MOBILE VIEW  | 

QUININE

Classification   |    Detailed evidence-based information

Therapeutic Toxic Class

    A) Quinine is used in the treatment of uncomplicated Plasmodium falciparum malaria by inhibiting nucleic acid synthesis, protein synthesis, and glycolysis in P. falciparum and can bind with hemazoin in parasitized erythrocytes.
    B) The FDA warns against the use of quinine for night time leg cramps due to the risk of serious and life-threatening reactions.

Specific Substances

    1) Chininum
    2) Quinine sulfate
    3) Quinina
    4) Molecular Formula: (C(20)H(24)N(2)O(2))2 H(2)SO(4)2H(2)O
    5) CAS 130-95-0 (anhydrous)
    1.2.1) MOLECULAR FORMULA
    1) QUININE SULFATE: (C20H24N2O2)2-H2SO4-2H2O

Available Forms Sources

    A) FORMS
    1) Quinine sulfate is available in a clear 324 mg capsules in bottles of 30, 100, 500 and 1000 (Prod Info QUALAQUIN(R) oral capsules, 2010).
    B) USES
    1) SUMMARY
    a) Quinine sulfate is used for the treatment of uncomplicated Plasmodium falciparum malaria. It has been found to be effective in geographical regions where resistance to chloroquine has been documented (Prod Info QUALAQUIN(R) oral capsules, 2010).
    b) Quinine is NOT approved for patients with severe or complicated P. falciparum malaria or the prevention of malaria (Prod Info QUALAQUIN(R) oral capsules, 2010).
    c) Quinine is NOT approved for the treatment or prevention of night time leg cramps (Prod Info QUALAQUIN(R) oral capsules, 2010; US Food and Drug Administration, 2010).
    2) SAFETY ANNOUNCEMENT
    a) In July 2010, the Food and Drug Administration (FDA), announced that due to continued reports of serious side effects in patients using quinine for the "off-label" treatment of night time leg cramps, warned against the use of quinine because of the risk for serious and life-threatening blood-related (ie, thrombocytopenia) reactions. Other adverse events include hemolytic-uremia syndrome/thrombotic thrombocytopenic purpura which can produce permanent kidney damage. The FDA recommends that patients taking quinine for night time leg cramps should discuss other treatment options with a healthcare professional (US Food and Drug Administration, 2010).
    1) Thirty-eight cases of serious adverse events (24 hematologic, 4 cardiovascular and 10 miscellaneous events) associated with quinine use for the treatment of night time leg cramps have been submitted to the FDA's Adverse Event Reporting System (AERS) from April 2005 to October 2008. Of the 24 hematologic cases, median time to onset of adverse events after starting quinine therapy was 13.5 days. Twenty-one patients developed thrombocytopenia (median platelet count: 4500 cells/microliter) with 4 patients developing thrombotic thrombocytopenic purpura (n=2) or idiopathic thrombocytopenic purpura (n=2). Most patients recovered with the discontinuation of quinine therapy; however, 2 deaths were reported (US Food and Drug Administration, 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: Quinine is indicated in the treatment of uncomplicated Plasmodium falciparum malaria. It is NOT approved for use in the treatment of night time leg cramps. In July 2010, the Food and Drug Administration warned against the use of "off-label" quinine for night time leg cramps because of the risk for serious and life-threatening blood-related (ie, thrombocytopenia) events. Quinine has been used less commonly for chloroquine-resistant malaria. It has also been used as an abortifacient and as an adulterant in heroin because of its bitter taste.
    B) PHARMACOLOGY: Quinine has anti- and prodysrhythmic effects secondary to inhibition of cardiac sodium and potassium channels. It also can act as an alpha-adrenergic blocker.
    C) TOXICOLOGY: Inhibition of the sodium channels may result in widening of the QRS complex, and inhibition of the potassium channels may predispose patients to torsades de pointes. Alpha-adrenergic blockade may cause hypotension. Because of its oxytocic effects, it may act as an abortifacient. Quinine is directly toxic to the retina and causes inhibition of hearing by various mechanisms. It also inhibits ATP-sensitive potassium channels in pancreatic beta cells causing insulin release similar to sulfonylureas, resulting in hyperinsulinemia and hypoglycemia. However, this is usually limited to high dose intravenous treatment.
    D) EPIDEMIOLOGY: Quinine exposure is uncommon; however, with severe exposure, manifestations are life-threatening.
    E) WITH THERAPEUTIC USE
    1) At therapeutic doses, patients often complain of skin changes, gastrointestinal upset, and various subtle neurological complaints, including headache, vision changes, hypoglycemia and alteration in hearing. Quinine-induced thrombocytopenia appears to be immune mediated, may be life threatening, and usually resolves within a week of discontinuation. Hemolytic uremic syndrome may occur in patients with G6PD deficiency and thrombotic thrombocytopenic purpura have also been reported. Immune-mediated pancytopenia and coagulopathy may occur at therapeutic doses of quinine. This may be associated with renal failure and the hemolytic uremic syndrome. Contact dermatitis may occur following topical administration.
    F) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: In mild overdose, patients present with skin flushing, gastrointestinal upset (nausea and vomiting is common), and cinchonism (tinnitus, deafness, vertigo, headache, and visual disturbances).
    2) SEVERE TOXICITY: Patients can develop ataxia, CNS depression, coma, seizures, respiratory arrest, hypotension, PR prolongation, QRS widening, QTc prolongation, ST depression, AV blocks, and various ventricular dysrhythmias. CNS toxicity seems to be more marked in children than adults; children frequently present with seizures following an overdose. Cardiac toxicity resembles toxicity secondary to quinidine. Retinal toxicity may also result approximately 9 to 10 hours postingestion. Patients complain of blurred vision, tunnel vision, visual field constriction, diplopia, alteration in color perception, photophobia, scotomata, and frank blindness. Respiratory depression may occur. Visual complaints usually resolve without treatment.
    0.2.4) HEENT
    A) WITH POISONING/EXPOSURE
    1) Decreased visual acuity and visual field constriction may progress to sudden blindness with nonreactive, dilated pupils following quinine overdose.
    2) Fixed dilated pupils are seen frequently in children following quinine overdose.
    3) Tinnitus and concentration-dependent hearing impairment are frequent.
    0.2.5) CARDIOVASCULAR
    A) WITH POISONING/EXPOSURE
    1) Sinoatrial, atrioventricular, and His-ventricular conduction delays may occur resulting in significant QRS and QT interval prolongation, PR prolongation, ST depression and T wave inversion on ECG. Ventricular fibrillation and ventricular tachycardia, may also occur. Torsade de pointes may lead to syncope.
    2) Hypotension occurs frequently following severe overdose. Heart failure may result.
    3) ONSET: Cardiotoxicity typically appears within 8 hours following ingestion of quinine.
    0.2.20) REPRODUCTIVE
    A) Quinine is classified as FDA pregnancy category C. Quinine crosses the placental barrier and yields measurable blood concentrations in the fetus. Numerous malformations and fetal anomalies have been reported in humans and animals. Quinine is excreted in human breast milk; however, the nursing infant receives a small amount of the maternal dose and toxicity has not been observed.

Laboratory Monitoring

    A) Specific laboratory studies for confirming quinine ingestion are not readily available in most clinical laboratories, and are not generally useful for guiding therapy.
    B) Monitor serum electrolytes, BUN, creatinine, glucose, CBC, CPK, urinalysis, and ECG.
    C) In the acutely ill patient, blood gas analysis may demonstrate metabolic acidosis. Lactate is also usually elevated.

Treatment Overview

    0.4.2) ORAL/PARENTERAL EXPOSURE
    A) MANAGEMENT OF MILD TO MODERATE TOXICITY
    1) Treatment is primarily supportive with IV fluids and antiemetics for gastrointestinal upset.
    B) MANAGEMENT OF SEVERE TOXICITY
    1) Treat QRS widening with ampules of sodium bicarbonate 1 to 2 mEq/kg rapid IV bolus with a goal serum pH of 7.45 to 7.55 until QRS narrows. Treat torsades de pointes with 2 g of magnesium and aggressive potassium supplementation. Overdrive pacing may be necessary. Cardiovert unstable ventricular dysrhythmias. Type Ib antiarrhythmic drugs like lidocaine (50 to 100 mg IV in adults and 1 mg/kg in children) may be utilized for ventricular dysrhythmias, but are often ineffective.
    2) Treat hypotension with IV fluids; add adrenergic vasopressors if hypotension persists.
    3) The management of retinal toxicity is controversial. It usually resolves without treatment. In general, stellate ganglion blocks are not effective. Oral vasodilatory drugs, nitrates, vasospastic drugs, and hyperbaric therapy have been utilized in some cases, but there have been no studies of efficacy.
    4) Treat seizures with aggressive benzodiazepine therapy; add propofol or barbiturates if seizures persist.
    5) Treat hypoglycemia with dextrose and consider octreotide (ADULTS: 50 to 100 mcg SubQ every 6 to 12 hours; CHILDREN: 1 mcg/kg SubQ every 6 hours).
    C) DECONTAMINATION
    1) PREHOSPITAL: Administer activated charcoal if the ingestion is recent and the patient is not vomiting and is alert.
    2) HOSPITAL: Administer activated charcoal if the patient is not vomiting and ingestion is recent. Consider gastric lavage for patients with massive, recent ingestions who are not vomiting and have a secure airway. Multi-dose activated charcoal can also be utilized (0.5 to 1 g/kg every 2 to 4 hours) if the patient is not vomiting and airway is secure.
    D) AIRWAY MANAGEMENT
    1) Perform early in patients with severe intoxication (ie, coma, CNS depression, seizures, or dysrhythmias).
    E) ANTIDOTE
    1) None
    F) ENHANCED ELIMINATION
    1) There is no clinical role for hemodialysis or hemoperfusion because quinine has an extensive volume of distribution and is highly protein bound.
    G) PATIENT DISPOSITION
    1) HOME CRITERIA: Home management is only indicated if the patient is asymptomatic, ingestion was minimal (therapeutic dose or less) and there is no suspicion for self-harm.
    2) OBSERVATION CRITERIA: Any patient with an intentional ingestion, or a significant ingestion greater than weight/age appropriate dosing, should be observed for 6 to 8 hours or until asymptomatic.
    3) ADMISSION CRITERIA: Admit any patient with altered mental status, hemodynamic instability, or vision changes.
    4) CONSULT CRITERIA: Consult ophthalmology if the patient has any visual disturbances. Involve a toxicologist or poison center immediately if there is concern for intentional or significant overdose.
    H) PITFALLS
    1) Pitfalls include use of other Ia, Ic, or III anti-arrhythmic drugs as they may worsen cardiotoxicity. Failure to replace potassium and magnesium may exacerbate the toxic effect of quinine on potassium channels and predispose patients to QTc prolongation and torsades de pointes.
    I) PHARMACOKINETICS
    1) Quinine's peak concentration is 1 to 3 hours following oral administration. Half-life after therapeutic oral doses is approximately 12 hours. Quinine is 93% to 95% protein bound and has a volume of distribution of 3 L/kg. Elimination may be increased in patients with hepatic or renal failure.
    J) TOXICOKINETICS
    1) Half-life of quinine following overdose can be greater than 24 hours.
    K) PREDISPOSING CONDITIONS
    1) Preexisting liver or renal disease may predispose patients to toxicity.
    L) DIFFERENTIAL DIAGNOSIS
    1) Other antimalarial agents, other type Ia antiarrhythmic agents, and tricyclic antidepressants

Range Of Toxicity

    A) TOXICITY: In adults, mild to moderate toxicity is reported after ingestion of 2 to 4 grams, severe toxicity after 4.5 to 9 grams, and fatalities after 5 to 9.75 grams.
    B) THERAPEUTIC DOSE: ADULTS: 648 mg orally every 8 hours for 7 days. CHILDREN: 10 mg/kg orally three times daily for 3 or 7 days.

Gastrointestinal

    3.8.2) CLINICAL EFFECTS
    A) NAUSEA AND VOMITING
    1) WITH THERAPEUTIC USE
    a) Nausea and vomiting are common side effects of quinine (Tiliakos & Waites, 1981).
    2) WITH POISONING/EXPOSURE
    a) Nausea has been reported following quinine overdose (Vusirikala et al, 2005).
    b) Persistent vomiting has occurred within 2 hours of toxic ingestions (Nordt & Clark, 1998).

Hepatic

    3.9.2) CLINICAL EFFECTS
    A) INJURY OF LIVER
    1) WITH THERAPEUTIC USE
    a) Hepatotoxicity with therapeutic quinine use has been reported (Perez et al, 1994) and has occurred within 24 hours of initiation of a quinine dose (Farver & Lavin, 1999), although onset generally occurs after 2 weeks of therapy. On discontinuance of quinine, liver enzyme values have returned to normal.

Genitourinary

    3.10.2) CLINICAL EFFECTS
    A) UREMIA
    1) WITH THERAPEUTIC USE
    a) Hemolytic uremic syndrome (HUS) has occasionally been reported as an adverse effect of quinine therapy. In these cases, the immune-mediated nature of the blood dyscrasia is specific to quinine. The presence of quinine-dependent antibodies to red cells, granulocytes or platelets has been demonstrated in some cases. There appears to be no correlation between the type and specificity of antibody and severity of renal failure (McDonald et al, 1997).
    b) CASE REPORT: HUS was reported in a 46-year-old woman after a single dose of 260 mg quinine (Hagley et al, 1992).
    1) SIGNS/SYMPTOMS: Nausea, vomiting and myalgias started 2 hours after the dose; 5 days later, the serum creatinine increased to 11.5 mg/dL.
    2) Dialysis was performed days 5 through 21; the patient recovered fully in 30 days.
    c) CASE REPORT: Following a single 300 mg dose of quinine, a 57-year-old woman presented with a 4 day history of abdominal pain, nausea, diarrhea, bruising and a transient morbilliform rash. Acute renal failure, hemolysis, and evidence of DIC were seen on laboratory data. The patient's serum reacted with normal red cells in an indirect antiglobulin test in the presence of quinine. A diagnosis of HUS was made. The patient recovered following 6 plasma exchanges, supportive hemodialysis and steroid therapy (McDonald et al, 1997).
    d) CASE REPORT: A 78-year-old woman developed acute renal failure, progressive coagulopathy, thrombocytopenia, and hemolysis with red blood cell fragmentation several hours after ingesting 150 mg of quinine for treatment of leg cramps. Laboratory analysis showed a serum creatinine of 0.58 mmol/L, elevated LDH and bilirubin serum levels, and a low haptoglobin concentration which is consistent with hemolytic-uremic syndrome. The patient gradually recovered following treatment with plasma exchange, corticosteroids, and hemodialysis, although the serum concentration continued to remain elevated at 0.17 mmol/L two months post ingestion (Morton, 2002).
    B) INTERSTITIAL NEPHRITIS
    1) WITH THERAPEUTIC USE
    a) Quinine-induced hemolysis and renal failure, due to acute interstitial nephritis, has been reported (Pawar et al, 1994).
    C) ACUTE RENAL FAILURE SYNDROME
    1) WITH POISONING/EXPOSURE
    a) A 21-year-old woman ingested 90 g of quinine as a single-dose in her eighth-week of pregnancy to induce abortion. The patient subsequently became drowsy, with dark red urine which was positive for occult blood and albumin. She became oliguric with blood urea climbing to 208 g/dL. Platelet count was reported as 43,000/cu mm; 24 hours after ingestion the patient had a seizure. Therapy with diuretics, blood transfusion and hemodialysis resulted in improvement of her condition with hospital discharge at 37 days (Notelovitz et al, 1970).

Acid-Base

    3.11.2) CLINICAL EFFECTS
    A) ACIDOSIS
    1) WITH POISONING/EXPOSURE
    a) Arterial blood gas analysis has revealed severe mixed respiratory and metabolic acidosis in a patient with seizures and hypotension after an overdose of 9 grams quinine (Pearson, 1998).

