MOBILE VIEW  | 

MUSHROOMS-CYCLOPEPTIDES

Classification   |    Detailed evidence-based information

Therapeutic Toxic Class

    A) Amatoxins are liver toxins found in the genera Amanita, Galerina and some Lepiota species. NOT all species in these genera contain the toxin, or in equal quantities. Cyclopeptide mushrooms are sometimes called Group 1 mushrooms. Amatoxins are water soluble, heat stable polypeptides that bind and inhibit RNA polymerase in kidney and liver cells in particular.

Specific Substances

    A) NOTE: SILIBININ STUDY
    1) For information on a silibinin clinical trial (start date: Nov 2009), please refer to individual product entry.
    2) Cyclopeptides-mushrooms
    3) Cyclopeptides (mushrooms)
    4) Amatoxins
    AMANITAS (synonym)
    1) Amanita bisporigera - "Destroying Angel" (synonym)
    2) Amanita brunescens (synonym)
    3) Amanita hygroscopica- "Pink-gilled Destroying Angel" (synonym)
    4) Amanita ocreata- "Destroying Angel" (synonym)
    5) Amanita phalloides - "The Death Cap" (synonym)
    6) Amanita suballiacea (synonym)
    7) Amanita tenuifolia (synonym)
    8) Amanita verna - "Destroying Angel" (synonym)
    9) Amanita virosa- "Destroying Angel" (synonym)
    10) Mushrooms-amanitins
    GALERINAS (synonym)
    1) Galerina autumnalis - "Deadly Galerina", 'Autumnal Galerina" (synonym)
    2) Galerina marginata - "Marginate Pholiota" (synonym)
    3) Galerina sulcipes - (Tropical species) (synonym)
    4) Galerina venenata - "Deadly Lawn Galerina" (synonym)
    5) Marginate Pholiota - Pholiota marginata (synonym)
    6) Autumnal Galerina - (common name for Galerina autumnalis) (synonym)
    7) Deadly Galerina - (common name for Galerina autumnalis) (synonym)
    8) Deadly Lawn Galerina - (common name for Galerina venenata) (synonym)
    LEPIOTAS (synonym)
    1) Lepiota castanea (synonym)
    2) Lepiota helveola (synonym)
    3) Lepiota subincarnata (synonym)
    4) Lepiota josserandii - "Deadly Parasol" (synonym)
    5) Lepiota subincarnata (synonym)
    6) Pholiota marginata - "Marginate Pholiota" (synonym)
    7) Deadly Parasol - (common name for Lepiota josserandii) (synonym)
    8) Conocybe filaris - (Pholiotina filaris) (synonym)
    9) Pholiotina filaris - (Conocybe filaris) (synonym)

Available Forms Sources

    A) FORMS
    1) AMATOXIN CONCENTRATIONS vary greatly in a definite species and from one mushroom to another.
    2) The following concentrations of amatoxins expressed in mg/g of dry tissue have been reported:
    1) A. phalloides, 2 to 7.3
    2) A. verna, 0.4 to 4.6
    3) A. virosa, 1.2 to 2.6
    4) A. bisporigera, 2.4
    5) Galerina autumnalis, 0.8 to 1.5
    6) Galerina marginata, 0.4

Life Support

    A) This overview assumes that basic life support measures have been instituted.

Clinical Effects

    0.2.1) SUMMARY OF EXPOSURE
    A) USES: Mushrooms of the genera Amanita, Galerina and some Lepiota species contain amatoxins. Not all species in these genera contain amatoxin. In the US, toxicity develops most commonly in amateur mushroom foragers who mistake these for edible mushrooms.
    B) TOXICOLOGY: Amatoxins inhibit RNA polymerase II within the nucleoplasm and inhibits both DNA and RNA transcription. Phallotoxin irreversibly polymerizes actin filaments by binding to F-actin. The target organ for amatoxin is the liver, causing fatty degeneration and centrilobular necrosis.
    C) EPIDEMIOLOGY: Exposures are rare in the United States, but there are several fatalities reported annually.
    D) WITH POISONING/EXPOSURE
    1) GASTROENTERITIS PHASE (PHASE I): Occurs 6 to 24 hours (mean, 12.3 hours) after ingestion. Symptoms may include nausea, vomiting, profuse diarrhea (may be bloody), abdominal pain, and electrolyte disturbances. These herald the onset of toxicity. Fever, tachycardia, hypovolemic shock, and metabolic acidosis are also seen.
    2) LATENT PHASE (PHASE II): Occurs 24 to 48 hours after ingestion. In this phase, there is resolution of the gastrointestinal symptoms and the patient appears clinically improved, though renal and hepatic function tests become abnormal.
    3) HEPATORENAL PHASE (PHASE III): Occurs 3 to 4 days after ingestion. There is worsening hepatic dysfunction with coma, jaundice with sclera icterus, hypoglycemia, altered mental status, metabolic acidosis, coagulopathy, spontaneous bleeding, and renal failure. Eventually, patients can go on to suffer from fulminant hepatic failure. Pulmonary injury in the form of acute respiratory distress syndrome (ARDS) occurs during this phase and typically is seen with severe hepatic dysfunction and coagulopathy. Neurologic sequelae occurs in conjunction with severe hepatic failure as well. These include coma, encephalopathy, confusion, and seizures. Either patients will make a slow recovery or will die within 6 to 16 days postingestion. Renal failure in this phase is common during fatal cases and occurs from direct nephrotoxicity or hypovolemia.
    4) Patients who die are more likely to develop hypotension, encephalopathy, mucosal hemorrhage, oliguria/anuria, hypoglycemia and thrombocytopenia during hospitalization than patients who survive. Laboratory findings associated with increased likelihood of fatal outcome include: hyponatremia, increased BUN, increased AST, ALT, bilirubin, LDH, and INR, and prolonged PT, or aPTT.
    0.2.20) REPRODUCTIVE
    A) In pregnancy, the fetus may develop a toxic hepatitis. Because it is unknown whether amatoxins are excreted in breast milk, nursing should be stopped.

Laboratory Monitoring

    A) Monitor serum glucose frequently in patients with evidence of hepatic injury. Monitor serial serum electrolytes, renal function, liver enzymes, coagulation studies, bilirubin, and urine output.
    B) AST, ALT, LDH, and serum bilirubin elevations are the earliest and best indicators of liver damage. Hepatic enzymes typically peak between the 60th and 72nd hours and then decrease. Transaminases may be low in cases of massive hepatic necrosis.
    C) Glucose, fibrinogen, and prothrombin time are the best indicators of hepatic failure.
    D) Amatoxins are detected in biologic fluids such as serum, urine, and emesis by radioimmunoassay or HPLC. These methods are not generally available in clinical practice. Serum levels of amatoxins do not correlate with the severity of symptoms of poisoning.

Treatment Overview

    0.4.2) ORAL/PARENTERAL EXPOSURE
    A) MANAGEMENT OF TOXICITY
    1) All patients with cyclopeptide mushroom ingestion have the potential to develop severe toxicity. Administer activated charcoal for recent ingestions, and consider multiple dose activated charcoal or nasogastric suction to interrupt enterohepatic recirculation of the toxins. N-acetylcysteine may be beneficial and is associated with little toxicity. Silibinin is currently undergoing a clinical trial for amatoxin poisoning. In studies, NAC and silibinin were the most effective agents. High-dose penicillin G has also been used. All of these therapies should be instituted as soon as possible in patients with the potential for significant toxicity.
    B) DECONTAMINATION
    1) Administer activated charcoal 25 to 50 g in adults and usually 25 g in children (1 to 12 years), and 1 g/kg in infants. Multiple dose activated charcoal may enhance elimination. After 50 g initial dose, give subsequent doses in 4-hour intervals at a rate of 12.5 g/hr. For children, after the initial dose of 25 g, administer subsequent doses at 4-hour intervals at a rate equivalent to 6.25 g/hr. Evaluate frequently for the presence of bowel sounds or signs of obstruction. Because this ingestion is potentially life-threatening, gastric lavage is indicated if the patient has not vomited spontaneously and it can be performed within a few hours of ingestion. Nasogastric suction between doses of charcoal may remove toxins eliminated in the bile.
    C) AIRWAY MANAGEMENT
    1) Perform early in patients unable to protect their airway.
    D) ANTIDOTE
    1) There are several drugs that have been proposed as antidotes in the management of cyclopeptide containing mushroom poisoning. None has been proven to have clinical efficacy in humans, but since the toxicity of these agents is limited, they are often used in suspected cyclopeptide mushroom ingestion. In studies, silibinin and NAC both administered as monotherapy, and silibinin with benzylpenicillin as bi-, tri-, and polytherapies were associated with the lowest mortalities. Overall, NAC and silibinin were the most effective agents.
    2) N-ACETYLCYSTEINE: High doses of N-acetylcysteine have been used in humans in the setting of amatoxin poisoning. The dose most often used is 150 mg/kg infusion over 60 minutes followed by 50 mg/kg infusion over 4 hours followed by 6.25 mg/kg/hour infusion which is continued until the patient has clinically improved.
    3) SILIBININ: Silibinin is being studied as a potential antidote for amatoxin. It is an extract from milk thistle and thought to inhibit the uptake of amatoxin into hepatocytes. The most common dose is 5 mg/kg IV loading dose followed by 20 mg/kg/day via continuous infusion. There is some human data showing promise of silibinin as an antidote, and it is currently available as part of an open label, multicenter clinical trial. To obtain silibinin, contact 866-520-4412.
    4) PENICILLIN G: Penicillin G appears to displace amatoxin from plasma protein binding sites and possibly inhibit its uptake into hepatocytes. The dose with some evidence of effectiveness is 300,000 to 1,000,000 units/day, though mortality is not significantly affected in preliminary studies.
    5) THIOCTIC ACID: Thioctic acid is a coenzyme in the Krebs cycle used in Eastern Europe for the treatment of amatoxin poisoning with some data showing a possible reduction in mortality. The dose is 50 to 150 mg every 6 hours. Its efficacy remains unproven. It has been reported to cause hypoglycemia and is not readily available in the US.
    E) VOLUME CONTRACTION/ELECTROLYTE DERANGEMENT
    1) Treat with aggressive intravenous fluid resuscitation. This serves to correct hypovolemia and prevent renal failure. Urine output should be closely monitored. Hypokalemia and metabolic acidosis should be corrected as well.
    F) HEPATIC INJURY
    1) Serum glucose levels should be monitored very closely. Intravenous boluses and/or infusions of dextrose should be given as indicated by the serum glucose levels. Coagulation status should be followed closely with laboratory testing. In the presence of coagulopathy or bleeding, give vitamin K and fresh frozen plasma. In cases of fulminant liver failure, consider liver transplantation. Several different criteria have been set forth to determine prognosis in hepatic failure. King's College criteria for non-paracetamol poisoning are as follows: either a prothrombin time over 100 sec (INR over 7), OR at least 3 of the following: PT over 50 sec (INR over 3.5), serum bilirubin over 300 micromol/L, age less than 10 or greater than 40 years, an interval between jaundice and encephalopathy over 7 days. Those that meet the "poor prognosis" will likely benefit from liver transplantation.
    2) NASOBILIARY DRAINAGE: Nasobiliary drainage has been purported as a possible therapeutic option for amatoxin and phallotoxin poisoning as a means to interrupt enterohepatic recirculation.
    3) MOLECULAR ABSORBENT REGENERATING SYSTEM (MARS): MARS is a method for removing protein-bound substances in patients with liver failure and hepatic encephalopathy. It employs an albumin-impregnated highly permeable dialyzer with albumin-containing dialysate recycled in a closed loop with a charcoal cartridge, an anion exchange resin absorber, and a conventional hemodialysis membrane. In Europe, it has been used in a small number of patients with fulminant hepatic failure secondary to cyclopeptide mushroom ingestion.
    4) FRACTIONATED PLASMA SEPARATION AND ADSORPTION SYSTEM (FPSA): FPSA is a method for filtering toxin-bearing proteins of molecular weight 300,00 daltons or less. In Europe it has been used in a small number of patients with fulminant hepatic failure secondary to cyclopeptide mushroom ingestion.
    G) ENHANCED ELIMINATION PROCEDURE
    1) Significant amounts of amatoxin are eliminated in the urine during the first 48 hours. Therefore, high urine output should be achieved in the first 48 hours. In addition, because amatoxins are cleared from the serum in the first 24 to 48 hours either by the kidneys or via uptake into hepatocytes, extracorporeal elimination techniques are not likely to be useful.
    H) PATIENT DISPOSITION
    1) HOME CRITERIA: Unless the potentially lethal mushroom species can be definitely ruled out, there should be no home treatment for mushroom intoxications.
    2) ADMISSION CRITERIA: Patients who develop gastroenteritis 6 to 24 hours postingestion should be sent to a medical care facility for evaluation and admitted until cyclopeptide ingestion can be excluded.
    3) CONSULT CRITERIA: Mushroom identification is best performed by a mycologist. If a sample of the mushroom can be retrieved, then it should be analyzed by a mycologist. After general treatment for poisonings has been administered, the regional poison center should be contacted for treatment recommendations. For those patients who develop hepatic failure, a transplant center should be notified to evaluate the patient for a possible liver transplantation.
    I) PITFALLS
    1) Much of the effective management for amatoxin poisoning is time-dependent, thus early diagnosis is essential.
    J) PHARMACOKINETICS
    1) The onset of gastrointestinal symptoms after amatoxin poisoning is typically between 6 to 24 hours, though some amatoxins have been found to be absorbed rapidly (as early as 90 to 120 minutes postingestion) into the serum. The volume of distribution in dogs has been found to range between 160 to 290 mL/kg and the elimination half-life was demonstrated to be 26.7 to 49.6 minutes.
    K) DIFFERENTIAL DIAGNOSIS
    1) Many mushroom species cause gastroenteritis; however, the onset is generally early (within 6 hours) after ingestions of mushrooms that only cause gastroenteritis. Gyromitra sp such as G. esculenta can cause delayed vomiting and diarrhea as well as hepatic and renal injury. Hapalopilus rutilans which has the toxin polyporic acid can cause hepatorenal failure.
    L) PROGNOSTIC FACTORS
    1) It appears that severity of coagulopathy is a reliable indicator of recovery or death in patients with Amanita phalloides poisoning. In one study, a reliable tool for deciding emergency transplantation was a prothrombin index of lower than 10% (an approximate INR of 6), 4 days or more after ingestion. No additional prognostic information were provided by encephalopathy and creatinine; therefore, it was proposed that encephalopathy should not be an absolute prerequisite for deciding transplantation. In another study, fatal outcomes were associated with low mean arterial pressure, encephalopathy, mucosal hemorrhage, oliguria-anuria, hypoglycemia, or thrombocytopenia during hospitalization. Low sodium values and elevated urea, AST, ALT, total bilirubin, LDH, prothrombin time, INR, and activated partial thromboplastin time (APTT) values were associated with poor outcome.

