was reduced to 4 mg/kg/hr. There were 5 treatment groups evaluated: normal saline infusion (control), calcium chloride as a loading dose and infusion, levosimendan 24 mcg/kg loading dose and 0.6 mcg/kg/min infusion (Levo-24), levosimendan 6 mcg/kg loading dose and 0.4 mcg/kg/min infusion (Levo-6), and levosimendan 0.4 mcg/kg/min infusion with calcium chloride loading dose and infusion (Levo + CaCl2). The results showed that although the Levo-6 and Levo-24 groups showed statistically significant improvement in blood pressure compared to the control group (p <0.05) from t=20 minutes, the rats continued to be hypotensive with no improvement in blood pressure from that observed at t=0 minutes (peak verapamil toxicity prior to treatment administration). Cardiac output was significantly higher in all treatment groups as compared with the control group from t=20 minutes, except for Levo-6, which showed a significant improvement from t=30 minutes, and heart rate was maintained at pre-toxicity levels in all treatment groups. Based on the results of this study, the authors conclude that further investigation is warranted in order to consider levosimendan as an effective agent for the treatment of verapamil poisoning (Graudins et al, 2008).
    3) METHYLENE BLUE
    a) CASE REPORT: A 25-year-old woman presented to the emergency department 1 hour after intentionally ingesting 40 10-mg amlodipine tablets. Vital signs indicated normal blood pressure (120/86 mmHg), but elevated pulse rate (110 bpm), and an ECG revealed sinus tachycardia. Approximately 2 to 3 hours after receiving activated charcoal, the patient became hypotensive (75/40 mmHg) and her pulse rate increased to 120 bpm. Despite supportive treatment with calcium gluconate, glucagon, vasopressors, and high dose insulin therapy, the patient's condition progressively worsened, resulting in shock. Approximately 16 hours post-ingestion, methylene blue was administered at 2 mg/kg over 20 minutes, followed by 1 mg/kg/hour infusion. One hour after initiating therapy, the patient's blood pressure increased to 90/75 mmHg and her pulse rate decreased to 90 bpm. The patient was gradually weaned off all vasopressors and high-dose insulin therapy, and methylene blue therapy was discontinued. She was discharged 6 days later without sequelae (Jang et al, 2011).

Enhanced Elimination

    A) SUMMARY
    1) Hemodialysis is unlikely to be effective because of the high degree of protein binding and volume of distribution of these agents (Prod Info NYMALIZE(TM) oral solution, 2013; Prod Info nicardipine HCl intravenous injection solution, 2012; Prod Info Cleviprex(R) intravenous injection, 2011; Prod Info isradipine oral capsules, 2011; Prod Info SULAR(R) extended release oral tablets, 2010). Case reports suggest that hemoperfusion, or albumin dialysis with the molecular adsorbents recirculating system (MARS), both of which can remove protein bound drugs, might be effective in severe overdose, but definitive evidence is lacking.
    B) HEMOPERFUSION
    1) In general, the large volumes of distribution and high protein binding of all calcium channel blocking agents would suggest hemodialysis or hemoperfusion would have limited usefulness in removal of significant quantities of these drugs.
    2) Hemoperfusion for three hours 35 minutes resulted in a prominent decrease in both plasma diltiazem and metoprolol serum concentrations.
    a) The 49-year-old woman had ingested 1200 mg diltiazem, 500 mg metoprolol and an undetermined amount of ethanol.
    b) The patient improved clinically while on dialysis with less need for cardiac pacing and pressor agents (Anthony et al, 1986).
    3) Five hours of combined hemodialysis and charcoal hemoperfusion was associated with a decline in verapamil concentration from 687 to 192 nanograms/mL in a 48-year-old with occult liver disease and anuric acute renal failure (Rosansky, 1991).
    a) The hypotension and acidosis did not resolve and the patient died 8 hours after the completion of dialysis and hemoperfusion.
    C) PLASMAPHERESIS
    1) CASE REPORTS: Two patients developed severe hypotension and bradycardia requiring resuscitation and transvenous pacing after ingesting 2.4 and 9.6 g of verapamil, respectively. Initial blood concentrations were 5180 and 1856 ng/mL, respectively. Plasmapheresis was begun within 4 hours of ingestion. In both cases, blood concentrations decreased markedly (from 5180 ng/mL to 1272 ng/mL after 4 hours in the first patient and from 1856 ng/mL to 485 ng/mL after 7 hours in the second patient). In the first patient, cardiovascular improvement was noted during therapy, however, the patient died 38 hours after exposure of multiorgan failure; verapamil blood concentration was below 500 ng/mL. The second patient survived with no permanent sequelae (Kuhlmann et al, 1999).
    D) ALBUMIN DIALYSIS
    1) CASE REPORT: A 55-year-old woman with refractory shock after ingestion of sustained release diltiazem (8.4 grams) was treated with albumin dialysis using the molecular adsorbents recirculating system (MARS). During the procedure, blood lactate levels declined, and vasopressors were tapered and then discontinued 6 hour after albumin dialysis was initiated (Pichon et al, 2006).
    2) Albumin dialysis with Molecular Adsorbents Recirculating System (MARS) therapy was performed on 3 patients (a 55-year-old woman, a 13-year-old girl, and a 43-year-old man) who developed refractory cardiogenic shock and acute renal failure after ingesting 8.4 g sustained-release diltiazem, 4.2 g sustained-release diltiazem, and 14.4 g slow-release verapamil, respectively. Serum diltiazem concentrations in the 55-year-old and the 13-year-old were 2,658 mcg/L and 8,580 mcg/L, respectively, and the serum verapamil concentration was 2,200 mcg/L. Following dialysis, serum calcium antagonist concentrations decreased significantly in all three patients, with full recovery of myocardial function and normalization of renal function (Pichon et al, 2011).
    3) CASE REPORT: A 70-year-old woman ingested an unknown amount of sustained release diltiazem and subsequently developed lactic acidosis and vasoplegic shock refractory to intensive supportive therapy, including administration of calcium salts, fluids, vasopressors, insulin/dextrose infusion, and insertion of an intra-aortic balloon pump. Her plasma diltiazem concentration increased from 2209 ng/mL at hospital admission to 20,856 ng/mL 42 hours post-admission and Molecular Adsorbents Recirculating System (MARS) therapy was initiated. During the first 8 hours MARS session, vasopressor doses could be reduced and plasma diltiazem concentration decreased to 8285 ng/mL with a half life of 7.3 hours during MARS. Plasma diltiazem concentration increased to 17170, and a second 8 hour MARS treatment was performed, during which vasopressors were further decreased and diltiazem concentration decreased to 6404 ng/mL with a half life of 9.4 hours during MARS. Vasoactive drugs were discontinued on the 6th day and the patient made a complete recovery (Belleflamme et al, 2012).
    E) PLASMA EXCHANGE
    1) CASE REPORT: A 22-year-old woman intentionally ingested 425 mg of amlodipine besylate and presented to the hospital with nausea, vomiting, hypotension, tachycardia, and chest tightness. She subsequently developed third-degree heart block and absent P-waves. Despite treatment with glucagon, insulin-dextrose infusions, vasopressors and inotropic agents, the patient's condition continued to deteriorate, with the development of acute renal failure and metabolic acidosis. She was initially treated with charcoal hemoperfusion and continuous venovenous hemodialysis over the next 24 hours with minimal response. Therapeutic plasma exchange (TPE) was then instituted over the following 2 days. Her serum amlodipine concentration, prior to the procedure, was 150 ng/mL (normal 3 to 11 ng/mL). Two hours after completing the procedure, her serum amlodipine concentration decreased to 40 ng/mL. Her blood pressure and heart rate also normalized over the following two weeks after completing two sessions of TPE. Amlodipine has a large volume of distribution, rebound of serum concentrations would be expected after TPE, but was not evaluated in this patient, it is unlikely that TPE substantially affected outcome in this case (Ezidiegwu et al, 2008).

Summary

    A) TOXICITY: The toxic dose is variable depending on the particular agent. The following doses are considered to be potentially toxic (IR = immediate release, SR = sustained release): BEPRIDIL: ADULT: greater than 300 mg; CHILD: any amount. ISRADIPINE: ADULT: greater than 20 mg; CHILD: greater than 0.1 mg/kg. NICARDIPINE: ADULT: greater than 40 mg IR or chewed SR, or greater than 60 mg SR; CHILD: 1.25 mg/kg or more. NIMODIPINE: ADULT: greater than 60 mg; CHILD: any amount. NISOLDIPINE: ADULT: greater than 30 mg; CHILD: any amount. Single ingestions of therapeutic adult doses in children have resulted in death. Patients with underlying cardiovascular disease and the elderly tend to be more susceptible to the cardiac effects. In general, ingestions of phenylalkylamines (eg, verapamil) and benzothiazepines (eg, diltiazem) are more serious than ingestions of dihydropyridines (eg, niCARdipine).