Hematologic

    3.13.2) CLINICAL EFFECTS
    A) BLOOD COAGULATION PATHWAY FINDING
    1) WITH POISONING/EXPOSURE
    a) HYPOPROTHROMBINEMIA has been reported after quinine overdose (Goldenberg & Wexler, 1988).
    B) HEMOLYTIC ANEMIA
    1) WITH THERAPEUTIC USE
    a) Hemolytic anemia may occur, particularly in patients with glucose-6-phosphate-dehydrogenase deficiency.
    C) PANCYTOPENIA
    1) WITH THERAPEUTIC USE
    a) Immune-mediated pancytopenia and coagulopathy may occur at therapeutic doses of quinine (Schmitt & Tomford, 1994). This may be associated with renal failure and the hemolytic uremic syndrome (Morton, 2002; Blayney, 1992; Story et al, 1994).
    b) Profound thrombocytopenia and neutropenia have been reported in a 40-year-old woman, in addition to menorrhagia, 2 days after starting quinine 300 mg every 6 hours as needed. Platelet-associated antibodies and neutrophil antibodies were detected. Bone marrow aspirate was reported to be normal. Five days after discontinuing quinine, WBC and platelet counts returned to normal (Chuah & Denaro, 2000).
    D) UREMIA
    1) WITH THERAPEUTIC USE
    a) Antibodies to platelets, red blood cells, T lymphocytes and neutrophils have been associated with the development of the hemolytic uremic syndrome following quinine use (Stroncek et al, 1992; Aster, 1993; Maguire et al, 1993; Hagley et al, 1992).
    E) THROMBOCYTOPENIC DISORDER
    1) WITH THERAPEUTIC USE
    a) SUMMARY: Quinine-induced thrombocytopenia appears to be immune mediated. In some patients, the events have been life-threatening or fatal. Cases of hemolytic uremic syndrome and thrombotic thrombocytopenic purpura have developed. Thrombocytopenia usually resolves within a week of quinine discontinuation, but is likely to reoccur with the administration of quinine from any source (Prod Info QUALAQUIN(R) oral capsules, 2010).
    b) FDA WARNING: In July 2010, the Food and Drug Administration (FDA), announced that due to continued reports of serious side effects in patients using quinine for the "off-label" treatment of night time leg cramps, warned against the use of quinine because of the risk for serious and life-threatening blood-related (ie, thrombocytopenia) reactions. Other adverse events include hemolytic-uremia syndrome/thrombotic thrombocytopenic purpura which can produce permanent kidney damage. The FDA recommends that patients taking quinine for night time leg cramps should discuss other treatment options with a healthcare professional (US Food and Drug Administration, 2010).
    1) Thirty-eight cases of serious adverse events (24 hematologic, 4 cardiovascular and 10 miscellaneous events) associated with quinine use for the treatment of night time leg cramps have been submitted to the FDA's Adverse Event Reporting System (AERS) from April 2005 to October 2008. Of the 24 hematologic cases, median time to onset of adverse events after starting quinine therapy was 13.5 days. Twenty-one patients developed thrombocytopenia (median platelet count: 4500 cells/microliter) with 4 patients developing thrombotic thrombocytopenic purpura (n=2) or idiopathic thrombocytopenic purpura (n=2). Most patients recovered with the discontinuation of quinine therapy; however, 2 deaths were reported (US Food and Drug Administration, 2010).
    c) CASE REPORT/FATALITY: A 70-year-old man was admitted to the ED with shortness of breath and hemoptysis for 24 hours. Of note, the patient had a prior history of quinine-induced thrombocytopenia after a short course of quinine, which responded to prednisone and immunoglobulin. Five days prior to the current admission, the patient had been prescribed quinine (300 mg daily) for leg cramps without knowing the patient's history (ie, no prior medical records available and the patient was a poor historian). He was immediately started on high-flow oxygen to maintain a pulse oximetry of 88%. An initial chest x-ray showed evidence of bilateral pulmonary hemorrhage. Petechiae and ecchymosis were observed over his limbs and he had a platelet count of less than 10 x E(9)/L. A platelet transfusion (4 units) was administered along with prednisone. Immunoglobulin was added after the patient failed to improve. Despite various interventions, the patient developed increased oxygen requirements and died. Although quinine-induced disseminated intravascular coagulation was present, there were no signs of thrombotic thrombocytopenia purpura and hemolytic uremic syndrome (Samaranayake & Yap, 2014).
    d) CASE REPORT: A 57-year-old presented to the ED with petechiae and thrombocytopenia (platelet count, 2 x 10(9)/L). Abundant megakaryocytes were seen in a bone marrow aspirate which was consistent with increased peripheral destruction of platelets via quinine-associated immune thrombocytopenia. Laboratory data confirmed therapeutic serum quinine levels, even though the patient denied quinine use. The authors report a Munchausen's syndrome in this patient (Abraham & Whitehead, 1998).
    F) DISSEMINATED INTRAVASCULAR COAGULATION
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: A 78-year-old woman developed disseminated intravascular coagulation (DIC) several hours after ingesting 150 mg of quinine for treatment of leg cramps. The coagulopathy resolved within 48 hours postingestion (Morton, 2002).

Dermatologic

    3.14.2) CLINICAL EFFECTS
    A) PHOTOSENSITIVITY
    1) WITH THERAPEUTIC USE
    a) Photosensitivity may be seen after therapeutic quinine ingestion (Ljunggren et al, 1992; Ferguson et al, 1987; Okun et al, 1994; Dawson, 1995; Ljunggren et al, 1992; Wagner et al, 1994).
    B) CONTACT DERMATITIS
    1) WITH THERAPEUTIC USE
    a) Contact dermatitis has been reported after topical application (Tapadinhas et al, 1994) (Valseechi et al, 1994).
    C) VASCULITIS
    1) WITH THERAPEUTIC USE
    a) Severe cutaneous vasculitis developed in a 60-year-old woman 3 weeks after beginning therapy with quinine sulfate (Price et al, 1992).

Endocrine

    3.16.2) CLINICAL EFFECTS
    A) HYPOGLYCEMIA
    1) WITH THERAPEUTIC USE
    a) Therapeutic usage of quinine has resulted in severe hypoglycemia (White et al, 1983) Harets et al, 1984) (Okitolonda et al, 1987; Limburg et al, 1993).
    1) However, this hypoglycemia may be a complication of falciparum malaria in children, reflecting severe disease and associated with a poor prognosis.
    b) CASE SERIES: A recent study in 76 children who received intravenous quinine for falciparum malaria did not find any association between quinine and hypoglycemia (Taylor et al, 1988).
    2) WITH POISONING/EXPOSURE
    a) Severe hypoglycemia has been reported following a quinine overdose (Wenstone et al, 1989).

Immunologic

    3.19.2) CLINICAL EFFECTS
    A) DISORDER OF IMMUNE FUNCTION
    1) WITH THERAPEUTIC USE
    a) Therapeutic quinine use was associated with a positive lupus anticoagulant test in one study (Bird et al, 1995).
    B) ACUTE ALLERGIC REACTION
    1) WITH THERAPEUTIC USE
    a) A variant form of quinine toxicity, with hypersensitivity mimicking septic shock, has been reported. Within 2 hours of a therapeutic quinine dose, a patient presented to the ED with sudden onset of fever, rigors, and back pain followed within hours by hypotension, metabolic acidosis, and disseminated intravascular coagulation. All cultures were reported to be negative. Within 24 to 36 hours all symptoms abated. This sequence occurred twice; both times it was preceded by a quinine dose 2 hours earlier. No further episodes occurred after stopping quinine (Schattner, 1998).

Summary Of Exposure

    A) USES: Quinine is indicated in the treatment of uncomplicated Plasmodium falciparum malaria. It is NOT approved for use in the treatment of night time leg cramps. In July 2010, the Food and Drug Administration warned against the use of "off-label" quinine for night time leg cramps because of the risk for serious and life-threatening blood-related (ie, thrombocytopenia) events. Quinine has been used less commonly for chloroquine-resistant malaria. It has also been used as an abortifacient and as an adulterant in heroin because of its bitter taste.
    B) PHARMACOLOGY: Quinine has anti- and prodysrhythmic effects secondary to inhibition of cardiac sodium and potassium channels. It also can act as an alpha-adrenergic blocker.
    C) TOXICOLOGY: Inhibition of the sodium channels may result in widening of the QRS complex, and inhibition of the potassium channels may predispose patients to torsades de pointes. Alpha-adrenergic blockade may cause hypotension. Because of its oxytocic effects, it may act as an abortifacient. Quinine is directly toxic to the retina and causes inhibition of hearing by various mechanisms. It also inhibits ATP-sensitive potassium channels in pancreatic beta cells causing insulin release similar to sulfonylureas, resulting in hyperinsulinemia and hypoglycemia. However, this is usually limited to high dose intravenous treatment.
    D) EPIDEMIOLOGY: Quinine exposure is uncommon; however, with severe exposure, manifestations are life-threatening.
    E) WITH THERAPEUTIC USE
    1) At therapeutic doses, patients often complain of skin changes, gastrointestinal upset, and various subtle neurological complaints, including headache, vision changes, hypoglycemia and alteration in hearing. Quinine-induced thrombocytopenia appears to be immune mediated, may be life threatening, and usually resolves within a week of discontinuation. Hemolytic uremic syndrome may occur in patients with G6PD deficiency and thrombotic thrombocytopenic purpura have also been reported. Immune-mediated pancytopenia and coagulopathy may occur at therapeutic doses of quinine. This may be associated with renal failure and the hemolytic uremic syndrome. Contact dermatitis may occur following topical administration.
    F) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: In mild overdose, patients present with skin flushing, gastrointestinal upset (nausea and vomiting is common), and cinchonism (tinnitus, deafness, vertigo, headache, and visual disturbances).
    2) SEVERE TOXICITY: Patients can develop ataxia, CNS depression, coma, seizures, respiratory arrest, hypotension, PR prolongation, QRS widening, QTc prolongation, ST depression, AV blocks, and various ventricular dysrhythmias. CNS toxicity seems to be more marked in children than adults; children frequently present with seizures following an overdose. Cardiac toxicity resembles toxicity secondary to quinidine. Retinal toxicity may also result approximately 9 to 10 hours postingestion. Patients complain of blurred vision, tunnel vision, visual field constriction, diplopia, alteration in color perception, photophobia, scotomata, and frank blindness. Respiratory depression may occur. Visual complaints usually resolve without treatment.

Vital Signs

    3.3.3) TEMPERATURE
    A) WITH THERAPEUTIC USE
    1) DRUG FEVER: Drug fever occurred in a 19-year-old and a 22-year-old who were treated for malaria with 600 mg of quinine 3 times/day for 4 to 10 days. One patient developed shaking chills and fever, which spiked to 104 degrees F, and the other patient developed epigastric burning, vomiting, dyspnea, weakness, fever and a 5% eosinophilia. Discontinuation of quinine resulted in quick disappearance of the fever, and in 1 patient rechallenge with 300 mg of quinine resulted in recurrence of the chills and fever within 6 hours (Schlutz et al, 1973).

Heent

    3.4.1) SUMMARY
    A) WITH POISONING/EXPOSURE
    1) Decreased visual acuity and visual field constriction may progress to sudden blindness with nonreactive, dilated pupils following quinine overdose.
    2) Fixed dilated pupils are seen frequently in children following quinine overdose.
    3) Tinnitus and concentration-dependent hearing impairment are frequent.
    3.4.3) EYES
    A) BLINDNESS
    1) WITH POISONING/EXPOSURE
    a) Following an overdose, onset of symptoms of oculotoxicity may vary from a few hours to a day or more (S Sweetman , 1999). In addition to cinchonism, quinine is thought to have either an ischemic action on the retinal vasculature or a direct toxic effect on the retina, causing constricted visual fields that can progress to blindness with dilated nonreactive pupils (Brinton et al, 1980; Dyson & Proudfoot, 1985; Nordt & Clark, 1998; Barrett & Solano, 2002).
    b) Central vision usually recovers first. Complete recovery of vision may take several months; pupils may remain dilated after recovery of vision (Wolf et al, 1992). Permanent visual deficits may remain (Prod Info QUALAQUIN(R) oral capsules, 2008).
    c) CASE REPORT: Visual function tests following quinine overdose in a 19-year-old girl suggest that quinine exerts a direct effect on the cells of the outer retina and pigment epithelium, as well as on the ganglion cells (Moloney et al, 1987).
    d) CASE SERIES: In a series of 165 patients overdosing on quinine, 70 (42%) developed visual symptoms 2 to 24 hours following ingestion. Of these patients, 39 (56%) subsequently complained of total blindness while 31 (44%) developed blurring of vision only.
    1) Nineteen patients with total vision loss developed permanent visual deficits, although none were completely blinded (however, two patients remained completely blind in one eye). Five patients died (Boland et al, 1985).
    e) CASE SERIES: In a case series of 96 patients with quinine intoxication, visual toxicity developed in only 1 (3%) of 37 patients with reported ingestions of less than 1 gram compared with 80% of the patients with reported ingestions of more than 5 g (Langford et al, 2003)
    f) CASE REPORT: Bilateral loss of vision, with dilated and nonreactive pupils, occurred in a 26-year-old man following ingestion of 5 g quinine and an unknown quantity of alcohol (Drake WM & Hiorns MP, 1994). The individual had been taking 300 mg quinine nightly for several years. The peak serum quinine level was 11 mg/L approximately 16 hours after ingestion.
    1) The administration of charcoal and isosorbide dinitrate resolved the patient's nausea and tinnitus, but did not restore normal vision. The patient's pupils remained dilated, with ischemic retinas two weeks later.
    g) Plasma concentrations associated with the visual deficits usually exceed 15 mcg/mL in less than 10 hours postingestion and greater than 10 mcg/mL after 10 hours. However, plasma concentrations appear to be inadequate to predict the type of visual disturbance.
    h) CASE SERIES: Bilateral stellate ganglion block was initiated in 34 patients with impaired visual acuity or blindness, however, response was poor with improvement of symptoms appearing in only 4 cases.
    1) Stellate ganglion blockade does not appear to be as effective as previously claimed for treating visual disturbances secondary to quinine overdose and it is not indicated regularly in quinine induced amblyopia (Boland et al, 1985).
    i) Blindness following quinine overdose may be delayed in onset for more than 12 hours. Visual function may return entirely or partially without use of modalities such as hemoperfusion or hemodialysis or stellate ganglion block (Smilkstein et al, 1987; Pearson, 1998).
    j) CASE REPORT: A 57-year-old man presented with confusion and bilateral blindness approximately 36 hours after ingesting approximately 7.2 g quinine sulfate (equivalent to 6 g quinine base) with an unknown amount of alcohol. The patient's past medical history included surgery on the left eye, 19 years ago, due to prolapse of the iris secondary to a traffic accident. Physical examination showed bilateral dilation and absent light reflexes. Visual acuity was reduced to light perception only. With supportive care, the patient's central vision partially returned, however visual field defects continued to persist six weeks after hospital discharge (Prasad et al, 2003). It is believed that methanol did not play a role in this patient's blindness because metabolic acidosis, a common occurrence following methanol intoxication, was not present.
    k) Complete blindness occurred in a 55-year-old woman 7 days after ingesting an unknown amount of quinine (Taylor, 2004).
    l) CASE REPORT: A 39-year-old woman presented with ocular toxicity (reduced visual acuity, dilated, sluggish pupils, and retinal arterial attenuation), 40 hours after taking 28 quinine sulfate tablets (300 mg each). She was treated with nimodipine (60 mg for 6 days), clonidine infusion (300 mcg/24 hours), and stellate ganglion block (SGB) (ropivacaine 1% 10 mL). Since the left eye was amblyopic, it was difficult to quantify the effect of treatment; however, the right visual acuity in the right eye improved from the ability to detect hand motions to 6/12 uncorrected and 9/12 with pinhole correction by the 7th day after admission. The right eye had impaired color vision and severely constricted visual field (Vusirikala et al, 2005).
    2) WITH THERAPEUTIC USE
    a) CASE REPORT: A patient developed blindness after receiving his second dose of intravenous quinine. Vision began to improve 6 hours later, and the following morning vision had returned to normal. The patient's quinine clearance was reduced, and vision began to return when total serum quinine levels peaked. The return of vision was associated with an increase in alpha1-acid glycoprotein levels (which is an acute phase reactant that rises as part of the systemic inflammatory reaction associated with malaria and also binds free quinine), resulting in a reduced fraction of unbound quinine (Di Perri et al, 2002).
    B) VERMIFORM PUPIL MOTION
    1) CASE REPORT: Forty-eight hours after a 9 g ingestion of quinine sulfate in a 28-year-old woman, vermiform pupil motion was noted in both eyes (Canning & Hague, 1988).
    C) MYDRIASIS
    1) Fixed dilated pupils are reported frequently in children following quinine overdose (Hla et al, 1987; Grattan-Smith et al, 1987).
    D) NYSTAGMUS
    1) Transitory latent nystagmus was more frequent following ingestion of 120 or 160 mg quinine than with placebo. A decrease in ocular flutter was also noted (Drewitt et al, 1993).
    E) VISUAL DETERIORATION
    1) CHRONIC TOXICITY: A 46-year-old man developed visual deterioration after drinking 4 liters of tonic water daily for 12 months. Visual acuity improved to normal within 6 months after he stopped drinking tonic water (Horgan & Williams, 1995).
    F) ANIMAL STUDIES
    1) ANIMAL STUDIES: The retinotoxic effect of quinine, with no evidence of acute retinal ischemia, has been confirmed in an experimental animal model of quinine blindness (Buchanan et al, 1987).
    3.4.4) EARS
    A) TINNITUS
    1) WITH POISONING/EXPOSURE
    a) Transient TINNITUS and bilateral nerve deafness have been observed after overdose (Langford et al, 2003; Lincoff, 1955; Braveman et al, 1948; Nordt & Clark, 1998).
    B) DEAFNESS
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: DEAFNESS and mutism occurred in a 14-year-old who overdosed on approximately 6.5 to 7.8 g of quinine sulfate. Symptoms lasted approximately 3 days (Schonwald & Shannon, 1991).
    b) CASE SERIES: In a case series of 96 patients with quinine intoxication, tinnitus and other auditory complaints developed in 17% of the patients with reported ingestions of less than 1 gram compared with 80% of the patients with reported ingestions of more than 5 g (Langford et al, 2003).
    c) Positional abnormalities have been documented at subtherapeutic quinine concentrations that may occur from drinking 1.6 L/day of tonic water (Jung et al, 1993).
    2) WITH THERAPEUTIC USE
    a) Dose-dependent hearing impairment was observed in volunteers taking therapeutic doses of quinine. No differences were observed when the drug was administered orally or intravenously (Paintaud et al, 1993).
    b) Hearing loss is typically reversible at therapeutic doses and occurs in 20% of patients taking prolonged courses of 200 to 300 mg daily. Sensorineural hearing loss affects high frequencies initially. By the time losses at conversational frequencies are noted, the hearing loss may be irreversible (Jung et al, 1993).