Range Of Toxicity

    A) TOXICITY: Amatoxins are highly potent toxins and are amongst the most lethal poisons known. The LD50 of the dose is 0.1 mg/kg which is equivalent to an ingestion of 6 to 7 mg of mushroom or a cap by an average person. The weight of an average Amanita phalloides cap is 60 g and the concentration of alpha-amanitin is usually 0.5 to 1.5 mg/g of mushroom. One Amanita cap is potentially lethal. Fifteen to 20 Galerina caps and 30 Lepiotas can kill a healthy adult. Mortality is 20% to 30% untreated (generally higher in children), but can be reduced to 5% with appropriate, aggressive, and intensive supportive care.

Summary Of Exposure

    A) USES: Mushrooms of the genera Amanita, Galerina and some Lepiota species contain amatoxins. Not all species in these genera contain amatoxin. In the US, toxicity develops most commonly in amateur mushroom foragers who mistake these for edible mushrooms.
    B) TOXICOLOGY: Amatoxins inhibit RNA polymerase II within the nucleoplasm and inhibits both DNA and RNA transcription. Phallotoxin irreversibly polymerizes actin filaments by binding to F-actin. The target organ for amatoxin is the liver, causing fatty degeneration and centrilobular necrosis.
    C) EPIDEMIOLOGY: Exposures are rare in the United States, but there are several fatalities reported annually.
    D) WITH POISONING/EXPOSURE
    1) GASTROENTERITIS PHASE (PHASE I): Occurs 6 to 24 hours (mean, 12.3 hours) after ingestion. Symptoms may include nausea, vomiting, profuse diarrhea (may be bloody), abdominal pain, and electrolyte disturbances. These herald the onset of toxicity. Fever, tachycardia, hypovolemic shock, and metabolic acidosis are also seen.
    2) LATENT PHASE (PHASE II): Occurs 24 to 48 hours after ingestion. In this phase, there is resolution of the gastrointestinal symptoms and the patient appears clinically improved, though renal and hepatic function tests become abnormal.
    3) HEPATORENAL PHASE (PHASE III): Occurs 3 to 4 days after ingestion. There is worsening hepatic dysfunction with coma, jaundice with sclera icterus, hypoglycemia, altered mental status, metabolic acidosis, coagulopathy, spontaneous bleeding, and renal failure. Eventually, patients can go on to suffer from fulminant hepatic failure. Pulmonary injury in the form of acute respiratory distress syndrome (ARDS) occurs during this phase and typically is seen with severe hepatic dysfunction and coagulopathy. Neurologic sequelae occurs in conjunction with severe hepatic failure as well. These include coma, encephalopathy, confusion, and seizures. Either patients will make a slow recovery or will die within 6 to 16 days postingestion. Renal failure in this phase is common during fatal cases and occurs from direct nephrotoxicity or hypovolemia.
    4) Patients who die are more likely to develop hypotension, encephalopathy, mucosal hemorrhage, oliguria/anuria, hypoglycemia and thrombocytopenia during hospitalization than patients who survive. Laboratory findings associated with increased likelihood of fatal outcome include: hyponatremia, increased BUN, increased AST, ALT, bilirubin, LDH, and INR, and prolonged PT, or aPTT.

Vital Signs

    3.3.2) RESPIRATIONS
    A) WITH POISONING/EXPOSURE
    1) Respirations are usually normal, but hyperventilation accompanies fulminant hepatitis. Hypoventilation and apnea may occur at the terminal phase of hepatic coma.
    3.3.3) TEMPERATURE
    A) WITH POISONING/EXPOSURE
    1) Amatoxins themselves do NOT alter temperature regulation. Temperature regulation disorders may occur in severe hepatic failure with encephalopathy.
    2) FEVER: Has been reported (Kervegant et al, 2013; Trabulus & Altiparmak, 2011). In a retrospective review of 144 amatoxin poisonings, 8 patients developed fever (Trabulus & Altiparmak, 2011).
    3.3.4) BLOOD PRESSURE
    A) WITH POISONING/EXPOSURE
    1) Hypovolemic shock with decreased central venous pressure may occur during the gastrointestinal phase. A functional and reversible renal failure often accompanies a hypovolemic shock secondary to gastrointestinal fluid losses.
    a) In patients with severe hepatocellular failure, shock is mostly related to hemorrhage or occurs in the terminal phase.
    3.3.5) PULSE
    A) WITH POISONING/EXPOSURE
    1) TACHYCARDIA is frequent when dehydration and hypovolemia are present.

Heent

    3.4.3) EYES
    A) WITH POISONING/EXPOSURE
    1) SCLERAL ICTERUS is associated with jaundice, but it may be the only manifestation of an increase in bilirubin.
    3.4.5) NOSE
    A) WITH POISONING/EXPOSURE
    1) EPISTAXIS may occur in patients with coagulation disorders related to severe hepatocellular damage. Thus, if intubation is necessary, the nasal route should be avoided because it may induce severe local hemorrhage.

Cardiovascular

    3.5.2) CLINICAL EFFECTS
    A) TACHYCARDIA
    1) WITH POISONING/EXPOSURE
    a) Tachycardia has been reported (Aygul et al, 2010). In the gastrointestinal phase, vomiting and diarrhea can produce severe fluid losses resulting in hypovolemic shock with tachycardia and decreased central venous pressure.
    b) In a retrospective review of 144 amatoxin poisonings, 7 patients developed tachycardia (Trabulus & Altiparmak, 2011).
    B) HYPOTENSIVE EPISODE
    1) WITH POISONING/EXPOSURE
    a) Patients may present with hypotension in the first phase of poisoning (Aygul et al, 2010; Scalzo et al, 1998). When shock occurs later, during the hepatitis phase, it is mostly due to hemorrhage secondary to severe coagulation disorders. Cardiovascular collapse also accompanies a severe hepatic failure at the terminal stage (Nicholson & Korman, 1997). Death may result from multiple organ failure (CDC, 1997; Nicholson & Korman, 1997).
    1) CASE REPORT: A 90-year-old woman, with recent onset of complete heart block treated with a permanent pacemaker, ingested mushrooms of the Amanita/Lepiota group. She developed hypotension refractory to pressors, in conjunction with fulminant liver failure. Lactic acidosis developed and she died on hospital day 5 (Feinfeld et al, 1994).
    b) In a retrospective review of 144 amatoxin poisonings, 21 patients developed hypotension (Trabulus & Altiparmak, 2011).
    C) BRADYCARDIA
    1) WITH POISONING/EXPOSURE
    a) In a retrospective review of 144 amatoxin poisonings, 2 patients developed bradycardia (Trabulus & Altiparmak, 2011).
    D) VENTRICULAR ARRHYTHMIA
    1) WITH POISONING/EXPOSURE
    a) In a retrospective review of 144 amatoxin poisonings, 2 patients developed ventricular dysrhythmia (Trabulus & Altiparmak, 2011).
    b) Transient impairment in cardiac systolic function developed in 3 of 45 patients with acute kidney injury and hepatic injury after ingesting amatoxin mushrooms. All 3 patients had acute reversible left ventricular dysfunction and mildly elevated cardiac enzymes (Altintepe et al, 2014).
    E) CARDIOGENIC SHOCK
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 24-year-old woman presented with abdominal pain, nausea, vomiting, and weakness 6 hours after ingesting Amanita phalloides mushrooms (unknown quantity). Despite supportive therapy, including 4 sessions of hemodialysis, her condition did not improve. Her condition deteriorated very quickly and she developed multiorgan failure, including liver, renal, and cardiac failure. At this time, she was orthopneic, cyanotic, somnolent, tachycardiac (130 beats/min), and hypotensive (BP 70/50 mmHg). An ECG revealed sinus tachycardia with nonspecific ST-T wave changes in anterior leads. Laboratory results revealed a prolonged prothrombin time (INR 5.19) and elevated liver enzymes and serum creatinine. Borderline cardiomegaly with bilateral pleural effusions at the costophrenic angles was observed in a chest x-ray. An echocardiogram showed a global left ventricular hypokinesia with left ventricular ejection fraction (EF) of 24%, end-diastolic diameter of 6.2 cm, and systolic pulmonary artery pressure of 50 mmHg. At this time, an intra-aortic balloon counterpulsation catheter was inserted and a marked improvement was noted within 1 hour. In addition, she was treated with 4 units of fresh-frozen plasma and a peritoneal dialysis catheter was inserted. Her condition continued to improve and both intra-aortic balloon counterpulsation and peritoneal dialysis were removed on day 5. She was discharged on day 12 (Aygul et al, 2010).

Respiratory

    3.6.2) CLINICAL EFFECTS
    A) APNEA
    1) WITH POISONING/EXPOSURE
    a) Respirations are usually normal, but hyperventilation accompanies fulminant hepatitis. Respiratory failure with hypoventilation or apnea may occur in patients presenting with hepatic coma and is a sign of poor prognosis.
    B) FIBROSIS OF LUNG
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Hemorrhagic pulmonary alveolitis was seen in a 14-year-old fatality with massive liver necrosis (Sanz et al, 1988).
    C) ACUTE LUNG INJURY
    1) WITH POISONING/EXPOSURE
    a) Adult respiratory distress syndrome (ARDS) may develop in the later stages of cyclopeptide mushroom poisoning, in conjunction with severe hepatic impairment and coagulopathies. ARDS resulting in death has been reported (Zevin et al, 1997; Ramirez et al, 1993).
    b) CASE REPORT: Seven days after eating Amanita phalloides mushrooms, a 32-year-old man developed adult respiratory distress syndrome requiring intubation and mechanical ventilation. He died 9 days after ingestion (CDC, 1997).

Neurologic

    3.7.2) CLINICAL EFFECTS
    A) HEPATIC ENCEPHALOPATHY
    1) WITH POISONING/EXPOSURE
    a) Neurologic symptoms are related to hepatic encephalopathy and may occur in patients developing severe hepatic failure. Encephalopathy usually occurs 5 to 7 days after ingestion (Mottram et al, 2010; Hydzik et al, 2008; Burton et al, 2002; CDC, 1997; Ramirez et al, 1993).
    b) CASE REPORT: A 54-year-old man who ingested 2 to 4 Amanita phalloides mushrooms, developed necrotic liver, encephalopathy, severe coagulopathy, and acute renal failure. He was treated with 7 sessions of Molecular Absorbents Recirculating Systems (MARS) starting 63 hours after ingesting mushrooms, as well as standard supportive care (eg, fluid resuscitation, NAC, oral activated charcoal). He also underwent liver transplantation 9 days postingestion. After the surgery, he experienced persistent acute renal failure (requiring hemodialysis), rhabdomyolysis, and heparin-induced thrombocytopenia. He recovered and was discharged home after 128 days of hospitalization (Kantola et al, 2009).
    c) CASE REPORT: A 65-year-old woman developed acute renal failure, liver encephalopathy, and hypoprothrombinemia after ingesting Amanita phalloides mushrooms. Following supportive care, her liver function recovered completely. However, acute renal failure persisted, necessitating hemodialysis (started on day 10). She still required hemodialysis at 2-year follow-up (Garrouste et al, 2009).
    d) In a retrospective review of 144 amatoxin poisonings, 10 patients developed encephalopathy (Trabulus & Altiparmak, 2011).
    e) CASE REPORT: A 71-year-old man presented with nausea, vomiting, diarrhea, and weakness within 48 hours of ingesting an unknown amount of Amanita mushrooms while camping in northwestern Iowa. Laboratory results revealed elevated liver enzymes, total bilirubin of 1.5 mg/dL (normal range, 0 to 1 mg/dL), INR of 1.3 (normal range, 0.8 to 1.2), and a serum creatinine of 2.7 mg/dL (normal range, 0.51 to 1.2 mg/dL). Despite treatment with IV N-acetylcysteine and penicillin G, he developed encephalopathy, acute renal injury, coagulopathy, and worsening transaminase elevations. He was transported to the liver transplant center and received multidose activated charcoal and IV silibinin (loading dose: 5 mg/kg over 1 hour, continued for 3 days at 20 mg/kg/day) starting 96 hours postingestion. His condition gradually improved by day 3 and he was discharged 5 days after presentation. On a follow-up visit a month later, his liver enzymes were normal (Gores et al, 2014).
    f) SOMNOLENCE
    1) Somnolence and confusion developed in a patient after ingesting a meal containing Lepiota brunneoincarnata mushrooms (Kervegant et al, 2013).
    2) Somnolence and confusion are the first signs of hepatic encephalopathy.
    g) COMA
    1) Coma usually follows somnolence and confusion. An altered state of consciousness is common in severe hepatic failure as part of a general CNS deterioration. Encephalopathy may progress to coma.
    2) In severe hepatic failure, coma may also be due to hypoglycemia secondary to glucose metabolism disorders.
    3) CASE REPORT: A 9-year-old boy became deeply comatose on the 5th day after amanita phalloides poisoning. Severe metabolic disorders included hypoglycemia, elevated serum lactate, total bilirubin of 7.3% and a prolonged prothrombin time (Langer et al, 1997).
    B) SEIZURE
    1) WITH POISONING/EXPOSURE
    a) Seizures may be observed in hepatic coma.
    b) Meningeal signs with anisocoria and miosis are often present prior to seizures (Mitchell & Rumack, 1978).
    C) RAISED INTRACRANIAL PRESSURE
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Langer et al (1997) reported intracranial hypertension on the 5th day following amanita phalloides ingestion in a 9-year-old boy which was adequately controlled for 12 hr. After 36 hours, intracranial hypertension (above 80 mm Hg) became resistant to therapy and the child died (Langer et al, 1997).
    D) NEUROPATHY
    1) WITH POISONING/EXPOSURE
    a) Polyneuropathy, developing 8 days after mushroom ingestion, has been reported in 5 patients. Loss of strength in the lower extremities, absence of deep tendon reflexes and alteration of pain, temperature and proprioceptive sensitivity was noted in all 5 patients. EMG readings showed a decrease in sensitive conduction speed and, to a lesser extent, motor speed. Two of these patients did not improve after 1 year (Ramirez et al, 1993).
    E) ASTHENIA
    1) WITH POISONING/EXPOSURE
    a) Asthenia has been reported in some patients after ingestion of these mushrooms (Kervegant et al, 2013; Krenova et al, 2007; Trabulus & Altiparmak, 2011).
    b) CASE SERIES: In a study of 34 patients with amanita phalloides poisoning, weakness developed in 4% of patients (Krenova et al, 2007).
    c) In a retrospective review of 144 amatoxin poisonings, 16 patients developed weakness (Trabulus & Altiparmak, 2011).
    F) VERTIGO
    1) WITH POISONING/EXPOSURE
    a) In a retrospective review of 144 amatoxin poisonings, 27 patients developed vertigo (Trabulus & Altiparmak, 2011).
    G) HEADACHE
    1) WITH POISONING/EXPOSURE
    a) In a retrospective review of 144 amatoxin poisonings, 10 patients developed headache (Trabulus & Altiparmak, 2011).

