Clin Liver Dis 11 (2007) 525–548

Acetaminophen Hepatotoxicity Anne M. Larson, MD, FACP, AGAF Division of Gastroenterology, Hepatology Section, University of Washington, 1959 NE Pacific Street, Box 356174, Seattle, WA 98195-6174, USA

Acetaminophen (N-acetyl-p-aminophenol or APAP), a mild nonnarcotic analgesic and antipyretic agent, is widely used as a pain reliever and fever reducer. It is available in hundreds of single-ingredient and combination over-the-counter (OTC) products, and also in numerous prescription products. Its use has achieved great popularity, with more than 1 billion tablets sold annually in the United States [1]. APAP was first introduced for prescription use in 1955, and subsequently approved by the Food and Drug Administration (FDA) in 1960 as an OTC 325-mg immediate-release tablet [2]. A 500-mg immediate-release capsule and tablet were approved in 1973 and 1975, respectively [2]. Based on available literature, the FDA determined that acetaminophen was generally safe and effective, and that the maximum safe daily dosage should be no greater than 4 g in a 24-hour period. APAP is effective and safe when consumed as recommended (1–4 g per day). By 1966, however, there were reports of fatal and nonfatal hepatic necrosis following large suicidal ingestions [3–5]. It is now well documented that ingestion of a single dose of more than 10 to 15 g can cause severe or fatal liver injury secondary to massive hepatic necrosis [6]. There are emerging data that ingestion of daily doses of less than 10 g on consecutive days carries a risk for liver injury in some individuals [7]. It has also been suggested that APAP hepatotoxicity may occur in those ingesting therapeutic doses under certain conditions, such as fasting or alcohol use (see later discussion) [8–15]. APAP may even cause transient asymptomatic elevation in serum aminotransferases at therapeutic doses [16,17].

Epidemiology The American Association of Poison Control Centers notes that APAPrelated calls make up about 10% of all calls to poison control centers. E-mail address: [email protected] 1089-3261/07/$ - see front matter Ó 2007 Elsevier Inc. All rights reserved. doi:10.1016/j.cld.2007.06.006 liver.theclinics.com

526

LARSON

Between 1995 and 1999, APAP was the leading cause of poisoning in the United States [2,7,18]. Annually during this time period there were more than 56,000 emergency visits for APAP poisoning (intentional and unintentional), leading to more than 26,000 hospitalizations and about 458 deaths. Of these, unintentional overdoses accounted for more than 13,000 emergency visits with more than 2,000 hospitalizations and 100 deaths [18]. Although a decrease in calls was seen between 1995 and 1999 (111,175 to 108,102), there were 133,125 APAP-related calls generated in 2004 [19]. The proportion of APAP-induced acute liver failure (ALF) cases has alarmingly increased between 1998 (21%) and 2003 (51%) [20]. About half of these cases were unintentional poisonings [19,20]. APAP poisoning remains is a significant concern in other countries also [21–23].

Drug metabolism General Exogenous products are hepatically metabolized predominantly by two mechanisms (Fig. 1) [24]. During phase 1 metabolism, polar groups are added to the molecule being processed by way of oxidation, reduction, or hydrolysis. These reactions are catalyzed by the cytochrome P450 system of mixed function oxidases (CYP), a family of membrane-bound hemoproteins located in the smooth endoplasmic reticulum of the centrilobular (zone 3) hepatocytes. There are numerous CYPs that participate in the metabolism of exogenous compounds. Although many drugs are converted to active compounds by this process, toxic byproducts, such as free radicals, may also be formed [25]. During phase 2 metabolism, molecules are conjugated with glucuronic acid, sulfates, or glutathione (GSH) by way of the UDPglucuronyl transferases, sulfotransferases, and glutathione S-transferases.

Phase I

+

H

NADP

CYP

NADP

enz

yme

s

Toxic Metabolite

Hepatotoxicity

Stable Metabolite

Drug

Phase II

Glutathione Glucuronide Tra Methyl ns fer Sulfate as es Conjugated Drug

Excretion

Fig. 1. Drug metabolism. Phase I reactions are catalyzed by the cytochrome P450s. Phase II reactions are catalyzed by one of several enzymes, such as UDP-glucuronyl-transferase, sulfotransferases, and glutathione S-transferases. The toxic metabolites that are formed can lead to hepatic injury.

ACETAMINOPHEN HEPATOTOXICITY

527

These enzymes are rarely responsible for toxic metabolite formation and their nontoxic products are generally ready for excretion [26]. Acetaminophen metabolism About 2% of APAP is excreted in the urine unchanged [27–30]. More than 90% is metabolized by way of conjugationdtwo thirds through glucuronidation (UDP-glucuronosyltransferases) and one third through sulfation (sulfotransferases) (Fig. 2) [29–31]. The nontoxic inactive conjugates are largely excreted in the urine and bile. Approximately 5% to 9% undergoes oxidative conversion by way of the CYPs (CYP1A2, CYP2A6, CYP2E1, CYP3A4) to the toxic metabolite N-acetyl-p-benzoquinone imine (NAPQI) [9,30,32–35]. CYP2E1 is the major source of NAPQI, but with less contribution from the remaining enzymes [30,36].

Fig. 2. Metabolism of acetaminophen. Most APAP is conjugated to either glucuronide or sulfate. That portion which is oxidized to NAPQI is further detoxified by glutathione transferase. If this system is overwhelmed, NAPQI binds to cellular targets leading to hepatocellular necrosis. (From Zimmerman HJ. Acetaminophen hepatotoxicity. Clin Liver Dis 1998;2:527; with permission.)

528

LARSON

NAPQI is a highly reactive two-electron species that can act as an electrophile or an oxidant. It is rapidly metabolized by conjugation to intracellular GSH forming a nontoxic APAP–GSH conjugate (3-[glutathioneS-yl]-APAP) [28,37,38]. Further processing leads to its urinary excretion as mercapturic acid and cysteine conjugates [27]. APAP is also oxidized by myeloperoxidase and the peroxidase function of cyclooxygenase-1 (COX-1), the clinical significance of which is unknown [27,39–41].

Acetaminophen hepatotoxicity APAP is an established dose-related hepatotoxin and can potentially lead to hepatocellular injury by three different mechanisms, individually or in combination. The most common method of toxicity is overdose (ingestion of more than 10 to 15 g in adults and 150 mg/kg in children), which overwhelms the hepatic detoxification process [42]. There are reports of toxicity occurring with doses less than 10 g and even at therapeutic doses [7,20,43–45]. Excessive CYP activation as a result of other medications or herbs may lead to increased free radical formation with the possibility of hepatocellular injury [46–48]. Depletion of GSH stores (by overdose, malnutrition [49,50], or alcohol ingestion [9,51]) results in the inability to detoxify many reactive metabolites and may increase risk for hepatotoxicity. The importance of this mechanism remains controversial [52,53]. The precise means of hepatocyte death remains speculative. The degree of hepatic toxicity correlates with the activity of the catalyzing enzyme systems and GSH availability. Therapeutic doses of APAP result in small quantities of NAPQI that are easily detoxified by GSH. Excessive doses of APAP lead to the saturation of the glucuronic acid or sulfate pathways, shunting more APAP into the CYP system. Increased metabolism of APAP by CYP2E1, in turn, increases the amount of NAPQI produced [54]. Glutathione stores within the liver are limited and may be depleted in an attempt to keep up with the increased NAPQI levels [37]. When GSH stores are reduced by 70% to 80%, the detoxification capacity of the liver is exceeded and NAPQI accumulates, thereby interacting with and destroying hepatocytes and other cells [55–57]. In the absence of GSH, covalent binding of NAPQI to the cysteine groups on hepatocyte macromolecules occurs forming NAPQI-protein adducts [58–60]. This process is the initial and irreversible step in the development of cell injury [61–63]. Glutathione depletion further contributes to cellular oxidant stress [27,64]. With NAPQI binding to critical cellular targets, such as mitochondrial proteins, mitochondrial dysfunction and loss of cellular ATP occurs [65,66]. The hepatocytes subsequently experience overall energy failure [67–70]. The ultimate result is alteration in calcium homeostasis, mitochondrial dysfunction with ATP depletion, DNA damage, and intracellular protein modification [28,62,69,71]. These events lead to necrotic cell death.

