29: Toxicology

Published on 23/06/2015 by admin

Filed under Emergency Medicine

Last modified 22/04/2025

Print this page

rate 1 star rate 2 star rate 3 star rate 4 star rate 5 star
Your rating: none, Average: 0 (0 votes)

This article have been viewed 1546 times

Section 29 Toxicology

Edited by Lindsay Murray

29.1 Approach to the poisoned patient

Introduction

Drug overdose in adults usually occurs in the context of self-poisoning, which may be either recreational or an act of deliberate self-harm.

Deliberate self-poisoning is one of the commonest reasons for general hospital admission in the UK1 and accounts for 1–5% of all public hospital admissions in Australia.2,3 The bulk of the medical management of cases presenting to hospital is carried out in the emergency department (ED), and the emergency physician is expected to be expert in the field. Although the management must vary considerably according to the nature and severity of the poisoning, some general principles apply.

Above all, it must be remembered that the acute overdose presentation is only a discrete time-limited event in the course of the underlying condition, which is usually psychiatric or social in origin.

Cardiovascular manifestations of poisoning include tachycardia, bradycardia, hypertension, hypotension, conduction defects and arrhythmias (Table 29.1.2). Bradycardia is relatively rarely observed and is associated with a number of potentially life-threatening ingestions. Tachycardia is commonly observed and is usually benign. It may be due to intrinsic sympathomimetic or anticholinergic effects of a drug, or a reflex response to hypotension or hypoxia. Hypotension is also commonly observed and may be due to a number of different causes (Table 29.1.2). Hypertension is unusual. Severe hypertension is usually associated with illicit drug use and is important because it may produce complications such as intracerebral haemorrhage.

Table 29.1.2 Cardiovascular effects of poisoning

CNS manifestations of poisoning include decreased level of consciousness, agitation or delirium, seizures and disordered temperature regulation. A decreased level of consciousness is a common presentation of poisoning and is associated with many drugs, some of which are listed in Table 29.1.1. Although usually a direct drug effect, CNS depression is occasionally secondary to hypoglycaemia, hypoxia or hypotension. Common causes of agitation or delirium following overdose are listed in Table 29.1.3. Toxic seizures are potentially life-threatening, and important causes are listed in Table 29.1.4.

Table 29.1.3 Toxic causes of agitation or delirium

Alcohol
Anticholinergic syndrome
Antidepressants

Atypical antipsychotic agents

Benzodiazepines and other sedative-hypnotics
Cannabis
Hallucinogenic agents
Serotonin syndrome
Sympathomimetic syndrome

Theophylline
Withdrawal syndromes

Table 29.1.4 Toxic causes of seizures

Amphetamines
Bupropion
Carbamazepine
Chloroquine
Cocaine
Isoniazid
Mefanamic acid
Theophylline
Tramadol
Tricyclic antidepressants
Venlafaxine

Hypothermia is usually a complication of environmental exposure secondary to a decreased level of consciousness or altered behaviour. Hyperthermia is a direct toxic effect and causes are listed in Table 29.1.5. Severe hyperthermia is rapidly lethal if not corrected.

Table 29.1.5 Toxic causes of hyperthermia

Amphetamines
Anticholinergics
Cocaine
MAO inhibitors
Salicylates
Serotonin syndrome

Metabolic and other manifestations of poisoning include hyper- and hypoglycaemia, hyper- and hyponatraemia, acidosis and alkalosis and hepatic failure.

Acute poisoning is distinguished from many other forms of acute illness in that, given appropriate supportive care over a relatively short period, a full recovery can usually be expected. A small number of potentially fatal poisonings may demonstrate progressive toxicity despite full supportive care. These are the so-called cellular toxins, and include colchicine, iron, salicylate, cyanide, paracetamol, theophylline and digoxin. In some of these cases early aggressive gastrointestinal decontamination, timely administration of antidotes or the institution of techniques of enhanced elimination may be life saving.

Mortality or morbidity may also result from specific complications of a poisoning. These include trauma, pulmonary aspiration, adult respiratory distress syndrome, rhabdomyolysis, renal failure and hypoxic encephalopathy. These complications usually occur prior to arrival in the ED.

Pulmonary aspiration frequently complicates a period of decreased level of consciousness or a seizure. It is a leading cause of in-hospital morbidity and mortality following overdose. This complication is characterized by rapid onset of dyspnoea, cough, fever, wheeze and cyanosis.

Rhabdomyolysis occurs as a direct toxic effect (rare) or secondary to excessive muscular hyperactivity, seizures, hyperthermia or prolonged coma with direct muscle compression. The urine is dark and acute renal failure can develop secondary to tubular deposition of myoglobin.

Assessment

Risk assessment

A risk assessment should be made as soon as possible in the management of the poisoned patient. Only resuscitation is a greater priority (see Table 29.1.6). Risk assessment is a distinct quantitative cognitive step through which the clinician attempts to predict the likely clinical course and potential complications for the individual patient at that particular presentation.4 An accurate risk assessment allows informed decision-making in regard to all subsequent management steps including duration and intensity of supportive care and monitoring, screening and specialized testing, decontamination, enhanced elimination, antidotes and disposition. Factors that are taken into account when formulating this risk assessment include: the agent(s), the dose, the time since ingestion, the clinical features present and patient factors (Table 29.1.6). Specialized testing may refine risk assessment. Access to specialized poisons information in the form of a poisons information centre or in-house databases is often necessary to formulate an accurate risk assessment.

Table 29.1.6 Risk assessment-based approach to poisoning

Supportive care and monitoring Investigations

Decontamination Enhanced elimination Antidotes Disposition

Reproduced from Toxicology Handbook. Murray L, Daly F, Little M, Cadogan M. Elsevier, Sydney 2007.

Physical examination

The focused physical examination of the poisoned patient aims to:

The initial physical examination of the overdose patient in many ways parallels the primary survey of the trauma patient. The airway, breathing and circulation are assessed and stabilized as necessary. The level of consciousness should be assessed, the presence of seizure activity noted and the blood glucose and temperature measured.

A more complete examination is carried out when the patient is stable. This should include a full neurological examination, including assessment of the level of consciousness and mental status, pupil size, muscle tone and movements and the presence or absence of focal neurological signs. Poisoning normally causes global CNS depression, and focal signs suggest an alternative diagnosis or a CNS complication such as cerebral haemorrhage.

Other features that should be specifically sought are any evidence of associated trauma, the state of hydration, the condition of the skin, in particular the presence of pressure areas, the presence or absence of bowel sounds and the condition of the urine.

Several toxic autonomic syndromes, or ‘toxidromes’, have been described in relation to poisoning. The principal ones are listed in Table 29.1.7. Identification of these syndromes may narrow the differential diagnosis in cases of unknown poisoning.5

Table 29.1.7 Toxic autonomic syndromes or ‘toxidromes’

Toxidrome Features Common causes
Anticholinergic Agitated delirium Antihistamines
Tachycardia Benztropine
Hyperthermia Carbamazepine
Dilated pupils Phenothiazines
Dry flushed skin Plant poisonings
Urinary retention Tricyclic antidepressants
Ileus  
Mixed cholinergic Brady- or tachycardia Organophosphates
Hypo- or hypertension Carbamates
Miosis or mydriasis  
Sweating  
Increased bronchial secretion  
Gastrointestinal hyperactivity  
Muscle weakness  
Fasciculations  
Mixed α- and β-adrenergic Hypertension Amphetamines
Tachycardia Cocaine
Mydriasis  
Agitation  
β-Adrenergic Hypotension Caffeine
Tachycardia Salbutamol
Hypokalaemia Theophylline
Hyperglycaemia  
Serotonin Altered mental status Amphetamines
Autonomic dysfunction Antihistamines
Fever Monoamine oxidase inhibitors
Hypertension NSSRIs
Sweating SSRIs
Tachycardia Tricyclic antidepressants (Usually combined overdose)
Motor dysfunction  
Hyperreflexia  
Hypertonia (esp. lower limbs)  
Myoclonus  

NSSRI, non-selective serotonin re-uptake inhibitor; SSRI, selective serotonin re-uptake inhibitor.

Treatment

The management of poisoning should be approached in a systematic way. Following initial resuscitation, further treatment is informed by the risk assessment (Table 29.1.6).

Resuscitation, supportive care and monitoring

Supportive care is the key element in the management of poisoning. The vast majority of poisonings result in temporary dysfunction of one or more of the body systems. If appropriate support of the system in question is instituted in a timely fashion and continued until the toxic substance is metabolized or excreted, a good outcome can be anticipated. In severe poisonings supportive care may be very aggressive, and possible interventions are listed in Table 29.1.8.

Table 29.1.8 Supportive care measures for the poisoned patient

Airway Endotracheal intubation
Breathing Supplemental oxygen
Ventilation
Circulation Intravenous fluids
Inotropes
Antihypertensives
Antiarrhythmics
Defibrillation/cardioversion
Cardiac pacing
Cardiopulmonary bypass
Metabolic Hypertonic dextrose
Hypertonic saline
Insulin/dextrose
Calcium salts
Sodium bicarbonate
Agitation/delirium Benzodiazepines
Butyrophenones
Seizures Benzodiazepines
Barbiturates
Body temperature External rewarming
External cooling
Impaired renal function Rehydration
Haemodialysis

The specific supportive management of a number of manifestations or complications of poisoning warrants further mention insofar as it may differ from the standard management of such conditions with other aetiologies.

Cardiopulmonary arrest from poisoning should be aggressively resuscitated. Direct current cardioversion is rarely successful in terminating toxic arrhythmias and should not take precedence over establishing adequate ventilation and oxygenation, cardiac compressions, correction of acidosis or hypovolaemia and the administration of specific antidotes. Resuscitative efforts should be continued beyond the usual timeframe. In cardiac arrest due to drugs with direct cardiac toxicity, the use of cardiopulmonary bypass or extracorporeal membrane oxygenation (ECMO) until the drug is metabolized may be life-saving.

In general, intravenous benzodiazepines are the drugs of choice for control of toxic seizures. Large doses may be required. Hypoxia and hypoglycaemia must be corrected if they are contributory factors. Patients with toxic seizures do not generally need long-term anticonvulsant therapy. Isoniazid-induced seizures are not controlled without administration of an adequate dose of the specific antidote, pyridoxine.

The management of pulmonary aspiration is essentially supportive, with supplemental oxygenation and intubation and mechanical ventilation if necessary. Neither prophylactic antibiotics nor corticosteroids have been shown to be helpful in the management of this condition, which is essentially a chemical pneumonitis.

Toxic hypertension rarely requires specific therapy. Most cases are mild and simple observation is sufficient. Agitation or delirium is a feature of many intoxications associated with hypertension, and adequate sedation with benzodiazepines usually lowers the blood pressure. Severe toxic hypertension is most likely in toxicity from cocaine or amphetamine-type drugs, and treatment may be indicated to avoid complications such as cardiac failure or intracerebral haemorrhage. The drug of choice in this situation is sodium nitroprusside by intravenous infusion. The extremely short duration of action of this vasodilator allows accurate control of hypertension during the toxic phase, and avoids the development of hypotension once toxicity begins to wear off.

Management of rhabdomyolysis consists of treatment of the causative factors, fluid resuscitation and careful monitoring of fluids and electrolytes. The role of mannitol and urinary alkalinization in reducing the risk of renal failure is not clear. Established acute renal failure requires haemodialysis, often for up to 6 weeks.

Decontamination

The aim of decontamination of the gastrointestinal tract is to bind or remove ingested material before it is absorbed into the circulation and able to exert its toxic effects. This is a very attractive concept and has long been considered one of the fundamental interventions in management of the overdose patient.

However, gastrointestinal decontamination should not be regarded as a routine procedure in the management of the patient presenting to the ED following an overdose. The decision to perform gastrointestinal decontamination and the choice of method should be based on an assessment of the likely benefit, the likely risk and the resources required. Gastrointestinal decontamination should only be considered where there is likely to be a significant amount of a significantly toxic material remaining in the gut. It is never indicated when the risk assessment predicts a benign course. Efforts at decontamination technique should never take precedence over the institution of appropriate supportive care.

Three basic approaches to gastrointestinal decontamination are available: gastric emptying, administration of an adsorbent and catharsis.

Gastric emptying can be attempted by the administration of an emetic, most commonly syrup of ipecac, or by gastric lavage. In volunteer studies both of these techniques removed highly variable amounts of marker substances from the stomach even if performed immediately after ingestion, and the effect diminished rapidly with time to the point of being negligible after one hour.6,7 Clinical outcome trials have failed to demonstrate improved outcome as a result of routine gastric emptying in addition to administration of activated charcoal, except, perhaps, in patients presenting unconscious within 1 h of ingestion.810

The principal adsorbent available to clinicians is activated charcoal (AC), which effectively binds most pharmaceuticals and chemicals, and is currently the decontamination method of choice for most poisonings. Materials that do not bind well to charcoal are listed in Table 29.1.9.

Table 29.1.9 Materials that do not bind well to activated charcoal

Alcohols

Corrosives

Hydrocarbons Metals and their salts

Charcoal is ‘activated’ by treatment in acid and steam at high temperature. This process removes impurities and greatly increases the surface area available for binding. Activated charcoal is packaged as a 50 g dose premixed with water or sorbitol, which is likely to be sufficient for the majority of ingestions. Adult patients are usually able to drink AC slurry from a cup. If the level of consciousness is too impaired to allow this, they should be intubated first. Administration of AC is absolutely contraindicated unless the patient has an intact or protected airway.

Volunteer studies demonstrate that the effect of AC diminishes rapidly with time and that the greatest benefit occurs if it is administered within 1 h. There is as yet no evidence that AC improves clinical outcome.11

There is no evidence to suggest that the addition of a cathartic such as sorbitol to AC improves clinical outcome.12,13

Apart from rarely employed endoscopic and surgical techniques, whole-bowel irrigation (WBI) is the most aggressive form of gastrointestinal decontamination. Polyethylene glycol solution (Golytely™) is administered via a nasogastric tube at a rate of 2 L/h until a clear rectal effluent is produced. This usually takes about 6 h and requires one-to-one nursing. In volunteer studies, this technique reduced the absorption of slow-release pharmaceuticals and so may be of benefit in life-threatening overdoses of these agents.13,14 Again, clinical benefit has not yet been conclusively demonstrated.15 The use of WBI has also been reported in the management of potentially toxic ingestions of iron, lead and packets of illicit drugs. Whole-bowel irrigation is contraindicated if there is evidence of ileus or bowel obstruction, and in patients who have an unprotected airway or haemodynamic compromise.16

Enhanced elimination

A number of techniques are available to enhance the elimination of toxins from the body. Their use is rarely indicated, as only a very few drugs capable of causing severe poisoning have pharmacokinetic parameters that render them amenable to these techniques (Table 29.1.10).