Gastrointestinal

    3.8.2) CLINICAL EFFECTS
    A) GASTROENTERITIS
    1) WITH POISONING/EXPOSURE
    a) Gastrointestinal symptoms, including nausea, vomiting, diarrhea, and abdominal pain, appear after a latent period of 6 to 24 hours (mean 12.3 hours) (Vanooteghem et al, 2014; Kervegant et al, 2013; Kantola et al, 2009; Hydzik et al, 2008; Himmelmann et al, 2001; Yamada et al, 1998; Scalzo et al, 1998; Omidynia et al, 1997; Serne et al, 1996; Aji et al, 1995; Cappell & Hassan, 1992; Rivett & Boon, 1988). One patient developed rectal bleeding after ingesting a meal containing Lepiota brunneoincarnata mushrooms (Kervegant et al, 2013).
    b) INCIDENCE: In a study of 205 Amanita intoxications, gastrointestinal symptoms were present in 199 (97%) patients (Floersheim et al, 1982).
    c) CASE REPORT: A 71-year-old man presented with nausea, vomiting, diarrhea, and weakness within 48 hours of ingesting an unknown amount of Amanita mushrooms while camping in northwestern Iowa. Laboratory results revealed elevated liver enzymes, total bilirubin of 1.5 mg/dL (normal range, 0 to 1 mg/dL), INR of 1.3 (normal range, 0.8 to 1.2), and a serum creatinine of 2.7 mg/dL (normal range, 0.51 to 1.2 mg/dL). Despite treatment with IV N-acetylcysteine and penicillin G, he developed encephalopathy, acute renal injury, coagulopathy, and worsening transaminase elevations. He was transported to the liver transplant center and received multidose activated charcoal and IV silibinin (loading dose: 5 mg/kg over 1 hour, continued for 3 days at 20 mg/kg/day) starting 96 hours postingestion. His condition gradually improved by day 3 and he was discharged 5 days after presentation. On a follow-up visit a month later, his liver enzymes were normal (Gores et al, 2014).
    B) NAUSEA AND VOMITING
    1) WITH POISONING/EXPOSURE
    a) There is a sudden onset of colicky abdominal pain rapidly followed by nausea, frequent vomiting, and diarrhea, usually beginning 10 to 12 hours after ingestion (Vanooteghem et al, 2014; Mottram et al, 2010; Hanrahan & Gordon, 1984; Gazzero & Goos, 1991; Scalzo et al, 1998; Nicholls et al, 1995; Cappell & Hassan, 1992; Nicholson & Korman, 1997; Omidynia et al, 1997).
    b) In a 15-year retrospective analysis of amatoxin mushroom poisoning (n=111), 93 (83.8%) patients developed nausea and vomiting, with the average delay in onset of 12.4 +/- 0.8 hours (Giannini et al, 2007).
    c) CASE SERIES: In a study of 34 patients with amanita phalloides poisoning, vomiting developed in 76% of patients (Krenova et al, 2007).
    d) In a retrospective review of 144 amatoxin poisonings, 139 patients developed nausea and 129 developed vomiting (Trabulus & Altiparmak, 2011).
    C) DIARRHEA
    1) WITH POISONING/EXPOSURE
    a) In many cases, diarrhea is severe, watery, and cholera-like (up to 2 to 4 L/day) (Mottram et al, 2010; Kaneko et al, 2001; Olesen, 1990; Nicholson & Korman, 1997; Omidynia et al, 1997) or may be bloody (Nicholls et al, 1995). In the absence of fluid replacement, diarrhea may induce rapid dehydration, hemoconcentration, and hypovolemic shock. Diarrhea may persist for 2 to 4 days.
    b) In a 15-year retrospective analysis of amatoxin mushroom poisoning (n=111), 102 (91.9%) patients developed diarrhea, with the average delay in onset of 16.7 +/- 1.8 hours (Giannini et al, 2007).
    c) CASE SERIES: In a study of 34 patients with amanita phalloides poisoning, diarrhea developed in 62% of patients; 22% experienced abdominal cramping (Krenova et al, 2007).
    d) In a retrospective review of 144 amatoxin poisonings, 82 patients developed diarrhea (Trabulus & Altiparmak, 2011).
    D) TOXIC MEGACOLON
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 71-year-old man inadvertently ingested mushrooms containing amanita phalloides, and developed typical gastrointestinal symptoms and laboratory evidence of severe hepatic damage. Although hepatic injury slowly resolved, the patient had evidence of toxic megacolon (ie, intractable diarrhea, dilatation of the intestine {6 cm noted in small intestine; 8 cm dilatation in the colon}). Antibiotic and systemic steroid therapy were not successful, but clinical improvement was observed with decompression of the colon via a catheter. No permanent sequelae was reported (Eyer et al, 2004).
    E) PANCREATITIS
    1) WITH POISONING/EXPOSURE
    a) Transient elevation of pancreatic enzyme levels, without the presence of clinical signs and symptoms of pancreatitis, were reported in a 6-year-old child approximately 5 days after ingesting the Galerina species of mushrooms (Kaneko et al, 2001).
    b) CASE REPORT: A 43-year-old woman, with a medical history significant for hepatitis B carrier status, developed fulminant hepatic failure with pancreatitis, coagulopathy, profound metabolic acidosis, and renal insufficiency, 36 hours after ingesting approximately 170 g of sauteed Lepiota subincarnata mushrooms. Following supportive care and a liver transplant, she was discharged home on day 12 (Mottram et al, 2010).
    F) ABDOMINAL PAIN
    1) WITH POISONING/EXPOSURE
    a) Abdominal pain developed in a patient after ingesting a meal containing Lepiota brunneoincarnata mushrooms (Kervegant et al, 2013).
    b) In a retrospective review of 144 amatoxin poisonings, 78 patients developed abdominal pain (Trabulus & Altiparmak, 2011).