ACETAMINOPHEN HEPATOTOXICITY

529

It has been shown in rodents, however, that covalent binding to hepatic proteins in isolation does not cause hepatotoxicity [67,72,73]. There is a growing body of evidence suggesting that inflammatory mediators (eg, IFNg) trigger the innate immune system, leading to influx of NK/NKT cells, neutrophils, Kupffer cells, and macrophages that participate in the development and propagation of hepatocyte injury [74–77]. Controversy remains as to whether neutrophils aggravate this process [78,79]. It has therefore been proposed that acetaminophen toxicity occurs in two phases: a metabolic phase and an oxidative phase [80]. Factors influencing acetaminophen hepatotoxicity Age The metabolism of APAP is age dependent [81]. Infants and children process APAP predominantly by way of sulfation. Metabolism by glucuronidation increases over time and becomes the predominant pathway between ages 5 and 12 years [81,82]. Children less than 5 years of age seem to be less susceptible to APAP hepatotoxicity, possibly because of lower production of NAPQI and a greater degree of conjugation [8,81,83–85]. Overdose in juveniles and older populations may have an increased incidence of hepatotoxicity and hepatotoxicity may occur at lower doses [86–89]. Genetics Polymorphisms exist in the cytochrome isoenzymes that may contribute to diminished metabolism, lack of metabolism, or excessive metabolism of a compound [90]. The clinical relevance of these polymorphisms in the setting of APAP ingestion is unknown. Chronic liver disease Patients who have chronic liver disease who do not regularly consume alcohol are not at increased risk for APAP-induced liver injury [15,91]. CYP activity in patients who have severe liver disease is unchanged or reduced and does not seem to be inducible [91–93]. Data are mixed as to whether GSH is reduced or increased in the setting of chronic liver disease [91]. Alcohol (therapeutic misadventure) The role of alcohol in the generation of APAP hepatotoxicity remains contentious [38,52,94,95]. Acute alcohol intake may actually be protective because the alcohol competes with APAP for CYP [10,96–100]. Chronic alcohol ingestion modifies CYP2E1 activity and depletes GSH levels [52,101]. Chronic alcohol ingestion increases CYP2E1 activity twofold, an effect that lasts for up to 10 days following abstinence [12,102–104]. Alcohol also inhibits the rate of GSH synthesis leading to accelerated GSH turnover and it impairs mitochondrial transport of GSH (with sequestration within the mitochondria) [105]. Both of these features would suggest that

530

LARSON

hepatotoxicity in this setting could occur with lesser amounts of APAP. Poor nutrition in alcoholics could also contribute to decreased GSH [9,106,107]. Chronic alcoholics do not seem to be at increased risk for toxicity following a single time-point overdose, particularly if they are treated within 8 hours or are below the probable toxicity line on the Rumack-Matthew nomogram (Fig. 3 and later discussion) [108–110]. Smaller doses, however, used with therapeutic intent for pain complaints, have been reported to result in severe or even fatal disease in chronic drinkers [9,11–13,51]. Other investigators have not found an association between alcohol use and APAP hepatotoxicity [111,112]. Nevertheless, the FDA has issued an alcohol warning to patients taking APAP if they are using more than three drinks a day [113]. Drugs The evidence for a significant clinical impact of drug-induced CYP induction on APAP hepatotoxicity remains incomplete [114]. Anticonvulsants and antituberculous medications induce the CYPs [46,115,116]. Isoniazid and halothane can enhance CYP2E1 [117]. Phenytoin, carbamazepine, and phenobarbital induce CYP3A4 and increase production of NAPQI [46,118–120]. Phenytoin and phenobarbital have also been shown to inhibit APAP glucuronidation in human hepatocytes [121]. Zidovudine and trimethoprim-

Fig. 3. Rumack-Matthew nomogram. This nomogram shows the relationship between plasma acetaminophen concentration, time after drug ingestion, and the risk for hepatotoxicity. The thick diagonal line of possible hepatotoxicity represents a 25% likelihood of disease. (Adapted from Rumack BH, Matthews H. Acetaminophen poisoning and toxicity. Pediatrics 1975;55:873; with permission.)

ACETAMINOPHEN HEPATOTOXICITY

531

sulfamethoxazole lead to impaired glucuronidation by competing for glucuronyl transferase, shunting more drug to CYP2E1 [122]. Cimetidine, an inhibitor of CYP2E1 [123] in animals, may decrease the risk for toxicity; however, human studies have not supported this [124,125]. Patients should be questioned about use of herbal supplements and complementary medication use [126]. Many of these drugs are hepatotoxic themselves (eg, INH), and few clinically significant drug–APAP interactions have been well documented. Nutritional status Fasting, which contributes to depletion of GSH and induction of CYP2E1, has been reported to enhance APAP toxicity, particularly in the setting of chronic alcohol consumption [9,127,128]. Fasting also leads to glycogen depletion with decreased glucuronidation, in turn causing enhanced production of toxic metabolites [9,12]. In a mouse model of nonalcoholic steatohepatitis, hepatic steatosis inhibited CYP2E1 induction and protected against APAP-induced liver injury [129]. Both would suggest that nutritional status contributes to APAP hepatotoxicity. Recent reviews, however, have concluded that there is insufficient evidence to support the theory of malnutrition and increased toxicity [53,130]. Tobacco use Tobacco smoke contains several potent inducers of CYP1A2 and has been shown to induce oxidative metabolism [131–133]. A retrospective report found tobacco use to be an independent risk factor for mortality following APAP poisoning (odds ratio 3.64; 1.23–10.75) [134]. The risk did not depend on the quantity of tobacco consumed. Mortality was even greater in those smokers also consuming alcohol [134]. Autoprotection Despite the well-known dose-related relationship to hepatotoxicity, there are individuals who report chronically ingesting ‘‘toxic’’ doses of APAP without ill effect [135]. These patients are most often consuming combination narcotic/APAP products for pain relief and have consumed gradually increasing quantities of the product over time [136–138]. The mechanism of this tolerance is unclear. The phenomenon has also been demonstrated in rodents [138].

Clinical presentation Clinical stages Several distinct stages occur following excessive APAP ingestion. The first 24 hours following ingestion (stage 1) are characterized by nonspecific gastrointestinal irritation with nausea, vomiting, abdominal pain, anorexia,

532

LARSON

lethargy, malaise, and diaphoresis [96]. Pallor and mild hepatic tenderness may be present on physical examination [54,139]. If measured, the laboratory studies are typically normal, but mild liver enzyme elevations may be seen as early as 8 to 12 hours following ingestion [140]. Patients may not present for medical care, and these early symptoms usually subside over the course of this period. If seen, patients may be mistakenly considered recovered. The latent phase (stage 2) develops over the next 24 to 72 hours following ingestion. Patients often appear well, but subclinical and biochemical evidence of hepatotoxicity begins to appear. The serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) start to increase. All patients who have hepatotoxicity develop elevated aminotransferases within 24 to 36 hours [140]. Patients may develop right upper quadrant pain and notice jaundice. Elevations in total bilirubin and prothrombin time (PT/ INR) occur. Nephrotoxicity and oliguria may develop during this stage. The hepatic stage (stage 3) develops 72 to 96 hours after ingestion. It is the phase of overt hepatocellular necrosis and death most commonly occurs during this time period [141]. Signs and symptoms vary depending on the severity of the liver injury, and if treated early this stage is minimal. Anorexia, nausea, vomiting, abdominal pain, malaise, and confusion (hepatic encephalopathy) may develop. Jaundice may appear or worsen and serum AST and ALT are severely elevated. The AST can be 10,000 IU/L or higher, and may be more elevated than the ALT. Levels up to 48,000 IU/L have been reported. The degree of aminotransferase elevation corresponds roughly to the degree of centrilobular hepatocellular damage. Elevation of the AST and ALT usually peaks by 72 hours after acute ingestion. Renal insufficiency occurs in more than 50% and is generally attributable to acetaminophen-induced acute tubular necrosis with a component of dehydration [139,142,143]. The exact mechanism of renal cellular injury is uncertain. Clinically, proteinuria, hematuria, and granular casts are seen [142]. Renal injury is unrelated to the degree of liver injury [144]. Hypophosphatemia secondary to renal phosphate loss may develop, even in the absence of liver damage. Other metabolic derangements may accompany this stage. Pancreatitis has been reported [145]. Hypoglycemia is a poor prognostic sign and is attributable to the damaged liver’s inability to mobilize glycogen stores and perform gluconeogenesis. There are also inappropriately high insulin levels seen [146]. Lactic acidosis usually presents more than 1 to 2 days after ingestion and is secondary to tissue hypoxia with poor hepatic clearance of lactate [147,148]. A PT/INR that continues to increase after days 3 to 4 is ominous and can be associated with up to 93% mortality [149,150]. Patients may develop progressive CNS symptoms of lethargy, confusion, and coma, and may require intubation. As encephalopathy progresses, cerebral edema develops and may result in cerebral herniation. Indicators of a poor outcome include development of acute liver failure, PT greater than 100 seconds, grade 3 or 4 encephalopathy, cerebral edema, renal failure, and metabolic acidosis (pH!7.3) (see later discussion). Death

ACETAMINOPHEN HEPATOTOXICITY

533

resulting from cerebral edema, multiorgan-system organ failure, or sepsis generally occurs 3 to 5 days following ingestion. Approximately 70% of patients survive acute liver failure and enter the recovery stage (stage 4) by 4 days to 2 weeks after ingestion. Most patients recover completely by 7 days, but recovery may be slower in those who were more severely ill [110]. Complete functional recovery can be expected. Differential diagnosis Although the history often leads to an accurate diagnosis, other causes of hepatotoxicity and ALF must be considered. Acute APAP hepatotoxicity is characterized by marked elevations in the aminotransferases, usually more than 3,000 IU/L. Other diagnoses to be considered include viral hepatitis, drug- or toxin-induced hepatitis, Reye syndrome, acute fatty liver of pregnancy, and ischemic hepatitis (‘‘shock liver’’). Alcoholic hepatitis and acute deterioration of chronic liver disease should be considered, but the aminotransferases are generally more modestly elevated (usually !500 IU/L). Chronic acetaminophen toxicity in a chronic alcohol user, however, may also present with markedly elevated enzymes. Histopathology Zone 3 (centrilobular) hepatocellular necrosisdthe classic hepatotoxic lesiondis characteristic of APAP injury. The centrilobular region is the area of greatest concentration of CYP2E1, and therefore the site of maximal production of NAPQI [151]. Also present may be passive congestion and scattered mononuclear and polynuclear lymphocytes. Once the patient recovers clinically, histologic improvement may take up to 3 months. Permanent liver damage is rare if the patient survives, with complete restoration of hepatic architecture the rule [152].

Treatment Assessment of overdose Prompt recognition of APAP intoxication is imperative to management and prevention of hepatocellular injury, liver failure, and death. Identifying patients who have intentionally taken substantial amounts of APAP is often less problematic than identifying those who have an unintentional overdose. The history of the amount of APAP ingested in both cases, however, is often unreliable. Patients may be unwilling or unable to admit to the amount or timing of the overdose. The vague or symptom-free latent phase may further confuse the clinical picture. Careful attention must be paid to the type of product ingested, quantity of APAP used, intent of the ingestion (intentional, unintentional), pattern of ingestion (single dose, repeated dosing), and other medications, drugs, herbs, or toxins consumed.

