Vous êtes sur la page 1sur 32

Clinical Pharmacokinetics 6: 161-192 (1981)

0312-5963/81/0005-0161/$08.00/0
© ADIS Press Australasia Pty Ltd. All rights reserved.

Pharmacokinetics of Drug Overdose

Jon Rosenberg, Neal L. Benowitz and Susan Pond


Clinical Pharmacology Unit of the Medical Service, San Francisco General Hospital Medical
Center, and Departments of Medicine and Psychiatry, University of California, San Francisco,
California

Summary Pharmacokinetics of drugs taken in overdose may differ from those observed following
therapeutic doses. Differences are due both to dose-dependent changes and to effects of drugs or
pathophysiological consequences of the overdose on kinetics. Dose-dependent changes in rate
and extent of absorption, bioavailability (saturation of first-pass metabolism), distribution
(saturation of protein binding sites) and metabolism are discussed. Gastrointestinal motility is
affected both by specific drug actions, such as delayed gastric emptying by anticholinergic
drugs, and by general nervous system depression caused by many drugs. Drug-induced cir-
culatory insufficiency may retard tissue distribution and reduce clearance. Disturbances in
blood and urine pH may alter distribution and clearance of weak acids and bases. Drug-in-
duced renal or hepatiC failure can significantly decrease clearance. Hypothermia is a common
complication of drug overdose and might retard distribution and also reduce clearance.
The data concerning pharmacokinetics during overdose are usually incomplete and difficult
to interpret. Doses and times of ingestion are uncertain, duration of blood and urine sampling is
often inadequate to distinguish absorption from distribution and elimination phases, active
metabolites are not measured, protein binding is not determined and clinical features of
patients not adequately described. We have, however, reviewed available data for salicylate,
paracetamol (acetaminophen), barbiturates, ethchlorvynol, glutethimide, chloral hydrate, tri-
cyclic antidepressants, lithium, phenytoin, ethanol, theophylline, digoxin, amphetamine and
phencyclidine.

An understanding of the pharmacokinetics of selection of methods to accelerate drug removal or


drugs taken in overdose could contribute to rational allow decisions to treat patients prophylactically for
medical management of many patients admitted to anticipated toxicity. The published data on phar-
hospitals each year after suicidal or accidental drug macokinetics after an overdose are often difficult to
ingestions. For example, blood concentration data interpret for the following reasons: dose and time of
might be used to predict the time course of coma or drug ingestion are rarely known reliably; a prolonged
other pharmacological or toxic effects. Similarly, absorption phase and relatively brief period of blood
pharmacokinetic data may facilitate evaluation and sampling in most patients make it difficult to dis-
Pharmacokinetics of Drug Overdose 162

tinguish continuing absorption from elimination; the on drug solubility and the amount of time the drug
identity, amounts, and concentrations of other drugs remains in the small intestine.
ingested are often not known or stated; and the pre- An overdose of a drug commonly delays its ab-
sence and potential effects of specific organ toxicities sorption, which may in turn delay the onset or peak
or disease states and their physiological effects on intensity of the drug's action. Mechanisms respons-
pharmacokinetics are commonly not evaluated. These ible for delayed drug absorption include formation of
factors may account for the often poor correlation be- a poorly soluble mass of drug, and a decrease in gas-
tween the clinical findings in overdose patients and trointestinal motility, the latter which will be dis-
the blood concentrations of ingested drugs. Other fac- cussed later (section 1.2. J). On occasion, bezoar for-
tors could also contribute to this poor correlation mation has necessitated operative removal of the drug
such as differences among subjects in their responses mass, either because of continuing drug absorption or
to various drug concentrations in blood or tissue. because of mechanical obstruction (Schwartz~ i 976).
However, careful studies of the time course of blood Theoretically, the percentage of drug reaching the
concentrations of drugs during overdose, identifica- systemic circulation after an overdose may be greater
tion and quantitation of active metabolites, measure- than after a therapeutic dose. This may occur because
ment of protein binding, and more careful recording of saturation of presystemic or first-pass metabolism
of clinical findings may allow better correlation of of a drug that normally exhibits significant first-pass
these findings with pharmacokinetic data. metabolism. Such drugs will include many of the nar-
The pharmacokinetics of most drugs taken in cotic analgesics, tricyclic antidepressants, phenothia-
overdose are often assumed to be similar to those ob- zines and ~-adrenoceptor antagonists. Even within
served after therapeutic doses, but may differ consi- the therapeutic dosage range, propranolol (Walle et
derably. Evidence that such differences occur and aI., 1978) appears to display dose-dependent increases
possible mechanisms for the differences are subjects in bioavailability.
of this review. Initial discussion will centre around Individual examples in which drug absorption ap-
the phases of drug absorption, distribution, and pears to be altered because of ingestion of an overdose
elimination and the ways that these processes may be are discussed in the section on specific drugs. How-
altered after drug overdose. The second part of the ever, even when reported, data on drug absorption in
review deals with specific drugs for which there are overdose patients are difficult to evaluate because of
reasonable data about the pharmacokinetics after an unreliable histories of the quantities of the drug in-
overdose. gested and because of intervening measures such as
emesis, lavage, or administration of charcoal or
cathartics.
1. Effects of Overdose on Pharmacokinetic
Processes J . J.2 Distribution
Drug distribution depends on route of administra-
1.1 Dose-dependent Kinetics tion, plasma and tissue protein binding, blood flow,
diffusion rate, and extent of partitioning to tissues
J . J. J Absorption and Bioavailability (Rowland, 1978). An alteration in drug distribution
The small intestine is the principal site of drug ab- with increasing drug concentration can have impor-
sorption. In general, the rate of drug absorption de- tant implications after an overdose. Even when the
pends on the rate of dissolution of the drug and the plasma concentration of drug is within the thera-
rate of gastric emptying, which may determine transit peutic range, such drugs as salicylate, phenylbutazone
time into the small intestine (Rowland, 1978). The (Aarbakke, 1978), naproxen (Runkel et al., 1976),
eventual amount of drug absorbed depends primarily and disopyramide (Cunningham et al., 1977) may
Pharmacokinetics of Drug Overdose 163

saturate plasma protein binding sites. Therefore, as lithium, which will be discussed in detail later. Also
blood concentration increases, the fraction of the drug discussed later will be the effect of haemodynamic,
that is unbound also increases. It is likely, although acid-base, and other pathophysiological phenomena
not well studied, that these and other drugs, par- on distribution of drug into tissues after drug over-
ticularly those that are highly protein-bound, will dose.
similarly saturate plasma protein binding sites and be
proportionately less bound after an overdose. Assum- 1.1.3 Elimination
ing no change in the drug binding to tissues, the Generally, processes in pharmacokinetics are
volume of distribution of a drug then increases. assumed to be first-order, so that the rate of elimina-
Because of variability in protein binding among sub- tion of the drug is directly proportional to the drug
jects, it may be difficult to relate total blood con- concentration in the plasma. The proportionality fac-
centration of drug to clinical effects. The concentra- tor for the elimination process is clearance, which is
tion of free drug might be more closely related to the parameter that relates the elimination rate to con-
clinical effects. centration of the drug. However, as virtually all drug
Estimations of drug clearance based on total blood metabolism and secretory pathways involve enzyme-
or plasma concentration of drug may be complicated catalysed or carrier-mediated systems, the capacity of
by the presence of concentration-dependent protein these systems may limit the rate of metabolism or
binding. For example, for a drug whose clearance of elimination. This has been shown after usual doses of
unbound drug is constant, a concentration-dependent ethanol (Lundquist and Wolthers, 1958) and after
decrease in protein binding will result in a relative therapeutic doses of salicylate (Levy et al., 1972) and
decrease in total blood concentration. Therefore, phenytoin (Arnold and Gerber, 1970). If biotransfor-
clearance calculated from total blood concentration of mation or elimination proceeds via a single pathway,
drug would increase. then the rate of elimination may be described by the
In the absence of changes in distribution, changes Michaelis-Menten equation (Levy et al., 1972;
in plasma concentration of drug usually reflect Wagner, 1971):
changes in the amount of drug in the body. However,
in the presence of concentration-dependent protein -dC Vm·C
(Eq. 1)
binding, changes in plasma concentration of drug dt Km + C
may be disproportionate to changes in the amount of
drug in the body, because distribution changes with where - dC/dt is the rate of decline of drug con-
concentration. Estimation of drug half-life becomes centration, C; V m is the theoretical maximum rate
less informative in the presence of dose-dependent when the enzyme or carrier is saturated; and Km is
protein binding. Half-life depends both on volume of the Michaelis constant. Km reflects the affinity of the
distribution and clearance, and reported differences in enzyme or carrier protein for the substrate.
half-life in overdose patients may reflect an alteration For low plasma concentrations (C« Km),
in one or both of these parameters. elimination proceeds as a first-order process because
Usually, the rate of diffusion into tissues is rapid equation 1 reduces to:
relative to the rates of its absorption and elimination.
Therefore, clinical effects of the drug parallel the rate -dC Vm·C
at which the drug enters the systemic circulation. (Eq.2)
dt Km
However, diffusion of a drug into tissues may be slow
and result in a significant lag time between peak Increasing doses and, thereby, increasing plasma
blood concentrations of drug and peak clinical mani- concentrations of drug, will ultimately lead to a con-
festations. One drug with these characteristics is centration-dependent elimination rate. Once C is
Pharmacokinetics of Drug Overdose 164

much greater than Km, elimination proceeds as a concentration-dependent kinetics exhibited by these
zero-order process as indicated: drugs is that the relative contribution of methods of
-dC extracorporeal drug removal to total body clearance
- - = Vm (Eq. 3) would increase as the drug concentration increases.
dt
This is so because endogenous drug clearance would
In this case, elimination occurs at a constant rate decrease as drug concentration increased. Further-
and the elimination is independent of drug concentra- more, as already mentioned, drug action may be
tion. The time courses of elimination of 3 different greatly prolonged because of an associated prolonged
doses of salicylate (fig. 1) illustrate the 3 general drug 'half-life'. A further implication of concentra-
kinetic situations. Elimination of the lowest dose tion-dependent kinetics would be that the area under
follows first-order kinetics. Conversely, the highest the plasma concentration-time curve after an oral
dose yields initial concentrations considerably abOVe dose may increase uisproportionately whh increasing
Km so that the concentration of drug declines initially dose. At sufficiently high dosage levels, the area
at a constant rate. The time required for the initial would be proportional to the square of the dose rather
concentration of drug to decrease by 50 % is depen-
dent on concentration and increases as concentration
increases.
Mechanisms of apparent saturation at sites of drug
metabolism or excretion may differ for different 4000-
drugs. Possible mechanisms include simple saturation

I ...·...·....
of the enzyme or carrier, inhibition of enzyme activity 2000L
by the drug, decrease in enzyme affinity or activity
-
due to a metabolic product of the drug, exhaustion of E100000 ' ..
co factors required for enzyme activity, and partial in- E 1 . . .·, .".
activation or destruction of enzyme or carrier. The
latter is illustrated by the self-catalysed destruction by
~
.~
500 -
I .".\ "'-"
., '. - Beginning of
first-order kinetics

drugs of the cytochrome P-450 haem, the terminal


electron acceptor in the hepatic microsomal mono- :1 2°O'?1' 0\ \.. ~,,,
"i • '/ \.

; ':1
oxygenase system (DeMatteis, 1970; Ivanetich et a!., .y

1978). Destruction is probably due to a metabolite


formed from the parent drug by metabolism in the ~~, ~,;~e".,
mono-oxygenase system. Some of the commonly

I- t .b- t~/?l- , t - , - , -~
used barbiturates have been shown to destroy P-450 <t: 20- It)

in this way (Levin et al., 1972). •


The drugs that have actually been demonstrated to ,-,-,
4 8 12 16 20 24 28
exhibit concentration-dependent pharmacokinetics in Time (h)
the overdosed patient include chloral hydrate/
trichloroethanol (Stalker et aI., 1978), ethchlorvynol
(Benowitz et aI., 1980), some barbiturates (Forrest et Fig. 1. Dose-dependency of salicylate elimination kinetics
a!., 1974; Prescott et al., 1973), theophylline (Kadlec in 1 subject.
Amount unexcreted estimated from initial dose minus
et aI., 1978), paracetamol/acetaminophen (Slattery
cumulative urinary excretion of salicylate and metabolites.
and Levy, 1979), and perhaps glutethimide (Maher, Vertical arrows indicate time after excretion at which 50% of
1970). These drugs are discussed in more detail in dose has been eliminated (from Levy: J. Pharm. Sci. 54: 959,
later sections of this review. One implication of the 1965; reproduced by permission of author and editor).
Pharmacokinetics of Drug Overdose 165

than to the dose itself (Gibaldi and Perrier, 1975). an overdose. Furthermore, although not definitely
Furthermore, the area under the curve after ad- proven to be effective in drug overdose, cathartics,
ministration of a fixed dose of the drug showing con- because of their effect in increasing gastrointestinal
centration-dependent elimination may also vary with motility, could decrease absorption of poorly soluble
the rate of absorption. The area under the plasma drugs. Finally, one drug may alter the absorption of
concentration-time curve would be in this case larger another, and toxicity of a drug taken in a mixed over-
the more rapidly the dose is absorbed. dose might thus be altered by the second drug. For ex-
Finally, under certain circumstances, drug elim- ample, after therapeutic doses of propoxyphene, the
ination may appear to be first-order but drug half-life absorption of concurrently ingested paracetamol
may increase with increasing dose. This may occur (acetaminophen) is slowed (Nimmo et a1., 1979). Pro-
either because of drug elimination by parallel bably propoxyphene delays gastric emptying and
capacity-limited and first-order processes, or because hence delivery of paracetamol to the small intestine.
of inhibition of metabolism of the parent drug by a Theoretically, decreasing the rate of absorption of
product (Gibaldi and Perrier, 1975). paracetamol may mitigate against some of the
hepatotoxic effects of the paracetamol that are related
in part to the rate of generation of a toxic metabolite.
1.2 Pathophysiology of Drug Overdose
1.2.2 Cardiovascular and Pulmonary Function
1.2.1 Gastrointestinal Motility Drug overdose is often complicated by com-
Many drugs taken in overdose decrease gastroin- promised cardiovascular and pulmonary function
testinal motility (Nimmo, 1976). The list includes that can in turn influence drug kinetics (Benowitz and
drugs with specific anticholinergic activity, those that Meister, 1976; Benowitz et al., 1979). Most com-
directly affect gastrointestinal smooth muscle, and monly, overdose patients develop hypotension due to
those that generally depress autonomic nervous a decrease in cardiac output and/ or vascular resis-
system activity. Conversely, the organophosphate in- tance. After a severe overdose of drugs such as tri-
secticides and drugs used to treat myasthenia gravis cyclic antidepressants, frank cardiogenic Shock may
that inhibit acetylcholinesterase may result in develop. However, drug overdosed patients may not
enhanced gastrointestinal motility. exhibit the increased activity of the sympathetic ner-
A decrease in gastrointestinal motility may both vous system which usually occurs in patients with
decrease the rate and increase the amount of drug ab- hypotension due to primary myocardial depression.
sorbed, or lead to prolonged, erratic, and delayed ab- Rather, the drugs may suppress sympathetic nervous
sorption. If a drug is poorly water-soluble, a decrease system activity which in part accounts for the
in gastrointestinal motility may increase the eventual decreased vascular resistance that accompanies many
amount of drug absorbed because of the length of drug overdoses. Occasionally, anticholinergic or sym-
time that the drug has to dissolve. Rate of dissolution pathomimetic drug overdose may lead to hyperten-
is often the limiting factor in drug absorption, sion with or without increased cardiac output.
especially of poorly water-soluble drugs such as Furthermore, noncardiogenic pulmonary oedema fre-
digoxin and phenytoin. It is not surprising, therefore, quently accompanies severe drug overdose, such as
that many patients continue to manifest drug toxicity with narcotics and salicylates. The oedema may
many days after a single ingestion of a drug, worsen hypoxaemia and acidosis and add to the
presumably because of persistence of the drug in the drug's depressant effect on the cardiovascular system.
small intestine with continuing absorption. Thus, the Each of these pathophysiological changes may
use of lavage and administration of activated charcoal alter pharmacokinetics. However, often the precise
may be indicated for as long as 12 to 24 hours after effects cannot be examined easily in overdose patients
Pharmacokinetics of Drug Overdose 166