Hepatic

    3.9.2) CLINICAL EFFECTS
    A) TOXIC HEPATITIS
    1) WITH POISONING/EXPOSURE
    a) Clinical signs of hepatocellular damage usually become evident only on the third to fourth day after ingestion. Clinical presentation may only include a mild jaundice and a mild hepatomegaly (Burton et al, 2002; Nicholls et al, 1995). Elevated liver enzyme levels are common, and sometimes are accompanied with coagulopathy (Stein et al, 2015; Gores et al, 2014; French et al, 2011; Aygul et al, 2010; Madhok et al, 2006; Lim et al, 2000; Cappell & Hassan, 1992; Scalzo et al, 1998; O'Brien & Khuu, 1996; Nicholson & Korman, 1997; Zevin et al, 1997; Aji et al, 1995; Serne et al, 1996; Ramirez et al, 1993).
    b) Fulminant hepatic failure, developing very quickly, often within 4 days, and requiring liver transplantation has been reported following severe intoxications (Stein et al, 2015; Hydzik et al, 2008; Donnelly et al, 2000; Zilker et al, 1999; Yamada et al, 1998).
    c) In a 15-year retrospective analysis of amatoxin mushroom poisoning (n=111), patients were graded according to the Poisoning Severity Score (PSS) and transaminase levels: PSS 1 (patients (n=62) with mild intoxication and transaminase of less than 1000 International Units/L); PSS 2 (patients (n=18) with moderate intoxication and transaminase of 1000 to 2000 International Units/L); PSS 3 (patients (n=31) with severe intoxication and transaminase of greater than 2000 International Units/L). The majority of patients reached the highest PSS 60 hours after mushroom ingestion. Two patients (graded as PSS 3) died; both patients were hospitalized very late (more than 60 hours after ingestion). The most clinically useful indicators of prognosis were hepatic transaminases and prothrombin activity (PTA). Overall, patients with PSS3 grade (higher transaminase levels) had lower PTA. In most patients, peak hepatic transaminase levels and minimum PTA were reached on day 3 postingestion. Patients treated within 36 hours of mushroom ingestion had lower transaminase peak, higher PTA, lower PSS grading, and earlier discharge from hospital. At follow-up, 12 to 180 months (89.6 +/- 6.7) after discharge, all patients had normal hepatic transaminase and PTA levels, including those classified as PSS 3 (Giannini et al, 2007).
    d) In severe cases, hepatitis follows an explosive course with marked jaundice, and hepatic coma. It may be accompanied by renal failure and cardiovascular collapse. In fatal cases death occurs within 6 to 16 days (mean 8 days). CDC (1997) reported 2 cases of poisoning, both developing hepatic encephalopathy and one developing renal failure, with death occurring 6 and 9 days after ingestion.
    e) PROGNOSTIC INDICATOR: In a small study (n=12; 4 fatalities; 8 recovered) of amanita phalloid poisonings, transaminase levels and severity of clinical features were examined. In all patients, transaminase levels were increased usually 2 days after exposure and the highest values were reached by days 4 to 6. In the group that died, the average value of AST was 4456 +/- 534 (range 2260-6328) and ALT was 3758 +/- 1054 (range 1870-6388) with a De Ritis index (AST to ALT ratio) generally higher than 1 (mean 1.41 +/- 0.28, range 0.96 to 1.6.). In the surviving group, the average value of AST was 271.5 +/- 110 (range 67-661) and ALT was 1235.37 +/- 212 (range 63-4641) with a De Ritis index of less than 1.0 (mean 0.32 +/- 0.0085; range 0.14 to 0.46). The authors concluded that aminotransferases are important biological markers, and suggested that monitoring transaminases and measuring their ratio may be of prognostic value (Petkovska et al, 2003).
    f) Liver biopsies carried out during the acute phase of the poisoning do NOT offer any substantial advantage over current clinical and biochemical criteria with respect to assessment of short- or medium-term prognosis. Prothrombin time appears to be a more useful prognostic marker for clinical outcome than serum aminotransferase levels, although close monitoring of both are recommended (Lim et al, 2000).
    1) Patients who recover from the acute stage of moderate to severe intoxication, have been reported to develop chronic active hepatitis (based on a study by Bartoloni, of 64 cases: 12.5% of all cases in the study, and 57.8% of those in the study classified with moderate to severe symptoms) (Bartoloni et al, 1985).
    2) In a study of 205 Amanita intoxications, hepatitis was present in 199 (97%). Fifty-two (25%) patients developed hepatic coma and the overall mortality was 22.4% (Floersheim et al, 1982).
    g) CASE REPORTS
    1) AMANITA BISPORIGERA
    a) CASE REPORT: An 18-year-old man unintentionally ingested 11 amanita bisporigera mushrooms, and developed hepatotoxicity. Laboratory findings approximately 20 hours after exposure included: aspartate aminotransferase (AST) 42 Units/L, total bilirubin 0.9 mg/dL, total protein 5.1 g/dL and ammonia level of 62 micromol/L. Due to concern of developing severe hepatotoxicity, the patient was transferred to a higher level of care. Severe liver dysfunction developed within 72 hours (AST 2459 U/L, ALT 3649, U/L, INR of 5.96, ammonia level of 67 micromol/L) and aggressive treatment included the placement of a nasobiliary drain to interrupt the enterohepatic circulation of the amatoxins. Total amatoxin (including alpha-amatoxin and beta-amatoxin) excreted from the bile was 4.03 mg over 3 days. Charcoal hemoperfusion was also performed to enhance elimination. The patient recovered completely with normal liver enzymes and was discharged to home on day 8 (Madhok et al, 2006).
    b) CASE REPORT: Fulminant hepatic failure with coagulopathy was reported in a teenager within one week of ingestion of 11 Amanita bisporigera mushrooms. ERCP was performed and charcoal hemoperfusion was instituted. Aggressive supportive therapy for hepatic failure was given, and the patient recovered and was discharged on hospital day 8 (Scalzo et al, 1998).
    c) CASE REPORT: A 55-year-old man presented with a 4-day history of nausea, vomiting, diarrhea, and progressive fatigue after ingesting an unknown amount of Amanita bisporigera mushrooms. Laboratory results revealed elevated serum creatinine 9.2 mg/dL (normal range, 0.6 to 1.3), elevated serum ALT (7846 Units/L; normal range, 20 to 65) and AST (2840 Units/L; normal range, 12 to 32), and elevated serum lipase level 1119 Units/L (normal range, 73 to 393). All other laboratory tests were normal. Following supportive care, including IV N-acetylcysteine and penicillin G, his condition gradually improved. All laboratory results were normal 3 weeks postingestion (Yarze & Tulloss, 2012).
    2) AMANITA PHALLOIDES
    a) CASE REPORT: A 58-year-old woman presented with an 8-hour history of diarrhea and vomiting a day after ingesting 6 boiled Amanita phalloides mushrooms. Despite aggressive treatment with IV silibinin 5 mg/kg 4 times daily, high-dose benzylpenicillin (1,000,000 International Units/kg/day), n-acetylcysteine, and IV fluids, she developed decompensated coagulopathic liver failure and later became encephalopathic. Five days postingestion, she died of fulminant liver failure (Lawton & Bhraonain, 2013).
    b) CASE REPORT: A 17-month-old boy presented with vomiting and diarrhea 9 hours after ingesting a meal containing Amanita phalloides mushrooms. All laboratory results were normal at presentation. Despite treatment with IV silibinin, activated charcoal, and vitamin K, his condition deteriorated rapidly and he developed hepatic encephalopathy stage II-III with somnolence (Glasgow coma score 13), coagulopathy, irritability, and inconsolable crying episodes. Based on his age, factor V levels (less than 10%), INR of 4.7, prothrombin time of greater than 100 ms, and persistent encephalopathy, both King's College and Clichy criteria for urgent liver transplantation were met 48 hours postingestion. Because of a catheter thrombus, hemofiltration with MARS (Molecular Adsorbent Recycling System) was not successful. While waiting for living donor liver transplantation, his condition continued to improve and he was discharged home 3 weeks postingestion. He did not have any signs of underlying or residual liver disease on follow-up visits (Garcia de la Fuente et al, 2011).
    c) CASE REPORT: A 65-year-old woman developed acute renal failure, hepatic encephalopathy, and hypoprothrombinemia after ingesting Amanita phalloides mushrooms. Following supportive care, her liver function recovered completely. However, acute renal failure persisted, necessitating hemodialysis (started on day 10). She still required hemodialysis at 2-year follow-up (Garrouste et al, 2009).
    d) CASE REPORT: A 9-year-old boy was admitted to the ED 2 days after ingestion of amanita phalloides with severe dehydration, hepatocellular necrosis and hypoglycemia. On the 4th day after ingestion, his clinical status declined with severe coagulopathy, coma and liver failure. He died on the 7th day with no signs of renal, cardiac or respiratory dysfunction (Langer et al, 1997).
    e) CASE REPORT: A 61-year-old woman ingested several mushrooms that had been frozen for approximately 7 to 8 months and presented, 36 hours later, with severe nausea, vomiting, and diarrhea. Laboratory analysis showed severe metabolic acidosis, serum creatinine of 248 mcmol/L, a prothrombin time of 47%, and elevated liver enzyme levels. A speculative diagnosis of Amanitas phalloides poisoning was confirmed due to urinary detection of amatoxin at a level of 37.3 mcg/L (measured approximately 4 days postingestion). Over the next 12 hours the patient's condition continued to deteriorate despite aggressive supportive measures. The patient died on hospital day 4 (approximately 6 days postingestion) from progressive liver and renal failure (Himmelmann et al, 2001).
    f) CASE SERIES: Severe hepatitis was reported in 8 patients following suspected Amanita phalloides ingestions. All 8 patients presented to the ED with profound nausea, vomiting, and diarrhea. Laboratory analysis showed ALT and AST elevations (mean peak levels were 7618 +/- 4037 International Units/L and 5488 +/- 4114 International Units/L, respectively), hyperbilirubinemia, and coagulopathy (mean peak prothrombin time was 31 seconds [ranging from 17 to 42 seconds]). All patients recovered without liver transplantation, following aggressive decontamination and supportive measures. It is believed that prevention of liver transplantation in these patients may have been due to early referral to a liver transplant center and early and aggressive supportive therapy (Rengstorff et al, 2003).
    g) CASE SERIES: In a study of 34 patients with amanita phalloides poisoning, hepatic failure with coagulopathy and encephalopathy developed in 24% of patients. One patient developed fulminant hepatic failure 2 days after ingesting mushrooms. He died of circulatory failure during liver transplantation (Krenova et al, 2007).
    h) In a retrospective review of 144 amatoxin poisonings, 14 patients with acute hepatic failure died (Trabulus & Altiparmak, 2011).
    3) GALERINA SPECIES
    a) CASE REPORT: Fulminant hepatic failure developed within 3 days after ingesting a Galerina species of mushroom. A liver biopsy, performed 90 hours postingestion, showed marked fatty degeneration and severe centrilobular necrosis. The patient gradually recovered, with normalization of liver enzyme levels, following plasma exchanges combined with continuous hemodiafiltration for 48 hours, as well as repeated administration of activated charcoal with a cathartic (Kaneko et al, 2001).
    4) LEPIOTA BRUNNEOINCARNATA
    a) CASE SERIES: A woman and her 2 children (an 8-year-old boy and an 11-year-old girl) ingested a meal containing Lepiota brunneoincarnata mushrooms and presented about 9 hours later with stomach cramps and vomiting. The mother reported that she ate most of the mushroom meal and her son ate a smaller portion and her daughter only had a bite. They had normal laboratory results and were discharged with a prescription for antiemetics and spasmolytic agents. The next day, the son's condition deteriorated and he developed asthenia, fever, abdominal pain, and watery diarrhea. He presented to a pediatric ED on day 4 with the same symptoms, as well as rectal bleeding, somnolence, and confusion. Laboratory results revealed major liver failure with elevated transaminase concentrations (aspartate transaminase, 1018 International Units/L; alanine transaminase, 3205 International Units/L) and a clotting disturbance (prothrombin time, 18 seconds [normal values: 10 to 14 seconds]). The mother and daughter also had transient minor liver impairment. All patients received activated charcoal, penicillin G (0.1 MIU/kg x 6/day for 12 hours), silibinin (5 mg/kg x 4/day for 24 hours), and N-acetylcysteine (IV loading dose for 150 mg/kg, followed by 50 mg/kg every 4 hours for 12 hours). They gradually improved and were discharged 11 days postingestion (Kervegant et al, 2013).
    5) LEPIOTA BRUNNEOLILACEA
    a) CASE REPORT: Meunier et al (1995) reported the case of a 27-year-old woman who presented to the ED 10 hours after ingestion of Lepiota brunneolilacea with nausea, vomiting and diarrhea. The following day the liver function tests were significantly abnormal except for a normal bilirubin. The prothrombin time was prolonged. Hepatocellular failure with coagulopathy developed rapidly. An EEG revealed grade 1 encephalopathy. A liver transplant was performed.
    6) LEPIOTA SUBINCARNATA
    a) CASE REPORT: A 43-year-old woman, with a medical history significant for hepatitis B carrier status, developed fulminant hepatic failure with pancreatitis, coagulopathy, profound metabolic acidosis, and renal insufficiency, 36 hours after ingesting approximately 170 g of sauteed Lepiota subincarnata mushrooms which she picked outside her home in Chicago. Following supportive care and a liver transplant, she was discharged home on day 12 (Mottram et al, 2010).
    7) OTHER
    a) CASE REPORT: A 3-year-old child developed fulminant hepatic failure with coagulopathy within 72 hours of mushroom ingestion. Despite aggressive therapy, her liver failure evolved into stage IV hepatic encephalopathy on day 5, and ventilator support was necessary. The patient died on day 11 of fulminant liver failure complicated by sepsis (O'Brien & Khuu, 1996).
    B) JAUNDICE
    1) WITH POISONING/EXPOSURE
    a) In a retrospective review of 144 amatoxin poisonings, 9 patients developed jaundice (Trabulus & Altiparmak, 2011).
    3.9.3) ANIMAL EFFECTS
    A) ANIMAL STUDIES
    1) HEPATOCELLULAR DAMAGE
    a) Mice given a single dose of alpha-amanitin, 0.6 mg/kg or 1.2 mg/kg, demonstrated hepatotoxicity based on liver histology and enzymes that were concentration dependent. Hyperbaric oxygen therapy provided no protection against liver damage (Thomas et al, 1997).

Genitourinary

    3.10.2) CLINICAL EFFECTS
    A) RENAL FAILURE SYNDROME
    1) WITH POISONING/EXPOSURE
    a) Two kinds of renal failure may be observed. During the gastrointestinal phase, a functional renal failure is frequently observed. It is associated with hypovolemia, and is secondary to fluid losses and hypoperfusion of the kidneys. This renal failure improves if dehydration and hypovolemia are treated rapidly and aggressively.
    b) In a retrospective review of 144 amatoxin poisonings, 19 patients developed acute renal failure; 5 patients developed oliguria-anuria (Trabulus & Altiparmak, 2011).
    c) It is sometimes possible to avoid acute tubular necrosis by the correct administration of fluids (Constantino et al, 1978) (Vesconi et al, 1985). A direct nephrotoxic effect of amatoxins is possible.
    d) AMANITA BISPORIGERA
    1) CASE REPORT: A 55-year-old man presented with a 4-day history of nausea, vomiting, diarrhea, and progressive fatigue after ingesting an unknown amount of Amanita bisporigera mushrooms. Laboratory results revealed elevated serum creatinine 9.2 mg/dL (normal range, 0.6 to 1.3), elevated serum ALT (7846 Units/L; normal range, 20 to 65) and AST (2840 Units/L; normal range, 12 to 32), and elevated serum lipase level 1119 Units/L (normal range, 73 to 393). All other laboratory tests were normal. Following supportive care, including IV N-acetylcysteine and penicillin G, his condition gradually improved. All laboratory results were normal 3 weeks postingestion (Yarze & Tulloss, 2012).
    e) AMANITA PHALLOIDES
    1) CASE REPORT: A 54-year-old man who ingested 2 to 4 Amanita phalloides mushrooms, developed necrotic liver, encephalopathy, severe coagulopathy, and acute renal failure. He was treated with 7 sessions of Molecular Absorbents Recirculating Systems (MARS) starting 63 hours after ingesting mushrooms, as well as standard supportive care (eg, fluid resuscitation, NAC, oral activated charcoal). He also underwent liver transplantation 9 days postingestion. After the surgery, he experienced persistent acute renal failure (requiring hemodialysis), rhabdomyolysis, and heparin-induced thrombocytopenia. He recovered and was discharged home after 128 days of hospitalization (Kantola et al, 2009).
    2) CASE SERIES: In a study of 34 patients with amanita phalloides poisoning, renal failure developed in 11% of patients (Krenova et al, 2007).
    3) Acute renal failure with anuria may occur at the third phase of poisoning, and may be accompanied by severe hepatitis with hepatic coma and hemorrhages, which is part of the hepatorenal syndrome. In one case severe oliguric renal failure occurred (approximately one week after ingestion) in conjunction with liver failure and ARDS (CDC, 1997). Nicholson & Korman (1997) reported a fatal case of severe renal impairment and marked lactic acidosis with hepatic failure and coagulopathies following an Amanita ingestion (Nicholson & Korman, 1997).
    a) CASE REPORT: A 27-year-old woman who ingested Lepiota brunneolilacea developed liver failure, requiring a liver transplant. Intravascular hemolysis with an associated renal failure developed 17 days after the liver transplant. Hemodialysis was preformed on 4 occasions in addition to transfusions of blood and packed cells and steroid therapy (Meunier et al, 1995).
    b) CASE REPORT: Progressively deteriorating renal function, necessitating continuous venovenous hemodialysis, occurred in a 62-year-old female who also developed hepatic failure, requiring liver transplantation, after ingesting several Lepiota josserandii mushrooms. It is believed that the patient's renal and hepatic failure were due to the amanitin contained within the mushrooms (Burton et al, 2002).
    4) CASE REPORT: A 65-year-old woman developed acute renal failure (serum creatinine concentration peaked at 929 mcmol/L on day 10), liver encephalopathy, and hypoprothrombinemia after ingesting Amanita phalloides mushrooms. Following supportive care, her liver function recovered completely. However, acute renal failure persisted, necessitating hemodialysis (started on day 10). On day 46, a kidney biopsy revealed the presence of massive acute tubular necrosis, mainly in the proximal convoluted tubule, severe tubular epithelial damage with tubular epithelial cell-shedding, apoptosis and tubular epithelium atrophy. Patient still required hemodialysis 2 years postingestion (Garrouste et al, 2009)
    5) CASE REPORT: A 61-year-old woman ingested several mushrooms that had been frozen for approximately 7 to 8 months and developed acute renal failure (approximately 48 hours postingestion), as well as progressive hepatic failure and hypoprothrombinemia. A diagnosis of Amanita phalloides poisoning was confirmed due to urinary detection of amatoxin at a level of 37.3 mcg/mL (measured approximately 4 days postingestion). Despite aggressive supportive care, the patient died on day 4 of hospitalization (approximately 6 days postingestion) (Himmelmann et al, 2001).
    6) CASE REPORT: Acute renal failure was reported in a patient following a suspected Amanita phalloides ingestion. The patient's renal function improved following 3 days of continuous veno-venous hemofiltration (Rengstorff et al, 2003).
    B) RENAL TUBULAR DISORDER
    1) WITH POISONING/EXPOSURE
    a) HYPOURICEMIA seen after A. phalloides poisoning in children is probably due to proximal tubule injury. A possible mechanism is tubular hypersecretion. The tubular injury/defect may endure long after liver function tests are normalized (Zawadzki et al, 1993).