534

LARSON

All patients suspected of APAP ingestion should undergo prompt measurement of serum APAP concentration. APAP is rapidly absorbed from the gastrointestinal tract within 30 to 60 minutes and reaches peak plasma values within 1 to 3 hours [29]. Ideally, plasma APAP levels should be obtained between 4 and 24 hours following ingestion. A negative value does not preclude a significant ingestion and false positive levels have been seen in patients who have hyperbilirubinemia (O10 mg/mL) [153–155]. NAPQI-protein adducts are released into the circulation following hepatic injury and can be measured by high-performance liquid chromatography [156]. The serum concentration of these adducts correlates positively with serum AST activity [156]. Adducts can be detected even in the setting of negative serum APAP levels. Adducts have been identified in some patients who have indeterminate ALF and no reported history of APAP ingestion, suggesting that these cases were missed APAP overdoses [157]. Because measurement of serum NAPQI-protein adducts reliably identified acetaminophen toxicity, they may be a useful diagnostic test for cases lacking historical data or other clinical information. Unfortunately, their measurement is not routinely available in most centers. There is no correlation between the dose of APAP reportedly ingested and the serum APAP concentration measured [158,159]. The most widely accepted approach to determine the risk for hepatotoxicity following acute single time-point ingestion is to plot the serum APAP concentration on the Rumack-Matthew nomogram (see Fig. 3). The original nomogram is a semilogarithmic graph of serum APAP concentration versus time from ingestion [42]. Patients whose APAP concentration exceeds the lowest plotted line postingestion have a probable risk for hepatotoxicity. The FDA required a lower safety line (25% lower than the original) be used to allow for errors in plasma assays and estimated ingestion time [53,160]. Patients who have levels above this line are considered to be at possible risk for hepatotoxicity, and it is this nomogram that is most commonly used in the United States [160]. Treatment is recommended if serum levels of APAP exceed this concentration [96]. Controversy remains, however, over which line to use [161]. The nomogram was developed for single time-point overdoses. It cannot be used to accurately predict risk for hepatotoxicity if patients have taken multiple large doses over time, if the precise time of ingestion is unknown, or if patients have been taking an extended-release product [162]. These patients represent a significant portion of APAP overdoses, particularly in the United States [19,20,163]. There have been attempts to develop treatment algorithms in these settings [164,165]. Management APAP hepatotoxicity is a potentially treatable illness. In fact, serious hepatotoxicity is rare if the antidote, N-acetylcysteine (NAC, Mucomyst, Acetadote) can be administered within 8 to 10 hours following an acute

ACETAMINOPHEN HEPATOTOXICITY

535

overdose, regardless of the initial APAP concentration [160,166–168]. Once signs of ALF (eg, encephalopathy, coagulopathy) have developed, however, the risk for complications and death increase significantly [109]. If the patient presents within 4 hours following an acute ingestion, gastric lavage may be useful in decreasing the pill load [169]. During this time period, activated charcoal reduces APAP absorption by 50% to 90% in simulated overdose, and decreases serum APAP concentration [170,171]. It can be administered in a single dose of 1 g per kilogram body weight. Activated charcoal also reduces NAC absorption somewhat; however, this has not been shown to decrease its overall effectiveness [172]. NAC is a glutathione precursor and was first used for the treatment of APAP poisoning in 1977 [173,174]. Its mechanism of action is threefold. It increases glutathione stores, it acts as a glutathione substitute that binds with NAPQI, and it enhances sulfate conjugation [28,175–179]. NAC also has anti-inflammatory, antioxidant, inotropic, and vasodilating effects that may further benefit the patient [180–183]. N-acetylcysteine is the treatment of choice following an APAP overdose. It is indicated when any of the following conditions apply [184]:  Serum APAP concentration following acute ingestion is above the ‘‘possible hepatic toxicity’’ line on the Rumack-Matthew nomogram  Estimated single ingestion of greater than 150 mg/kg (7.5 g) in an adult, or there is a delay in acquiring a serum APAP concentration  Time of ingestion is not known and the serum APAP level is greater than 10 mg/mL  Laboratory evidence of any hepatotoxicity and a history of supratherapeutic APAP ingestion  Repeated supratherapeutic ingestion and a serum APAP concentration greater than 10 mg/mL It is best to err on the side of treating when not all the factors are clear, because NAC is relatively safe. Treatment should be started within 8 to 10 hours of an acute APAP overdose, but it is still indicated as late as 24 hours after ingestion [109,160,168,180]. In patients who develop no evidence of liver injury, NAC may be safely discontinued [185–188]. Current evidence suggests that even late administration of NAC is beneficial to patients who develop ALF [180–183]. In these patients, NAC treatment should be continued until clinical improvement occurs and the INR is less than 2.0 [34,182]. Oral administration The standard of care is a promptly started 72-hour course of oral NAC. This medication is given as a loading dose of 140 mg/kg followed by a maintenance dose of 70 mg/kg every 4 hours for a total of 17 doses (total dose 1330 mg/kg) [160]. If a dose is lost because of emesis, it must be repeated. If the 4-hour plasma APAP concentration is below the toxic level, further maintenance doses can be discontinued [160].

536

LARSON

Intravenous administration Intravenous NAC was first available in Europe and Canada, and approved for use in the United States in 2004 [182,184,189]. Its use is recommended in the following situations [184]:  Patients who cannot tolerate oral NAC because of intractable vomiting and for whom further delay would result in decreased NAC efficacy  Patients who cannot use enteral NAC (eg, corrosive ingestion, gastrointestinal bleeding, obstruction, or perforation)  Patients who have ALF, because intravenous NAC results in higher serum concentrations and has been shown to be beneficial in this setting [190]  Patients who are pregnant. The higher serum concentrations achieved may facilitate transplacental delivery to the fetus. The superiority of intravenous to oral NAC in pregnancy is theoretical, however. Placental transfer of NAC has not been shown to occur in animal studies [191]. The loading dose consists of 150 mg/kg over 15 minutes, followed by 50 mg/kg over the next 4 hours and 100 mg/kg over the 16 hours thereafter. The total dose received is 300 mg/kg over 20 hours. This dosing regimen remains controversial, and others have suggested a higher loading dose with 140 mg/kg followed by 70 mg/kg every 4 hours for 12 doses [192]. Oral and intravenous regimens show similar efficacy when started within 10 hours of acetaminophen overdose [160,166,192,193]. When treatment is delayed beyond 10 hours, however, the oral regimen may be more effective, predominantly because of its larger total dose (1330 versus 300 mg/kg) [160,166,167,192,194]. Oral NAC has a bad taste and pungent odor (rotten eggs). Its predominant side effects include nausea, diarrhea, and rash. Vomiting is common. Nausea and vomiting can be minimized by diluting the NAC solution with juice or soda, holding one’s breath while taking the medication, or nasogastric tube administration. Adverse events following infusion of intravenous NAC range from 0.2% to 21% and include nausea, flushing, rash, pruritus, bronchospasm, rhinorrhea, hemolysis, fevers, chills, angioedema, hypotension, and anaphylaxis [184,187,194–200]. Many of these side effects occur when the drug is used endotracheally as a mucolytic, and asthmatics may be at particular risk [200]. Most of the reactions are mild and resolve with discontinuation of the infusion. Occasionally, patients require further treatment (ie, antihistamine or epinephrine), and rare deaths have been reported [196,197]. Slowing the infusion rate of the initial dose does not decrease efficacy or lower the incidence of adverse events [168]. Cimetidine inhibits cytochrome P-450 and theoretically would be believed to decrease NAPQI formation; however, its use as an adjunct to NAC has shown no benefit [124,125]. Hemodialysis seems to have limited efficacy and does not prevent hepatotoxicity. Further care remains supportive.

ACETAMINOPHEN HEPATOTOXICITY

537

There has been a recent suggestion that patients who have APAP poisoning and lack of significant symptoms could be discharged home with instructions to self-administer NAC [201]. In the current climate of American health care, this may become more commonplace. Careful follow-up would be essential, however. Treatment of the therapeutic misadventure is more difficult. Patients who have been ingesting APAP over a period of time often present with fully developed liver injury or acute liver failure. Acute liver failure Patients who develop ALF should be managed in specialty centers where liver transplantation is available. Improved survival has been demonstrated in patients being managed at these centers [109]. NAC should be administered until transplantation or recovery, evidenced by an INR less than 2.0 [199,202]. It is inadvisable to use fresh frozen plasma to treat the coagulopathy, because the INR is an important prognostic indicator of patient status. Use of plasma is recommended only for line placement and active bleeding. Mortality More than 90% of all patients who have APAP poisoning can be expected to recover completely [109]. Patients at risk for hepatotoxicity and who are not treated with NAC have an overall mortality rate of 5.3% to 24% [32,166] and greater than 50% in those falling into the high risk category. The mortality rate is generally less than 1% if NAC is given in a timely fashion [160]. In patients who develop ALF who do not receive life-saving transplant, mortality approaches 30% [20,149,163]. Liver transplantation Orthotopic liver transplantation (OLT) is indicated in those who progress to severe ALF. APAP poisoning is the leading cause of drug-induced liver failure in the United States requiring OLT [163]. Because a significant number of patients survive without transplantation, however, it becomes necessary to determine who will ultimately die without transplantation. Prognostic factors have been identified that facilitate decision-making with regard to necessity of liver transplantation. Concern has been raised regarding support after transplantation and the risk that those who make suicide attempts may repeat the process, even after a transplant. Some centers rarely or never offer transplantation for acetaminophen poisoning for this reason. One of the most widely used prognostic models was developed at King’s College in London [109,203–205]. Because those who have APAP hepatotoxicity have a more favorable outcome, their prognostic model was divided accordingly (Fig. 4) [203]. Ninety percent of patients who had APAPinduced ALF whose blood pH failed to correct to greater than 7.3 with