because of the rapidly changing clinical status. may not be based on the effect of alkalaemia on the
Therefore, we might consider the effects of more partitioning of drug between drug and the myocar-
stable altered haemodynamic states, such as cardiac dium.
failure, on pharmacokinetics (Benowitz and Meister, Alteration of urinary pH by a pathophysiological
1976). Cardiac failure substantially decreases the process or by therapeutic intervention may alter renal
volume of distribution and clearance of some drugs. clearance of some drugs. If renal clearance can be in-
A decrease in cardiac output decreases perfusion of creased to a sufficient fraction of total body clearance
organs, such as skeletal muscle, that show major up- to alter pharmacokinetics, then an alteration of urin-
take of drugs. Conversely, autonomic reflex adjust- ary pH can be used as a therapeutic intervention to
ments direct a greater fraction of cardiac output and hasten drug elimination after an overdose. The renal
hence drug to critical organs such as brain and heart. clearance of saJicylate is increased in an alkaline
Reduced hepatic and renal blood flow and! Oi tiSSUe urine. and in uverdused patient" the eiirnination ha!f-
hypoxia also decrease hepatic and renal drug life can be shortened significantly (Morgan and Polak,
clearance. Thus, in the overdose patient with a low 197 \). Similarly, alkalinisation of the urine increases
cardiac output, we would predict that blood con- the renal clearance of phenobarbitone significantly
centrations would be higher and more persistent than (Waddell and Butler, 1957; Hadden et ai., 1969). The
expected from the dose, due both to diminished tissue renal elimination of the basic drug amphetamine IS
distribution and drug clearance. sensitive to changes in urinary pH. Clearance in-
creases and plasma half-life decreases with urine
1.2.3 Acid-base Disturbances acidification (Davis et ai., 197 \). Patients with
Alterations in acid-base balance, whether drug-in- amphetamine psychosis treated by acidifying the
duced or not, may alter drug pharmacokinetics and urine had shorter half-lives of amphetamine and
toxicity. For example, blood pH may alter distribu- shorter duration of symptoms than those treated with
tion of drug into organs such as the brain, as illus- alkalinising the urine (Anggard et aI., 1973). In con-
trated by salicylate. A small decrease in blood pH trast, the contribution of renal clearance to the total
below 7.4 can result in a large increase in the propor- body clearance of phencyclidine is unknown, and the
tion of salicylate that is non-ionised and available to role of urinary acidification as proposed by Aronow
diffuse into the brain. Both in salicylate-intoxicated et al. (I 978) for the treatment of patients overdosed
animals (Hill, 1973) and in patients (Gabow et aI., with phencyclidine is uncertain.
1978), salicylate toxicity correlates with extent of
acidaemia. Waddell and Butler (\957) demonstrated 1.2.4 Renal Function
in dogs that lowering blood pH increased brain levels Although a significant percentage of cases of acute
and anaesthetic effects of phenobarbitone. Acid-base renal failure is reported to be caused by nephrotoxins
status has also been reported to affect the course of (Maher, 1976), drug overdose per se is not often com-
tricyclic antidepressant overdose (Brown, I 976a). In plicated by acute renal failure. The substances most
experimental animals and in tricyclic antidepressant often implicated are drugs with nephrotoxicity at ap-
drug-overdosed patients, cardiac arrhythmias have proximately therapeutic levels (e.g. antibiotics such as
been reported to be reduced by administration of aminoglycosides) or unusual poisons (e.g. ethylene
alkali to maintain blood pH equal to or greater than glycol, carbon tetrachloride, inorganic mercury,
7.4 (Brown, I 976a,b). Some investigators recom- Amanita phalloides mushroom). Renal failure may be
mend infusion of sodium bicarbonate until patients induced through a variety of mechanisms, which we
become alkalaemic up to a pH of 7.5. However, why will review briefly. Pharmacokinetics in renal disease
an alkaline pH should mitigate against the toxicity of were recently reviewed by Levy (I977) and in this
tricyclic antidepressants is unclear. The mechanism journal by Fabre and colleagues (Fabre and Balant,
Pharmacokinetics of Drug Overdose 167

1976; Fabre et a!., 1980) and will not be discussed renal complication in these patients (Cunningham et
here. a!., 1980). Snake and spider venoms can produce
Drugs producing acute proximal tubular necrosis renal failure because of haemolysis, haemoglobinuria,
include antibiotics, particularly aminoglycosides, and shock (Russell et aI., 1975).
halogenated hydrocarbon solvents such as carbon Although abrupt decreases in renal function have
tetrachloride and tetrachloroethylene, metals includ- been associated with the administration of aspirin and
ing mercury, arsenic and iron, mushrooms and other prostaglandin inhibitors (Kimberly et a!.,
various snake venoms (Maher, 1976). Paracetamol 1978), acute renal failure is uncommon in patients
overdose may result in acute hepatic necrosis as well with salicylate overdose. However, evidence of neph-
as acute renal failure, but the former usually pre- rotoxicity such as celluria and proteinuria is usually
dominates (Rumack and Matthew, 1975). observed (Temple, 1978).
The deposition of crystals may lead to intrarenal Accidental or suicidal ingestion of inorganic mer-
tubular obstruction and tubular necrosis. Most curic salts frequently causes acute renal failure due to
patients surviving the acute toxicity of ethylene glycol proximal tubular necrosis (Maher, 1976). Renal func-
poisoning develop renal failure typical of acute tubu- tion is particularly important with respect to treat-
lar necrosis (Parry and Wallach, 1974). It has been ment of mercury poisoning because the chelating an-
assumed that oxalate crystal deposition in the renal tidote, dimercaprol (BAL), is eliminated by the kidney
tubules causes acute tubular necrosis, although this and may accumulate with renal failure. Accumula-
has been questioned recently. Ethylene glycol itself is tion of BAL or the mercury-BAL complex may be
relatively non-toxic, so it appears that metabolism is nephrotoxic as well (Maher, 1976). Renal insufficien-
necessary for nephrotoxicity. The first step in its cy was a common complication of lithium overdose
metabolism to oxalate is mediated by alcohol de- in one series of patients (Schou et ai., 1968), as dis-
hydrogenase. Administration of ethanol, which com- cussed in section 2.5, although acute renal failure se-
petes for this enzyme, decreases the rate of metabolite condary to lithium intoxication has been infrequently
production and may be of value in prevention of acute reported (Lavender et aI., 1973). Because the
tubular necrosis (Parry and Wallach, 1974). Ethanol clearance of lithium is primarily renal, renal failure
should be started within 4 to 6 hours of ingestion, may alter the course of lithium intoxication.
because ethylene glycol is metabolised rapidly and has
a short half-life (3 hours in non-human primates [Par- 1.2.5 Hepatic Function
ry and Wallach, 1974]). Single doses of a few drugs or toxins may lead to
A number of drugs are associated with rhab- acute hepatic necrosis. Important examples include
domyolysis, myoglobinuria, and acute renal failure. paracetamol (Proudfoot and Wright, 1970), iron
These include stimulant drugs such as amphetamines (Gleason et a!., 1979) and Amanita phal/oides
or phencyclidine, narcotics, sedatives, and drugs that mushrooms (Becker et a!., 1976). The presence of
produce seizures such as tricyclic antidepressants acute or pre-existing chronic liver disease in an over-
(Cogen et a!., 1978; Eneas et a!., 1979; Komer et aI., dosed patient has obvious pharmacokinetic implica-
1976). Treatment of myoglobinuria should be a con- tions for many drugs. However, precise details of the
sideration in the treatment of overdose with these alterations in pharmacokinetics by liver disease will
drugs. The necrotising angiitis reported to be associ- not be discussed here because of the many reviews
ated with intravenous stimulant abuse may also cause and reports already available (Schenker et a!., 1975;
acute renal failure (Citron et a!., 1970). Although Blaschke, 1977). In contrast, chronic ingestion of
rhabdomyolysis and acute renal failure have been drugs such as barbiturates or phenytoin may enhance
reported occasionally in heroin users, a chronic the metabolism of other drugs by the hepatic micro-
sclerosing glomerulonephritis is the more common somal mono-oxygenase system. Enhanced metabol-
Pharmacokinetics of Drug Overdose 168

ism of a second drug may be important if the drug is pothermia by causing peripheral vasodilatation, a
metabolised to toxic intermediates. An example phenomenon observed after most types of sedative-
would be the combination of hepatic microsomal hypnotic overdose and after ingestion of sym-
enzyme-inducing drugs and paracetamol. In animals, patholytic and cholinomimetic drugs.
phenobarbitone increases the production of the toxic Hypothermia could alter drug disposition. Blood
metabolite of paracetamol and hence hepatic necrosis flow to and motility of the gastrointestinal tract are
(Mitchell et aI., 1973). A retrospective study of reduced by hypothermia, but effects on drug absorp-
patients with paracetamol overdose suggested greater tion have not been studied. Hypothermia is associated
susceptibility to the hepatotoxic effects of paracetamol with haemodynamic changes that could affect drug
in patients taking anticonvulsant medication (Wright distribution. Most studies have been performed dur-
and Prescott, 1973). ing external cooling or hypothermic cardiopulmonary
Apart from direct hep~Tic injury or lIiterlltion of bypass and not dmine drl1e-indl1ce(i hypothermia.
drug-metabolising enzymes, drug pharmacokinetics These two types of hypothermia may lead to signifi-
may be altered by a reduction in hepatic blood flow. cantly different haemodynamic changes, particularly
Most often, hepatic blood flow will be reduced in regard to sympathetic nervous system activity. For
because of the hypotension associated with many example, immersion of human volunteers in water at
drug overdoses. In turn, reduced hepatic blood flow 10°C is associated with an approximately 3-fold in-
may decrease systemic clearance and presystemic crease in plasma noradrenaline (norepinephrine) con-
elimination of drugs that undergo extensive first-pass centration and a similar increase in metabolic rate
biotransformation in the liver. Thus, the bio- (Johnson et aI., 1977). In contrast, an overdose of
availability of these drugs may be greater than antici- sedative-hypnotic or narcotic drugs decreases meta-
pated leading to higher blood concentrations of drug bolic rate and probably does not increase peripheral
than expected. Drugs that may exhibit this type of vascular resistance to the same extent as external
pharmacokinetic behaviour include lignocaine (lido- cooling (Shubin and Weil, 1965).
caine), some ~-adrenoceptor antagonists, opiate anal- In various animal studies, external cooling
gesics, and the tricyclic antidepressants. The relation- resulted in increased total peripheral resistance and
ships between drug kinetics and hepatic blood flow decreased regional blood flow, in particular to brain
have been reviewed recently by George (1979) and and kidney (Black et aI., 1976; Popovic and Popovic,
the pharmacokinetics of drugs in abnormal haemo- 1974). Heart rate and cardiac output decreased in pro-
dynamic states by Benowitz and Meister (1976). portion to the decrease in myocardial oxygen con-
sumption. There are few studies of drug distribution
J.2.6 Hypothermia during hypothermia. McAllister et al. (1979) com-
Hypothermia, or a body temperature below 35°C, pared the kinetics of propranolol in anaesthetised
is a common complication of drug overdose. Mechan- dogs maintained at 37°C to those maintained at 26°C
isms include decreased heat production, increased using a cold water bath. The apparent volume of dis-
heat loss, and exposure to low temperature (Popovic tribution (Vd~) was reduced by more than 3-fold by
and Popovic, 1974). Sedative-hypnotic and narcotic hypothermia, presumably as a result of peripheral
drugs may decrease metabolic rate. Hypothermia vasoconstriction. In an accompanying study of
after overdose with these drugs reflects diminished patients on chronic oral propranolol therapy undergo-
heat production. These drugs also increase heat loss ing coronary artery bypass surgery using hypother-
by inhibiting shivering. Even in therapeutic doses, mic cardiopulmonary bypass, plasma propranolol
phenothiazines produce poikilothermia, which, concentrations, corrected for haemodilution, rose
coupled with exposure to a cool environment, may slightly during hypothermia. The investigators at-
result in hypothermia. Drugs can also produce hy- tributed this change to a decrease in volume of dis-
Pharmacokinetics of Drug Overdose 169

tribution, but because plasma volume was not effects of digoxin on the heart; the amount of digoxin
measured after cooling, the validity of the haemodilu- necessary to produce ventricular fibrillation in hy-
tion correction is open to quest~on. Nurmand et al. pothermic dogs (24 to 28°C) was twice that in nor-
(\965) reported decreased distribution of thiopentone mothermic dogs (Beyda et aI., 1961). It is of interest
(thiopental), but not amylobarbitone (amobarbital), that digitalis drugs may reverse the effects of hy-
into fatty tissue during hypothermia and unchanged pothermia on cardiac conduction (Nahum and
distribution of both drugs into other organs. Phillips, 1959). The magnitude and duration of action
Hypothermia may decrease the clearance of drugs, of suxamethonium (succinylcholine) increased in cats
particularly by liver metabolism, as blood flow to the whose body temperatures were lowered from 36 to
liver is decreased as cardiac output decreases. 30°C (Wislicki, 1960). This may be due at least in
Furthermore, intrahepatic distribution of blood may part to a decrease in the rate of hydrolysis of suxa-
also be altered, and the metabolic rate of hepatic cells, methonium by plasma cholinesterase.
as reflected by oxygen uptake, diminished by hy- In summary, hypothermia may result in altera-
pothermia (Krarup and Larsen, 1972). The rates of tions of the disposition and pharmacodynamics of
metabolism of many drugs by hepatic microsomes drugs, either taken in overdose or administered in
are slowed as temperatures are decreased below 37°C therapeutic doses to hypothermic patients. These po-
(Fuhrman et aI., 1944; Kaiser et aI., 1968; McAllister tential alterations should be considered in the treat-
et aI., 1978; Rink et aI., 1956). Studies in vivo ment of these patients.
demonstrated that the clearance of propranolol in
dogs cooled to 26°C is reduced to 50 % of that at
37°C. Similarly, a fall in body temperature of I to 2. Pharmacokinetics of Specffic Drugs
2°C in cats is associated with a 26 % decrease in rate During Overdose
of elimination of ethanol (Krarup and Larsen, 1972).
Renal drug clearance may be altered by hypother- 2.1 Salicylate
mia. The activity of energy requiring metabolic pro-
cesses such as active tubular reabsorption and secre- Salicylate overdose was one of the first and best
tion may be reduced (Black et aI., 1976; Popovic and characterised examples of the clinical application of
Popovic, 1974). Renal blood flow and glomerular pharmacokinetics to the diagnosis and treatment of
filtration rate decrease as cardiac output decreases. drug overdose. A concise review of the pharmaco-
Thus, clearance of drugs that are filtered or secreted kinetics of salicylate was published recently (Levy,
may decrease. In contrast, clearance of drugs that are 1978). Salicylate disposition is dependent on the con-
actively reabsorbed may increase. However, we know centration of salicylate in the blood and on urine pH,
of no studies of the effects of hypothermia on drug with large intersubject variability.
elimination by the kidney. Absorption of salicylates in therapeutic doses is
A number of investigators have reported altered usually rapid and complete in less than I hour. Large
effects of drugs during hypothermia but have not single doses of aspirin may delay gastric emptying
measured the concentrations of the drugs. For exam- and dissolution of drug by irritating the gastric
ple, hypothermia prolonged the sleeping time of mice mucosa, resulting in rising salicylate concentrations
after anaesthetic doses of pentobarbitone (Fuhrman, for as long as 24 hours after ingestion (Levy, 1978).
1946). In hypothermic dogs, the pressor responses to Hydrolysis of acetylsalicylic acid in the plasma to
adrenaline (epinephrine), noradrenaline (norepineph- salicylate is rapid with a plasma half-life of IS to 20
rine) and angiotensin were delayed in onset, minutes (Rowland et aI., 1972).
diminished in magnitude, but prolonged in duration Protein binding of salicylate is dose-dependent. At
(Rubenstein, 1961). Hypothermia decreased the low plasma concentrations (e.g. below 80)Jg/ml) in
Pharmacokinetics of Drug Overdose 170