Acid-Base

    3.11.2) CLINICAL EFFECTS
    A) ACIDOSIS
    1) WITH POISONING/EXPOSURE
    a) During the gastrointestinal phase, metabolic acidosis may occur as a result of bicarbonate losses from the gut. Metabolic acidosis, hypotension, and severe liver dysfunction were reported 6 days after ingestion of Amanita phalloides mushrooms (CDC, 1997) and within a week of ingestion of Amanita bisporigera (Scalzo et al, 1998).
    b) CASE REPORT: Severe metabolic acidosis (pH 7.05) occurred in a 61-year-old female several hours after ingesting several mushrooms that had been dried and frozen for approximately 7 to 8 months. The diagnosis of Amanita phalloides poisoning was confirmed by detection of amatoxin in the patient's urine (Himmelmann et al, 2001).
    c) CASE REPORT: Severe metabolic acidosis (pH 6.98, pCO2 17.9 mmHg, bicarbonate 10 mEq/L, lactate 10.9 mmol/L, anion gap 28 mEq/L) developed in a 43-year-old woman who also experienced fulminant hepatic failure with pancreatitis, coagulopathy, profound metabolic acidosis, and renal insufficiency, 36 hours after ingesting approximately 170 g of sauteed Lepiota subincarnata mushrooms. Following supportive care and a liver transplant, she was discharged home on day 12 (Mottram et al, 2010).
    B) LACTIC ACIDOSIS
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Lactic acidosis has been reported during the hepatic phase in a 90-year-old woman. The patient was in fulminant hepatic failure (Feinfeld et al, 1994).
    b) CASE REPORT: A 68-year-old man was reported with marked lactic acidosis and severe renal impairment during the hepatic phase of Amanita phalloides poisoning. He died 24 hours after hospital admission of multiorgan failure (Nicholson & Korman, 1997).

Hematologic

    3.13.2) CLINICAL EFFECTS
    A) BLOOD COAGULATION PATHWAY FINDING
    1) WITH POISONING/EXPOSURE
    a) Coagulation defects with hypofibrinogenemia and hypoprothrombinemia occur in hepatic failure and may result in local or general bleeding.
    b) Usually the earliest indication of coagulopathy is persistent bleeding from intravenous puncture sites (Bivins et al, 1985). Other types of bleeding include epistaxis and gastrointestinal hemorrhage.
    c) Manifested by marked coagulation defects secondary to impaired synthesis of clotting factors, this condition usually signifies a poor prognosis in that it indicates extensive hepatocellular damage (Floersheim, 1987; Scalzo et al, 1998; Serne et al, 1996).
    1) CASE REPORT: One study reported a case of a 27-year-old woman who developed severe hepatocellular failure associated with coagulopathy following ingestion of Lepiota brunneolilacea. Following a liver transplant, intravascular hemolysis developed with an associated renal failure (Meunier et al, 1995).
    d) CASE SERIES: A woman and her 2 children (an 8-year-old boy and an 11-year-old girl) ingested a meal containing Lepiota brunneoincarnata mushrooms and presented about 9 hours later with stomach cramps and vomiting. The mother reported that she ate most of the mushroom meal and her son ate a smaller portion and her daughter only had a bite. They had normal laboratory results and were discharged with a prescription for antiemetics and spasmolytic agents. The next day, the son's condition deteriorated and he developed asthenia, fever, abdominal pain, and watery diarrhea. He presented to a pediatric ED on day 4 with the same symptoms, as well as rectal bleeding, somnolence, and confusion. Laboratory results revealed major liver failure with elevated transaminase concentrations (aspartate transaminase, 1018 International Units/L; alanine transaminase, 3205 International Units/L) and a clotting disturbance (prothrombin time, 18 seconds [normal values: 10 to 14 seconds]). The mother and daughter also had transient minor liver impairment. All patients received activated charcoal, penicillin G (0.1 MIU/kg x 6/day for 12 hours), silibinin (5 mg/kg x 4/day for 24 hours), and N-acetylcysteine (IV loading dose for 150 mg/kg, followed by 50 mg/kg every 4 hours for 12 hours). They gradually improved and were discharged 11 days postingestion (Kervegant et al, 2013).
    e) CASE REPORT: A 54-year-old man who ingested 2 to 4 Amanita phalloides mushrooms, developed necrotic liver, encephalopathy, severe coagulopathy, and acute renal failure. He was treated with 7 sessions of Molecular Absorbents Recirculating Systems (MARS) starting 63 hours after ingesting mushrooms, as well as standard supportive care (eg, fluid resuscitation, NAC, oral activated charcoal). He also underwent liver transplantation 9 days postingestion. After the surgery, he experienced persistent acute renal failure (requiring hemodialysis), rhabdomyolysis, and heparin-induced thrombocytopenia. He recovered and was discharged home after 128 days of hospitalization (Kantola et al, 2009).
    f) CASE REPORT: A 65-year-old woman developed acute renal failure, liver encephalopathy, and hypoprothrombinemia after ingesting Amanita phalloides mushrooms. Following supportive care, her liver function recovered completely. However, acute renal failure persisted, necessitating hemodialysis (started on day 10). She still required hemodialysis at 2-year follow-up (Garrouste et al, 2009).
    g) CASE REPORT: A 61-year-old woman developed a decrease in prothrombin time, from 47% to 32% despite administration of vitamin K, approximately 2 days after ingesting several mushrooms that had been frozen for approximately 7 to 8 months. The speculative diagnosis of Amanitas phalloides was confirmed by urinary detection of amatoxin at a level of 37.3 mcg/L (measured approximately 4 days postingestion). Despite aggressive supportive care, the patient continued to deteriorate with a prothrombin time of less than 10% on day 3 of hospitalization (approximately 6 days postingestion) and subsequently died on day 4 of hospitalization (Himmelmann et al, 2001).
    h) CASE SERIES: In a study of 34 patients with amanita phalloides poisoning, hepatic failure with coagulopathy and encephalopathy developed in 24% of patients. One patient developed fulminant hepatic failure 2 days after ingesting mushrooms. He died of circulatory failure during liver transplantation (Krenova et al, 2007).
    i) CASE REPORT: A 33-year-old woman developed hepatic failure with coagulopathy and encephalopathy after ingesting Amanita phalloides mushrooms. She underwent liver dialysis and a successful liver transplantation was performed 6 days postingestion (Hydzik et al, 2008).
    j) CASE REPORT: A 71-year-old man presented with nausea, vomiting, diarrhea, and weakness within 48 hours of ingesting an unknown amount of Amanita mushrooms while camping in northwestern Iowa. Laboratory results revealed elevated liver enzymes, total bilirubin of 1.5 mg/dL (normal range, 0 to 1 mg/dL), INR of 1.3 (normal range, 0.8 to 1.2), and a serum creatinine of 2.7 mg/dL (normal range, 0.51 to 1.2 mg/dL). Despite treatment with IV N-acetylcysteine and penicillin G, he developed encephalopathy, acute renal injury, coagulopathy, and worsening transaminase elevations. He was transported to the liver transplant center and received multidose activated charcoal and IV silibinin (loading dose: 5 mg/kg over 1 hour, continued for 3 days at 20 mg/kg/day) starting 96 hours postingestion. His condition gradually improved by day 3 and he was discharged 5 days after presentation. On a follow-up visit a month later, his liver enzymes were normal (Gores et al, 2014).
    B) DISSEMINATED INTRAVASCULAR COAGULATION
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Disseminated intravascular coagulation (DIC) was observed in a patient who died from massive liver necrosis (Sanz et al, 1988).

Dermatologic

    3.14.2) CLINICAL EFFECTS
    A) JAUNDICE
    1) WITH POISONING/EXPOSURE
    a) Mild jaundice with sclera icterus is present in most patients at the hepatic phase of the intoxication. Marked jaundice occurs when hepatitis progresses to severe hepatic failure.

Endocrine

    3.16.2) CLINICAL EFFECTS
    A) HYPERINSULINISM
    1) WITH POISONING/EXPOSURE
    a) Serum insulin and C-peptide levels were elevated after ingestion (Kelner & Alexander, 1987).
    B) HYPOGLYCEMIA
    1) WITH POISONING/EXPOSURE
    a) In severe hepatic failure, glucose metabolism is often disturbed.
    1) Spontaneous hypoglycemia results from impaired glycogenolysis and gluconeogenesis (Bivins et al, 1985). Insulin and C-peptide concentrations were elevated in several patients (Kelner & Alexander, 1987).
    b) CASE REPORT: A 9-year-old boy was reported to have severe hypoglycemia and hepatic failure on the second day after ingestion of amanita phalloides (Langer et al, 1997).
    c) CASE REPORT: Hypoglycemia was reported in a 61-year-old woman who had ingested several mushrooms that had previously been frozen for approximately 7 to 8 months (Himmelmann et al, 2001).
    d) Hyperglycemia may also occur when highly concentrated dextrose solutions are infused.
    e) In in vivo and in vitro studies it was noted that amanita phalloides toxins can induce insulin release. Although the exact effect of amanita poisoning related to clinical presentation (ie, insulin levels and glucose metabolism) cannot be determined, the results indicate that amanita toxins can induce a direct insulin-releasing effect and a cytotoxic effect on beta cells (DeCarlo et al, 2003).
    f) In a retrospective review of 144 amatoxin poisonings, 8 patients developed hypoglycemia (Trabulus & Altiparmak, 2011).
    C) DISORDER OF ENDOCRINE SYSTEM
    1) WITH POISONING/EXPOSURE
    a) Serum calcitonin levels were elevated in conjunction with hypocalcemia in 4 patients who ingested alpha-amanitin containing mushrooms (Kelner & Alexander, 1987).
    D) HYPERPARATHYROIDISM
    1) WITH POISONING/EXPOSURE
    a) Serum parathyroid hormone was elevated at first (during hypocalcemia), but normalized when the hypocalcemia was corrected (Kelner & Alexander, 1987).
    E) HYPOTHYROIDISM
    1) WITH POISONING/EXPOSURE
    a) Serum thyroxine concentrations were depressed in 4 patients who ingested alpha-amanitin containing mushrooms (Kelner & Alexander, 1987).

Reproductive

    3.20.1) SUMMARY
    A) In pregnancy, the fetus may develop a toxic hepatitis. Because it is unknown whether amatoxins are excreted in breast milk, nursing should be stopped.
    3.20.3) EFFECTS IN PREGNANCY
    A) HEPATOCELLULAR DAMAGE
    1) FETAL DAMAGE/FIRST TRIMESTER is reported:
    a) CASE REPORT: Kaufmann et al (1978) reported an Amanita phalloides ingestion in a 25-year-old woman at 9 weeks gestation. The patient developed a toxic hepatitis (transaminase 1800 Units/L). After life-threatening maternal toxicity was treated, a therapeutic abortion was carried out at 12 weeks gestation. Histologic examination of fetal liver showed cellular damage related to amanitin toxicity. Amatoxins appear to cross the placental barrier (Kaufmann et al, 1978).
    2) Pregnant women, acutely poisoned with amanitans, have survived. However, there are often histochemical, histological, and immunomorphological changes that adversely impact the developing fetus (Hubenova & Stankova, 1992).
    B) GROWTH RETARDED
    1) CASE SERIES: Twenty-two women with cyclopeptide mushroom poisoning during pregnancy (5, 8, and 9 during the first, second and third trimester, respectively) were compared to controls (no mushroom poisoning). Twenty of the pregnancies went to term. Lower mean birth weights (but not gestational age) were reported in the mushroom-poisoned group as compared to the control series (Timar & Czeizel, 1997).
    C) LACK OF EFFECT
    1) FIRST TRIMESTER exposure resulted in NO fetal abnormalities.
    a) CASE REPORT: A 22-year-old woman in the 11th week of pregnancy was diagnosed with medium severity Amanita phalloides poisoning (eg, vomiting, diarrhea, persistent increases in liver enzymes, severe decreases in prothrombin factor V, II, VII, and X). Following treatment with intravenous hydration, silymarine and N-acetylcysteine, her condition gradually improved. On day 10, no fetal abnormalities were seen during an obstetric examination. She delivered a healthy baby in the 38th week of pregnancy (Boyer et al, 2001).
    2) SECOND TRIMESTER exposure resulted in NO fetal abnormalities in the acute phase of poisoning:
    a) CASE REPORT: A 26-year-old woman in her 23rd week of pregnancy was diagnosed with medium severity Amanita phalloides intoxication, requiring hydration, forced diuresis, and administration of silibinin, penicillin, thioctic acid, hydrocortisone, vitamin K and fresh frozen plasma. No fetal abnormalities were seen on sonographic and obstetric controls. A healthy baby was born in the 38th week of pregnancy (Nagy et al, 1994).
    b) CASE REPORT: A 27-year-old woman who was 20 weeks pregnant was admitted for amanita poisoning and developed hepatic encephalopathy. Despite aggressive supportive care and extracorporeal detoxification by hemodialysis plus hemoperfusion (performed daily), the patient's clinical status (hepatic and neuro function) did not improve. Uterine contraction was noted on day 5, with threatened abortion anticipated. The patient then underwent three courses of albumin dialysis using MARS intermittently starting from the 8th day of hospitalization. Liver function was noted to immediately improve, as well as clinical features; uterine contraction ceased. The patient was discharged with normal liver function and fetal evaluation appeared normal. Upon follow-up, a healthy baby was born at 36 weeks and had normal development at 7 months (Wu & Wang, 2004).
    3) THIRD TRIMESTER exposure resulted in lack of effect in the fetus:
    a) CASE REPORT: A 21-year-old woman in the eighth month of pregnancy, ingested Amanita phalloides. Blood tested positive for amatoxin, but the amniotic fluid tested negative. She gave birth to a healthy baby without biochemical evidence of hepatotoxicity (Belliardo et al, 1983).
    b) CASE REPORT: Four family members presented with gastrointestinal symptoms 2 days after ingesting Amanita phalloides mushrooms. One patient, a pregnant woman (at 39 weeks' gestation) developed only elevated liver enzymes and was treated with silibin (2 g/24 hour for 3 days). She delivered a healthy baby who had a slight temporary increase in liver enzymes activity. All samples of amniotic fluid, umbilical vein blood, peripheral venous blood, and urine of mother and newborn tested negative for amatoxin. Two family members died from liver failure and lethal brain edema (Wacker et al, 2009).
    3.20.4) EFFECTS DURING BREAST-FEEDING
    A) BREAST MILK
    1) Because it is unknown whether amatoxins are excreted in breast milk, breast feeding mothers who have ingested Amanita, asymptomatic or not, should be advised to stop nursing until it is determined whether or not they have been poisoned.