Table 29.1.10 Techniques of enhanced elimination

Technique Suitable toxin
Repeat-dose activated charcoal Carbamazepine
Dapsone
Phenobarbitone
Phenytoin
Salicylate
Theophylline
Urinary alkalinization Phenobarbitone
Salicylate
Haemodialysis Ethylene glycol
Lithium
Methanol
Salicylate
Theophylline
Haemoperfusion Theophylline

Repeat-dose AC (25–50 g every 3–4 h) may enhance drug elimination by interrupting the enterohepatic circulation or by ‘gastrointestinal dialysis’. Gastrointestinal dialysis is the movement of a toxin across the gastrointestinal wall from the circulation into the gut down a concentration gradient that is maintained by charcoal binding. For this technique to be effective, a drug must undergo considerable enterohepatic circulation or, in the case of ‘gastrointestinal dialysis’, have a small volume of distribution, small molecular weight, low protein binding, slow endogenous elimination and bind to charcoal.16,17 The advantages of this technique are that it is non-invasive and simple to carry out.

Alkalinization of the urine enhances urinary excretion of drugs that are filtered at the glomerulus and are unable to be reabsorbed across the tubular epithelium when in an ionized form at alkaline pH. For elimination to be effectively enhanced by this method, the drug must be predominantly eliminated by the kidneys in the unchanged form, have a low pKa, be distributed mainly to the extracellular fluid compartment and be minimally protein bound.18

Haemodialysis (HD) and haemoperfusion (HP) are both very invasive techniques and for that reason are reserved for potentially life-threatening intoxications. Only a small number of drugs that have small volumes of distribution, slow endogenous clearance rates, small molecular weights (HD) and bind to charcoal (HP) will have their rates of elimination significantly enhanced by these procedures.

Antidotes

Very few drugs have effective antidotes. Occasionally, however, timely use of an antidote may be life saving or substantially reduce morbidity, time in hospital or resource requirements. Antidotes that may be indicated in the ED setting are listed in Table 29.1.11. However, it must be remembered that antidotes are also drugs, and are frequently associated with adverse effects of their own. An antidote should only be used where a specific indication exists, and then only at the correct dose, by the correct route, and with appropriate monitoring. Because many antidotes are so infrequently used, obtaining sufficient supplies when the need arises can be difficult. Every ED must review its stocking of antidotes and have a plan for obtaining further supplies should the need arise.

Table 29.1.11 Useful emergency antidotes

Poisoning Antidote
Atropine Physostigmine
Benzodiazepines Flumazenil
Cyanide Dicobalt edetate, hydroxocobalamin
Digoxin Digoxin-specific Fab fragments
Insulin Dextrose
Iron Desferoxamine
Isoniazid Pyridoxine
Methaemoglobinaemia Methylene blue
Methanol and ethylene glycol Ethanol, fomepizole
Organophosphates and carbamates Atropine, oximes
Opioids Naloxone
Paracetamol N-acetyl cysteine
Sulphonylureas Dextrose, octreotide
Tricyclic antidepressants Sodium bicarbonate
Warfarin, brodifacoum Vitamin K

Clinical investigation

Investigations should only be performed if they are likely to affect the management of the patient. They are employed as either screening tests or for specific purposes.

In poisoning, screening tests aim to identify occult toxic ingestions for which early specific treatment might improve outcome. The recommended screening tests for acute poisoning are the 12-lead ECG and the serum paracetamol level. The ECG is used to exclude conduction defects, which may predict potentially life-threatening cardiotoxicity. The serum paracetamol is useful to ensure that paracetamol poisoning is diagnosed within the time available for effective antidotal treatment.

Other specific investigations may be indicated to exclude important differential diagnoses, confirm a specific poisoning for which significant complications might be anticipated, assess the severity of intoxication, assess response to treatment or assess the need for a specific antidote or enhanced elimination technique.

The patient with only minor manifestations of poisoning may require no other blood tests apart from a screening paracetamol level. Pregnancy should be excluded in women of childbearing age by serum or urine β-HCG if necessary. More seriously ill patients may require electrolyte, renal and liver function tests and a full blood count, creatine kinase and arterial blood gases. Urinalysis reveals myoglobinuria in significant rhabdomyolysis.

Routine qualitative drug screening of urine or blood in the overdose patient is rarely useful in planning management.

Measurement of serum drug concentrations is only useful if this provides important diagnostic or prognostic information, or assists in planning management. Some drug levels that may be useful are listed in Table 29.1.12. For most cases, drug overdose management is guided by clinical findings and not by drug levels. Some drugs commonly taken in overdose for which serum concentrations are of no value in planning management are listed in Table 29.1.13.

Table 29.1.12 Drug levels that may be helpful in the management of selected cases of overdose

Carbamazepine
Digoxin
Dilantin
Lithium
Iron
Paracetamol
Phenobarbitone
Salicylate
Theophylline
Valproate

Table 29.1.13 Drug levels that are not helpful in the management of overdose

CNS drugs Cardiovascular drugs
Antidepressants ACE inhibitors
Benzodiazepines Beta-blockers
Benztropine Calcium channel blockers
Cocaine Clonidine
Newer antipsychotics  
Opiates  
Phenothiazines  

Radiology has a limited role in the management of overdose. A chest X-ray is indicated in any patient with a significantly decreased level of consciousness, seizures or hypoxia. It may show evidence of pulmonary aspiration. A computerized tomography scan of the head may be indicated to exclude other intracranial pathology in the patient with an altered mental status. The abdominal X-ray is useful in evaluating overdose of radio-opaque metals including iron, lithium, potassium, lead and arsenic.

Disposition

Both the medical and the psychiatric disposition of the overdose patient must be considered. A good risk assessment is essential to determining timely and safe disposition.

The majority of overdose patients who remain stable at 4–6 h after the ingestion do not need further close monitoring and may be admitted to a non-monitored bed until manifestations of toxicity completely resolve. An emergency observation ward is ideal for this purpose.

Any patient who develops clinical manifestations of intoxication severe enough to require the institution of specific supportive care measures requires admission to an intensive care environment. A few patients will require admission for prolonged monitoring based on the history of the ingestion. For example, anyone with a history of ingestion of colchicine, organophosphates, slow-release theophylline or slow-release calcium channel blockers requires admission because of the possibility of delayed onset of severe toxicity.

Psychiatric evaluation of deliberate self-poisoning cases is indicated as soon as the patient’s medical condition permits. All such patients must be continuously supervised until the psychiatric evaluation has taken place.

References

1 Hawton K, Fagg J. Trends in deliberate self-poisoning and self-injury in Oxford. British Medical Journal. 1992;304:1409-1411.

2 McGrath J. A survey of deliberate self-poisoning. Medical Journal of Australia. 1989;150:317-322.

3 Pond SM. Prescription for poisoning. Medical Journal of Australia. 1995;162:174-175.

4 Murray L, Daly F, Little M, Cadogan M, editors. Toxicology handbook. Sydney: Elsevier, 2007.

5 Kulig K. Initial management of ingestion of toxic substances. New England Journal of Medicine. 1992;326:1677.

6 Krenzelok EP, McGuigan M, Lheureux P. Position statement: ipecac syrup. American Academy of Clinical Toxicology; European Association of Poisons Centres and Clinical Toxicologists. Journal of Toxicology – Clinical Toxicology. 1997;35(7):699-709.

7 Vale JA. Position statement: gastric lavage. American Academy of Clinical Toxicology; European Association of Poisons Centres and Clinical Toxicologists. Journal of Toxicology – Clinical Toxicology. 1997;35(7):711-719.

8 Kulig K, Bar-Or D, Kantrill SV, et al. Management of acutely poisoned patients without gastric emptying. Annals of Emergency Medicine. 1990;14:562-567.

9 Merigian KS, Woodard M, Hedges JR, et al. Prospective evaluation of gastric emptying in the self-poisoned patient. American Journal of Emergency Medicine. 1990;8:479-483.

10 Pond SM, Lewis-Driver DJ, Williams G, et al. Gastric emptying in acute overdose: a prospective randomised controlled trial. Medical Journal of Australia. 1995;163:345-349.

11 Chyka PA, Seger D. Position statement: single-dose activated charcoal. American Academy of Clinical Toxicology; European Association of Poisons Centres and Clinical Toxicologists. Journal of Toxicology – Clinical Toxicology. 1997;35(7):721-741.

12 Barceloux D, McGuigan M, Hartigan-Go K. Position statement: cathartics. American Academy of Clinical Toxicology; European Association of Poisons Centres and Clinical Toxicologists. Journal of Toxicology – Clinical Toxicology. 1997;35(7):743-752.

13 Kirshenbaum LA, Mathew SC, Sitar DS, et al. Whole-bowel irrigation versus activated charcoal in sorbitol for the ingestion of modified-release pharmaceuticals. Clinical Pharmacology Therapy. 1989;46:264-271.

14 Smith SW, Ling LJ, Halstenson CE. Whole-bowel irrigation as a treatment for acute lithium overdose. Annals of Emergency Medicine. 1991;20:536-539.

15 Tenenbein M. Position statement: whole bowel irrigation. American Academy of Clinical Toxicology; European Association of Poisons Centres and Clinical Toxicologists. Journal of Toxicology – Clinical Toxicology. 1997;35(7):753-756.

16 Pond SM. Role of repeated oral doses of activated charcoal in clinical toxicology. Medical Toxicology. 1986;1:3-11.

17 Chyka PA. Multiple-dose activated charcoal and enhancement of systemic drug clearance: summary of studies in animals and human volunteers. Clinical Toxicology. 1995;33:399-405.

18 Winchester JF. Active methods for detoxification. In Haddad LM, Shannon MW, Winchester JF, editors: Clinical management of poisoning and drug overdose, 3rd edn, Philadelphia: WB Saunders, 1998.

19 Whyte IM, Dawson AH, Buckley NA, et al. A model for the management of self-poisoning. Medical Journal of Australia. 1997;167:142-146.

20 Lee V, Kerr JF, Braitberg G, et al. Impact of a toxicology service on a metropolitan teaching hospital. Emergency Medicine. 2001;13:37-42.

29.2 Cardiovascular drugs

Pharmacokinetics

Standard CCB preparations are rapidly absorbed from the gastrointestinal tract, with onset of action occurring within 30 min.1 Pharmacokinetic parameters are shown in Table 29.2.1. Verapamil and diltiazem undergo significant first-pass hepatic clearance. Verapamil is metabolized to norverapamil, which possesses 15–20% of verapamil’s pharmacological activity and is renally excreted. Diltiazem is metabolized to deacetyldiltiazem, which has half the potency of the parent compound and undergoes biliary excretion. The elimination half-lives of all CCBs may be prolonged following massive overdose. Amlodipine has a longer plasma half-life (30–50 h) than other CCBs.

Importantly, slow-release preparations of both verapamil and diltiazem are widely prescribed and are associated with much longer times to peak plasma concentration and clinical effect.

Absorption of β-blockers is rapid, with peak clinical effects occurring within 1–4 h. Pharmacokinetic parameters of the principal β-blockers are detailed in Table 29.2.2. Agents with high lipid solubility, such as propranolol, penetrate the blood–brain barrier better than the water-soluble agents, and hence cause greater central nervous system (CNS) toxicity.

Pathophysiology

CCBs antagonize the entry of extracellular calcium into cardiac and smooth muscle, but not skeletal muscle. Upon entry into cells, calcium participates in mechanical, electrical and biochemical reactions. It is involved in excitation–contraction of cardiac and smooth muscles, as well as phase 0 depolarization in the sinus and atrioventricular (AV) nodes by calcium influx through channels.2 CCBs affect myocardial contractility and slow conduction through the sinus and AV nodes. Contraction of smooth muscle is mediated by calcium influx, which is inhibited by CCBs. This results in vasodilatation and secondary reflex tachycardia from an increase in sympathetic activity.

The different classes of CCB have somewhat different pharmacological and toxic effects, as a consequence of their different binding characteristics to the dihydropyridine (DHP) receptors. Verapamil, a phenylalkylamine, produces more profound cardiac conduction defects and equal reductions in systemic vascular resistance when compared with other CCBs on a mg/kg basis.1 Verapamil is more likely to produce symptomatic decreases in blood pressure, heart rate and cardiac output than diltiazem, a benzothiazepine. The DHPs, which include amlodipine, felodipine, lercanidipine and nifedipine preferentially bind to vascular smooth muscle and predominantly decrease systemic and coronary vascular resistance. With the exception of felodipine, they also produce a reflex tachycardia by the unloading of baroreceptors.

β-blockers prevent the binding of catecholamines to β receptors (β1, β2). β1 receptors are located in the myocardium, kidney and eye, and β2 receptors in adipose tissue, pancreas, liver and both smooth and skeletal muscle. β1 stimulation produces increased chronotropy and inotropy in the heart, increased renin secretion in the kidney and increased aqueous humor production. β2 stimulation relaxes smooth muscle in the blood vessels, bronchial tree, intestinal tract and uterus.

Blockade of β receptors results in increased intracellular cAMP concentrations, with a resultant blunting of the metabolic, chronotropic and inotropic effects of catecholamines. Some β-blockers, especially propranolol, may also impede sodium entry via myocardial fast inward sodium channels, thus slowing phase 0 of the action potential. This results in a prolonged QRS duration on the electrocardiogram and produces cardiotoxicity in overdose more like that of the tricyclic antidepressants.

The different β-blockers have slightly differing pharmacological properties, including selectivity for β adrenoreceptors, intrinsic sympathomimetic activity and membrane-stabilizing activity. The relative affinity for β adrenoreceptors may influence expression of toxicity. Atenolol, esmolol and metoprolol are β1-selective agents, and therapeutic use of these drugs is less likely to produce the peripheral vasoconstriction, bronchospasm and disturbances in glucose homoeostasis that result from β2 inhibition. However, pharmacological specificity decreases with increasing dose.3 Several β-blockers have partial agonist activity such that, although they block the β receptor to catecholamines, they also weakly stimulate the receptor. This partial agonist activity may have a protective effect in overdose.