normal subjects, approximately 94 % of salicylate is The tissue distribution of salicylate is also pH de-
bound to albumin (Borga et al., 1976). Protein bind- pendent. Salicylic acid has a pKa of approximately
ing decreases with increasing plasma concentration of 3.0. Because only a small percentage (0.004%) of
salicylate, decreasing plasma concentration of salicylate is not ionised at pH 7.4, small changes in
albumin, and uraemia (Borg a et al., 1976; Ekstrand pH result in large changes in the proportion of drug
et al., 1979; Lowenthal et al., 1974; Yacobi and Levy, that is not ionised and enters tissue. The correlation
1977). There is also large intersubject variability, between arterial bicarbonate and partition of salicyl-
some of which may be genetically determined. Figure ate into the cerebrospinal fluid of infants with salicyl-
2 shows estimations of the increasing volume of dis- ate intoxication is shown in figure 3. Hill (I 973)
tribution with increasing dose in patients with sali- demonstrated the increase in toxicity of salicylate
cylate overdose. As protein binding decreases, sali- with acidaemia and the protective effect of
cyiate distrihution io the hrain, a key target organ in aikaiaemia. The ciinicai significance and TherapeUTic
salicylate toxicity (Hil~, 1973), probably increases. implications of salicylate overdose kinetics were re-
cently reviewed by Temple (J 978) and Done (J 978).
Both respiratory alkalosis and metabolic acidosis are
complications of salicylate overdose. Evaluation of
patients requires knowledge of both concentrations of
salicylate in plasma and arterial pH. Patients with
0.4 alkalaemia often do well with supportive care,
whereas patients with acidaemia should be treated
]>
.-
13
with bicarbonate or, if necessary, haemodialysis .
---
...J •
12
Gabow et al. (I978) recently observed a correlation
';;' 0.3 between incidence of acidaemia and presence of
o
·s.0 neurological symptoms in salicylate intoxicated
·c
1ii adults.
iJ
'0 The elimination of salicylate also varies with dose
~ 0.2 and urinary pH. The dose-dependency of the 2 quan-
:::J
'0
>
titatively most important metabolic pathways has
1: been demonstrated at therapeutic concentrations of
~
~ salicylate. The renal clearance of salicylate is depen-
~0.1 dent on urinary pH. With increasing blood con-
centrations of salicylate, the elimination of salicylate
by the kidney assumes increasing importance. Altera-
tion of urinary pH with antacid therapy has been
100 200 300 shown to alter salicylate levels significantly in
Dose (as salicylic acid) [mg /kg} patients taking antirheumatic doses of aspirin (Levy
and Leonards, 1971; Levy et aI., 1975). The renal ex-
cretion of salicylate is quantitatively more important
Fig. 2. Increasing volume of distribution with increasing after an overdose than after therapeutic doses.
salicylate dose. Alkalinisation of the urine is usually recommended in
• - Accidental intoxication (arrow indicating under-estima-
salicylate overdosed patients (Done, 1978; Temple,
tion of dose due to delay in urine collection); • - chronic in-
toxication; 0 - healthy adult male (from Levy and Yaffe: Ped.
1978). Various treatment regimens have been studied
54: 7813, 1974; reproduced by permission of author and edi- by Lawson et al. (1969) and Morgan and Polak
tor). (J 97 1). In the latter study, there was a 4-fold increase
Pharmacokinetics of Drug Overdose 171

2.2 Paracetamol (acetaminophen)

15 The popularity of paracetamol has increased


• rapidly over the past 20 years. Overdosage of the

--
::; drug is now well documented and has caused many
C"
LU deaths. The predominant complication of overdosage
S is hepatic necrosis and failure, although renal, pan-
~.,<: 10
•• creatic, and cardiac complications also may be ob-
0
.0
lii • • served (Prescott and Wright, 1973; Proudfoot and
u
:c Wright, 1970; Rumack and Matthew, 1975;
(ij
:::l
• Williams and Davis, 1977).
t)
« 5 •• r = +0.90
intercept = 1.26
Paracetamol is absorbed rapidly after therapeutic
slope = 2.84 doses but may be absorbed more slowly as the dose

• increases. Rawlins and coworkers (1977) demon-
strated that plasma concentrations of paracetamol
reach a maximum at 0.5 to I hour after dosing with
2 3 4 5 500mg and 1000mg, respectively, but continue to
Serum/CSF salicylate ratio rise for 2 hours after a 2000mg dose. Oral
bioavailability was incomplete at all dose levels,
presumably due to first-pass metabolism, and was
Fig. 3. Influence of acid-base status (expressed as actual significantly less after 500mg (63 %) than after
serum bicarbonate concentration) on entry of salicylate into I OOOmg (89 %) or 2000mg (87 %). Precise estimates
the CNS (expressed as serum to CSF salicylate concentration
of the percentage of dose absorbed after an overdose
ratio) in 10 infants with salicylate overdose (from Buchanan
and Rabinowitz: J. Ped. 84: 391, 1974; reproduced by permis-
of paracetamol are not available because of the
sion of author and editor). unreliable histories of the amount of drug ingested by
overdosed patients and because of either spontaneous
or induced vomiting after the drug has been taken.
in renal clearance of salicylate for each rise of I unit Despite this, indirect estimates of the dose of
in urine pH. Thus, renal clearance of salicylate in- paracetamol ingested indicate that the mean dose ab-
creased from 16 to 64ml! min as urine pH increased sorbed is higher in patients with liver damage than in
from 6.5 to 7.5. The renal clearance of salicylate also those without, although individual susceptibility to
increased with increasing urine flow, an effect that liver damage varies widely (Prescott and Wright,
diminished as urine pH rose (Morgan and Polak, 1973). As little as 6.2g produced a severe hepatic
197 J). lesion in I patient, whereas 15.3g did not produce
Because of saturable metabolism and increasing liver damage in another.
volume of distribution, the apparent plasma half-life The probable mechanism of paracetamol hepato-
of salicylate increases with increasing dosage. The toxicity has been elucidated primarily by Mitchell and
time for plasma concentrations of salicylate to coworkers (1974). A small fraction of paracetamol is
decrease by one-half averages 2.5 hours after 0.3g, in- metabolised by hepatic microsomal enzymes to N-hy-
creases to 5 to 7 hours after I g, and may be as long as droxyparacetamol. This, in turn, is converted to a
24 hours after an overdose. Because renal clearance of reactive metabolite, which is primarily conjugated to
salicylate is a major determinant of elimination in the glutathione and excreted as mercapturic acid. How-
overdosed patient, the half-life will vary with the ever, the reactive metabolite can also bind covalently
urine pH (fig. 4). to liver cell proteins if hepatic levels of glutathione
Pharmacokinetics of Drug Overdose 172

x
28

24

20

16
£
~ t2 l- ..........
I
:t::
~
III ~.
lii
Q)
8 x ~
>- I •
<> • I
~
(/) 4 ••

6.0 6.5 7.0 7.5 8.0
Urine pH

Fig. 4. Influence of urine pH on salicylate half-life during mannitol-lactate (x) and acetazolamide-bicarbonate (.) treatment
(from Morgan and Polak: elin. Sci. 41: 475, 1971; reproduced by permission of author and editor).

have been depleted to 30 % or less of normal values N-hydroxyparacetamol. The elimination half-life of
(Mitchell et aI., 1973). The binding to liver cell pro- paracetamol is prolonged in overdosed patients, par-
teins causes the hepatic damage. Individual suscep- ticularly when it is associated with hepatic injury
tibility to paracetamol hepatotoxicity probably relates (fig. 5). Prescott et al. (J 97 1) measured plasma con-
in part to the amount of reactive metabolites gener- centrations of paracetamol at 2 points in time, 8
ated, which in turn may depend on the state of hours apart, in patients who did and did not develop
enzyme induction in the individual (for example, by liver damage, as well as in normal subjects after
alcohol, sedatives, or anticonvulsants). Protein mal- therapeutic doses. The plasma concentrations at 4 and
nutrition may deplete hepatic levels of glutathione 12 hours were higher in patients with liver damage
and thus also increase susceptibility to liver cell than in those without. Furthermore, the plasma
necrosis. paracetamol half-life in the 17 patients with liver
The metabolic profile and disposition kinetics of damage was more than twice as long 0.6 ± 0.8
paracetamol are dose-dependent (Davis et aI., I 976; hours) than in the 13 patients without liver damage
Slattery and Levy, 1979). The compound is elimi- (2.9 ± 0.3 hours) and more than 3 times as long as
nated from the body primarily by formation of the half-life in 17 normal subjects (2.0 ± 0.1 hours).
glucuronide and sulphate conjugates, both of which The prolonged half-life could reflect effects of early
are saturable biotransformation pathways. A small hepatic injury and, in fact, has been suggested to be a
fraction ofthe drug is excreted unchanged in the urine predictor of the development of clinical liver damage.
and, as mentioned, a small fraction is metabolised to However, enzyme saturation or limited availability of
Pharmacokinetics of Drug Overdose 173

glucuronic acid or sulphate could also explain the dosed patients. This may be because of poor correla-
prolonged half-life at high paracetamol concentra- tion between blood concentration and pharmacologi-
tions. N-Acetylcysteine has been used successfully in cal effects and the now recognised inappropriateness
preventing or decreasing hepatic damage and increas- of tabulating 'toxic' and 'lethal' plasma or blood con-
ing survival after paracetamol poisoning (Prescott et centrations of drugs. These tabulations are grossly in-
a\., 1977). This drug may act as a scavenger for the accurate because of the problems, particularly after
hepatotoxic metabolite of paracetamol, thus prevent- ingestion of large single doses, of delayed onset of
ing binding of the metabolite to liver proteins pharmacological effects, complex concentration-effect
(Rumack and Peterson, 1978). In addition, N-acetyl- relationships, and tolerance (Prescott et aI., 1973).
cysteine could increase the capacity of the sulphate However, some generalisations about the phar-
conjugation pathway by acting as a sulphate source macokinetics of barbiturates can be made (Breimer,
(Slattery and Levy, 1979). 1974; Harvey, 1975) and may help in the treatment
of barbiturate overdose.
The historical classification of the barbiturates into
2.3 Sedative-Hypnotics long, intermediate, short, and ultrashort acting
categories is based upon the duration of action of a
2.3.1 Barbiturates single dose of each barbiturate in rabbits. However,
Despite the extensive literature available on bar- the duration of action is determined by dose, rate of
biturates, surprisingly little has been published on the distribution, and elimination, and does not neces-
pharmacokinetic behaviour of these drugs in over- sarily correlate with terminal elimination phase half-

1000
- - Uver damage
- - - - No liver damage
300

15
E
a;'"
<J 30
~
"'-
~E
E~ 10
"'-
'" ::L
e.g
al"~
Cl~ 3
i~
-5 8
:58 32 36
4 8 12 16 20 24 28
Hours after overdose

Fig. 5. Paracetamol elimination in patients with and without liver damage (from Prescott et al.: Lancet 1; 519. 1971;
reproduced by permission of author and editor).
Pharmacokinetics of Drug Overdose 174

life. For example, the average half-life of drugs


classified as intermediate acting barbiturates varies
from 3 to 7 hours for hexabarbitone to 34 to 42
hours for butobarbitone. The onset and termination 40
of drug effect are determined by the physicochemical
properties of barbiturates that affect both distribution
and elimination. There are major differences in these
properties between the short and long acting barbitur- 30

ates. The short acting barbiturates are lipid-soluble


and rapidly achieve high levels in the brain. Thus, the
onset of action is within minutes and this fact makes c
'E 20
-.&_-.. .
thionpntonp"- l1<:pflli "n"p<:thpti(' ""pnt Rprli<:trihlltion 1::::-
..s
~&&&~t'_&.~~&.- -~-.-. ~.&_~.- -o-·-~· -----~.--- ---~--
/

of thiopentone from the central nervous system to a>


0
other parts of the body is primarily responsible for <:
e
the termination of its anaesthetic effect. The slower '"
Q)
U 10
rate of elimination probably determines the duration
of residual pharmacological action. The longer acting
barbiturates are less lipid-soluble, may take longer to
produce their effect, and depend more on drug
elimination than redistribution for termination of 2 4 6
effect. Urine flow (ml/min)

In addition to their higher lipid solubility, the


shorter acting barbiturates are more highly protein-
Fig. 6. Influence of urine flow and pH on phenobarbitone
bound (35 to 65 %) than their longer acting counter- renal clearance in dogs.
parts (Harvey, 1975). They are eliminated primarily Ll - urine pH 7.8-8.0; 0 - urine pH below 7.0. Diuresis
by hepatic metabolism and most of the metabolites was induced by oral water, intravenous mercaptomerin or in-
are inactive. Little unchanged drug appears in the travenous sodium sulphate itrom Waddell and Butler: J. Clin.
Invest. 36: 1217, 1957, reproduced by permission of author
urine. These weak acids are mostly non-ionised at
and editor).
physiological urine pH and are reabsorbed by the
renal tubule. Renal excretion may be increased
slightly, although insignificantly, by forced diuresis clearance of phenobarbitone to values as high as
and is not affected by alkalinisation of the urine (Had- l7ml/ min (Hadden et al., 1969). This adds signifi-
den et al., 1969). Long acting barbiturates, such as cantly to the usual renal clearance of phenobarbitone
phenobarbitone, are less protein-bound and have which is approximately I to 3ml/ min under physio-
lower pKas than the shorter acting drugs. Their lower logical conditions of urinary flow and pH. However,
lipid solubility is associated with a smaller volume of forced diuresis incurs the risk of pulmonary oedema
distribution. They are metabolised more slowly by (Goodman et al., 1976).
the liver and a greater fraction of unchanged drug is After therapeutic doses, most of a barbiturate dose
excreted by the kidney. At least for phenobarbitone, is absorbed within an hour; therefore, drug removal
forced diuresis and alkalinisation of the urine increase by emesis or lavage should be attempted early. How-
renal clearance. The contributions of urine pH and ever, because delayed gastric emptying may lead to
flow to renal clearance of phenobarbitone are protracted absorption, gastric lavage and administra-
demonstrated in figure 6. In overdosed patients, tion of charcoal should still be given to patients who
forced alkaline diuresis may increase the renal have ingested barbiturates 4 to 8 hours earlier.
Pharmacokinetics of Drug Overdose 175