Therapeutic Dose

    7.2.1) ADULT
    A) SPECIFIC SUBSTANCE
    1) CLEVIDIPINE: Initially, 1 to 2 mg/hour as an intravenous infusion, with a desired therapeutic response occurring at 4 to 6 mg/hour. Doses up to 32 mg/hour may be required for patients with severe hypertension. Due to lipid load restrictions, no more than 1000 mL or an average of 21 mg/hour over a 24-hour period is recommended (Prod Info Cleviprex(R) intravenous injection, 2011).
    2) ISRADIPINE: Recommended initial dose is 2.5 mg orally twice daily. May be titrated for response in increments of 5 mg/day at 2 to 4 week intervals, up to a maximum daily dose of 20 mg (Prod Info isradipine oral capsules, 2011).
    3) NICARDIPINE
    a) IMMEDIATE-RELEASE CAPSULES: Initially, 20 mg orally three times daily, titrated up to a maximum dose of 40 mg orally three times daily (Prod Info nicardipine HCl oral capsules, 2013).
    b) SUSTAINED-RELEASE CAPSULES: Initially, 30 mg orally twice daily; titrated up to a maximum of 60 mg twice daily (Prod Info CARDENE(R) SR oral sustained release capsules, 2010).
    c) INJECTION: For patients not receiving oral niCARdipine, initially 50 mL/hour (5 mg/hour) via slow continuous IV infusion; may be increased by 25 mL/hour (2.5 mg/hour) every 5 to 15 minutes up to a maximum of 150 mL/hour (15 mg/hour) (Prod Info nicardipine HCl intravenous injection solution, 2012)
    1) Once the blood pressure goal has been achieved via rapid titration, decrease the infusion rate to 30 mL/hour (3 mg/hour) (Prod Info nicardipine HCl intravenous injection solution, 2012).
    4) NIMODIPINE
    a) CAPSULES: The recommended dose is 60 mg orally every 4 hours for 21 consecutive days. Administer at least one hour before or 2 hours following a meal (Prod Info nimodipine oral capsules, 2012)
    1) Nimodipine is used to improve neurological outcome after subarachnoid hemorrhage. Nimodipine capsules should not be used by intravenous or other parenteral routes. Deaths and serious adverse events, including cardiac arrest, cardiovascular collapse, hypotension, and bradycardia have been reported following the intravenous administration of the contents of nimodipine capsules. If the patient can not swallow the capsule, the content of a capsule may be emptied by making a hole in both ends of a capsule with an 18-gauge needle. The contents should then be administered through the patient's naso-gastric tube and washed down the tube with 30 mL of normal saline solution (0.9%) (US Food and Drug Administration, 2006).
    b) ORAL SOLUTION: The recommended dose is 20 mL (60 mg) orally every 4 hours for 21 consecutive days. Administer one hour before or 2 hours following a meal (Prod Info NYMALIZE(TM) oral solution, 2013).
    5) NISOLDIPINE: Initially 17 mg orally once daily; may be increased in increments of 8.5 mg/week or longer, up to a maximum dose of 34 mg once daily (Prod Info SULAR(R) extended release oral tablets, 2010).
    7.2.2) PEDIATRIC
    A) SPECIFIC SUBSTANCE
    1) CLEVIDIPINE
    a) Safety and effectiveness have not been established in pediatric patients (Prod Info Cleviprex(R) intravenous injection, 2011).
    2) ISRADIPINE
    a) Rapid acting and useful for acute hypertension. Suspension may be compounded for infants and young children. Standard-release dosage form has shorter duration (6 to 8 hours) of effect in pediatric patients, requiring multiple daily dosing(Flynn & Pasko, 2000).
    b) Suggested initial dose in children is 0.05 to 0.15 mg/kg every 6 to 8 hours; maximum dose is 0.8 mg/kg/day up to 20 mg/day (Flynn & Pasko, 2000).
    3) NICARDIPINE
    a) HYPERTENSION, SEVERE ACUTE
    1) Initial dose: 0.5 to 1 mcg/kg/minute continuous IV infusion titrated every 15 to 30 minutes until goal blood pressure achieved (Sahney, 2006; Flynn et al, 2001; Tenney & Sakarcan, 2000; Treluyer et al, 1993). Initial doses of 5 mcg/kg/minute have been used to achieve rapid blood pressure control, then titrated down to maintain blood pressure control (Michael et al, 1998; Tobias et al, 1995).
    2) Titrate dose slowly in patients with heart failure or renal or hepatic impairment (Prod Info CARDENE(R) IV Premixed Injection (0.1 mg/mL) IV injection, 2010).
    3) Maintenance dose: Usual maintenance doses range between 0.5 to 4 mcg/kg/minute continuous IV infusion (Flynn & Tullus, 2009; Flynn et al, 2001; Tenney & Sakarcan, 2000; Tobias et al, 1996; Treluyer et al, 1993).
    4) Titrate dose to response. Blood pressure will begin to decrease within minutes of starting the infusion, reaching half of its ultimate decrease in approximately 45 minutes (adult data) (Prod Info CARDENE(R) IV Premixed Injection (0.1 mg/mL) IV injection, 2010). In studies in children, mean time to desired MAP was 5 to 10 minutes (Tobias et al, 1996; Tobias et al, 1995).
    4) NIMODIPINE
    a) ORAL SOLUTION: Safety and effectiveness have not been established in pediatric patients(Prod Info NYMALIZE(TM) oral solution, 2013).
    b) TABLETS: Safety and effectiveness have not been established in pediatric patients (Prod Info nimodipine oral capsules, 2012).
    5) NISOLDIPINE
    a) Safety and efficacy in pediatric patients have not been established (Prod Info SULAR(R) extended release oral tablets, 2010).

Minimum Lethal Exposure

    A) CASE REPORTS
    1) NICARDIPINE
    a) An 89-year-old man died after inadvertent intravenous administration of 40 mL niCARdipine hydrochloride (containing 60 mg niCARdipine) instead of 40 mL of 50% glucose (Ikegaya et al, 2002).

Maximum Tolerated Exposure

    A) GENERAL/SUMMARY
    1) Patients with the following inadvertent single substance ingestions are considered to have the potential to develop toxicity and should be referred to a healthcare facility (IR = immediate release, SR = sustained release) (Olson et al, 2005):
    a) BEPRIDIL: ADULT: greater than 300 mg; CHILD: any amount
    b) ISRADIPINE: ADULT: greater than 20 mg; CHILD: greater than 0.1 mg/kg
    c) NICARDIPINE: ADULT: greater than 40 mg IR or chewed SR, or greater than 60 mg SR; CHILD: 1.25 mg/kg or more
    d) NIMODIPINE: ADULT: greater than 60 mg; CHILD: any amount
    e) NISOLDIPINE: ADULT: greater than 30 mg; CHILD: any amount
    B) CASE REPORTS
    1) BARNIDIPINE
    a) Acute myocardial infarction occurred in a 25-year-old woman who intentionally ingested 500 mg of barnidipine. With supportive therapy, the patient recovered (Guvenc et al, 2010).
    2) BENIDIPINE
    a) CHILD: A 16-month-old child developed metabolic acidosis, tachycardia, Kussmaul respiration, and fever following an inadvertent ingestion of two 4-mg benidipine tablets (0.8 mg/kg). With supportive therapy, the patient recovered and was discharged approximately 2 days post-ingestion (Akbayram et al, 2012).
    3) BEPRIDIL
    a) Survival following a single dose of 1600 milligrams without significant symptomatology is noted by the manufacturer (Prod Info Vascor(R), bepridil HCl, 1994).
    4) CLEVIDIPINE
    a) During clinical trials, the maximum dose administered was 106 mg/hour or a total dose of 1153 mg (Prod Info Cleviprex(R) intravenous injection, 2011).
    5) ISRADIPINE
    a) ADULT
    1) The manufacturer reported lethargy and sinus tachycardia following overdose ingestions of 20 to 100 mg, and transient hypotension with the 100 mg dose. Reversible effects of flushing, tachycardia with ST depression, and hypotension were also reported following an overdose ingestion of 200 mg isradipine, with concomitant ethanol ingestion (Prod Info isradipine oral capsules, 2011).
    b) CHILD
    1) A 2-year-old boy survived a single 2.5 mg dose, although blood pressure dropped to 68/40 mmHg. Therapy included lavage and charcoal, with fluids to maintain blood pressure. No conduction defects were observed, and recovery was complete within 24 hours (Spiller & Ramoska, 1992).
    6) LERCANIDIPINE
    a) CASE REPORT: A 49-year-old man developed bradycardia and severe hypotension, refractory to IV fluids, calcium and glucagon, after intentionally ingesting 560 mg of slow-release lercanidipine. Following hyperinsulinemic euglycemia therapy and norepinephrine administration, the patient's blood pressure normalized and he was discharged approximately 2 days post-ingestion (Hadjipavlou et al, 2011).
    7) NICARDIPINE
    a) ADULT
    1) Two separate adult ingestions of 600 mg immediate-release niCARdipine and 2160 mg of sustained-release niCARdipine resulted in symptoms of hypotension, bradycardia, palpitations, flushing, drowsiness, confusion, and slurred speech. Both patients recovered without sequelae (Prod Info nicardipine HCl oral capsules, 2013; Prod Info CARDENE(R) SR oral sustained release capsules, 2010).
    2) Headache, flushing, and sinus tachycardia were reported in a 39-year-old woman who was 36 weeks pregnant and inadvertently received an overdose infusion of niCARdipine at 20 to 25 mg/hour over a 10-hour period, instead of the prescribed 2 mg/hour, to treat severe hypertension (Aya et al, 1996).
    b) CHILD
    1) A child who ingested half the contents of a 30 mg immediate-release capsule remained asymptomatic (Prod Info CARDENE(R) SR oral sustained release capsules, 2010).