Cardiovascular

    3.5.1) SUMMARY
    A) WITH POISONING/EXPOSURE
    1) Sinoatrial, atrioventricular, and His-ventricular conduction delays may occur resulting in significant QRS and QT interval prolongation, PR prolongation, ST depression and T wave inversion on ECG. Ventricular fibrillation and ventricular tachycardia, may also occur. Torsade de pointes may lead to syncope.
    2) Hypotension occurs frequently following severe overdose. Heart failure may result.
    3) ONSET: Cardiotoxicity typically appears within 8 hours following ingestion of quinine.
    3.5.2) CLINICAL EFFECTS
    A) ELECTROCARDIOGRAM ABNORMAL
    1) WITH THERAPEUTIC USE
    a) Ventricular tachycardia, torsade de pointes, and cardiac arrest were reported in a patient who ingested 600 mg of quinine daily for 7 days in combination with mefloquine (Langford et al, 2003).
    2) WITH POISONING/EXPOSURE
    a) Sinus tachycardia and minor ECG changes appear to be the most common cardiovascular effects following overdose. Toxic ECG manifestations include prolongation of PR, QRS and QT intervals, ST depression, and T wave inversion. QT prolongation greater than 50% suggests toxicity. Severe poisoning may result in conduction abnormalities and ventricular dysrhythmias (Vusirikala et al, 2005).
    b) CONDUCTION DEPRESSION: Sinoatrial, atrioventricular, and His-ventricular conduction delays are common. Ventricular tachycardia and ventricular fibrillation occur with severe toxicity.
    c) Sinus bradycardia may occur but anticholinergic effects usually predominate, resulting in sinus tachycardia.
    d) Cardiovascular effects are similar to quinidine but less common. Deaths from dysrhythmias do occur (Dyson & Proudfoot, 1985; Goldenberg & Wexler, 1988).
    e) ONSET: Cardiotoxicity typically appears within 8 hours following ingestion of quinine. Cardiotoxicity delayed until 25 hours after ingestion has been reported (Bodenhamer & Smilkstein, 1993). Prolonged QRS has been reported in a patient who presented approximately 1 hr post-ingestion of 9 grams quinine (Pearson, 1998).
    f) CASE REPORT: Following ingestion of 5.5 grams of quinine, a 51-year-old woman experienced a normal sinus rhythm on ECG with a QRS interval of 0.109 sec and a prolonged corrected QT interval of 0.533 sec. After charcoal administration, a second ECG 3 hours later showed the corrected QT interval to remain prolonged at 0.509 sec, and 30 hr later ECG still showed a prolonged corrected QT interval of 0.520 sec. The patient left the hospital against medical advice 36 hr post ingestion (Nordt & Clark, 1998).
    g) CASE REPORT: Following an overdose of quinine of 9.75 grams, a 32-year-old man presented to the ED. A wide complex tachycardia associated with hypotension was noted, which persisted in spite of pacing, cardioversion, fluids and vasopressors. The patient died approximately 64 hours after the ingestion (Morrison et al, 2003; Morrison et al, 2001).
    B) CARDIAC ARREST
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Asystolic cardiac arrest was reported in a 16-month-old who ingested 600 mg of quinine. Several episodes occurred over a 5 hour period (Hla et al, 1992).
    C) SYNCOPE
    1) WITH POISONING/EXPOSURE
    a) Quinine syncope is attributed to transient torsade de pointes (Swiryn & Kimm, 1983).
    b) Occurrence of syncope or sudden death correlates with QT prolongation but does not correlate with toxic quinine levels or other ECG abnormalities (Bauman et al, 1984).
    D) HYPOTENSIVE EPISODE
    1) WITH POISONING/EXPOSURE
    a) Hypotension occurs from alpha receptor-blockade and depressed myocardial contractility. Heart failure may result (Goldenberg & Wexler, 1988). Within a few hours of ingestion of 9 grams quinine, an adult experienced a drop in blood pressure to 85/65 mmHg; he was resuscitated with 3500 mL intravenous fluids (Pearson, 1998).
    b) CASE REPORT: A 32-year-old man developed hypotension (99/45 mmHg) accompanied by wide complex tachycardia after ingesting 30 325-mg quinine tablets (a total of 9.75 g). Despite decontamination with activated charcoal and administration of fluids, vasopressors, cardioversion, and pacing, the patient died approximately 64 hours postingestion (Morrison et al, 2003).

Respiratory

    3.6.2) CLINICAL EFFECTS
    A) ACUTE RESPIRATORY INSUFFICIENCY
    1) WITH POISONING/EXPOSURE
    a) Massive overdose of quinine has resulted in respiratory depression (Bateman & Dyson, 1986).
    B) ACUTE LUNG INJURY
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: A 45-year-old woman, with a history of rheumatoid arthritis, presented with dyspnea, wheezing, a dry nonproductive cough, orthopnea, fever, chills, and pleuritic chest discomfort approximately 45 minutes after ingesting a single 325-mg dose of quinine for treatment of nocturnal leg cramps. Physical examination revealed diminished bilateral breath sounds with diffuse expiratory wheezing and basilar crackles, and pulse oximetry showed an oxygen saturation of 79%. A chest X-ray showed bilateral diffuse alveolar infiltrates, consistent with non-cardiogenic pulmonary edema. The patient recovered following administration of 100% oxygen, nebulized albuterol, and intravenous administration of corticosteroids (Krantz et al, 2002).
    2) WITH POISONING/EXPOSURE
    a) Adult respiratory distress syndrome (ARDS) was reported in a fatal case (Wenstone et al, 1989).
    C) PULMONARY HEMORRHAGE
    1) WITH THERAPEUTIC USE
    a) CASE REPORT/FATALITY: A 70-year-old man was admitted to the ED with shortness of breath and hemoptysis for 24 hours. Of note, the patient had a prior history of quinine-induced thrombocytopenia after a short course of quinine, which responded to prednisone and immunoglobulin. Five days prior to the current admission, the patient had been prescribed quinine (300 mg daily) for leg cramps without knowing the patient's history (ie, no prior medical records available and the patient was a poor historian). He was immediately started on high-flow oxygen to maintain a pulse oximetry of 88%. An initial chest x-ray showed evidence of bilateral pulmonary hemorrhage. Petechiae and ecchymosis were observed over his limbs and he had a platelet count of less than 10 x E(9)/L. A platelet transfusion (4 units) was administered along with prednisone. Immunoglobulin was added after the patient failed to improve. Despite various interventions, the patient developed increased oxygen requirements and died. Although quinine-induced disseminated intravascular coagulation was present, there were no signs of thrombotic thrombocytopenia purpura and hemolytic uremic syndrome (Samaranayake & Yap, 2014).

Neurologic

    3.7.2) CLINICAL EFFECTS
    A) COMA
    1) WITH POISONING/EXPOSURE
    a) Coma and cinchonism may occur with quinine (Bateman & Dyson, 1986; Valman & White, 1977; Friedman, 1980; Garrod & Judson, 1981).
    B) SEIZURE
    1) WITH POISONING/EXPOSURE
    a) Quinine overdose may produce grand mal seizures (Valman & White, 1977; Friedman, 1980; Garrod & Judson, 1981; Pearson, 1998).
    b) CHILDREN: Central nervous system toxicity seems to be more marked in children than adults; children frequently present with seizures following an overdose of quinine (Hla et al, 1987; Grattan-Smith et al, 1987).
    C) ATAXIA
    1) WITH POISONING/EXPOSURE
    a) Quinine may produce ataxia (Bateman & Dyson, 1986; Valman & White, 1977; Friedman, 1980; Garrod & Judson, 1981).
    D) PARESTHESIA
    1) WITH POISONING/EXPOSURE
    a) Quinine overdose may produce paresthesias (Valman & White, 1977; Friedman, 1980; Garrod & Judson, 1981).
    E) CLOUDED CONSCIOUSNESS
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Confusion and delirium occurred in a 57-year-old man after ingesting approximately 7.2 g quinine sulfate (equivalent to 6 g quinine base) and an unknown amount of alcohol (Prasad et al, 2003).

Reproductive

    3.20.1) SUMMARY
    A) Quinine is classified as FDA pregnancy category C. Quinine crosses the placental barrier and yields measurable blood concentrations in the fetus. Numerous malformations and fetal anomalies have been reported in humans and animals. Quinine is excreted in human breast milk; however, the nursing infant receives a small amount of the maternal dose and toxicity has not been observed.
    3.20.2) TERATOGENICITY
    A) CONGENITAL ANOMALY
    1) Numerous malformations and fetal anomalies have been reported. Quinine has often been used as a home abortifacient, resulting in a 59% incidence of congenital anomalies and 16% of maternal death (Dannenberg et al, 1983).
    2) Among 21 cases of attempted abortion through self administration of large doses (1 to 4 g total) of quinine during early pregnancy, primary defects included hydrocephalus, limb defects, as well as facial and heart anomalies (Schardein, 1976). Optic nerve hypoplasia, congenital deafness, and auditory nerve hypoplasia have been reported after quinine administration in the first trimester of pregnancy (Fong et al, 1984; Nishimura & Tanimura, 1976). Reports from geographic areas where malaria is prevalent and quinine is widely used indicate that congenital deafness is rare and no more common than in non-malarious areas (Nishimura & Tanimura, 1976).
    B) LACK OF EFFECT
    1) Published data on over 1000 pregnant women who were exposed to quinine showed no increase in teratogenic effects over the background rate in the general population. However, the majority of these women were not in their first trimester (Prod Info QUALAQUIN(R) oral capsules, 2013).
    2) The overall incidence of congenital malformations was similar for the quinine group (1.4%) compared with the control group (1.7%) in a 1999 study of offspring of pregnant Thailand women who either had P. falciparum malaria and were treated with quinine sulfate 10 mg/kg 3 times daily for 7 days at any point during pregnancy (n=633) or did not have malaria and were not exposed to antimalarial drugs during pregnancy (n=2201) (Prod Info QUALAQUIN(R) oral capsules, 2013).
    3) No increased risk of structural birth defects was observed (1.9%, 2 fetal malformations) in an epidemiologic survey of 104 mother-child pairs in which the mother was exposed to quinine during the first 4 months of gestation (Prod Info QUALAQUIN(R) oral capsules, 2013).
    4) A pregnant woman in her third trimester was diagnosed with babesiosis and treated with quinine 650 mg three times daily. Therapy lasted seven days. Two weeks later, she delivered a healthy, 2.7 kg baby girl (Feder et al, 2003).
    C) ANIMAL STUDIES
    1) In animal studies with quinine at doses in the range of the maximum human recommended dose (MHRD) given subQ or IM, teratogenic effects have been demonstrated. In rabbits administered maternal doses of 100 mg/kg/day (approximately 0.5 times the MHRD based on body surface area (BSA)) or higher, there were increases in utero death. In rabbits administered 130 mg/kg/day (approximately 1.3 times the MHRD based on BSA), increased rates of degenerated auditory nerve and spiral ganglion, and CNS anomalies such as anencephaly and microcephaly occurred. In dogs administered doses of 15 mg/kg/day (approximately 0.25 times the MHRD based on BSA) increases in utero death were reported. In guinea pigs administered maternal doses of 200 mg/kg (approximately 1.4 times the MHRD based on BSA), increased rates of hemorrhage and mitochondrial change in cochlea occurred in offspring. However, in rats and monkeys, no teratogenic effects were demonstrated at maternal doses up to 300 mg/kg/day (approximately equal to the MHRD based on BSA) and 200 mg/kg/day (approximately 2 times the MHRD based on BSA), respectively. In rats, maternal oral doses of 20 mg/kg/day (approximately 0.1 times the MHRD based on BSA) resulted in offspring with impaired growth, lower body weights at birth and during lactation, and delayed physical development of teeth eruption and eye opening during lactation (Prod Info QUALAQUIN(R) oral capsules, 2013).
    3.20.3) EFFECTS IN PREGNANCY
    A) PREGNANCY CATEGORY
    1) The manufacturer has classified quinine as FDA pregnancy category C (Prod Info QUALAQUIN(R) oral capsules, 2013)
    2) Pregnant women with P falciparum malaria have a higher risk of morbidity and mortality compared with pregnant women in the general population. Malaria during pregnancy has resulted in increased rates of fetal loss (ie, spontaneous abortion and stillbirth), preterm labor and delivery, intrauterine growth retardation, low birth weight and maternal death (Prod Info QUALAQUIN(R) oral capsules, 2013).
    B) PLACENTAL BARRIER
    1) Quinine crosses the placental barrier and yields measurable blood concentrations in the fetus. Placental cord plasma quinine concentrations were between 1 and 4.6 mg/L (mean of 2.4 mg/mL) and the mean ratio of cord plasma to maternal plasma quinine concentrations was 0.32 +/- 0.14 in 8 women who delivered live infants between 1 and 6 days subsequent to quinine therapy initiation (Prod Info QUALAQUIN(R) oral capsules, 2013).
    C) SPONTANEOUS ABORTION
    1) The incidence of spontaneous abortion was 3.1 times lower (confidence interval 95%; 2.1 to 4.7) for the quinine group (3.5%) compared with the control group (10.9%) in a 1999 study of pregnant Thailand women who either had P. falciparum malaria and were treated with quinine sulfate 10 mg/kg 3 times daily for 7 days at any point during pregnancy (n=633) or did not have malaria and were not exposed to antimalarial drugs during pregnancy (n=2201) (Prod Info QUALAQUIN(R) oral capsules, 2013).
    D) HYPOGLYCEMIA
    1) Hypoglycemia resulting from increased pancreatic secretion of insulin, has been associated with quinine use, particularly during pregnancy (Prod Info QUALAQUIN(R) oral capsules, 2013). Intravenous quinine hydrochloride therapy was associated with the occurrence of hypoglycemia and hyperinsulinemia in 7 of 12 women with severe falciparum malaria during the third trimester of pregnancy (Looareesuwan et al, 1985).
    E) LACK OF EFFECT
    1) STILLBIRTH- The incidence of stillbirth at greater than 28 weeks gestation was not significantly different for the quinine group (1.6%) compared with the control group (1.8%) in a 1999 study of offspring of pregnant Thai women who either had P. falciparum malaria and were treated with quinine sulfate 10 mg/kg 3 times daily for 7 days at any point during pregnancy (n=633) or did not have malaria and were not exposed to antimalarial drugs during pregnancy (n=2201) (Prod Info QUALAQUIN(R) oral capsules, 2013).
    2) A study describing a 7-day course with a combination of quinine sulfate 10 mg/kg every 8 hours plus clindamycin 5 mg/kg every 8 hours included 65 pregnant Thai women with acute, uncomplicated falciparum malaria in the second and third trimesters. Pregnancy outcomes were available for 118 of the 129 women treated with quinine/clindamycin (with a comparator group treated with artesunate); 115 were singleton births, with 1 stillbirth in each group, unrelated to drug exposure. There were no differences in birthweight, placental weight, gestational ages, or proportions of low birthweight. Seventy-two of the 115 infants had 1-year follow-up with no differences between treatment exposure for milestone ages for sitting, crawling or walking (McGready et al, 2001).
    3.20.4) EFFECTS DURING BREAST-FEEDING
    A) BREAST MILK
    1) In 1 study of 25 lactating women being treated with quinine 10 mg/kg every 8 hours for 1 to 10 days, toxicity was not observed and the estimated amount that nursing infants received from the maternal quinine dose was less than 2 to 3 mg/day (less than 0.4%) via breast milk (Prod Info QUALAQUIN(R) oral capsules, 2013).
    3.20.5) FERTILITY
    A) SPERM
    1) Sperm motility was decreased and the proportion of sperm with abnormal morphology was increased in a published study in 5 men who received quinine 600 mg three times daily (Prod Info QUALAQUIN(R) oral capsules, 2013).
    B) ANIMAL STUDIES
    1) In animal studies, quinine produced testicular toxicity in mice at a single intraperitoneal dose approximately 0.75 times the maximum recommended human dose (MRHD) of 32 mg/kg/day and in rats at an IM dose approximately 0.05 times the daily MRHD based on body surface area (Prod Info QUALAQUIN(R) oral capsules, 2013).