Some data are available on barbiturate elimination protein binding of ethchlorvynol in normal subjects is
and half-life after drug overdose. The terminal half- 62 % and does not vary over a 4-fold increase in con-
life of many of the intermediate acting barbiturates in centration of ethchlorvynol in plasma (Benowitz et
overdosed patients is shorter than the half-lives aI., 1980). The ratio of ethchlorvynol concentrations
measured in normal persons after therapeutic doses in cerebrospinal fluid and plasma, which probably
(Prescott et aI., 1973). Furthermore, Martin et al. reflects partitioning of unbound drug, has been
(J 979) found that the mean serum half-life of reported to be 0.42, which is consistent with the pro-
phenobarbitone in chronic sedative-hypnotic abusers tein binding data (Teehan et a!., 1970). After haemo-
is 57.5 ± 4.9 hours versus 86.3 hours in normal perfusion in I patient, the plasma levels of ethchlor-
volunteers. Prescott and coworkers (J 973) and For- vynol rose significantly and peaked 6 hours after the
rest et al. (J 974) also demonstrated that in some over- end of haemoperfusion (Benowitz et aI., 1980). This
dosed patients the rate of elimination of the ingested reflects the high tissue affinity and slow redistribution
barbiturate increased as the plasma concentration of of ethchlorvynol from tissues to plasma.
drug decreased. They proposed that acute barbiturate After a therapeutic dose of ethchlorvynol, the
ingestion induces microsomal enzyme activity, lead- plasma concentration-time curve is biexponential,
ing to an increased rate of barbiturate elimination. An with an average terminal half-life of 24 hours and a
alternative explanation for these observations could clearance from 120 to 140ml/ min (Cummins et aI.,
be that high levels of barbiturates exhibit capacity- 1971). Half-lives of ethchlorvynol in overdosed
limited kinetics (Sumner et aI., 1975). It has also been patients have been reported to be substantially longer
suggested that high concentrations of barbiturates than those observed after a single therapeutic dose.
result in destruction of cytochrome P-450 haem Teehan and coworkers (J 970) reported estimated
(Levin et al., 1972), as discussed previously (section half-lives in 4 patients overdosed with ethchlorvynol
1.1.3 ). ranging from 21 to 105 hours with an average of 72
There is recent evidence that phenobarbitone un- hours. We observed in I patient, that at high con-
dergoes enterohepatic or enteroenteric recirculation, centrations of ethchlorvynol in plasma, elimination
and that this cycle can be interrupted by administra- approached zero order (Benowitz et aI., 1980). The
tion of large multiple doses of activated charcoal. observed rate of elimination was 30mg/L/day, cor-
Neuvonen and coworkers (J 980) administered char- responding to 1.6g/day in our patient. The plasma
coal, I I 8g in 5 divided doses after administration of concentrations of ethchlorvynol reported by Tozer et
phenobarbitone, and observed a shortening of the al. (J 974) also showed that in the posthaemodialysis
half-life from 110 to 20 hours. The half-lives of car- period the decline of plasma concentrations was
bamazepine, phenylbutazone and dapsone were also linear, at a rate of 24.8mg/L/day. Log-linear decline
shortened following administration of charcoal. of ethchlorvynol concentrations after an overdose has
Charcoal was effective in increasing the elimination also been described (Ogilvie et aI., 1966); however,
of dapsone in 2 overdosed patients. Overdoses of the drug was measured by an assay that measured
other drugs with long half-lives and possible entero- ethchlorvynol and metabolites. Renal clearance of
hepatic recirculation, such as phenytoin, might be ethchlorvynol is small and makes an inconsequential
similarly affected. This is an important area for future contribution to overall drug removal (Cummins et aI.,
investigation. 1971; Gibson and Wright, 1972; Westervelt, 1966).

2.3.2 Ethchlorvynol 2.3.3 Glutethimide


Ethchlorvynol overdose, although relatively un- Acute glutethimide intoxication may be in-
common, is associated with prolonged coma and distinguishable from that produced by overdosage
substantial morbidity (Teehan et aI., 1970). Mean with other sedative-hypnotic drugs. However, the
Pharmacokinetics of Drug Overdose 176

clinical course can be complicated by an as yet unex- 2.3.4 Chloral Hydrate


plained cyclic variation in the depth of coma (Maher Blood concentrations of chloral hydrate or its
et al., 1962). Possible explanations include delayed metabolites are rarely reported in patients intoxicated
and intermittent absorption of glutethimide and with this drug. Chloral hydrate is rapidly reduced by
enterohepatic recirculation of glutethimide and/ or of alcohol dehydrogenase to trichloroethanol, the active
active metabolites. Glutethimide is extensively meta- agent. Trichloroethanol is glucuronidated and the
bolised and is excreted primarily in the urine as conjugate is excreted by the kidneys. The half-life of
glucuronidated metabolites. 4-Hydroxyglutethimide trichloroethanol in normal subjects is 8.2 to 13.6
(4-0HG) is active, and in mice produces a sedative hours, the half-life in I overdose patient was 35
effect equal to or greater than the parent compound hours at the highest concentration of trichloroethanol
(Ambre and Fischer, 1974). After therapeutic doses and decreased as plasma concentration declined
of gillthetimicil\ seciation correiaTes weii with pia<;ma (Staiker eT ai., i'ln).
concentrations of glutethimide, and the metabolite (4-
OHG) does not contribute significantly to the effect of
glutethimide (Crow et al., 1977). However, although 2.4 Tricyclic Antidepressants
the data are somewhat conflicting, plasma levels of
glutethimide do not correlate or only correlate Compared with most other drugs, the mortality
roughly with the degree of depression of the central from overdosage with tricyclic antidepressant (TCA)
nervous system in patients who have taken an over- drugs is high. A review of toxicological features in
dose of glutethimide (Chazan and Garella, 1971; 106 severely or fatally intoxicated patients, with dis-
Wright and Roscoe, 1970). Hansen and coworkers cussion of pharmacokinetic aspects, was published by
(1975) demonstrated that 4-0HG accumulates in the Bickel (I 975). Pharmacokinetic considerations are
plasma of patients who have ingested an overdose of important in understanding the time course of the
glutethimide. In 1 patient, they found that coma onset of the signs and symptoms and of the duration
deepened despite a decreasing plasma level of of the potential toxicity. These issues have practical
glutethimide, but the 4-0HG levels were increasing at consequences because continuous cardiac monitoring,
that time; the patient's clinical state correlated best usually in an intensive care unit, is advisable as long
with the sum of the concentrations of both as there is evidence for or a probability of significant
glutethimide and 4-0HG in the plasma. intoxication.
Apart from the accumulation of the active meta- The tricyclic antidepressants (TCAs) are strongly
bolite, the long duration of coma induced by an over- anticholinergic and therefore depress gastrointestinal
dose of glutethimide has been attributed to delayed motility. This may alter the rate of TCA absorption.
drug absorption or to saturation of biotransformation In therapeutic doses, TeAs have been shown to delay
pathways (Maher et ai., 1962; McDonald et al., 1963; the absorption of other drugs (Consolo et al., 1970;
Schreiner et al., 1958). The mean half-life in normal Morgan et al., 1975). Time of peak concentrations
persons is 11.6 hours (Curry et al., 1971). The half- after therapeutic doses of various TCAs range from 3
life in 1 patient reported by Hansen et al. (1975) who to 12 hours. Little data are available concerning the
had taken an overdose was 10 hours. Maher (1970) effects of larger doses of TCAs on their own absorp-
reported a mean half-life in 37 intoxicated patients of tion. In an overdosed patient in whom data were ob-
40.1 ± 5.5 hours. These estimations were only tained, peak concentrations cif nortriptyline occurred
based on paired serum concentrations of glute- 17 hours after ingestion, which is consistent with
thimide, but probably do reflect some saturation of delayed absorption (Sjoqvist et ai., 1972). Evaluation
the biotransformation pathways of glutethimide at of a potentially serious TCA overdose must include
higher drug concentrations. assessment of gastrointestinal activity.
Pharmacokinetics of Drug Overdose 177

Once absorbed, TeAs are extensively bound to tive Oandhyala et al., 1977; Potter et al., 197 9a, b).
body tissues. As a consequence, blood concentrations Thus, a full understanding of the relationship be-
are quite low and only a tiny proportion of the total tween the pharmacokinetics and the pharmaco-
dose is available for elimination by metabolism, urin- dynamics of TeAs in the overdosed patients requires
ary excretion or extracorporeal techniques. High con- measurement of concentrations of parent drugs and
centrations of TeAs have been detected in body active metabolites and quantitative assessment of
tissues as long as 8 days after ingestion (Bickel, their relative activity.
1975).
The time course of TCAs in the blood after an
overdose has been examined in a few patients (Spiker 2.5 Lithium
and Biggs, 1976). Data from 3 of 4 patients after
amitriptyline overdose showed half-lives of 60.3, Appreciation of the pharmacokinetics of lithium
73.5, and 81.4 hours, which are considerably longer may result in better understanding of the course of
than the apparent normal half-life range of 16 to 24 lithium toxicity and offers the possibility of early
hours (Hollister, 1978). The half-life in the fourth diagnosis and treatment of severe overdose. The clini-
patient was 25.3 hours. In 2 patients with des- cal pharmacokinetics of lithium were reviewed re-
ipramine overdose, the terminal half-lives were 26.3 cently (Amidsen, 1977). Pharmacokinetic features of
and 26.5 hours and are at the upper limit of the nor- particular relevance to the overdose situation are the
mal range. Raw data were not published in most narrow therapeutic range of concentrations of lithium
cases, so we cannot assess whether or not the ter- in blood, the dependence of renal clearance of lithium
minal decline phase included continued slow absorp- on sodium balance, and the slow rate of transfer of
tion. In addition, the range of half-lives after thera-
peutic doses is quite wide (Hollister, 1978). For these
reasons, it is difficult to know if the elimination half-
lives in overdosed patients are significantly different
from those observed after therapeutic doses. Of in- 15.0
terest is a pregnant patient who ingested an overdose
of nortriptyline. She had a normal half-life of nortrip- 10

tyline of 17 hours, but the child, born the day after


the overdose, had a prolonged half-life of 56 hours
(Sjoqvist et ai., 1972). 5
Many questions remain in interpreting blood con-
centrations of TeAs after an overdose. It is well
known that tertiary TCAs such as amitriptyline and
imipramine, are demethylated to active secondary
TCAs, nortriptyline and desipramine. Total (tertiary
plus secondary) TeA concentrations in severely in- eNS
E Admission depression
toxicated patients are usually greater than 1)lg/ ml ~ + +
(Petit et al., 1977). The ratio of plasma concentrations ~ 1~~~--~~~~~~--~~~
of parent to demethylated TeA is higher with acute Days
overdose compared with therapeutic overdosing, and
might be useful in differentiating these circumstances Fig. 7. Serum lithium concentrations in a patient following
(Bailey et al., 1978). Recent data suggest that some of acute overdose (from Achong et al.: Canad. Med. Ass. J. 112:
the hydroxylated metabolites of TCAs may also be ac- 868, 1975; reproduced by permission of author and editor).
Pharmacokinetics of Drug Overdose 178

lithium in and out of tissues. A common cause of curs from 8 to 24 hours after an intraperitoneal dose
lithium intoxication during chronic treatment is a (Frazer et aI., 1973; Wraae, 1978). Figure 8 shows
change in the patient's sodium balance, usually in- the time course of lithium in serum and brain from 1
duced by diuretic therapy, a change in diet, or gastro- study. This long distribution time may explain why
intestinal disease with vomiting or diarrhoea. patients, after an acute lithium overdose, may be
The absorption of an overdose of lithium may be asymptomatic despite serum concentrations of lith-
prolonged, perhaps due to the formation of relatively ium of 4mEq/L or greater (Achong et aI., 1975).
insoluble aggregates of the lithium salt. In 1 patient, Studies in rats show that erythrocyte concentra-
the maximum plasma concentration of lithium occur- tions of lithium more closely approximate those of
red 45 hours after ingestion of a massive dose of brain than do serum levels (Frazer et aI., 1973).
lithium carbonate (Achong et aI., 1975) [fig. 7]. The Measurement of the concentration of lithium in red
onset of toxicity wao;; aiso deiayed in this patient, biood ceUs (R HeI, or R He : serum nlTios, hllS been
resulting in failure to appreciate the severity of the suggested as a means of monitoring both therapeutic
overdose. and toxic responses (Elizur et aI., 1972). This might
The clinical course of a patient after lithium over- prove to be most useful in patients after an acute
dose is influenced by the distribution kinetics of overdose, but to our knowledge no such data have
lithium. In particular, the delay in onset and pro- been published. Elizur et aI. (1972) reponed
longed course of neurotoxicity of lithium may be ex- RBC : plasma lithium concentration ratios in a series
plained by the slow rate of transfer of lithium into of patients receiving lithium on a chronic basis. One
and out of the brain. The predominant toxicity of patient demonstrated neurotoxicity despite a normal
lithium is neurological and is presumably related to plasma concentration of lithium. This patient's
the concentration of lithium in the brain. In patients RBC : plasma lithium concentration ratio was higher
who become intoxicated during the course of chronic than that for all other patients, which is consistent
administration of lithium, severity of intoxication with the hypothesis that the RBC lithium concentra-
correlates well with serum concentrations of lithium tion is a better predictor of neurological effects than
(Schou et aI., 1968). In these patients, altered mental the plasma lithium concentration.
status, seizures and coma are most often associated In patients with normal renal function, the renal
with serum concentrations above 2.0mEq/L. Studies clearance oflithium ranges from 10 to 40ml/ min and
in rats show that at steady -state, brain: serum the plasma terminal half-life from 20 to 50 hours
lithium concentration ratios are approximately 1.0, (Amdisen, 1977). In overdosed patients, the elimina-
whereas cerebrospinal fluid: serum ratios are ap- tion half-life may be prolonged to 30-100 hours,
proximately 0.4 (Wraae, 1978). Elimination of presumably due to decreased renal clearance of
lithium from the brain (into the CSF) may occur at a lithium (Schou et aI., 1968). This may, in turn, be due
slower rate than from serum (into urine), as seen in to a consequence of lithium-induced nephrotoxicity
figure 8 (Frazer et al., 1973), so that recovery of these (Amdisen, 1977). Therefore, once patients become in-
patients is usually later than that predicted from a fall toxicated, the course may be prolonged both by slow
in the serum concentration of lithium (Schou et aI., intrinsic clearance of lithium from the brain and
1968). decreased renal clearance of the drug.
In contrast, in patients who become intoxicated In summary, patients who manifest signs of lith-
after a single acute overdose, severity of intoxication ium overdose who have been taking the drug chronic-
does not correlate well with serum concentrations of ally must be distinguished from those who have taken
lithium. This is due at least in part to the slow dis- a single acute overdose. In the former, toxicity should
tribution of lithium from the blood into the brain. In be anticipated at an earlier time and at a lower serum
rats, maximal concentration of lithium in brain oc- concentration of lithium. In the latter, prolonged ab-
Pharmacokinetics of Drug Overdose 179

3.25 3.25

3.00 Plasma 3.00


RBC
2.75 2.75
Brain

2.50 2.50

2.25 2.25

2.00 2.00
/~\ 1.75
,
1.75 \
\

1.50 \
, 1.50
\
,
,,
1.25 1.25
]>
.......
.B' 100
-I.. , \

1.00
0
E
"", , ~~
....... 0.75 ,. .' " 0.75
'},
....J

w
C'
"... " i
EE 0.50 " .........
,
~
f····· ..... ..... 0.50
.... '''I
................~
:::l
:i:
:S 0.25 0.25

2 8 16 2 8 16 24
a. Time (h) b.

Fig. 8. Distribution of lithium in plasma, red blood cell and brain.