Serum Plasma Blood Concentrations

    7.5.1) THERAPEUTIC CONCENTRATIONS
    A) THERAPEUTIC CONCENTRATION LEVELS
    1) SPECIFIC TOXIN
    a) BEPRIDIL
    1) THERAPEUTIC concentrations of approximately 1000 to 2000 nanograms/mL are listed by the manufacturer (Prod Info Vascor(R), bepridil HCl, 1994).
    b) NICARDIPINE
    1) Plasma/serum range considered therapeutic was stated to be 0.07 to 0.1 mcg/mL (Schulz & Schmoldt, 2003).
    2) Plasma concentrations following intravenous niCARdipine in doses of 5 and 10 mg administered over 10 minutes were 68 and 123 mcg/L, respectively (Rousseau et al, 1984).
    7.5.2) TOXIC CONCENTRATIONS
    A) TOXIC CONCENTRATION LEVELS
    1) SPECIFIC TOXIN
    a) ISRADIPINE
    1) CASE REPORT: A 5-year-old child, who was taking isradipine 2.5 mg twice daily for treatment of hypertension, presented with bradycardia and abdominal distention. Thirty minutes after hospital admission, the patient developed a ventricular escape rhythm (20 bpm), became apneic, then developed asystole. The patient responded to cardiac resuscitation and temporary pacing, with spontaneous cardiac activity returning approximately 12 hours post-admission (Romano et al, 2002). Serum isradipine concentration, obtained one hour post-admission, was 260.7 ng/mL. Twenty hours later, a second serum isradipine concentration was 27.4 ng/mL. The normal therapeutic concentration in adults receiving multiple daily doses is 2.2 to 8.21 ng/mL.

Toxicity Information

    7.7.1) TOXICITY VALUES
    A) ISRADIPINE
    1) LD50- (ORAL)MOUSE:
    a) 216 mg/kg (RTECS , 2000)
    2) LD50- (ORAL)RAT:
    a) >3 g/kg (RTECS , 2000)
    B) NICARDIPINE
    1) LD50- (ORAL)MOUSE:
    a) 305 mg/kg (RTECS , 2000)
    2) LD50- (ORAL)RAT:
    a) 320 mg/kg (RTECS , 2000)
    C) NIMODIPINE
    1) LD50- (ORAL)MOUSE:
    a) 940 mg/kg (RTECS , 2000)
    2) LD50- (ORAL)RAT:
    a) 2738 mg/kg (RTECS , 2000)
    D) NISOLDIPINE
    1) LD50- (ORAL)MOUSE:
    a) 411 mg/kg (RTECS , 2000)
    2) LD50- (ORAL)RAT:
    a) 1257 mg/kg (RTECS , 2000)

Pharmacologic Mechanism

    A) SUMMARY: Calcium antagonists selectively inhibit membrane transport of calcium during the slow inward excitation-contraction coupling phase in cardiac and vascular smooth muscle. Intracellular calcium ion outflow may also be speeded through stimulation of ATP dependent Ca and Na-K pumps.
    1) Negative inotropic (contractility) effects on the myocardium are usually compensated for by reflex sympathetic nervous system mechanisms.
    2) Antiarrhythmic effects are due to delayed antegrade conduction through the AV node, although direct SA node effects may also be involved.
    3) Dilation of coronary and peripheral arteries and arterioles decreases afterload and also prevents coronary arterial spasm; other protective cardiac effects unrelated to coronary blood flow have been proposed.
    4) Antiplatelet effects may also provide beneficial effects in ischemic heart disease (Ferrari & Visioli, 1991).
    B) CARDIOVASCULAR
    1) Calcium antagonists selectively inhibit membrane transport of calcium during the slow inward excitation-contraction coupling phase in smooth muscle leading to coronary and peripheral vasodilation. In general, they have a negative inotropic (contractility) effect on the myocardium (Singh et al, 1978) not usually manifested with therapeutic doses due to compensation of the sympathetic nervous system.
    2) Nimodipine is chemically similar to nifedipine but preferentially dilates brain vessels (Schluter, 1986).

Toxicologic Mechanism

    A) CARDIOVASCULAR EFFECTS
    1) Cardiovascular effects that occur in overdose, as related to the specific class of calcium channel blockers, are as follows (Verbrugge & vanWezel, 2007):
    a) Vasodilation - Dihydropyridines (eg, niCARdipine) > phenylalkalamines (eg, verapamil) > benzothiazepines (eg, diltiazem)
    b) Chronotropic suppression (SA node) - Phenylalkalamines = benzothiazepines >>>dihydropyridines
    c) Suppression of conduction (AV node) - Phenylalkalamines = benzothiazepines >>>dihydropyridines
    d) Inotropy (contractility) - Phenylalkalamines >>benzothiazepines>dihydropyridines
    B) HYPERGLYCEMIA
    1) Calcium channel antagonists, in general, decrease pancreatic insulin secretion and induce systemic insulin resistance, resulting in hyperglycemia (DeWitt & Waksman, 2004). An in vitro study showed that dysregulation of the phosphatidylinositol-3-kinase (PI3K) pathway, an insulin-dependent pathway, may contribute to the development of insulin resistance, resulting in hyperglycemia in the setting of calcium channel antagonist toxicity (Bechtel et al, 2008).
    C) METABOLIC ACIDOSIS
    1) Decreased insulin secretion, increased insulin resistance, and poor tissue perfusion and substrate delivery may be related to the occurrence of metabolic acidosis associated with calcium channel blocker (CCB) poisoning (DeWitt & Waksman, 2004). Another contributing factor may be the CCBs interference with glucose catabolism via inhibition of calcium-stimulated mitochondrial activity, thereby leading to lactate production and ATP hydrolysis.
    D) PSYCHOSIS
    1) Excessive dopaminergic influences may be responsible since calcium antagonism leads to stimulation of tyrosine hydroxylase activity in dopaminergic neurons (Kahn, 1986); however, confusion and disorientation due to hypoperfusion may be mistaken for psychosis.

Physical Characteristics

    A) CLEVIDIPINE: Milky-white formulated as an oil-in-water emulsion (Prod Info Cleviprex(R) intravenous injection, 2011).
    B) ISRADIPINE is a yellow, fine crystalline powder, which is odorless or has a faint characteristic odor, and is freely soluble in acetone, chloroform, and methylene chloride; soluble in ethanol; and practically insoluble in water (less than 10 mg/L at 37 degrees C) (Prod Info isradipine oral capsules, 2011).
    C) NICARDIPINE HYDROCHLORIDE is a greenish-yellow, odorless, crystalline powder that is freely soluble in chloroform, methanol, and glacial acetic acid; sparingly soluble in anhydrous ethanol; slightly soluble in n-butanol, water, 0.01 M potassium dihydrogen phosphate, acetone, and dioxane; very slightly soluble in ethyl acetate; and practically insoluble in benzene, ether, and hexane; and has a melting point of about 169 degrees C (Prod Info nicardipine HCl oral capsules, 2013).
    D) NIMODIPINE is a yellow crystalline substance that is practically insoluble in water (Prod Info nimodipine oral capsules, 2012).
    E) NISOLDIPINE: A yellow crystalline substance, soluble in ethanol but practically insoluble in water (Prod Info SULAR(R) extended release oral tablets, 2010).

Molecular Weight

    A) CLEVIDIPINE: 456.3 (Prod Info Cleviprex(R) intravenous injection, 2011)
    B) ISRADIPINE: 371.39 (Prod Info isradipine oral capsules, 2011)
    C) NICARDIPINE HYDROCHLORIDE: 515.99 (Prod Info nicardipine HCl oral capsules, 2013)
    D) NIMODIPINE: 418.5 (Prod Info nimodipine oral capsules, 2012)
    E) NISOLDIPINE: 388.4 (Prod Info SULAR(R) extended release oral tablets, 2010)