Monitoring Parameters Levels

    4.1.1) SUMMARY
    A) Specific laboratory studies for confirming quinine ingestion are not readily available in most clinical laboratories, and are not generally useful for guiding therapy.
    B) Monitor serum electrolytes, BUN, creatinine, glucose, CBC, CPK, urinalysis, and ECG.
    C) In the acutely ill patient, blood gas analysis may demonstrate metabolic acidosis. Lactate is also usually elevated.
    4.1.2) SERUM/BLOOD
    A) BLOOD/SERUM CHEMISTRY
    1) TOXIC LEVELS: Plasma concentrations over 5 mcg/mL have been associated with cinchonism. Levels above 10 mcg/mL are associated with visual impairment, and levels above 16 mcg/mL are associated with cardiac dysrhythmias (Dyson & Proudfoot, 1985; Boland et al, 1985; White et al, 1982; Bateman et al, 1985).
    2) Specific laboratory studies for confirming quinine ingestion are not readily available in most clinical laboratories, and are not generally useful for guiding therapy.
    3) Monitor serum electrolytes, BUN, creatinine, glucose, CBC, and CPK.
    B) ACID/BASE
    1) In the acutely ill patient, blood gas analysis may demonstrate metabolic acidosis. Lactate is also usually elevated.
    4.1.4) OTHER
    A) OTHER
    1) ECG
    a) Obtain an ECG and institute continuous cardiac monitoring.

Methods

    A) MULTIPLE ANALYTICAL METHODS
    1) Usually analyzed in biologic fluid by fluorometry after protein precipitation (Brodie & Undenfriend, 1943) or after solvent extraction (Hall et al, 1973).
    2) Other methods include ultraviolet spectrophotometry (measured at 250 nm) and thin-layer chromatography (Clarke, 1969).

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 experiencing clinical manifestations of quinine overdose (ie, altered mental status, hemodynamic instability, or vision changes) should be hospitalized in an intensive care facility with immediate attention given to vital signs (blood pressure, pulse, respiration) and careful monitoring of ECG, neurological status, and respiratory status. The patient should remain in intensive care until symptoms have resolved and the ECG has returned to normal. This may take approximately 24 to 48 hours (Huston & Levinson, 2006).
    B) Patients should be warned of possible blindness but reassured that some recovery of sight frequently occurs.
    1) A 44-year-old man who ingested 6.8 g of quinine sulfate 5 hours before admission, became totally blind 1 hour before admission. Following gastric lavage acid diuresis and bilateral stellate ganglion block some light perception returned.
    a) No improvement was noted in the next 3 hours. Concurrent resin hemoperfusion and hemodialysis for the next 6 hours was associated with return to almost normal vision (Gibbs et al, 1985).
    b) This report, however, may be simply an observation on the natural course of amblyopia after quinine overdose with onset after 4 to 10 hours and partial or total reversal after 12 to 18 hours (Heath, 1985).
    6.3.1.2) HOME CRITERIA/ORAL
    A) Home management is only indicated if the patient is asymptomatic, ingestion was minimal (therapeutic dose or less) and there is no suspicion for self-harm.
    6.3.1.3) CONSULT CRITERIA/ORAL
    A) Consult ophthalmology if the patient has any visual disturbances. Involve a toxicologist or poison center immediately if there is concern for intentional or significant overdose.
    6.3.1.4) PATIENT TRANSFER/ORAL
    A) The patient should remain under careful close observation until:
    1) There is no evidence of central nervous system toxicity such as seizures or CNS depression,
    2) Acid-base disturbances are corrected,
    3) Electrolytes are normal,
    4) The electrocardiogram has returned to baseline,
    5) And there is no hypoglycemia.
    6.3.1.5) OBSERVATION CRITERIA/ORAL
    A) Any patient with an intentional ingestion, or a significant ingestion greater than weight/age appropriate dosing, should be observed for 6 to 8 hours or until asymptomatic.
    B) For children who ingest quinine, the weight based and age based therapeutic dose should be considered. If the ingested amount exceeds the therapeutic dose or if the exact amount ingested is unknown, then the child should be evaluated in an emergency department as well as receive cardiac monitoring. The child should be observed for 6 hours after the time of ingestion (Huston & Levinson, 2006).
    C) All symptomatic patients should remain in intensive care until clinical manifestations of the overdose have resolved and the ECG has returned to normal. This may take approximately 24 to 48 hours (Huston & Levinson, 2006).
    D) Patients may require ophthalmologic follow-up for many months after discharge from the hospital.
    E) Patients should be warned of possible blindness, but reassured that some recovery of sight frequently occurs.

Monitoring

    A) Specific laboratory studies for confirming quinine ingestion are not readily available in most clinical laboratories, and are not generally useful for guiding therapy.
    B) Monitor serum electrolytes, BUN, creatinine, glucose, CBC, CPK, urinalysis, and ECG.
    C) In the acutely ill patient, blood gas analysis may demonstrate metabolic acidosis. Lactate is also usually elevated.