Rats were given intraperitoneal injection of lithium chloride (a) 2mEq/kg or (b) 4mEq/kg. Each mean value represents the four
determinations. each bar the SEM (from Frazer et al.: J. Psychiat. Res. 10: l. 1973; reproduced by permission of author and edi·
tor).

sorption and slow distribution of the drug into the clearance and halve the elimination rate (Amdisen
brain combine to delay or prevent the onset of tox· and Skjoldborg, 1969; von Hartitzsch et al., 1972;
icity. Serial determinations of serum levels of lithium Wilson et al., 1971). Accelerated removal of lithium
may be necessary to predict the course of overdose. would be most effective if performed before lithium
A major therapeutic decision involves the use of enters the brain. Once neurotoxicity is evident,
dialysis. Haemodialysis increases lithium clearance removal of lithium from the blood may be of less
by 30 to 1OOml/ min, and peritoneal dialysis provides benefit because of its slow transfer out of the brain.
a lithium clearance of 13 to 15ml/ min; these tech- Both death (Achong et al., 1975) and permanent
niques may more than double endogenous lithium neurological damage (von Hartitzsch et al., 1972)
Pharmacokinetics of Drug Overdose 180

have been reported in patients exposed to toxic levels 1973; Wilson et al., 1979). As the CSF concentration
of lithium for 2 days or more before dialysis was in- would be expected to be directly proportional to the
itiated. Dialysis techniques might be applied earlier to free phenytoin plasma concentration, these data sug-
patients known to be taking lithium chronically who gest that protein binding might have been decreased
have signs of severe intoxication on admission. in these patients.
Restoration and maintenance of adequate body Because of dose-dependent elimination, actual
sodium will optimise the renal clearance of lithium. plasma half-lives of phenytoin cannot be calculated.
Although increasing sodium intake and excretion However, the time for the plasma concentration of
above this rate may increase the clearance of lithium phenytoin to decrease by half has been estimated in
slightly, there is no evidence that forced diuresis sig- intoxicated patients and is usually greater than 60
nificantly increases lithium excretion or favourably hours (Holcomb et aI., 1972; Wilder, 1973) and can
influences the course of overdose (Thomsen and be up to 9.6 days (Gill et ai., ! ~ I ~,I and 1.1. days
Schou, 1968). (Wilson et aI., 1979), although prolonged absorption
and distribution probably contribute to this.

2.6 Phenytoin
2.7 Ethanol
Overdose with phenytoin is relatively common.
Patients often remain intoxicated for a prolonged Ethanol is a common concomitant ingestion in
period of time, primarily due to the dose-dependent many overdose cases, and is the primary intoxicant in
elimination kinetics of phenytoin. A recent review of some cases, mostly in children and non-tolerant
the pharmacokinetics of phenytoin is available adults. A single ingestion of a large amount of
(Richens, 1979). Phenytoin is poorly water-soluble ethanol results in delayed gastric emptying, probably
and decreases gastrointestinal motility, thus resulting as a result of gastric irritation. Consequently, absorp-
in prolonged absorption after ingestion of a large tion may be delayed, and toxicity may occur at a
single dose. Peak plasma concentrations of phenytoin variable time after ingestion. The prediction of peak
occur between 24 and 48 hours after an overdose plasma concentrations of ethanol based upon dose
(Holcomb et al., 1972; Pruitt et aI., 1975; Wilder et and apparent volume of distribution is useful in the
aI., 1973; Wilson et aI., 1979) compared with 3 to 12 management of ethanol ingestion in children. For ex-
hours after therapeutic doses (Richens, 1979). ample, consider a child ingesting 20ml of a commer-
Approximately 92 % of phenytoin is bound to cial product containing 90 % ethanol by weight, a
plasma protein, and binding is decreased in patients maximum dose of 18g of ethanol. Using an apparent
with uraemia, and perhaps acidaemia (Pruitt et al., volume of distribution of 0.6L/kg, the estimated
1975). Although the binding of phenytoin to albumin maximum plasma concentration of ethanol 1 to 2
in vitro does not vary with the phenytoin concentra- hours after ingestion is 1OOmg/ dl. This level is con-
tion (Pruitt et al., 1975), the possibility of concen- sistent with moderate intoxication, but does not indi-
tration-dependent protein binding of phenytoin in cate more serious consequences. The child might be
the overdosed patient has not been assessed ade- discharged after a short period of observation. This
quately. example illustrates the point that only small volumes
In 2 overdose cases, plasma and CSF phenytoin of ethanol are necessary to produce intoxication in a
concentrations were measured simultaneously; the small child, and that pharmacokinetic calculations
resultant ratios were lower than expected, and the may assist in predicting the degree of intoxication. A
degree of intoxication seemed out of proportion to the similar use of the volume of distribution to calculate
plasma concentrations of phenytoin (Wilder et aI., a loading dose of ethanol in the treatment of metha-
Pharmacokinetics of Drug Overdose 181

nol and ethylene glycol intoxication is discussed significantly to the overall elimination rate, but is not
below. clinically recommended.
The elimination of ethanol is generally considered We know of only a few cases in which blood con-
to follow zero-order kinetics. When studied carefully, centrations of alcohol have been characterised after an
however, the rate of disappearance of ethanol appears overdose. In I patient, the decline of blood concentra-
to follow Michaelis-Menten kinetics, and the effect of tions of ethanol from 780 to 200mg/dl appeared
concentration on elimination rates can, for the most to be first-order with a half-life of about 200min
part, be predicted by the Michaelis-Menten equation (Hammond et al., 1973). In another case, however,
as described in section 1.1.3, equation I. As ethanol with a peak blood concentration of ethanol of
concentrations increase, the rate of disappearance 63Smg/dl, elimination appeared to be zero-order
continues to increase. However, as Vm is ap- with a rate of 34mg/dl/hour (Lindblad and Olsson,
proached, the increases become smaller, giving the 1976).
appearance of a fixed- or zero-order elimination rate. An additional use of ethanol pharmacokinetics for
For example, assume that for a particular individual overdosed patients is in the treatment of methanol
the Vm equals 7Smg/kg/h and km equals and ethylene glycol poisoning. The toxic metabolites
13.8mg/dl(McCoy et aI., 1979; Vestaletal., 1977; of these compounds account for most of the mor-
Wilkinson et aI., 1976): at blood concentrations of bidity and mortality resulting from their ingestion
ethanol of 10, SO, 100, and SOOmg/ dl, the rate of dis- (McMartin et aI., 1980; Parry and Wallach, 1974).
appearance, V, will be 31.S, S8.8, 6S.9 and These metabolites are generated by the alcohol
72.8mg/kg/h, respectively. dehydrogenase enzyme system. Ethanol has a greater
First-order elimination processes, which are of affinity for alcohol dehydrogenase than methanol and
minor importance at lower concentrations, may also ethylene glycol. At blood concentrations of ethanol of
contribute to ethanol elimination in the overdosed 100 to 200mg/ dl, much of the metabolism of
patient. Feinman and coworkers (I978) suggested methanol and ethylene glycol is inhibited, so that the
that the microsomal ethanol oxidising system less toxic parent compounds may be eliminated by the
becomes important at higher concentrations of kidney and by haemodialysis. The loading dose of
ethanol. It has also been suggested, although not ethanol is determined by the volume of distribution,
demonstrated, that renal excretion can contribute sig- while the maintenance dose is determined by the
nificantly to total elimination during the overdosed elimination rate, which is dependent on both V m and
period (Hammond et aI., 1973). Because ethanol dis- km (equation I). Assuming an average volume of dis-
tributes into total body water, ethanol concentrations tribution of 0.6L1kg, a loading dose of 0.6g/kg will
in urine are the same as those in blood, and renal result in an average blood concentration of ethanol of
clearance approximates urine flow. Assuming a flow 100mg/dl.
of 100ml/hour, with a steady blood concentration of As reviewed recently by McCoy et aI. (I 979),
ethanol of 100 or 500mg/ dl, the rate of the renal maintenance doses will depend on whether patients
ethanol elimination would be 100mg or SOOmg/ are chronic drinkers (Vm = 7Smg/kg/h for
hour, respectively. Compared with the body's non-drinkers and 17Smg/kg/h for drinkers) and
usual metabolic capacity of about 7g/houdor greater may need to be adjusted for ethanol losses due to
during the overdose period), renal excretion at normal haemodialysis. Thus, to maintain a blood ethanol
urine flow plays a minor role in total elimination. concentration of I OOmg/ dl, a non-drinker would re-
However, during a forced diuresis of 1000mi of quire 66mg/kg/h and a drinker IS4mg/kg/h.
urine/hour in a patient with a blood ethanol con- Assuming ethanol clearance of 120ml/min by
centration of SOOmg/ dl, Sg/hour might be elimi- dialysis, an extra 7.2g ethanol/h would be necessary
nated by the kidney. Such a diuresis might contribute to compensate for dialysis losses.
Pharmacokinetics of Drug Overdose 182

2.8 Theophylline phylline were infused intravenously to 20 children


with asthma. Differences in steady-state serum con-
Theophylline intoxication most commonly occurs centrations were higher than those predicted from the
during therapeutic administration primarily due to dose differences. The daily dose was increased by
disease-induced alterations in its kinetics. However, 50 % in 1 child, but the serum concentration of
theophylline overdose due to suicidal or accidental theophylline increased by more than 300 % and
ingestion is not uncommon. Good correlation has seizures developed. Few data are available on over-
been reported between serum concentrations of dose kinetics. Kadlec et ai. (1978) reported a peak
theophylline and therapeutic (Mitenko and Ogilvie, serum concentration of theophylline of 102.4J.1g/ ml
1973) and toxic effects (Jacobs et aI., 1976; Zwillich in a 17-month old child and biphasic first-
et aI., 1975). Major morbidity and mortality occur as order elimination curve in which serum levels
a re.~mit of neurotoxicity. Zwiiiich et aL (j 975) _________ "vpr
rlp{'iinptl ..... _a _'~fl h,,"r.,
_...... _." w,th
~ ........... "_ ..h"lf~l;fp
a_.... "r 1 'J_.'V'h .hr
A&&_ ............ ... ",
....... ,
.&~_

reported that 8 patients with seizures had serum con- followed by a decline over the next 12 hours with a
centrations of theophylline ranging from 25~g/ ml to half-life of 5.4 hours (fig. 9). Vaucher et al. (1977)
70~g/ml, with a mean of 53J.1g/ml compared with reported 2 children, ages 15 and 5 months, who had
the recommended therapeutic concentrations of 10 to monophasic first-order elimination (half-lives of 8.5
20J.lg/ml. and 9 hours) at peak serum concentrations of
The kinetics of theophylline in children may be theophylline of 65 and 141 J.lg/ ml, respectively. A
dose-dependent, as suggested by Weinberger and possible explanation for this discrepancy is that
Ginchansky (1977). Two different doses of theo- Vaucher used a spectrophotometric assay for theo-

tl/2 = 12.6h
100
kg = O.057h- 1
80
60 Lethargy T •
Vomiting .·t.
40 Haematemesis Seizure •
t
CSF
20

tl/2 = 5.4h
10 kg = O.128h- 1
8
6

TIme (h)

Fig. 9. Serum and CSF theophylline concentration in a 17-month-old child following acute theophylline overdose (from Kadlec
et al.: Ann. Allergy 41: 337, 1978; reproduced by permission of author and editor).
Pharmacokinetics of Drug Overdose 183

phylline, which may have also measured metabolites, was initially longer (43 hours) compared with the ter-
whereas Kadlec et al. used a specific high perfor- minal portion (J 5 hours) (Hobson and Zettner, 1973).
mance liquid chromatographic assay. It is likely that this pattern results from delayed and
continuing gastrointestinal absorption. On the other
hand, several patients exhibited early half-lives of
2.9 Digoxin drug that were shorter than terminal half-lives
(Ahlmark, 1976; Bertler et aI., 1973; Smith and
Digoxin overdose occurs in 2 contexts: the Willerson, 1971). It is likely that the early short half-
chronic, usually gradual, situation, and the acute life represents continuing tissue distribution. At
overdose situation. The first, and by far the most therapeutic doses, the distribution half-life is usually
common form of overdose, occurs in a patient taking about 5 hours, so that distribution is essentially com-
digoxin chronically for treatment of arrhythmias or plete by 24 hours (Reuning et aI., 1973). Conceivably,
cardiac failure. Both cardiac disease and increasing tissue uptake is slower during the overdose so that the
age are associated with reduced creatinine clearance tissue uptake phase, which is still faster than the rate
and decreased renal and total clearance of digoxin. of elimination, is observed for several days. In many
When renal function becomes markedly reduced, of these patients, the reported half-lives after overdose
tissue binding decreases with a decrease in the volume were somewhat shorter than the half-lives reported
of distribution (Reuning et aI., 1973). Another in- after therapeutic doses of digoxin. In most, the short
creasingly recognised cause of digoxin intoxication is half-lives are probably a function of sampling over
coadministration of quinidine, which increases the too short a time interval, thereby including the dis-
serum concentration of digoxin by reducing both tribution phase. However, in I case, the terminal
clearance and extracardiac tissue binding of digoxin half-life of IS hours did appear to be unusually short
(Bigger, 1979). (Hobson and Zettner, 1973). Whether this short half-
Less common is suicidal or accidental overdose of life is a function of the pathophysiology of the over-
a single large dose of digoxin. We reviewed 10 such dose or merely represents normal interindividual
cases in which pharmacokinetic data were obtained variability could only have been experimentally ex-
(Ahlmark, 1976; Bertler et aI., 1973; Hobson and amined by performing postrecovery pharmacokinetic
Zettner, 1973; Rumack et aI., 1974; Smith and studies.
Willerson, 1971): 5 patients had cardiac diseases, and
5 were initially healthy. Two of the patients with car-
diac disease, who were the eldest at 67 and 78 years 2.10 Narcotics
of age, had reduced renal function (creatinine
clearance 49 and 40ml/min, respectively), whereas Although narcotic overdoses are relatively com-
the rest of the cardiac patients had normal renal func- mon, little is known of the pharmacokinetics of
tion. The patients with cardiac disease and reduced opiate drugs in the acutely overdosed patient. Of most
renal function had the longest serum half-lives of relevance is the biological half-life of the administered
digoxin at 36 and 60 hours, respectively, whereas in opiate, because of the relatively short half-life of the
the other overdosed patients, ages 2112 months to 65 competitive narcotic antagonist naloxone. Naloxone
years of age, the calculated serum half-lives of digoxin has a plasma half-life of approximately 90 minutes
ranged from 6 to 33 hours. and an even shorter biological half-life (Fishman et
When an adequate number of blood samples were aI., 1973). After therapeutic doses, the half-lives of
obtained, the shape of the plotted decline of digoxin most narcotics are longer than 90 minutes (Berko-
concentration over time after acute overdose was witz, 1976). Therefore, repetitive injections or con-
usually not simply logarithmic. In I case the half-life tinuous intravenous infusions of naloxone may be
Pharmacokinetics of Drug Overdose 184

necessary to achieve the desired reversal of opiate in doses of 30mg/hour and 60mg/hour (Nomof et
effects in overdosed patients. aI., 1977). Blood concentrations of codeine in 1
patient after an intravenous overdose decreased only
2.10.1 Heroin and Morphine slightly over the first 48 hours, followed by a more
The most commonly abused narcotic, heroin rapid fall with a half-life of approximately 6 hours
(diamorphine), is rapidly metabolised to morphine, (Huffman and Ferguson, 1975). Shock and hepatic
which has a plasma half-life of approximately 2 hours damage during this patient's early course may have
(Spector and Vessell, I 972). Weare not aware of any contributed to the initial slow rate of disappearance of
pharmacokinetic studies in overdosed patients. codeine. The normal elimination half-life of codeine is
Morphine is a weak base and the occurrence of respir- 2.4 to 2.9 hours (Findlay et aI., 1978).
atory acidosis due to overdose may influence the dis-
tribution of morphine. Finck et ai. (i 9771 measured 2.jO.4 Propoxyphene
the volume of distribution and brain concentration of Propoxyphene overdose is increasing in incidence
morphine in dogs with blood at normal pH (7 AI) and and is distinguished from most other narcotic over-
in those with respiratory acidosis (pH 7.15). They doses by the occurrence of seizures (Schou et aI.,
hypothesised that the decrease in systemic pH would 1978). A major metabolite, norpropoxyphene, is car-
result in a decrease in non-ionised morphine from 23 diotoxic and its effects are not antagonised by nalox-
to 14 %. Although the observed volume of distribu- one (Nickander and Smits, 1973). In normal subjects,
tion was smaller in the acidaemic dogs, the brain con- the plasma half-life of propoxyphene is 8 to 24 hours
centration of morphine actually increased, possibly whereas that of norpropoxyphene is 18 to 36 hours
due to increases in cerebral blood flow secondary to (Gram et al., 1979; Wolen et aI., 1975). Therefore,
hypercapnia. Because 10% or less of morphine is ex- norpropoxyphene accumulates in chronic users
creted unchanged in urine (Berkowitz, 1976), changes (Baselt et aI., 1975). The use of hyperventilation for
in urinary pH do not substantially affect total management of cardiac toxicity due to propoxyphene
morphine clearance. overdose has been proposed by Schou et aI. (J 978).
The authors reported that 5 patients with severe pro-
2.10.2 Methadone poxyphene intoxication who were treated with hyper-
From 5 to 22 % of a dose of methadone, which is ventilation had higher maximum serum levels of pro-
also a weak base, may be excreted unchanged into poxyphene and norpropoxyphene, and had longer
alkaline and acid urine, respectively (Baselt and plasma half-lives of propoxyphene (mean of 30.5
Casarett, I 972). Spontaneous changes in urine pH hours) than 6 less severely overdosed patients who
result in 3-fold differences in renal clearance of were treated conservatively (J 7.9 hours). Because
methadone in methadone-maintained patients studied plasma levels of drug and metabolite were determined
by Bellward et aI. (1977). Since methadone has a long only after induction of respiratory alkalosis, it is not
half-life, often > 24h, a continuous infusion of nalo- possible to distinguish between concentration-depen-
xone (0.04mg/mO may be necessary to antagonise dent and pH-dependent effects on the disposition of
methadone effects over the course of the overdose. propoxyphene in these patients.