General Bibliography

    1) AMA Department of DrugsAMA Department of Drugs: AMA Evaluations Subscription, American Medical Association, Chicago, IL, 1992.
    2) Agarwal A, Yu SW, Rehman A, et al: Hyperinsulinemia euglycemia therapy for calcium channel blocker overdose: a case report. Tex Heart Inst J 2012; 39(4):575-578.
    3) Agoston S, Maestrone E, & van Hezik EJ: Effective treatment of verapamil intoxication with 4-aminopyridine in the cat. J Clin Invest 1984; 73:1291-1296.
    4) Akbayram S, Akgun C, Dogan M, et al: Benidipine hydrochloride intoxication in a child. J Emerg Med 2012; 42(6):700-701.
    5) 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.
    6) American College of Medical Toxicology: ACMT position statement: guidance for the use of intravenous lipid emulsion. J Med Toxicol 2016; Epub:Epub-.
    7) Anon: Warning labeling changes for new heart drug Posicor, US FDA Talk Paper, US Dept Health & Human Services, Rockville, MD, 1997.
    8) Anthony T, Jastremski M, & Elliot W: Charcoal hemoperfusion for the treatment of a combined diltiazem and metoprolol overdose. Ann Emerg Med 1986; 15:1344-1348.
    9) Artigas A, Bernard GR, Carlet J, et al: The American-European consensus conference on ARDS, part 2: ventilatory, pharmacologic, supportive therapy, study design strategies, and issues related to recovery and remodeling.. Am J Respir Crit Care Med 1998; 157:1332-1347.
    10) Aya AG, Bruelle P, Lefrant JY, et al: Accidental nicardipine overdosage without serious maternal or neonatal consequence. Anaesth Intensive Care 1996; 24(1):99-101.
    11) Aya AGM, Bruelle P, & Lefrant JY: Accidental nicardipine overdosage without serious maternal or neonatal consequence. Anaesth Intens Care 1996a; 24:99-101.
    12) Azendour H , Belyamani L , Atmani M , et al: Severe amlodipine intoxication treated by hyperinsulinemia euglycemia therapy. J Emerg Med 2010; 38(1):33-35.
    13) Bania TC, Blaufeux B, & Hughes S: Calcium and digoxin vs calcium alone for severe verapamil toxicity. Acad Emerg Med 2000; 7:1089-1096.
    14) Bania TC, Chu J, Perez E, et al: Hemodynamic effects of intravenous fat emulsion in an animal model of severe verapamil toxicity resuscitated with atropine, calcium, and saline. Acad Emerg Med 2007; 14(2):105-111.
    15) Barrow PM, Houston PL, & Wong DT: Overdose of sustained-release verapamil. Br J Anaesth 1994; 72:361-365.
    16) Barry JD, Durkovich D, Cantrell L, et al: Vasopressin treatment of verapamil toxicity in the porcine model. J Med Toxicol 2005; 1(1):3-10.
    17) Bechtel LK, Haverstick DM, & Holstege CP: Verapamil toxicity dysregulates the phosphatidylinositol 3-kinase pathway. Acad Emerg Med 2008; 15(4):368-374.
    18) Belfort M, Saade G, Moise KJ Jr, et al: Nimodipine in the management of preeclampsia: maternal and fetal effects. Am J Obstet Gynecol 1994; 171:417-24.
    19) Belleflamme M, Hantson P, Gougnard T, et al: Survival despite extremely high plasma diltiazem level in a case of acute poisoning treated by the molecular-adsorbent recirculating system. Eur J Emerg Med 2012; 19(1):59-61.
    20) Belson MG, Gorman SE, & Sullivan K: Calcium channel blocker ingestions in children. Am J Emerg Med 2000; 18:581-586.
    21) Bizovi K, Stork C, & Joyce D: Pacemaker use in critically ill calcium channel blocker overdoses (abstract). J Toxicol Clin Toxicol 1998; 36:509.
    22) Boyer EW, Duic PA, & Evans A: Hyperinsulinemia/euglycemia therapy for calcium channel blocker poisoning. Ped Emerg Care 2002; 18:36-37.
    23) Braun S, Boyko V, & Behar S: Calcium channel blocking agents and risk of cancer in patients with coronary heart disease. J Amer Coll Cardiol 1998; 31:804-808.
    24) Brayer AF & Wax P: Accidental ingestion of sustained release calcium channel blockers in children. Vet Hum Toxicol 1998; 40:104-106.
    25) Brophy GM, Bell R, Claassen J, et al: Guidelines for the evaluation and management of status epilepticus. Neurocrit Care 2012; 17(1):3-23.
    26) Brower RG, Matthay AM, & Morris A: Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. N Eng J Med 2000; 342:1301-1308.
    27) Buckley N, Dawson AH, & Howarth D: Slow-release verapamil poisoning: use of polyethylene glycol whole-bowel lavage and high-dose calcium. Med J Aust 1993; 158:202-204.
    28) Buckley NA, Whyte IM, & Dawson AH: Overdose with calcium channel blockers (letter). Br Med J 1994; 308:1639.
    29) Candell J, Valle V, & Soler M: Acute intoxication with verapamil. Chest 1979; 75:200-201.
    30) Cantrell FL, Clark RF, & Manoguerra AS: Determining triage guidelines for unintentional overdoses with calcium channel antagonists. Clin Toxicol (Phila) 2005; 43(7):849-853.
    31) Caravati EM, Knight HH, & Linscott MS: Esophageal laceration and charcoal mediastinum complicating gastric lavage. J Emerg Med 2001; 20:273-276.
    32) Carbonne B, Jannet D, Touboul C, et al: Nicardipine treatment of hypertension during pregnancy. Obstet Gynecol 1993; 81:908-914.
    33) Carcas AJ, Abad-Santos F, de Rosendo JM, et al: Nimodipine transfer into human breast milk and cerebrospinal fluid. Ann Pharmacother 1996; 30(2):148-150.
    34) Castro JrG, Tsai PWC & Ozaki MM et al: Nifedipine overdose. EAPCCT XVI Int Congress Vienna (Apr), 1994.
    35) Cataletto M: Respiratory Distress Syndrome, Acute(ARDS). In: Domino FJ, ed. The 5-Minute Clinical Consult 2012, 20th ed. Lippincott Williams & Wilkins, Philadelphia, PA, 2012.
    36) Cavagnaro F, Aglony M, & Rios JC: A suicide attempt with an oral calcium channel blocker. Vet Human Toxicol 2000; 42:99-100.
    37) 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.
    38) 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.
    39) Choonara IA & Rane A: Therapeutic drug monitoring of anticonvulsants state of the art. Clin Pharmacokinet 1990; 18:318-328.
    40) Chyka PA, Seger D, Krenzelok EP, et al: Position paper: Single-dose activated charcoal. Clin Toxicol (Phila) 2005; 43(2):61-87.
    41) Clark RF & Hanna TC: Calcium channel blocker toxicity. Top Emerg Med 1993; 15:15-26.
    42) Cohen HJ, Pieper CF, & Hanlon JT: Calcium channel blockers and cancer. Amer J Med 2000; 108:210-215.
    43) Cosbey SH & Carson DJL: A fatal case of amlodipine poisoning. J Analytical Toxicol 1997; 21:221-222.
    44) Cumpston KL , Aks SE , Sigg T , et al: Whole bowel irrigation and the hemodynamically unstable calcium channel blocker overdose: primum non nocere. J Emerg Med 2010; 38(2):171-174.
    45) De Cicco M, Macor F, & Robieux I: Pharmacokinetic and pharmacodynamic effects of high-dose continuous intravenous verapamil infusion: Clinical experience in the intensive care unit. Crit Care Med 1999; 27:332-339.
    46) DeRoos FJ: Calcium Channel Blockers. In: Nelson LS, Lewin NA, Howland MA, et al, eds. Goldfrank's Toxicological Emergencies, 9th ed. McGraw Hill Medical, New York, NY, 2011.
    47) DeWitt CR & Waksman JC: Pharmacology, pathophysiology and management of calcium channel blocker and beta-blocker toxicity. Toxicol Rev 2004; 23:223-238.
    48) Devasahayam J , Pillai U , & Uppaluri C : Acute severe intestinal obstruction secondary to amlodipine toxicity. Qjm 2012; 105(5):467-469.
    49) Dimich I, Profeta J, Jurado R, et al: Reversal of the adverse cardiovascular effects of intravenous diltiazem in anesthetized dogs. J Cardiothorac Anesth 1988; 2(4):455-462.
    50) Donovan JW, O'Donnell S, & Burkhart K: Calcium channel blocker overdose causing mesenteric ischemia (abstract). J Toxicol Clin Toxicol 1999; 37:628.
    51) Doyon S & Roberts JR: The use of glucagon in a case of calcium channel blocker overdose. Ann Emerg Med 1993; 22:1229-1233.
    52) Durward A, Guerguerian AM, Lefebvre M, et al: Massive diltiazem overdose treated with extracorporeal membrane oxygenation. Pediatr Crit Care 2003; 4(3):372-376.
    53) Elliot CG, Colby TV, & Kelly TM: Charcoal lung. Bronchiolitis obliterans after aspiration of activated charcoal. Chest 1989; 96:672-674.
    54) Engebretsen KM, Kaczmarek KM, Morgan J, et al: High-dose insulin therapy in beta-blocker and calcium channel-blocker poisoning. Clin Toxicol (Phila) 2011; 49(4):277-283.