Clinical features

Cardiovascular

Metabolic

β-blockers

In one large series of patients with β-blocker overdose, 30–40% of patients remained asymptomatic and only 20% developed severe toxicity.4 Toxicity is more likely to develop after ingestion of propranolol, in patients with pre-existing cardiac disease or where there is co-ingestion of other drugs with effects on the cardiovascular system, especially CCBs and cyclic antidepressants.4,5 If β-blocker toxicity is to develop, it is usually observed within 6 h of ingestion.5,6

Sinus node suppression and conduction abnormalities and decreased contractility are typical. First-degree AV block, AV dissociation, right bundle branch block and intraventricular conduction delay have been reported.

Propranol overdose is characterized by cardiotoxicity including prolongation of the QRS interval and ventricular arrhythmias that more closely resemble tricyclic antidepressant overdose; a consequence of the sodium channel blocking effects.

Sotalol has both β-blocker activity and class III antiarrhythmic properties. Class III drugs lengthen the duration of the QT interval owing to prolongation of the action potential in His-Purkinje tissue. Therefore, ventricular arrhythmias are more common with sotalol.

Hypotension occurs as a result of negative inotropic effect. In addition, CNS effects, such as depressed conscious level and seizures, can occur, especially with the more lipid-soluble and membrane-depressant agents such as propranolol. Hypoglycaemia is reported following atenolol overdose.

Treatment

The primary aim in both β-blocker and CCB toxicity is to restore perfusion to vital organs by increasing cardiac output, and the methods used are similar.

Supportive management may include airway and ventilatory support, intravenous fluid administration, transcutaneous or transvenous pacing and administration of inotropes. Severe cases may require placement of a Swan–Ganz catheter and invasive blood pressure monitoring.

Oral-activated charcoal should be administered as soon as practicable to all patients presenting within 2–4 h of ingestion, and to all those presenting after ingestion of slow-release preparations. More aggressive decontamination, with whole-bowel irrigation, is indicated following overdose with slow-release CCBs.7

A number of drugs play a role in the management of significant CCB or β-blocker poisoning, although none is a completely effective antidote. Suggested doses are shown in Table 29.2.4.

Table 29.2.4 Useful drugs in the management of CCB and β-blocker toxicity

  CCBs β-Blockers
Calcium 0.5–1 g (5–10 mLs) calcium chloride or 1–2 g (10–20 mLs) calcium gluconate i.v. over 5–10 minutes. Repeat every 10–15 minutes as required. Further administration guided by serum calcium concentrations.  
Catecholamines Adrenaline (epinephrine) infusion started at 1 μg/kg/min and titrate to maintain organ perfusion. Isoprenaline or adrenaline (epinephrine) infusion titrated to maintain organ perfusion.
Glucagon A bolus dose of 5–10 mg followed by an infusion of 1–5 mg/h. A bolus dose of 5–10 mg followed by an infusion of 1–5 mg/h.
Hyperinsulinaemia euglycaemia Actrapid 1 U/kg i.v. bolus followed by 0.5–1 U/kg/hr infusion. Give with 50% dextrose 50 mL followed by infusion to maintain euglycaemia. Actrapid 1 U/kg i.v. bolus followed by 0.5–1 U/kg/hr infusion. Give with 50% dextrose 50 mL followed by infusion to maintain euglycaemia.

Calcium, an inotropic agent, is the initial drug of choice for CCB toxicity. Administration must be closely monitored, with ionized calcium measured 30 min after commencing the infusion, and then second-hourly.8 Catecholamines are useful in attempting to restore adequate tissue perfusion.

Glucagon, a polypeptide hormone of pancreatic origin, enhances myocardial performance by increasing intracellular cAMP concentrations. This increase in cAMP triggers the release of cAMP-dependent protein kinase, which activates the calcium channels, causing an increase in heart rate and myocardial contractility. It works independently from that of the β-adrenoreceptor stimulation of the heart. Use of glucagon is supported only by case reports and some animal studies. There are no clinical trials supporting its efficacy in either calcium channel or beta-blocker poisoning and its role in management of these poisoning is questioned.9 It is frequently difficult to source adequate stocks of glucagon for use as an inotropic agent.

Hyperinsulinaemic euglycaemia therapy (HIET) is increasingly advocated as therapy for hypotension unresponsive to fluids, calcium salts and inotropes. This therapy is supported by animal work10,11 and promising initial human case reports12 but again clinical trials are lacking. Insulin administration switches cardiac cell metabolism from fatty acids to carbohydrates. It restores calcium fluxes and improves myocardial contractility. The recommended initial dose of actrapid is 1 U/kg i.v. followed by an infusion of 0.5–1 U/kg/h. This should be accompanied by an initial bolus dose of 50 mL 50% dextrose followed by an infusion to maintain euglycaemia.13

Severe propranolol toxicity is usually due to sodium channel blockade and treatment as for tricyclic antidepressant poisoning, including intubation, ventilation and sodium bicarbonate, is appropriate.

There are no clinically effective methods of enhancing the elimination of CCBs or β-blockers.

Clinical features

Two distinct clinical presentations of digoxin toxicity are observed: acute and chronic. Both are characterized by cardiac arrhythmias, and virtually all types of arrhythmia have been reported in the context of digoxin toxicity.14

Acute digoxin overdose in adults is usually intentional. The therapeutic margin for digoxin is relatively narrow, and any ingestion with suicidal intent is regarded as potentially life-threatening.

The non-cardiac manifestations of toxicity are nausea and vomiting and hypokalaemia. Nausea and vomiting occur early and may be the presenting complaint. The most common cardiac manifestations are sinus bradycardia, sinoatrial node arrest and first-, second- or third-degree heart block. Ventricular tachycardia and fibrillation may occur. In significant acute overdose progressive worsening of the conduction disturbance over a period of hours is usually observed.

Chronic digoxin toxicity may be precipitated by therapeutic errors, intercurrent illnesses that decrease renal elimination of digoxin or by drug interactions. Common drug interactions include those with quinidine, CCBs, amiodarone and indometacin. The patient is commonly elderly. Reduced muscle mass and reduced renal function in the elderly mean that both the volume of distribution and rate of elimination of digoxin may be substantially reduced.

Nausea and vomiting are also common manifestations of chronic digoxin toxicity and are frequent presenting symptoms. Neurological manifestations are characteristic of chronic toxicity and include visual disturbances, weakness and fatigue. The most common cardiovascular manifestations of chronic digoxin toxicity are arrhythmias, and these may be sinus bradycardia, atrial fibrillation with slowed ventricular response or a junctional escape rhythm, atrial tachycardia with block and ventricular tachycardia and fibrillation.

Death from digoxin toxicity results from pump failure, severe cardiac conduction impairment or ventricular arrhythmia.

Clinical investigation

The most important investigations are the ECG, serum electrolytes and creatinine and serum digoxin concentration.

The ECG is invaluable in documenting the type and severity of any cardiac conduction defect. Serial ECGs may demonstrate worsening of the cardiac conduction defects as toxicity progresses.

In acute poisoning the serum potassium rises as Na-K ATPase function is progressively impaired. Hyperkalaemia denotes significant acute digoxin toxicity. Prior to the availability of a specific antidote for digoxin poisoning, a serum potassium concentration >5.5 mEq/L was associated with a high probability of lethal outcome.16 Hyperkalaemia is not usually observed in chronic digoxin toxicity. In fact, these patients are frequently hypokalaemic and hypomagnesaemic secondary to chronic diuretic use. Both these electrolyte disorders are important as they exacerbate digoxin toxicity.

Serum digoxin concentrations are extremely useful in assessing and confirming toxicity, but must be carefully interpreted in the context of the clinical presentation. They do not accurately correlate with clinical toxicity. Therapeutic concentrations are usually quoted as 0.6–2.3 nmol/L (0.5–1.8 μµg/L). Significant chronic toxicity may be associated with relatively minor elevations of the serum digoxin concentration. This is particularly the case in the presence of pre-existing cardiac disease, hypokalaemia or hypomagnesaemia. Following acute overdose the serum digoxin concentration is relatively high compared to tissue concentrations, until distribution is completed by 6–12 h post ingestion. However, early concentrations greater than 15 nmol/L indicate serious poisoning.

Treatment

The best outcome is associated with early recognition of digoxin toxicity.

For chronic toxicity with minimal symptoms, management may involve no more than observation, cessation of digoxin administration, correction of hypokalaemia and hypomagnesaemia and appropriate management of any factors that contributed to the development of toxicity. However, presence of any cardiovascular system effects, particularly in elderly patients, is an indication for the administration of Fab fragments of digoxin-specific antibodies. The potential lethality of chronic digoxin poisoning is often underestimated with the result that digoxin antibody fragments are inappropriately withheld.16 From a purely economic view, the reduction in length of stay as a result of treatment with digoxin-specific antibodies outweighs the expense of the therapy.17

Following acute overdose the patient should be initially managed in a monitored area with full resuscitative equipment available. Immediate attention to the airway, breathing and circulation may be required. Intravenous access should be established and blood sent for urgent electrolytes and serum digoxin concentration. Although digoxin is well bound by charcoal, administration is usually difficult because of repetitive vomiting, and attempts should not detract from other interventions.

The specific antidote to digoxin poisoning is Fab fragments of digoxin-specific antibodies, which should be administered as soon as possible in any potentially life-threatening digoxin intoxication. Commonly accepted indications for the administration of Fab fragments are listed in Table 29.2.5.

Table 29.2.5 Indications for administration of Fab fragments of digoxin-specific antibodies following acute overdose

Hyperkalaemia (K > 5.0 mmol/L) associated with digoxin toxicity
History of ingestion of more than 10 mg of digoxin
Haemodynamically unstable cardiac arrhythmia
Cardiac arrest from digoxin toxicity
Serum digoxin concentration greater than 15 nmol/L

Fab fragments of digoxin-specific antibodies

These are derived from IgG antidigoxin antibodies produced in sheep. Removal of the Fc fragments of the antibodies greatly reduces the potential for hypersensitivity reactions and contributes to the remarkable safety profile of the product. Intravenously administered Fab fragments bind digoxin in the intravascular space on a mole-for-mole basis. As binding continues, digoxin moves down a concentration gradient from the tissue compartments to the intravascular compartment. Bound digoxin is inactive. A clinical response is usually observed within 20–30 min of administration. The Fab–digoxin complexes are excreted in the urine.

The extraordinary clinical efficacy of digoxin-specific fragments has been well documented in a multi-centre study.17 The same study also demonstrated the safety of the product, with the only adverse reactions reported being hypokalaemia (4% incidence) and worsening of congestive cardiac failure (3%).

The correct dose of Fab fragments may be calculated on the basis that 40 mg (one vial) will bind 0.6 mg of digoxin. If the dose ingested is unknown and/or a steady-state serum digoxin concentration is not available, dosing of Fab fragments must be empiric. Following acute overdose a reasonable approach to empiric dosing is to give five vials initially and then repeat until a clinical response is observed. Smaller doses (two vials) are usually sufficient to reverse the effects of chronic toxicity.

It is important that ED staff are aware of the amount and location of supplies of Fab fragments within their own institution, and know the most rapid way to acquire further stocks should the need arise.

Serum digoxin concentrations will be extremely high following the administration of Fab fragments because most assays measure both bound and unbound digoxin.

References

1 Robertson RM, Robertson D. Drugs used for the treatment of myocardial ischaemia. In: Gilman AG, Hardman JG, Limbird LE, et al, editors. Goodman and Gilman’s: The pharmacological basis of therapeutics. 9th edn. New York: Pergamon Press; 1996:770.

2 Antman EM, Stone PH, Muller JE, et al. Calcium channel blocking agents in the treatment of cardiovascular diseases: Part E Basic and clinical electrophysiological effects. Annals of Internal Medicine. 1980;93:875-885.

3 Lewis RV, McDevitt DG. Adverse reactions and interactions with beta-adrenoreceptor blocking drugs. Medical Toxicology. 1986;1:343-361.

4 Taboulet P, Cariou A, Berdeaux A, et al. Pathophysiology and management of self-poisoning with beta-blockers. Journal of Toxicology and Clinical Toxicology. 1993;31:531-551.

5 Reith DM, Dawson AH, Epid D, et al. Relative toxicity of beta blockers in overdose. Journal of Toxicology and Clinical Toxicology. 1996;34:273-278.

6 Love J, Howell JM, Litovitz TL, et al. Acute beta blocker overdose: factors associated with the development of cardiovascular morbidity. Journal of Toxicology and Clinical Toxicology. 2000;38:275-281.

7 Buckley N, Dawson AH, Howarth D, et al. Slow release verapamil poisoning. Use of polyethylene glycol whole bowel lavage and high dose calcium. Medical Journal of Australia. 1993;158:202.

8 Pertoldi F, D’Orlando L, Mercante WP. Electromechanical dissociation 48 hours after atenolol overdose: usefulness of calcium chloride. Annals of Emergency Medicine. 1998;31:777-781.

9 Bailey B. Glucagon in beta-blocker and calcium channel blocker overdoses: a systematic review. Journal of Toxicology Clinical Toxicology. 2003;41:595-602.

10 Kline JA, Leonova E, Raymond RM. Beneficial myocardial metabolic effects of insulin during verapamil toxicity in the anesthetized canine. Critical Care Medicine. 1995;23:1251-1263.

11 Holger JS, Engerbretsen KM, Fritzlar SJ, et al. Insulin versus vasopressin and epinephrine to treat b-blocker toxicity. Clinical Toxicology. 2007;45:396-401.

12 Yuan I, Kerns WP, Tomaszewski CA, et al. Insulin glucose as adjunctive therapy for severe calcium channel antagonist poisoning. Journal of Toxicology Clinical Toxicology. 1999;37:463-474.

13 Megarbane B, Karyo S, Baud FJ. The role of insulin and glucose (hyperinsulinaemia/euglycaemia) therapy in acute calcium and beta-blocker poisoning. Toxicology Reviews. 2004;23(4):214-222.