2.10.3 Codeine
Codeine (methylmorphine) is in part metabolised 2.11 Stimulants
to morphine. The metabolism may be dose-depen- (Amphetamine and Phencyclidine)
dent, as suggested by the observation that maximum
rates of urinary excretion of codeine and metabolites, Although the use of amphetamines is apparently
measured during intravenous infusions, were similar decreasing, that of phencyclidine is increasing and
Pharmacokinetics of Drug Overdose 185

r~sults in considerable morbidity and mortality. 60mg/kg, and then increased again so that mortality
Because both drugs are weak bases, their distribution at 1OOmg/kg equalled that at 40mg/kg. Pretreatment
and renal elimination may be affected by pH. with inhibitors of hepatic oxidative metabolism
The renal elimination of amphetamine is sensitive abolished the triphasic response curve. This observa-
to changes in urinary pH. Approximately 60 % of an tion supports the suggestion that metabolites of
intravenously administered dose of amphetamine is amphetamine may contribute to its toxicity.
excreted unchanged into acid urine, whereas less than The issue of manipulation of acid-base status in
20 % is usually excreted into alkaline urine (Davis et the treatment of phencyclidine (PCP) intoxication is
aI., 1971). However, amphetamine is also metabol- important, controversial, and requires further in-
ised, primarily deaminated but also hydroxylated, to vestigation. We know of no published studies of the
potentially toxic metabolites. Davis et ai. (1971) pharmacokinetics of PCP in man. Plasma concentra-
demonstrated a shorter plasma half-life of am- tions of PCP after intravenous administration have
phetamine with acid urine (8 to 10.5 hours) than with been studied in dogs and monkeys, but neither renal
alkaline urine (16 to 31 hours). Anggard et al. (1973) clearance nor the effect of pH on drug distribution
acidified the urine of 3 patients and alkalinised the was reported (Wilson and Domino, 1978). Thus, the
urine of 4 patients with amphetamine psychosis. The relevant data are confined to overdosed patients in
plasma half-life of amphetamine in the 3 with acid whom the doses are unknown. However, a few con-
urine was 7 to 14 hours, and the symptoms cleared clusions can be made from these studies. Urinary
rapidly. The plasma half-life of the 4 with alkaline acidification clearly increases the renal clearance of
urine was 19 to 34 hours, and psychosis was PCP (Aronow et ai., 1978; Domino and Wilson,
prolonged. As a consequence of decreased renal 1977; Done et ai., 1977; Done et ai., 1978). Gastric
elimination of amphetamine, the formation of basic acidification increases the partitioning of phen-
polar metabolites increases; these metabolites may be cyclidine into gastric contents (Done et al., 1978).
active and contribute to toxicity. Although these After the recommendations of these authors, urinary
results provide a rationale for acidifying the urine of acidification and gastric suction have been used
patients with amphetamine psychosis, its use has not widely in the treatment of phencyclidine overdose
been studied in patients with acute amphetamine (Rappolt et ai., 1979). However, the contribution of
overdose. In such cases, morbidity is related to the techniques that increase renal or gastric clearance to
acute cardiovascular effects of the drug rather than to total body clearance is uncertain and may, in fact, be
the time course of persistence of the drug in the body. small. In the few cases in which the time course of
Acute amphetamine and phencyclidine overdose has serum concentrations of phencyclidine was reported,
been associated with rhabdomylosis and excretion of the concentrations did not appear to be substantially
myoglobin in urine (Cogen et al., 1978). It has been altered by these techniques (Done et aI., 1978). It is
postulated that in acid urine a greater amount of fer- difficult to evaluate the improvement in clinical status
rihemate dissociates from the globin of myoglobin, after treatment because all patients reported were so
resulting in more nephrotoxicity (Eneas et al., 1979). treated (Aronow et ai., 1978).
The risks of urine acidification in the presence of In 1 patient, phencyclidine concentrations in
myoglobinuria may outweigh the potential benefits serum and CSF were determined sequentially at vary-
(Barton et al., 1980). ing serum pHs (Done et ai., 1978). At pH 7.4, the
An interesting aspect of amphetamine toxicity of CSF : serum ratio was 140: 66ng/ ml, or approx-
uncertain clinical significance is the triphasic lethal imately 2: 1; at pH 7.2, this ratio was 8: SOng/ml,
dose curve obtained in mice (James and Franklin, or approximately 1 : 6. An immediate improvement
1978). The lethality of amphetamine in mice in- in unspecified central nervous system effects was
creased up to 40mg/kg, decreased between 40 and reported. These investigators recommended mainte-
Pharmacokinetics of Drug Overdose 186

nance of serum pH at approximately 7.3 during ad- urinary acidification or alkalinisation, haemoperfu-
ministration of acid to effect urinary acidification. sion or haemodialysis should be preceded and
This is a potentially hazardous procedure (Barton et followed by adequate periods of blood concentration
aI., 1980), and its ultimate benefit to the outcome of measurement in order to assess quantitative removal
the patient remains to be documented. beyond that attributable to endogenous processes and
to detect possible late rebound of drug concentrations.
Direct measurement of drug in faeces, urine, haemo-
3. Design a/Clinical Studies perfusion cartridge or dialysate solutions, as ap-
propriate, are also most useful.
In reviewing clinical studies of the pharmaco-
kinetics of drug overdose, a number of problems
thai make resuiis difficuii io evaiuate were noted. We A ckn()wiedgement.~
offer the following suggestions for future studies:
1. Frequent blood sampling in the first few days Preparation of this review was supported in part by
and continued blood sampling over the entire course research grants DAO 1696 and DA02453 from the United
States National Institutes of Health.
of recovery after overdose may permit better charac-
terisation of absorption and terminal elimination
kinetics.
2. Measuring concentrations of both ingested Re/erences
drug and known or potentially active metabolites and
relating these concentrations to quantitative measures Aarbakke. J.: Clinical pharmacokinetics of phenylbutazone. Clini-
cal Pharmacokinetics 3: 369-380 (1978).
of neurological depression or other clinical observa-
Achong, M.R.; Fernandez. F.G. and McLeod, PJ.: Fatal self-
tions may suggest relationships which could later be poisoning with lithium carbonate. Canadian Medical Associa-
analysed by covariant analyses. tion Journal 112: 868-870 (1975).
3. Measuring the concentrations of several drugs Ahlmark, G.: Extreme digitalis intoxication. Acta Medica Scan-
dinavica 200: 423-425 (1976).
over the course of an overdose may be necessary to
Amhre, .r..l. and Fischer, 1...1.: Identification and activity of the hy-
explain clinical observations when multiple drugs droxy metabolite that accumulates in the plasma of humans in-
have been ingested. toxicated with glutethimide. Drug Metabolism and Disposition
4. Measurement of protein binding in the drug- 2: 151-158 (1974).
overdosed patient may demonstrate that with 'higher- Amdisen. A.: Serum level monitoring and clinical phar-
than-therapeutic' blood concentrations or in the pre- macokinetics of lithium. Clinical Pharmacokinetics 2: 73-92
(t 977).
sence of other drugs andlor physiological distur- Amdisen. A. and Skjoldborg. H.: Hemodialysis for lithium
bances such as acidaemia, hypo- or hyperthermia, poisoning. Lancet 2: 213 (1969).
free drug concentrations differ from that expected Anggard, E.; Jonsson, L.-E.; Hogmark, A.-L. and Gunne, L.-M.:
from data obtained in normal subjects. Amphetamine metabolism in amphetamine psychosis. Clinical
5. For overdoses such as with lithium where Pharmacology and Therapeutics 14: 870-879 (t 973).
Arnold, K. and Gerber, N.: The rate of decline of diphenylhydan-
serious toxicity can occur long after peak blood con- toin in human plasma. Clinical Pharmacology and Thera-
centrations, studying the relationship between blood peutics II: 121-134 (1970).
concentrations measured early in the course of over- Aronow, R.; Miceli, J.N. and Done. A.K.: Clinical observations
dose and outcome, may provide a rational basis for during phencyclidine intoxication and treatment based on ion-
trapping; in Petersen and Stillman (Eds) Phencyclidine (PCP)
interventions such as haemodialysis before serious
Abuse: An Appraisal. pp.218-228 (NIDA Research
toxicity is manifest. Monograph 21.1978). .
6. Manipulations designed to accelerate drug Bailey. D.N.; Van Dyke, c.; Langov. R.A. and Jatlow. P.1.: Tri-
removal, such as repeated administration of charcoal, cyclic antidepressants: plasma levels and clinical findings in
Pharmacokinetics of Drug Overdose 187

overdose. American Journal of Psychiatry 135: 1325-1328 Breimer, D.D.: Pharmacokinetics of Hypnotic Drugs (Drukkerij-
(1978). Uitgeverij Brakkenstein, Nijmegen 1974).
Barton, CH.; Sterling, M.L. and Vaziri, N.D.: Rhabdomyolysis Brown, T.CK.: Sodium bicarbonate treatment for tricyclic anti-
and acute renal failure associated with phencyclidine intoxica- depressant arrhythmias in children. Medical Journal of
tion. Archives of Internal Medicine 140: 568-569 (1980). Australia 2: 380-382 (I 976a).
Baselt, R.C and Casarett, L.J.: Urinary excretion of methadone in Brown, T.CK.: Tricyclic antidepressant overdosage: experimental
man. Clinical Pharmacology and Therapeutics 13: 64-70 studies on the management of circulatory complications. Clini-
(972). cal Toxicology 9: 255-272 (I 976b).
Baselt, R.C; Wright, J.A.; Turner, J.E. and Cravey, R.H.: Pro- Buchanan, N. and Rabinowitz, L.: Infantile salicylism - a reap-
poxyphene and norpropoxyphene concentrations in fatalities praisal. Journal of Pediatrics 84: 391-395 (1974).
associated with propoxyphene hydrochloride and propoxy- Chazan, J.A. and Garella, S.: Glutethimide intoxication: a
phene napsylate. Archives of Toxicology 34: 145-152 (1975). prospective study of 70 patients treated conservatively without
Becker, CE.; Tong, T.G.; Boerner, U.; Roe, L.L.; Scott, R.A.; hemodialysis. Archives of Internal Medicine 128: 215-220
MacQuarie, M. B. and Bartter, F.: Diagnosis and treatment of 0970.
Amanita phalloides-type mushroom poisoning. Western Jour- Citron, B.P.; Halpern, M.; McCarson, M.; Lundberg, G.D.; Mc-
nal of Medicine 125: 100-109 (976). Cormick, R.; Pincas, U.; Tatter, D. and Haverback, B.1.:
Bellward, G.D.; Warren, P.M.; Howald, P.; Axelson, J.E. and Necrotizing angiitis associated with drug abuse. New England
Abbott, F.S.: Methadone maintenance: effects of urinary pH Journal of Medicine 283: 1003-1011 (1970).
on renal clearance in chronic high and low doses. Clinical Cogen, F.; Rigg, G.; Simmons, J.L. and Domino, E.F.: Phen-
Pharmacology and Therapeutics 22: 92-99 (1977). cyclidine associated acute rhabdomyolysis. Annals of Internal
Benowitz, N.; Abolin, C; Tozer, T.; Rosenberg, J.; Rogers, W.; Medicine 88: 210-220 (1978).
Pond, S.; Schoenfeld, P. and Humphreys, M.: Resin hemoper- Consolo, S.; Morselli, D.L.; Zaccala, M. and Garattini, S.: Delayed
fusion in ethchlorvynol overdose. Clinical Pharmacology and absorption of phenylbutazone caused by desmethylimipramine
Therapeutics 27: 236-242 (980). in humans. European Journal of Pharmacology 10: 239-242
Benowitz, N.L.; Rosenberg, J. and Becker, CE.: Cardiopulmon- (1970).
ary castrophes in drug-overdosed patients. Medical Clinics of Crow, J.W.; Lain, P.; Bachner, F.; Shaeman, D.W. and Azarnoff,
North America 63: 267-296 (1979). D.L.: Gluthethimide and 4-0H glutethimide: phar-
Benowitz, N.L. and Meister, D.W.: Pharmacokinetics in patients macokinetics and effect on performance in man. Clinical Phar-
with cardiac failure. Clinical Pharmacokinetics I: 389-405 macology and Therapeutics 22: 458-464 (977).
(976). Cummins, L.M.; Martin, Y.C and Scherfling, E.E.: Serum and
Berkowitz, B.A.: The relationship of pharmacokinetics to phar- urine levels of ethchlorvynol in man. Journal of Pharmaceutic-
macological activity: morphine, methadone and naloxone. al Science 60: 261 (1971).
Clinical Pharmacokinetics I: 219-230 (976). Cunningham, E.E.; Brentiens, J.R.; Zielezny, M.A.; Andres, G.A.
Bertler, A.; Gustafson, A. and Redfors, A.: Massive digoxin intox- and Venuto, R.C.: Heroin nephropathy, a clinicopathologic
ication. Report of two cases with pharmacokinetic correla- and epidemiologic study. American Journal of Medicine 68:
tions. Acta Medica Scandinavica 194: 245-249 (1973). 47-53 (1980).
Beyda, EJ.; Jung, M. and Bellet, S.: Effect of hypothermia on the Cunningham, J.L.; When, D.D.; Shudo, \. and Azarnoff. D.L.:
tolerance of dogs to digitalis. Circulation Research 9: 129-135 The effects of urine pH and plasma protein binding on the
(1960. renal clearance of disopyramide. Clinical Pharmacokinetics 2:
Bickel, M.H.: Poisoning by tricyclic antidepressant drugs. General 373-383 (1977).
and pharmacokinetic considerations. International Journal of Curry, S.H.; Riddall, D.; Gordon, 1.S.; Simpson, G.; Binns, F.E.;
Clinical Pharmacology II: 145-176 (1975). Rondel, R.K. and McMartin, C: Disposition of glutethimide
Bigger, J.T.: The quinidine-digoxin interaction: what do we know in man. Clinical Pharmacology and Therapeutics 12: 849-857
about it? New England Journal of Medicine 30 I: 779-781 (1970.
(979). Davis, J.M.; Kopin, 1.J.; Lemberger, L. and Axelrod, J.: Effects of
Black, P.R.; Van Devanter, S. and Cohn, L.: Effects of hypother- urinary pH on amphetamine metabolism. Annals of the New
mia on systemic and organ system metabolism and function. York Academy of Sciences 179: 493-50 I (1971).
Journal of Surgical Research 20: 49-63 (1976). Davis, M.; Labadarios, D. and Williams, R.S.: Metabolism of
Blaschke, T.F.: Protein binding and kinetics of drugs in liver paracetamol after therapeutic and hepatotoxic doses in man.
disease. Clinical Pharmacokinetics 2: 32-44 (977). Journal of International Medical Research 4 (Suppl. 4): 40-45
Borga, 0.; Odar-Cederlof, J.; Ringberger, V.A. and Norlin, A.: (1976).
Protein binding of salicylate in uremic and normal plasma. DeMatteis, F.: Rapid loss of cytochrome P-450 and haem caused
Clinical Pharmacology and Therapeutics 20: 464-475 (1976). in the liver by the porphyrogenic agent 2-a1lyl-2-
Pharmacokinetics of Drug Overdose 188

