Physical Address
304 North Cardinal St.
Dorchester Center, MA 02124
Toxicology is a broad, multidisciplinary science where the goal is to determine the effects of chemical agents on living systems. Innumerable potential toxins can inflict harm, including pharmaceuticals, herbals, household products, environmental agents, occupational chemicals, drugs of abuse, and chemical terrorism threats. Each year millions of human exposure cases are reported worldwide. The Centers for Disease Control and Prevention (CDC) has reported that poisoning (both intentional and unintentional) is one of the leading causes of injury-related death in the United States in all adult age groups. From the beginnings of written history, poisons and their effects have been well described. Paracelsus (1493–1541) correctly noted that “Alle Dinge sind Gift, und nichts ist ohne Gift; allein die Dosis macht, daß ein Ding kein Gift sei,” which means, “Everything is a poison; there is nothing which is not. Only the dose differentiates a poison.” As life in the modern era has become more complex, so has the study of poisons, their identification, and their treatments.
This chapter provides a general overview of clinical toxicology and the laboratory services necessary to support the care of poisoned patients. Because a comprehensive discussion of all aspects of toxicology is beyond the scope of this chapter, the clinical significance and toxicity of only a select number of common drugs, drugs of abuse, and other chemicals are discussed.
In practice, it is neither possible nor necessary to test for all of the hundreds or thousands of clinical toxins that may be encountered. In reality less than 25 substances account for 80% or more of cases of intoxication treated in most emergency departments. Moreover, some drugs are encountered rarely in some locations but with relatively high frequency in others. For example, phencyclidine (PCP) use is almost non-existent in some areas but is responsible for a relatively high number of intoxications in a few large metropolitan cities. Thus the scope of clinical toxicology testing provided by the laboratory will depend on the pattern of local drug use and on the available resources of the institution and should be developed in consultation with the appropriate clinical staff.
The value of drug and/or substance testing (screening) is well established (1) in the workplace, (2) for some athletic competitions, (3) to monitor drug use during pregnancy, (4) to evaluate drug exposure and/or withdrawal in newborns, (5) to monitor patients in pain management and drug abuse treatment programs, and (6) to aid in the prompt diagnosis of toxicity for a select number of drugs or agents for which a specific antidote or treatment modality is required ( Table 43.1 ). In many other instances of drug toxicity, the value of drug screening, especially on an emergency basis, is more controversial.
Toxin | Potential Antidote/Treatment |
---|---|
Acetaminophen | N -acetylcysteine |
Aluminum | Deferoxamine |
Anticholinergics | Physostigmine |
Arsenic | 2,3-Dimercaptosuccinic acid (DMSA), dimercaprol (BAL) |
Barbiturates | Multiple-dose oral activated charcoal, sodium bicarbonate |
Benzodiazepines | Flumazenil |
β-Adrenergic blockers | Calcium, glucagon, high-dose insulin |
Calcium channel blockers | Calcium, glucagon, high-dose insulin |
Carbamates | Atropine |
Carbamazepine | Multiple-dose oral activated charcoal, extracorporeal techniques |
Cardiac glycosides | Anti-digoxin Fab fragments |
Carbon monoxide | Oxygen (normobaric or hyperbaric) |
Copper | d-Penicillamine |
Cyanide | Hydroxocobalamin, nitrites, thiosulfate |
Ethylene glycol | Fomepizole (4-methylpyrazole), ethanol, hemodialysis |
Heparin | Protamine sulfate |
Iron | Deferoxamine |
Isoniazid | Pyridoxine |
Lead | EDTA, dimercaprol (BAL), 2,3-dimercaptosuccinic acid (DMSA), d-penicillamine |
Mercury | dimercaprol (BAL), 2,3-dimercaptosuccinic acid (DMSA), d-penicillamine |
Methanol | Fomepizole (4-methylpyrazole) or ethanol, hemodialysis |
Methemoglobin | Methylene blue, vitamin C |
Methotrexate | Leucovorin |
Opioids | Naloxone |
Organophosphates | Atropine, pralidoxime |
Sulfonylureas | Glucose, octreotide |
Salicylates | Sodium bicarbonate, hemodialysis |
Tricyclic antidepressants | Sodium bicarbonate |
Theophylline | Multiple-dose oral activated charcoal, extracorporeal techniques |
Tissue plasminogen activator (tPA), streptokinase, urokinase | Aminocaproic acid |
Warfarin | Phylloquinone (vitamin K1), plasma |
Approaches to drug testing vary from the provision of just a few specific tests (e.g., acetaminophen, salicylate, ethanol, digoxin, iron) to testing for additional targeted groups of drugs (e.g., stimulant panel and coma panel) or to a more comprehensive general drug screen that might include 100 or so drugs and/or substances. For all of these situations, it is imperative that the laboratory communicate with the physician concerning the scope (and limitation) of the service and the proper timing and selection of specimens; when possible, the laboratory should assist with interpretation of results. At a minimum, the laboratory should clearly identify and indicate limit of detection for the drugs that it has the capability of detecting. Otherwise, the report of a “negative” result for a drug screen could be misleading.
To operate effectively, the laboratory should be closely associated with the health care team directly managing the patient. Through close and collaborative work, clinical information provided will help to guide appropriate ordering of tests and to ensure that interpretation of results is complete and accurate. For example, the team caring for the patient should provide the following information with the laboratory request:
The time and date of the suspected exposure along with the time and date of sample collection.
History from the patient or witnesses that might aid in identification of the toxin.
Assessment of the physical state of the patient at the time of presentation.
Such information is useful to guide test selection and interpretation of results.
Due to the wide range of chemical diversity among toxicologic compounds, no single analytical technique is adequate for broad-spectrum drug detection. Combinations of several analytical approaches are generally required. These may include simple, inexpensive, and rapid spot tests; immunoassays (see Chapter 26 ); and chromatographic and/or mass spectrometric techniques (see Chapters 19 , 20 , and 21 )—high-performance liquid chromatography (HPLC), gas chromatography (GC), gas chromatography-mass spectrometry (GC-MS or GC-MS/MS), liquid chromatography-mass spectrometry (LC-MS, LC-MS/MS), and high-resolution mass spectrometry (HRMS, time of flight [TOF], Orbitrap). LC-MS/MS and TOF are increasingly utilized in clinical and forensic settings. GC-MS and headspace GC remain widely used definitive confirmatory procedures. Confirmatory testing is mandatory for forensic drug testing (e.g., workplace drug testing).
Speed of analysis, or turnaround time (TAT), and availability of tests are critical issues in clinical toxicology. A drug analysis that requires several hours to complete or that is not available at all hours of the day is of little value in a clinical emergency. Alternatively, a rapid test that provides false information could result in erroneous diagnostic and therapeutic decisions. For numerous agents, quantitative determinations guide management during a clinical emergency. These agents include acetaminophen (paracetamol), salicylate, ethanol, methanol, isopropanol, ethylene glycol, carbamazepine, phenytoin, valproic acid, phenobarbital, iron, lithium, theophylline, and digoxin, and in whole blood, carboxyhemoglobin and methemoglobin. Results for these determinations should be available as close as possible to 1 hour of receiving the specimen.
Proper selection of analytical methods and interpretation of results require knowledge of the pharmacology and pharmacokinetics of the toxins of interest. For example, the potential hepatotoxicity of acetaminophen is related to the concentration of unmetabolized drug. Conversely, delta-9-tetrahydrocannabinol (THC)—a metabolite of marijuana—is measured in urine as an indication of marijuana use. Knowledge of immunoassay screen cross reactivity is essential for laboratorians to guide providers with ordering drug panels using alternative methodologies incorporating detection of compounds not typically detected by immunoassay screens.
When a health care team initially evaluates a patient who presents with a potential toxicologically induced health problem, the final diagnosis is often determined by (1) reviewing the history, (2) performing a directed physical examination, (3) using ancillary tests (e.g., electrocardiogram [ECG], radiology), and (4) applying a rational and evidence-based approach to laboratory testing. Often no specific antidote or treatment is available for a poisoned patient and careful supportive care is the most appropriate intervention.
All patients who present with potential toxicity should be thoroughly assessed, and it is imperative that clinicians follow a standard “ABC” approach with attention to “airway, breathing, and circulation,” respectively. The patient’s airway should be open and adequate ventilation ensured. If the patient’s airway is not secure and endotracheal tube intubation is considered, the first diagnostic test that should be performed is a rapid bedside glucose concentration. Hypoglycemia can result in coma or new-onset seizures, thereby mimicking a toxic etiology. In addition, numerous toxins are clinically associated with hypoglycemia (e.g., sulfonylureas, ethanol, Mentha pulegium ). Clinical effects induced by hypoglycemia can be rapidly reversed with intravenous glucose, thus preventing unnecessary and costly procedures and testing.
Too often, health care providers are lulled into a false sense of security when a patient presents with altered mental status and oxygen saturations on pulse oximetry that are adequate on high-flow oxygen. If the patient has inadequate ventilation or a poor gag reflex, then the patient may be at risk for progressive CO 2 narcosis or aspiration, respectively, and yet may be maintaining adequate oxygen saturation on supplemental oxygen. Capnography (monitoring of the concentration or partial pressure of carbon dioxide in the respiratory gases) should be considered for use in appropriate cases. It is imperative that the health care team avoid the common complication of aspiration pneumonitis by assuring that the airway is secured if necessary. , An arterial blood gas (ABG) can rapidly aid the health care team in determining the need for intubation and mechanical ventilation. The ABG can also provide valuable information regarding the patient’s acid-base status and can help the clinician begin to generate a differential diagnosis. For example, in the scenario of a febrile toxic patient who presents with an altered mental status, a normal ABG (lack of acidosis) eliminates the possibility of uncoupling of oxidative phosphorylation as a cause of that patient’s fever. Finally, an ABG with co-oximetry can rapidly assist in determining other toxic etiologies, such as carbon monoxide poisoning (depending on the timing of the blood draw in relation to the exposure) and methemoglobinemia.
The initial treatment of hypotension in all toxic patients consists of the administration of intravenous fluids. The patient’s pulmonary status should be closely monitored to ensure that pulmonary edema (a rare complication in poisoned patients) does not develop as fluids are infused. Symptomatic toxic patients should be placed on continuous cardiac monitoring with pulse oximetry, and the health care team must perform frequent neurologic checks to ensure continued protection of the airway. Acutely poisoned patients should receive a large-bore peripheral intravenous line, and all symptomatic patients should have a second line placed in the peripheral or central venous system, depending on the severity of their clinical status. At this time in patient care, blood can be drawn and sent for appropriate laboratory diagnostic testing. Placement of a urinary catheter should be considered early in the care of hemodynamically unstable poisoned patients to monitor urinary output as an indicator of adequate perfusion. A rapid bedside urine dipstick can provide helpful information quickly as health care team members await further laboratory testing. For example, a urine specific gravity will give insight into the patient’s initial hydration status, and the appearance of tea-colored urine positive for blood may indicate the presence of myoglobinuria in a comatose patient with rhabdomyolysis.
The secondary survey involves a thorough examination of the entire patient. For adequate access to a toxic patient, the patient must be completely undressed. Exposure of the patient ensures that a complete physical examination is performed. If the patient is not completely undressed, an important diagnostic clue may be missed. For example, skin lesions consistent with pressure necrosis on the back of a comatose patient may indicate the need to obtain other testing including a urine myoglobin concentration. A comatose drug abuser may have attached transdermal drug patches (e.g., fentanyl, clonidine) in atypical locations (e.g., gluteal sulcus) that when found can rapidly lead to a diagnosis, avoiding the need for further laboratory testing. Besides completing a thorough physical review of all organ systems, the secondary survey involves reviewing items brought with the patient (e.g., medication bottles, drug paraphernalia). Searching carefully through the patient’s clothing may assist in providing clues that change the plan for specific laboratory tests or explain specific laboratory findings. For example, the discovery of a cough and cold product in the patient’s pocket that contains dextromethorphan could explain the clinical presentation of an agitated patient with hyperreflexia whose initial urine toxicology screen was positive for phencyclidine but later was found negative on confirmation.