14 Moorman JR, Pritchett ELC. The arrhythmias of digitalis intoxication. Archives of Internal Medicine. 1985;145:1289-1292.

15 Bismuth C, Gaultier M, Conso F, et al. Hyperkalemia in acute digitalis poisoning: prognostic significance and therapeutic implications. Clinical Toxicology. 1973;6:153-162.

16 Marik PE, Fromm L. A case series of hospitalised patients with elevated digoxin levels. American Journal of Medicine. 1998;105(2):110-115.

17 DiDomenico RJ, Walton SM, Sanoski CA, et al. Analysis of the use of digoxin immune Fab for the treatment of non-life-threatening digoxin toxicity. Journal of Cardiovascular Pharmacology & Therapeutics. 2000;5(2):77-85.

18 Antman EM, Wenger FL, Butler VP, et al. Treatment of 150 cases of life threatening digitalis intoxication with digoxin specific Fab antibody fragments: final report of multicenter study. Circulation. 1990;81:1744-1752.

19 Seger D. Clonidine Toxicity Revisited. Clinical Toxicology. 2002;40(2):145-155.

20 Nichols MH, King WD, James LP. Clonidine poisoning in Jefferson County, Alabama. Annals of Emergency Medicine. 1997;29:511-517.

21 Erickson SJ, Duncan A. Clonidine poisoning – an emerging problem: Epidemiology, clinical features, management and preventative strategies. Journal of Paediatrics and Child Health. 1998;34(3):280-282.

22 McVey FK, Corke CF. Extracorporeal circulation in the management of massive propranolol overdose. Anaesthesia. 1991;46:744-746.

29.3 Central nervous system drugs

Benzodiazepines

Pharmacology

Benzodiazepines possess a shared structure comprising a benzene ring fused to a diazepine ring. The pharmacologically significant benzodiazepines also demonstrate a 5-aryl substituent.

Most benzodiazepines are highly lipid soluble and rapidly absorbed following oral administration. The more water-soluble agents, such as temazepam and oxazepam, are more slowly absorbed. Following ingestion, peak plasma concentrations are reached within 90 min for midazolam and diazepam, compared to 120–180 min for temazepam and oxazepam. Following intramuscular injection absorption of benzodiazepines is often erratic, except for lorazepam and midazolam.

Plasma protein binding is variable. Diazepam has the highest (99%) and alprazolam the lowest (70%) plasma protein binding. The unbound fraction is able to cross the blood–brain barrier and interact with specific receptors in the CNS. Benzodiazepines are widely distributed to the body tissues, with volumes of distribution ranging from 0.3 to 5.5 L/kg.

The duration of action for benzodiazepines depends on a number of factors, including the rate of redistribution from the CNS compartment to the body tissues, the metabolism and excretion of the drug and the sensitivity of the benzodiazepine receptor to its agonist. Drugs that are lipophobic tend to have a shorter measured plasma half-life but a longer duration of action. This reflects slower redistribution from the CNS compartment. Lipophilic drugs rapidly redistribute to body fat and muscle and, therefore, have a rapid onset but relatively short duration of CNS effect together with relatively longer plasma half-lives. Repeated dosing eventually saturates peripheral body stores and this ‘stored’ drug may then leach out resulting in prolonged pharmacological activity.

The metabolism of most benzodiazepines includes both phase I oxidative and phase II conjugative processes, with phase I producing pharmacologically active metabolites. The major metabolite of diazepam is desmethyldiazepam, which is pharmacologically active and has a longer half-life than its parent compound. Lorazepam, temazepam and oxazepam only undergo phase II metabolism.

Gamma-aminobutyric acid (GABA) is an inhibitory neurotransmitter found predominantly in the basal ganglia, the hippocampus, hypothalamus, cerebellum and the dorsal horn of the spinal cord.1 GABA interacts with two receptors, GABA-A and GABA-B, resulting in the influx of chloride through a ligand-gated ion channel. The former receptor is the predominant site of benzodiazepine action. By binding to the GABA-A receptor complex at a specific site, benzodiazepines enhance the binding of GABA at GABA-A, which in turn opens more chloride channels, and hence produces their sedative, hypnotic, anxiolytic and anticonvulsant effects. GABA-B is mainly involved in feedback mechanisms and the control of muscle tone. GABA receptor subunits have been identified, and the sensitivity and specificity of individual benzodiazepines is determined by their interaction with these subunits. Tolerance develops to most of the effects of benzodiazepines, and may be associated with downregulation of GABA receptors.

Clinical features

Overdose

Death as a result of pure benzodiazepine overdose is very uncommon.2 When death is reported, it is usually in the setting of a mixed overdose including other CNS depressants such as alcohol, antidepressants, phenothiazines and narcotics. The most common manifestation of overdose is drowsiness, which may progress to stupor depending on patient characteristics, co-ingestants and dose. Coma is uncommon. Other features characteristic of benzodiazepine overdose are respiratory depression, hypothermia and hypotension, though these are not usually life-threatening.3 The duration of effect varies from 6 to 36 h, depending on the drug.

Respiratory insufficiency in the setting of benzodiazepine overdose may be due to an increase in upper airway resistance and work of breathing rather than central apnoea.4

The duration of benzodiazepine effect varies between 6 and 36 h depending on the agents(s) involved. The effects on the CNS are exacerbated if there is co-ingestion of other CNS depressants such as alcohol, antidepressants, phenothiazines or narcotics. A fatal outcome is more likely in this setting. Acute alcohol ingestion tends to delay benzodiazepine metabolism whereas chronic alcohol ingestion induces metabolic pathways and may increase clearance rates of these drugs.5

Non-benzodiazepine sedative-hypnotics

Antipsychotic Drugs

Pharmacology

This large group of drugs can be classified as typical or atypical (Table 29.3.1), according to structure (Table 29.3.2) or according to neuroreceptor-binding affinity. The latter may offer the most reliable prediction of the risk of toxicity.18

Table 29.3.1 Typical and atypical antipsychotic drugs

Typical Atypical
Chlorpromazine Mesoridazine
Fluphenazine Thioridazine
Perphenazine Clozapine
Prochlorperazine Olanzapine
Trifluoperazine Risperidone
Haloperidol Remoxipride
Thiothixene Loxapine
Molindone Quetiapine

Table 29.3.2 Structural classification of the antipsychotics

Structural class Generic name
Phenothiazines  
Aliphatic Chlorpromazine
Triflupromazine
Promethazine
Piperazine Fluphenazine
Perphenazine
Prochlorperazine
Trifluoperazine
Piperidine Mesoridazine
Thioridazine
Butyrophenone Haloperidol
Thioxanthene Droperidol
Chlorprothixene
Thiothixene
Dihydroindolone Molindone
Dibenzoxazepine Loxapine
Clozapine
Olanzapine
Diphenylbutylpiperidine Pimozide
Benzisoxazole Risperidone
Benzamides Sulpiride
Remoxipride

Atypical drugs are defined as such on clinical and pharmacological grounds. Clinically, they produce fewer extrapyramidal side effects and tardive dyskinesias. For this reasons the newer atypical antipsychotic agents have largely replaced the traditional agents as first-line treatment of schizophrenia. Pharmacologically, they may be regarded as atypical for a variety of reasons including low D2-dopamine receptor potency, low D2-receptor occupancy in the mesolimbic and nigrostriatal areas and high affinities for M1-muscarinic, D1– and D4-dopamine and 5-HT1A- and 5-HT2A-serotonin receptors.19 This produces three broad functional groups of atypical antipsychotics: the D2-, D3-receptor antagonists such as amisulpiride, the D2, α1, 5-HT2A-receptor antagonists such as risperidone, and the broad-spectrum multiple receptor antagonists such as clozapine, quetiapine and olanzapine.19

The therapeutic and predominant toxic effects of these drugs are related to their blockade of the D2-subtype dopamine receptors. These are located throughout the brain in the basal ganglia, hypothalamus, pituitary, medulla and the mesocortical and mesolimbic pathways. The antipsychotic effect of a drug is mediated by its blockade of D2 receptors in the mesocortical and mesolimbic pathways. The development of extrapyramidal effects is closely related to a drug’s affinity with D2 receptors in the basal ganglia. D2-receptor blockade in the pituitary can cause elevated prolactin levels, with resulting galactorrhoea and gynaecomastia. Blockade of D2 receptors in the hypothalamus affects body temperature regulation: hypothermia or hyperthermia may result. The strong antiemetic effect of some antipsychotic agents is regulated through D2-receptor blockade in the medulla.

The blockade of other neuroreceptors and the relative ratio to D2-receptor blockade predicts the likelihood of adverse effects at therapeutic dosing and in overdose. Blockade of α1-adrenergic receptors results in postural hypotension of varying degrees, depending on binding affinity. Significant α2-receptor blockade occurs with clozapine but the clinical importance of this is not clear. H1-histamine receptor blockade correlates with sedation and, to a lesser extent, hypotension. Sedation, along with delirium, hallucinations, mydriasis, flushing, dry skin, urinary retention and ileus, is seen with M1-acetylcholine receptor blockade. Agents that possess a relatively higher anticholinergic activity compared to dopaminergic activity have a lower risk for inducing extrapyramidal side effects. Examples include chlorpromazine, thioridazine, clozapine and olanzapine. The reverse is also true: those drugs with a higher dopaminergic effect in relation to their anticholinergic activity have a higher risk of inducing extrapyramidal side effects, e.g. fluphenazine, prochlorperazine and haloperidol. Serotonin antagonism may be an important mechanism in the antipsychotic action and responsible for the low incidence of extrapyramidal side effects seen with the newer atypical antipsychotics such as clozapine and olanzapine. These drugs, which tend to have a high 5-HT2A antagonism in relation to D2 antagonism, can be given in smaller doses to produce the same therapeutic effect.

Phenothiazine antipsychotics also have a quinidine-like effect and can produce a variety of ECG changes, both at therapeutic doses and in overdose. Thioridazine and mesoridazine are considered to be the most cardioactive, and also possess calcium channel blocking ability, which may contribute to the cardiotoxicity observed in overdose of these agents.

Generally, the pharmacokinetics of this heterogeneous group of drugs are similar. They are rapidly and well absorbed after oral administration. Peak plasma concentrations occur between 1 and 6 h following oral administration and from 30 to 60 min following intramuscular administration. Those agents that possess a considerable anticholinergic effect may show delayed absorption after ingestion. Most antipsychotics exhibit a relatively high plasma protein binding of between 75% and 99%, are widely distributed to the tissues and have high volumes of distribution, ranging from 10 to 40 L/kg. Clozapine and risperidone are exceptions with smaller volumes of distribution (2 and 1 L/kg respectively). Only about 1% of an ingested dose is excreted unchanged in the urine, with the majority of drugs undergoing extensive hepatic metabolism, some with significant enterohepatic circulation.

Clinical features

Adverse effects

Adverse effects following therapeutic dosing may be idiosyncratic or dose-related, and may occur after initiation of the medication in question or late into a course of treatment.

Extrapyramidal movement disorders

Up to 90% of patients receiving antipsychotic medication will experience some extrapyramidal side effects, and these often result in the cessation of treatment.20 Of the four recognized extrapyramidal syndromes, acute dystonia, parkinsonism and akathisia are reversible and tend to occur early in a course of treatment. Tardive dyskinesia is irreversible but occurs after months to years of treatment. Clozapine, olanzapine and quetiapine are not associated with extrapyramidal syndromes.19

The pathophysiology of acute dystonic reactions is not fully understood, but involves disruption of the dopaminergic-cholinergic-GABA balance in the basal ganglia. Reactions are idiosyncratic and equally frequent following a single therapeutic ingestion or an overdose. Risk factors for developing an acute dystonic reaction following antipsychotic medication are the use of antipsychotic drugs with a high D2-dopaminergic, low M1-muscarinic and low 5-HT2A-serotonergic receptor binding affinity; young and male patients; the use of depot preparations and the recent use of alcohol.21,22 Reactions may present in varied forms and may be spasmodic or sustained, but are always involuntary. The muscles of the face, trunk and neck are commonly involved, but other sites may also be affected. About half of all cases occur within 48 h of dosing.20 The overall incidence of acute dystonic reactions varies considerably: rates of 3.5% have been reported for chlorpromazine, and 16% for haloperidol.21

Akathisia is dose-related, can occur at any age, and tends to appear some days after beginning treatment. It is thought to be due to D2-dopaminergic blockade in the mesocortical pathways.23 Drug-induced parkinsonism is more common in the elderly and tends to be seen with high-potency agents that block the postsynaptic D2-dopaminergic receptors in the nigrostriatal area. Tardive dyskinesia appears after months or years of antipsychotic treatment. It is seen with all antipsychotics except clozapine, and has a prevalence of between 27% and 35% in patients on long-term therapy.24 It is thought to be the result of an increased number and sensitivity of dopaminergic receptors in the nigrostriatal area of the brain, a response to long-term blockade.

Seizures

Antipsychotic drugs lower the seizure threshold.25 They also produce EEG changes that vary depending on the agent.26 Organic brain disease, epilepsy, drug-associated seizures and polypharmacy are risk factors for the development of seizures. They are more likely with chlorpromazine, clozapine and loxapine.

Neuroleptic malignant syndrome

This idiosyncratic adverse reaction to antipsychotic medication therapy is rare. It occurs early in the treatment course or after changes in dose. Neuroleptic malignant syndrome (NMS) has been reported with all typical antipsychotics but is particularly associated with higher potency drugs such as haloperidol and fluphenazine. In the atypical group, NMS has been reported with clozapine, olanzapine and risperidone.2830 Pooled data studies suggest the incidence is somewhere between 0.07% and 0.2%, although some have described incidences of up to 12.2%.