538

LARSON

Acetaminophen-Induced ALF • Arterial pH <7.3 (irrespective of the grade of encephalopathy) OR • Grade III or IV encephalopathy AND • Prothrombin time >100 seconds AND • Serum creatinine >3.4 mg/dL (301 µmol/L)

All Other Causes of ALF • Prothrombin time >100 seconds (irrespective of the grade of encephalopathy) OR Any three of the following (irrespective of the grade of encephalopathy) 1. Age <10 years or >40 years 2. Etiology: non-A, non-B hepatitis, halothane hepatitis, idiosyncratic drug reactions 3. Duration of jaundice before onset of encephalopathy >7 days 4. Prothrombin time >50 seconds 5. Serum bilirubin >18 mg/dL (308 µmol/L)

Fig. 4. King’s College Criteria: criteria predictive of a poor outcome in the setting of hepatotoxicity from acetaminophen and from all other causes. (Adapted from O’Grady JG, Alexander GJM, Hayllar KM, et al. Early indicators of prognosis in fulminant hepatic failure. Gastroenterology 1989;97:439.)

resuscitation perished without transplantation. For those who met the second set of three criteria, a mortality rate of 81% without transplantation was seen. These criteria have been validated in other cohorts [109]. In addition, it has been shown that patients who develop cerebral edema carry a mortality rate of greater than 90% [109]. The PT/INR is also a useful prognostic indicator of mortality in APAP-induced ALF, particularly if it continues to increase after day four [150]. Others have supported the measurement of coagulation factor V as the best prognostic indicator of survival [206,207]; however, this has generally been viewed as less accurate than the King’s College criteria [206], and its use remains controversial.

Summary Acetaminophen is a safe and effective medication, and given the vast amount consumed it carries a relatively low rate of hepatotoxicity. It can be used safely in patients who have chronic liver disease, although the maximum safe dose remains unclear. The complete mechanism of hepatocellular injury and cell death remains to be defined. Misuse of the product is the most frequent cause of hepatotoxicity. That nearly half of overdoses in the United States are unintentional is of significant concern, and patients should be educated regarding the potential for liver injury. Determining which patients are at risk for severe hepatotoxicity remains difficult and the development of assays for NAPQI-protein adducts may aid in identifying these individuals. Prompt medical intervention minimizes liver injury

ACETAMINOPHEN HEPATOTOXICITY

539

and death. Liver transplantation remains a viable option for those who develop acute liver failure. References [1] Nourja P, Willey M. Epidemiology of acetaminophen-related overdose. Dept of health and human services, center for drug evaluation and research, food and drug administration. FDA 2002; Available at: http://www.fda.gov/. Accessed February 26, 2007. [2] FDA Background information for September 19–20, 2002, NDAC Meeting. FDA 2002; Available at: http://www.fda.gov/ohrms/dockets/ac/02/briefing/3882b1.htm. Accessed June 26, 2007. [3] Thomson JS, Prescott LF. Liver damage and impaired glucose tolerance after paracetamol overdosage. Br Med J 1966;2(5512):506–7. [4] Davidson DG, Eastham WN. Acute liver necrosis following overdose of paracetamol. Br Med J 1966;2(5512):497–9. [5] Boyd EM, Bereczky GM. Liver necrosis from paracetamol. Br J Pharmacol Chemother 1966;26(3):606–14. [6] McJunkin B, Barwick KW, Little WC, et al. Fatal massive hepatic necrosis following acetaminophen overdose. JAMA 1976;236(16):1874–5. [7] NDAC meeting Sept. 19–20, 2002 transcript. FDA 2007; Available at: http://www.fda.gov/ ohrms/dockets/ac/02/transcripts/3882T1.htm. AND http://www.fda.gov/ohrms/dockets/ ac/02/transcripts/3882T2.htm. Accessed June 26, 2007. [8] Zimmerman HJ. Effects of aspirin and acetaminophen on the liver. Arch Intern Med 1981; 141(3 spec no):333–42. [9] Whitcomb DC, Block GD. Association of acetaminophen hepatotoxicity with fasting and ethanol use. JAMA 1994;272(23):1845–50. [10] Slattery JT, Nelson SD, Thummel KE. The complex interaction between ethanol and acetaminophen. Clin Pharmacol Ther 1996;60(3):241–6. [11] Seeff LB, Cuccherini BA, Zimmerman HJ, et al. Acetaminophen hepatotoxicity in alcoholics. A therapeutic misadventure. Ann Intern Med 1986;104(3):399–404. [12] Zimmerman HJ, Maddrey WC. Acetaminophen (paracetamol) hepatotoxicity with regular intake of alcohol: analysis of instances of therapeutic misadventure. Hepatology 1995; 22(3):767–73. [13] Maddrey WC. Hepatic effects of acetaminophen. Enhanced toxicity in alcoholics. J Clin Gastroenterol 1987;9(2):180–5. [14] Schiodt FV, Rochling FA, Casey DL, et al. Acetaminophen toxicity in an urban county hospital. N Engl J Med 1997;337(16):1112–7. [15] Benson GD. Hepatotoxicity following the therapeutic use of antipyretic analgesics. Am J Med 1983;75(5A):85–93. [16] Watkins PB, Kaplowitz N, Slattery JT, et al. Aminotransferase elevations in healthy adults receiving 4 grams of acetaminophen daily: a randomized controlled trial. JAMA 2006; 296(1):87–93. [17] Kuffner EK, Temple AR, Cooper KM, et al. Retrospective analysis of transient elevations in alanine aminotransferase during long-term treatment with acetaminophen in osteoarthritis clinical trials. Curr Med Res Opin 2006;22(11):2137–48. [18] FDA. Internal analgesic, antipyretic, and antirheumatic drug products for over-thecounter human use; proposed amendment of the tentative final monograph; required warnings and other labeling. 2007; Available at: http://www.fda.gov/OHRMS/DOCKETS/ 98fr/E6-21855.htm. Accessed June 26, 2007. [19] Watson WA, Litovitz TL, Rodgers GC Jr, et al. 2004 annual report of the American Association of Poison Control Centers toxic exposure surveillance system. Am J Emerg Med 2005;23(5):589–666.

540

LARSON

[20] Larson AM, Polson J, Fontana RJ, et al. Acetaminophen-induced acute liver failure: results of a United States multicenter, prospective study. Hepatology 2005;42(6):1364–72. [21] Litovitz TL, Klein-Schwartz W, Dyer KS, et al. 1997 annual report of the American Association of Poison Control Centers toxic exposure surveillance system. Am J Emerg Med 1998;16(5):443–97. [22] Gow PJ, Smallwood RA, Angus PW. Paracetamol overdose in a liver transplantation centre: an 8-year experience. J Gastroenterol Hepatol 1999;14(8):817–21. [23] Ayonrinde OT, Phelps GJ, Hurley JC, et al. Paracetamol overdose and hepatotoxicity at a regional Australian hospital: a 4-year experience. Intern Med J 2005;35(11):655–60. [24] Park BK, Pirmohamed M, Kitteringham NR. The role of cytochrome P450 enzymes in hepatic and extrahepatic human drug toxicity. Pharmacol Ther 1995;68(3):385–424. [25] Walgren JL, Mitchell MD, Thompson DC. Role of metabolism in drug-induced idiosyncratic hepatotoxicity. Crit Rev Toxicol 2005;35(4):325–61. [26] Nelson SD. Mechanisms of acetaminophen-induced liver disease. In: Kaplowitz N, DeLeve LD, editors. Drug-induced liver disease. New York: Marcel Dekker Inc; 2003. p. 287–325. [27] Graham GG, Scott KF, Day RO. Tolerability of paracetamol. Drug Saf 2005;28(3):227–40. [28] Mitchell JR, Thorgeirsson SS, Potter WZ, et al. Acetaminophen-induced hepatic injury: protective role of glutathione in man and rationale for therapy. Clin Pharmacol Ther 1974;16(4):676–84. [29] Forrest JA, Clements JA, Prescott LF. Clinical pharmacokinetics of paracetamol. Clin Pharmacokinet 1982;7(2):93–107. [30] Manyike PT, Kharasch ED, Kalhorn TF, et al. Contribution of CYP2E1 and CYP3A to acetaminophen reactive metabolite formation. Clin Pharmacol Ther 2000;67(3):275–82. [31] Watkins PB, Seeff LB. Drug-induced liver injury: summary of a single topic clinical research conference. Hepatology 2006;43(3):618–31. [32] Corcoran GB, Mitchell JR, Vaishnav YN, et al. Evidence that acetaminophen and N-hydroxyacetaminophen form a common arylating intermediate, N-acetyl-p-benzoquinoneimine. Mol Pharmacol 1980;18(3):536–42. [33] Thummel KE, Lee CA, Kunze KL, et al. Oxidation of acetaminophen to N-acetyl-p-aminobenzoquinone imine by human CYP3A4. Biochem Pharmacol 1993;45(8):1563–9. [34] Makin A, Williams R. The current management of paracetamol overdosage. Br J Clin Pract 1994;48(3):144–8. [35] Chen W, Koenigs LL, Thompson SJ, et al. Oxidation of acetaminophen to its toxic quinone imine and nontoxic catechol metabolites by baculovirus-expressed and purified human cytochromes P450 2E1 and 2A6. Chem Res Toxicol 1998;11(4):295–301. [36] Sarich T, Kalhorn T, Magee S, et al. The effect of omeprazole pretreatment on acetaminophen metabolism in rapid and slow metabolizers of S-mephenytoin. Clin Pharmacol Ther 1997;62(1):21–8. [37] Mitchell JR, Jollow DJ, Potter WZ, et al. Acetaminophen-induced hepatic necrosis. IV. Protective role of glutathione. J Pharmacol Exp Ther 1973;187(1):211–7. [38] Kaplowitz N. Acetaminophen hepatoxicity: what do we know, what don’t we know, and what do we do next? Hepatology 2004;40(1):23–6. [39] O’Brien PJ, Khan S, Jatoe SD. Formation of biological reactive intermediates by peroxidases: halide mediated acetaminophen oxidation and cytotoxicity. Adv Exp Med Biol 1991;283:51–64. [40] Potter DW, Hinson JA. The 1- and 2-electron oxidation of acetaminophen catalyzed by prostaglandin H synthase. J Biol Chem 1987;262(3):974–80. [41] Harvison PJ, Egan RW, Gale PH, et al. Acetaminophen and analogs as cosubstrates and inhibitors of prostaglandin H synthase. Chem Biol Interact 1988;64(3):251–66. [42] Rumack BH, Matthew H. Acetaminophen poisoning and toxicity. Pediatrics 1975;55(6): 871–6. [43] Eriksson LS, Broome U, Kalin M, et al. Hepatotoxicity due to repeated intake of low doses of paracetamol. J Intern Med 1992;231(5):567–70.