    55) Erickson FC, Ling LJ, & Grande GA: Diltiazem overdose: case report and review. J Emerg Med 1991; 9:357-366.
    56) Evans JSM & Oram MP: Neurological recovery after prolonged verapamil-induced cardiac arrest. Anaes Intens Care 1999; 27:653-655.
    57) Ezidiegwu C, Spektor Z, Nasr MR, et al: A case report on the role of plasma exchange in the management of a massive amlodipine besylate intoxication. Ther Apher Dial 2008; 12(2):180-184.
    58) FDA: Poison treatment drug product for over-the-counter human use; tentative final monograph. FDA: Fed Register 1985; 50:2244-2262.
    59) Fant JS, James LP, & Fiser RT: The use of glucagon in nifedipine poisoning complicated by clonidine ingestion. Ped Emerg Care 1997; 13:417-419.
    60) Fauville JP, Hantson P, & Honore P: Severe diltiazem poisoning with intestinal pseudo-obstruction: case report and toxicological data. Clin Toxicol 1995; 33:273-277.
    61) Ferner RE, Monkman S, & Riley J: Pharmacokinetics and toxic effects of nifedipine in massive overdose. Hum Exp Toxicol 1990; 9:309-311.
    62) Ferrari R & Visioli O: Calcium channel blockers and ischemic heart disease: theoretical expectations and clinical experience. Eur Heart J 1991; 12:18-24.
    63) Flynn JT & Pasko DA: Calcium channel blockers: pharmacology and place in therapy of pediatric hypertension. Pediatr Nephrol 2000; 15:302-316.
    64) Flynn JT & Tullus K: Severe hypertension in children and adolescents: pathophysiology and treatment. Pediatr Nephrol 2009; 24(6):1101-1112.
    65) Flynn JT, Mottes TA, Brophy PD, et al: Intravenous nicardipine for treatment of severe hypertension in children. J Pediatr 2001; 139(1):38-43.
    66) Franxman TJ, Al-Nabhan M, Cavallazzi RS, et al: Lipid emulsion therapy for verapamil overdose. Ann Intern Med 2011; 154(4):292.
    67) Frierson J, Bailly D, & Shultz T: Refractory cardiogenic shock and complete heart block after suspected verapamil-SR and atenolol overdose. Clin Cardiol 1991; 14:933-935.
    68) Gerloni R & Copetti R: Easily reversible hypoxemia and hypotension induced by nimodipine. Eur J Emerg Med 2004; 11(5):295-297.
    69) Ghosh S & Sircar M: Calcium channel blocker overdose: Experience with amlodipine. Indian J Crit Care Med 2008; 12(4):190-193.
    70) Gokel Y, Paydas S, & Duru M: High-dose verapamil-trandolapril induced rhabdomyolysis and acute renal failure (letter). Am J Emerg Med 2000; 18:738-739.
    71) Golej J, Boigner H, Burda G, et al: Severe respiratory failure following charcoal application in a toddler. Resuscitation 2001; 49:315-318.
    72) Graff GR, Stark J, & Berkenbosch JW: Chronic lung disease after activated charcoal aspiration. Pediatrics 2002; 109:959-961.
    73) Graudins A, Najafi J, & Rur-SC MP: Treatment of experimental verapamil poisoning with levosimendan utilizing a rodent model of drug toxicity. Clin Toxicol (Phila) 2008; 46(1):50-56.
    74) Greene SL, Gawarammana I, Wood DM, et al: Relative safety of hyperinsulinaemia/euglycaemia therapy in the management of calcium channel blocker overdose: a prospective observational study. Intensive Care Med 2007; 33(11):2019-2024.
    75) Guvenc TS , Gurkan U , Guzelburc O , et al: Barnidipine intoxication causing acute myocardial infarction. Am J Emerg Med 2010; 28(4):541-543.
    76) Haas CF: Mechanical ventilation with lung protective strategies: what works?. Crit Care Clin 2011; 27(3):469-486.
    77) Haddad LM: Resuscitation after nifedipine overdose exclusively with intravenous calcium chloride. Amer J Emerg Med 1996; 14:602-603.
    78) Hadjipavlou G, Hafeez A, Messer B, et al: Management of lercanidipine overdose with hyperinsulinaemic euglycaemia therapy: case report. Scand J Trauma Resusc Emerg Med 2011; 19(1):8.
    79) Hantsch C, Seger D, & Meredith T: Calcium channel antagonist toxicity and hypercalcemia (abstract). J Toxicol Clin Toxicol 1997; 35:495-496.
    80) Harris CR & Filandrinos D: Accidental administration of activated charcoal into the lung: aspiration by proxy. Ann Emerg Med 1993; 22:1470-1473.
    81) Haynes RC, Murad F, & Gilman AG: The Pharmacological Basis of Therapeutics, Macmillan Publishing Co, New York, NY, 1985, pp 1521.
    82) Hegenbarth MA & American Academy of Pediatrics Committee on Drugs: Preparing for pediatric emergencies: drugs to consider. Pediatrics 2008; 121(2):433-443.
    83) Henderson A, Stevenson N, & Hackett LP: Diltiazem overdose in an elderly patient: efficacy of adrenaline. Anesth Intens Care 1992; 20:507-510.
    84) Hendren WG, Schieber RS, & Garrettson LK: Extracorporeal bypass for the treatment of verapamil poisoning. Ann Emerg Med 1989; 18:984-987.
    85) Henrikson CA & Chandra-Strobos N: Calcium channel blocker overdose mimicking an acute myocardial infarction. Resuscitation 2003; 59:361-364.
    86) Herbert JX, O'Malley C, & Tracey JA: Verapamil overdosage unresponsive to dextrose/insulin therapy (abstract). J Toxicol Clin Toxicol 2001; 39:293-294.
    87) Herrington DM, Insley BM, & Weinman GG: Nifedipine overdose. Am J Med 1986; 81:344-346.
    88) Hofer CA, Smith JK, & Tenholder MF: Verapamil intoxication: a literature review of overdoses and discussion of therapeutic options. Am J Med 1993; 95:431-438.
    89) Holzer M, Sterz F, & Schoerkhuber W: Successful resuscitation of a verapamil-intoxicated patient with percutaneous cardiopulmonary bypass. Crit Care Med 1999; 27:2818-2823.
    90) Howarth DM, Dawson AH, & Smith AJ: Calcium channel blocking drug overdose: am Australian series. Human Exp Toxicol 1994; 13:161-166.
    91) Humbert VH Jr, Munn NJ, & Hawkins RF: Noncardiogenic pulmonary edema complicating massive diltiazem overdose. Chest 1991; 99:258-260.
    92) Hung YM & Olson KR: Acute amlodipine overdose treated by high dose intravenous calcium in a patient with severe renal insufficiency. Clin Toxicol (Phila) 2007; 45(3):301-303.
    93) Hvidberg EF & Dam M: Clinical pharmacokinetics of anticonvulsants. Clin Pharmacokinet 1976; 1:161.
    94) Ikegaya H, Kobayashi M, Sakurada K, et al: Detection of the calcium antagonist nicardipine and its metabolites by gas chromatography-mass spectrometry. Forensic Sci Int 2002; 130(1):25-28.
    95) Isbister GK: Delayed asystolic cardiac arrest after diltiazem overdose; resuscitation with high dose intravenous calcium. Emerg Med J 2002; 19(4):355-357.
    96) Jaeger A, Sauder P, & Kopferschmitt J: Diltiazem acute poisoning: hemodynamic and kinetic study (abstract 191). Vet Hum Toxicol 1989; 31:377.
    97) Jang DH, Nelson LS, & Hoffman RS: Methylene Blue in the Treatment of Refractory Shock From an Amlodipine Overdose. Ann Emerg Med 2011; Epub:Epub-.
    98) Jick H, Jick S, & Derby LE: Calcium-channel blockers and risk of cancer. Lancet 1997; 349:525-528.
    99) Kahn JK: Nifedipine-associated acute psychosis. Am J Med 1986; 81:705-706.
    100) Kamijo Y, Yoshida T, Ide A, et al: Mixed venous oxygen saturation monitoring in calcium channel blocker poisoning: tissue hypoxia avoidance despite hypotension. Am J Emerg Med 2006; 24(3):357-360.
    101) Kanagarajan K, Marraffa JM, Bouchard NC, et al: The use of vasopressin in the setting of recalcitrant hypotension due to calcium channel blocker overdose. Clin Toxicol (Phila) 2007; 45(1):56-59.
    102) Kanamasa K, Kimura A, & Miyataka M: Incidence of cancer in postmyocardial infarction patients treated with short-acting nifedipine and diltiazem. Cancer 1999; 85:1369-1374.
    103) Kaplan RC, Heckbert SR, Koepsell TD, et al: Use of calcium channel blockers and risk of hospitalized gastrointestinal tract bleeding.. Arch Intern Med 2000; 160:1849-1855.
    104) Kirshenbaum LA, Mathews SC, & Sitar DS: Whole-bowel irrigation versus activated charcoal in sorbitol for the ingestion of modified-release pharmaceuticals. Clin Pharmacol Ther 1989; 46:264-271.
    105) 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.
    106) Kline JA, Leonova E, & Raymond RM: Beneficial myocardial metabolic effects of insulin during verapamil toxicity in the anesthetized canine. Crit Care Med 1995; 23:1251-1263.