Typically, patients are male (male : female ratio 2 : 1), with symptoms developing over 1–3 days. Risk factors associated with the development of NMS include the use of high-potency agents and depot preparations, organic brain disease, past history of NMS, dehydration and interactions with other drugs such as lithium and anticholinergics.31 The characteristic clinical features are a temperature ≥38 °C, muscle rigidity, altered consciousness and autonomic dysfunction. Other features that may be seen are an elevated creatine kinase, leukocytosis, elevated hepatic transaminases, renal failure and metabolic acidosis. There is no specific test to confirm or exclude the diagnosis, which is reliant upon clinical and historical data. Alternative diagnoses must be excluded, especially infection (including meningitis and encephalitis). Other differential diagnoses include heat stroke, thyrotoxicosis, intracranial haemorrhage, phaeochromocytoma, tetanus, serotonin syndrome, drug overdose (MAOI, sympathomimetics and lithium), substance/alcohol withdrawal and malignant hyperthermia. The mortality rate has been reduced from 30% to 5–11% mainly as a result of improved intensive supportive care.32 Death is usually secondary to respiratory or cardiovascular failure; however, renal failure secondary to myoglobinuria, arrhythmias, pulmonary embolism and disseminated intravascular coagulation are also reported.

Overdose

Most patients with serious poisoning display manifestations of cardiovascular and/or CNS toxicity. Isolated antipsychotic overdose is rarely fatal. Peak toxicity is usually seen from 2 to 6 h following ingestion but may be delayed especially after thioridazine overdose.33 Delayed onset of life-threatening cardiotoxicity is also reported following amisulpride overdose.34 CNS effects vary greatly, depending on individual susceptibility, dose ingested and the presence of co-ingestants. Lethargy is common to most patients, with effects potentially progressing to confusion, ataxia, coma and seizures. Ingestion of more than 300 mg of olanzapine or 3 g of quetiapine is likely to cause CNS depression significant enough to require intubation. Seizures are more often seen following overdose with loxapine or clozapine, and are usually generalized.35 Paradoxically, agitation may also be observed, especially in the setting of mixed overdose, and following overdose with clozapine, olanzapine or thioridazine.

Life-threatening cardiotoxicity is unusual, except in the setting of piperidine phenothiazine overdoses, e.g. thioridazine, which is associated with QRS widening, QT prolongation, ventricular tachycardia and torsade des pointes.36,37 Torsades des pointes is also reported in large haloperidol overdoses, either following deliberate self-poisoning or in the setting of excessive intravenous therapy in critically ill patients.38 More common cardiovascular effects are hypotension (initially postural) and tachycardia. Uncommon effects are hypertension and bradycardia. ECG abnormalities are not unusual in significant overdoses and can range from simple ventricular ectopics to conduction abnormalities, QRS widening, ventricular tachycardia and torsades des pointes. QT prolongation has been reported in the setting of thioridazine overdose and more recently quetiapine and amisulpride ingestions.34,3941 The prolonged corrected QT observed in a number of reported quetiapine overdoses may be the result of the underlying sinus tachycardia observed, rather than an indicator of significant cardiotoxicity.39,40 Amisulpride overdose can result in severe cardiotoxicity, characterized by intraventricular conduction abnormalities, QT prolongation and torsades des pointes.34

Temperature abnormalities may also occur, and commonly manifest as mild hypothermia. Hyperthermia may be seen in the setting of a high environmental temperature and seizures. Following ingestions of aliphatic and piperidine phenothiazines, clozapine and olanzapine, significant anticholinergic toxicity may occur.

The diagnosis of antipsychotic drug overdose is based on a history of ingestion and the presence of symptoms and signs in keeping with the expected findings, as outlined above. Qualitative serum and urine drug screening can be used to detect the presence of many of the antipsychotic drugs, but a high false–negative rate makes these screens notoriously unreliable. Quantitative levels may also be performed, but the results do not correlate with clinical findings and do affect management. The differential diagnosis of antipsychotic drug overdose includes meningitis and other CNS infections, stroke and head injury, as well as other drug toxicities, including tricyclic antidepressants, sedatives, alcohols, anticholinergic drugs and anticonvulsants. Many other agents have also been reported as causing acute dystonic reactions, including tricyclic antidepressants, antihistamines and anticonvulsants.

Treatment

The management of antipsychotic drug overdose is essentially supportive. Patients should undergo an initial resuscitation period with a careful airway assessment and, if necessary, endotracheal intubation. Ventilation should be supported with supplemental oxygen and mechanical ventilation if indicated. Hypotension should be treated with Trendelenburg positioning, intravenous fluids and, if resistant, inotropic agents, preferably with some α-agonist properties. All patients should be placed on a cardiac monitor, have a 12-lead ECG recorded and an intravenous cannula inserted, with blood being drawn for full blood examination, electrolytes, creatine kinase and renal function. If paracetamol overdose is suspected appropriate levels should be measured.

Administration of activated charcoal should be considered unless there has been considerable delay in presentation. Multidose charcoal has not been shown to be of benefit in antipsychotic drug overdose. The use of extracorporeal blood purification techniques to enhance drug elimination is not effective, owing to the large volume of distribution and high tissue-protein binding of these drugs.

Seizures should be treated with benzodiazepines such as diazepam or clonazepam as the first-line agents. For resistant seizure activity phenobarbitone may be needed.

Cardiac arrhythmias should be treated according to advanced cardiac life support protocols. However, type IA antiarrhythmics should be avoided in the setting of QRS widening or conduction abnormalities, as they may exacerbate the toxicity. Serum alkalinization, to a pH of 7.45–7.55, should be performed in the presence of significant QRS widening or life-threatening arrhythmias. Intravenous magnesium and chemical or electrical overdrive pacing may be required to control torsades des pointes.

Clinical features

Adverse effects

The adverse effects of bupropion are relatively mild compared to other antidepressants. Mild hypertension has been noted, but has usually occurred in the already hypertensive.44 Postural hypotension has also been observed in sporadic patients.45 QRS or QT prolongation is not seen with bupropion at therapeutic doses.46 Neurological side effects occur more commonly with headache, insomnia, agitation and seizures being reported.47,48 Minor gastrointestinal irritation and priapism are also reported.

Tricyclic antidepressants

Tricyclic antidepressants (TCAs) have long been the leading cause of death from prescription drug overdose. However, they are increasingly being replaced in clinical practice by newer agents, which appear to be significantly safer in overdose. The TCAs currently available in Australia are listed in Table 29.3.3. Reported mortality rates for intentional TCA overdose range from 2% to 5%.54 The vast majority of successful TCA suicides do not reach hospital but die at home.55 The ingestion of 10 mg/kg or more of a TCA is potentially fatal, though there are differences in toxicity within the group. In Australia, doxepin is associated with the greatest lethality.56

Table 29.3.3 Tricyclic antidepressants available in Australia

Amitriptyline
Clomipramine
Dothiepin
Doxepin
Imipramine
Nortriptyline
Trimipramine

Clinical features

The clinical features of TCA overdose include anticholinergic, cardiovascular and CNS effects. The type and severity of clinical manifestations are dose-related (Table 29.3.4) Onset is usually rapid and, following large ingestions, rapid deterioration in clinical status within 1–2 h is characteristic.

Table 29.3.4 Tricyclic antidepressants: dose-related risk assessment

Dose Effect
<5 mg/kg Minimal symptoms
5–10 mg/kg Drowsiness and mild anticholinergic effects Major toxicity not expected
>10 mg/kg Potential for all major effects to occur within 2–4 h of ingestion
>30 mg/kg Severe toxicity with pH-dependent cardiotoxicity and coma expected to last >24 h

Adapted from Toxicology Handbook. Murray L, Daly F, Little M, Cadogan M (eds), Sydney: Elsevier; 2007.

The clinical features of central and peripheral anticholinergic toxicity are described elsewhere in this book. Anticholinergic delirium is most commonly observed following a modest TCA overdose or early in the course of more significant ingestions. Large overdoses usually lead to coma, which obscures any evidence of anticholinergic delirium. Seizures are characteristic of TCA overdose and usually occur early in the clinical course. Overall the rate is quoted to be 3–4%.59 Myoclonic jerking is also associated with TCA overdose.

Sinus tachycardia is commonly observed following TCA overdose and is usually due to the anticholinergic effects of the TCA, rather than sodium channel blockade. More serious cardiac arrhythmias can develop as a consequence of the effects on the fast sodium channels and cardiac depolarization and conduction. These include supraventricular tachycardia (with or without aberrancy), ventricular tachycardia, torsades des pointes (augmented by potassium channel blocking effects) and ventricular fibrillation. Junctional or idioventricular rhythms, second- or third-degree heart block or asystole can also occur.60 Hypotension is commonly observed and is due to both peripheral vasodilatation and impaired myocardial contractility.

Clinical investigation

Serum TCA concentrations correlate poorly with the clinical severity of TCA intoxication. The single most important investigation in assessing the patient following a TCA overdose is the 12-lead ECG. The degree of prolongation of the QRS interval is predictive of the risk of both ventricular arrhythmias and seizures.61 The positive and negative predictive values of ECG changes in TCA poisoning in one study were 66% and 100%, respectively.62 A QRS duration of >120 ms in the setting of a TCA overdose indicates cardiotoxicity. A terminal R wave >3 mm in lead aVR may be a more useful predictor of seizures or arrhythmias than QRS duration.63 A patient may exhibit significant CNS toxicity despite a normal ECG.

Treatment

The management of TCA poisoning is largely supportive. In particular, it involves maintenance of the airway, ventilation and blood pressure and control of ventricular arrhythmias and seizures.

The potential for rapid deterioration in clinical status must be appreciated and patients with a history of recent TCA overdose should be managed in a closely monitored environment. Intravenous access should be established, supplemental oxygen administered and cardiac monitoring commenced on arrival. There should be a relatively low threshold for performing endotracheal intubation in the patient with deteriorating mental status because hypoxia and acidosis exacerbate cardiotoxicity. Patients with a decreased level of consciousness or anticholinergic symptoms should undergo urinary catheterization.

Oral activated charcoal should be administered to all patients with significant ingestions after the airway is secured (if necessary). All the TCAs have very large volumes of distribution, and so techniques of enhancing elimination are not helpful.

Hypotension should initially be managed with i.v. fluids. If blood pressure fails to respond to infusions of crystalloid or colloid, then sodium bicarbonate should be tried even in the presence of a normal QRS. If there is still no response inotropes should be started. The ideal inotrope is one that will overcome α-adrenergic blockade and have little stimulatory effect on β receptors. For these reasons, noradrenaline (norepinephrine) is usually regarded as the inotrope of choice. Dopamine is best avoided as it stimulates β receptors (and may lead to a paradoxical decrease in blood pressure) and, as an indirectly acting sympathomimetic, it will become ineffective when neuronal stores of noradrenaline (norepinephrine) are depleted in the presence of a potent reuptake pump inhibitor such as TCAs.64

Seizures, delirium and hyperthermia should be controlled using standard techniques.

Flumazenil should be avoided in the setting of a TCA overdose because its action may precipitate refractory seizures and increase morbidity and mortality.65

Disposition

Patients with a history of TCA ingestion and who have received oral-activated charcoal but show no signs of toxicity after 6 h of observation are safe for medical discharge and ready for psychiatric evaluation.68 Those with significant cardiovascular or CNS toxicity should be admitted to an intensive care environment. Those with mild CNS manifestations only should be observed in hospital until these resolve.

Serotonin reuptake inhibition with α2-adrenergic antagonism

Clinical features

Adverse effects

The most common adverse effects attributed to the SSRIs are gastrointestinal symptoms, sexual dysfunction, headache, insomnia, jitteriness, dizziness and fatigue.70 Inappropriate antidiuretic hormone secretion is also reported, particularly in the elderly.71 The adverse effect most likely to result in presentation to the ED is the development of serotonin syndrome (see below) as a result of an interaction between two drugs that enhance serotonergic activity or where there has been an insufficient ‘wash-out’ period between ceasing one such drug and commencing another.

Symptoms usually begin shortly after the commencement of a serotoninergic drug, or the administration of two different classes of drugs that increase serotonin levels synergistically, for example lithium and fluoxetine. In addition, potential drug interaction may arise when the appropriate ‘change-over’ period between drugs is not observed.77 A severe form of the syndrome may develop some hours following overdose with an SSRI or, more commonly, following overdose with multiple serotonergically active drugs.7880

The diagnosis of serotonin syndrome is clinical and based upon the presence of the triad of alteration in behaviour-cognitive ability, autonomic function and neuromuscular activity. A grading system has been proposed.81 In its most benign form the patient experiences anxiety and apprehension, but altered sensorium with confusion occurs in 50% of reported cases.69 Seizures may occur.79 Abnormal neuromuscular activity, caused by increased brainstem and spinal-cord serotonin levels, manifests as increased rigidity (more in the lower than the upper limbs), hyperreflexia, involuntary jerks and resting extremity tremor. Hyperthermia, secondary to increased muscle activity is a common feature and may lead to confusion with NMS. Diaphoresis, diarrhoea and rigors are common. Cardiovascular instability may occur. Although most patients recover, fatalities are reported.79,80 There is no correlation with drug levels, and serotonin syndrome remains a clinical diagnosis.69 The differential diagnosis includes NMS, acute dystonia, hyperadrenergic states (e.g. cocaine toxicity), anticholinergic syndrome and malignant hyperthermia. Decision algorithms have been developed to help the clinician distinguish serotonin syndrome from other conditions.82

Carbamazepine

Clinical features

Onset of clinical features of carbamazepine toxicity may be delayed many hours following overdose due to delayed absorption.86,87 The clinical features are predominantly neurological and include CNS depression which may progress to coma, drowsiness, ataxia, nystagmus and dystonia. Paradoxical seizures are also reported in severe poisoning.88,89 Carbamazepine toxicity may also manifest as the anticholinergic syndrome, although the delirium may be masked by coma as the intoxication progresses.

Minor ECG changes may be observed in severe carbamazepine poisoning but significant cardiovascular effects are rare.90

Phenytoin

Valproic acid

Pharmacology

Valproic acid is a simple monocarboxylic acid, chemically unrelated to any other class of antiepileptic drug. Its mechanism of action is thought to involve but not be limited to a decrease in breakdown of GABA-A and increased conversion of GABA from glutamate.95

Oral absorption of valproic acid is rapid with peak levels usually occurring within 4 h but this may be delayed following overdose.96 The volume of distribution is very small at 0.13–0.23 L/kg and there is extensive plasma protein binding which may be saturated in overdose. The drug undergoes extensive hepatic metabolism and has active metabolites. The elimination half-life of 7–15 h may be prolonged in overdose.97

Clinical presentation

The clinical course following valproate overdose is dose-dependent. Ingestions of less than 200 mg/kg are usually asymptomatic or result in minor drowsiness only.98 For ingestions >200 mg/kg, coma may develop and, for ingestions >400 mg/kg, there is a risk of prolonged profound coma and metabolic disorders including hyperammonaemia, hypernatraemia, hypocalcaemia and bone-marrow depression.99 Death from cerebral oedema is reported.