ACETAMINOPHEN HEPATOTOXICITY

541

[44] Bonkovsky HL, Kane RE, Jones DP, et al. Acute hepatic and renal toxicity from low doses of acetaminophen in the absence of alcohol abuse or malnutrition: evidence for increased susceptibility to drug toxicity due to cardiopulmonary and renal insufficiency. Hepatology 1994;19(5):1141–8. [45] Moling O, Cairon E, Rimenti G, et al. Severe hepatotoxicity after therapeutic doses of acetaminophen. Clin Ther 2006;28(5):755–60. [46] Bray GP, Harrison PM, O’Grady JG, et al. Long-term anticonvulsant therapy worsens outcome in paracetamol-induced fulminant hepatic failure. Hum Exp Toxicol 1992;11(4): 265–70. [47] Goldfinger R, Ahmed KS, Pitchumoni CS, et al. Concomitant alcohol and drug abuse enhancing acetaminophen toxicity. Report of a case. Am J Gastroenterol 1978;70(4): 385–8. [48] Mutlib AE, Goosen TC, Bauman JN, et al. Kinetics of acetaminophen glucuronidation by UDP-glucuronosyltransferases 1A1, 1A6, 1A9 and 2B15. Potential implications in acetaminophen-induced hepatotoxicity. Chem Res Toxicol 2006;19(5):701–9. [49] Price VF, Jollow DJ. Effect of glucose and gluconeogenic substrates on fasting-induced suppression of acetaminophen glucuronidation in the rat. Biochem Pharmacol 1989; 38(2):289–97. [50] Chanda S, Mehendale HM. Role of nutrition in the survival after hepatotoxic injury. Toxicology 1996;111(1–3):163–78. [51] McClain CJ, Kromhout JP, Peterson FJ, et al. Potentiation of acetaminophen hepatotoxicity by alcohol. JAMA 1980;244(3):251–3. [52] Prescott LF. Paracetamol, alcohol and the liver. Br J Clin Pharmacol 2000;49(4):291–301. [53] Rumack BH. Acetaminophen hepatotoxicity: the first 35 years. J Toxicol Clin Toxicol 2002;40(1):3–20. [54] Lee SS, Buters JT, Pineau T, et al. Role of CYP2E1 in the hepatotoxicity of acetaminophen. J Biol Chem 1996;271(20):12063–7. [55] Linden CH, Rumack BH. Acetaminophen overdose. Emerg Med Clin North Am 1984;2(1): 103–19. [56] Prescott LF. Paracetamol overdosage. Pharmacological considerations and clinical management. Drugs 1983;25(3):290–314. [57] Gemborys MW, Mudge GH, Gribble GW. Mechanism of decomposition of N-hydroxyacetaminophen, a postulated toxic metabolite of acetaminophen. J Med Chem 1980;23(3): 304–8. [58] James LP, Mayeux PR, Hinson JA. Acetaminophen-induced hepatotoxicity. Drug Metab Dispos 2003;31(12):1499–506. [59] Roberts DW, Pumford NR, Potter DW, et al. A sensitive immunochemical assay for acetaminophen-protein adducts. J Pharmacol Exp Ther 1987;241(2):527–33. [60] Pumford NR, Hinson JA, Potter DW, et al. Immunochemical quantitation of 3-(cysteinS-yl)acetaminophen adducts in serum and liver proteins of acetaminophen-treated mice. J Pharmacol Exp Ther 1989;248(1):190–6. [61] Gibson JD, Pumford NR, Samokyszyn VM, et al. Mechanism of acetaminophen-induced hepatotoxicity: covalent binding versus oxidative stress. Chem Res Toxicol 1996;9(3): 580–5. [62] Cohen SD, Khairallah EA. Selective protein arylation and acetaminophen-induced hepatotoxicity. Drug Metab Rev 1997;29(1–2):59–77. [63] Pumford NR, Hinson JA, Benson RW, et al. Immunoblot analysis of protein containing 3-(cystein-S-yl)acetaminophen adducts in serum and subcellular liver fractions from acetaminophen-treated mice. Toxicol Appl Pharmacol 1990;104(3):521–32. [64] Roberts LJ, Morrow JD. Analgesic-antipyretic and antiinflammatory agents and drugs employed in the treatment of gout. In: Brunton L, Lazo J, Parker K, editors. Goodman and Gilman’s the pharmacological basis of therapeutics. 10th edition. New York: McGrawHill; 2001. p. 687–731.

542

LARSON

[65] Harman AW, Kyle ME, Serroni A, et al. The killing of cultured hepatocytes by N-acetyl-pbenzoquinone imine (NAPQI) as a model of the cytotoxicity of acetaminophen. Biochem Pharmacol 1991;41(8):1111–7. [66] Jaeschke H, Knight TR, Bajt ML. The role of oxidant stress and reactive nitrogen species in acetaminophen hepatotoxicity. Toxicol Lett 2003;144(3):279–88. [67] Tirmenstein MA, Nelson SD. Subcellular binding and effects on calcium homeostasis produced by acetaminophen and a nonhepatotoxic regioisomer, 30 -hydroxyacetanilide, in mouse liver. J Biol Chem 1989;264(17):9814–9. [68] Andersson BS, Rundgren M, Nelson SD, et al. N-acetyl-p-benzoquinone imine-induced changes in the energy metabolism in hepatocytes. Chem Biol Interact 1990;75(2):201–11. [69] Burcham PC, Harman AW. Acetaminophen toxicity results in site-specific mitochondrial damage in isolated mouse hepatocytes. J Biol Chem 1991;266(8):5049–54. [70] Park BK, Kitteringham NR, Maggs JL, et al. The role of metabolic activation in druginduced hepatotoxicity. Annu Rev Pharmacol Toxicol 2005;45:177–202. [71] Jaeschke H, Bajt ML. Intracellular signaling mechanisms of acetaminophen-induced liver cell death. Toxicol Sci 2006;89(1):31–41. [72] Boobis AR, Tee LB, Hampden CE, et al. Freshly isolated hepatocytes as a model for studying the toxicity of paracetamol. Food Chem Toxicol 1986;24(6–7):731–6. [73 Grewal KK, Racz WJ. Intracellular calcium disruption as a secondary event in acetaminophen-induced hepatotoxicity. Can J Physiol Pharmacol 1993;71(1):26–33. [74] Blazka ME, Wilmer JL, Holladay SD, et al. Role of proinflammatory cytokines in acetaminophen hepatotoxicity. Toxicol Appl Pharmacol 1995;133(1):43–52. [75] Liu ZX, Govindarajan S, Kaplowitz N. Innate immune system plays a critical role in determining the progression and severity of acetaminophen hepatotoxicity. Gastroenterology 2004;127(6):1760–74. [76] Liu ZX, Kaplowitz N. Role of innate immunity in acetaminophen-induced hepatotoxicity. Expert Opin Drug Metab Toxicol 2006;2(4):493–503. [77] Ishida Y, Kondo T, Ohshima T, et al. A pivotal involvement of IFN-gamma in the pathogenesis of acetaminophen-induced acute liver injury. FASEB J 2002;16(10):1227–36. [78] Cover C, Liu J, Farhood A, et al. Pathophysiological role of the acute inflammatory response during acetaminophen hepatotoxicity. Toxicol Appl Pharmacol 2006;216(1):98–107. [79] Jaeschke H. How relevant are neutrophils for acetaminophen hepatotoxicity? Hepatology 2006;43(6):1191–4. [80] Hinson JA, Reid AB, McCullough SS, et al. Acetaminophen-induced hepatotoxicity: role of metabolic activation, reactive oxygen/nitrogen species, and mitochondrial permeability transition. Drug Metab Rev 2004;36(3–4):805–22. [81] Miller RP, Roberts RJ, Fischer LJ. Acetaminophen elimination kinetics in neonates, children, and adults. Clin Pharmacol Ther 1976;19(3):284–94. [82] van der Marel CD, Anderson BJ, van Lingen RA, et al. Paracetamol and metabolite pharmacokinetics in infants. Eur J Clin Pharmacol 2003;59(3):243–51. [83] Peterson RG, Rumack BH. Age as a variable in acetaminophen overdose. Arch Intern Med 1981;141(3 spec no):390–3. [84] Rumack BH. Acetaminophen overdose in children and adolescents. Pediatr Clin North Am 1986;33(3):691–701. [85] Rivera-Penera T, Gugig R, Davis J, et al. Outcome of acetaminophen overdose in pediatric patients and factors contributing to hepatotoxicity. J Pediatr 1997;130(2):300–4. [86] Nelson SD. Molecular mechanisms of the hepatotoxicity caused by acetaminophen. Semin Liver Dis 1990;10(4):267–78. [87] Rumore MM, Blaiklock RG. Influence of age-dependent pharmacokinetics and metabolism on acetaminophen hepatotoxicity. J Pharm Sci 1992;81(3):203–7. [88] Tarloff JB, Khairallah EA, Cohen SD, et al. Sex- and age-dependent acetaminophen hepato- and nephrotoxicity in Sprague-Dawley rats: role of tissue accumulation, nonprotein sulfhydryl depletion, and covalent binding. Fundam Appl Toxicol 1996;30(1):13–22.