    107) Kline JA, Raymond RM, & Leonova ED: Insulin improves heart function and metabolism during non-ischemic cardiogenic shock in awake canines. Cardiovasc Res 1997; 289-298.
    108) Kline JA, Tomaszewski CA, & Schroeder JD: Insulin is a superior antidote for cardiovascular toxicity induced by verapamil in the anesthetized canine. J Pharmacol Exp Ther 1993; 267:744-750.
    109) Kollef MH & Schuster DP: The acute respiratory distress syndrome. N Engl J Med 1995; 332:27-37.
    110) Koppel C, Fahron G & Kruger A et al: Prolonged resuscitation efforts in serious metoprolol and verapamil overdose. EAPCCT XVII Int Congress Poland, 1995.
    111) Krenzelok EP: Acute calcium channel blocker overdosage. Clin Tox Forum 1991; 3:1-6.
    112) Krick SE, Gums JG, & Grauer K: Severe verapamil (sustained-release) overdose. DICP Ann Pharmacother 1990; 24:705-706.
    113) Kuhlmann U, Schoenemann H, Muller TF, et al: Plasmapheresis in fatal overdose with verapamil. Intensive Care Med 1999; 25(12):1473-1473.
    114) Laine K, Kivisto KT, & Neuvonen PJ: Effect of delayed administration of activated charcoal on the absorption of conventional and slow-release verapamil. Clin Toxicol 1997a; 35:263-268.
    115) Laine K, Kivisto KT, & Neuvonen PJ: Effect of delayed administration of activated charcoal on the absorption of conventional and slow-release verapamil. J Toxicol Clin Toxicol 1997; 35:263-268.
    116) Lam YM , Tse HF , & Lau CP : Continuous calcium chloride infusion for massive nifedipine overdose. Chest 2001; 119(4):1280-1282.
    117) Lapatto-Reiniluoto O, Kivisto KT, & Neuvonen PJ: Efficacy of activated charcoal versus gastric lavage half an hour after ingestion of moclobemide, temazepam, and verapamil. Eur J Clin Pharmacol 2000; 56:285-288.
    118) 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.
    119) Lawless MJ, Mofenson HC, & Caraccio TR: Delayed toxicity in sustained-release nifedipine overdose despite whole bowel irrigation (WBI) (abstract). J Toxicol Clin Toxicol 1997; 35:498.
    120) Lee DC, Greene T, & Dougherty T: Fatal nifedipine ingestions in children. J Emerg Med 2000; 19:359-361.
    121) Leesar MA, Martyn R, & Talley JD: Noncardiogenic pulmonary edema complicating massive verapamil overdose. Chest 1994; 105:606-607.
    122) Leone M, Charvet A, Delmas A, et al: Terlipressin: a new therapeutic for calcium-channel blockers overdose (letter). J Crit Care 2005; 20:114-115.
    123) Levine M, Boyer EW, Pozner CN, et al: Assessment of hyperglycemia after calcium channel blocker overdoses involving diltiazem or verapamil. Crit Care Med 2007; 35(9):2071-2075.
    124) Levine M, Curry SC, Padilla-Jones A, et al: Critical Care Management ofVerapamil and Diltiazem Overdose With a Focus on Vasopressors: A 25-Year Experience at a Single Center. Ann Emerg Med 2013; Epub:Epub-.
    125) Lheureux PE, Zahir S, Gris M, et al: Bench-to-bedside review: Hyperinsulinaemia/euglycaemia therapy in the management of overdose of calcium-channel blockers. Crit Care (Lond) 2006; 10(3):212-.
    126) Liang CW, Diamond SJ, & Hagg DS: Lipid rescue of massive verapamil overdose: a case report. J Med Case Rep 2011; 5:399-399.
    127) Lieberman P, Nicklas RA, Oppenheimer J, et al: The diagnosis and management of anaphylaxis practice parameter: 2010 update. J Allergy Clin Immunol 2010; 126(3):477-480.
    128) Link MS, Berkow LC, Kudenchuk PJ, et al: Part 7: Adult Advanced Cardiovascular Life Support: 2015 American Heart Association Guidelines Update for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2015; 132(18 Suppl 2):S444-S464.
    129) Loddenkemper T & Goodkin HP: Treatment of Pediatric Status Epilepticus. Curr Treat Options Neurol 2011; Epub:Epub.
    130) Louagie HK, Vandewoude KM, Vogelaers DP, et al: The measurement of nisoldipine serum concentration in a case of severe nisoldipine overdose. J Toxicol Clin Toxicol 1998; 36(1-2):125-127.
    131) Luscher TF, Noll G, & Sturmer T: Calcium gluconate in severe verapamil intoxication (letter). N Eng J Med 1994; 330:718-720.
    132) MacDonald D & Alguire PC: Case report: fatal overdose with sustained-release verapamil. Am J Med Sci 1992; 303:115-117.
    133) Magdalan J: New treatment methods in verapamil poisoning: experimental studies. Polish J Pharmacol 2003; 55:425-432.
    134) Mahr NC, Valdes A, & Lamas G: Use of glucagon for acute intravenous diltiazem toxicity. Amer J Cardiol 1997; 79:1570-1571.
    135) Malcolm N, Callegari P, & Goldberg J: Massive diltiazem overdosage: clinical and pharmacokinetic observations. Drug Intell Clin Pharm 1986; 20:888.
    136) Manno EM: New management strategies in the treatment of status epilepticus. Mayo Clin Proc 2003; 78(4):508-518.
    137) McMillan R: Management of acute severe verapamil intoxication. J Emerg Med 1988; 6:193-196.
    138) Melanson P, Shir RD, & DeRoos F: Intra-aortic balloon counterpulsation in calcium channel blocker overdose (abstract). Vet Hum Toxicol 1993; 35:345.
    139) Meyer M, Stremski E, & Scanlon M: Verapamil-induced hypotension reversed with dextrose-insulin (abstract). J Toxicol Clin Toxicol 2001; 39:500.
    140) Michael J, Groshong T, & Tobias JD: Nicardipine for hypertensive emergencies in children with renal disease. Pediatr Nephrol 1998; 12(1):40-42.
    141) Mitchell LB, Schroeder JS, & Mason JW: Comparative clinical electrophysiologic effects of diltiazem, verapamil and nifedipine: a review. Am J Cardiol 1982; 49:629-635.
    142) Morimoto S, Sasaki S, & Kiyama M: Sustained-release diltiazem overdose. J Hum Hypertension 1999; 13:643-644.
    143) Moroni F, Mannaioni PF, & Dolara A: Calcium gluconate and hypertonic sodium chloride in a case of massive verapamil poisoning. Clin Toxicol 1980; 17:395-400.
    144) Morris DL & Goldschlager N: Calcium infusion for reversal of adverse effects of intravenous verapamil. JAMA 1983; 249:3212-3213.
    145) Mullins ME, Horowitz BZ, & Linden DH: Life-threatening interaction of mibefradil and Beta-blockers with dihydropyridine caclim channel blockers. JAMA 1998; 280:157-158.
    146) NHLBI ARDS Network: Mechanical ventilation protocol summary. Massachusetts General Hospital. Boston, MA. 2008. Available from URL: http://www.ardsnet.org/system/files/6mlcardsmall_2008update_final_JULY2008.pdf. As accessed 2013-08-07.
    147) 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.
    148) Niezabitowski LM, Nguyen BN, & Gums JG: Extended-release nifedipine bezoar identified one year after discontinuation. Ann Pharmacother 2000; 34:862-864.
    149) None Listed: Position paper: cathartics. J Toxicol Clin Toxicol 2004; 42(3):243-253.
    150) Nordlander M, Sjoquist PO, Ericsson H, et al: Pharmacodynamic, pharmacokinetic and clinical effects of clevidipine, an ultrashort-acting calcium antagonist for rapid blood pressure control. Cardiovasc Drug Rev 2004; 22(3):227-250.
    151) Olson KR, Erdman AR, Woolf AD, et al: Calcium channel blocker ingestion: an evidence-based consensus guideline for out-of-hospital management. Clin Toxicol (Phila) 2005; 43(7):797-822.
    152) Ori Y, Korzets A, & Caneti M: Lymphocytic intracellular calcium in a patient with complicated verapamil overdose. Amer J Med Sci 2000; 319:63-67.
    153) Paetow GM, Austad EJ, Olives TD, et al: Use of Tissue Perfusion Monitoring to Titrate High Dose Insulin in Beta Blocker and Calcium Channel Blocker Overdose. Clin Toxicol (Phila) 2015; 53(7):705-706.
    154) Papadopoulos J & O'Neil MG: Utilization of a glucagon infusion in the management of a massive nifedipine overdose. J Emerg Med 2000; 18:453-455.
    155) Passal DB & Crespin FH: Verapamil poisoning in an infant. Pediatrics 1984; 73:543-545.
    156) Pearigen PD & Benowitz NL: Poisoning due to calcium antagonists. Experience with verapamil, diltiazem and nifedipine. Drug Safety 1991; 6:408-430.
    157) 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.