Toxic syndromes (“toxidromes”) are clinical syndromes that are essential for the successful recognition of poisoning patterns. A toxidrome is the constellation of clinical signs and symptoms that suggests a specific class of poisoning. An important component of the secondary survey is to determine whether a specific toxic syndrome is present. The most commonly encountered toxidromes include (1) anticholinergic, (2) cholinergic, (3) opioid, (4) sedative-hypnotic, and (5) sympathomimetic ( Table 43.2 ). Many toxidromes have several overlapping features. For example, anticholinergic findings are highly similar to sympathomimetic findings, with an exception being the effects on sweat glands: anticholinergic agents produce warm, flushed dry skin, but sympathomimetic agents produce diaphoresis. Toxidrome findings may also be affected by individual variability, comorbid conditions, and coingestants. For example, tachycardia associated with sympathomimetic or anticholinergic toxidromes may be absent in a patient who is concurrently taking beta-antagonist medications. Additionally, although toxidromes may be applied to classes of drugs, one or more toxidrome findings may be absent for some individual agents within these classes. For instance, meperidine is an opioid analgesic, but it does not induce miosis, which helps to define the “classic” opioid toxidrome. When accurately identified, the toxidrome may provide invaluable information for diagnosis and subsequent treatment, although the many limitations impeding acute toxidrome diagnosis must be carefully considered.
Toxidrome | Symptom |
---|---|
Anticholinergic |
|
Cholinergic |
|
Opioid |
|
Sedative-hypnotic |
|
Sympathomimetic |
|
Anticholinergic agents block the neurotransmitter, acetylcholine, inhibiting parasympathetic central and peripheral nerve impulses. Characteristics of the anticholinergic syndrome have long been taught using the old medical adage, “dry as a bone, blind as a bat, red as a beet, hot as a hare, and mad as a hatter” (some add Bladder loses its tone, and Heart runs alone) which correspond with a symptomatic person’s anhidrosis, mydriasis, flushing, fever, and delirium, respectively.
Depending on the dose and time post exposure, various central nervous system (CNS) effects may manifest from an anticholinergic agent. Restlessness, apprehension, abnormal speech, confusion, agitation, tremor, picking movements, ataxia, stupor, and coma all have been described following exposure to various anticholinergics. When manifesting delirium, the individual will often stare into space and mutter, fluctuating between occasional lucid intervals with appropriate responses and then descriptions of vivid hallucinations. Phantom behaviors, such as plucking or picking in the air or at garments, are characteristic. Hallucinations are prominent, and they may be benign, entertaining, or terrifying to the patient experiencing them. Exposed patients may have conversations with hallucinated figures and/or they may misidentify persons they typically know well. Simple tasks typically performed well by the exposed person may become difficult. Motor coordination, perception, cognition, and new memory formation are altered.
Mydriasis causes photophobia; impairment of near vision occurs because of loss of accommodation and reduced depth of field secondary to ciliary muscle paralysis and pupillary enlargement. Tachycardia and exacerbated heart rate responses to exertion are expected. Systolic and diastolic blood pressure may show moderate elevation. A decrease in capillary tone may cause skin flushing. Intestinal motility slows, resulting in nausea, vomiting, and decreased bowel sounds. All glandular cells become inhibited, resulting in dry mucous membranes of the mouth and inhibition of sweating with resultant dry skin. Urination may be difficult, and urinary retention may occur. The exposed patient’s temperature may become elevated from an inability to sweat and dissipate heat. In warm climates, this may result in marked hyperthermia.
Numerous substances can cause the anticholinergic syndrome (see also section Agents Related to the Anticholinergic Toxidrome). More common agents include antihistamines, tricyclic antidepressant (TCA) drugs, phenothiazines, selective serotonin and norepinephrine reuptake inhibitors (SSRIs and SNRIs), and cyclobenzaprine. Several plants of the nightshade family contain deadly alkaloids. Examples include: Belladonna ( Atropa belladonna ), Jimson weed/Devil’s Snare (Datura stramonium), which contain atropine, hyoscyamine, and scopolamine. Mandrake and Henbane also are members of this family of plants.
Acetylcholine is a neurotransmitter found throughout the CNS, including (1) the sympathetic and parasympathetic autonomic ganglia, (2) the postganglionic parasympathetic nervous system, and (3) the skeletal muscle motor end plate. Acetylcholine binds to and activates muscarinic and nicotinic receptors. Activating muscarinic receptors stimulates or inhibits cellular function at visceral smooth muscle, cardiac muscle, and secretory glands. Alternatively, nicotinic receptors are present at postsynaptic membranes in autonomic ganglia and at skeletal muscle motor end plates. The enzyme acetylcholinesterase (AChE) regulates the activity of acetylcholine within the synaptic cleft. Acetylcholine binds to the active site of AChE, where the enzyme rapidly hydrolyzes acetylcholine to choline and acetic acid. These hydrolyzed products rapidly dissociate from AChE, so that the enzyme is free to act on another molecule.
The respiratory effects of cholinergic poisoning tend to be dramatic and are considered to be the major factor leading to the death of its victims. Respiratory failure typically occurs as a triad of increased airway resistance, neuromuscular failure, and depression of central respiratory centers. Profuse watery nasal discharge, marked salivation, bronchorrhea, and bronchoconstriction result in a prolonged expiratory phase, cough, and wheezing. Because of the widespread presence of cholinergic receptors in the brain, cholinergic poisoning can produce great variation in neurologic signs and symptoms, including centrally mediated respiratory failure, coma, and seizures. Cholinergic cardiotoxicity can result in two clinical scenarios: a period of intense sympathetic activity that results in sinus tachydysrhythmias, or a period of increased parasympathetic tone that leads to bradydysrhythmias, prolongation of the PR interval, and atrioventricular block. Muscular symptoms may be vague and may consist of muscular weakness and difficulty with ambulation that can progress to muscular fasciculations and subsequent paralysis. Cholinergic agents cause constriction of both the sphincter muscle of the iris and the ciliary muscle of the lens, as well as stimulation of the lacrimal gland, resulting in lacrimation and miosis. The dermal sweat glands are innervated by sympathetic muscarinic receptors. When these receptors are stimulated, profuse sweating occurs. Cholinergic gastrointestinal and genitourinary symptoms may result in nausea, vomiting, abdominal cramps, tenesmus, and involuntary defecation and urination. Two mnemonics have been developed to help recall cholinergic clinical effects: DUMB BELS (diarrhea, urination, miosis, bradycardia, and bronchorrhea-bronchoconstriction, emesis, lacrimation, sweating-salivation) and SLUDGE (salivation, lacrimation, urination, defecation, GI distress, emesis-eye findings, miosis).
Agents that cause these cholinergic clinical effects include organophosphates and carbamates (see also section Agents Related to the Cholinergic Toxidrome). These agents, which include pesticides, medications (Irinotecan), some mushroom species, and chemical warfare agents (e.g., nerve agents), inhibit the activity of AChE, thereby prolonging the action of acetylcholine at muscarinic and nicotinic receptors. Cholinergic clinical effects can also follow after poisoning with direct nicotinic receptor agonists (e.g., nicotine, lobeline, conine, and arecoline) and specific drugs with direct muscarinic activity (e.g., methacholine, bethanechol, cevimeline).
Opioids can induce coma, respiratory depression, bradycardia, hypotension, hypothermia, miosis, pulmonary edema, decreased bowel sounds, and decreased reflexes. Common causes of this syndrome, in which CNS depression, respiratory depression, and miosis are the cardinal three signs, include morphine, diacetylmorphine (heroin), fentanyl, hydrocodone, hydromorphone, methadone, and oxycodone. Meperidine, and tramadol toxicity has been associated with mydriasis and not with miosis. Numerous drugs can mimic the opioid syndrome triad by inducing coma, respiratory depression, and miosis, including alpha-2 agonists (e.g., clonidine, oxymetazoline) and antipsychotics.
Sedative-hypnotics are a broad class of drugs (e.g., benzodiazepines, barbiturates, meprobamate, ethchlorvynol, zolpidem) that can induce sedation, respiratory depression, hypotension, hyporeflexia, nystagmus, dysarthria, staggering gait, apnea, and coma. Numerous sedative-hypnotics have been utilized to commit drug-facilitated crimes (see below), such as the solution of chloral hydrate in ethanol (historically called a “Mickey Finn”). There are few diagnostic clinical features that distinguish the drugs in this large class from one another. These features will be described in the sections below.
Norepinephrine is the neurotransmitter for postganglionic sympathetic fibers (adrenergic) that enervate skin, eyes, heart, lungs, gastrointestinal tract, exocrine glands, and some neuronal tracts in the CNS. Physiologic responses to activation of the adrenergic system are complex and depend on the type of receptor (α 1 , α 2 , β 1 , β 2 ) activated; some are excitatory and others have opposing inhibitory responses. Stimulation of the sympathetic nervous system produces CNS excitation (agitation, anxiety, tremors, delusions, and paranoia), tachycardia, hypertension, mydriasis, hyperpyrexia, and diaphoresis. In severe cases, seizures, cardiac arrhythmias, and coma may occur. Examples of drugs that produce an excitatory sympathomimetic response include amphetamines, cocaine, phencyclidine, ephedrine, methcathinone, and pseudoephedrine.
Toxin-induced hyperthermic syndromes are potentially devastating and require rapid management. Even though the patient’s temperature is one of the vital signs, the temperature often is not obtained in clinical practice. Fever in a poisoned patient can be associated with several hyperthermic syndromes: sympathomimetic toxicity, uncoupling of oxidative phosphorylation, serotonin syndrome, neuroleptic malignant syndrome, malignant hyperthermia, anticholinergic poisoning, and withdrawal syndromes. Sympathomimetics, such as amphetamines and cocaine, may produce hyperthermia as the result of excess serotonin and dopamine, leading to thermal deregulation. Uncoupling of oxidative phosphorylation, as seen in severe salicylate poisoning, occurs when the process of oxidative phosphorylation is disrupted, leading to heat generation and a reduced ability to aerobically generate adenosine-5′-triphosphate (ATP).
Serotonin syndrome occurs when a relative excess of serotonin is present at both peripheral and central serotonergic receptors. Patients may present with hyperthermia, alterations in mental status, and neuromuscular abnormalities (rigidity, hyperreflexia, clonus), although individual variability is noted in these findings. Serotonin syndrome is associated with drug interactions such as those associated with the combination of monoamine oxidase (MAO) inhibitors and meperidine, but it may also occur with single-agent therapeutic dosing or overdosing with serotonergic agents.
Neuroleptic malignant syndrome is a condition caused by relative deficiency of dopamine within the CNS. It has been associated with dopamine receptor antagonists and the withdrawal of dopamine agonists such as levodopa/carbidopa products. Malignant hyperthermia occurs when genetically susceptible individuals are exposed to specific depolarizing neuromuscular blocking agents or volatile general anesthetics. It clinically is associated with elevated temperature, tachycardia, muscle rigidity, and hypercarbia. Anticholinergic poisoning may result in hyperthermia through impairment of normal cooling mechanisms such as sweating. Withdrawal syndromes can produce excessive adrenergic responses (e.g., sedative-hypnotic withdrawal, ethanol withdrawal) and subsequent heat generation. It should be noted that opioid withdrawal is not associated with fever or altered mental status. Overall, differentiating between the toxic hyperthermic syndromes may be challenging, and additional causes of hyperthermia such as heat stroke and infection should be explored.