ACETAMINOPHEN HEPATOTOXICITY

543

[89] Alander SW, Dowd MD, Bratton SL, et al. Pediatric acetaminophen overdose: risk factors associated with hepatocellular injury. Arch Pediatr Adolesc Med 2000;154(4): 346–50. [90] Ueshima Y, Tsutsumi M, Takase S, et al. Acetaminophen metabolism in patients with different cytochrome P-4502E1 genotypes. Alcohol Clin Exp Res 1996;20(Suppl 1): 25A–8A. [91] Benson GD, Koff RS, Tolman KG. The therapeutic use of acetaminophen in patients with liver disease. Am J Ther 2005;12(2):133–41. [92] Farrell GC, Cooksley WG, Powell LW. Drug metabolism in liver disease: activity of hepatic microsomal metabolizing enzymes. Clin Pharmacol Ther 1979;26(4):483–92. [93] Guengerich FP, Turvy CG. Comparison of levels of several human microsomal cytochrome P-450 enzymes and epoxide hydrolase in normal and disease states using immunochemical analysis of surgical liver samples. J Pharmacol Exp Ther 1991;256(3):1189–94. [94] Rumack BH. Acetaminophen misconceptions. Hepatology 2004;40(1):10–5. [95] Lee WM. Acetaminophen and the U.S. Acute Liver Failure study group: lowering the risks of hepatic failure. Hepatology 2004;40(1):6–9. [96] Rumack BH, Peterson RC, Koch GG, et al. Acetaminophen overdose. 662 cases with evaluation of oral acetylcysteine treatment. Arch Intern Med 1981;141(3 spec no):380–5. [97] Altomare E, Leo MA, Lieber CS. Interaction of acute ethanol administration with acetaminophen metabolism and toxicity in rats fed alcohol chronically. Alcohol Clin Exp Res 1984;8(4):405–8. [98] Thummel KE, Slattery JT, Nelson SD. Mechanism by which ethanol diminishes the hepatotoxicity of acetaminophen. J Pharmacol Exp Ther 1988;245(1):129–36. [99] Thummel KE, Slattery JT, Nelson SD, et al. Effect of ethanol on hepatotoxicity of acetaminophen in mice and on reactive metabolite formation by mouse and human liver microsomes. Toxicol Appl Pharmacol 1989;100(3):391–7. [100] Schmidt LE, Dalhoff K, Poulsen HE. Acute versus chronic alcohol consumption in acetaminophen-induced hepatotoxicity. Hepatology 2002;35(4):876–82. [101] Schiodt FV, Lee WM, Bondesen S, et al. Influence of acute and chronic alcohol intake on the clinical course and outcome in acetaminophen overdose. Aliment Pharmacol Ther 2002; 16(4):707–15. [102] Black M, Raucy J. Acetaminophen, alcohol, and cytochrome P-450. Ann Intern Med 1986; 104(3):427–9. [103] Perrot N, Nalpas B, Yang CS, et al. Modulation of cytochrome P450 isozymes in human liver, by ethanol and drug intake. Eur J Clin Invest 1989;19(6):549–55. [104] Lucas D, Menez C, Girre C, et al. Decrease in cytochrome P4502E1 as assessed by the rate of chlorzoxazone hydroxylation in alcoholics during the withdrawal phase. Alcohol Clin Exp Res 1995;19(2):362–6. [105] Lauterburg BH, Velez ME. Glutathione deficiency in alcoholics: risk factor for paracetamol hepatotoxicity. Gut 1988;29(9):1153–7. [106] Lieber CS. Biochemical factors in alcoholic liver disease. Semin Liver Dis 1993;13(2): 136–53. [107] Newman TJ, Bargman GJ. Acetaminophen hepatotoxicity and malnutrition. Am J Gastroenterol 1979;72(6):647–50. [108] Cheung L, Potts RG, Meyer KC. Acetaminophen treatment nomogram. N Engl J Med 1994;330(26):1907–8. [109] Makin AJ, Wendon J, Williams R. A 7-year experience of severe acetaminophen-induced hepatotoxicity (1987–1993). Gastroenterology 1995;109(6):1907–16. [110] Bizovi KE, Smilkstein MJ. Acetaminophen. In: Goldfrank LR, Flomenbaum NE, Lewin NA, et al., editors. Goldfrank’s toxicologic emergencies. 7th edition. New York (NY): McGraw-Hill; 2002. p. 480–501. [111] Makin A, Williams R. Paracetamol hepatotoxicity and alcohol consumption in deliberate and accidental overdose. QJM 2000;93(6):341–9.

544

LARSON

[112] Kuffner EK, Dart RC, Bogdan GM, et al. Effect of maximal daily doses of acetaminophen on the liver of alcoholic patients: a randomized, double-blind, placebo-controlled trial. Arch Intern Med 2001;161(18):2247–52. [113] FDA. Alcohol warnings for pain relievers and fever reducers. 1998; Available at: http:// www.fda.gov/bbs/topics/NEWS/NEW00659.html. Accessed March 1, 2007. [114] Toes MJ, Jones AL, Prescott L. Drug interactions with paracetamol. Am J Ther 2005;12(1): 56–66. [115] Nolan CM, Sandblom RE, Thummel KE, et al. Hepatotoxicity associated with acetaminophen usage in patients receiving multiple drug therapy for tuberculosis. Chest 1994;105(2): 408–11. [116] Nicod L, Viollon C, Regnier A, et al. Rifampicin and isoniazid increase acetaminophen and isoniazid cytotoxicity in human HepG2 hepatoma cells. Hum Exp Toxicol 1997;16(1): 28–34. [117] Moulding TS, Redeker AG, Kanel GC. Acetaminophen, isoniazid, and hepatic toxicity. Ann Intern Med 1991;114(5):431. [118] Minton NA, Henry JA, Frankel RJ. Fatal paracetamol poisoning in an epileptic. Hum Toxicol 1988;7(1):33–4. [119] McClements BM, Hyland M, Callender ME, et al. Management of paracetamol poisoning complicated by enzyme induction due to alcohol or drugs. Lancet 1990;335(8704): 1526. [120] Suchin SM, Wolf DC, Lee Y, et al. Potentiation of acetaminophen hepatotoxicity by phenytoin, leading to liver transplantation. Dig Dis Sci 2005;50(10):1836–8. [121] Kostrubsky SE, Sinclair JF, Strom SC, et al. Phenobarbital and phenytoin increased acetaminophen hepatotoxicity due to inhibition of UDP-glucuronosyltransferases in cultured human hepatocytes. Toxicol Sci 2005;87(1):146–55. [122] Shriner K, Goetz MB. Severe hepatotoxicity in a patient receiving both acetaminophen and zidovudine. Am J Med 1992;93(1):94–6. [123] Levine M, Bellward GD. Effect of cimetidine on hepatic cytochrome P450: evidence for formation of a metabolite-intermediate complex. Drug Metab Dispos 1995;23(12): 1407–11. [124] Slattery JT, McRorie TI, Reynolds R, et al. Lack of effect of cimetidine on acetaminophen disposition in humans. Clin Pharmacol Ther 1989;46(5):591–7. [125] Burkhart KK, Janco N, Kulig KW, et al. Cimetidine as adjunctive treatment for acetaminophen overdose. Hum Exp Toxicol 1995;14(3):299–304. [126] Estes JD, Stolpman D, Olyaei A, et al. High prevalence of potentially hepatotoxic herbal supplement use in patients with fulminant hepatic failure. Arch Surg 2003;138(8):852–8. [127] Price VF, Miller MG, Jollow DJ. Mechanisms of fasting-induced potentiation of acetaminophen hepatotoxicity in the rat. Biochem Pharmacol 1987;36(4):427–33. [128] Kurtovic J, Riordan SM. Paracetamol-induced hepatotoxicity at recommended dosage. J Intern Med 2003;253(2):240–3. [129] Ito Y, Abril ER, Bethea NW, et al. Dietary steatotic liver attenuates acetaminophen hepatotoxicity in mice. Microcirculation 2006;13(1):19–27. [130] Lauterburg BH. Analgesics and glutathione. Am J Ther 2002;9(3):225–33. [131] Dong SX, Ping ZZ, Xiao WZ, et al. Effect of active and passive cigarette smoking on CYP1A2-mediated phenacetin disposition in Chinese subjects. Ther Drug Monit 1998; 20(4):371–5. [132] Seo KW, Park M, Kim JG, et al. Effects of benzothiazole on the xenobiotic metabolizing enzymes and metabolism of acetaminophen. J Appl Toxicol 2000;20(6):427–30. [133] MacLeod SL, Nowell S, Massengill J, et al. Cancer therapy and polymorphisms of cytochromes P450. Clin Chem Lab Med 2000;38(9):883–7. [134] Schmidt LE, Dalhoff K. The impact of current tobacco use on the outcome of paracetamol poisoning. Aliment Pharmacol Ther 2003;18(10):979–85. [135] Bromer MQ, Black M. Acetaminophen hepatotoxicity. Clin Liver Dis 2003;7(2):351–67.