Monitoring Parameters Levels

    4.1.1) SUMMARY
    A) Monitor serum glucose frequently in patients with evidence of hepatic injury. Monitor serial serum electrolytes, renal function, liver enzymes, coagulation studies, bilirubin, and urine output.
    B) AST, ALT, LDH, and serum bilirubin elevations are the earliest and best indicators of liver damage. Hepatic enzymes typically peak between the 60th and 72nd hours and then decrease. Transaminases may be low in cases of massive hepatic necrosis.
    C) Glucose, fibrinogen, and prothrombin time are the best indicators of hepatic failure.
    D) Amatoxins are detected in biologic fluids such as serum, urine, and emesis by radioimmunoassay or HPLC. These methods are not generally available in clinical practice. Serum levels of amatoxins do not correlate with the severity of symptoms of poisoning.
    4.1.2) SERUM/BLOOD
    A) SPECIFIC AGENT
    1) Serum levels of amatoxins do not correlate with severity of poisoning and are probably not clinically useful. Generally, a short latency period between the mushroom ingestion and the first clinical symptoms indicates serious poisoning (Sabeel et al, 1995).
    2) In a group of 29 patients studied by Vesconi et al (1985), serum amatoxins were detectable in 65% of the patients (Vesconi et al, 1985) even as late as 30 hours postingestion. Concentrations were 0.5 to 24 ng/mL. Only 4 of 19 had concentrations greater than 3 ng/mL.
    3) One study found serum concentrations of alpha amanitin between 70 and 90 ng/mL in patients with Amanita phalloides intoxication (Pastorello & Tolentino, 1982).
    4) One source found alpha and beta amanitin in serum only in one-third of the patients (Jaeger et al, 1988).
    a) Concentrations were mean 73 +/- 69 ng/mL for alpha amanitin (range 8 to 190 ng/mL) and mean 55.6 +/- 60 ng/mL for beta-amanitin (range 16 to 162 ng/mL).
    b) In all cases except one, amanitins were only found in serum before the 48th hour following ingestion.
    c) In 45 patients, amatoxins were found in the plasma of 11 and the urine of 22. Amatoxins were present in the plasma until the 36th hour, but could be found in urine for up to 4 days using HPLC (Jaeger et al, 1989).
    5) One study found amatoxins in the serum of intoxicated patients up to 36 hours postingestion (Busi et al, 1979).
    6) Another study detected amatoxins in the serum of intoxicated patients in 70% of the cases before 12 hours postingestion and in 50% of the cases before 24 hours postingestion (Velvaert & Schlatter-Lanz, 1982).
    7) The Meixner test, a simple colorimetric spot test for detection of amatoxin (sensitive with samples as small as 2 mcg), had a 100% rate of false-positive reactions with samples containing psilocybin and 5-substituted tryptamines in one study. The findings indicate that the test has limited clinical utility because of the false-positive readings. In addition, a negative reading is not indicative that a mushroom is safe; potential toxicity following ingestion may still exist (Beuhler et al, 2004).
    B) BLOOD/SERUM CHEMISTRY
    1) Elevated AST, ALT, LDH, and serum bilirubin are the first and best indicators of liver damage and should be monitored throughout the course of the illness. Lim et al (2000) reported that initial serum aminotransferase levels may not predict clinical outcome, but that prothrombin time may be a more useful prognostic marker, although close monitoring of both are recommended.
    a) Hepatic enzymes usually reach a peak between the 60th and 72nd hours and then decrease. Enzyme activity may be low in massive liver necrosis (Bivins et al, 1985).
    b) Alkaline phosphatase (AP) activity is often within normal limits. An AP ISO enzyme study will show increases in the hepatic-2 fraction as an indicator of severity of hepatic damage (Parra et al, 1992).
    c) Szponar et al (1999) reported that decreased prothrombin index, significant increase of bilirubin, and increased ALT, AST and AFP levels correlated with the clinical stage of illness.
    d) PROGNOSTIC INDICATOR: In a small study (n=12; 4 fatalities; 8 recovered) of amanita phalloid poisonings, transaminase levels and severity of clinical features were examined. In all patients, increased transaminase levels were increased usually 2 days after exposure and the highest values were reached by days 4 to 6. In the group that died, the average value of AST was 4456 +/- 534 (range 2260-6328) and ALT was 3758 +/- 1054 (range 1870-6388) with a De Ritis index (AST to ALT ratio) generally higher than 1 (mean 1.41 +/- 0.28, range 0.96 to 1.6.). In the surviving group, the average value of AST was 271.5 +/- 110 (range 67-661) and ALT was 1235.37 +/- 212 (range 63-4641) with a De Ritis index of less than 1.0 (mean 0.32 +/- 0.0085; range 0.14 to 0.46). The authors concluded that aminotransferases are important biological markers, and suggested that monitoring transaminases and measuring their ratio may be of prognostic value (Petkovska et al, 2003).
    2) Monitor serum electrolytes, urea, and serum creatinine in order to rapidly detect hypokalemia, hypochloronatremia, metabolic acidosis, and renal failure.
    3) Serum glucose should be monitored hourly at the bedside in order to detect and to correct rapidly developing hypoglycemia (Bivins et al, 1985).
    C) ACID/BASE
    1) Monitor blood gases to rapidly detect metabolic acidosis.
    D) COAGULATION STUDIES
    1) Monitor coagulation parameters (INR or PT), especially the clotting factors synthesized by the liver, i.e., fibrinogen, prothrombin. A prothrombin level below 10% is an index of poor prognosis (Floersheim, 1987; Lim et al, 2000).
    4.1.3) URINE
    A) URINARY LEVELS
    1) URINE AMATOXIN LEVELS: In the same group of patients, urine concentrations ranged from 0.5 to 56 ng/mL. Initial catheterization to obtain concentrated urine found levels as high as 180 ng/mL (Vesconi et al, 1985).
    a) In serial determinations, urinary amatoxins were found 24 to 66 hours postingestion. Total urinary output of amatoxins in this period ranged from 12,000 to 23,000 ng.
    2) In the same group of patients, amatoxins were detected in urine in 15 of 24 (62%) patients. Alpha amanitin was present 14 times and beta amanitin 10 times (Jaeger et al, 1993; Jaeger et al, 1988).
    a) In some cases, serial determinations showed that amanitins were excreted up to 72 to 96 hours postingestion.
    b) Concentrations as high as 4800 ng/mL (alpha amanitin) and 4300 ng/mL (beta amanitin) were observed.
    c) Mean total amounts excreted in urine: alpha amanitin (0.95 mg) and beta amanitin (1.7 mg).
    3) Amatoxins were detected by radioimmunoassay in urine of 100% of the cases tested before the 24th hour and in 80% of the cases tested before the 48th hour postingestion (Velvaert & Schlatter-Lanz, 1982).
    4) Using HPLC, Jaeger et al (1993) found amatoxins in the urine until day 4 (Jaeger et al, 1993).
    5) In a 15-year retrospective analysis of amatoxin mushroom poisoning (n=111), the toxin was identified in the urine of only 62 patients (55.9%). The patients in whom amitoxin was not detected in urine (n=49) were hospitalized later (40.8 +/- 4.5 hours postingestion) than those whose urine tested positive (35.1 +/- 2.5 hours postingestion) (p less than 0.05). The authors suggested that a negative result in urine collected more than 36 hours after mushroom ingestion cannot rule out poisoning (Giannini et al, 2007).
    4.1.4) OTHER
    A) OTHER
    1) EEG
    a) In patients presenting with seizures, EEG should be considered.
    2) 13C-METHACETIN BREATH TEST (13C-MBT)
    a) 13C-MBT has been used experimentally to evaluate cytochrome P450-dependent liver function in patients with Amanita phalloides poisoning. Methacetin, a substrate that undergoes extensive first-pass clearance, is metabolized by cytochrome P450 (isoform CYP1A2) into carbon dioxide by single dealkylation. The appearance of 13CO2 in exhaled breath suggests that 13C-methacetin has undergone oxidation. Exhaled breath samples are collected and 13C content are measured using an isotope ratio mass spectrometer. The breath test shows a cumulative percentage of the administered dose of 13C recovered over time, which corresponds to 13C-methacetin cumulative oxidation percentage and reflects metabolic liver capacity (Hydzik et al, 2008).
    1) A woman with liver injury after ingesting Amanita phalloides mushrooms received 13C-methacetin through a nasogastric tube, before the first and after the second liver dialysis sessions. Her results were compared with the result of a control group. The breath test results revealed that 40 min after 13C-methacetin ingestion, the mean cumulative oxidation percentage for the patient was 0.09% on the fourth day and 0.02% on the fifth day after mushroom ingestion; however, this parameter was 10.5 +/- 3.8% in healthy controls (Hydzik et al, 2008).

Radiographic Studies

    A) MULTIDETECTOR COMPUTED TOMOGRAPHY (MDCT)
    1) Precontrast MDCT images of 3 children with mushroom poisoning revealed diffuse reduction of hepatic attenuation compared with the spleen. Homogeneous contrast enhancement of the liver was observed on contrast-enhanced MDCT images. All children recovered following supportive treatment. A follow-up precontrast MDCT image of one patient showed normal hepatic density (Cakir et al, 2007).

Methods

    A) OTHER
    1) AMATOXINS IN MUSHROOMS
    a) WIELAND TEST (Meixner Test) (Meixner, 1979) -
    1) Perform indoors away from sunlight and excessive heat.
    2) Squeeze a drop of juice from fresh mushroom tissue onto a piece of pulp paper such as newsprint. A garlic press is recommended. If only a small fragment of tissue is available, mash it onto the paper so the juice will be absorbed.
    3) Encircle the wet spot with a pencil to mark location.
    4) Dry the spot with gentle warm air such as from a hair dryer.
    5) Add a drop of concentrated hydrochloric acid to the dry spot. The presence of amatoxins is indicated by the formation of a blue color.
    a) Test a control solution (without amatoxins) on paper outside the circle to eliminate the possibility of newsprint that ordinarily turns blue after exposure to hydrochloric acid; false positives do occur.
    b) A false positive reaction may also occur by drying the spot at temperatures greater than 63 degrees C or by exposure to sunlight (Lampe & McCann, 1987).
    c) Psilocybin, bufotenine, and certain terpenes will also give false positives.
    d) Run a similar test on filter paper which is lignin-free. Lignin-free paper will NOT elicit the response even if the amatoxin is present (Lampe & McCann, 1987).
    6) For dry mushrooms, crush a part in absolute methanol and apply a drop of the methanol to the paper.
    7) This test may NOT be used on mushroom parts that have been removed from the stomach (Lampe & McCann, 1987).
    8) The test's sensitivity is 0.2 mg/mL (Lampe & McCann, 1987).
    b) MELZER'S REAGENT -
    1) Melzer's reagent is used to detect an amyloid reaction in cyclopeptide containing Amanitas.
    2) REAGENT CONTAINS: 1.5 g iodine, 5 g potassium iodide, 100 g (mL) chloral hydrate; add to 100 mL of warm water. Do NOT boil.
    3) A drop of this solution is placed on the spores of suspected mushrooms. The cyclopeptide-containing species spores turn blue (amyloid), other mushroom types turn brown (dextrinoid) or do NOT change color (non-amyloid) (Lincoff & Mitchell, 1977).
    c) OTHER METHODS
    1) Column chromatography procedure on Sephadex LH-20 followed by thin-layer chromatography has been used for the determination of amatoxins in A. phalloides (Faulstich et al, 1975).
    2) One study applied high-performance thin-layer chromatography for the determination of alpha, beta, and gamma amanitins in A. phalloides and A. verna (Seeger & Stijve, 1979).
    B) IMMUNOASSAY
    1) Radioimmunoassay methods for detecting amatoxins in biologic fluids have been developed (Andres & Frei, 1987; Faulstich et al, 1975; pp 1233-1234).
    a) Antibodies against amanitin were obtained in rabbits (Faulstich et al, 1975; Andres & Frei, 1987) or in rats (pp 1233-1234).
    b) The test used either 3H tracer (Faulstich et al, 1975; pp 1233-1234) or 125I tracer (Andres & Frei, 1987).
    c) The sensitivity of radioimmunoassay is 0.1 ng/mL for serum and 0.25 ng/mL for urine (Vesconi et al, 1985; Fiume, 1980); 0.1 ng/mL for serum and 1 mg/mL for urine (Andres & Frei, 1987).
    C) CHROMATOGRAPHY
    1) HPLC assays have been developed for detection of alpha and beta amanitins in human serum, urine, or gastrointestinal fluids (Pastorello & Tolentino, 1982; Jehl et al, 1985; Caccialanza et al, 1985).
    a) Sensitivity is 6 to 10 mg/mL (Jehl et al, 1985; Caccialanza et al, 1985).
    2) One study described a validated electrospray liquid chromatographic-mass spectrometric assay method for the determination of alpha- and beta-amanitin in urine following immunoaffinity extraction (IAE-LC-MS). Level of detection and limit of quantification for a- and b-amanitin was reported to be 2.5 ng/mL and 5.0 ng/mL, respectively (Maurer et al, 2000).