    158) Perez E, Chu J, Bania T, et al: L-carnitine increases survival in a murine model of severe verapamil toxicity. Acad Emerg Med 2011; 18(11):1135-1140.
    159) Perkins CM: Serious verapamil poisoning. Treatment with intravenous calcium gluconate. Br Med J 1978; 21:1127.
    160) Pichon N, Dugard A, Clavel M, et al: Extracorporeal Albumin Dialysis in Three Cases of Acute Calcium Channel Blocker Poisoning With Life-Threatening Refractory Cardiogenic Shock. Ann Emerg Med 2011; Epub:Epub.
    161) Pichon N, Francois B, Chevreuil C, et al: Albumin dialysis: a new therapeutic alternative for severe diltiazem intoxication. Clin Toxicol (Phila) 2006; 44(2):195-196.
    162) Pollack MM, Dunbar BS, & Holbrook PR: Aspiration of activated charcoal and gastric contents. Ann Emerg Med 1981; 10:528-529.
    163) Product Information: CALAN(R) SR oral sustained release caplets, verapamil HCl oral sustained release caplets. G.D. Searle (per FDA), New York, NY, 2013.
    164) Product Information: CARDENE(R) IV Premixed Injection (0.1 mg/mL) IV injection, nicardipine HCL premixed injection (0.1 mg/mL) IV injection. EKR Therapeutics, Inc, Bedminster, NJ, 2010.
    165) Product Information: CARDENE(R) SR oral sustained release capsules, nicardipine HCl oral sustained release capsules. EKR Therapeutics, Inc. (per DailyMed), Bedminster, NJ, 2010.
    166) Product Information: Cleviprex(R) intravenous injection, clevidipine intravenous injection. The Medicines Company (per DailyMed), Parsippany, NJ, 2011.
    167) Product Information: Isuprel(TM) intravenous injection, intramuscular injection, subcutaneous injection, intracardiac injection, isoproterenol HCl intravenous injection, intramuscular injection, subcutaneous injection, intracardiac injection. Hospira, Inc. (per FDA), Lake Forest, IL, 2013.
    168) Product Information: NYMALIZE(TM) oral solution, nimodipine oral solution. Arbor Pharmaceuticals, Inc. (per FDA), Atlanta, GA, 2013.
    169) Product Information: SULAR(R) extended release oral tablets, nisoldipine extended release oral tablets. Shionogi Pharma, Inc, Atlanta, GA, 2010.
    170) Product Information: Vascor(R), bepridil HCl. McNeil Pharmaceuticals, Springhouse, PA, 1994.
    171) Product Information: diazepam IM, IV injection, diazepam IM, IV injection. Hospira, Inc (per Manufacturer), Lake Forest, IL, 2008.
    172) Product Information: dobutamine HCl 5% dextrose intravenous injection, dobutamine HCl 5% dextrose intravenous injection. Baxter Healthcare Corporation (per DailyMed), Deerfield, IL, 2012.
    173) Product Information: dopamine hcl, 5% dextrose IV injection, dopamine hcl, 5% dextrose IV injection. Hospira,Inc, Lake Forest, IL, 2004.
    174) Product Information: inamrinone intravenous solution, inamrinone intravenous solution. Bedford Laboratories, Bedford, OH, 2002.
    175) Product Information: isradipine oral capsules, isradipine oral capsules. Watson Pharma, Inc. (per manufacturer), Corona, CA, 2011.
    176) Product Information: lorazepam IM, IV injection, lorazepam IM, IV injection. Akorn, Inc, Lake Forest, IL, 2008.
    177) Product Information: nicardipine HCl intravenous injection solution, nicardipine HCl intravenous injection solution. American Regent, Inc. (per DailyMed), Shirley, NY, 2012.
    178) Product Information: nicardipine HCl oral capsules, nicardipine HCl oral capsules. Mylan Pharmaceuticals Inc. (per DailyMed), Morgantown, WV, 2013.
    179) Product Information: nimodipine oral capsules, nimodipine oral capsules. Caraco Pharmaceutical Laboratories, Ltd. (per DailyMed), Detroit, MI, 2012.
    180) Product Information: norepinephrine bitartrate injection, norepinephrine bitartrate injection. Sicor Pharmaceuticals,Inc, Irvine, CA, 2005.
    181) Product Information: phenylephrine HCl subcutaneous injection, intramuscular injection, intravenous injection, phenylephrine HCl subcutaneous injection, intramuscular injection, intravenous injection. West-Ward Pharmaceuticals (per DailyMed), Eatontown, NJ, 2011.
    182) Quezado Z, Lippmann M, & Wertheimer J: Severe cardiac, respiratory, and metabolic complications of massive verapamil overdose. Crit Care Med 1991; 19:436-438.
    183) RTECS : Registry of Toxic Effects of Chemical Substances. National Institute for Occupational Safety and Health. Cincinnati, OH (Internet Version). Edition expires 2000; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    184) Ramoska EA, Spiller HA, & Winter M: A one-year evaluation of calcium channel blocker overdoses: toxicity and treatment. Ann Emerg Med 1993; 22:196-200.
    185) Rankin RJ & Edwards IR: Overdose of sustained release verapamil (letter). NZ Med J 1990; 103:165.
    186) Ranniger C & Roche C: Are one or two dangerous? Calcium channel blocker exposure in toddlers. J Emerg Med 2007; 33(2):145-154.
    187) Rau NR, Nagaraj MV, Prakash PS, et al: Fatal pulmonary aspiration of oral activated charcoal. Br Med J 1988; 297:918-919.
    188) Roberts D, Honcharik N, & Sitar DS: Diltiazem overdose: pharmacokinetics of diltiazem and its metabolites and effect of multiple dose charcoal therapy. Clin Toxicol 1991; 29:45-52.
    189) Robinson HP, Jacoby KJ, Prekker ME, et al: Combined therapy with high-dose insulin and vasoactive drugs for beta blocker/calcium channel blocker overdose: characteristics, complications and outcomes (abstracts). Clin Toxicol (Phila) 2015; 53(7):651.
    190) Rodgers GC, Al-Mahasneh QM, & White SL: Treatment of severe sustained release verapamil poisoning with cardiac pacing: a case report (abstract 190). Vet Hum Toxicol 1989; 31:377.
    191) Rogers IR & Prpic R: Profound symptomatic bradycardia associated with combined mibefradil and beta-blocker therapy. MJA 1998; 169:425-427.
    192) Romano MJ, Gaylor A, & Sang CJ: Life-threatening isradipine poisoning in a child. Pharmacotherapy 2002; 22(6):766-770.
    193) Rona R, Cortinovis B, Marcolin R, et al: Extra-corporeal life support for near-fatal multi-drug intoxication: a case report. J Med Case Reports 2011; 5:231.
    194) Rosansky SJ: Verapamil toxicity-treatment with hemoperfusion (letter). Ann Intern Med 1991; 114:340-341.
    195) Rousseau MF, Etienne J, Van Mechelen H, et al: Hemodynamic and cardiac effects of nicardipine in patients with coronary artery disease. J Cardiovasc Pharmacol 1984; 6:833-839.
    196) Sahney S: A review of calcium channel antagonists in the treatment of pediatric hypertension. Paediatr Drugs 2006; 8(6):357-373.
    197) SamiKarti S, Ulusoy H, Yandi M, et al: Non-cardiogenic pulmonary oedema in the course of verapamil intoxication. Emerg Med J 2002; 19(5):458-459.
    198) Samniah N & Schlaeffer F: Cerebral infarction associated with oral verapamil overdose. Clin Toxicol 1988; 26:365-369.
    199) Sanders P, Walker J, & Craig RJ: Mibefradil (Posicor) induced sinus arrest. Aust NZ J Med 1998; 28:836-837.
    200) Sandroni C, Cavallaro F, Addario C, et al: Successful treatment with enoximone for severe poisoning with atenolol and verapamil: a case report. Acta Anaesthesiologica Scandinavica 2004; 48:790-792.
    201) Saravu K & Balasubramanian R: Near-fatal amlodipine poisoning. J Assoc Phys India 2004; 52:156-157.
    202) Schluter G: Toxicological investigations with nimodipine: summary of relevant studies. Arzneimittelforsch 1986; 36:1733-1735.
    203) Schulz M & Schmoldt A: Therapeutic and toxic blood concentrations of more than 800 drugs and other xenobiotics. Pharmazie 2003; 58(7):447-474.
    204) Schwab M, Oetzel C, & Morike K: Using trade names. A risk factor for accidental drug overdose. Arch Intern Med 2002; 162:1065-1066.
    205) 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.
    206) Seki H, Takeda S, Kinoshita K, et al: Long-term treatment with nicardipine for severe eclampsia. Int J Gynecol Obstet 2002; 76:135-141.
    207) Shah AR & Passalacqua BR: Case report: sustained-release verapamil overdose causing stroke: an unusual complication. Am J Med Sci 1992; 304:357-359.
    208) Shepherd G & Klein-Schwartz W: High-dose insulin therapy for calcium-channel blocker overdose. Ann Pharmacother 2005; 39:923-930.