Oral Exposure

    6.5.1) PREVENTION OF ABSORPTION/PREHOSPITAL
    A) ACTIVATED CHARCOAL
    1) PREHOSPITAL ACTIVATED CHARCOAL ADMINISTRATION
    a) Consider prehospital administration of activated charcoal as an aqueous slurry in patients with a potentially toxic ingestion who are awake and able to protect their airway. Activated charcoal is most effective when administered within one hour of ingestion. Administration in the prehospital setting has the potential to significantly decrease the time from toxin ingestion to activated charcoal administration, although it has not been shown to affect outcome (Alaspaa et al, 2005; Thakore & Murphy, 2002; Spiller & Rogers, 2002).
    1) In patients who are at risk for the abrupt onset of seizures or mental status depression, activated charcoal should not be administered in the prehospital setting, due to the risk of aspiration in the event of spontaneous emesis.
    2) The addition of flavoring agents (cola drinks, chocolate milk, cherry syrup) to activated charcoal improves the palatability for children and may facilitate successful administration (Guenther Skokan et al, 2001; Dagnone et al, 2002).
    2) CHARCOAL DOSE
    a) Use a minimum of 240 milliliters of water per 30 grams charcoal (FDA, 1985). Optimum dose not established; usual dose is 25 to 100 grams in adults and adolescents; 25 to 50 grams in children aged 1 to 12 years (or 0.5 to 1 gram/kilogram body weight) ; and 0.5 to 1 gram/kilogram in infants up to 1 year old (Chyka et al, 2005).
    1) Routine use of a cathartic with activated charcoal is NOT recommended as there is no evidence that cathartics reduce drug absorption and cathartics are known to cause adverse effects such as nausea, vomiting, abdominal cramps, electrolyte imbalances and occasionally hypotension (None Listed, 2004).
    b) ADVERSE EFFECTS/CONTRAINDICATIONS
    1) Complications: emesis, aspiration (Chyka et al, 2005). Aspiration may be complicated by acute respiratory failure, ARDS, bronchiolitis obliterans or chronic lung disease (Golej et al, 2001; Graff et al, 2002; Pollack et al, 1981; Harris & Filandrinos, 1993; Elliot et al, 1989; Rau et al, 1988; Golej et al, 2001; Graff et al, 2002). Refer to the ACTIVATED CHARCOAL/TREATMENT management for further information.
    2) Contraindications: unprotected airway (increases risk/severity of aspiration) , nonfunctioning gastrointestinal tract, uncontrolled vomiting, and ingestion of most hydrocarbons (Chyka et al, 2005).
    6.5.2) PREVENTION OF ABSORPTION
    A) ACTIVATED CHARCOAL
    1) CHARCOAL ADMINISTRATION
    a) Consider administration of activated charcoal after a potentially toxic ingestion (Chyka et al, 2005). Administer charcoal as an aqueous slurry; most effective when administered within one hour of ingestion.
    2) CHARCOAL DOSE
    a) Use a minimum of 240 milliliters of water per 30 grams charcoal (FDA, 1985). Optimum dose not established; usual dose is 25 to 100 grams in adults and adolescents; 25 to 50 grams in children aged 1 to 12 years (or 0.5 to 1 gram/kilogram body weight) ; and 0.5 to 1 gram/kilogram in infants up to 1 year old (Chyka et al, 2005).
    1) Routine use of a cathartic with activated charcoal is NOT recommended as there is no evidence that cathartics reduce drug absorption and cathartics are known to cause adverse effects such as nausea, vomiting, abdominal cramps, electrolyte imbalances and occasionally hypotension (None Listed, 2004).
    b) ADVERSE EFFECTS/CONTRAINDICATIONS
    1) Complications: emesis, aspiration (Chyka et al, 2005). Aspiration may be complicated by acute respiratory failure, ARDS, bronchiolitis obliterans or chronic lung disease (Golej et al, 2001; Graff et al, 2002; Pollack et al, 1981; Harris & Filandrinos, 1993; Elliot et al, 1989; Rau et al, 1988; Golej et al, 2001; Graff et al, 2002). Refer to the ACTIVATED CHARCOAL/TREATMENT management for further information.
    2) Contraindications: unprotected airway (increases risk/severity of aspiration) , nonfunctioning gastrointestinal tract, uncontrolled vomiting, and ingestion of most hydrocarbons (Chyka et al, 2005).
    B) GASTRIC LAVAGE
    1) INDICATIONS: Consider gastric lavage with a large-bore orogastric tube (ADULT: 36 to 40 French or 30 English gauge tube {external diameter 12 to 13.3 mm}; CHILD: 24 to 28 French {diameter 7.8 to 9.3 mm}) after a potentially life threatening ingestion if it can be performed soon after ingestion (generally within 60 minutes).
    a) Consider lavage more than 60 minutes after ingestion of sustained-release formulations and substances known to form bezoars or concretions.
    2) PRECAUTIONS:
    a) SEIZURE CONTROL: Is mandatory prior to gastric lavage.
    b) AIRWAY PROTECTION: Place patients in the head down left lateral decubitus position, with suction available. Patients with depressed mental status should be intubated with a cuffed endotracheal tube prior to lavage.
    3) LAVAGE FLUID:
    a) Use small aliquots of liquid. Lavage with 200 to 300 milliliters warm tap water (preferably 38 degrees Celsius) or saline per wash (in older children or adults) and 10 milliliters/kilogram body weight of normal saline in young children(Vale et al, 2004) and repeat until lavage return is clear.
    b) The volume of lavage return should approximate amount of fluid given to avoid fluid-electrolyte imbalance.
    c) CAUTION: Water should be avoided in young children because of the risk of electrolyte imbalance and water intoxication. Warm fluids avoid the risk of hypothermia in very young children and the elderly.
    4) COMPLICATIONS:
    a) Complications of gastric lavage have included: aspiration pneumonia, hypoxia, hypercapnia, mechanical injury to the throat, esophagus, or stomach, fluid and electrolyte imbalance (Vale, 1997). Combative patients may be at greater risk for complications (Caravati et al, 2001).
    b) Gastric lavage can cause significant morbidity; it should NOT be performed routinely in all poisoned patients (Vale, 1997).
    5) CONTRAINDICATIONS:
    a) Loss of airway protective reflexes or decreased level of consciousness if patient is not intubated, following ingestion of corrosive substances, hydrocarbons (high aspiration potential), patients at risk of hemorrhage or gastrointestinal perforation, or trivial or non-toxic ingestion.
    C) MULTIPLE-DOSE ACTIVATED CHARCOAL
    1) Despite quinine's relatively large volume of distribution, high protein binding, and poor in vitro adsorption by charcoal (White et al, 1982; Corby & Decker, 1974), studies have shown enhanced elimination with multiple-dose-charcoal regimens in therapeutic doses and overdoses (Lockey & Bateman, 1989; Prescott et al, 1989). It should be considered in patients with potentially life threatening overdose.
    a) Multiple dose charcoal has not been shown to affect outcome after quinine overdose. It is recommended in patients with severe toxicity and those with rising levels despite initial decontamination.
    b) Lockey & Bateman (1989) demonstrated an increase of oral clearance by 56% and decrease in half-life from 8.23 to 4.55 hours after therapeutic doses of quinine and MDC 50 grams every 4 hours (Lockey & Bateman, 1989).
    c) In a study of quinine overdose patients, the same regimen of MDC resulted in a mean half-life of 8.1 hours, compared to 26 hours in previous reports of overdosed patients (Prescott et al, 1989).
    d) A half-life of 33 hours prior to charcoal decreased to 10 hours after charcoal administration in a single patient (Prescott et al, 1989).
    2) MULTIPLE DOSE ACTIVATED CHARCOAL
    a) ADULT DOSE: Optimal dose not established. After an initial dose of 50 to 100 grams of activated charcoal, subsequent doses may be administered every 1, 2 or 4 hours at a dose equivalent to 12.5 grams/hour (Vale et al, 1999), do not exceed: 0.5 g/kg charcoal every 2 hours (Ghannoum & Gosselin, 2013; Mauro et al, 1994). There is some evidence that smaller more frequent doses are more effective at enhancing drug elimination than larger less frequent doses (Park et al, 1983; Ilkhanipour et al, 1992). PEDIATRIC DOSE: Optimal dose not established. After an initial dose of 25 to 50 grams in children aged 1 to 12 years (or 0.5 to 1 gram/kilogram body weight) (Chyka & Seger, 1997), subsequent doses may be administered every 1, 2 or 4 hours (Vale et al, 1999) in a dose equivalent to 6.25 grams/hour in children 1 to 12 years old.
    b) Activated charcoal should be continued until the patient's clinical and laboratory parameters, including drug concentrations if available, are improving (Vale et al, 1999). The patient should be frequently assessed for the ability to protect the airway and evidence of decreased peristalsis or intestinal obstruction.
    c) Use of cathartics has not been shown to increase drug elimination and may increase the likelihood of vomiting. Routine coadministration of a cathartic is NOT recommended (Vale et al, 1999).
    d) AGENTS AMENABLE TO MDAC THERAPY: The following properties of a drug that are likely to allow MDAC therapy to be effective include: small volume of distribution, low protein binding, prolonged half-life, low intrinsic clearance, and a nonionized state at physiologic pH (Chyka, 1995; Ghannoum & Gosselin, 2013).
    e) Vomiting is a common adverse effect; antiemetics may be necessary.
    f) CONTRAINDICATIONS: Absolute contraindications include an unprotected airway, intestinal obstruction, a gastrointestinal tract that is not intact and agents that may increase the risk of aspiration (eg, hydrocarbons). Relative contraindications include decreased peristalsis (eg, decreased bowel sounds, abdominal distention, ileus, severe constipation) (Vale et al, 1999; Mauro et al, 1994).
    g) COMPLICATIONS: Include constipation, intestinal bleeding, bowel obstruction, appendicitis, charcoal bezoars, and aspiration which may be complicated by acute respiratory failure, adult respiratory distress syndrome or bronchiolitis obliterans (Ghannoum & Gosselin, 2013; Ray et al, 1988; Atkinson et al, 1992; Gomez et al, 1994; Mizutani et al, 1991; Pollack et al, 1981; Harris & Filandrinos, 1993; Elliot et al, 1989; Mina et al, 2002; Harsch, 1986; Rau et al, 1988; Golej et al, 2001; Graff et al, 2002).
    6.5.3) TREATMENT
    A) MONITORING OF PATIENT
    1) Specific laboratory studies for confirming quinine ingestion are not readily available in most clinical laboratories, and are not generally useful for guiding therapy.
    2) Monitor serum electrolytes, BUN, creatinine, glucose, CBC, CPK, urinalysis, and ECG.
    3) In the acutely ill patient, blood gas analysis may demonstrate metabolic acidosis. Lactate is also usually elevated.
    B) SEIZURE
    1) SUMMARY
    a) Attempt initial control with a benzodiazepine (eg, diazepam, lorazepam). If seizures persist or recur, administer phenobarbital or propofol.
    b) Monitor for respiratory depression, hypotension, and dysrhythmias. Endotracheal intubation should be performed in patients with persistent seizures.
    c) Evaluate for hypoxia, electrolyte disturbances, and hypoglycemia (or, if immediate bedside glucose testing is not available, treat with intravenous dextrose).
    2) DIAZEPAM
    a) ADULT DOSE: Initially 5 to 10 mg IV, OR 0.15 mg/kg IV up to 10 mg per dose up to a rate of 5 mg/minute; may be repeated every 5 to 20 minutes as needed (Brophy et al, 2012; Prod Info diazepam IM, IV injection, 2008; Manno, 2003).
    b) PEDIATRIC DOSE: 0.1 to 0.5 mg/kg IV over 2 to 5 minutes; up to a maximum of 10 mg/dose. May repeat dose every 5 to 10 minutes as needed (Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008).
    c) Monitor for hypotension, respiratory depression, and the need for endotracheal intubation. Consider a second agent if seizures persist or recur after repeated doses of diazepam .
    3) NO INTRAVENOUS ACCESS
    a) DIAZEPAM may be given rectally or intramuscularly (Manno, 2003). RECTAL DOSE: CHILD: Greater than 12 years: 0.2 mg/kg; 6 to 11 years: 0.3 mg/kg; 2 to 5 years: 0.5 mg/kg (Brophy et al, 2012).
    b) MIDAZOLAM has been used intramuscularly and intranasally, particularly in children when intravenous access has not been established. ADULT DOSE: 0.2 mg/kg IM, up to a maximum dose of 10 mg (Brophy et al, 2012). PEDIATRIC DOSE: INTRAMUSCULAR: 0.2 mg/kg IM, up to a maximum dose of 7 mg (Chamberlain et al, 1997) OR 10 mg IM (weight greater than 40 kg); 5 mg IM (weight 13 to 40 kg); INTRANASAL: 0.2 to 0.5 mg/kg up to a maximum of 10 mg/dose (Loddenkemper & Goodkin, 2011; Brophy et al, 2012). BUCCAL midazolam, 10 mg, has been used in adolescents and older children (5-years-old or more) to control seizures when intravenous access was not established (Scott et al, 1999).
    4) LORAZEPAM
    a) MAXIMUM RATE: The rate of intravenous administration of lorazepam should not exceed 2 mg/min (Brophy et al, 2012; Prod Info lorazepam IM, IV injection, 2008).
    b) ADULT DOSE: 2 to 4 mg IV initially; repeat every 5 to 10 minutes as needed, if seizures persist (Manno, 2003; Brophy et al, 2012).
    c) PEDIATRIC DOSE: 0.05 to 0.1 mg/kg IV over 2 to 5 minutes, up to a maximum of 4 mg/dose; may repeat in 5 to 15 minutes as needed, if seizures continue (Brophy et al, 2012; Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008; Sreenath et al, 2009; Chin et al, 2008).
    5) PHENOBARBITAL
    a) ADULT LOADING DOSE: 20 mg/kg IV at an infusion rate of 50 to 100 mg/minute IV. An additional 5 to 10 mg/kg dose may be given 10 minutes after loading infusion if seizures persist or recur (Brophy et al, 2012).
    b) Patients receiving high doses will require endotracheal intubation and may require vasopressor support (Brophy et al, 2012).
    c) PEDIATRIC LOADING DOSE: 20 mg/kg may be given as single or divided application (2 mg/kg/minute in children weighing less than 40 kg up to 100 mg/min in children weighing greater than 40 kg). A plasma concentration of about 20 mg/L will be achieved by this dose (Loddenkemper & Goodkin, 2011).
    d) REPEAT PEDIATRIC DOSE: Repeat doses of 5 to 20 mg/kg may be given every 15 to 20 minutes if seizures persist, with cardiorespiratory monitoring (Loddenkemper & Goodkin, 2011).
    e) MONITOR: For hypotension, respiratory depression, and the need for endotracheal intubation (Loddenkemper & Goodkin, 2011; Manno, 2003).
    f) SERUM CONCENTRATION MONITORING: Monitor serum concentrations over the next 12 to 24 hours. Therapeutic serum concentrations of phenobarbital range from 10 to 40 mcg/mL, although the optimal plasma concentration for some individuals may vary outside this range (Hvidberg & Dam, 1976; Choonara & Rane, 1990; AMA Department of Drugs, 1992).
    6) OTHER AGENTS
    a) If seizures persist after phenobarbital, propofol or pentobarbital infusion, or neuromuscular paralysis with general anesthesia (isoflurane) and continuous EEG monitoring should be considered (Manno, 2003). Other anticonvulsants can be considered (eg, valproate sodium, levetiracetam, lacosamide, topiramate) if seizures persist or recur; however, there is very little data regarding their use in toxin induced seizures, controlled trials are not available to define the optimal dosage ranges for these agents in status epilepticus (Brophy et al, 2012):
    1) VALPROATE SODIUM: ADULT DOSE: An initial dose of 20 to 40 mg/kg IV, at a rate of 3 to 6 mg/kg/minute; may give an additional dose of 20 mg/kg 10 minutes after loading infusion. PEDIATRIC DOSE: 1.5 to 3 mg/kg/minute (Brophy et al, 2012).
    2) LEVETIRACETAM: ADULT DOSE: 1000 to 3000 mg IV, at a rate of 2 to 5 mg/kg/min IV. PEDIATRIC DOSE: 20 to 60 mg/kg IV (Brophy et al, 2012; Loddenkemper & Goodkin, 2011).
    3) LACOSAMIDE: ADULT DOSE: 200 to 400 mg IV; 200 mg IV over 15 minutes (Brophy et al, 2012). PEDIATRIC DOSE: In one study, median starting doses of 1.3 mg/kg/day and maintenance doses of 4.7 mg/kg/day were used in children 8 years and older (Loddenkemper & Goodkin, 2011).
    4) TOPIRAMATE: ADULT DOSE: 200 to 400 mg nasogastric/orally OR 300 to 1600 mg/day orally divided in 2 to 4 times daily (Brophy et al, 2012).
    C) VENTRICULAR ARRHYTHMIA
    1) Treat QRS widening and/or ventricular dysrhythmias with intravenous sodium bicarbonate. A reasonable starting dose is 1 to 2 mEq/kilogram as an intravenous bolus. Repeat as necessary to maintain arterial pH 7.45 to 7.55. Monitor arterial blood gases, serum electrolytes, and serial ECGs and institute continuous cardiac monitoring. Avoid Class 1A antiarrhythmics such as disopyramide and procainamide. Phenytoin is theoretically preferable since it increases AV conduction.
    2) Treatment of ventricular tachycardia (especially torsade de pointes variant) may require D-C cardioversion, overdrive pacing (Anderson & Mason, 1978), or isoproterenol infusion to decrease temporal dispersion in refractoriness.
    D) TORSADES DE POINTES
    1) SUMMARY
    a) Withdraw the causative agent. Hemodynamically unstable patients with Torsades de pointes (TdP) require electrical cardioversion. Emergent treatment with magnesium (first-line agent) or atrial overdrive pacing is indicated. Detect and correct underlying electrolyte abnormalities (ie, hypomagnesemia, hypokalemia, hypocalcemia). Correct hypoxia, if present (Drew et al, 2010; Neumar et al, 2010; Keren et al, 1981; Smith & Gallagher, 1980).
    b) Polymorphic VT associated with acquired long QT syndrome may be treated with IV magnesium. Overdrive pacing or isoproterenol may be successful in terminating TdP, particularly when accompanied by bradycardia or if TdP appears to be precipitated by pauses in rhythm (Neumar et al, 2010). In patients with polymorphic VT with a normal QT interval, magnesium is unlikely to be effective (Link et al, 2015).
    2) MAGNESIUM SULFATE
    a) Magnesium is recommended (first-line agent) for the prevention and treatment of drug-induced torsades de pointes (TdP) even if the serum magnesium concentration is normal. QTc intervals greater than 500 milliseconds after a potential drug overdose may correlate with the development of TdP (Charlton et al, 2010; Drew et al, 2010). ADULT DOSE: No clearly established guidelines exist; an optimal dosing regimen has not been established. Administer 1 to 2 grams diluted in 10 milliliters D5W IV/IO over 15 minutes (Neumar et al, 2010). Followed if needed by a second 2 gram bolus and an infusion of 0.5 to 1 gram (4 to 8 mEq) per hour in patients not responding to the initial bolus or with recurrence of dysrhythmias (American Heart Association, 2005; Perticone et al, 1997). Rate of infusion may be increased if dysrhythmias recur. For persistent refractory dysrhythmias, a continuous infusion of up to 3 to 10 milligrams/minute in adults may be given (Charlton et al, 2010).
    b) PEDIATRIC DOSE: 25 to 50 milligrams/kilogram diluted to 10 milligrams/milliliter for intravenous infusion over 5 to 15 minutes up to 2 g (Charlton et al, 2010).
    c) PRECAUTIONS: Use with caution in patients with renal insufficiency.
    d) MAJOR ADVERSE EFFECTS: High doses may cause hypotension, respiratory depression, and CNS toxicity (Neumar et al, 2010). Toxicity may be observed at magnesium levels of 3.5 to 4.0 mEq/L or greater (Charlton et al, 2010).
    e) MONITORING PARAMETERS: Monitor heart rate and rhythm, blood pressure, respiratory rate, motor strength, deep tendon reflexes, serum magnesium, phosphorus, and calcium concentrations (Prod Info magnesium sulfate heptahydrate IV, IM injection, solution, 2009).
    3) OVERDRIVE PACING
    a) Institute electrical overdrive pacing at a rate of 130 to 150 beats per minute, and decrease as tolerated. Rates of 100 to 120 beats per minute may terminate torsades (American Heart Association, 2005). Pacing can be used to suppress self-limited runs of TdP that may progress to unstable or refractory TdP, or for override refractory, persistent TdP before the potential development of ventricular fibrillation (Charlton et al, 2010). In a case series overdrive pacing was successful in terminating TdP associated with bradycardia and drug-induced QT prolongation (Neumar et al, 2010).
    4) POTASSIUM REPLETION
    a) Potassium supplementation, even if serum potassium is normal, has been recommended by many experts (Charlton et al, 2010; American Heart Association, 2005). Supplementation to supratherapeutic potassium concentrations of 4.5 to 5 mmol/L has been suggested, although there is little evidence to determine the optimal range in dysrhythmia (Drew et al, 2010; Charlton et al, 2010).
    5) ISOPROTERENOL
    a) Isoproterenol has been successful in aborting torsades de pointes that was resistant to magnesium therapy in a patient in whom transvenous overdrive pacing was not an option (Charlton et al, 2010) and has been successfully used to treat torsades de pointes associated with bradycardia and drug induced QT prolongation (Keren et al, 1981; Neumar et al, 2010). Isoproterenol may have a limited role in pharmacologic overdrive pacing in select patients with drug-induced torsades de pointes and acquired long QT syndrome (Charlton et al, 2010; Neumar et al, 2010). Isoproterenol should be avoided in patients with polymorphic VT associated with familial long QT syndrome (Neumar et al, 2010).
    b) DOSE: ADULT: 2 to 10 micrograms/minute via a continuous monitored intravenous infusion; titrate to heart rate and rhythm response (Neumar et al, 2010).
    c) PRECAUTIONS: Correct hypovolemia before using; contraindicated in patients with acute cardiac ischemia (Prod Info Isuprel(TM) intravenous injection, intramuscular injection, subcutaneous injection, intracardiac injection, 2013).
    1) Contraindicated in patients with preexisting dysrhythmias; tachycardia or heart block due to digitalis toxicity; ventricular dysrhythmias that require inotropic therapy; and angina. Use with caution in patients with coronary insufficiency (Prod Info Isuprel(TM) intravenous injection, intramuscular injection, subcutaneous injection, intracardiac injection, 2013).
    d) MAJOR ADVERSE EFFECTS: Tachycardia, cardiac dysrhythmias, palpitations, hypotension or hypertension, nervousness, headache, dizziness, and dyspnea (Prod Info Isuprel(TM) intravenous injection, intramuscular injection, subcutaneous injection, intracardiac injection, 2013).
    e) MONITORING PARAMETERS: Monitor heart rate and rhythm, blood pressure, respirations and central venous pressure to guide volume replacement (Prod Info Isuprel(TM) intravenous injection, intramuscular injection, subcutaneous injection, intracardiac injection, 2013).
    6) OTHER DRUGS
    a) Mexiletine, verapamil, propranolol, and labetalol have also been used to treat TdP, but results have been inconsistent (Khan & Gowda, 2004).
    7) AVOID
    a) Avoid class Ia antidysrhythmics (eg, quinidine, disopyramide, procainamide, aprindine), class Ic (eg, flecainide, encainide, propafenone) and most class III antidysrhythmics (eg, N-acetylprocainamide, sotalol) since they may further prolong the QT interval and have been associated with TdP.
    E) HYPOTENSIVE EPISODE
    1) Pure or predominant alpha agonists may be more effective in managing hypotension. These include norepinephrine or metaraminol.
    2) Isoproterenol may be used to treat refractory bradycardia or heart block with slow ventricular response, while a temporary pacemaker is being inserted.
    3) The MAST suit may be beneficial (Hoffman, 1983) as a temporary measure for hypotension by increasing systemic vascular resistance. Favorable results using an intraaortic balloon pump for refractory hypotension has also been reported (Shub et al, 1978).
    4) NOREPINEPHRINE
    a) PREPARATION: 4 milligrams (1 amp) added to 1000 milliliters of diluent provides a concentration of 4 micrograms/milliliter of norepinephrine base. Norepinephrine bitartrate should be mixed in dextrose solutions (dextrose 5% in water, dextrose 5% in saline) since dextrose-containing solutions protect against excessive oxidation and subsequent potency loss. Administration in saline alone is not recommended (Prod Info norepinephrine bitartrate injection, 2005).
    b) DOSE
    1) ADULT: Dose range: 0.1 to 0.5 microgram/kilogram/minute (eg, 70 kg adult 7 to 35 mcg/min); titrate to maintain adequate blood pressure (Peberdy et al, 2010).
    2) CHILD: Dose range: 0.1 to 2 micrograms/kilogram/minute; titrate to maintain adequate blood pressure (Kleinman et al, 2010).
    3) CAUTION: Extravasation may cause local tissue ischemia, administration by central venous catheter is advised (Peberdy et al, 2010).
    F) BLINDNESS AND/OR VISION IMPAIRMENT LEVEL
    1) STELLATE GANGLION BLOCK
    a) Blindness usually resolves without treatment. Stellate ganglion block (SGB) has never been proven beneficial in any clinical study and is potentially dangerous. Its use is NOT recommended.
    b) Stellate ganglion block is theorized to improve regional blood supply (Valman & White, 1977; Boscoe et al, 1983; Thomas, 1984).
    c) Since the primary toxic effect of quinine is direct damage of photoelectric cells, any benefit of stellate block is probably minimal or coincident with spontaneous visual return, and the procedure has serious complications (Boland et al, 1985; Dyson & Proudfoot, 1985).
    d) Treatment of quinine overdose including stellate ganglion block had no apparent benefit in a series of 25 patients (Dyson et al, 1985a). This was recently confirmed (Bacon et al, 1988).
    e) In one case report, a late presentation of ocular quinine toxicity was managed with a combination of vasodilatory treatments (oral nimodipine, clonidine infusion, and SGB) (Vusirikala et al, 2005).
    1) A 39-year-old woman with ocular toxicity (reduced visual acuity, dilated, sluggish pupils, and retinal arterial attenuation), presented late for ophthalmology review 40 hours after taking 28 quinine sulfate tablets (300 mg each). She was treated with nimodipine (60 mg for 6 days), clonidine infusion (300 mcg/24 hours), and stellate ganglion block (SGB) (ropivacaine 1% 10 mL). Since the left eye was amblyopic, it was difficult to quantify the effect of treatment; however, the unilateral block was performed late without complication on the side of the nonamblyopic eye and the right visual acuity improved 48 hours after the treatment (Vusirikala et al, 2005).
    2) NITRATES
    a) Retinal arterial constriction is often noted with quinine toxicity.
    b) Hla et al (1992) treated a case of blindness (16-month-old who ingested 600 mg) with intravenous isosorbide dinitrate. An ophthalmological review 6 weeks post discharge showed full restoration of sight.
    c) A 36-year-old man presented with blindness 8 to 10 hours after quinine overdose (Moore et al, 1992). There was some evidence of visual recovery 1 to 2 hours after intravenous nitrates were begun, and he eventually had total visual recovery.
    1) Since resolution of ocular toxicity is spontaneous in a significant number of patients, it is unclear what role nitrate played in this patient's course.
    3) VASOSPASM THERAPY
    a) In a case of quinine poisoning resulting in bilateral visual loss with retinal arteriolar constriction, fluctuating visual loss suggested an element of vasospasm. The authors chose to use IV nimodipine (0.01 mg/kg/hr) for vasospasm, and IV hydration with 0.9% saline to maintain a central venous pressure of at least 9 mmHg. In addition, IV noradrenaline was administered to maintain a systolic blood pressure of 140 to 180 mmHg. Over the next 24 hours, the patient's vision improved to 6/9 bilaterally and did not deteriorate again. Within 12 hours of the therapy, retinal blood flow was noted to be improved on direct fundoscopy (Barrett & Solano, 2002).