Life Support

    A) Support respiratory and cardiovascular function.

Patient Disposition

    6.3.1) DISPOSITION/ORAL EXPOSURE
    6.3.1.1) ADMISSION CRITERIA/ORAL
    A) If a patient develops a gastroenteritis syndrome 6 to 24 hours after ingestion of mushrooms, one should ALWAYS consider that this patient may be poisoned by amatoxin-containing mushrooms. The patient should immediately be admitted and treated.
    B) If a group of people ate the same type of mushroom and one presents with symptoms, consider the possibility that the others who are NOT yet symptomatic also may have been poisoned and will require treatment (Bivins et al, 1985).
    C) Severity classifications (4 grades)
    1) Grade 1: GI upset only, no indications of liver or kidney failure. Symptomatic treatment only (Spoerke & Rumack, 1994).
    2) Grade 2: Show all signs of intoxication, with a mild to moderate rise in transaminases (less than 500 units/L). Symptomatic treatment only (Spoerke & Rumack, 1994).
    3) Grade 3: Severe hepatic damage with a great increase in transaminases (> 500 units/L), plus an impaired plasma clotting function (e.g. a prolonged prothrombin time) (Spoerke & Rumack, 1994). Can be divided into two groups based on bilirubin values:
    a) Grade 3a: Bilirubin rise is mild or absent (Spoerke & Rumack, 1994).
    b) Grade 3b: Bilirubin rise is steep and continuous (> 5 mg/dL) (Spoerke & Rumack, 1994).
    c) These patients (especially Grade 3b), are at risk and should be transferred to a facility where liver transplant is possible (Spoerke & Rumack, 1994).
    4) Grade 4: Steep rise in transaminases, accompanied by a steep decline in clotting function, a steep rise in bilirubin, and the onset of kidney dysfunction. These patients have a poor prognosis, and many die in spite of intensive care (Spoerke & Rumack, 1994).
    6.3.1.2) HOME CRITERIA/ORAL
    A) Unless the potentially lethal mushroom species can be definitely ruled out, there should be no home treatment for mushroom intoxications.
    6.3.1.3) CONSULT CRITERIA/ORAL
    A) Mushroom identification is best performed by a mycologist. If a sample of the mushroom can be retrieved, then it should be analyzed by a mycologist. After general treatment for poisonings has been administered, the regional poison center should be contacted for treatment recommendations. For those patients who develop hepatic failure, a transplant center should be notified to evaluate the patient for a possible liver transplantation.

Monitoring

    A) Monitor serum glucose frequently in patients with evidence of hepatic injury. Monitor serial serum electrolytes, renal function, liver enzymes, coagulation studies, bilirubin, and urine output.
    B) AST, ALT, LDH, and serum bilirubin elevations are the earliest and best indicators of liver damage. Hepatic enzymes typically peak between the 60th and 72nd hours and then decrease. Transaminases may be low in cases of massive hepatic necrosis.
    C) Glucose, fibrinogen, and prothrombin time are the best indicators of hepatic failure.
    D) Amatoxins are detected in biologic fluids such as serum, urine, and emesis by radioimmunoassay or HPLC. These methods are not generally available in clinical practice. Serum levels of amatoxins do not correlate with the severity of symptoms of poisoning.