isopropylacetamide. FEBS Letters 6: 343-345 (1970). Frazer, A.; Mendels, 1.; Secunda, S.K.; Cochrane, C.M. and
Domino, E.F. and Wilson, A.E.: Effects of urine acidification on Bianchi, c.P.: The prediction of brain lithium concentrations
plasma and urine phencyclidine levels in overdosage. Clinical from plasma or erythrocyte measures. Journal of Psychiatric
Pharmacology and Therapeutics 22: 421-424 (1977). Research 10: 1-7 (1973).
Done, A.K.: Aspirin overdosage: incidence, diagnosis, and Fuhrman, F.A.: The effect of body temperature on drug action.
management. Pediatrics 62 (SuppU: 890-897 (1978). Physiology Review 26: 247-276 (1946).
Done, A.K.; Aronow; R.; Miceli, J.N. and Lin, D.C.K.: Phar- Fuhrman, F.A.; Crismon, 1.M.; Fuhrman, G.1. and Field, J.: The
macokinetic observations in the treatment of phencyclidine effect of temperature on the inactivation of epinephrine in vivo
poisoning: A preliminary report, in Rumack and Temple (Eds) and in vitro. Journal of Pharmacology and Experimental
Management of the Poisoned Patient, pp. 79-102 (Science Therapeutics 8: 323-334 (J 944).
Press, Princeton 1977). Gabow, P.A.; Anderson, R.1.; Potts, D.E. and Schrier, R.W.:
Done, A.K.; Aronow, R. and Miceli, J.N.: The pharmacokinetics Acid-base disturbances in the saliCYlate-intoxicated adult.
of phencyclidine in overdosage and its treatment, in Petersen Archives of Internal Medicine 138: 1481-1484 (978).
and Stillman (Eds) Phencyclidine (PCP) .l\buse: i\.n /\ppraisal. George, C.F.: Drug kinetics and hepatic blood flow. Clinical Phar·
pp.21 0-217 (NIDA Research Monograph 21, 1978). macokinetics 4: 433-448 (J 979).
Ekstrand, R.; Alvan, G. and Borga, 0.: Concentration dependent Gibaldi, M. and Perrier, D.: Nonlinear pharmacokinetics; in Phar-
plasma protein binding of salicylate in rheumatoid patients. macokinetics pp.215-228 (Marcel Dekker, New York 1975).
Clinical Pharmacokinetics 4: 137-143 (1979). Gibson, P.F. and Wright, N.: Ethchlorvynol in biological fluids:
Elizur, A.; Shopsin, B.; Gershon, S. and Ehlenberger, A.: Intra- specificity of assay methods. Journal of Pharmaceutical
extra-cellular lithium ratios and clinical course in affective Science 61. 169 (972).
states. Clinical Pharmacology and Thherapeutics 13: 947-952 Gill, M.A.; Kein, J.W.; Kaneko, J.; McKeon, J. and Oavis, c.:
(1972). Phenytoin overdose kinetics. Western Journal of Medicine
Eneas, J.F.; Schoenfeld, P.Y. and Humphreys, M.H.: The effect of 128: 246-248 (1978).
infusion of mannitol-sodium bicarbonate on the clinical course Gleason, W.A.; deMello, D.E.; deCastro, F.1. and Connors, J.1.:
of myoglobinuria. Archives of Internal Medicine 139: Acute hepatic failure in severe iron poisoning. Journal of
801-805 (979). Pediatrics 95: 138-140 (979).
Fabre, J. and Balant, L.: Renal failure, drug pharmacokinetics and Goodman, J.M.; Bischel, M.D.; Wagers, P.W. and Barbour, B.H.:
drug action. Clinical Pharmacokinetics I: 99-120 (976). Barbiturate intoxication - morbidity and mortality. Western
Fabre, J.; Fox, H.M.; Dayer, P. and Balant, L.: Differences in Journal of Medicine 124: 179-186 (976).
kinetic properties of drugs: Implications as to the selection of a Gram, L.F.; Schou, J.; Way, W.L.; Heltberg, J. and Bodin, N.O.:
particular drug for use in patients with renal failure (special d-Propoxyphene kinetics after single oral and intravenous
cmphasi, un antibiutics ami p-adr~nuc~plur blUl.;king ageni>;). uus~s in man. Clinical Pharmacology and Therapeutics 26:
Clinical Pharmacokinetics 5: 441-464 (1980). 473-482 (J 979).
Feinman, L.; Baraona, E.; Matsuzaki, S.; Korsten, M. and Lieber, Hadden, J.; Johnson, K.; Smith, S.; Price, L. and Giardina, E.:
C.S.: Concentration dependence of ethanol metabolism in vivo Acute barbiturate intoxication. Concepts of management.
in rats and man. Alcohol: Clinical Experimental Research 2: Journal of the American Medical Association 209: 893-900
381-385 (978). (1969).
Finck, A.D.; Berkowitz, B.A.; Hempstead, J. and Ngai, S.H.: Hammond, K.B.; Rumack, B.H. and Rodgerson, D.O.: Blood
Pharmacokinetics of morphine: Effects of hypercarbia on ethanol: a report of unusually high levels in a living patient.
serum and brain morphine concentrations in the dog. Journal of the American Medical Association 226: 63-64
Anesthesiology 47: 407-410 (977). (973).
Findlay, J.W.A.; Jones, E.C.; Butz, R.F. and Welch, R.M.: Hansen, A.R.; Kennedy, F.A. and Ambre, J.J.: Glutethimide
Plasma codeine and morphine concentrations after therapeutic poisoning: a metabolite contributes to morbidity and mortality.
oral doses of codeine-containing analgesics. Clinical Phar- New England Journal of Medicine 292: 250-252 (J 975).
macology and Therapeutics 24: 60-68 (1978). Harvey, S.c.: Hypnotics and sedatives. The barbiturates; in Good-
Fishman, J.; Roffwarg, H. and Hellman, L.: Disposition of nalox,- man and Gilman (Eds) The Pharmacological Basis of Thera-
one- 7,8 'H in normal and narcotic dependent men. Journal of peutics, pp.102-122 (MacMillan, New York 1975).
Pharmacology and Experimental Therapeutics 187: 575-580 Hill, J.B.: Salicylate intoxication. New England Journal of
(1973). Medicine 288: 1110-1113 (973).
Forrest, J.A.H.; Roscoe, P.; Stevenson, I.H. and Prescott, L.F.: Hobson, J.D. and Zettner, A.: Digoxin serum half-life following
Abnormal drug metabolism following barbiturate and suicidal digoxin poisoning. Journal of the American Medical
paracetamol overdose. British Medical Journal 4: 499-502 Association 223: 147-149(973).
(1974). Holcomb, R.; Lynn, R.; Harvey, B.; Sweetman, B.J. and Gerber,
Pharmacokinetics of Drug Overdose 189

N.: Intoxication with 55-diphenylhydantoin (Dilantin). Jour- diuresis in the treatment of acute salicylate pOisoning in adults.
nal of Pediatrics 80: 627-632 (1972). Quarterly Journal of Medicine 38: 31-48 (1969).
Hollister, L.E.: Tricyclic antidepressants. New England Journal of Levin, W.; Sernatinger, E.; Jacobson, M. and Kuntzman, R.:
Medicine 299: 1106-1109 (1978). Destruction of cytochrome P450 by secobarbital and other
Huffman, D.H. and Ferguson, R.L.: Acute codeine overdose: cor- barbiturates containing allyl groups. Science 176: 1341-1343
respondence between clinical course and codeine metabolism. (1972).
Johns Hopkins Medical Journal 136: 183-1 86 (1975). Levy, G.: Clinical pharmacokinetics of aspirin. Pediatrics 62
Ivanetich, K.M.; Lucas, S.; Marsh, J.A.; Ziman, M.R.; Katz, 1.0. (Supp!): 867-872 (1978).
and Bradshaw, J.1.: Organic compounds: their interaction with Levy, G.: Pharmacokinetics in renal disease. American Journal of
and degradation of hepatic microsomal drug-metabolizing Medicine 62: 461-465 (1977).
enzymes in vitro. Drug Metabolism and Disposition 6: Levy, G.: Pharmacokinetics of salicylate elimination in man. Jour-
218-225 (1978). nal of Pharmaceutical Sciences 54: 959-967 (1965).
Jacobs, M.H.; Senior, R.M. and Kessler, G.: Clinical experience Levy, G.; Lampman, T. and Kamath, B.L.: Decreased serum
with theophylline: relationships between dosage, serum con- salicylate concentrations in children with rheumatic fever
centration and toxicity. Journal of the American Medical treated with antacids. New England Journal of Medicine 293:
Association 235: 198J-1986 (1976). 323-325 (1975).
James, R.e. and Franklin, M.R.: The triphasic amphetamine lethal Levy, G. and Yaffe, S.1.: Relationship between dose and apparent
dose curve in mice and its possilile relationship to drug meta- volume of distribution of salicylate in children. Pediatrics 54:
bolism. Toxicology and Applied Pharmacology 44: 63-73 713-717 «974).
(1978). Levy, G.; Tsuchiya, T. and Amsel, L.P.: Limited capacity for
Jandhyala, B.S.; Steenberg, M.L.; Perel, J.M.; Manian, A.A. and salicyl phenolic glucuronide formation and its effect on the
Buckley, J. P.: Effects of several tricyclic antidepressants on the kinetics of salicylate elimination in man. Clinical Phar-
hemodynamics and myocardial contractility of the macology and Therapeutics 13: 258-268 (1972).
anesthetized dog. European Journal of Pharmacology 42: Levy, G. and Leonards, J.R.: Urine pH and salicylate therapy.
403-410(1977). Journal of the American Medical Association 217: 81 (197 I).
Johnson, D.G.; Hayward, J.S.; Jacobs, T.P.; Collis, M.L.; Ecker- Lindblad, B. and Olsson, R.: Unusually high blood levels of
son, J.D. and Williams, R.H.: Plasma norepinephrine alcohol. Journal of the American Medical Association 236:
responses of man in cold water. Journal of Applied Physiology 1600-1602 (1976).
43: 216-220 (1977). Lowenthal, P:T.; Briggs, W.A. and Levy, G.: Kinetics of salicylate
Kadlec, G.1.; Jarboe, e.H.; Pollard, S.1. and Sublett, J.L.: Acute elimination by anephric patients. Journal of Clinical Investiga-
theophylline intoxication. Biphasic first order elimination tion 54: 1221-1226 (1974).
kinetics in a child. Annals of Allergy 41: 337-339 (1978). Lundquist, F. and Wolthers, H.: The kinetics of alcohol elimina-
Kaiser, S.e.; Kelvington, E.1.; Kunig, R. and Randolph, M.M.: tion in man. Acta Pharmacologia and Toxicologia 14: 265
Drug metabolism in hypothermia. Uptake, metabolism and ex- (1958).
cretion of C 14 -procaine by the isolated, perfused rat liver. Jour- Maher, J.F.: Toxic nephropathy; in Brenner and Rector (Eds) The
nal of Pharmacology and Therapeutics 164: 396-404 « 968). Kidney, pp.1355-1395 (Saunders, Philadelphia 1976).
Kimberly, R.P.; Bowden, R.E.; Keiser, H.R. and Plotz, P.H.: Maher, J.F.: Determinants of serum half-life of glutethimide in in-
Reduction of renal function by newer nonsteroidal anti-in- toxicated patients. Journal of Pharmacology and Experimental
flammatory drugs. American Journal of Medicine 64: Therapeutics 174: 450-455 (1970).
804-807 (1978). Maher, J.F.; Schreiner, G.E. and Westervelt, F.B., Jr.: Acute
Koffler, A.; Friedler, R.M. and Massry, S.G.: Acute renal failure glutethimide intoxication: I. Clinical experience (22 patients)
due to nontraumatic rhabdomyolysis. Annals of Internal compared to acute barbiturate intoxication (63 patients).
Medicine 85: 23-28 (1976). American Journal of Medicine 33: 70-81 (1962).
Krarup, N. and Larsen, J.A.: The effect of slight hypothermia on Martin, P.E.; Kapar, B.M.; Whiteside, E.A. and Sellers, E.M.: In-
liver function as measured by the elimination rate of ethanol, travenous phenobarbital ttherapy in barbiturate and other hyp-
the hepatic uptake and excretion of indocyanine green and bile nosedative withdrawal reactions: a kinetic approach. Clinical
formation. Acta Physiologica Scandinavica 84: 396-407 Pharmacology and Therapeutics 26: 256-264 (1979).
(1972). McAllister, R.G.; Bourne, D.W.; Tan, T.G.; Erickson, J.L.;
Lavender, S.; Brown, J.N. and Berrill, W.T.: Acute renal failure Wachtel, e.e. and Todd, E.P.: Effects of hypothermia on
and lithium intoxication. Postgraduate Medical Journal 49: propranolol kinetics. Clinical Pharmacology and Therapeutics
277-279 (\ 97 3). 25: 1-7 (1979).
Lawson, A.A.H.; PrOUdfoot, A.T.; Brown, S.S.; Macdonald, McAllister, R.G.; Tan, T.G. and Todd, E.P.: Effect of hypother-
R.H.; Fraser, A.G.; Cameron, J.c. and Matthew, H.: Forced mia on the metabolism of propranolol, quinidine and
Pharmacokinetics of Drug Overdose 190