Enhanced Elimination

    A) DIURESIS
    1) From 5% to 20% of quinine is excreted unchanged in the urine. In one case report (Sabto et al, 1981) forced diuresis over 75 hours accounted for removal of about 18% of a 9 g dose. The real danger of fluid overload with the myocardial depressant effects of quinine do NOT allow forced acid diuresis to be generally recommended, although some may have a slight benefit with increased urine flow.
    B) HEMODIALYSIS
    1) Hemodialysis has shown minimal effectiveness and peritoneal dialysis almost none.
    a) In one report, hemodialysis removed only 30 mg and 26 mg of quinine (Sabto et al, 1981).
    b) In another report, peritoneal dialysis removed 1.6 mg/hour over the first 48 hours and a 2L plasma exchange procedure (plasmapheresis) removed 8.5 mg (Floyd et al, 1974).
    c) Combined hemodialysis and hemoperfusion were not shown to be effective in enhancing elimination after ingestion of 6 g (Martin et al, 1992).
    d) None of these modalities can be currently recommended.
    C) HEMOPERFUSION
    1) Hemoperfusion is also ineffective in increasing quinine elimination (Morgan et al, 1983; Bateman et al, 1985; Heath, 1985).
    D) MULTIPLE-DOSE ACTIVATED CHARCOAL
    1) Despite quinine's relatively large volume of distribution, high protein binding, and poor in vitro adsorption by charcoal (White et al, 1982; Corby & Decker, 1974), studies have shown enhanced elimination with multiple-dose-charcoal regimens in therapeutic doses and overdoses (Lockey & Bateman, 1989; Prescott et al, 1989). It should be considered in patients with potentially life threatening overdose.
    a) Multiple dose charcoal has not been shown to affect outcome after quinine overdose. It is recommended in patients with severe toxicity and those with rising levels despite initial decontamination.
    b) Lockey & Bateman (1989) demonstrated an increase of oral clearance by 56% and decrease in half-life from 8.23 to 4.55 hours after therapeutic doses of quinine and MDC 50 g every 4 hours (Lockey & Bateman, 1989).
    c) In a study of quinine overdose patients, the same regimen of MDC resulted in a mean half-life of 8.1 hours, compared to 26 hours in previous reports of overdosed patients (Prescott et al, 1989).
    d) A half-life of 33 hours prior to charcoal decreased to 10 hours after charcoal administration in a single patient (Prescott et al, 1989).
    2) MULTIPLE DOSE ACTIVATED CHARCOAL
    a) ADULT DOSE: Optimal dose not established. After an initial dose of 50 to 100 grams of activated charcoal, subsequent doses may be administered every 1, 2 or 4 hours at a dose equivalent to 12.5 grams/hour (Vale et al, 1999), do not exceed: 0.5 g/kg charcoal every 2 hours (Ghannoum & Gosselin, 2013; Mauro et al, 1994). There is some evidence that smaller more frequent doses are more effective at enhancing drug elimination than larger less frequent doses (Park et al, 1983; Ilkhanipour et al, 1992). PEDIATRIC DOSE: Optimal dose not established. After an initial dose of 25 to 50 grams in children aged 1 to 12 years (or 0.5 to 1 gram/kilogram body weight) (Chyka & Seger, 1997), subsequent doses may be administered every 1, 2 or 4 hours (Vale et al, 1999) in a dose equivalent to 6.25 grams/hour in children 1 to 12 years old.
    b) Activated charcoal should be continued until the patient's clinical and laboratory parameters, including drug concentrations if available, are improving (Vale et al, 1999). The patient should be frequently assessed for the ability to protect the airway and evidence of decreased peristalsis or intestinal obstruction.
    c) Use of cathartics has not been shown to increase drug elimination and may increase the likelihood of vomiting. Routine coadministration of a cathartic is NOT recommended (Vale et al, 1999).
    d) AGENTS AMENABLE TO MDAC THERAPY: The following properties of a drug that are likely to allow MDAC therapy to be effective include: small volume of distribution, low protein binding, prolonged half-life, low intrinsic clearance, and a nonionized state at physiologic pH (Chyka, 1995; Ghannoum & Gosselin, 2013).
    e) Vomiting is a common adverse effect; antiemetics may be necessary.
    f) CONTRAINDICATIONS: Absolute contraindications include an unprotected airway, intestinal obstruction, a gastrointestinal tract that is not intact and agents that may increase the risk of aspiration (eg, hydrocarbons). Relative contraindications include decreased peristalsis (eg, decreased bowel sounds, abdominal distention, ileus, severe constipation) (Vale et al, 1999; Mauro et al, 1994).
    g) COMPLICATIONS: Include constipation, intestinal bleeding, bowel obstruction, appendicitis, charcoal bezoars, and aspiration which may be complicated by acute respiratory failure, adult respiratory distress syndrome or bronchiolitis obliterans (Ghannoum & Gosselin, 2013; Ray et al, 1988; Atkinson et al, 1992; Gomez et al, 1994; Mizutani et al, 1991; Pollack et al, 1981; Harris & Filandrinos, 1993; Elliot et al, 1989; Mina et al, 2002; Harsch, 1986; Rau et al, 1988; Golej et al, 2001; Graff et al, 2002).

Case Reports

    A) ACUTE EFFECTS
    1) CARDIOTOXICITY: Bodenhamer & Smilkstein (1993) reported delayed cardiotoxicity following ingestion of 16,250 mg of quinine in a 49-year-old woman. Symptoms appeared 11.5 hours following ingestion, with conduction abnormalities, ectopy, and torsades de pointes occurring 25 hours after ingestion (Bodenhamer & Smilkstein, 1993).
    2) VISUAL DAMAGE: Two patients, a 38-year-old man and a 60-year-old woman, suffered visual damage following quinine overdoses. The vision loss was only partially reversible (Murray & Jay, 1983). A 26-year-old man developed bilateral loss of vision after ingesting 20 quinine tablets and an unknown amount of alcohol (Drake WM & Hiorns MP, 1994).
    3) FATALITY: A 17-year-old man ingested 5 g of quinine and developed deafness within several hours. Upon presentation, he collapsed and was noted to have broad complex tachycardia, which normalized over 8 hours. The case was complicated with hypokalemia (2.2 mmol/L) and hypoglycemia (2.7 mmol/L). Later, chest x-ray revealed non-cardiogenic pulmonary edema. He was treated with antibiotics and inotropic agents. Renal dysfunction developed on the ninth day and he died from hypoxic cardiac arrest on the twelfth day. Necropsy revealed ARDS. The peak quinine level was 17.8 mg/L (Wenstone et al, 1989).
    4) PREGNANCY: A 21-year-old woman ingested 90 g of quinine in her eighth week of pregnancy to induce abortion. The patient subsequently became drowsy, with dark red urine which was positive for occult blood and albumin. Subsequently the patient became oliguric and her blood urea climbed to 208 g/dL. Platelet count was known to be 43,000/cu mm and at 24 hours postingestion the patient had a seizure. The patient was treated with diuretics, blood transfusion and hemodialysis with improvement of her condition resulting in hospital discharge at 37 days (Notelovitz et al, 1970).

Summary

    A) TOXICITY: In adults, mild to moderate toxicity is reported after ingestion of 2 to 4 grams, severe toxicity after 4.5 to 9 grams, and fatalities after 5 to 9.75 grams.
    B) THERAPEUTIC DOSE: ADULTS: 648 mg orally every 8 hours for 7 days. CHILDREN: 10 mg/kg orally three times daily for 3 or 7 days.

Therapeutic Dose

    7.2.1) ADULT
    A) UNCOMPLICATED PLASMODIUM FALCIPARUM
    1) ORAL: The usual oral dose for the treatment of uncomplicated P falciparum malaria is 648 mg (2 capsules) every 8 hours for 7 days (Prod Info QUALAQUIN(R) oral capsules, 2014).
    B) SAFETY ANNOUNCEMENT
    1) Quinine is NOT approved for use in the treatment or prevention of night time leg cramps. In some cases, patients receiving "off-label" quinine have developed serious adverse events and life-threatening blood related (ie, thrombocytopenia) events (Prod Info QUALAQUIN(R) oral capsules, 2010; US Food and Drug Administration, 2010). Thirty-eight cases of serious adverse events (24 hematologic, 4 cardiovascular and 10 miscellaneous events) associated with quinine use for the treatment of night time leg cramps have been submitted to the Food and Drug Administration's Adverse Event Reporting System (AERS) from April 2005 to October 2008 (US Food and Drug Administration, 2010).
    7.2.2) PEDIATRIC
    A) UNCOMPLICATED PLASMODIUM FALCIPARUM
    1) ORAL
    a) 16 YEARS OF AGE OR OLDER: For the treatment of uncomplicated P falciparum malaria, the recommended dose is 648 mg (2 capsules) every 8 hours for 7 days (Prod Info QUALAQUIN(R) oral capsules, 2014)
    b) LESS THAN 16 YEARS OF AGE: For the treatment of uncomplicated, chloroquine-resistant malaria, the recommended dose is 10 mg/kg of quinine sulfate orally three times daily for 3 or 7 days. The duration of treatment is dependent on the region where the infection was acquired: 7 days, if the infection was acquired in Southeast Asia; 3 days, if the infection was acquired elsewhere (Centers for Disease Control and Prevention, 2009).
    2) INTRAVENOUS
    a) An intravenous loading dose of quinine (20 mg/kg) followed by 10 mg/kg every 8 hours has been suggested in children with cerebral malaria (Van der Torn et al, 1998).

Minimum Lethal Exposure

    A) CASE REPORTS
    1) ADULT
    a) Ingestion of 8 grams of quinine sulfate resulted in fatality in a 24-year-old man who received hemoperfusion 10 hours postingestion (Goldenberg & Wexler, 1988).
    b) Ingestion of 5 grams of quinine bisulfate was fatal in a 17-year-old male (Wenstone et al, 1989).
    c) Following the ingestion of 9.75 grams of quinine, a 32-year-old man presented to the ED with blurred vision, deafness and dilated pupils. A syndrome of extreme peripheral vasodilation and a wide complex tachycardia developed that did not respond to aggressive therapies. The patient died 64 hours after the ingestion (Morrison et al, 2001).
    B) ACUTE
    1) LDLo - (UNREPORTED) HUMAN, Male: 294 mg/kg (RTECS , 2001)
    2) Death has occurred in a child after ingestion of 1.5 grams (Bateman et al, 1985)

Maximum Tolerated Exposure

    A) CASE REPORTS
    1) Visual symptoms have occurred in adults who ingested 2 grams but the average adult would probably tolerate up to 3 or 4 grams with only mild cinchonism (Fong et al, 1984; Murray & Jay, 1983) .
    2) A 51-year-old male was reported to have a prolonged QRS (135 milliseconds) approximately one hour after the ingestion of 9 grams quinine. Subsequently, hypotension, blindness, a grand mal seizure, and a severe mixed respiratory and metabolic acidosis developed. Following supportive therapy the patient recovered, with normal vision reported by day 5 (Pearson, 1998).
    3) Severe bilateral visual loss with no light perception was reported in a 53-year-old man between 24 and 36 hours after an intentional overdose of 4.5 grams quinine. No co-ingestants were taken. Ophthalmologic examination revealed cherry-red spots at the macula consistent with a period of retinal arteriolar constriction. The authors suspected vasospasm due to fluctuating visual loss, and initiated the following therapies: nimodipine (to treat vasospasm), and induced hypertension, hemodilution and hypervolemia, with resolution of symptoms over the next 24 hours (Barrett & Solano, 2002).