Interpretation of the ECG in the poisoned patient can significantly facilitate appropriate laboratory testing, diagnosis, and management of the poisoned patient, because numerous substances can cause ECG changes. Despite the fact that drugs have widely varying indications for therapeutic use, many unrelated drugs share common cardiac electrocardiographic effects if taken at therapeutic doses or in overdose. Potential toxins can be placed into broad classes on the basis of their cardiac effects. For example, agents that block cardiac potassium efflux channels and agents that block cardiac fast sodium channels can lead to characteristic changes in cardiac indices consisting of QT prolongation and QRS prolongation, respectively. The recognition of specific ECG changes associated with other clinical data (toxidromes) can be potentially lifesaving.
Radiologic testing is sometimes used to diagnose complications associated with poisonings, such as aspiration pneumonitis and anoxic brain injury. The use of radiology has also been advocated to detect the presence of potentially radiopaque poisons. For example, O’Brien and associates studied the detectability of 459 different tablets and capsules using plain radiography. Investigators used a ferrous sulfate tablet as a control in grading the radiopacity of other tablets. Overall, of the wide variety of pills tested, only 6.3% were graded as having radiopacity the same as or greater than ferrous sulfate; 29.6% were regarded as having at least moderate opacity; and the largest remaining portion of pills (64%) was regarded as no more than minimally detectable. Based on this and other studies, the indiscriminate use of plain abdominal x-rays is not justified, and a negative film should not be relied upon to rule out potential toxic pill ingestion, especially if enough time is given to allow the pills to dissolve.
Obtaining a basic metabolic panel in all poisoned patients is recommended and is an important initial screening test. When low serum bicarbonate is discovered on a metabolic panel, the clinician should determine whether an elevated anion gap exists. The formula most commonly used for the anion gap (AG) calculation is as follows :
The primary cation (sodium) and anions (chloride and bicarbonate) are represented in the equation. Other serum cations are not commonly included in this calculation because either their concentrations are relatively low (e.g., potassium) or assigning a number to represent their respective contribution is difficult (e.g., magnesium, calcium). Similarly, a multitude of other serum anions (e.g., sulfate, phosphate, organic anions) are also difficult to measure and quantify in an equation. , These “unmeasured” ions represent the anion gap calculated using the previous equation. The reference limit for this anion gap is accepted to be 8 to 16 mmol/L, but it has been suggested that because of changes in the technique used to measure chloride, these limits should be lowered to 6 to 14 mmol/L. Practically speaking, an increase in the anion gap beyond an accepted reference limit, accompanied by a metabolic acidosis, represents an increase in unmeasured endogenous (e.g., lactate) or exogenous (e.g., salicylates) anions. Clinically useful mnemonics for causes of high anion gap metabolic acidoses are the classic MUDPILES (representing Methanol, Uremia, Diabetic (as well as alcoholic and starvation) ketoacidosis, Propylene glycol, Iron, Isoniazid,/Inhalants, Lactate, Ethylene glycol, and Salicylate) and the more recently proposed GOLD MARK (Glycols [ethylene and propylene], Oxoproline, l -lactate, d -lactate, Methanol, Aspirin, Renal failure, and Ketoacidosis). For further discussion of these causes of high anion gap metabolic acidoses the reader is referred to Chapter 50 .
It is imperative that clinicians who admit poisoned patients initially presenting with an increased anion gap metabolic acidosis investigate the cause of that acidosis. Many symptomatic poisoned patients may have an initial mild metabolic acidosis upon presentation caused by processes resulting in elevated serum lactate (e.g., transient hypoxia or hypovolemia). However, with adequate supportive care (e.g., oxygenation and hydration), the anion gap acidosis should steadily improve. If, despite adequate supportive care, an anion gap metabolic acidosis worsens in a poisoned patient, the clinician should consider continued absorption of exogenous acids (e.g., salicylate), formation of acidic metabolites (e.g., ethylene glycol, methanol, toluene metabolites), and cellular ischemia with worsening lactic acidosis (e.g., cyanide) as potential causes.
The main osmotically active constituents of serum are Na + , Cl − HCO 3 − , glucose, and urea. Several empirical formulae based on measurement of these substances have been used to estimate the serum osmolality. In practice, one has not shown itself to be superior to the others, yet each equation demonstrates significant differences in the osmolal gap reference interval. Therefore reference intervals must be validated on appropriate patient populations. Two commonly used formulas (in conventional and SI units) are presented here:
or
The difference between the actual osmolality (OSMm), measured by freezing-point depression, and the calculated osmolality (OSMc) is referred to as delta-osmolality, or the osmolal gap (OSMg).
Elevated OSMg implies the presence of unmeasured osmotically active substances. Volatile alcohols (ethanol, methanol, isopropanol, acetone, and ethylene glycol) when present at significant concentrations increase serum osmolality, thus resulting in an increased OSMg. The calculation of OSMg is commonly used as a screen. However, it is important to remember that volatile alcohols are not detected when osmolality is measured with a vapor pressure osmometer. Therefore for the purpose of determining the OSMg, only osmolality measurements based on freezing-point depression are acceptable. Most clinical osmometers measure the temperature at which a solution freezes (freezing point depression). When 1000 mosmol of a small solute(s) is added to 1 kg of water, the freezing point of the solution will be depressed 1.86 °C. The freezing point of plasma water is −0.521 °C, therefore plasma osmolality is (0.521/1.86) 280 mosmol/kg.
What constitutes a normal osmolal gap is widely debated. Traditionally, a normal gap has been defined as 10 mOsm/kg or less. The original source of this value is an article by Smithline and Gardner, which declared that this number was pure convention. A further clinical study has not shown this assumption to be correct. However, large variability is seen in the normal population. Researchers have found the OSMg to vary from −9 to +5 mOsm/kg ; from −13.5 to +8.9 ; and from −10 to +20 mOsm/kg, depending on the population studied. An important point to consider is that the day-to-day coefficient of variance of sodium is 1%. This analytical variance alone may account for the variation found in patients’ osmolal gaps.
One would expect that each 100 mg/dL (21.7 mmol/L) of ethanol (molecular weight = 46.068 g/mol) in serum results in an approximate increase of 21.7 mOsm/kg. However, this is not found to be the case. Applying a correction factor of 0.83 to the ethanol value will more closely approximate the contribution of ethanol to the OSMg. By considering this effect of ethanol on the serum osmolality, it is possible to determine what portion of an increased osmolal gap is due to ethanol. The contribution of ethanol to the measured osmolality can be calculated (ethanol, mg/dL/4.6 × 0.83) and included in the preceding formula for delta osmolality calculation. However, it has been observed that ethanol and methanol do not follow a completely predictable relationship with OSMg. In severe ethanol and methanol intoxication, OSMg increases with increasing concentration, making it appear that something is present besides the alcohol. ,
A significant residual osmolal gap (>10 mOsm/kg) would suggest the possible presence of isopropanol, methanol, acetone, or ethylene glycol or other osmotically active substance. This information, in conjunction with the presence or absence of metabolic acidosis or serum acetone, or other clinical indicators, is helpful to the clinician when specific measurements of alcohols other than ethanol are not available on an emergency basis ( Table 43.3 ). It must be realized that ketones and substances administered to patients such as polyethylene glycol (burn cream), mannitol (osmotic diuretic), and propylene glycol (solvent for diazepam and phenytoin) may increase serum osmolality. Osmolal gap may also be increased in advanced chronic kidney disease or other clinical conditions.
Alcohol | Serum Osmolal Gap | Metabolic Acidosis With Anion Gap | Serum Acetone | Urine Oxalate |
---|---|---|---|---|
Ethanol | + | − | − | − |
Methanol | + | + | − | − |
Isopropanol | + | − | + | − |
Ethylene glycol | + | + | − | + |
For the diagnosis of ethanol intoxication, OSMg has lost its usefulness because ethanol can be measured quickly on most chemistry analyzers. However, because other toxic alcohols can be measured only by chromatographic techniques that take much longer to perform, it is still useful. Unfortunately, OSMg as a screening method is insensitive to low, yet potentially clinically significant, concentrations of ethylene glycol (<50 mg/dL) and methanol (<30 mg/dL).
Screening procedures are designed for the relatively rapid and generally qualitative detection of a particular drug or other toxic substance/s. In general, screening tests have adequate clinical sensitivity but may not be highly specific. Thus a negative result yielded by a screening procedure may rule out with reasonable certainty the presence of clinically significant concentrations of a particular analyte/drug but not necessarily the entire class of drugs. However, because of possible interferences, a positive result should be considered “presumptive positive” and should be confirmed by an alternate procedure of greater specificity.
Spot tests are less frequently employed now because some have been largely replaced by rapid immunoassays that may be performed at the point of care (POC) or in the central laboratory. For a more comprehensive description, refer to previous editions of this textbook.
Different types of immunoassays are useful in screening specimens for drugs. In some cases, these assays are relatively specific for a single drug (LSD), but in others, several drugs of a similar class are detected (e.g., opiates). The detection limit for various members of a class of drugs or the degree of cross-reactivity for similar drugs varies, and each manufacturer of immunoassay reagents should be consulted for specific information. These assays are easy to perform; many are available for use on automated instrumentation and may be able to provide “semiquantitative” results. For the vast majority of drugs of abuse, immunoassays are the methods of choice for initial screening. However, for a more comprehensive drug screening, chromatographic procedures complement immunoassays.
Numerous POC drug test devices for urine (and oral fluid) are designed for easy, rugged, and portable use by nontechnical personnel. Although these devices are relatively simple to use, proper training of nonlaboratory users is important for optimal performance (see Chapter 30 ). , These noninstrumental immunoassay test devices are designed for use at the site of collection; results are available within minutes and are variously configured to detect only one drug or many drugs simultaneously. The spectrum of drugs tested commonly includes the traditional SAMHSA (Substance Abuse and Mental Health Services Administration) or d 5 (National Institute on Drug Abuse, amphetamine, cocaine, marijuana, opiates, and phencyclidine) but may also include barbiturates, benzodiazepines, buprenorphine, methamphetamine, methadone, methylenedioxymethamphetamine (MDMA), oxycodone, fentanyl, and TCAs.
As previously cited, such devices are also available for measurement of acetaminophen and salicylate in serum or whole blood. Evaluations for some of these test devices for urine and oral fluid , have been published. A comprehensive review of on-site drug testing is also available. The assay principles of these POC test devices include the following:
Sequential competitive binding microparticle capture immunoassay
Homogeneous microparticle capture immunochromatography
Solid-phase competitive sequential enzyme immunoassay
Latex-agglutination-inhibition immunoassay
A more detailed description of these methods can be found in Chapter 30 and in the manufacturer package insert for each specific test kit.
Planar chromatography, commonly known as thin-layer chromatography (TLC), is a versatile procedure that requires no instrumentation and thus is operationally relatively simple and inexpensive. However, application of TLC to drug screening requires considerable experience and skill to recognize drug and metabolite patterns and various color hues for detection; it has largely been replaced by other chromatographic techniques. For a more comprehensive description, refer to previous editions of this textbook.
Also known as gas liquid chromatography, GC is relatively rapid, capable of resolving a broad spectrum of drugs, and is widely used for qualitative and quantitative drug analysis. Capillary columns, because of their high efficiency, are analytical columns that are commonly used for drug detection by GC (see Chapter 19 ). In many instances, nonderivatized drugs have good GC properties when capillary columns are used, but in other instances, derivatization to a less polar or more volatile compound is necessary. Common detectors for drug detection by GC are flame ionization and alkali flame ionization (nitrogen phosphorus [NP]) detectors and mass spectrometers, which provide the greatest accuracy of identification. Numerous methods for general drug screening by GC-MS have been published, but one comprehensive method that can be adapted to multiple body fluids and tissues is referenced.