L-carnitine

L-carnitine is an amino acid carrier molecule used to transport long-chain fatty acids across to mitochondria. It is synthetized chiefly in liver and kidney. It is available in oral and intravenous forms and appears to have an acceptable safety profile.100 It is postulated that L-carnitine could provide benefit in patients with concomitant hyperammonemia encephalopathy and/or hepatotoxicity as there is some evidence that it reduces ammonia concentrations in acute valproate overdose.101,102 While L-carnitine has been used in a number of case reports definitive evidence of efficacy is lacking.

Newer antiepileptic drugs

A number of new antiepileptic drugs with differing pharmacokinetic properties and mechanisms of action have been introduced into clinical practice over the last decade. These include oxcarbazepine, gabapentin, felbamate, vigabatrin, topiramate and tiagabine. The toxicity profiles of these drugs in overdose are not yet well established. Gabapentin, felbamate and lamotrigine are reported to cause only minor CNS effects in overdose.99103 Vigabatrin overdose has resulted in severe agitation.104

References

1 Goodchild CS. GABA receptors and benzodiazepines. British Journal of Anaesthesia. 1993;71:127-133.

2 Greenblatt DJ, Allen MD, Noel BJ, et al. Acute overdosage with benzodiazepine derivatives. Clinical Pharmacology and Therapy. 1977;21:497-514.

3 Busto U, Kaplan HL, Sellers EM. Benzodiazepine-associated emergencies in Toronto. American Journal of Psychiatry. 1980;137:224-227.

4 Gueye PN, Lofaso F, Borron SW, et al. Mechanism of respiratory insufficiency in pure or mixed drug-induced coma involving benzodiazepines. Clinical Toxicology. 2002;40:35-47.

5 Tanaka E. Toxicological interactions between alcohol and benzodiazepines. Clinical Toxicology. 2002;40:69-75.

6 Haverkos GP, DiSalvo RP, Imhoff TE. Fatal seizures after flumazenil administration in a patient with mixed overdose. Annals of Pharmacotherapy. 1994;28:1347-1349.

7 Gaulier JM, Merle G, Lacassie E, et al. Fatal intoxications with chloral hydrate. Journal of Forensic Sciences. 2001;46:1507-1509.

8 Regouby Y, Delomez G, Tisserant A. First-degree heart block caused by voluntary zopiclone poisoning. Therapie. 1990;45:162.

9 Bramness JG, Arnestad M, Karinen R, et al. Fatal overdose of zopiclone in an elderly woman with bronchogenic coma. Journal of Forensic Sciences. 2001;46:1247-1249.

10 Boniface PF, Russel SG. Two cases of fatal zopiclone overdose. Journal of Analytical Toxicology. 1996;20:131-133.

11 Meram D, Descotes J. Acute poisoning with zolpidem. Revue Medicine Interne. 1989;10:466.

12 Gock SB, Wong SHY, Nuwayhid N, et al. Acute zolpidem overdose – report of two cases. Journal of Analytical Toxicology. 1999;23:559-562.

.

13 Ahmad Z, Herepath M, Ebden P. Diagnostic utility of flumazenil in coma with suspected poisoning. British Medical Journal. 1991;302:292.

14 Lheureux P, Debailleul G, De Witte O, et al. Zolpidem intoxication mimicking narcotic overdoes: response to flumazenil. Human and Experimental Toxicology. 1990;9:105-107.

15 Lheureux P. Continuous flumazenil for zolpidem toxicity – commentary. Clinical Toxicology. 1998;36:745-746.

16 Graham SR, Day RO, Lee R, et al. Overdose with chloral hydrate: a pharmacological and therapeutic review. Medical Journal of Australia. 1988;149:686-688.

17 Zahedi A, Grant MH, Wong DT. Successful treatment of chloral hydrate cardiac toxicity with propranolol. American Journal of Emergency Medicine. 1999;17:490-491.

18 Black JL, Richelson E, Richardson JW. Antipsychotic agents: a clinical update. Mayo Clinic Proceedings. 1985;60:777.

19 Burns MJ. The pharmacology and toxicology of atypical antipsychotic agents. Clinical Toxicology. 2001;39:1-14.

20 Casey DE, Keepers GA. Neuroleptic side effects: acute extrapyramidal syndromes and tardive dyskinesia. Casey DE, Christensen AV, editors. Psychopharmacology: current trends. 1988, 74-83. Springer-Verlag

21 Rupniak NJ, Jenner P, Marsden CD. Acute dystonia induced by neuroleptic drugs. Psychopharmacology. 1986;88:403.

22 Swett C. Drug induced dystonia. American Journal of Psychiatry. 1975;132:532.

23 Marsden CD, Jenner P. The pathophysiology of extrapyramidal side effects of neuroleptic drugs. Psychological Medicine. 1980;10:55.

24 Yassa R, Ananth J, Cordozo S, et al. Tardive dyskinesia in an outpatient population: prevalence and predisposing factors. Canadian Journal of Psychiatry. 1983;28:391.

25 Marks RC, Luchins DJ. Antipsychotic medications and seizures. Psychiatric Medicine. 1991;9:37.

26 Logothetis J. Spontaneous epileptic seizures and electroencephalographic changes in the course of phenothiazine therapy. Neurology. 1967;17:869.

27 Reilly JG, Ayis SA, Ferrier IN, et al. QTc-interval abnormalities and psychotropic drug therapy in psychiatric patients. Lancet. 2000;355:1048-1052.

28 Kariagianis JL, Phillips LC, Hogan KP, et al. Clozapine-associated neuroleptic malignant syndrome: two new cases and a review of the literature. Annals of Pharmacotherapy. 1999;3(5):623-630.

29 Levin GM, Lazowick AL, Powell HS. Neuroleptic malignant syndrome with risperidone. Journal of Clinical Psychopharmacology. 1996;16:192-193.

30 Burkhard PR, Vingerhoets FLG. Olanzapine-induced neuroleptic malignant syndrome (letter). Archives of General Psychiatry. 1999;56:101-102.

31 Nierenberg D, Disch M, Manheimer E, et al. Facilitating prompt diagnosis and treatment of the neuroleptic malignant syndrome. Clinical Pharmacology and Therapy. 1991;50:580.

32 Shalev A, Hermesh H, Munitz H. Mortality from neuroleptic malignant syndrome. Journal of Clinical Psychiatry. 1989;50:18.

33 Blaye IL, Donatini B, Hall M, et al. Acute overdosage with thioridazine: a review of the available clinical exposure. Veterinary and Human Toxicology. 1993;35:147-150.

34 Isbister GK, Murray L, John S, et al. Amisulpride deliberate self-poisoning causing severe cardiac toxicity including QT prolongation and torsades de pointes. Medical Journal of Australia. 2006;184:354-356.

35 Haag S, Spigset O, Edwardsson H, et al. Prolonged sedation and slowly decreasing clozapine serum concentrations after an overdose. Journal of Clinical Psychopharmacology. 1999;19:282-284.

36 Buckley NA, Whyte IM, Dawson AH. Cardiotoxicity is more common in thioridazine overdose than with other neuroleptics. Clinical Toxicology. 1995;33:199-204.

37 Schmidt W, Lang K. Life-threatening dysrhythmias in severe thioridazine poisoning treated with physostigmine and transient atrial pacing. Critical Care Medicine. 1997;25:1925-1930.

38 Sharma ND, Roman HS, Padhi D, et al. Torsades de pointes associated with intravenous haloperidol in critically ill patients. American Journal of Cardiology. 1998;81:238-240.

39 Ward DI. Two cases of amisulpride overdose: A cause for prolonged QT syndrome. Emergency Medical Association. 2005;17:274-276.

40 Balit CR, Isbister GK, Hackett PL, et al. Quetiapine Poisoning: A Case Series. Annals of Emergency Medicine. 2003;42:751-758.

41 Hunfeld NGM, Westerman EM, Boswijk DJ, et al. Quetiapine in overdosage. a clinical and pharmacokinetic analysis of 14 cases. Therapeutic Drug Monitoring. 2006;28:185-189.

42 Bryant SG, Guernsey BG, Ingrim NB. Review of bupropion. Clinical Pharmacology. 1983;2:525-537.

43 Lai AA, Schroeder DH. Clinical pharmacokinetics of bupropion: a review. Journal of Clinical Psychiatry. 1983;44:82-84.

44 Roose SP, Dalack GW, Glassman AH, et al. Cardiovascular effects of bupropion in depressed patients with heart disease. American Journal of Psychiatry. 1991;148:512-516.

45 Szuba MP, Leuchter AF. Falling backward in two elderly patients taking bupropion. Journal of Clinical Psychiatry. 1992;53:157-159.

46 Wenger TL, Stern WC. The cardiovascular profile of bupropion. Journal of Clinical Psychiatry. 1983;44:176-182.

47 Settle EC, Stahl SM, Batey SR, et al. Safety profile of sustained-release bupropion in depression: results of three clinical trials. Clinical Therapeutics. 1999;21:454-463.

48 Davidson J. Seizures and bupropion: a review. Journal of Clinical Psychiatry. 1989;50:256-261.

49 Balit CR, Lynch CN, Isbister GK. Bupropion poisoning: a case series. Medical Journal of Australia. 2003;178:61-63.

50 Druteika D, Zed PJ. Cardiotoxicity following bupropion overdose. Annals of Pharmacotherapy. 2002;36:1791-1795.

51 Biswas AK, Zabrocki LA, Mayes KL, et al. Cardiotoxicity associated with intentional ziprasidone and bupropion overdose. Journal of Toxicology, Clinical Toxicology. 2003;41:101-104.

52 Isbister GK, Balit CR. Bupropion overdose: QTc prolongation and its clinical significance. Annals of Pharmacotherapy. 2003;37:999-1002.

53 Harris CR, Gualtieri J, Stark G. Fatal bupropion overdose. Clinical Toxicology. 1997;35:321-324.

54 Mills KC. Tricyclic antidepressants. In: Tintinalli JE, Ruiz E, Krome RL, editors. Emergency medicine: a comprehensive study guide. 4th edn. Ohio: McGraw Hill; 1996:740.

55 Buckley NA, Dawson AH, Whyte IM, et al. Six years of self-poisoning in Newcastle: 19871992. Medical Journal of Australia. 1995;162:190-193.

56 Buckley NA, Dawson AH, Whyte IM, et al. Greater toxicity of dothiepin in overdose than of other tricyclic antidepressants. Lancet. 1994;343:159-162.

57 Curry SC. Neurotransmitter principles. In: Goldfrank LR, Weissman RS, Flomenbaum NE, Howard MA, Lewin NA, Hoffman RS, editors. Goldfrank’s toxicologic emergencies. 5th edn. Norwalk, Connecticut: Appleton and Lange; 1994:231-257.

58 Chan BC, Graudins A, Whyte IA, et al. Serotonin syndrome resulting from drug interactions. Medical Journal of Australia. 1998;169:523-525.

59 Wedin GP, Odra GM, Klein-Schwartz W, et al. Relative toxicity of cyclic antidepressants. Annals of Emergency Medicine. 1986;15:797.

60 Dzuikas LJ, Vohra J. Tricyclic antidepressant poisoning. Medical Journal of Australia. 1991;154:344-350.

61 Boehnert MT, Lovejoy FH. Value of the QRS duration versus the serum drug level in predicting seizures and ventricular arrhythmias after an acute overdose of tricyclic antidepressants. New England Journal of Medicine. 1985;313:474-479.

62 Niemann JT, Benson HA, Rothstein RJ, et al. Electrocardiographic criteria for tricyclic antidepressant cardiotoxicity. American Journal of Cardiology. 1986;57:1154-1159.

63 Liebelt EL, Francis PD, Woolf AD. ECG lead aVR versus QRS interval in predicting seizures and arrhythmias in acute tricyclic antidepressant toxicity. Annals of Emergency Medicine. 1995;26:195-201.

64 Buchamn AL, Dauer J, Giederan J. The use of vasoactive agents in the treatment of antidepressant overdose. Journal of Clinical Psychopharmacology. 1990;10:409-413.

65 Spivey WH. Flumazenil and seizures: Analysis of 33 cases. Clinical Therapy. 1992;14(2):292-305.

66 Nattel S, Mittleman M. Treatment of ventricular tachyarrhythmias from amitriptyline toxicity in dogs. Journal of Pharmacology and Experimental. Therapy. 1984;1231:430-435.

67 Nattel S, Keable H, Sasyniuk BI. Experimental amytriptyline intoxication: electrophysiologic manifestations and management. Journal of Cardiovascular Pharmacology. 1984;6:83-89.

68 Callaham M, Kassel D. Epidemiology of fatal tricyclic antidepressant ingestion: implications for management. Annals of Emergency Medicine. 1985;14:1-9.

69 Mills K. Serotonin toxicity: a comprehensive review for emergency medicine. Topics in Emergency Medicine. 1993;15:54-73.

70 Woodrum ST, Brown CS. Management of SSRI-induced sexual dysfunction. Annals of Pharmacotherapy. 1998;32:1209-1215.

71 Kirchner V, Silver LE, Kelly CA. Selective serotonin reuptake inhibitors and hyponatremia: review and proposed mechanisms in the elderly. Journal of Psychopharmacology. 1998;12:396-400.

72 Whyte IM, Dawson AH, Buckley NA. Relative toxicity of venlafaxine and selective serotonin reuptake inhibitors in overdose compared to tricyclic antidepressants. Quarterly Journal of Medicine. 2003;96:369-374.

73 Isbister GK, Bowe SJ, Dawson A, et al. Relative toxicity of selective serotonin re-uptake inhibitors (SSRIs) in overdose. Clinical Toxicology. 2004;42:277-285.

74 Isbister GK Fridberg LE, Duffull SB. Application of pharmacokinetic-pharmacodynamic modelling in the management of QT abnormalities after citalopram overdose. Intensive Care Medicine. 2006;32:1060-1065.