verapamil. Clinical Pharmacology and Therapeutics 23: 121 Ogilvie, R.I.; Douglas, D.E.; Lochead, J.R.; Moscovich, M.D. and
([978). Kaye, M.: Ethchlorvynol (Placidyll intoxication and its treat-
McCoy, H.G.; Cipolle, R.1.; Ehlers, S.M.; Sawchuk, R.J. and ment by hemodialysis. Canadian Medical Association Journal
Zaske, D.E.: Severe methanol poisoning. Application of a 95: 954-956 (1966).
pharmacokinetic model for ethanol therapy and hemodialysis. Parry, M.F. and Wallach, R.: Ethylene glycol poisoning.
American Journal of Medicine 67: 804-807 ([ 979). American Journal of Medicine 57: 143-150 ([974).
McDonald, D.F.; Green, W.M. and Kretchmar, L.: Experiences in Petit, J.M.; Spiker, D.G.; Ruwitch, J.F.; Ziegler, V.E.; Weiss,
acute glutethimide (Doriden) intoxication. Investigations in A.N. and Biggs, J.T.: Tricyclic antidepressant plasma levels
Urology I: 127-133 ([963). and adverse effects after overdose. Clinical Pharmacology and
McMartin, K.E.; Ambre, J.1. and Tephly, T.R.: Methanol poison- Therapeutics 21: 47-51 ([977).
ing in human subjects. Role for formic acid in the metabolic Popovic, V. and Popovic, P.: Hypothermia in Biology and
acidosis. American Journal of Medicine 68: 414-418 ([ 980). Medicine (Grune and Stratton, New York 1974).
Mitchell, J.R.; Thorgeirsson, S.S.; Potter, W.Z.; Jollow, D.J. and Potter, W.Z.; Calil, H.; Zavadil, A.; Jusko, W.; Suftin, T. and
Keiser. 1-1.: ACetaniinophen-induced hepatic injur;: protective RapapuJ i, L Aciive hyuroxyiateO metaboiites of tricvciic anti-
role of glutathione in man and rationale for therapy. Clinical depressants in man (Abstract). Clinical Pharmacology and
Pharmacology and Therapeutics 16: 676-684 ([ 974). Therapeutics 25: 242 ([ 979a).
Mitchell, J.R.; Jollow, 0.1.; Potter, W.Z.; Davis, D.C.; Gillette, Potter, W.Z.; Cali!, H.M.; Manian, A.A.; Zavadil, A.P. and
J.R. and Brodie, B.B.: Acetaminophen-induced hepatic Goodwin, F.K.: Hydroxylated metabolites of tricyclic anti-
necrosis. I. Role of drug metabolism. Journal of Phar- depressants: Preclinical assessment of activity. Biological Psy-
macology and Experimental Therapeutics 187: 185-194 chiatry 14: 601-611 ([ 97 9b).
([973). Prescott, L.F. and Wright, N.: The effects of hepatic and renal
Mitenko, P.A. and Ogilvie, R.I.: Pharmacokinetics of intravenous damage on paracetamol excretion following overdosage. A
theophylline. Clinical Pharmacology and Therapeutics 14: pharmacokinetic study. British Journal of Pharmacology 49:
509-513 ([973). 602-613 ([ 973).
Morgan, A.G. and Polak, A.: The excretion of salicylate in salicyl- Prescott, L.F.; Park, J.; Ballantyne, A.; Adriaenssens, P. and
ate poisoning. Clinical Science 41: 475-484 (I 9711. Proudfoot, A.T., Treatment of paracetamol (acetaminophen)
Morgan, J.D.; Rivera-Calimlim, L.; Messiha, F.; Sandaresan, P.R. poisoning with N-acetylcysteine. Lancet 2: 432-434 ([ 977).
and Trabert, N.: Imipramine-mediated interference with Prescott, L.F.; Roscoe, P. and Forrest, J.A.H.: Plasma concentra-
levodopa absorption from the gastrointestinal tract in man. tions and drug toxicity in man; in Davies and Prichard (Eds)
Neurology 25: 1029-1034 ([ 975). Biological Effects of Drugs in Relation to their Plasma Con-
Nahum, L.H. and Phillips, R.: The effect of hypothermia on centrations, pp.51-81 (Macmillan, London 1973).
digitalis intoxication. Connecticut Medicine 23: 568-572 Prescott, L.F.; Koscoe, P.; Wright, N. and Brown, S.S.: Plasma
([ 959). paracetamol half-life and hepatic necrosis in patients with
Neuvonen, P.1.; Elonen, E. and Mattila, M.1.: Orally given char- paracetamol overdosage. Lancet I: 519-522 ([ 97l).
coal increases the rate of elimination of phenobarbital, car- Proudfoot, A.T. and Wright, N.: Acute paracetamol poisoning.
bamazepine, phenylbutazone, and dapsone in man (Abstract). British Medical Journal 4: 557-558 ([ 970).
Clinical Pharmacology and Therapeutics 27: 275 ([ 980). Pruitt, A.W.; Zwiven, G.T.; Patterson, J.H.; Dayton, P.G.; Cook,
Nickander, R. and Smits, S.: Some effects of a-d-norpropoxy- C.E. and Wall, M.E.: A complex pattern of disposition of
phene: the major metabolite of a-d-propoxyphene (Darvon). phenytoin in severe intoxication. Clinical Pharmacology and
Pharmacologist 15: 203 ([ 97 3). Therapeutics 18: 112-120 ([ 975).
Nimmo, W.S.: Drugs, diseases and altered gastric emptying. Clini- Rappolt, R.T. Sr.; Gay, O.R. and Farris, R.D.: Emergency
cal Pharmacokinetics I: 189-203 ([ 976). management of acute phencyclidine intoxication. Journal of
Nimmo, W.S.; Heading, R.C.; Wilson, J. and Prescott, L.F.: the American College of Emergency Physicians 812: 68-76
Reversal of narcotic-induced delay in gastric emptying and ([ 979).
paracetamol absorption by naloxone. British Medical Journal Rawlins, M.D.; Henderson, D.B. and Hijab, A.R.: Phar-
2: 1189 ([ 979). macokinetics of paracetamol (acetaminophen) after intraven-
Nomof, N.; Elliott, H.W. and Parker, K.D.: Actions and metabol- ous and oral administration. European Journal of Clinical
ism of codeine (methylmorphine) administration by con- Pharmacology II: 283-286 ([ 977).
tinuous intravenous infusion to humans. Clinical Toxicology Reuning, R.H.; Sams, R.A. and Notari, R.E.: Role of phar-
II: 517-529 ([ 977). macokinetics in drug dosage adjustment. I. Pharmacologic
Nurmand, L.B.; Pervik, S.G. and Pyarna, R.A.: Distribution of effect kinetics and apparent volume of distribution of digoxin.
barbiturates in the organism under conditions of artificial hy- Journal of Clinical Pharmacology 13: 127-141 ([973).
pothermia. Farmakol. Toksikol. 28: 534-535 ([ 965). Richens, A.: Clinical pharmacokinetics of phenytoin. Clinical
Pharmacokinetics of Drug Overdose 191

Pharmacokinetics 4: 153-169 (1979). Slattery, J.T. and Levy, G.: Acetaminophen kinetics in acutely
Rink, R.A.; Gray, I.; Rueckart, R.R. and Slocum, H.e.: The effect poisoned patients. Clinical Pharmacology and Therapeutics
of hypothermia on morphine metabolism in an isolated per- 25: 184-195 (I 979).
fused liver. Anesthesiology 17: 377-384 (1956). Smith, T.W. and Willerson, J.T.: Suicidal and accidental digoxin
Rowland, M.: Drug administration and regimens; in Melmon and ingestion. Report of five cases with serum digoxin level cor-
Morrelli (Eds) Clinical Pharmacology, pp.25-70 (McMillan, relations. Circulation 54: 29-36 (197 n
New York 1978). Spector, S. and Vessell, E.S.: Disposition of morphine in man.
Rowland, M.; Riegelman, S. and Harris, P.A.: Absorption kinetics Science 174: 421-422 (1972).
of aspirin in man follOWing oral administration of an aqueous Spiker, D.G. and Biggs, J.T.: Tricyclic antidepressants. Prolonged
solution. Journal of Pharmaceutical Science 61: 379-385 plasma levels after overdose. Journal of the American Medical
(1972). Association 236: 1711-1712 (J 976).
Rubenstein, E. H.: Vascular responses to adrenaline, noradrenaline Stalker, N.E.; Gambertoglio, J.G.; Fukumitsu, CJ.; Naughton,
and angiotensin in hYpothermic dogs. Acta Physiologica J.L. and Benet, L.Z.: Acute massive chloral hydrate intoxica-
Latino Americana II: 33-37 (1961). tion treated with hemodialysis: a clinical pharmacokinetic
Rumack, B.H. and Peterson, R.G.: Acetaminophen overdose: inci- analysis. Journal of Clinical Pharmacology 18: 136-142
dence, diagnosis, and management in 416 patients. Pediatrics (J 978).
62 (Suppll: 898-903 (1978). Sumner, D.J.; Kalk, J. and Whiting, B.: Metabolism of barbiturate
Rumack, B.H. and Matthew, H.: Acetaminophen poisoning and after overdosage. British Medical Journal I: 335 (1975).
toxicity. Pediatrics 55: 871-876 (1975). Teehan, B.P.; Maher, J.F.; Carey, J.J.H.; Flynn, P.D. and
Rumack, B.H.; Wolfe, R.R. and Gilfrich, H.: Phenytoin Schreiner, G.E.: Acute ethchlorvynol (Placidyli!l» intoxication.
(diphenylhydantoin) treatment of massive digoxin overdose. Annals of Internal Medicine 72: 875-882 (J 970).
British Heart Journal 36: 405-408 (1974). Temple, A.R.: Pathophysiology of aspirin overdosage toxicity,
Runkel, R.; Chaplin, M.D.; Sevelius, H.; Ortega, E. and Segre, E.: with implications for management. Pediatrics 62 (Suppl.):
Pharmacokinetics of naproxen overdoses. Clinical Phar- 873-876 (1978).
macology and Therapeutics 20: 269-277 (1976). Thomsen, K. and Schou, M.: Renal lithium excretion in man.
Russell, F.E.; Carlson, R.W.; Wainschel, J. and Osborne, A.H.: American Journal of Physiology 215: 823-827 (J 968).
Snake venom poisoning in the United States. Journal of the Tozer, T.N.; Witt, L.D.; Gee, L.; Tong, T.G. and Gambertoglio,
American Medical Association 233: 341-344 (1975). J.: Evaluation of hemodialysis for ethchlorvynol (Placidyll
Schenker, S.; Hoyumpa, A.M. and Wilkinson, G.R.: The effect of overdose. American Journal of Hospital Pharmacy 31:
parenchymal liver disease on the disposition and elimination of 986-989 (1974).
sedatives and analgesics. Medical Clinics of North America Vaucher, Y.; Lightner, E.S. and Watson, P.D.: Theophylline
57: 887-896 (1975). poisoning. Journal of Pediatrics 90: 827-830 (1977).
Schou, J.; Angelo, H.; Dam, W.; Jensen, K. and Christensen, Vestal, R.E.; McGuire, E.A.; Tobin, J.D.; Andres, R.; Norris,
J.M.: Pharmacokinetics of dextropropoxyphene in acute A.H. and Mezey, E.: Aging and ethanol metabolism. Clinical
poisoning. Archives of Toxicology (Suppl.ll: 343-346 (1978). Pharmacology and Therapeutics 21: 343-354 (1977).
Schou, M.; Amidsen, A. and Trap-Jensen, J.: Lithium poisoning. von Hartitzsch, B.; Hoenich, N.A.; Leigh, RJ.; Wilkinson, R.;
American Journal of Psychiatry 125: 520-527 (1968). Frost, T.H.; Weddel, A. and Posen, G.A.: Permanent neuro-
Shreiner, G.E.; Berman, L.B.; Kovach, R. and Bloomer, H.A.: logical sequelae despite hemodialysis for lithium intoxication.
Acute glutethinide (Doviden) poisoning. The use of bemegride British Medical Journal 4: 757-759 (1972).
(Megimide) and hemodialysis. American Medical Association Waddell, W. J. and Butler, T.e.: The distribution and excretion of
Archives of Internal Medicine 101: 899-911 (1958). phenobarbital. Journal of Clinical Investigation 36:
Schwartz, H.S.: Acute meprobamate poisoning with gastrotomy 1217-1226 (1957).
and removal of a drug-containing mass. New England Journal Wagner, J.G.: Biopharmaceutics and Relevant Pharmacokinetics,
of Medicine 295: 1177-1178 (1976). p.249 (Drug Intelligence, Hamilton, l11inois 1971).
Shubin, H. and Wei!, M.H.: The mechanism of shock following Walle, T.; Conradi, E.C; Wal)e, U.K.; Fagan, T.C and Gaffney,
suicidal doses of barbiturate, narcotics and tranquilizing drugs. T.E.: The predictable relationship between plasma levels and
with observations on the effects of treatment. American Jour- dose during chronic propranolol therapy. Clinical Phar-
nal of Medicine 38: 853-863 (I965). macoogy and Therapeutics 24: 668-677 (I 978).
Sj6qvist, F.; Bergfors, P.G.; Borga, 0.; Lind, M. and Ygge, H.: Weinberger, M. and Ginchansky, E.: Dose-dependent kinetics of
Plasma disappearance of nortriptyline in a newborn infant theophylline disposition in asthmatic children. Journal of
following placental transfer from an intoxicated mother: Evi- Pediatrics 91: 820-824 (I 977).
dence for drug metabolism. Journal of Pediatrics 80: 496-500 Westervelt, F.B.: Ethchlorvynol (Placidyli!l» intoxication: ex-
(1972). perience with five patients, including treatment with
Pharmacokinetics of Drug Overdose 192

hemodialysis. Annals of Internal Medicine 64: 1229-1236 Wolen. R.L.; Ziege. E.A. and Gruber. C.M.: Determination of
(1966). propoxyphene and norpropoxyphene by chemical ionization
Wilder. BJ.; Buchanan. R.A. and Serrano. E.E.: Correlation of mass fragmentography. Clinical Pharmacology and Thera-
acute diphenylhydantoin intoxication with plasma levels and peutics 17: 15-20 (1975).
metabolite excretion. Neurology 23: 1329-1332 (1973). Wraae. 0.: The pharmacokinetics of lithium in the brain.
Wilkinson. P.K.; Sedman. AJ.; Sakmar. B.S.; Earhart. R.H.:. cerebrospinal fluid and serum of the rat. British Journal of
Weidler. DJ. and Wagner. J.G.: Blood ethanol concentrations Pharmacology 64: 273-279 (1978).
during and following constant-rate intravenous infusion of Wright, N. and Prescott, L.F.: Potentiation by previous drug ther-
alcohol. Clinical Pharmacology and Therapeutics 19: 213-223 apy of hepatotoxicity following paracetamol overdosage. Scot-
(1976). tish Medical Journal 18: 56-58 (1973).
Williams. R. and Davis. M.: Pathology and biochemistry of Wright, N. and Roscoe, P.: Acute glutethimide poisoning: conser-
paracetamol-induced liver damage; in Duncan and Leonard vative management of 31 patients. Journal of the American
(Eds) Proceedings of the European SOCiety of Toxicology; Medical Association 214: 1704-1706 (t 970).
Clinical Toxicology. Inteinational Congress Series No. 417. Yacobi, A. and Levy, G.: Intra individual relationships between
Vol. 18. pp.27-40 (Excerpta Medica. Amsterdam 1977). serum protein binding of drugs in normal human subjects,
Wilson. A.E. and Domino, E.F.: Plasma phencyclidine phar- patients with impaired renal function, and rats. Journal of
macokinetics in dog and monkey using a gas chromatography Pharmaceutical Science 66: 1285-1288 (977).
selected ion monitoring assay. Biomedical Mass Spectrometry Zwillich, C.W.; Sutton, F.D.; Neff, T.A.; Cohn, W.M.; Matthay,
5: I 12-116 (1978). R.A. and Weinberger, M.M.: Theophylline-induced seizures
Wilson. J.H.P.; Donker. AJ.M.; Van der Hem. G.K. and in adults. Annals of Internal ~1edicine 82: 784-787 (1975).
Wientjes, J.: Peritoneal dialysis for lithium poisoning. British
Medical Journal 2: 749-750 (1971).
Wilson. J.T.; Huff. J.G. and Kilroy. A.W.: Prolonged toxicity
following acute phenytoin overdose in a child. Journal of
Pediatrics 95: 135-138 (1979).
Wislicki. L.: Effects of hypothermia and hyperthermia on the ac-
tion of neuromuscular blocking agents. I. Suxamethonium. Author's address: Dr Neal L. Benowitz. San Francisco General
Archives of International Pharmacodynamics 126: 68-78 Hospital 5H6, 1001 Potrero Avenue, San Francisco, California
(1960). 94110 (USA).

Vous aimerez peut-être aussi