The resolving power of HPLC (see Chapter 19 ) for separating widely divergent chemical constituents has been applied to the complex challenge of comprehensive drug screening in biological fluids. Advantages of HPLC over GC include the ability to analyze polar compounds without derivatization (e.g., morphine, benzoylecgonine [BE]) and thermally labile drugs (e.g., chlordiazepoxide). The advent of diode array detectors that provide a spectral scan of compounds as they elute from the column greatly increased the discriminatory power of this technique. LC-MS or LC-MS/MS and high-resolution mass spectrometry (HRMS—TOF, orbitrap) currently play a limited role in comprehensive screening, but are rapidly gaining in popularity over GC-MS for drug confirmations. , Comprehensive drug screens using HRMS require highly trained technical staff and are typically used in high-volume hospital laboratory settings or at reference laboratories ( Chapters 19 and 20 ).
The toxic, pharmacologic, biochemical, and analytical characteristics of several individual drugs and toxins are discussed in this section.
Carbon monoxide and methemoglobin-forming agents interfere with oxygen transport, resulting in cellular hypoxia. Cyanide interferes with oxygen use and therefore causes an apparent cellular hypoxia.
Carbon monoxide (CO) is a colorless, odorless, tasteless gas that is a product of incomplete combustion of carbonaceous material. Common exogenous sources of carbon monoxide include cigarette smoke, gasoline engines, and improperly ventilated home heating units. Small amounts of carbon monoxide are produced endogenously in the metabolic conversion of heme to biliverdin. This endogenous production of carbon monoxide is accelerated in hemolytic anemias.
When inhaled, carbon monoxide combines tightly with the heme-Fe 2+ of hemoglobin to form carboxyhemoglobin. The binding affinity of hemoglobin for carbon monoxide is about 250 times greater than that for oxygen. Therefore high concentrations of carboxyhemoglobin limit the oxygen content of blood. Moreover, the binding of carbon monoxide to a hemoglobin subunit increases the oxygen affinity for the remaining subunits in the hemoglobin tetramer. Thus at a given tissue P O 2 value, less oxygen dissociates from hemoglobin when carbon monoxide is also bound, shifting the hemoglobin-oxygen dissociation curve to the left. Consequently, carbon monoxide not only decreases the oxygen content of blood, it also decreases oxygen availability to tissue, thereby producing a greater degree of tissue hypoxia than would result from an equivalent reduction in oxyhemoglobin due to hypoxia alone. , Carbon monoxide may also bind to other heme proteins, such as myoglobin and mitochondrial cytochrome oxidase a 3 ; this may limit oxygen use when tissue P O 2 is very low. ,
The toxic effects of carbon monoxide are a result of tissue hypoxia. Organs with high oxygen demand, such as heart and brain, are most sensitive to hypoxia and thus account for the major clinical sequelae of carbon monoxide poisoning. It must be emphasized that the carboxyhemoglobin concentration, although helpful in diagnosis, does not always correlate with the clinical findings or prognosis. , Factors other than carboxyhemoglobin concentration that contribute to toxicity include length of exposure, metabolic activity, and underlying disease, especially cardiac or cerebrovascular disease. Moreover, low carboxyhemoglobin concentrations relative to the severity of poisoning may be observed if the patient was removed from the carbon monoxide–contaminated environment a significant time before blood sampling.
An insidious effect of carbon monoxide poisoning is the delayed development of neuropsychiatric sequelae, which may include personality changes, motor disturbances, and memory impairment. These manifestations do not correlate with the length of exposure or with the maximum blood carboxyhemoglobin concentration. ,
Treatment for carbon monoxide poisoning involves removal of the individual from the contaminated area and administration of oxygen. The half-life (t ½ ) of carboxyhemoglobin in the body is variable, and attempts to determine the exact elimination t ½ for CO based on the inhaled oxygen concentration have not been validated. In room air the approximate half-life is about 4 to 5 hours, and during hyperbaric oxygen therapy it is as short as 12 to 20 minutes. The findings from randomized controlled human studies are not consistent regarding the benefit of hyperbaric oxygen for carbon monoxide poisoned patients and there is therefore no consensus regarding its role in treatment.
Carbon monoxide may be released from hemoglobin and then measured by GC, or it may be determined indirectly as carboxyhemoglobin by spectrophotometry. Gas chromatographic methods are accurate and precise even for very low concentrations of carbon monoxide. Spectrophotometric methods are rapid, convenient, accurate, and precise, except at very low concentrations of carboxyhemoglobin (<2 to 3%).
Gas chromatographic methods measure the carbon monoxide content of blood. When blood is treated with potassium ferricyanide, carboxyhemoglobin is converted to methemoglobin, and carbon monoxide is released into the gas phase. Measurement of the released carbon monoxide may be performed by GC using a molecular sieve column and a thermal conductivity detector. A lower detection limit is achieved by incorporating a reducing catalyst (e.g., nickel) between the GC column and the detector to convert carbon monoxide to methane. The methane may then be detected with a flame ionization detector. A very low detection limit may be achieved with the use of a heated mercuric oxide reaction chamber between the GC column and an ultraviolet light detector. As carbon monoxide elutes from the column, it reacts with mercuric oxide to form mercury gas, which has a high molar absorptivity at 254 nm. In practice, the carbon monoxide binding capacity is also determined after an aliquot of the blood specimen is treated with carbon monoxide to saturate the hemoglobin. The results are then expressed as percent of carboxyhemoglobin:
GC methods are accurate and precise and are considered to be reference procedures. Reference limits for carboxyhemoglobin in rural nonsmokers are about 0.5%; for urban nonsmokers, 1 to 2%; and for smokers, 5 to 6%. Values may be increased by about 3% HbCO in cases of hemolytic anemia.
Spectrophotometric methods rely on the characteristic spectral absorption properties of carboxyhemoglobin. , Among several such methods, the most popular are based on automated, multiwavelength measurements of several hemoglobin species. Automated, multicomponent spectrophotometric methods are most rapid and convenient for the determination of carboxyhemoglobin and other hemoglobin species. Spectrophotometric methods generally compare favorably with gas chromatographic procedures at carboxyhemoglobin concentrations greater than 2 to 3%, but their precision is poor below these concentrations. Therefore they are sufficiently accurate and precise for measurement of carbon monoxide after exogenous exposure but are too insensitive to detect the increased endogenous production of carbon monoxide that occurs in hemolytic anemia.
Fetal hemoglobin has slightly different spectral properties than adult hemoglobin. Consequently, falsely high carboxyhemoglobin values of 4 to 7% may occur when blood from neonates is measured by some spectrophotometric methods utilizing fewer wavelengths. Moreover, erroneous results may occur with lipemic specimens, with bilirubin, and in the presence of methylene blue (see section below on “Methemoglobin-Forming Agents”).
The heme iron in hemoglobin is normally present in the ferrous state (Fe 2+ ). When oxidized to the ferric state (Fe 3+ ), methemoglobin is formed, and this form of hemoglobin cannot bind oxygen. Congenital methemoglobinemia may result from a deficiency of NADH-methemoglobin reductase or, more rarely, from hemoglobin variants (hemoglobin M) in which heme iron is both more susceptible to oxidation and more resistant to reduction by the methemoglobin reductase system (for more information on methemoglobinemia, refer to Chapters 40 and 78 ).
An acquired (toxic) methemoglobinemia may be caused by various drugs and chemicals ( Table 43.4 ). Additionally, oxides of nitrogen and other oxidant combustion products make smoke inhalation a potential cause of methemoglobinemia. The normal percentage of methemoglobin is less than 1.5% of total hemoglobin. The severity of symptoms usually correlates with measured methemoglobin levels; methemoglobin percentages up to 20% may cause slate-gray cutaneous discoloration, cyanosis, and chocolate-brown blood. Percentages between 20 and 50% may cause dyspnea, exercise intolerance, fatigue, weakness, and syncope. More severe symptoms of dysrhythmias, seizures, metabolic acidosis, and coma are associated with methemoglobin percentages of 50 to 70%, and greater than 70% may be lethal. , All of these symptoms are a consequence of hypoxia associated with the diminished O 2 content of the blood, and with a decreased O 2 dissociation from hemoglobin species in which some, but not all, subunits contain heme iron in the ferric state (i.e., shift of dissociation curve to the left). The P O 2 is normal in these patients, and therefore so is the calculated hemoglobin oxygen saturation. Thus a normal P O 2 in a cyanotic patient is a significant indication for the possible presence of methemoglobinemia. Direct measurement of methemoglobin is important in these cases and may be performed by the manual spectrophotometric method of Evelyn and Malloy or by automated multiwavelength measurements with a co-oximeter (see section on “Carbon Monoxide”).
Local anesthetics | Analgesics | Miscellaneous |
---|---|---|
Benzocaine | Phenazopyridine | Aminophenol |
Lidocaine | Phenacetin | Aniline, p -chloraniline |
Prilocaine | Nitrites and nitrates | Bromates |
Antimicrobials | Ammonium nitrate | Chlorates |
Chloroquine | Amyl nitrite | 4-Dimethyl-amino-phenolate (4-DMAP) |
Dapsone | Butyl nitrite | Metoclopramide |
Primaquine | Isobutyl nitrite | Nitrobenzene |
Sulfonamides | Potassium nitrate | Nitroethane |
Trimethoprim | Sodium nitrate | Nitroglycerin |
Nitrogen oxides | Phenazopyridine | |
Nitric oxide | Potassium permanganate | |
Nitrogen dioxide | Propanil |
Specific therapy for toxic methemoglobinemia involves the administration of methylene blue, which acts as an electron transfer agent in the NADPH-methemoglobin reductase reaction, thereby increasing the activity of this system several-fold. Methylene blue and sulfhemoglobin cause spectral interference in the measurement of methemoglobin with some co-oximeters , but not with the Evelyn-Malloy method. Ascorbic acid can also reverse methemoglobin by an alternate metabolic pathway, but it is of minimal use acutely because of its slow action.
Methemoglobin can be measured in blood manually, , or by automated multiwavelength measurements with a co-oximeter. Methemoglobin interferes with the noninvasive pulse oximetry method, measuring the absorbance of light at 660 nm (oxyhemoglobin) and 940 nm (deoxyhemoglobin). Because methemoglobin is not stable at room temperature, specimens should be kept on ice or refrigerated but not frozen. The stability of methemoglobin at 4 °C has not been well studied. Some sources indicate significant decreases in methemoglobin concentration after 4 to 8 hours, whereas others report little or no change after 24 hours. Freezing results in an increase in methemoglobin concentration.
Cyanide consists of one atom of carbon bound to one atom of nitrogen by three molecular bonds (C[N). Inorganic cyanides (also known as cyanide salts) contain cyanide in the anion form ( CN − ) and are used in numerous industries, such as metallurgy, photographic developing, plastic manufacturing, fumigation, and mining. Organic compounds that have a cyano group bonded to an alkyl residue are called nitriles; for example, acetonitrile, also known as methyl cyanide (CH 3 CN). Hydrogen cyanide (HCN) is a colorless gas at standard temperature and pressure with a rarely reported associated “bitter almond” odor. Cyanogen gas, a dimer of cyanide, reacts with water and breaks down into the cyanide anion. Many plants, such as Manihot spp. (cassava), Linum spp., Lotus spp., Prunus spp., Sorghum spp., and Phaseolus spp., contain cyanogenic glycosides. Iatrogenic cyanide poisoning may occur during use of nitroprusside as a vasodilator given to reduce blood pressure and afterload. Each nitroprusside molecule contains five cyanide molecules, which are slowly released in vivo. If endogenous sulfate stores are depleted, as in the malnourished or postoperative patient, cyanide may accumulate even with therapeutic nitroprusside infusion rates (2 to 10 μg/kg/min) infused for long periods of time.