Serum Plasma Blood Concentrations

    7.5.2) TOXIC CONCENTRATIONS
    A) TOXIC CONCENTRATION LEVELS
    1) CONCENTRATION LEVEL
    a) LEVELS NOT RESULTING IN TOXICITY -
    1) No serious cardiac or neurologic toxicity was observed in patients with serum levels of 10 to 20 mcg/mL during a 3 day treatment with combined intravenous and oral therapy (White et al, 1982).
    a) This suggests tolerance or altered distribution during continuous exposure, or transient high concentrations of unbound drug early in the distribution phase of acute ingestion.
    b) TOXIC LEVELS -
    1) Plasma concentrations over 5 mcg/mL have been associated with cinchonism. Levels above 10 mcg/mL are associated with visual impairment, and levels above 16 mcg/mL are associated with cardiac dysrhythmias (Dyson & Proudfoot, 1985; Boland et al, 1985; White et al, 1982; Bateman et al, 1985).
    2) Survival with residual retinal damage occurred in a patient with a level of 20.4 mcg/mL (Bateman et al, 1985).
    3) Survival with cardiac dysrhythmias and temporary blindness and tinnitus is reported in a patient with a level of 17 mcg/mL following an estimated overdose of 5.5 grams (Nordt & Clark, 1998).
    4) A serum quinine level of 45 mcg/mL was reported between 24 and 36 hours after the ingestion of 4.5 grams of quinine in a 53-year-old man. Quinine ocular toxicity was treated using therapy for vasospasm, and his symptoms improved over the next 24 hours (Barrett & Solano, 2002).
    c) FATAL LEVELS -
    1) Death was reported in a patient with an initial quinine level of 22.2 mcg/mL (Goldenberg & Wexler, 1988).
    2) A peak level of 17.8 mcg/mL was reported in a fatal case (Wenstone et al, 1989).
    3) Postmortem serum quinine level of 15 mg/L was reported in a 32-year-old male who ingested 9.75 grams of quinine 64 hours prior to death (Morrison et al, 2003; Morrison et al, 2001).

Toxicity Information

    7.7.1) TOXICITY VALUES
    A) ANIMAL DATA
    1) LD50- (INTRAPERITONEAL)MOUSE:
    a) 115 mg/kg (RTECS, 2006)

Pharmacologic Mechanism

    A) Quinine is used for muscle leg cramps. It has also been used anecdotally to induce abortion (Dannenberg et al, 1983). Quinine overdose is similar to quinidine overdose with less cardiotoxicity.

Toxicologic Mechanism

    A) Quinine is a Class 1A antiarrhythmic, less potent than quinidine. Cardiac effects include sodium channel blockade, phase O depression, conduction delay, and prolonged repolarization (Bateman & Dyson, 1986).
    B) Quinine is an alpha adrenoreceptor antagonist and may have oxytocic activity (Bateman & Dyson, 1986).
    C) Ocular toxicity due to quinine may be due to retinal ischemia secondary to vasospasm and/or a direct toxic effect on the retinal cells (Barrett & Solano, 2002).

Physical Characteristics

    A) QUININE is very bitter. The bitterness threshold of quinine hydrochloride is 1:30,000.
    B) QUININE BISULFATE is a white crystalline powder or colorless crystals that effloresce in dry air and are freely soluble in water and sparingly soluble in alcohol (S Sweetman , 1999).
    C) QUININE HYDROCHLORIDE has a bitterness threshold of 1:30,000.
    D) QUININE SULFATE is a bitter tasting, white, odorless, crystalline powder that darkens on exposure to light and is slightly soluble in alcohol, ether, chloroform, and water (Prod Info QUALAQUIN(R) oral capsules, 2011).

Ph

    A) QUININE: 8.8 (saturated aqueous solution)
    B) QUININE BISULFATE: 2.8 to 3.4 (1% aqueous solution) (S Sweetman , 1999)
    C) QUININE HYDROCHLORIDE: approximately 6
    D) QUININE SULFATE: 6.2 (saturated aqueous solution)

Molecular Weight

    A) QUININE: 324.41
    B) QUININE HYDROCHLORIDE: 360.88
    C) QUININE SULFATE: 782.96 (Prod Info QUALAQUIN(R) oral capsules, 2011)