    209) Sim MT & Stevenson FT: A fatal case of iatrogenic hypercalcemia after calcium channel blocker overdose. J Med Toxicol 2008; 4(1):25-29.
    210) Singh BN, Ellrodt G, & Peter CT: Verapamil: A review of its pharmacological properties and therapeutic use. Drugs 1978; 15:169-197.
    211) Smith SW, Ling LJ, & Halstenson CE: Whole-bowel irrigation as a treatment for acute lithium overdose. Ann Emerg Med 1991; 20:536-539.
    212) Snook CP, Sigvaldason K, & Kristinsson J: Severe atenolol and diltiazem overdose. Clin Toxicol 2000; 38:661-665.
    213) Sorensen HT, Olsen JH, & Mellemkjaer L: Cancer risk and mortality in users of calcium channel blockers. Cancer 2000; 89:165-170.
    214) Sorrentino A: Update on pediatric resuscitation drugs: high dose, low dose, or no dose at all. Curr Opin Pediatr 2005; 17(2):223-226.
    215) Spiller HA & Ramoska EA: Isradipine ingestion in a 2 year old (abstract). Vet Hum Toxicol 1992; 34:329.
    216) Spiller HA, Meyers A, & Ziemba T: Delayed onset of cardiac arrhythmias from sustained-release verapamil. Ann Emerg Med 1991; 20:201-203.
    217) Spiller HA, Milliner BA, & Bosse GM: Amlodipine fatality in an infant with postmortem blood levels. J Med Toxicol 2012; 8(2):179-182.
    218) Sporer KA & Manning JJ: Massive ingestion of sustained-release verapamil with a concretion and bowel infarction. Ann Emerg Med 1993; 22:603-605.
    219) Spurlock BW, Virani NA, & Henry CA: Verapamil overdose. West J Med 1991; 154:208-211.
    220) 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.
    221) St-Onge M, Ajmo I, Poirier D, et al: L-Carnitine for the Treatment of a Calcium Channel Blocker and Metformin Poisoning. J Med Toxicol 2013; Epub:Epub-.
    222) Stanek EJ, Nelson CE, & DeNofrio D: Amlodipine overdose. Ann Pharmacotherapy 1997; 31:853-855.
    223) Stolbach A & Hoffman RS: Respiratory Principles. In: Nelson LS, Hoffman RS, Lewin NA, et al, eds. Goldfrank's Toxicologic Emergencies, 9th ed. McGraw Hill Medical, New York, NY, 2011.
    224) Sun DK, Reiner D, & Frishman W: Adverse dermatologic reactions from antiarrhythmic drug therapy. J Clin Pharmacol 1994; 34:953-966.
    225) Szekely LA, Thompson BT, & Woolf A: Use of partial liquid ventilation to manage pulmonary complications of acute verapamil-sustained release poisoning. Clin Toxicol 1999; 37:475-479.
    226) Takahashi h, Ohashi n, & Motokawa K: Poisoning caused by the combined ingestion of nifedipine and metoprolol. Clin Toxicol 1993; 31:631-637.
    227) Taketomo CK, Hodding JH, & Kraus DM (Eds): Pediatric Dosage Handbook, 4th. Lexi-Comp, Inc, Cleveland, OH, 1997.
    228) Tanen DA, Ruha AM, & Curry SC: Hypertonic sodium bicarbonate is effective in the acute management of verapamil toxicity in a swine model. Ann Emerg Med 2000; 36:547-553.
    229) Taylor JR, Streetman DS, & Castle SS: Medication Bezoars: a literature review and report of a case. Ann Pharmacother 1998; 32:940-946.
    230) Tebbutt S, Harvey M, Nicholson T, et al: Intralipid prolongs survival in a rat model of verapamil toxicity. Acad Emerg Med 2006; 13(2):134-139.
    231) Tenenbein M, Cohen S, & Sitar DS: Whole bowel irrigation as a decontamination procedure after acute drug overdose. Arch Int Med 1987; 147:905-907.
    232) Tenney F & Sakarcan A: Nicardipine is a safe and effective agent in pediatric hypertensive emergencies. Am J Kidney Dis 2000; 35(5):E20-.
    233) Tobias JD, Hersey S, Mencio GA, et al: Nicardipine for controlled hypotension during spinal surgery. J Pediatr Orthop 1996; 16(3):370-373.
    234) Tobias JD, Lowe S, & Deshpande JK: Nicardipine: perioperative applications in children. Paediatr Anaesth 1995; 5(3):171-176.
    235) Tonks AM: Nimodipine levels in breast milk. N Z J Surg 1995; 65:693-694.
    236) Treluyer JM, Hubert P, Jouvet P, et al: Intravenous nicardipine in hypertensive children. Eur J Pediatr 1993; 152(9):712-714.
    237) Tuka V, Ricarova B, Janota T, et al: Surprisingly delayed escalation of severe verapamil poisoning. Eur J Emerg Med 2009; 16(2):100-102.
    238) US Food and Drug Administration: Alert for Healthcare Professionals: Nimodipine (marketed as Nimotop). US Food and Drug Administration. Rockville, MDAvailable from URL: http://www.fda.gov/cder/drug/InfoSheets/HCP/nimodipineHCP.htm.
    239) Vadlamudi L & Wijdicks EFM: Multifocal myoclonus due to verapamil overdose. Neurol 2002; 58:984-985.
    240) Vale JA, Kulig K, American Academy of Clinical Toxicology, et al: Position paper: Gastric lavage. J Toxicol Clin Toxicol 2004; 42:933-943.
    241) Vale JA: Position Statement: gastric lavage. American Academy of Clinical Toxicology; European Association of Poisons Centres and Clinical Toxicologists. J Toxicol Clin Toxicol 1997; 35:711-719.
    242) Varpula T, Rapola J, Sallisalmi M, et al: Treatment of serious calcium channel blocker overdose with levosimendan, a calcium sensitizer. Anesth Analg 2009; 108(3):790-792.
    243) Verbrugge LB & vanWezel HB: Pathophysiology of verapamil overdose: new insights in the role of insulin. J Cardiothorac Vasc Anesth 2007; 21(3):406-409.
    244) Watling SM, Crain JL, & Edwards TD: Verapamil overdose: case report and review of the literature. Ann Pharmacother 1992; 26:1373-1377.
    245) Welch CD, Knoerzer RE, & Lewis GS: Verapamil and acebutolol overdose results in asystole: intra-aortic balloon pump provides mechanical support. J Extra-Corporeal Tech 1992; 24:36-37.
    246) Wells CD, Luckritz TC, Rady MY, et al: Bezoar formation requiring endoscopic removal after intentional overdose of extended-release nifedipine. Pharmacotherapy 2006; 26(12):1802-1805.
    247) Wells TG, Graham CJ, & Moss MM: Nifedipine poisoning in a child. Pediatrics 1990; 86:91-94.
    248) West PL, McKeown NJ, & Hendrickson RG: Iatrogenic lipid emulsion overdose in a case of amlodipine poisoning. Clin Toxicol (Phila) 2010; 48(4):393-396.
    249) White RD, Goldsmith RS, & Rodrigues R: Plasma ionic calcium levels following injection of chloride, gluconate, and gluceptate salts of calcium. J Thorac Cardiovasc Surg 1976; 71:609-613.
    250) Whitebloom D & Fitzharris J: Nifedipine overdose. Clin Cardiol 1988; 11:505-506.
    251) Wilffert B, Boskma RJ, vanderVoort PH, et al: 4-Aminopyridine (fampridine) effectively treats amlodipine poisoning: a case report. J Clin Pharm Ther 2007; 32(6):655-657.
    252) Williamson KM & Dunham GD: Plasma concentrations of diltiazem and desacetyldiltiazem in an overdose situation. Ann Pharmacother 1996; 30:608-611.
    253) Willson DF, Truwit JD, Conaway MR, et al: The adult calfactant in acute respiratory distress syndrome (CARDS) trial. Chest 2015; 148(2):356-364.
    254) Wilson DF, Thomas NJ, Markovitz BP, et al: Effect of exogenous surfactant (calfactant) in pediatric acute lung injury. A randomized controlled trial. JAMA 2005; 293:470-476.
    255) Wood DM, Wright KD, Jones AL, et al: Metaraminol (Aramine(R)) in the management of a significant amlodipine overdose. Human Exp Toxicol 2005; 24:377-381.
    256) Young AC, Velez LI, & Kleinschmidt KC: Intravenous fat emulsion therapy for intentional sustained-release verapamil overdose. Resuscitation 2009; 80(5):591-593.
    257) Yuan TH, Kerns WP, & Tomaszewski CA: Insulin-glucose as adjunctive therapy for severe calcium channel antagonist poisoning. Clin Toxicol 1999; 37:463-474.
    258) Zhou H, Liu Y, Li GQ, et al: A novel dosing regimen for calcium infusion in a patient of massive overdose of sustained-release nifedipine. Am J Med Sci 2013; 345(3):248-251.
    259) da Silva OA, de Melo RA, & Jorge Filho JP: Verapamil acute self-poisoning. Clin Toxicol 1979; 14:361-367.
    260) 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.
    261) de Faire & Lundman T: Serious verapamil poisoning (letter). Br Med J 1978; 2:1574.
    262) de Faire U & Lundman T: Attempted suicide with verapamil. Eur J Cardiol 1977; 6:195-198.
    263) ter Wee PM, Kremer Hovinga TK, Uges DRA, et al: 4-Aminopyridine and haemodialysis in the treatment of verapamil intoxication. Human Toxicol 1985; 4:327-329.