Hydrocyanic acid binds to hemoglobin. The hydrocyanic acid bound in the erythrocyte is in equilibrium with free hydrocyanic acid in the serum at a ratio of 10:1. Cyanide in serum readily crosses all biological membranes and avidly binds to heme iron (Fe 3+ ) in the cytochrome oxidase -a 3 complex within mitochondria. , When bound to cytochrome oxidase -a 3 , cyanide is a competitive inhibitor that causes decoupling of oxidative phosphorylation. Patients exposed to toxic concentrations of cyanide may exhibit rapid onset of symptoms typical of cellular hypoxia—flushing, headache, nausea and vomiting, anxiety, confusion, and collapse, initial hypertension and tachycardia progressing to hypotension, cyanosis bradycardia, and apnea, coma, seizures, complete heart block, and death if the dose is sufficiently large.
Hydroxycobalamin or the cyanide antidote kit should be administered as soon as cyanide poisoning is suspected. Hydroxocobalamin, a vitamin B 12 precursor, is a metalloprotein with a central cobalt atom that complexes cyanide, forming cyanocobalamin (vitamin B 12 ). Cyanocobalamin is eliminated in the urine or releases the cyanide moiety at a rate sufficient to allow detoxification by rhodanese. The cyanide antidote kit contains amyl nitrite, sodium nitrite, and sodium thiosulfate. Thiosulfate donates the sulfur atoms necessary for rhodanese-mediated cyanide biotransformation to thiocyanate. The mechanism of nitrite is less clear. The traditional rationale relies on the ability of nitrite to generate methemoglobin. Because cyanide has a higher affinity for methemoglobin than for cytochrome a 3 , cytochrome oxidase function is restored.
Cyanide determination was traditionally performed by spectrometry following microdiffusion separation. However, more recently chromatographic methods have been employed. GC has used both headspace gas and direct injection sampling coupled with electron capture (EC), nitrogen selective, or mass spectrometric detection methods. Liquid chromatography followed by fluorescence (FD) and mass spectrometric detection have also been described. ,
Several alcohols are toxic and medically important; they include ethanol, methanol, isopropanol, acetone (also a metabolite of isopropanol), and ethylene glycol.
Ethanol is the most widely used and often abused chemical substance. Consequently, measurement of ethanol is one of the more frequently performed tests in the toxicology laboratory. Ethanol is considered a CNS depressant whose effects vary depending on the blood ethanol concentration ( Table 43.5 ) but are also heavily influenced by an individual’s tolerance. Symptoms vary from euphoria and decreased inhibitions, to increased disorientation and incoordination, and then to coma and death. A blood alcohol concentration of 80 mg/dL (0.08 g/100 mL or 0.08% w/v, 17.4 mmol/L) or less (50 mg/dL in some states) has been established as the statutory limit for operation of a motor vehicle in most countries.
Blood Alcohol Concentration, % (mg/dL) | Influence | Clinical Signs/Symptoms |
---|---|---|
0.01–0.05 10–50 |
Subclinical | Influence/effects not apparent or obvious Behavior appears normal Impairment detectable by special tests |
0.03–0.12 (30–120) |
Euphoria | Mild euphoria increased sociability, talkativeness, self-confidence decreased inhibitions mild sensorimotor impairment Slowed information processing Loss of efficiency in finer performance tests Impairment of perception and memory |
0.09–0.25 (90–250) |
Excitement | Emotional instability; loss of critical judgment comprehension Decreased sensory response; increased reaction time Reduced visual acuity, peripheral vision, and glare recovery Sensorimotor incoordination; impaired balance Drowsiness |
0.18–0.30 (180–300) |
Confusion | Disorientation, mental confusion; dizziness Exaggerated emotional states (fear, rage, grief, etc.) Disturbances of vision (diplopia, etc.) and of perception of color, form, motion, dimensions Increased pain threshold Ataxia; dysarthria, apathy, lethargy |
0.25–0.40 (250–400) |
Stupor | General inertia; approaching loss of motor functions Markedly decreased response to stimuli Marked muscular incoordination; inability to stand or walk Vomiting; incontinence of urine and feces Impaired consciousness; sleep or stupor |
0.35–0.50 (350–500) |
Coma | Unconsciousness; coma; anesthesia Depressed or absent reflexes Subnormal temperature Impairment of circulation and respiration Possible death |
>0.45 (>450) |
Death | Possible death from respiratory arrest |
Because of many factors, not all individuals experience the same degree of CNS dysfunction at similar blood alcohol concentrations. Moreover, the CNS actions of ethanol are more pronounced when the blood ethanol concentration is increasing (absorptive phase) than when it is declining (elimination phase), in part because of the phenomenon of acute tolerance. In addition, heavy alcohol use leads to a more chronic form of tolerance. When consumed with other CNS depressant drugs, ethanol exerts a potentiation or synergistic depressant effect. This can occur at relatively low alcohol concentrations, and numerous deaths have resulted from combined ethanol and drug ingestion.
The pharmacologic mechanisms for the CNS depressant actions of ethanol are complex and incompletely understood, but probably involve both enhancement of major inhibitory neurons and impairment of excitatory neurons. The principal CNS inhibitory neuronal system is mediated by the neurotransmitter γ-aminobutyric acid (GABA). When GABA binds to its postsynaptic receptor subtype GABA A , this oligomeric ion-gated complex “opens” to allow inward flux of Cl, leading to membrane hyperpolarization and subsequent decreased electrical response. This GABA-mediated inhibitory response is enhanced by ethanol and sedative, hypnotic, and anesthetic agents, including barbiturates, benzodiazepines, and volatile anesthetics. Neuronal nicotinic acetylcholine receptors also may be prominent molecular targets of alcohol. Both enhancement and inhibition of nicotinic acetylcholine receptor function have been reported depending on receptor subunit concentration and the concentrations of ethanol tested. Ethanol also inhibits the function of the N -methyl-d-aspartate (NMDA)- and kainate-receptor subtypes; however, α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA) receptors are largely resistant to ethanol.
The aforementioned chronic tolerance to ethanol is considered to be mediated by ethanol-induced increased responsiveness and upregulation in the synthesis of NMDA receptors, attained by concomitant downregulation and desensitization through phosphorylation of GABA A and glutamate receptors. , , Largely because of these adaptive changes, abrupt withdrawal from chronic, heavy ethanol use leads to a physical abstinence syndrome that has prominent features which are the opposite of those produced by intoxication. This include features of CNS excitation, anxiety, irritability, insomnia, muscle tremor and cramps, hallucinations, and increased temperature, blood pressure, heart rate, and seizures. This syndrome can be fatal if the patient is not monitored properly.
Ethanol is metabolized principally by liver alcohol dehydrogenase (ADH) to acetaldehyde, which is subsequently oxidized to acetic acid by aldehyde dehydrogenase ( Fig. 43.1 ). The rate of elimination of ethanol from blood approximates a zero-order process. This rate varies among individuals, averaging about 15 mg/dL/h for males and 18 mg/dL/h for females. , At both low (<20 mg/dL) and high (>300 mg/dL) ethanol concentrations, elimination more closely resembles first-order kinetics, and it is accelerated at high concentrations. The elimination rate is also influenced by drinking habits (e.g., alcoholics have increased elimination rates caused by enzyme induction).
Ethanol is a teratogen, and alcohol consumption during pregnancy can result in a baby with fetal alcohol spectrum disorder (FASD). FASD is an umbrella term that describes the variety of effects that can occur in an individual whose mother drank alcohol during pregnancy. These effects may include physical, mental, behavioral, and/or learning disabilities with possible lifelong implications and are 100% preventable when a woman completely abstains from alcohol during her pregnancy.
Isopropanol is readily available to the general population as a 70% aqueous solution for use as rubbing alcohol, but can also be found in cleaners, disinfectants, antifreezes, cosmetics, solvents, inks, and pharmaceuticals. It has about twice the CNS depressant action as ethanol. Isopropanol has a short t ½ of 2.5 to 3.0 hours and is rapidly metabolized by ADH. Since it is a secondary alcohol (the hydroxyl group is attached to a central, rather than a terminal, carbon), consequently it is metabolized to a ketone, not an acid, and therefore ingestions do not cause significant metabolic acidosis (see Table 43.3 ). , Acetone is eliminated much more slowly, therefore concentrations of acetone in serum often exceed those of isopropanol during the elimination phase following isopropanol ingestion. Acetone has CNS depressant activity similar to that of ethanol, and because of its longer t ½ , it may prolong the apparent CNS effects of isopropanol intoxications.
Interestingly, like most enzymatic reactions, the conversion of isopropanol to acetone by ADH exists in an equilibrium; its balance is dependent on the ratio of NAD+ and NADH. In patients with severe diabetic ketoacidosis there is an excess of NADH, and the equilibrium reaction favors the formation of isopropanol. Therefore it is not uncommon in these patients to see not only the acetone expected in the diabetic ketoacidosis, but small amounts of isopropanol.
Methanol is used as a solvent in several commercial products, as a constituent in windshield wiper fluid, copy machine fluids, fuel additives (octane boosters), paint remover or thinner, antifreeze, canned heating sources, de-icing fluid, shellacs, and varnishes. It may be consumed intentionally by individuals as an ethanol substitute or accidentally when present as a contaminant in illegal whiskey, or inadvertently by children.
The CNS effects of methanol are substantially less severe than those of ethanol. Methanol is oxidized by liver ADH (at about one tenth the rate of ethanol) to formaldehyde. Formaldehyde in turn is rapidly oxidized by aldehyde dehydrogenase to formic acid. Formic acid’s main mechanism of toxicity is its binding to cytochrome oxidase and blockade of oxidative phosphorylation. This leads to anaerobic metabolism and development of lactic acidosis. In addition, metabolism of methanol increases the NADH/NAD + ratio, which favors the conversion of pyruvate to lactate and thereby worsens lactic acidosis. Falling pH also favors the undissociated form of formic acid (as opposed to formate ion), which moves more readily across tissue barriers. Therefore at lower pH, more formic acid can enter the brain and ocular tissues, worsening CNS depression and causing retinal and optic nerve injury, resulting in the characteristic profound metabolic acidosis, optic neuropathy, potential blindness, CNS bleeds, and death. ,
Ethylene glycol is present in antifreeze products, de-icing products, detergents, paints, and cosmetics. It may be ingested accidentally or for the purpose of inebriation or suicide. Because it tastes sweet, some animals are attracted to it. Veterinarians are often familiar with ethylene glycol toxicity because of cases involving dogs or cats that drank radiator fluid.
Ethylene glycol has initial CNS effects resembling those of ethanol. Ethylene glycol is metabolized by ADH to glycolaldehyde which is then metabolized to glycolic, glyoxylic, and oxalic acids. , Oxalate readily precipitates with calcium to form insoluble calcium oxalate crystals. Tissue injury is caused by widespread deposition of oxalate crystals and the toxic effects of glycolic and glyoxylic acids. , , Clinically, ethylene glycol poisoning is characterized by CNS depression, metabolic acidosis, and renal failure; however, multiple other organ systems may be affected. Poisonings have historically been divided into three stages, although the timing and stages may overlap. The first stage typically begins 30 minutes to 12 hours after ingestion due to the intoxicating effects of the ethylene glycol and may range from mild CNS depression to coma. , Ethylene glycol is directly irritating to the GI tract, so abdominal pain, nausea, and vomiting may be present. The formation of toxic metabolites generally takes 4 to 12 hours. The CNS effects of glycolic acid and calcium oxalate crystals include cerebral edema, basal ganglia hemorrhagic infarction, and meningoencephalitis. Hypocalcemia, which occurs when calcium combines with oxalate, may contribute to seizures. Metabolic acidosis appears as toxic metabolites are generated. The second stage begins 12 to 24 hours after ingestion and is characterized by metabolic acidosis, oxalate crystal deposition in tissues, multi-organ system failure, including heart failure, acute lung injury, and myositis. Most deaths occur during this stage. The third stage is often delayed 24 to 72 hours after ingestion and is characterized by renal failure due to calcium oxalate crystal deposition in the proximal tubules, the most common major complication of serious ethylene glycol poisoning. And if patients survive, delayed neuropathies may occur 5 to 20 days after ethylene glycol poisoning.