ACETAMINOPHEN HEPATOTOXICITY

545

[136] Strubelt O, Siegers CP, Volpel M, et al. Studies on the mechanism of paracetamol-induced protection against paracetamol hepatotoxicity. Toxicology 1979;12(2):121–33. [137] Poulsen HE, Thomsen P. Long-term administration of toxic doses of paracetamol (acetaminophen) to rats. Liver 1988;8(3):151–6. [138] Shayiq RM, Roberts DW, Rothstein K, et al. Repeat exposure to incremental doses of acetaminophen provides protection against acetaminophen-induced lethality in mice: an explanation for high acetaminophen dosage in humans without hepatic injury. Hepatology 1999; 29(2):451–63. [139] Jones AF, Vale JA. Paracetamol poisoning and the kidney. J Clin Pharm Ther 1993;18(1): 5–8. [140] Singer AJ, Carracio TR, Mofenson HC. The temporal profile of increased transaminase levels in patients with acetaminophen-induced liver dysfunction. Ann Emerg Med 1995; 26(1):49–53. [141] Bessems JG, Vermeulen NP. Paracetamol (acetaminophen)-induced toxicity: molecular and biochemical mechanisms, analogues and protective approaches. Crit Rev Toxicol 2001;31(1):55–138. [142] Curry RW Jr, Robinson JD, Sughrue MJ. Acute renal failure after acetaminophen ingestion. JAMA 1982;247(7):1012–4. [143] von Mach MA, Hermanns-Clausen M, Koch I, et al. Experiences of a poison center network with renal insufficiency in acetaminophen overdose: an analysis of 17 cases. Clin Toxicol (Phila) 2005;43(1):31–7. [144] Mour G, Feinfeld DA, Caraccio T, et al. Acute renal dysfunction in acetaminophen poisoning. Ren Fail 2005;27(4):381–3. [145] Mofenson HC, Caraccio TR, Nawaz H, et al. Acetaminophen induced pancreatitis. J Toxicol Clin Toxicol 1991;29(2):223–30. [146] Record CO, Chase RA, Alberti KG, et al. Disturbances in glucose metabolism in patients with liver damage due to paracetamol overdose. Clin Sci Mol Med 1975;49(5):473–9. [147] Heinig RE, Clarke EF, Waterhouse C. Lactic acidosis and liver disease. Arch Intern Med 1979;139(11):1229–32. [148] Bihari D, Gimson AE, Lindridge J, et al. Lactic acidosis in fulminant hepatic failure. Some aspects of pathogenesis and prognosis. J Hepatol 1985;1(4):405–16. [149] Bernal W, Wendon J, Rela M, et al. Use and outcome of liver transplantation in acetaminophen-induced acute liver failure. Hepatology 1998;27(4):1050–5. [150] Harrison PM, O’Grady JG, Keays RT, et al. Serial prothrombin time as prognostic indicator in paracetamol induced fulminant hepatic failure. BMJ 1990;301(6758):964–6. [151] Tsutsumi M, Lasker JM, Shimizu M, et al. The intralobular distribution of ethanol-inducible P450IIE1 in rat and human liver. Hepatology 1989;10(4):437–46. [152] Baeg NJ, Bodenheimer HC Jr, Burchard K. Long-term sequelae of acetaminophen-associated fulminant hepatic failure: relevance of early histology. Am J Gastroenterol 1988;83(5):569–71. [153] Bertholf RL, Johannsen LM, Bazooband A, et al. False-positive acetaminophen results in a hyperbilirubinemic patient. Clin Chem 2003;49(4):695–8. [154] Polson J, Orsulak PJ, Wians F. Elevated bilirubin may cause false positive acetaminophen levels in hepatitis patients [abstract]. Hepatology 2004;40:496A. [155] Beuhler MC, Curry SC. False positive acetaminophen levels associated with hyperbilirubinemia. Clin Toxicol (Phila) 2005;43(3):167–70. [156] Muldrew KL, James LP, Coop L, et al. Determination of acetaminophen-protein adducts in mouse liver and serum and human serum after hepatotoxic doses of acetaminophen using high-performance liquid chromatography with electrochemical detection. Drug Metab Dispos 2002;30(4):446–51. [157] Davern TJ, James LP, Hinson JA, et al. Measurement of serum acetaminophen-protein adducts in patients with acute liver failure. Gastroenterology 2006;130(3):687–94. [158] Read RB, Tredger JM, Williams R. Analysis of factors responsible for continuing mortality after paracetamol overdose. Hum Toxicol 1986;5(3):201–6.

546

LARSON

[159] Ambre J, Alexander M. Liver toxicity after acetaminophen ingestion. Inadequacy of the dose estimate as an index of risk. JAMA 1977;238(6):500–1. [160] Smilkstein MJ, Knapp GL, Kulig KW, et al. Efficacy of oral N-acetylcysteine in the treatment of acetaminophen overdose. Analysis of the national multicenter study (1976 to 1985). N Engl J Med 1988;319(24):1557–62. [161] Reid D, Hazell W. Paracetamol poisoning: which nomogram should we use? Emerg Med (Fremantle) 2003;15(5–6):486–96. [162] Tan C, Graudins A. Comparative pharmacokinetics of Panadol extend and immediaterelease paracetamol in a simulated overdose model. Emerg Med Australas 2006;18(4): 398–403. [163] Ostapowicz G, Fontana RJ, Schiodt FV, et al. Results of a prospective study of acute liver failure at 17 tertiary care centers in the United States. Ann Intern Med 2002;137(12): 947–54. [164] Daly FF, O’Malley GF, Heard K, et al. Prospective evaluation of repeated supratherapeutic acetaminophen (paracetamol) ingestion. Ann Emerg Med 2004;44(4):393–8. [165] Sivilotti ML, Yarema MC, Juurlink DN, et al. A risk quantification instrument for acute acetaminophen overdose patients treated with N-acetylcysteine. Ann Emerg Med 2005; 46(3):263–71. [166] Prescott LF, Illingworth RN, Critchley JA, et al. Intravenous N-acetylcysteine: the treatment of choice for paracetamol poisoning. Br Med J 1979;2(6198):1097–100. [167] Prescott LF, Illingworth RN, Critchley JA, et al. Intravenous N-acetylcysteine: still the treatment of choice for paracetamol poisoning. Br Med J 1980;280(6206):46–7. [168] Kerr F, Dawson A, Whyte IM, et al. The Australasian clinical toxicology investigators collaboration randomized trial of different loading infusion rates of N-acetylcysteine. Ann Emerg Med 2005;45(4):402–8. [169] Bond GR, Requa RK, Krenzelok EP, et al. Influence of time until emesis on the efficacy of decontamination using acetaminophen as a marker in a pediatric population. Ann Emerg Med 1993;22(9):1403–7. [170] Underhill TJ, Greene MK, Dove AF. A comparison of the efficacy of gastric lavage, ipecacuanha and activated charcoal in the emergency management of paracetamol overdose. Arch Emerg Med 1990;7(3):148–54. [171] Rose SR, Gorman RL, Oderda GM, et al. Simulated acetaminophen overdose: pharmacokinetics and effectiveness of activated charcoal. Ann Emerg Med 1991;20(10):1064–8. [172] Spiller HA, Krenzelok EP, Grande GA, et al. A prospective evaluation of the effect of activated charcoal before oral N-acetylcysteine in acetaminophen overdose. Ann Emerg Med 1994;23(3):519–23. [173] Prescott LF, Park J, Ballantyne A, et al. Treatment of paracetamol (acetaminophen) poisoning with N-acetylcysteine. Lancet 1977;2(8035):432–4. [174] Peterson RG, Rumack BH. Treating acute acetaminophen poisoning with acetylcysteine. JAMA 1977;237(22):2406–7. [175] Buckpitt AR, Rollins DE, Mitchell JR. Varying effects of sulfhydryl nucleophiles on acetaminophen oxidation and sulfhydryl adduct formation. Biochem Pharmacol 1979;28(19): 2941–6. [176] Lin JH, Levy G. Sulfate depletion after acetaminophen administration and replenishment by infusion of sodium sulfate or N-acetylcysteine in rats. Biochem Pharmacol 1981;30(19): 2723–5. [177] Miners JO, Drew R, Birkett DJ. Mechanism of action of paracetamol protective agents in mice in vivo. Biochem Pharmacol 1984;33(19):2995–3000. [178] Corcoran GB, Todd EL, Racz WJ, et al. Effects of N-acetylcysteine on the disposition and metabolism of acetaminophen in mice. J Pharmacol Exp Ther 1985;232(3):857–63. [179] Slattery JT, Wilson JM, Kalhorn TF, et al. Dose-dependent pharmacokinetics of acetaminophen: evidence of glutathione depletion in humans. Clin Pharmacol Ther 1987;41(4): 413–8.