75 Insel TR, Roy BF, Cohen RM, et al. Possible development of the serotonin syndrome in man. American Journal of Psychiatry. 1982;139:954-955.

76 Sternbach H. The serotonin syndrome. American Journal of Psychiatry. 1991;148:705-714.

77 DeVane DL. Pharmacokinetics of the selective serotonin reuptake inhibitors. Journal of Clinical Psychiatry. 1992;53:13-19. (Supplement)

78 Kaminski CA, Robbins MS, Weibley RE. Sertraline intoxication in a child. Annals of Emergency Medicine. 1994;23:1371-1374.

79 Kline SS, Mauro LS, Scala-Barnett DM, et al. Serotonin syndrome versus neuroleptic malignant syndrome as a cause of death. Clinical Pharmacology. 1989;8:510-514.

80 Power BM, Pinder M, Hackett LP, et al. Fatal serotonin syndrome following a combined overdose of moclobemide, clomipramine and fluoxetine. Anaesthetics and Intensive Care. 1995;23:499-502.

81 Dunkley EJ, Isister GK, Sibbritt D, et al. The Hunter Serotonin Toxicity Criteria: simple and accurate diagnostic decision rules for serotonin toxicity. Quarterly Journal of Medicine. 2003;96:635-642.

82 Boyer EW, Shannon M. The serotonin syndrome. New England Journal of Medicine. 2005;352:1112-1120.

83 Graudins A, Aaron CK. Delayed peak serum valproic acid in massive divalproex overdose treatment with charcoal hemoperfusion. Journal of Toxicology, Clinical Toxicology. 1996;34:335-341.

84 Guze BH, Baxter LR. The serotonin syndrome: case responsive to propranolol (letter). Journal of Clinical Psychopharmacology. 1986;6:119-120.

85 Geer SC, Baldessarini RJ. Motor effects of serotonin in the central nervous system. Life Sciences. 1980;27:1435-1451.

86 Sandyk R. L dopa induced ‘serotonin syndrome’ in a parkinsonian patient on bromocriptine (letter). Journal of Clinical Psychopharmacology. 1980;6:194-195.

87 Sullivan JB, Rumack BH, Peterson RG. Acute carbamazepine toxicity resulting from overdose. Neurology. 1981;31:621-624.

.

88 Sethna M, Solomon G, Cedarbaum J, et al. Successful treatment of massive carbamazepine overdose. Epilepsia. 1989;30:71-73.

89 Hojer J, Malmlund HO, Berg A. Clinical features in 28 consecutive cases of laboratory confirmed massive poisoning with carbamazepine alone. Clinical Toxicology. 1993;3:449-458.

90 Tibballs J. Acute toxic reaction to carbamazepine: clinical effects and serum concentrations. Journal of Pediatrics. 1992;121:295-299.

91 Apfelbaum JD, Caravati EM, Kerns WP, et al. Cardiovascular effects of carbamazepine toxicity. Annals of Emergency Medicine. 1995;25:631-635.

92 Morgan F. Fortnightly review: drug treatment of epilepsy. British Medical Journal. 1999;318:106-109.

93 Maloteaux EG. Pharmacological management of epilepsy. Mechanism of action, pharmacokinetic drug interactions, and new drug discovery possibilities. International Journal of Clinical Pharmacology and Therapeutics. 1988;36:181-184.

94 Peruca E, Gram L, Avanzi G, Dulac O. Antiepileptic drugs as a cause of worsening seizures. Epilepsia. 1998;39:5-17.

95 Russell MA, Bousvaros G. Fatal results from diphenylhydantoin administered intravenously. Journal of the American Medical Association. 1968;20:2118-2119.

96 Curry SC, Mills KC, Graeme KA. Neurotransmitters. Goldfrank LR, editor. Goldfranks toxicologic emergencies, 7th edn. 2002, 133-165. New York: McGraw-Hill

97 Brubacher JR, Dahghani P, McKnight D. Delayed toxicity following ingestion of enteric-coated divalproex sodium (Epival). Journal of Emergency Medicine. 1999;17:463-467.

98 Garnier R, Boudignat O, Fournier PE. Valproate poisoning. Lancet. 1982;2:97.

99 Anderson GO, Ritland S. Life-threatening intoxication with sodium valproate. Clinical Toxicology. 1995;33:279-284.

100 LoVecchio F, Shriki J, Samaddar R. L-carnitine was safely administered in the setting of valproate toxicity. Am J Emerg Med. 2005.

101 Yehya N, Saldarini CT, Koski ME, et al. Valproate-induced hyperammonemic encephalopathy. [Case Reports. Letter] Journal of the American Academy of Child & Adolescent Psychiatry. 2004;43(8):926-927.

102 Perez A, McKay CA. Role of carnitine in valproic acid toxicity. Journal of Toxicology Clinical Toxicology, 2003;1901-2100.

103 Nagel TR, Schunk JE. Felbamate overdose: a case report and discussion of a new antiepileptic drug. Pediatric Emergency Care. 1995;11:369-371.

104 O’Donnel J, Bateman ND. Lamotrigine overdose in an adult. Clinical Toxicology. 2000;38:659-660.

29.4 Lithium

Clinical features

Chronic lithium toxicity

Chronic lithium toxicity may develop in association with prolonged excessive dosing or, more commonly, as a result of impaired lithium excretion due to intercurrent illness or a drug interaction. Lithium excretion is impaired in renal failure and congestive cardiac failure because of reduced filtration at the glomerulus and also in water or sodium depletion states because of increased reabsorption of sodium (and lithium) in the proximal tubule. A number of drugs including non-steroidal anti-inflammatory drugs (NSAIDs), selective serotonin reuptake inhibitors (SSRIs), angiotesin converting enzyme (ACE) inhibitors, thiazide diuretics and topiramate may also impair lithium excretion.

The clinical features of chronic lithium toxicity are almost exclusively neurological and the following severity grading system is widely used:1

The differential diagnosis for this presentation is broad and includes non-convulsive status epilepticus, serotonin and neuroleptic malignant syndromes, electrolyte abnormalities and CNS pathologies such as sepsis.

Lithium toxicity is generally not associated with significant cardiovascular effects although delayed onset of conduction disturbances is reported.2 Minor benign ECG changes are more commonly observed.3

Chronic lithium therapy is also associated with nephrogenic diabetes insipidus and hypothyroidism, which may complicate the clinical presentation of toxicity.

Acute lithium overdose

Patients who take a significant overdose of lithium carbonate as with any other metal salt, develop rapid onset of gastrointestinal toxicity characterized by nausea, vomiting, abdominal pain and diarrhoea. This gastrointestinal disturbance can be very severe and may result in significant fluid and electrolyte losses. It is usually observed where more than 25 g are ingested but can occur following smaller doses. Gastrointestinal upset is not a prominent feature of chronic lithium toxicity.

Acute lithium overdose is much less likely to result in significant neurotoxicity than is chronic lithium toxicity.4 Neurotoxicity could theoretically slowly develop following acute overdose if renal clearance were sufficiently impaired so as to allow redistribution of sufficient lithium from the intravascular compartment to tissue compartments before it could be excreted. This situation may develop if there is pre-existing renal failure or if inadequate fluid resuscitation leads to dehydration, sodium depletion or renal impairment as a consequence of the fluid losses from gastrointestinal toxicity.

Clinical investigation

Essential laboratory investigations in the assessment of lithium toxicity are serum electrolytes, renal function and serum lithium concentration. Serial serum lithium concentrations are often required. Other investigations are performed as indicated to evaluate and manage intercurrent disease processes and to exclude important differential diagnoses.

Therapeutic serum lithium concentrations are generally quoted as 0.6–1.2 mEq/L, although clinical evidence of lithium toxicity can be observed at concentrations within this range, particularly in the elderly.5 More commonly in cases of chronic intoxication, mild toxicity is observed at lithium concentrations of 1.5–2.5 mEq/L, severe toxicity at concentrations of 2.5 to 3.5 mEq/L, and life-threatening toxicity at concentrations >3.5 mEq/L. Following acute overdose, serum lithium concentrations do not correlate with clinical severity as they do not reflect CNS concentrations; however, when performed serially, they are useful in guiding management. Peak serum lithium concentrations >4.0 mEq/L are frequently observed following acute overdose in patients who do not go on to develop neurotoxicity.

Treatment

Chronic lithium toxicity

The diagnosis of lithium toxicity should be considered in any individual on lithium therapy who presents to the emergency department unwell, in particular with evidence of neurological dysfunction. The diagnosis should be confirmed or excluded by ordering a serum lithium concentration as part of the initial work-up. A precipitating illness that has resulted in impaired lithium excretion will usually be present and require assessment and treatment on its own merits.

Appropriate supportive care measures should be instituted on arrival. Once the diagnosis of chronic lithium toxicity is confirmed, further care is oriented towards management of the precipitating medical condition and enhancing lithium excretion by optimizing renal function and correcting any water or sodium deficits with intravenous normal saline. Therapy with lithium carbonate and any drugs contributing to lithium toxicity should be immediately discontinued.6

Enhanced elimination of lithium by haemodialysis may be attempted in severe or worsening chronic lithium neurotoxicity. The aim of this intervention is to minimize the duration of neurological dysfunction and avoid permanent neurological sequelae. Lithium has physicochemical and pharmacokinetic properties that render it very suitable for enhancing elimination by haemodialysis: low molecular weight, high water solubility, small volume of distribution, no plasma protein binding and an endogenous renal clearance rate much lower than that achieved by haemodialysis.7 There is, however, no evidence that haemodialysis improves clinical outcome or survival rates.

The indications for haemodialysis are difficult to define. It should be considered in any patient with an elevated serum lithium concentration and severe or life-threatening neurotoxicity. It may be considered in the patient with less severe toxicity in whom adequate renal function and a falling lithium concentration are unable to be established with initial fluid resuscitation. Once instituted haemodialysis should be continued until the serum lithium is <1 mEq/L. Some rebound in serum lithium may be noted after intermittent haemodialysis is discontinued, which may be avoided if continuous arterio-venous (AV) or veno-venous (VV) haemodiafiltration is sustained for >16 h.8,9 The decision to dialyse can usually be made some 8–12 h after admission.7

Acute lithium overdose

In contrast to chronic toxicity, the vast majority of acute poisonings can be managed solely with good supportive care. Intravenous access should be established and infusion of normal saline commenced during the initial assessment. Administration should be sufficient to correct any sodium or water deficits arising as a result of the toxic gastroenteritis and to ensure a good urine output. Excessive administration of normal saline or attempts at forced diuresis do not further enhance lithium excretion.10 A serum lithium concentration, renal function and electrolytes should be performed as part of the initial assessment and repeated as necessary to guide further management. In particular, the serum lithium should be followed until falling and <2 mEq/L.

Activated charcoal does not bind lithium well and need not be administered unless there has been a significant co-ingestion. Sodium polystyrene sulfonate has been proposed as an effective alternative absorbent but is not widely used and repeated administration can cause hypokalaemia.11 On the basis of a single volunteer study, whole-bowel irrigation has been recommended for overdose of extended-release preparations12 but the gastrointestinal upset renders this intervention technically difficult in patients with large ingestions.

Haemodialysis is rarely indicated following acute overdose in the patient with normal renal function who receives good supportive care. It may be necessary in the presence of renal failure or in the patient who goes on to develop neurotoxicity in the presence of a slowly falling serum lithium concentration.

Disposition and prognosis

Patients with chronic lithium intoxication require admission for management of their fluid and electrolyte status, monitoring of renal function and serum lithium concentration and management of intercurrent illnesses. Ideally, admission should be to an institution with a capacity to perform haemodialysis where toxicity is moderate or severe. Following haemodialysis, neurological recovery may be delayed well beyond the removal of lithium and permanent neurological deficits are reported.13,14

Acute lithium overdose usually has an excellent outcome with good supportive care and may be admitted to a non-monitored setting for intravenous fluids and monitoring of fluid and electrolytes and lithium concentrations. The asymptomatic patient with normal renal function and lithium level falling to below 2 mEq/L is fit for medical discharge. This usually occurs within 24 h. Psychiatric evaluation is mandatory and may take place whilst waiting for lithium levels to fall.

References

1 Hansen HE, Amdisen A. Lithium intoxication. Quarterly Journal of Medicine. 1978;47:123-144.

2 Waring WS. Delayed cardiotoxicity in chronic lithium poisoning: discrepancy between serum lithium concentrations and clinical status. Basic and Clinical Pharmacology and Toxicology. 2007;100(5):353-355.

3 Tilkian AG, Schroeder JS, Kao JJ. Cardiovascular effects of lithium in man: a review of the literature. American Journal of Medicine. 1976;61:665-667.

4 Oakley PW, Whyte IM, Carter GL. Lithium toxicity: an iatrogenic problem in susceptible individuals. Australian & New Zealand Journal of Psychiatry. 2001;35:833-840.

5 Strayhorn JM, Nash JL. Severe neurotoxicity despite ‘therapeutic’ serum lithium levels. Diseases of the Nervous System. 1977;38:107-111.

6 Eyer F, Pfab R, Felgenhauer N, et al. Lithium poisoning: pharmacokinetics and clearance during different therapeutic measures. Journal of Clinical Psychopharmacology. 2006;26(3):325-330.

7 Jaeger A, Saunder P, Kopferschmidt J, et al. When should dialysis be performed in lithium poisoning? A kinetic study in 14 cases of lithium poisoning. Clinical Toxicology. 1993;31(3):429-447.

8 LeBlanc M, Raymond M, Bonnardeau A, et al. Lithium poisoning treated by high-performance continuous arteriovenous and venovenous hemodiafiltration. American Journal of Kidney Disease. 1996;27:365-372.

9 Waring WS. Management of lithium toxicity. Toxicology Reviews. 2006;25(4):221-230.

10 Amidsen A. Clinical features and management of lithium poisoning. Medical and Toxicological Adverse Drug Experiences. 1988;3:18-32.

11 Roberge RJ, Martin TG, Schneider S. Use of sodium polystyrene sulfonate in a lithium overdose. Annals of Emergency Medicine. 1993;22:1911-1915.

12 Smith S, Ling L, Halstenson C. Whole-bowel irrigation as a treatment for acute lithium overdose. Annals of Emergency Medicine. 1991;20:536-539.