In cases of isopropanol poisoning, there is little value in altering isopropanol metabolism because its major metabolite, acetone, is no more toxic than isopropanol itself, and slowing isopropanol metabolism would only serve to prolong the overall CNS depressant effect. For this reason, supportive care is the treatment of choice. In cases of severe isopropanol intoxication, dialysis may be indicated.
The basic principles of treatment for both methanol and ethylene glycol poisoning are essentially the same; correct the acidosis, reduce the formation of toxic metabolites (formaldehyde and formic acid from methanol; glycolaldehyde, glycolic acid, and oxalic acid from ethylene glycol), and enhance the clearance of the parent compound and its toxic metabolites from circulation. Serum concentrations of methanol ≥ 20 mg/dL (6 mmol/L) or ethylene glycol ≥ 20 mg/dL (3 mmol/L) warrants intervention. Mainstays of therapy include the administration of the ADH competitive inhibitors fomepizole (Antizol) or ethanol. They will compete with methanol or ethylene glycol for ADH and thereby impede the formation of the toxic metabolites. Hemodialysis may be required if the measured methanol or ethylene glycol concentrations are greater than 50 mg/dL in order to remove the parent compound and toxic metabolites. , Adjunctive treatment with B vitamins (including folate) is often provided, as these vitamins are thought to assist in the clearance of the toxic metabolites, although definitive evidence to support this hypothesis does not currently exist. ,
Serum, plasma, and whole blood are suitable specimens for the determination of ethanol. Alcohol distributes into the aqueous compartments of blood; because the water content of serum is greater than that of whole blood, higher alcohol concentrations are obtained with serum as compared with whole blood. Experimentally, the serum-to-whole blood ethanol ratio is 1.18 (1.10 to 1.35) and varies slightly with hematocrit. Therefore laboratories that perform alcohol determinations should make clear the choice of specimen.
Because of the volatile nature of alcohols, specimens should be kept capped to avoid evaporative loss. Blood may be stored, when properly sealed, for 14 days at room temperature or at 4 °C, with or without preservative. For longer storage or for nonsterile postmortem specimens, sodium fluoride should be used as a preservative to minimize changes in ethanol concentration.
To measure ethanol in serum/plasma, enzymatic analysis is the method of choice for many laboratories. In this method, ADH catalyzes the oxidation of ethanol to acetaldehyde and NAD is reduced to NADH. With this reaction, the formation of NADH can be measured at 340 nm, and is proportional to the amount of ethanol in the specimen :
Under most assay conditions, ADH is reasonably specific for ethanol, with interferences by isopropanol, acetone, methanol, and ethylene glycol of typically less than 1%. As a precaution the venipuncture site is recommended to be cleansed with an alcohol-free disinfectant, such as aqueous benzalkonium chloride.
Spuriously increased results for ethanol have been described in the presence of high concentrations of lactate dehydrogenase (LDH) together with elevated lactate. , The basis of the interference stems from increased blood concentrations of lactate in conjunction with increased concentrations of LDH. The increased lactate concentration may result from both clinical pathologies and from trauma, especially tissue hypoperfusion. The following reaction describes the source of the interference:
As NADH can be detected in the assay, it is easy to see how increased lactate and LDH could result in a positive ethanol. However, not all ethanol assays are known to have this interference. It is recognized that the lactate and LDH must hit some critical concentration for the interference to occur with those sensitive assays. However, the concentration of lactate and LDH necessary to cause a false positive ethanol in the enzymatic assay for ethanol and the magnitude of the ethanol results generated is very much manufacturer dependant. , Theoretically other dehydrogenases and substrates may cause similar interference.
In clinical laboratories, serum (or plasma) is the most common specimen for ethanol analysis by ADH methods; this method also performs well with urine or oral fluid. Whole blood determinations can be made directly or a precipitation step may be required before analysis to avoid interference from hemoglobin. Results from these methods generally compare closely with those from gas chromatographic methods. For more information about these methods, see “Analysis of Volatile Alcohols” section, later.
During the early part of the 20th century, Dr. Erik M.P. Widmark, a Swedish physician, did much of the foundational research regarding alcohol pharmacokinetics in the human body. In addition, he developed an algebraic equation that can be used to estimate the amount of alcohol consumed by an individual or the associated blood alcohol concentration when the values of the other variables are given , :
N = number of drinks
W = body weight (kg)
ρ (rho) = volume of distribution (L/kg) (0.68 for males, 0.55 for females)
C t = blood alcohol concentration (kg/L)
β = rate of ethanol elimination (0.15 g/L/h)
t = time since first drink (h)
d = specific gravity of alcohol (0.8)
Z = amount of ethanol alcohol per drink (L) (15 mL of ethanol in a standard drink)
Note that it may be necessary to convert the units from those more commonly reported. It is important to remember that this formula should only be used after completion of alcohol absorption, and when equilibrium has been reached between blood and body tissue.
Frequently the time since the first drink is unknown; the formula can be modified to estimate the number of drinks in an individual’s system at the time of the test.
The rate of elimination in the average person is commonly estimated at 0.015 g/100 mL/h (range, 0.010 to 0.030 g/100 mL/h). , Retrograde extrapolation is an estimation of a subject’s alcohol concentration at a prior time, derived from a blood alcohol concentration measured at a later time. This process may be applied when certain assumptions are made concerning absorption rates, elimination rates, and patterns of alcohol consumption, including drinking duration and volume consumed. Unfortunately, to be forensically useful and scientifically valid, such extrapolations may require facts about the person and that person’s alcohol consumption, as well as related information that often is not available. Consequently, significant legal debate surrounds the validity and accuracy of retrograde extrapolation.
Statutory laws for driving under the influence of alcohol were originally based on the concentration of ethanol in venous whole blood. Because the collection of blood is invasive and requires intervention by medical personnel, the determination of alcohol in expired air (breath) has become the mainstay of evidential alcohol measurements. , Clinical interest in determination of breath alcohol at the POC is growing. The fundamental principle for use of breath analysis is that alcohol in capillary alveolar blood rapidly equilibrates with alveolar air in a ratio of approximately 2100:1 (blood:breath). This blood-to-breath ratio may actually be closer to 2300:1 but is also very variable. Nevertheless, in the United States, evidential breath alcohol measurements are based on the ratio of 2100:1. The lower blood-to-breath ratio will predict a slightly lower than actual blood alcohol concentration; its use therefore is not prejudicial. To alleviate confusion and uncertainty surrounding the conversion from breath to blood alcohol concentration, the traffic laws in many countries specify per se limits for blood and/or breath.
Before breath alcohol analysis, a deprivation period of 15 minutes is required to allow for clearance of any residual alcohol that may have been present in the mouth (e.g., very recent drinking, use of alcohol-containing mouthwash, vomiting of alcohol-rich gastric fluid). Duplicate tests performed 5 to 10 minutes apart and within 20 mg/dL (0.02%) are used as an additional safeguard against mouth alcohol contamination.
During the period of active alcohol absorption, generally 30 to 60 minutes depending on a variety of factors, , , and before peak blood alcohol concentration is obtained, the alcohol concentration in arterial blood will be initially higher than that in peripheral venous blood, and the converse is true in the postabsorptive phase. Because end-expiratory air equilibrates with pulmonary alveolar/capillary blood, the breath alcohol concentration more closely reflects that of arterial alcohol ; however, the difference between arterial and venous blood is within the analytical error of most assays.
Determination of ethanol in expired air requires specialized breath alcohol analyzers. Several commercial evidential breath alcohol measurement devices are available. Principles of measurement used in such analyzers include (1) infrared absorption spectrometry (most common), (2) dichromate-sulfuric acid oxidation-reduction (photometric), (3) GC (flame ionization or thermal conductivity detection), (4) electrochemical (EC) oxidation (fuel cell), and (5) metal oxide semiconductor sensors. Breath alcohol devices also may be used for the medical evaluation of patients at the POC (e.g., emergency department).
Oral fluid (saliva) may be easily and noninvasively collected; therefore interest is growing in its use for ethanol measurements and for the detection of drugs, but it is not a frequently used sample for ethanol determinations (see section on “Detection of Drugs of Abuse Using Other Types of Specimens”).
Urine has been used as an alternative, less invasive specimen for the determination of alcohol use. During the postabsorptive phase following alcohol ingestion, the concentration of alcohol in urine is roughly 1.3 times that in blood. However, the use of urine alcohol measurements to estimate blood concentrations is discouraged because the ratio of 1.3 is highly variable and perhaps more important, the urine alcohol concentration may better reflect an average of the blood alcohol concentration during the period in which urine is collected in the bladder. The detection of alcohol in urine represents ingestion of alcohol within the previous 8 to 12 hours.
Ethyl glucuronide (EtG), ethyl sulfate (EtS), and phosphatidylethanols (PEth) are biomarkers of ethanol consumption. EtG and EtS are phase II metabolites of ethanol detected in urine. EtG is a phase II metabolite of ethanol formed through the UDP-glucuronosyltransferase catalyzed conjugation of ethanol with glucuronic acid. EtS is also formed directly by the conjugation of ethanol with sulfate group. PEth are detected in whole blood and are formed by phospholipase D in the presence of ethanol.
Because EtG has a long urinary elimination time (≤80 hours), its specificity for ethanol exposure, and the low detection limits, EtG is utilized as a marker of recent ethanol intake in a variety of clinical and legal settings, including medical monitoring for relapse, emergency department patient evaluation, postmortem assessment, and transportation accident investigation. ,
Monitoring both EtG and EtS in urine improves sensitivity when using ethanol biomarkers for monitoring recent drinking. There are challenges associated with establishing appropriate cut-off concentrations capable of distinguishing between drinking and incidental exposure such as nonbeverage sources of ethanol exposure (i.e., hand sanitizers, mouthwash), , nonuniform laboratory reporting limits, sample stability, and microbial activity may complicate interpretation of results. EtG can be produced in postspeciman collection, and upper respiratory infections and beta glucuronidase hydrolysis in urine sample with certain stains of Escherichia coli may lower levels of EtG; however, mEtS seems to not be affected. However, some interpretive guides have been proposed. EtG greater than 1000 ng/mL may indicate heavy drinking in the previous 48 hours or light drinking the same day. An EtG from 500 to 1000 ng/mL may indicate heavy drinking in the previous 3 days, light drinking in the past 24 hours, or intense “extraneous exposure” within 24 hours. Values less than 500 ng/mL may indicate heavy drinking in the previous 3 days, light drinking in the past 36 hours, or recent “extraneous” exposure.
PEth are a group of phospholipids with a common phosphoethanol head group with two fatty acid chains which differ in chain length and degree of unsaturation. PEth is a promising marker because of PEth’s persistence in blood for as long as 3 weeks after even only a few days of moderately heavy drinking (about four drinks per day). Therefore PEth may be utilized in conjunction with EtG and EtS to discriminate incidental ethanol exposure from moderate to heavy “binge” alcohol drinking. ,
Methanol poisoning can be lethal if not recognized early. Unfortunately, in some instances, a latent period can be as long as 12 to 24 hours before toxicity is recognized, making laboratory identification of this poisoning critical. Development of gas chromatographic methods for volatiles in 1964 157, was a significant step in the recognition and treatment of this very toxic alcohol.