ACETAMINOPHEN HEPATOTOXICITY

547

[180] Harrison PM, Keays R, Bray GP, et al. Improved outcome of paracetamol-induced fulminant hepatic failure by late administration of acetylcysteine. Lancet 1990;335(8705): 1572–3. [181] Harrison PM, Wendon JA, Gimson AE, et al. Improvement by acetylcysteine of hemodynamics and oxygen transport in fulminant hepatic failure. N Engl J Med 1991;324(26): 1852–7. [182] Keays R, Harrison PM, Wendon JA, et al. Intravenous acetylcysteine in paracetamol induced fulminant hepatic failure: a prospective controlled trial. BMJ 1991;303(6809): 1026–9. [183] Jones AL. Mechanism of action and value of N-acetylcysteine in the treatment of early and late acetaminophen poisoning: a critical review. J Toxicol Clin Toxicol 1998;36(4): 277–85. [184] Burns MJ, Friedman SL, Larson AM. Acetaminophen (paracetamol) intoxication in adults. In: Rose BD, editor. UpToDate. Available at: http://www.uptodate.com. Accessed June 26, 2007. [185] Woo OF, Mueller PD, Olson KR, et al. Shorter duration of oral N-acetylcysteine therapy for acute acetaminophen overdose. Ann Emerg Med 2000;35(4):363–8. [186] Tsai CL, Chang WT, Weng TI, et al. A patient-tailored N-acetylcysteine protocol for acute acetaminophen intoxication. Clin Ther 2005;27(3):336–41. [187] Yip L, Dart RC, Hurlbut KM. Intravenous administration of oral N-acetylcysteine. Crit Care Med 1998;26(1):40–3. [188] Betten DP, Cantrell FL, Thomas SC, et al. A prospective evaluation of shortened course oral N-acetylcysteine for the treatment of acute acetaminophen poisoning. Ann Emerg Med 2007, in press. [189] Bronstein AC, Linden CH, Hall AH. Intravenous N-acetylcysteine for acute acetaminophen poisoning [abstract]. Vet Hum Toxicol 1985;27:316. [190] Holdiness MR. Clinical pharmacokinetics of N-acetylcysteine. Clin Pharmacokinet 1991; 20(2):123–34. [191] Selden BS, Curry SC, Clark RF, et al. Transplacental transport of N-acetylcysteine in an ovine model. Ann Emerg Med 1991;20(10):1069–72. [192] Smilkstein MJ, Bronstein AC, Linden C, et al. Acetaminophen overdose: a 48-hour intravenous N-acetylcysteine treatment protocol. Ann Emerg Med 1991;20(10):1058–63. [193] Kociancic T, Reed MD. Acetaminophen intoxication and length of treatment: how long is long enough? Pharmacotherapy 2003;23(8):1052–9. [194] Perry HE, Shannon MW. Efficacy of oral versus intravenous N-acetylcysteine in acetaminophen overdose: results of an open-label, clinical trial. J Pediatr 1998;132(1):149–52. [195] Ho SW, Beilin LJ. Asthma associated with N-acetylcysteine infusion and paracetamol poisoning: report of two cases. Br Med J (Clin Res Ed) 1983;287(6396):876–7. [196] Mant TG, Tempowski JH, Volans GN, et al. Adverse reactions to acetylcysteine and effects of overdose. Br Med J (Clin Res Ed) 1984;289(6439):217–9. [197] Dawson AH, Henry DA, McEwen J. Adverse reactions to N-acetylcysteine during treatment for paracetamol poisoning. Med J Aust 1989;150(6):329–31. [198] Flanagan RJ, Meredith TJ. Use of N-acetylcysteine in clinical toxicology. Am J Med 1991; 91(3C):131S–9S. [199] Vale JA, Proudfoot AT. Paracetamol (acetaminophen) poisoning. Lancet 1995;346(8974): 547–52. [200] Bailey B, McGuigan MA. Management of anaphylactoid reactions to intravenous N-acetylcysteine. Ann Emerg Med 1998;31(6):710–5. [201] Dean BS, Bricker JD, Krenzelok EP. Outpatient N-acetylcysteine treatment for acetaminophen poisoning: an ethical dilemma or a new financial mandate? Vet Hum Toxicol 1996; 38(3):222–4. [202] Piperno E, Berssenbruegge DA. Reversal of experimental paracetamol toxicosis with N-acetylcysteine. Lancet 1976;2(7988):738–9.

548

LARSON

[203] O’Grady JG, Alexander GJ, Hayllar KM, et al. Early indicators of prognosis in fulminant hepatic failure. Gastroenterology 1989;97(2):439–45. [204] O’Grady JG, Langley PG, Isola LM, et al. Coagulopathy of fulminant hepatic failure. Semin Liver Dis 1986;6(2):159–63. [205] Munoz SJ. Prothrombin time in fulminant hepatic failure. Gastroenterology 1991;100(5 Pt 1):1480–1. [206] Izumi S, Langley PG, Wendon J, et al. Coagulation factor V levels as a prognostic indicator in fulminant hepatic failure. Hepatology 1996;23(6):1507–11. [207] Pereira LM, Langley PG, Hayllar KM, et al. Coagulation factor V and VIII/V ratio as predictors of outcome in paracetamol induced fulminant hepatic failure: relation to other prognostic indicators. Gut 1992;33(1):98–102.

Acetaminophen Hepatotoxicity - Clinics In Liver Disease

Acetaminophen (N-acetyl-p-aminophenol or APAP), a mild nonnarcotic analgesic and antipyretic agent, is widely used as a pain reliever and fever reducer. It is available in hundreds of single-ingredient and combination over-the-counter (OTC) products, and also in numerous prescription prod- ucts. Its use has achieved ...

437KB Sizes 0 Downloads 249 Views

Recommend Documents

Management of coagulopathy in patients with liver disease ...
Page 3 of 3. Mehta. Table 5: Approximate blood product transfusion require- ments for liver transplant. Non-cirrhotics (including salvage). Blood 2.8 (3.5). Fresh frozen plasma 4.1 (4.1). Platelets Occasional. Cryoprecipitate Occasional. Cirrhotics (

pdf-0946\drug-induced-liver-disease-second-edition-by-neil ...
Whoops! There was a problem previewing this document. Retrying. ... or edit this item. pdf-0946\drug-induced-liver-disease-second-edition-by-neil-kaplowitz.pdf.

pdf-0729\coping-with-liver-disease-by-mark-greener.pdf ... - Drive
pdf-0729\coping-with-liver-disease-by-mark-greener.pdf. pdf-0729\coping-with-liver-disease-by-mark-greener.pdf. Open. Extract. Open with. Sign In. Main menu.

Advances in Extracorporeal Ventilation - Anesthesiology Clinics
Mechanical ventilation remains the signature tool of critical care and has greatly contributed to the tremendous progress in the treatment of critically ill patients.

Neurodegenerative disease: Tracking disease progress in ... - Nature
Mar 15, 2011 - Johnston, S. C. et al. National Stroke. Association recommendations for systems of care for TIA. Ann. Neurol. doi:10.1002/ ana.22332. 2. Fussman, C., Rafferty, A. P., Lyon-Callo, S.,. Morgenstern, L. B. & Reeves, M. J. Lack of associat

Metabolic Issues in Liver Transplantation
usually within normal limits in patients receiving good preoperative care. ' Intraoperative ...... cellular injury after cardiopulmonary bypass. N Engl J Med. 1991 ...

veterinary clinics
however, the profession must carefully examine the current model of noneco- nomic damages in .... care and treatment and serve as a basis for review, study and evaluation of the care .... The paperless office is a popular trend in veterinary practice

Pain Management Clinics
Nov 26, 2017 - Administrative Office. When available, this may be accomplished through an online ..... nursing school, a physician assistant program, or an outpatient clinic associated with any of the foregoing schools or .... clinic, and doing so sh

Acute Kidney Injury in the Neonate - Clinics in Perinatology
Siegel Career Development Grant and by the Pediatric and Infant Center for Acute ... Development of new, more precise biomarkers is a major focus of current ...

veterinary clinics
minutes before skin incision creation, the term perioperative antimicrobial use is also appropriate [39]. ..... [33] Beal MW, Brown DC, Shofer FS. The effects of ...

[PDF Online] Liver Detox Cleanse :The Natural Liver ...
This piece originally appeared in Lauren O’Neal’s email newsletter Activism for Non Activists It is reprinted here with permission Okay let’s talk ...

Neuroimaging of Dementia - Neurologic Clinics
b Dent Neurologic Institute, 3980A Sheridan Drive, Amherst, NY 14226, USA. * Corresponding author. E-mail address: [email protected]. KEYWORDS.

Care in Advanced Huntington's disease
capacity to make the decision themselves, a best interest decision by the .... There is also an online forum Message Board, where family members may post ...... earmarked savings account or a funeral plan (though terms and costs should be.

The social context of neonatal pain - Clinics in Perinatology
aDepartment of Psychology, Life Sciences Centre, Dalhousie University Halifax, ... bSchool of Occupational Therapy, 5869 University Avenue, Forrest Building,.

OTP Clinics in Utah APR 2018 - Web Version.pdf
Page 1 of 2. COUNTY NAME & ADDRESS PHONE DOSING HOURS CONTACT INFORMATION OTHER. Carbon. County. Operation Recovery. 77 South 600 ...

Fatty Liver Disease.pdf
There was a problem previewing this document. Retrying... Download. Connect more apps... Try one of the apps below to open or edit this item. Fatty Liver ...

Liver Discharge Instructions.pdf
There was a problem previewing this document. Retrying... Download. Connect more apps... Try one of the apps below to open or edit this item. Liver Discharge ...

Monitoring of Liver Enzymes in Patients On Chinese Medicine Mazin ...
Cao He Che (Rhizoma Polygoni Bistortae) 12g. Da Qing ... Gan Cao (Radix Glycyrrhizae Uralensis) 6g ... Ji Xue Teng (Radix et Caulis Jixueteng) 30g .... Monitoring of Liver Enzymes in Patients On Chinese Medicine Mazin Al-Khafaji 62-6.pdf.