13 Verdoux H, Bougeois M. A case of lithium neurotoxicity with irreversible cerebellar syndrome. Journal of Nervous and Mental Disorders. 1990;178:761.

14 Shou M. Long lasting neurological sequelae after lithium intoxication. Acta Psychiatrica Scandinavica. 1984;70:594.

29.5 Antihistamine and anticholinergic poisoning

Introduction

Anticholinergic toxicity is a common side effect of many pharmaceutical agents, natural remedies and plants, both in therapeutic dosing and in overdose (see Table 29.5.1). Symptoms and signs may range from mild manifestations of the syndrome (e.g. dry mouth and blurred vision) to severe anticholinergic delirium with agitation, hallucinations and aggressive behaviour.

Table 29.5.1 Anticholinergic agents

Pharmaceuticals

Antipsychotic agents

Cyclic antidepressants

First-generation H1-receptor blockers

Others

Botanicals Datura spp.

The antihistamine agents are a diverse group of drugs that can be broadly classified, based upon receptor specificity, into H1– and H2-receptor antagonists. The H1-receptor antagonists are widely used in the treatment of allergic conditions and nasal congestion, as over-the-counter sleep aids and as antiemetics. This group can be further divided into the ‘first-generation’ agents, which tend to be more lipophilic and are more sedating, and the ‘second-generation’ or non-sedating agents. The H2-receptor antagonists are primarily used in the treatment of peptic ulcer disease and gastro-oesophageal reflux, but are also used in conjunction with H1 antagonists in the treatment of severe allergic reactions.

Antihistamine agents are relatively easy to obtain and frequently used in overdose for attempted suicide and abused recreationally for their sedating and anticholinergic effects. The incidence of antihistamine poisoning and abuse in Australia is not well characterized. Other prescription drugs may also result in anticholinergic toxicity both in therapeutic dosing and in overdose. These may also be intentionally abused for their anticholinergic effects.14 Chinese and traditional herbal medicines may result in anticholinergic toxicity either directly from the herbal agent ingested or as a result of contamination with anticholinergic agents such as atropine or scopolamine.57 The intentional abuse of botanicals (e.g. Datura spp.) may also present with anticholinergic toxicity.810 In view of the easy availability of many of these pharmaceutical and herbal agents, the emergency physician should include a detailed drug history in the evaluation of any patient presenting with evidence of mental status change and anticholinergic symptoms and signs. In particular, polypharmacy and drug interactions between multiple agents with the potential for anticholinergic effects should be included in the differential diagnosis of elderly patients presenting with mental status changes.

Pharmacodynamics and pharmacokinetics

The H1-antagonists are a diverse group of agents that reversibly block the action of histamine at H1 receptors. High lipid solubility results in central nervous system (CNS) penetration and sedation. The first generation agents also block muscarinic, α-adrenergic and serotonergic receptors. Local anaesthetic effects due to sodium channel blockade may mimic the antiarrhythmic properties of class 1A antiarrhythmic agents.11 Diphenhydramine, dimenhydrinate and cyproheptadine, in particular, may prolong the cardiac muscle cell action potential duration by this mechanism.12,13

The second generation H1-antagonists (fexofenadine, loratadine) have much less CNS penetration and are more histamine receptor specific with little or no effect at other receptor subtypes.11

All the H1-antagonists are well absorbed orally with peak serum concentrations occurring within 2 to 4 h. Absorption may be delayed in overdose due to anticholinergic effects seen with the first-generation agents. Bioavailability is limited by significant first-pass metabolism. Some agents may be converted to active metabolites (e.g. hydroxyzine). Volume of distribution and protein binding are generally high. Elimination half-lives for the first generation agents are between 2 and 6 h.11 The second generation agents generally have longer half-lives (e.g. loratadine 8.3 h).11

The H2-antagonist agents are generally well tolerated with few side effects with therapeutic dosing. Cimetidine inhibits hepatic microsomal enzyme metabolism and reduces the metabolism of drugs eliminated by this pathway. This may result in increased serum concentrations and clinical effects of co-ingested medications.

All drugs with anticholinergic side effects have the potential to slow gastric emptying and produce gastrointestinal ileus when taken in overdose. As a result, absorption of these agents may be slowed and result in the potential for prolonged toxicity.

Clinical features

The anticholinergic toxidrome is usually manifest by a combination of peripheral and central muscarinic, cholinergic receptor blockade. Peripheral effects may include sinus tachycardia, cutaneous vasodilatation and flushing, low-grade temperature, warm dry skin with an absence of axillary sweat, dry mucous membranes, gastrointestinal ileus and urinary retention. CNS effects include mydriasis with blurred vision due to the inhibition of visual accommodation, delirium, confusion, visual hallucinations, incoherent speech, agitation, combativeness, aggression and coma.14 Patients presenting with anticholinergic syndrome will often have an impaired perception of their environment. This may result in behaviour that could injure the patient. Anticholinergic symptoms and signs may be prominent with ingestion of first-generation antihistamines.1517 Even therapeutic doses of some of the H1-antagonist agents may be sufficient to produce an anticholinergic delirium in susceptible individuals (especially the elderly and children). Topical use of these agents, particularly on broken skin surfaces, may also result in anticholinergic delirium.18,19

In patients who present to hospital several hours following poisoning with an anticholinergic agent, the peripheral features of the toxidrome may be absent.2022 This may also occur in elderly people with mild-to-moderate anticholinergic delirium resulting from the side effects of therapeutic drug administration.22

Other manifestations of H1-antagonist toxicity may include CNS and cardiovascular effects, and rhabdomyolysis. Overdose of first-generation agents commonly produces drowsiness, sedation, confusion, agitation and ataxia. Large ingestions may result in coma.23,24 Seizures may also occur. Pheniramine, a commonly abused antihistamine in Australia, appears to be more proconvulsant than other agents following overdose, with a reported incidence of seizures of 30%.17 Seizures have been reported with other first-generation H1-antagonists, such as diphenhydramine, in doses as small as 150 mg in children.23,25 Fatal doses of diphenhydramine in adults range from 20 to 40 mg/kg.26 Doxylamine poisoning may result in non-traumatic rhabdomyolysis.27,28 Hypotension, due to α-receptor blockade, can occur following large ingestions of first-generation agents. Conduction defects are infrequent following poisoning with first-generation H1-antagonists. Diphenhydramine and dimenhydrinate poisoning can result in QRS-interval prolongation, broad-complex tachycardia and ventricular arrhythmias similar to that seen in cyclic antidepressant poisoning.12,13 This effect has not been reported with other first-generation H1-antagonists.

Overdose with H2-antagonists, such as cimetidine, usually results in little or no evidence of toxicity. Doses of up to 15 g have failed to produce clinical toxicity.29

Treatment

The mainstay of therapy for poisoning with anticholinergic agents is supportive care in a safe environment. Comatose or hypoventilating patients should have appropriate airway intervention and ventilatory support. Hypotension should be treated initially with intravenous crystalloid boluses. Hypotension refractory to fluids may necessitate the use of pressor agents such as noradrenaline. Agitation and seizures can be controlled using parenteral benzodiazepines in the first instance. Barbiturates (thiopentone, phenobarbitone) may be considered in refractory cases.

Gastrointestinal decontamination, if indicated, should be performed with a single-dose of oral activated charcoal. The benefit of activated charcoal in patients presenting with minimal or no signs of toxicity more than 2 h following ingestion is doubtful. Methods of enhancing elimination of antihistamines are ineffective because of their large volumes of distribution and high protein binding.

The reversible acetylcholinesterase inhibitor physostigmine has been used in the management and diagnosis of anticholinergic agitation and delirium.3032 Physostigmine rapidly reverses the effects of anticholinergic delirium and may prevent the need for escalating doses of benzodiazepines to control agitation. Physostigmine may decrease the need for other interventions such as cerebral computerized tomography scanning and lumbar puncture in patients with suspected anticholinergic delirium.31

When using physostigmine, an initial test dose 0.5 mg i.v. is followed by 1.0–2.0 mg over the following 3–5 min in an adult. A partial response may necessitate further 0.5–1.0 mg boluses. Clinical effects may last from 30 to 120 min. Caution should be exercised in using physostigmine in patients with suspected acute cyclic antidepressant poisoning or ECG evidence of cardiac conduction delay because of the risk of precipitating cardiac asystole.33,34

Broad-complex tachycardia resulting from severe poisoning with diphenhydramine or dimenhydrinate should be treated with serum alkalinization with intravenous sodium bicarbonate boluses (0.5–1.0 mmol/kg) as for severe cyclic antidepressant poisoning.12,13 Symptomatic bradycardia and high degree atrioventricular block should be initially treated with atropine. Unresponsive cases may need cardiac pacing or inotropic support. Class-1a, -1c or -3 anti-arrhythmic agents should be avoided in cases of antihistamine-induced arrhythmias.

References

1 Acri AA, Henretig FM. Effects of risperidone in overdose. American Journal of Emergency Medicine. 1998;16:498-501.

.

2 Fisher RS, Cysyk B. A fatal overdose of carbamazepine: case report and review of literature. Journal of Toxicology Clinical Toxicology. 1988;26:477-486.

3 Graudins A, Peden G, Dowsett RP. Massive overdose with controlled-release carbamazepine resulting in delayed peak serum concentrations and life-threatening toxicity. Emergency Medicine (Fremantle). 2002;14:89-94.

4 Yang CC, Deng JF. Anticholinergic syndrome with severe rhabdomyolysis – an unusual feature of amantadine toxicity. Intensive Care Medicine. 1997;23:355-356.

5 Chan TY. Anticholinergic poisoning due to Chinese herbal medicines. Veterinary & Human Toxicology. 1995;37:156-157.

6 Chan JC, Chan TY, Chan KL, et al. Anticholinergic poisoning from Chinese herbal medicines. Australian & New Zealand Journal of Medicine. 1994;24:317-318.

7 Chan TY, Tang CH, Critchley JA. Poisoning due to an over-the-counter hypnotic, Sleep-Qik (hyoscine, cyproheptadine, valerian). Postgraduate Medical Journal. 1995;71:227-228.

8 Finlay P. Anticholinergic poisoning due to Datura candida. Tropical Doctor. 1998;28:183-184.

9 Hanna JP, Schmidley JW, Braselton WE. Datura delirium. Clinical Neuropharmacology. 1992;15:109-113.

10 Mahler DA. Anticholinergic poisoning from Jimson weed. Journal of the American College of Emergency Physicians. 1976;5:440-442.

11 Rimmer SJ, Church MK. The pharmacology and mechanism of action of histamine H1 antagonists. Clinical and Experimental Allergy. 1990;20:3.

12 Clark RF, Vance MV. Massive diphenhydramine poisoning resulting in a wide-complex tachycardia: successful treatment with sodium bicarbonate. Annals of Emergency Medicine. 1992;21:318-321.

13 Farrell M, Heinrichs M, Tilelli JA. Response of life threatening dimenhydrinate intoxication to sodium bicarbonate administration. Journal of Toxicology – Clinical Toxicology. 1991;29:527-535.

14 Feldman MD. The syndrome of anticholinergic intoxication. American Family Physician. 1986;34:113-116.

15 Jones IH, Stevenson J, Jordan A, et al. Pheniramine as an hallucinogen. Medical Journal of Australia. 1973;1:382-386.

16 Jones J, Dougherty J, Cannon L. Diphenhydramine-induced toxic psychosis. American Journal of Emergency Medicine. 1986;4:369-371.

17 Buckley NA, Whyte IM, Dawson AH, et al. Pheniramine-a much abused drug. Medical Journal of Australia. 1994;160:188-192.

18 Schipior PG. An unusual case of antihistamine intoxication. Journal of Pediatrics. 1967;71:589-591.

19 Reilly JFJr., Weisse ME. Topically induced diphenhydramine toxicity. Journal of Emergency Medicine. 1990;8:59-61.

20 Rupreht J, Dworacek B. Central anticholinergic syndrome in anesthetic practice. Acta Anaesthesiologica Belgica. 1976;27:45-60.

21 Koppel C, Hopfe T, Menzel J. Central anticholinergic syndrome after ofloxacin overdose and therapeutic doses of diphenhydramine and chlormezanone. Journal of Toxicology – Clinical Toxicology. 1990;28:249-253.

22 Feinberg M. The problems of anticholinergic adverse effects in older patients. Drugs Aging. 1993;3:335-348.

23 Koppel C, Ibe K, Tenczer J. Clinical symptomatology of diphenhydramine overdose: an evaluation of 136 cases in 1982 to 1985. Journal of Toxicology – Clinical Toxicology. 1987;25:53-70.

24 Koppel C, Tenczer J, Ibe K. Poisoning with over-the-counter doxylamine preparations: an evaluation of 109 cases. Human Toxicology. 1987;6:355-359.

26 Krenzelok EP, Anderson GM, Mirick M. Massive diphenhydramine overdose resulting in death. Annals of Emergency Medicine. 1982;11:212-213.

27 Mendoza FS, Atiba JO, Krensky AM, et al. Rhabdomyolysis complicating doxylamine overdose. Clinical Paediatrics. 1987;26:595-597.

28 Frankel D, Dolgin J, Murray BM. Non-traumatic rhabdomyolysis complicating antihistamine overdosage. Clinical Toxicology. 1990;31:493.

29 Krenzelok EP, Litovitz T, Lippold KP, et al. Cimetidine toxicity: an assessment of 881 cases. Annals of Emergency Medicine. 1987;16:1217-1221.

30 Beaver KM, Gavin TJ. Treatment of acute anticholinergic poisoning with physostigmine. American Journal of Emergency Medicine. 1998;16:505-507.

31 Burns MJ, Linden CH, Graudins A, et al. A comparison of physostigmine and benzodiazepines for the treatment of anticholinergic poisoning. Annals of Emergency Medicine. 2000;35:374-381.

32 Mendelson G. Pheniramine aminosalicylate overdosage. Reversal of delirium and choreiform movements with tacrine treatment. Archives of Neurology. 1977;34:313.

33 Pentel P, Peterson CD. Asystole complicating physostigmine treatment of tricyclic antidepressant overdose. Annals of Emergency Medicine. 1980;9:588-590.

34 Suchard JR. Assessing physostigmine’s contraindication in cyclic antidepressant ingestions. Journal of Emergency Medicine. 2003;25:185-191.