Flame ionization GC remains the most common method for the detection and quantitation of volatile alcohols in biological samples. Not only does it distinguish between ethanol, methanol, isopropanol, and acetone, it has the capability to measure concentrations as low as 10 mg/dL (0.01%). Specimens are prepared by a variety of methods; the two most common are direct injection and headspace analysis. Direct injection involves injection of a sample prepared by diluting it with an aqueous solution of internal standard (thus reducing the amount of matrix introduced into the GC). Repeated injection of biological aqueous matrix into the GC will cause buildup on the injector and front of the analytical column, requiring frequent maintenance and column replacement. This can be alleviated by the use of headspace injection. The volatility of the alcohols is used to separate them from the matrix. Specifically, the “Gas Law” states that at a given temperature, the amount of volatile substance in the air space above the liquid—headspace—is proportional to the concentration of the volatile alcohol in the solution. Therefore the sample in the headspace allows for calculation of the concentration in the specimen.
Headspace gas chromatographic analysis is another excellent method for the measurement of methanol, isopropanol, acetone, and ethanol. In addition, an adaptation of this technique may be used to measure formate, the toxic metabolite of methanol, after esterification to methyl formate. Conversely, direct injection GC is the method of choice for ethylene glycol because it has a higher boiling point and is not as amenable to headspace analysis. A modification of the GC procedure described in 2005 has the potential of combining both toxic alcohols in a single GC analysis. Assessment of ethylene glycol metabolites is a clinically useful method that allows for the simultaneous determination of ethylene glycol and glycolic acid; such a method that is free from interference by propylene glycol (a diluent for parenteral drugs) or 2,3-butanediol (may be present in serum from some alcoholics) would be highly desirable. , Similar techniques are used to measure volatile alcohols in blood, serum, oral fluid, urine, other clinical specimens, and postmortem specimens (e.g., vitreous fluid, skeletal muscle).
Analgesics are substances that relieve pain without causing loss of consciousness. When used in excess, analgesics such as acetaminophen and salicylate can result in a toxic response.
Acetaminophen ( N -acetyl- p -aminophenol; paracetamol) has analgesic and antipyretic actions. In common with the group of drugs referred to as nonsteroidal anti-inflammatory drugs (NSAIDs; e.g., aspirin, ibuprofen, indomethacin), the pharmacologic actions of acetaminophen are related to its nonselective inhibition of cyclooxygenase enzymes (COX). This results in decreased production of prostaglandins, which are important mediators of inflammation, pain (low to moderate), and fever. Contrary to other NSAIDs, acetaminophen has very weak anti-inflammatory activity—a consequence of its weak inhibition of peripheral tissue cyclooxygenase compared with that in the brain. In normal doses, acetaminophen is safe and effective, but it may cause severe hepatic toxicity or death when consumed in overdose quantities. Less frequently, nephrotoxicity with or without associated hepatotoxicity may also occur.
Acetaminophen is normally metabolized in the liver to glucuronide (50 to 60%) and sulfate (<30%) conjugates. A smaller amount (<10%) is metabolized by a cytochrome P450 mixed-function oxidase pathway that is thought to involve formation of a highly reactive intermediate ( Fig. 43.2 ), N -acetyl- p -benzoquinone imine (NAPQI). This intermediate normally undergoes electrophilic conjugation with glutathione and then subsequent transformation to cysteine and mercapturic acid conjugates of acetaminophen. With acetaminophen overdose, the sulfation pathway becomes saturated; consequently, a greater portion is metabolized by the P450 mixed-function oxidase pathway. When the tissue stores of glutathione become depleted, arylation of cellular molecules by the benzoquinoneimine intermediate leads to hepatic necrosis.
The initial clinical findings in acetaminophen toxicity can be absent or relatively mild and nonspecific (nausea, vomiting, and abdominal discomfort) and thus are not predictive of impending hepatic necrosis, which typically begins 24 to 36 hours after toxic ingestion and becomes most severe by 72 to 96 hours. Although uncommon with severe overdose, coma and metabolic acidosis may occur before development of hepatic necrosis.
Specific therapy for acetaminophen overdose is the administration of N-acetylcysteine (NAC), which probably acts as a glutathione substitute. NAC may also provide substrate to replenish hepatic glutathione or to enhance sulfate conjugation, or both. The time of administration of NAC is critical. Maximum efficacy is observed when NAC is administered within 8 hours, and efficacy declines with time, therefore it is most effective when administered before hepatic injury occurs, as signified by elevations of AST and ALT. However, NAC treatment may provide beneficial effects even after liver injury has occurred, presumably through its ability to improve tissue oxygen delivery and use.
The measurement of serum acetaminophen concentrations becomes paramount for proper assessment of the severity of overdose and for appropriate decision making for antidotal therapy. If serum acetaminophen results are not available locally within 8 hours of suspected ingestion, treatment with NAC should begin until levels are available. The Rumack-Matthew nomogram relates serum acetaminophen concentration and time following acute ingestion to the probability of hepatic necrosis ( Fig. 43.3 ).
Several qualifications pertain to the use of this nomogram. First, to utilize the nomogram, blood samples should not be obtained earlier than 4 hours after ingestion to ensure that absorption is complete. Second, the nomogram applies only to an acute single ingestion and not to chronic ingestion. Toxicity from chronic ingestion of acetaminophen or other drugs is cumulative and typically occurs at lower blood concentrations than in acute overdose. Third, the nomogram is not useful if the time of ingestion is unknown or is considered unreliable. In this case, when the exact time of ingestion is unknown, clinicians should err on the side of treating with NAC until the acetaminophen concentration is not detectable and no transaminase elevation is seen. Fourth, if acetaminophen is coingested with a substance that may delay absorption (i.e., anticholinergic), the patient should be clinically monitored for clinical effects. If, for example, no anticholinergic signs or symptoms develop after the 4-hour acetaminophen concentration is measured, one may assume that absorption will not be delayed and the concentration can be plotted normally. If, however, the patient develops anticholinergic signs and symptoms, and the acetaminophen concentration is detectable, that patient should be treated with NAC as absorption is most likely delayed, and the concentration should not be plotted. Alcoholic patients, fasting or malnourished patients, and patients on long-term therapy with microsomal enzyme-inducing drugs (e.g. phenobarbital) may have increased susceptibility to acetaminophen hepatotoxicity, presumably as a result of induction of cytochrome P450 (see later) and, in the case of alcoholics or fasting patients, depletion of glutathione (see later). In these cases, it has been proposed that the decision line in the nomogram should be lowered by 50 to 70%. , Others do not advocate any change in the therapeutic decision line for such patients with acute ingestion. These risk factors may be more important in chronic acetaminophen poisoning. Although therapeutic guidelines for chronic acetaminophen poisoning have not been established, it is recommended to administer NAC if the transaminases are elevated or the acetaminophen level is greater than10 μg/mL (66.2 μmol/L), until improvement is noted.
An area of some controversy is whether acetaminophen screening should be performed on all intentional overdose patients. One of the most worrisome aspects of acetaminophen poisoning is that initial clinical symptoms (e.g., nausea, vomiting, abdominal pain) may be vague or even absent in the first 24 hours. , This possible delay in diagnosis is particularly problematic because the antidote, NAC, has been shown to be most effective when initiated within the first 8 hours. Studies recommend screening all patients with suicidal ingestion and those with altered mental status in whom ingestion is suspected. ,
Many spectrophotometric methods are available for the determination of acetaminophen. , In general, these methods are relatively easy to perform but are subject to various interferences such as bilirubin or bilirubin byproducts absorbing at similar wavelengths. , Some methods measure the nontoxic metabolites and parent acetaminophen, and thus may produce especially misleading results. Therefore only methods specific for parent acetaminophen should be used. Immunoassays are widely used for this purpose, as they are rapid, easily performed, and accurate. A different spectrophotometric approach uses arylacylamide amidohydrolase to hydrolyze acetaminophen (but not conjugates) to p -aminophenol and acetate. Subsequent formation of the absorbing species depends on the reaction of generated p -aminophenol with 8-hydroxyquinoline or o-cresol. Arylacylamide amidohydrolase methods are susceptible to interference by NAC, bilirubin, and immunoglobulin (Ig)M monoclonal immunoglobulins. Most chromatographic methods are very accurate and are considered reference procedures. A qualitative, one-step lateral flow immunoassay (cut-off of 25 μg/mL) may be suitable for point-of-care application, yet it has a low positive predictive value.
Acetylsalicylic acid (aspirin) has analgesic, antipyretic, and anti-inflammatory properties. These therapeutic benefits derive from its ability to inhibit biosynthesis of prostaglandins by acetylation of active site serine and subsequent irreversible inhibition of cyclooxygenase enzymes (COX-1; COX-2 isoenzymes). Salicylate, the metabolite of aspirin, also reduces prostaglandin synthesis by uncertain mechanisms. Aspirin also interferes with platelet aggregation and thus prolongs bleeding time. The platelet inhibitory effect is a consequence of the ability of aspirin to acetylate and irreversibly inhibit platelet cyclooxygenase, thereby reducing the formation of thromboxane A 2 , a potent mediator of platelet aggregation. Platelets have little or no capacity for protein synthesis; therefore the duration of this enzyme inhibition is the normal life span of the platelets (8 to 11 days). Because of this platelet inhibitory activity, low-dose aspirin has been recommended as prophylactic therapy for some individuals at risk for thromboembolic disease. , An epidemiologic association has been noted between aspirin ingestion and Reye syndrome in children and adolescents with viral infection (e.g., varicella, influenza). Therefore aspirin use in these patients is done so cautiously. However, because of the therapeutic benefits and the overall lack of serious side effects at normal doses in most patients, aspirin is widely available and frequently consumed.
Absorption of normal doses of regular aspirin from the GI tract is generally rapid, with peak serum concentration achieved within an hour. This peak value may be delayed for 12 hours or longer for enteric-coated or slow-release formulations. Once absorbed, aspirin has a very short half-life (t ½ = 15 minutes) because of its rapid hydrolysis to salicylate. Salicylate is eliminated mainly by conjugation with glycine to form salicyluric acid, and to a lesser extent with glucuronic acid to form phenol and acyl glucuronides. A very small amount is hydroxylated to gentisic acid. These metabolic pathways may become saturated even at high therapeutic doses. Consequently, serum salicylate concentration may increase disproportionately with dosage. At high therapeutic or toxic doses, the salicylate elimination half-life is prolonged (15 minutes to 30 hours versus 2 to 3 hours at low dose) and a much larger portion of the dose is excreted in urine as salicylate. Therapeutic serum salicylate concentrations are generally lower than 60 mg/L (0.5 mmol/L) for analgesic-antipyretic effects, and 150 to 300 mg/L (1.0 to 2.0 mmol/L)for anti-inflammatory actions.
Salicylates directly stimulate the central respiratory center and thereby cause hyperventilation and respiratory alkalosis. Additionally, salicylates cause uncoupling of oxidative phosphorylation. As a result, heat production (hyperthermia), oxygen consumption, and metabolic rate may be increased. Salicylates also enhance anaerobic glycolysis but inhibit the Krebs cycle and transaminase enzymes, which leads to accumulation of organic acids and thus to metabolic acidosis. ,
The primary acid-base disturbance observed with salicylate overdosage depends on age and severity of intoxication. Respiratory alkalosis predominates in children over the age of 4 years and in adults, except in very severe cases that may progress through a mixed respiratory alkalosis–metabolic acidosis to metabolic acidosis. In children younger than age 4, the initial period of respiratory alkalosis may be brief and therefore may not be observed; in such cases, metabolic acidosis predominates. , CNS depression is more pronounced when acidemia is severe, which is a consequence of increased brain uptake of nonionized salicylic acid. Respiratory acidosis, a result of severe CNS depression or pulmonary edema, may sometimes occur and is indicative of a poor prognosis.