General Bibliography

    1) AMA Department of DrugsAMA Department of Drugs: AMA Evaluations Subscription, American Medical Association, Chicago, IL, 1992.
    2) Abraham R & Whitehead S: Factitious quinine-induced thrombocytopenia. MJA 1998; 168:19-20.
    3) Alaspaa AO, Kuisma MJ, Hoppu K, et al: Out-of-hospital administration of activated charcoal by emergency medical services. Ann Emerg Med 2005; 45:207-12.
    4) American Heart Association: 2005 American Heart Association Guidelines for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2005; 112(24 Suppl):IV 1-203. Available from URL: http://circ.ahajournals.org/content/vol112/24_suppl/. As accessed 12/14/2005.
    5) Anderson JL & Mason JW: Successful treatment by overdrive pacing of recurrent quinidine syncope due to ventricular tachycardia. Am J Med 1978; 64:715-718.
    6) Aster RH: Quinine sensitivity: a new cause of the hemolytic uremic syndrome. Ann Intern Med 1993; 119:243-244.
    7) Atkinson SW, Young Y, & Trotter GA: Treatment with activated charcoal complicated by gastrointestinal obstruction requiring surgery. Br Med J 1992; 305:563.
    8) Auprayoon P, Sukontason K, & Na-Bangchang K: Pharmacokinetics of quinine in chronic liver disease. Br J Clin Pharmacol 1995; 40:494-497.
    9) Bacon P, Spalton DJ, & Smith SE: Blindness from quinine toxicity. Br J Ophthalmol 1988; 72:219-224.
    10) Barrett NA & Solano T: Quinine ocular toxicity: treatment of blindness using therapy for vasospasm. Anaesth Intensive Care 2002; 30:234-235.
    11) Baselt RC: Disposition of Toxic Drugs and Chemicals in Man, 5th ed, Chemical Toxicology Institute, Foster City, CA, 2000.
    12) Bateman DN & Dyson EH: Quinine toxicity. Adv Drug React Ac Pois Rev 1986; 4:215-233.
    13) Bateman DN, Blain PG, & Woodhouse KW: Pharmacokinetics and clinical toxicity of quinine overdosage: lack of efficacy of techniques to enhance elimination. Q J Med 1985; 54:125-131.
    14) Bauman JL, Bauernfeind RA, & Strasberg B: Torsades de pointes due to quinidine: observations in 31 patients. Am Heart J 1984; 107:425-430.
    15) Berglund F: Toxicity of quinine. Toxicology 1989; 58:237-238.
    16) Bird MR, O'Neill AI, & Buchanan RRC: Lupus anticoagulant in the elderly may be associated with both quinine and quinidine usage. Pathology 1995; 27:136-139.
    17) Blayney DW: Quinine-associated immune thrombopenia, neutropenia, and renal failure in a patient with Klinefelter's syndrome (letter). Blood 1992; 80:2686.
    18) Bodenhamer JE & Smilkstein MJ: Delayed cardiotoxicity following quinine overdose: a case report. J Emerg Med 1993; 11:279-285.
    19) Boland ME, Roper SMB, & Henry JA: Complications of quinine poisoning. Lancet 1985; 1:384-385.
    20) Boscoe MJ, Calver DM, & Keyte C: Quinine overdose. Prevention of visual damage by stellate ganglion block. Anaesthesia 1983; 38:669-671.
    21) Braveman BL, Koransky DS, & Kulvin MM: Quinine amaurosis. Am J Ophthalmol 1948; 31:731-733.
    22) Brinton GS, Norton EWD, & Zahn JR: Ocular quinine toxicity. Am J Ophthalmol 1980; 90:403-410.
    23) Brodie BB & Undenfriend S: The estimation of quinine in human plasma with a note on the estimation of quinidine. J Pharm Exp Ther 1943; 78:154-158.
    24) Brophy GM, Bell R, Claassen J, et al: Guidelines for the evaluation and management of status epilepticus. Neurocrit Care 2012; 17(1):3-23.
    25) Buchanan TAS, Lyness RW, & Collins AD: An experimental study of quinine blindness. Eye 1987; 1:522-524.
    26) Canning CR & Hague S: Ocular quinine toxicity. Br J Ophthalmol 1988; 72:23-26.
    27) Caravati EM, Knight HH, & Linscott MS: Esophageal laceration and charcoal mediastinum complicating gastric lavage. J Emerg Med 2001; 20:273-276.
    28) Centers for Disease Control and Prevention: Guidelines for Treatment of Malaria in the United States. Centers for Disease Control and Prevention. Atlanta, GA. 2009. Available from URL: http://www.cdc.gov/malaria/resources/pdf/treatmenttable73109.pdf. As accessed 2010-07-21.
    29) 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.
    30) Charlton NP , Lawrence DT , Brady WJ , et al: Termination of drug-induced torsades de pointes with overdrive pacing. Am J Emerg Med 2010; 28(1):95-102.
    31) 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.
    32) Choonara IA & Rane A: Therapeutic drug monitoring of anticonvulsants state of the art. Clin Pharmacokinet 1990; 18:318-328.
    33) Chuah TL & Denaro C: A case of quinine-induced thrombocytopenia and neutropenia. Aust NZ J Med 2000; 30:96.
    34) Chyka PA & Seger D: Position statement: single-dose activated charcoal. American Academy of Clinical Toxicology; European Association of Poisons Centres and Clinical Toxicologists. J Toxicol Clin Toxicol 1997; 35:721-741.
    35) Chyka PA, Seger D, Krenzelok EP, et al: Position paper: Single-dose activated charcoal. Clin Toxicol (Phila) 2005; 43(2):61-87.
    36) Chyka PA: Multiple-dose activated charcoal and enhancement of systemic drug clearance: summary of studies in animals and human volunteers. J Toxicol Clin Toxicol 1995; 33(5):399-405.
    37) Clark DJ: Clinical curio: warfarin and tonic water. Br Med J 1983; 110:788.
    38) Clarke EGC: In Isolation and Identification of Drugs, Pharmaceutical Press, London, England, 1969, pp 532-533.
    39) Corby DG & Decker WJ: Management of acute poisoning with activated charcoal. Pediatrics 1974; 54:324-328.
    40) Dagnone D, Matsui D, & Rieder MJ: Assessment of the palatability of vehicles for activated charcoal in pediatric volunteers. Pediatr Emerg Care 2002; 18:19-21.
    41) Dannenberg AL, Dorfman SF, & Johnson J: Use of quinine for self-induced abortion. South Med J 1983; 76:846-849.
    42) Dawson TAJ: Side effect of quinine for nocturnal cramps (letter). BMJ 1995; 310:738.
    43) Di Perri G, Allegranzi B, & Bonora S: Quinine-induced blindness reversed by an increase in alpha1-acid glycoprotein level (letter). Ann Intern Med 2002; 136:339.
    44) Drake WM & Hiorns MP: Quinine overdose (abstract 518). Adis International Limited (Sept), 1994.
    45) Drew BJ, Ackerman MJ, Funk M, et al: Prevention of torsade de pointes in hospital settings: a scientific statement from the American Heart Association and the American College of Cardiology Foundation. J Am Coll Cardiol 2010; 55(9):934-947.
    46) Drewitt PN, Butterworth KR, & Springall CD: Toxicity threshold of quinine hydrochloride following low-level repeated dosing in healthy volunteers. Fd Chem Toxic 1993; 31:235-245.
    47) Dyson EH & Proudfoot AT: Death and blindness due to overdose of quinine. Br Med J 1985; 291:31-33.
    48) Dyson EH, Proudfoot AT, & Bateman DN: Quinine amblyopia: is current management appropriate?. J Toxicol Clin Toxicol 1985a; 23:571-578.
    49) Elliot CG, Colby TV, & Kelly TM: Charcoal lung. Bronchiolitis obliterans after aspiration of activated charcoal. Chest 1989; 96:672-674.
    50) Evans ANW, Brooke OG, & West RJ: The ingestion by pregnant women of substances toxic to the foetus. Practitioner 1980; 224:315.
    51) FDA: Poison treatment drug product for over-the-counter human use; tentative final monograph. FDA: Fed Register 1985; 50:2244-2262.
    52) Farver DK & Lavin MN: Quinine-induced hepatotoxicity. Ann Pharmacother 1999; 33:32-34.
    53) Feder HM, Lawlor M, & Krause PJ: Babesiosis in pregnancy (letter). New Engl J Med 2003; 349(2):195-196.
    54) Ferguson J, Addo HA, & Johnson BE: Quinine-induced photosensitivity: clinical and experimental studies. Br J Dermatol 1987; 117:631-640.
    55) Floyd M, Hill AVL, & Ormston BJ: Quinine amblyopia treated by hemodialysis. Clin Nephrol 1974; 2:44-46.
    56) Fong LP, Kaufman DV, & Galbraith JEK: Ocular toxicity of quinine. Med J Aust 1984; 141:528-529.
    57) Friedman L: Clinical observations on quinine toxicity. Ann Ophthalmol 1980; 12:640-642.
    58) Garrod GD & Judson JA: Fatal quinine poisoning: a case report. NZ Med J 1981; 94:215-216.
    59) Ghannoum M & Gosselin S: Enhanced poison elimination in critical care. Adv Chronic Kidney Dis 2013; 20(1):94-101.
    60) Gibbs JL, Trafford A, & Sharpstone P: Quinine amblyopia treated by combined haemodialysis and activated resin haemoperfusion. Lancet 1985; 1:752-753.
    61) Goldenberg AM & Wexler LF: Quinine overdose: review of toxicity and treatment. Clin Cardiol 1988; 11:716-718.
    62) Golej J, Boigner H, Burda G, et al: Severe respiratory failure following charcoal application in a toddler. Resuscitation 2001; 49:315-318.
    63) Gomez HF, Brent JA, & Munoz DC: Charcoal stercolith with intestinal perforation in a patient treated for amitriptyline ingestion. J Emerg Med 1994; 12:57-60.
    64) Graff GR, Stark J, & Berkenbosch JW: Chronic lung disease after activated charcoal aspiration. Pediatrics 2002; 109:959-961.
    65) Grattan-Smith TM, Gillis J, & Kilham H: Quinine poisoning in children. Med J Aust 1987; 147:93-95.
    66) Guenther Skokan E, Junkins EP, & Corneli HM: Taste test: children rate flavoring agents used with activated charcoal. Arch Pediatr Adolesc Med 2001; 155:683-686.
    67) Hagley MT, Hosney IA, & Hulisz DT: Hemolytic-uremic syndrome associated with ingestion of quinine. Am J Nephrol 1992; 12:192-195.
    68) Hall AP, Czerwinski AW, & Madonia EC: Human plasma and urine quinine levels following tablets, capsules and intravenous infusion. Clin Pharm Ther 1973; 14:580-585.
    69) Harris CR & Filandrinos D: Accidental administration of activated charcoal into the lung: aspiration by proxy. Ann Emerg Med 1993; 22:1470-1473.
    70) Harsch HH: Aspiration of activated charcoal. N Engl J Med 1986; 314:318.
    71) Heath A: Resin haemoperfusion for quinine poisoning. Lancet 1985; 1:1224.
    72) Hegenbarth MA & American Academy of Pediatrics Committee on Drugs: Preparing for pediatric emergencies: drugs to consider. Pediatrics 2008; 121(2):433-443.
    73) Hla KK, Leahy N, & Henry JA: Accidental quinine poisoning in children under five. Vet Hum Toxicol 1987; 29(Suppl 2):121-123.
    74) Hla KK, Moore DF, & Henry JA: Intravenous nitrates a potential antidote for quinine ocular toxicity (abstract), EAPCCT, XV Congress, Istanbul, Turkey, 1992, pp 33.
    75) Hoffman JR: Overdose with cardiotherapeutic agents. Geriatrics 1983; 38:51-65.
    76) Horgan SE & Williams RW: Chronic retinal toxicity due to quinine in Indian tonic water (letter). Eye 1995; 9:637-663.
    77) Huston M & Levinson M: Are one or two dangerous? Quinine and quinidine exposure in toddlers. J Emerg Med 2006; 31(4):395-401.
    78) Hvidberg EF & Dam M: Clinical pharmacokinetics of anticonvulsants. Clin Pharmacokinet 1976; 1:161.
    79) Ilkhanipour K, Yealy DM, & Krenzelok EP: The comparative efficacy of various multiple-dose activated charcoal regimens. Am J Emerg Med 1992; 10:298-300.
    80) Jung TTK, Rhee C-K, & Lee CS: Ototoxicity of salicylate, nonsteroidal anti-inflammatory drugs, and quinine. Otolaryngol Clin N Am 1993; 26:791-810.
    81) Keren A, Tzivoni D, & Gavish D: Etiology, warning signs and therapy of torsade de pointes: a study of 10 patients. Circulation 1981; 64:1167-1174.
    82) Khan IA & Gowda RM: Novel therapeutics for treatment of long-QT syndrome and torsade de pointes. Int J Cardiol 2004; 95(1):1-6.
    83) 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.
    84) Krantz MJ, Dart RC, & Mehler PS: Transient pulmonary infiltrates possibly induced by quinine sulfate. Pharmacother 2002; 22:775-778.
    85) Langford NJ, Good AM, Laing WJ, et al: Quinine intoxications reported to the Scottish Poisons Information Bureau 1997-2002: a continuing problem. Br J Clin Pharmacol 2003; 56:576-578.
    86) Limburg PJ, Katz H, & Grant CS: Quinine-induced hypoglycemia. Ann Intern Med 1993; 119:218-219.
    87) Lincoff MH: Quinine amblyopia. Arch Ophthalmol 1955; 53:382-384.
    88) 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.
    89) Ljunggren B, Hindsen M, & Isaksson M: Systemic quinine photosensitivity with photoepicutaneous cross-reactivity to quinidine. Contact Derm 1992; 26:1-4.
    90) Lockey D & Bateman DN: Effect of oral activated charcoal on quinine elimination. Br J Clin Pharmacol 1989; 27:92-94.
    91) Loddenkemper T & Goodkin HP: Treatment of Pediatric Status Epilepticus. Curr Treat Options Neurol 2011; Epub:Epub.
    92) Looareesuwan S, White NJ, Phillips RE, et al: Quinine and severe falciparum malaria in late pregnancy. Lancet 1985; 2:4-7.
    93) Maguire RB, Stroncek DF, & Campbell AC: Recurrent pancytopenia, coagulopathy, and renal failure associated with multiple quinine-dependent antibodies. Ann Intern Med 1993; 119:215-217.
    94) Manno EM: New management strategies in the treatment of status epilepticus. Mayo Clin Proc 2003; 78(4):508-518.
    95) Martin TG, Michelson EA, & Benitez JG: Unsuccessful abortion attempt with quinine results in blindness (abstract). Vet Hum Toxicol 1992; 34:333.
    96) Mauro LS, Nawarskas JJ, & Mauro VF: Misadventures with activated charcoal and recommendations for safe use. Ann Pharmacother 1994; 28(7-8):915-924.
    97) McDonald SP, Shanahan EM, & Thomas AC: Quinine-induced hemolytic uremic syndrome. Clin Nephrol 1997; 47:397-400.
    98) McGready R, Cho T, et al: Randomized comparison of quinine-clindamycin versus artesunate in the treatment of falciparum malaria in pregnancy. Trans R Soc Trop Med Hyg 2001; 95(6):651-656.
    99) Mina B, Dym JP, & Kuepper F: Fatal inhalational anthrax with unknown source of exposure in a 61-year-old woman in New York City. J Am Med Assoc 2002; 287:858-862.
    100) Mizutani T, Naito H, & Oohashi N: Rectal ulcer with massive hemorrhage due to activated charcoal treatment in oral organophosphate poisoning. Hum Exp Toxicol 1991; 10:385-386.
    101) Moloney JBM, Hillery M, & Fenton M: Two year electrophysiology follow-up in quinine amblyopia: a case report. Acta Ophthalmol 1987; 65:731-734.
    102) Moore D, Marshall J, & Henry JA: Research into quinine ocular toxicity (letter). Br J Ophth 1992; 76:703.
    103) Morgan MDL, Rainsford DJ, & Pusey CD: The treatment of quinine poisoning with charcoal hemoperfusion. Postgrad Med J 1983; 59:365-367.
    104) Morrison LD, Berumen K, & Velez LI: Death by quinine (abstract). Clin Toxicol 2001; 39:507-508.
    105) Morrison LD, Velez LI, Shepherd G, et al: Death by quinine. Vet Human Toxicol 2003; 45:303-306.
    106) Morton AP: Quinine-induced disseminated intravascular coagulation and haemolytic-uraemic syndrome (letter). Med J Aust 2002; 176:351.
    107) Murray SB & Jay JL: Loss of sight after self poisoning with quinine. Br Med J 1983; 287:1700.
    108) 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.
    109) Nishimura H & Tanimura TNishimura H & Tanimura T: Clinical Aspects of the Teratogenicity of Drugs, Excerpta Medica, Amsterdam, 1976.
    110) None Listed: Position paper: cathartics. J Toxicol Clin Toxicol 2004; 42(3):243-253.
    111) Nordt SP & Clark RF: Acute blindness after severe quinine poisoning (letter). Am J Emerg Med 1998; 16:214-215.
    112) Notelovitz M, Dalrymple D, & Funston M: Acute renal failure following quinine poisoning. S Afr Med J 1970; 44:649-652.
    113) Okitolonda W, Delacollette C, & Melengreau M: High incidence of hypoglycaemia in African patients treated with intravenous quinine for severe malaria. Br Med J 1987; 295:716-718.
    114) Okun MM, Henner M, & Paulson C: A quinine-induced drug reaction of photosensitive distribution with histological features mimicking mycosis fungoides. Clin Exp Dermatol 1994; 19:246-248.
    115) Paintaud G, Alvan G, & Ericsson O: The reproducibility of quinine bioavailability. Br J Clin Pharmacol 1993; 35:305-307.
    116) Park GD, Radomski L, & Goldberg MJ: Effects of size and frequency of oral doses of charcoal on theophylline clearance. Clin Pharmacol Ther 1983; 34:663-666.
    117) Pawar R, Jacobs GH, & Smith MC: Quinine-associated acute interstitial nephritis. Am J Kidney Dis 1994; 24:211-214.
    118) Pearson S: Quinine toxicity. Emerg Med 1998; 10:317-323.
    119) 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.
    120) Perez JA Jr, Stryker J, & Arsura EL: Probable quinine-induced hepatotoxicity. West J Med 1994; 160:59-60.
    121) Perticone F, Ceravolo R, & Cuccurullo O: Prolonged magnesium sulfate infusion in the treatment of ventricular tachycardia in acquired long QT syndrome. Clin Drug Inverst 1997; 13:229-236.
    122) Pollack MM, Dunbar BS, & Holbrook PR: Aspiration of activated charcoal and gastric contents. Ann Emerg Med 1981; 10:528-529.
    123) Prasad RS, Kodali VRR, Khuraijam GS, et al: Acute confusion and blindness from quinine toxicity. Eur J Emerg Med 2003; 10:353-356.
    124) Prescott LF, Hamilton AR, & Heyworth R: Treatment of quinine overdosage with repeated oral charcoal. Br J Clin Pharmacol 1989; 27:95-97.
    125) Price EJ, Bevan JS, & Rees A: Quinine-induced cutaneous vasculitis. BJCP 1992; 46:138-139.
    126) 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.
    127) Product Information: QUALAQUIN(R) oral capsules, quinine sulfate oral capsules. AR Scientific, Inc. (per FDA), Philadelphia, PA, 2013.
    128) Product Information: QUALAQUIN(R) oral capsules, quinine sulfate oral capsules. AR Scientific, Inc., Philadelphia, PA, 2010.
    129) Product Information: QUALAQUIN(R) oral capsules, quinine sulfate oral capsules. AR Scientific, Inc., Philadelphia, PA, 2011.
    130) Product Information: QUALAQUIN(R) oral capsules, quinine sulfate oral capsules. AR Scientific,Inc, Philadelphia, PA, 2008.
    131) Product Information: QUALAQUIN(R) oral capsules, quinine sulfate oral capsules. Caraco Pharmaceutical Laboratories, Ltd. (per FDA), Detroit, MI, 2014.
    132) Product Information: diazepam IM, IV injection, diazepam IM, IV injection. Hospira, Inc (per Manufacturer), Lake Forest, IL, 2008.
    133) Product Information: lorazepam IM, IV injection, lorazepam IM, IV injection. Akorn, Inc, Lake Forest, IL, 2008.
    134) Product Information: magnesium sulfate heptahydrate IV, IM injection, solution, magnesium sulfate heptahydrate IV, IM injection, solution. Hospira, Inc. (per DailyMed), Lake Forest, IL, 2009.
    135) Product Information: norepinephrine bitartrate injection, norepinephrine bitartrate injection. Sicor Pharmaceuticals,Inc, Irvine, CA, 2005.
    136) RTECS : Registry of Toxic Effects of Chemical Substances. National Institute for Occupational Safety and Health. Cincinnati, OH (Internet Version). Edition expires 2001; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    137) RTECS: Registry of Toxic Effects of Chemical Substances. National Institute for Occupational Safety and Health. Cincinnati, OH (Internet Version). Edition expires 2006; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    138) Rau NR, Nagaraj MV, Prakash PS, et al: Fatal pulmonary aspiration of oral activated charcoal. Br Med J 1988; 297:918-919.
    139) Ray MJ, Padin DR, & Condie JD: Charcoal bezoar. Small-bowel obstruction secondary to amitriptyline overdose therapy. Dig Dis Sci 1988; 33:106-107.
    140) Reimold WV, Larbig D, & Kochsiek K: Hypokaliamie und herzrhythmusstorungen infolge chininvergiftung. Dtsch Med Wochenschr 1970; 95:517-521.
    141) S Sweetman : Martindale: The Complete Drug Reference (electronic version). The Pharmaceutical Press. London, UK (Internet Version). Edition expires 1999; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    142) Sabto J, Pierce RM, & West RH: Hemodialysis, peritoneal dialysis, plasmapheresis and forced diuresis for the treatment of quinine overdose. Clin Nephrol 1981; 16:264-268.
    143) Samaranayake CB & Yap E: Fatal quinine-induced thrombocytopenia from pulmonary haemorrhage. Intern Med J 2014; 44(4):423-425.
    144) Satjchuk JT, Milhail R, & Jewel JS: Electronystagmographic findings in long-term low-dose quinine ingestion. Arch Otolaryngol 1984; 110:788.
    145) Schardein JLSchardein JL: Drugs as Teratogens, CRC Press, Cleveland, OH, 1976.
    146) Schattner A: Quinine hypersensitivity simulating sepsis. Am J Med 1998; 104:488-490.
    147) Schlutz M, Zinneman HH, & Wall WH: Drug fever caused by quinine and quinidine. Minnesota Med 1973; 56:668-670.
    148) Schmitt SK & Tomford JW: Quinine-induced pancytopenia and coagulopathy (letter). Ann Intern Med 1994; 120:90-91.
    149) Schonwald S & Shannon M: Unsuspected quinine intoxication presenting as acute deafness and mutism. Am J Emerg Med 1991; 9:318-320.
    150) 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.
    151) Shub C, Gay GT, & Sidell PM: The management of acute quinidine intoxication. Chest 1978; 73:173-178.
    152) Smilkstein MJ, Kulig KW, & Rumack BH: Acute toxic blindness: unrecognized quinine poisoning. Ann Emerg Med 1987; 16:98-101.
    153) Smith WM & Gallagher JJ: "Les torsades de pointes": an unusual ventricular arrhythmia. Ann Intern Med 1980; 93:578-584.
    154) Spiller HA & Rogers GC: Evaluation of administration of activated charcoal in the home. Pediatrics 2002; 108:E100.
    155) 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.
    156) Story D, Hall A, & Older P: Quinine-induced haemolytic uraemic syndrome (letter). Anaesth Intens Care 1994; 22:114-115.
    157) Stroncek DF, Vercellotti GM, & Hammerschmidt DE: Characterization of multiple quinine-dependent antibodies in a patient with episodic hemolytic uremic syndrome and immune agranulocytosis. Blood 1992; 80:241-248.
    158) Swiryn S & Kimm SS: Quinidine-induced syncope. Arch Int Med 1983; 143:314-316.
    159) Tapadinhas C, Dias M, & Conchon I: Contact dermatitis due to quinine-containing hair lotion. Contact Derm 1994; 31:127.
    160) Taylor S: Quinine intoxications: a continuing problem (letter). Br J Clin Pharmacol 2004; 57:817.
    161) Taylor TE, Molyneux ME, & Wirima JJ: Blood glucose levels in Malawian children before and during the administration of intravenous quinine for severe falciparum malaria. N Engl J Med 1988; 319:1040-1047.
    162) Thakore S & Murphy N: The potential role of prehospital administration of activated charcoal. Emerg Med J 2002; 19:63-65.
    163) Thomas D: Forced acid diuresis and stellate ganglion block in the treatment of quinine poisoning. Anaesthesia 1984; 39:257-260.
    164) Tiliakos N & Waites TF: Multiform quinidine toxicity. South Med J 1981; 74:1267-1268.
    165) US Food and Drug Administration: FDA Drug Safety Communication: New risk management plan and patient Medication Guide for Qualaquin (quinine sulfate). US Food and Drug Administration. Rockville, MD. 2010. Available from URL: http://www.fda.gov/Drugs/DrugSafety/PostmarketDrugSafetyInformationforPatientsandProviders/ucm218202.htm. As accessed 2010-07-08.
    166) Vale JA, Krenzelok EP, & Barceloux GD: Position statement and practice guidelines on the use of multi-dose activated charcoal in the treatment of acute poisoning. J Toxicol Clin Toxicol 1999; 37:731-751.
    167) Vale JA, Kulig K, American Academy of Clinical Toxicology, et al: Position paper: Gastric lavage. J Toxicol Clin Toxicol 2004; 42:933-943.
    168) 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.
    169) Valman HB & White DC: Stellate block for quinine blindness in a child. Br Med J 1977; 1:1065.
    170) Van der Torn M, Thuma PE, & Mabeza GF: Loading dose of quinine in African children with cerebral malaria. Trans Roy Soc Trop Med Hyg 1998; 92:325-331.
    171) Vusirikala B, Williams TA, & Ram ARR: A late presentation of ocular quinine toxicity managed with a combination of vasodilatory treatments (letter). Eye 2005; 19(7):801-802.
    172) Wagner GH, Diffey BL, & Ive FA: 'I'll have mine with a twist of lemon'. Quinine photosensitivity from excessive intake of tonic water (letter). Br J Dermatol 1994; 131:734-735.
    173) Watt G, Na-Nakorn A, & Bateman DN: Amplification of quinine cardiac effects by the resistance-reversing agent prochlorperazine in falciparum malaria. Am J Trop Med Hyg 1993; 49:645-649.
    174) Wenstone R, Bell M, & Mostafa SM: Fatal adult respiratory distress syndrome after quinine overdose (letter). Lancet 1989; 1:1143-1144.
    175) White NJ, Looareesuwan S, & Warrell DA: Quinine pharmacokinetics and toxicity in cerebral and uncomplicated falciparina malaria. Am J Med 1982; 73:564-572.
    176) White NJ, Warrel DA, & Chanthavanich P: Severe hypoglycemia and hyperinsulinemia in falciparium malaria. N Engl J Med 1983; 309:61-66.
    177) Wolf LR, Otten EJ, & Spadafora MP: Cinchonism: Two case reports and review of acute quinine toxicity and treatment. J Emerg Med 1992; 10:295-301.