Following acute salicylate overdose, patients initially may be asymptomatic, especially if that product is enteric coated. Salicylate toxic patients may develop nausea, vomiting, abdominal pain, tinnitus, tachypnea, oliguria, and altered mental status ranging from agitation to lethargy to coma. Toxic doses of aspirin may form concretions or bezoars and produce pylorospasm, thereby delaying absorption. Serum salicylate in such instances may not reach maximum concentration for 6 hours or longer. , Therefore a concentration drawn soon after the original ingestion may not be reflective of the potential peak concentration—an important consideration when assessment of the severity of toxicity is based on such measurements. Use of salicylate concentrations to guide management must be done cautiously and only in conjunction with careful evaluation of a patient’s clinical status. The nomogram developed by Done is no longer recommend in the management of the salicylate poisoned patient. Toxic salicylate concentrations alone are of poor prognostic value; however, certain clinical findings predict a poor prognosis, including pulmonary edema, fever, coma, and acidosis. Initial serial concentrations should be performed every 2 hours while the patient is monitored clinically. When the concentrations begin to decline and the patient’s clinical status is improved, concentrations can be measured less frequently.
The need to screen all intentional overdose patients for salicylates is debated. , , Diagnosis of salicylate poisoning based solely on clinical examination is not without pitfalls. Although large, acute ingestions are usually detected through history and clinical symptoms, chronic salicylate toxicity often is more difficult to diagnose. Chronic intoxication can present in a similar fashion as acute exposures, yet such exposures typically are more insidious and therefore are often misdiagnosed. In these cases, patients present with nonspecific symptoms such as fever, abdominal pain, and encephalopathy and subsequently are misdiagnosed with surgical abdomen, myocardial infarction, sepsis, encephalitis, and alcoholic ketoacidosis. , Because products containing salicylates are readily available, the clinical effects of salicylate toxicity are nonspecific, and lack of metabolic acidosis does not rule out the potential for salicylate toxicity, clinicians should have a low threshold for obtaining serum salicylate concentrations. Some suggest that in any patient with a history of salicylate ingestion or possessing characteristic signs or symptoms of salicylate poisoning, a serum salicylate concentration should be obtained. Early identification of salicylate toxicity can be lifesaving.
Treatment for salicylate intoxication is directed toward (1) decreasing further absorption, (2) increasing elimination, and (3) correcting acid-base and electrolyte disturbances. Activated charcoal binds aspirin and prevents its absorption. Elimination of salicylate may be enhanced by alkaline diuresis and in severe cases by hemodialysis. Sodium bicarbonate may be given to alleviate metabolic acidosis and enhance elimination. Indications for emergent hemodialysis include serum salicylate greater than 1000 mg/L, CNS changes, intractable metabolic acidosis, pulmonary edema, hepatic failure with coagulopathy, and renal failure.
Classic methods for the measurement of salicylate in serum are based on the method of Trinder. These procedures rely on the reaction between salicylate and Fe 3+ to form a colored complex that is measured at 540 nm. To lessen endogenous background interference, a protein precipitation step or a serum blank is necessary. Nevertheless, blank readings equivalent to about 20 to 25 mg/L are generally observed. Moreover, interference by salicylate metabolites, endogenous compounds, and some drugs, especially structurally related drugs such as diflunisal (difluorophenyl salicylate), may occur. Azide, present as a preservative in some commercial control sera, also causes interference. Despite these limitations, photometric methods continue to be successfully used to assess salicylate overdose. The Trinder method results agreed very closely with those of a reference HPLC procedure. However, significant interference with the Trinder method was observed for one patient, who consumed an overdose of dichloralphenazone. Thus for best interpretation of test results, as much information as possible should be obtained regarding drug ingestion history.
Other methods for salicylate quantitation include fluorescent polarization immunoassay and salicylate hydroxylase–mediated photometric techniques. These procedures are subject to some of the same interferences as the Trinder method, but the salicylate hydroxylase method is considered more specific and has been adapted to automated analyzers. Gas and liquid chromatographic methods are the most specific methods for salicylate, , but their general availability, especially for emergency use, is limited. A qualitative, one-step lateral flow immunoassay (cut-off of 100 μg/mL) is commercially available for point-of-care application but has a low positive predictive value (0.47).
The units reported with each concentration should be documented before management decisions are made. Laboratories may alternatively report concentrations in terms of mg/dL and mg/L. This important distinction, which involves a ten-fold difference in concentration, is infamous for causing confusion. In extreme cases of these miscommunications, hemodialysis has been ordered for patients thought to have astronomically high salicylate concentrations that were later proven to be nontoxic.
The TCAs, the phenothiazines, and the antihistamines have divergent therapeutic applications; however in overdose, they often share similar anticholinergic and antihistaminic toxidromes as principal components of their overall toxic effects.
TCAs, so named because of their three-ring structure ( Fig. 43.4 ), represent a class of drugs frequently prescribed for the treatment of depression and neuropathic pain (see Chapter 42 ). The TCAs have been largely supplanted by the newer, less toxic selective serotonin reuptake inhibitors (SSRIs) and other atypical agents for depression, which now are accepted broadly as drugs of first choice, particularly for medically ill or potentially suicidal patients and for the elderly and the young. Fatalities are much less common since modern antidepressants have widely replaced these drugs. However, because of their continued use and narrow therapeutic range, TCAs are frequently associated with severe or fatal toxicity following overdose.
TCAs block neuronal uptake of serotonin and/or norepinephrine. TCAs have many other pharmacologic actions that apparently do not contribute to the therapeutic effects but do contribute to the side effects. TCAs have at least moderate affinity for blockade of α 1 -adrenergic receptors, much less for α 2 , and virtually none for β-receptors, leading to vasodilation and hypotension. TCAs have sedative effects and the potential for seizures related to antihistamine activity. TCAs exert central and peripheral anticholinergic effects (dry skin and mouth, flushing, hyperpyrexia, dilated pupils, constipation, urinary retention, and decreased GI motility) through their blockade of M1 muscarinic receptors. TCAs close GABA channels resulting in an increased risk of seizures.
Cardiovascular effects, the most serious manifestation of TCA overdose, accounts for the majority of fatalities. Several mechanisms may contribute to cardiovascular toxicity :
Anticholinergic effects and inhibition of neuronal reuptake of catecholamines result in tachycardia and potential for early hypertension.
Peripheral α-adrenergic blockade causes vasodilation and contributes to hypotension.
Cardiac fast sodium channel blockade and potassium efflux channel blockade resulting in QRS and QT prolongation, respectively, and risk for decreased cardiac output and ventricular dysrhythmias.
Metabolic and/or respiratory acidosis that may further contribute to inhibiting the cardiac fast sodium channel.
Some tolerance to the sedative and autonomic effects of TCAs tends to develop with continued drug use. Occasionally, patients show physical dependence, with malaise, chills, muscle aches, and sleep disturbance following abrupt discontinuation, particularly of high doses. Some withdrawal effects may reflect increased cholinergic activity following its inhibition. Some of these reactions have been confused with clinical worsening of depressive symptoms. Emergence of agitated or manic reactions has been observed after abrupt discontinuation of TCAs.
In general, antidepressants are associated with several clinically important drug interactions (e.g., serotonin syndrome), and they potentiate the effects of alcohol and probably other sedatives. TCAs are oxidized by hepatic cytochrome P450 (CYP) microsomal enzymes, followed by conjugation with glucuronic acid. The N -demethylated metabolites of several tricyclic antidepressants are pharmacologically active and may accumulate in concentrations approaching or exceeding those of the parent drug, to contribute variably to overall pharmacodynamic activity.
Tricyclic antidepressants are measured by chromatographic or immunoassay methods. Immunoassays are rapid and relatively easy to perform but may be subject to interference by other drugs, such as chlorpromazine, thioridazine, cyclobenzaprine, and diphenhydramine, and are not able to identify which TCA is being quantitated. In cases of overdose, qualitative identification (serum or urine) is sufficient, because the severity of intoxication is more reliably indicated by an increase in the QRS interval than by the serum drug concentration.
Cyclobenzaprine, a tricyclic amine structurally very similar to amitriptyline (see Fig. 43.4 ), is used as a centrally acting skeletal muscle relaxant. Similar to amitriptyline, cyclobenzaprine causes sedation, produces central and peripheral muscarinic blockade, and potentiates adrenergic actions. In overdose, cyclobenzaprine may cause a typical anticholinergic toxidrome and cardiac arrhythmias, hypotension, and coma. The analytical distinction between amitriptyline and cyclobenzaprine is often difficult. Cyclobenzaprine cross-reacts with immunoassays for tricyclic antidepressants and can co-elute or co-migrate with amitriptyline in HPLC and TLC. However, cyclobenzaprine and amitriptyline have different ultraviolet spectra; therefore they may be distinguished by HPLC using a diode array detector by multi-wavelength scanning or dual-wavelength discrimination. , However, amitriptyline and cyclobenzaprine are well resolved using capillary column GC- or LC-MS/MS and may be distinguished by careful examination of their respective mass spectra.
The antipsychotic drugs are generally used for primary psychiatric disorders, for example (1) schizophrenia, (2) bipolar disorder, (3) schizoaffective disorder, and (4) psychotic depression. In addition to their psychotherapeutic effects, these drugs have a number of other actions, so that certain members of this group are used as antiemetics (prochlorperazine), as antihistaminics (promethazine), and for sedation or potentiation of analgesia and general anesthesia. Antipsychotic compounds are traditionally divided and subdivided according to their chemical structure ( Table 43.6 and Fig. 43.5 ). ,
Antipsychotics | Examples , , , |
---|---|
Classical Antipsychotics | |
Phenothiazines |
|
Thioxanthenes |
|
Dibenzoxazepine | Loxapine |
Dihydroindoles | Molindone |
Butyrophenones |
|
Diphenylbutylpiperidines | Pimozide |
Benzamides | Sulpiride |
Atypical Antipsychotics | |
Dibenzodiazepine derivatives |
|
Benzothiazepine derivatives |
|
Benzisoxazole derivatives | Risperidone |
Benzisothiazol piperazine | Ziprasidone |
Imidazolinone derivatives | Sertindole |
The primary pharmacologic effect of all antipsychotic drugs is thought to be blockade of D2 receptors in the CNS. , It has been shown that classical antipsychotics reduce psychotic symptoms and that this correlates with their affinity for the D2 receptor. However, the drugs bind to many other receptors, including histamine (H 1 , H 2 ), GABA A , muscarinic receptors, α 1 - and α 2 -adrenoreceptors, and sodium and potassium voltage-gated ion channels. The atypical antipsychotics on the other hand have a different mechanism that my involve other dopamine receptors, serotonin receptors, or both.
The principal manifestations of phenothiazine toxicity involve the CNS and the cardiovascular system. , The most common effects in significant phenothiazine overdose include (1) sedation, (2) hypotension, (3) anticholinergic effects, and (4) ECG changes. Phenothiazines are relatively safe and rarely cause death when ingested alone.
All of the neuroleptic drugs are metabolized in the liver. Many have active metabolites and complex metabolic pathways. The main enzymes involved in metabolism are cytochrome P450 (CYP) enzymes, specifically CYP1A2, CYP2D6, and CYP3A4. , , Many sources of variation are found in CYP-mediated metabolism; however, where multiple enzymes are involved, such variability has only a relatively small effect on clearance and drug concentrations.
Toxicity is strongly correlated with peak serum concentrations and thus usually occurs within the first 4 to 6 hours after ingestion of these rapidly absorbed drugs. Neuroleptics may be detected by chromatographic methods. GC chromatographic methods are used to measure antipsychotic drugs, and NP, EC, and MS detectors are the detection systems of choice. HPLC, LC-MS, and LC-MS/MS and TOF methods are also utilized.
Become a Clinical Tree membership for Full access and enjoy Unlimited articles
If you are a member. Log in here