Oral Exposure

    6.5.1) PREVENTION OF ABSORPTION/PREHOSPITAL
    A) ACTIVATED CHARCOAL
    1) PREHOSPITAL ACTIVATED CHARCOAL ADMINISTRATION
    a) Consider prehospital administration of activated charcoal as an aqueous slurry in patients with a potentially toxic ingestion who are awake and able to protect their airway. Activated charcoal is most effective when administered within one hour of ingestion. Administration in the prehospital setting has the potential to significantly decrease the time from toxin ingestion to activated charcoal administration, although it has not been shown to affect outcome (Alaspaa et al, 2005; Thakore & Murphy, 2002; Spiller & Rogers, 2002).
    1) In patients who are at risk for the abrupt onset of seizures or mental status depression, activated charcoal should not be administered in the prehospital setting, due to the risk of aspiration in the event of spontaneous emesis.
    2) The addition of flavoring agents (cola drinks, chocolate milk, cherry syrup) to activated charcoal improves the palatability for children and may facilitate successful administration (Guenther Skokan et al, 2001; Dagnone et al, 2002).
    2) CHARCOAL DOSE
    a) Use a minimum of 240 milliliters of water per 30 grams charcoal (FDA, 1985). Optimum dose not established; usual dose is 25 to 100 grams in adults and adolescents; 25 to 50 grams in children aged 1 to 12 years (or 0.5 to 1 gram/kilogram body weight) ; and 0.5 to 1 gram/kilogram in infants up to 1 year old (Chyka et al, 2005).
    1) Routine use of a cathartic with activated charcoal is NOT recommended as there is no evidence that cathartics reduce drug absorption and cathartics are known to cause adverse effects such as nausea, vomiting, abdominal cramps, electrolyte imbalances and occasionally hypotension (None Listed, 2004).
    b) ADVERSE EFFECTS/CONTRAINDICATIONS
    1) Complications: emesis, aspiration (Chyka et al, 2005). Aspiration may be complicated by acute respiratory failure, ARDS, bronchiolitis obliterans or chronic lung disease (Golej et al, 2001; Graff et al, 2002; Pollack et al, 1981; Harris & Filandrinos, 1993; Elliot et al, 1989; Rau et al, 1988; Golej et al, 2001; Graff et al, 2002). Refer to the ACTIVATED CHARCOAL/TREATMENT management for further information.
    2) Contraindications: unprotected airway (increases risk/severity of aspiration) , nonfunctioning gastrointestinal tract, uncontrolled vomiting, and ingestion of most hydrocarbons (Chyka et al, 2005).
    6.5.2) PREVENTION OF ABSORPTION
    A) SUMMARY: Administer activated charcoal 25 to 50 g in adults and usually 25 g in children (1 to 12 years), and 1 g/kg in infants. Multiple dose activated charcoal may enhance elimination. After 50 g initial dose, give subsequent doses in 4-hour intervals at a rate of 12.5 g/hr. For children, after the initial dose of 25 g, administer subsequent doses at 4-hour intervals at a rate equivalent to 6.25 g/hr. Evaluate frequently for the presence of bowel sounds or signs of obstruction. Because this ingestion is potentially life-threatening, gastric lavage is indicated if the patient has not vomited spontaneously and it can be performed within a few hours of ingestion. Nasogastric suction between doses of charcoal may remove toxins eliminated in the bile.
    B) ACTIVATED CHARCOAL
    1) CHARCOAL ADMINISTRATION
    a) Consider administration of activated charcoal after a potentially toxic ingestion (Chyka et al, 2005). Administer charcoal as an aqueous slurry; most effective when administered within one hour of ingestion.
    2) CHARCOAL DOSE
    a) Use a minimum of 240 milliliters of water per 30 grams charcoal (FDA, 1985). Optimum dose not established; usual dose is 25 to 100 grams in adults and adolescents; 25 to 50 grams in children aged 1 to 12 years (or 0.5 to 1 gram/kilogram body weight) ; and 0.5 to 1 gram/kilogram in infants up to 1 year old (Chyka et al, 2005).
    1) Routine use of a cathartic with activated charcoal is NOT recommended as there is no evidence that cathartics reduce drug absorption and cathartics are known to cause adverse effects such as nausea, vomiting, abdominal cramps, electrolyte imbalances and occasionally hypotension (None Listed, 2004).
    b) ADVERSE EFFECTS/CONTRAINDICATIONS
    1) Complications: emesis, aspiration (Chyka et al, 2005). Aspiration may be complicated by acute respiratory failure, ARDS, bronchiolitis obliterans or chronic lung disease (Golej et al, 2001; Graff et al, 2002; Pollack et al, 1981; Harris & Filandrinos, 1993; Elliot et al, 1989; Rau et al, 1988; Golej et al, 2001; Graff et al, 2002). Refer to the ACTIVATED CHARCOAL/TREATMENT management for further information.
    2) Contraindications: unprotected airway (increases risk/severity of aspiration) , nonfunctioning gastrointestinal tract, uncontrolled vomiting, and ingestion of most hydrocarbons (Chyka et al, 2005).
    C) GASTRIC LAVAGE
    1) INDICATIONS: Consider gastric lavage with a large-bore orogastric tube (ADULT: 36 to 40 French or 30 English gauge tube {external diameter 12 to 13.3 mm}; CHILD: 24 to 28 French {diameter 7.8 to 9.3 mm}) after a potentially life threatening ingestion if it can be performed soon after ingestion (generally within 60 minutes).
    a) Consider lavage more than 60 minutes after ingestion of sustained-release formulations and substances known to form bezoars or concretions.
    2) PRECAUTIONS:
    a) SEIZURE CONTROL: Is mandatory prior to gastric lavage.
    b) AIRWAY PROTECTION: Place patients in the head down left lateral decubitus position, with suction available. Patients with depressed mental status should be intubated with a cuffed endotracheal tube prior to lavage.
    3) LAVAGE FLUID:
    a) Use small aliquots of liquid. Lavage with 200 to 300 milliliters warm tap water (preferably 38 degrees Celsius) or saline per wash (in older children or adults) and 10 milliliters/kilogram body weight of normal saline in young children(Vale et al, 2004) and repeat until lavage return is clear.
    b) The volume of lavage return should approximate amount of fluid given to avoid fluid-electrolyte imbalance.
    c) CAUTION: Water should be avoided in young children because of the risk of electrolyte imbalance and water intoxication. Warm fluids avoid the risk of hypothermia in very young children and the elderly.
    4) COMPLICATIONS:
    a) Complications of gastric lavage have included: aspiration pneumonia, hypoxia, hypercapnia, mechanical injury to the throat, esophagus, or stomach, fluid and electrolyte imbalance (Vale, 1997). Combative patients may be at greater risk for complications (Caravati et al, 2001).
    b) Gastric lavage can cause significant morbidity; it should NOT be performed routinely in all poisoned patients (Vale, 1997).
    5) CONTRAINDICATIONS:
    a) Loss of airway protective reflexes or decreased level of consciousness if patient is not intubated, following ingestion of corrosive substances, hydrocarbons (high aspiration potential), patients at risk of hemorrhage or gastrointestinal perforation, or trivial or non-toxic ingestion.
    D) MULTIPLE DOSE ACTIVATED CHARCOAL
    1) Multiple dose activated charcoal is recommended by some authors (Buchwald, 1989) and its use is supported by evidence of the enterohepatic circulation of the amatoxins (Giannini et al, 2007; Busi et al, 1979; Vesconi et al, 1985; Faulstich et al, 1985; O'Brien & Khuu, 1996). It has never been shown to affect outcome or decrease mortality.
    2) MULTIPLE DOSE ACTIVATED CHARCOAL
    a) ADULT DOSE: Optimal dose not established. After an initial dose of 50 to 100 grams of activated charcoal, subsequent doses may be administered every 1, 2 or 4 hours at a dose equivalent to 12.5 grams/hour (Vale et al, 1999), do not exceed: 0.5 g/kg charcoal every 2 hours (Ghannoum & Gosselin, 2013; Mauro et al, 1994). There is some evidence that smaller more frequent doses are more effective at enhancing drug elimination than larger less frequent doses (Park et al, 1983; Ilkhanipour et al, 1992). PEDIATRIC DOSE: Optimal dose not established. After an initial dose of 25 to 50 grams in children aged 1 to 12 years (or 0.5 to 1 gram/kilogram body weight) (Chyka & Seger, 1997), subsequent doses may be administered every 1, 2 or 4 hours (Vale et al, 1999) in a dose equivalent to 6.25 grams/hour in children 1 to 12 years old.
    b) Activated charcoal should be continued until the patient's clinical and laboratory parameters, including drug concentrations if available, are improving (Vale et al, 1999). The patient should be frequently assessed for the ability to protect the airway and evidence of decreased peristalsis or intestinal obstruction.
    c) Use of cathartics has not been shown to increase drug elimination and may increase the likelihood of vomiting. Routine coadministration of a cathartic is NOT recommended (Vale et al, 1999).
    d) AGENTS AMENABLE TO MDAC THERAPY: The following properties of a drug that are likely to allow MDAC therapy to be effective include: small volume of distribution, low protein binding, prolonged half-life, low intrinsic clearance, and a nonionized state at physiologic pH (Chyka, 1995; Ghannoum & Gosselin, 2013).
    e) Vomiting is a common adverse effect; antiemetics may be necessary.
    f) CONTRAINDICATIONS: Absolute contraindications include an unprotected airway, intestinal obstruction, a gastrointestinal tract that is not intact and agents that may increase the risk of aspiration (eg, hydrocarbons). Relative contraindications include decreased peristalsis (eg, decreased bowel sounds, abdominal distention, ileus, severe constipation) (Vale et al, 1999; Mauro et al, 1994).
    g) COMPLICATIONS: Include constipation, intestinal bleeding, bowel obstruction, appendicitis, charcoal bezoars, and aspiration which may be complicated by acute respiratory failure, adult respiratory distress syndrome or bronchiolitis obliterans (Ghannoum & Gosselin, 2013; Ray et al, 1988; Atkinson et al, 1992; Gomez et al, 1994; Mizutani et al, 1991; Pollack et al, 1981; Harris & Filandrinos, 1993; Elliot et al, 1989; Mina et al, 2002; Harsch, 1986; Rau et al, 1988; Golej et al, 2001; Graff et al, 2002).
    E) NASOGASTRIC SUCTION
    1) Intermittent gastroduodenal aspiration (between charcoal administrations) may be indicated in order to remove toxins eliminated in bile (Busi et al, 1979; Jaeger et al, 1988).
    6.5.3) TREATMENT
    A) SUPPORT
    1) MANAGEMENT OF TOXICITY
    a) All patients with cyclopeptide mushroom ingestion have the potential to develop severe toxicity. Administer activated charcoal for recent ingestions, and consider multiple dose activated charcoal or nasogastric suction to interrupt enterohepatic recirculation of the toxins. N-acetylcysteine may be beneficial and is associated with little toxicity. Silibinin is currently undergoing a clinical trial for amatoxin poisoning. In studies, NAC and silibinin were the most effective agents. High-dose penicillin G has also been used. All of these therapies should be instituted as soon as possible in patients with the potential for significant toxicity.
    B) MONITORING OF PATIENT
    1) Monitor serum glucose frequently in patients with evidence of hepatic injury. Monitor serial serum electrolytes, renal function, liver enzymes, coagulation studies, bilirubin, and urine output.
    2) AST, ALT, LDH, and serum bilirubin elevations are the earliest and best indicators of liver damage. Hepatic enzymes typically peak between the 60th and 72nd hours and then decrease. Transaminases may be low in cases of massive hepatic necrosis.
    3) Glucose, fibrinogen, and prothrombin time are the best indicators of hepatic failure.
    4) Amatoxins are detected in biologic fluids such as serum, urine, and emesis by radioimmunoassay or HPLC. These methods are not generally available in clinical practice. Serum levels of amatoxins do not correlate with the severity of symptoms of poisoning.
    C) FLUID/ELECTROLYTE BALANCE REGULATION
    1) Treatment of symptomatic patients may require vigorous and immediate correction of dehydration and hypovolemia. It is essential in these cases in order to prevent renal failure and start forced diuresis (Vesconi et al, 1985; Bivins et al, 1985).
    2) Administration of plasma expanders and fluids should be guided by monitoring of blood pressure, central venous pressure, and urinary output.
    3) Correction of hypokalemia (by potassium chloride diluted in solutions of dextrose 5%, or NaCl 0.9%) and of metabolic acidosis (by sodium bicarbonate solution 1.4%) should be guided by repeated laboratory analyses.
    D) ANTIDOTE
    1) SUMMARY
    a) There are several drugs that have been proposed as antidotes in the management of cyclopeptide containing mushroom poisoning. In studies, silibinin and NAC each administered as monotherapy, and silibinin with benzylpenicillin as bi-, tri-, and polytherapies were associated with the lowest mortalities. Overall, NAC and silibinin were the most effective agents.
    b) N-ACETYLCYSTEINE: High doses of N-acetylcysteine have been used in humans in the setting of amatoxin poisoning. The dose most often used is 150 mg/kg infusion over 60 minutes followed by 50 mg/kg infusion over 4 hours followed by 6.25 mg/kg/hour infusion which is continued until the patient has clinically improved.
    c) SILIBININ: Silibinin is being studied as a potential antidote for amatoxin. It is an extract from milk thistle and thought to inhibit the uptake of amatoxin into hepatocytes. The most common dose is 5 mg/kg IV loading dose followed by 20 mg/kg/day via continuous infusion. There is some human data showing promise of silibinin as an antidote, and it is currently available as part of an open label, multicenter clinical trial. To obtain silibinin, contact 866-520-4412.
    d) PENICILLIN G: Penicillin G appears to displace amatoxin from plasma protein binding sites and possibly inhibit its uptake into hepatocytes. The dose with some evidence of effectiveness is 300,000 to 1,000,000 units/day, though mortality is not significantly affected in preliminary studies.
    e) THIOCTIC ACID: Thioctic acid is a coenzyme in the Krebs cycle used in Eastern Europe for the treatment of amatoxin poisoning with some data showing a possible reduction in mortality. The dose is 50 to 150 mg every 6 hours. Its efficacy remains unproven. It has been reported to cause hypoglycemia and is not readily available in the US.
    2) Several drugs have been proposed for management of amatoxin poisoning: silibinin, N-acetylcysteine, high-dose penicillin, thioctic acid, steroids, cytochrome C, and hyperbaric oxygen (Rengstorff et al, 2003; Burton et al, 2002; Parish & Doering, 1986; Floersheim, 1987).
    3) In studies, silibinin and NAC each administered as monotherapy, and silibinin with benzylpenicillin as bi-, tri-, and polytherapies were associated with the lowest mortalities. Combinations of benzylpenicillin with thioctic acid, steroid, and other drugs, except for silibinin as bi-, tri-, and polytherapies, had the highest mortality/lowest efficacy. It appeared that silibinin monotherapy and silibinin in combination with benzylpenicillin were not significantly different. Benzylpenicillin as monotherapy was not effective. Overall, NAC and silibinin were the most effective agents (Enjalbert et al, 2002).
    4) CASE SERIES: A series of 111 adult patients with amatoxin poisoning were treated with the following regimen (Giannini et al, 2007):
    1) Careful correction of fluid, electrolyte, glucose, and acid base imbalances
    2) correction of altered coagulation factors with fresh frozen plasma as needed and intravenous vitamin K1 20 to 40 mg/day in patients with INR greater than 2.1
    3) multiple dose activated charcoal 20 to 40 g every 4 hours for at least 3 days after ingestion
    4) Fluid therapy (1 liter crystalloid/10 kg/day) and mannitol 18% (0.25 to 0.5 g/kg/hr) to maintain urine output of 200 ml/hr for 2 days after ingestion
    5) intravenous dexamethasone (8 to 16 mg/day)
    6) intravenous glutathione (4.8 g/day in two divided doses)
    7) continuous intravenous administration of penicillin g (1,000,000 international units/kg for the first day, then 500,000 international units/kg for the next 2 days)
    8) metoclopramide for nausea and vomiting (up to 10 mg three times/day
    a) In this series, only two patients died, both of whom presented to the hospital more than 60 hours after ingestion with high transaminase and bilirubin concentrations and low prothrombin activity. Patients who were hospitalized within 36 hours of ingestion had lower peak aminotransferase concentrations and higher prothrombin activity, and were discharged earlier, suggesting that early use of this treatment regimen may impact the severity of illness (Giannini et al, 2007).
    E) ACETYLCYSTEINE
    1) N-acetylcysteine may be beneficial and is associated with little toxicity.
    2) One study reported the use of NAC in 11 patients with various degrees of amanita poisoning. NAC therapy was started early, during the gastrointestinal phase of illness. NAC doses used were suggested by acetaminophen poisoning treatment. Additional treatment also included the use of hemodiaperfusion, high dose penicillin, and supportive care. Ten of the patients recovered and one patient with pre-existing chronic hepatitis B died due to hepatic failure (Montanini et al, 1999).
    a) DOSES: This is the standard FDA-approved dosing regimen for the treatment of acetaminophen poisoning. LOADING DOSE: 150 mg/kg in 200 mL of 5% dextrose, infuse intravenously over 60 minutes. MAINTENANCE DOSE: 50 mg/kg in 500 mL of 5% dextrose, infuse intravenously over 4 hours followed by 100 mg/kg (6.25 mg/kg/hour) in 1000 mL of 5% dextrose, infuse intravenously over 16 hours (Daly et al, 2008; Prod Info ACETADOTE(R) IV injection, 2006; Prescott et al, 1979).
    3) Two patients ingested a small piece (the size of a thumbnail) of Amanita phalloides mushroom and developed elevated liver enzymes and coagulopathy. Both patients recovered following treatment with N-acetylcysteine and IV silibinin (French et al, 2011).
    4) In one case series, 8 patients with hepatic failure secondary to Amanita phalloides poisoning underwent 21 fractionated plasma separation and adsorption system (Promethesus; FPSA) procedures (1 to 4 each; mean durations, 6.5 hours). All patients also received fluid replacement, penicillin G, and N-acetylcysteine. Silymarin was given to one patient. Following the first FPSA treatment, improvement of the biochemical parameters was observed. Six patients recovered completely. One patient with grade 3 encephalopathy underwent liver transplantation. There were no major adverse effects during the procedures. Another patient with grade 4 encephalopathy died (Vardar et al, 2010).
    5) ANIMAL STUDIES: N-acetylcysteine (NAC) was investigated in mice to see if it would prevent the toxicity of alpha amatoxins. Toxicity in the treated group and the non-treated group was almost identical. The authors concluded that single dose NAC has no protective effect in mice (Schneider et al, 1989).
    F) SILIBININ
    1) The hepatoprotective and antagonist effects of silibinin against amatoxins have been confirmed in experimental models. Silibinin is thought to inhibit the penetration of the amatoxins into liver cells (Jahn et al, 1980; Faulstich et al, 1980a; Floersheim, 1987).
    2) The most common dose is 5 mg/kg IV loading dose followed by 20 mg/kg/day via continuous infusion.
    3) Silibinin is NOT available as a licensed drug in the US. Silibinin oral capsules and ampules for intravenous use are available in Europe under the trade names Legalon(R) and Legalon(R)SIL, respectively.
    a) An open-label, multicenter, clinical trial (start date: February 2010; anticipated end date December, 2015), sponsored by Madaus Inc, (a division of Madaus GmbH, Cologne, Germany), is evaluating the safety and efficacy of intravenous silibinin (Legalon(R) SIL) for treating patients with amatoxin mushroom poisoning diagnosed by history, GI symptoms, elevated liver enzymes, and/or diagnostic assay. Patients will be treated with 5 mg/kg loading dose of silibinin followed by 20 mg/kg/day via continuous infusion. Treatment will be discontinued when coagulopathy has resolved, and when liver enzyme concentrations have significantly improved. Patients will be monitored for 7 to 14 days after the end of silibinin therapy with follow up lab studies. For more information on this clinical trial, check the following website: http://clinicaltrials.gov/ct2/show/NCT00915681 (Madaus Inc., 2009).
    b) To obtain silibinin, a 24-hour hotline is available for physicians: 866-520-4412 (Madaus Inc., 2009).
    4) One study reported on a total of 18 cases of poisoning by Amanita phalloides, that were treated by combined chemotherapy. After attempted primary elimination of the amatoxin all patients received silibinin as basic therapy (mainly by infusion, with two cases orally). All patients survived except one which was the result of a massive dose of toxin with suicidal intent (Hruby et al, 1983a).
    5) Two patients ingested a small piece (the size of a thumbnail) of Amanita phalloides mushroom and developed elevated liver enzymes and coagulopathy. Both patients recovered following treatment with N-acetylcysteine and IV silibinin (French et al, 2011).
    6) Silymarin (Legalon(R) 70) capsules 1.4 to 4.2 grams/day for 4 days may also be given but this treatment may be useless if the patient presents with vomiting or is treated with oral charcoal (Silibinin for intravenous use is prepared in ampules by Dr Madaus and Company, Cologne, West Germany).
    7) Silibinin is often combined with other therapy, such as penicillin, ascorbic acid, and hemodialysis (Lheureux et al, 1992) so the exact benefit is difficult to determine in a specific intoxication. One study found no significant difference between silibinin monotherapy and silibinin plus penicillin therapy for treatment of amanita intoxication in humans (Strenge-Hesse et al, 1999).
    8) Complexation of the poorly absorbed silybin has created Silipide which is orally administered and shows higher pharmacologic activity in animal models of hepatic injury (Pifferi, 1991; Conti et al, 1991; Marena & Lampertico, 1991).
    9) CASE REPORT: A 71-year-old man presented with nausea, vomiting, diarrhea, and weakness within 48 hours of ingesting an unknown amount of Amanita mushrooms while camping in northwestern Iowa. Laboratory results revealed elevated liver enzymes, total bilirubin of 1.5 mg/dL (normal range, 0 to 1 mg/dL), INR of 1.3 (normal range, 0.8 to 1.2), and a serum creatinine of 2.7 mg/dL (normal range, 0.51 to 1.2 mg/dL). Despite treatment with IV N-acetylcysteine and penicillin G, he developed encephalopathy, acute renal injury, coagulopathy, and worsening transaminase elevations. He was transported to the liver transplant center and received multidose activated charcoal and IV silibinin (loading dose: 5 mg/kg over 1 hour, continued for 3 days at 20 mg/kg/day) starting 96 hours postingestion. His condition gradually improved by day 3 and he was discharged 5 days after presentation. On a follow-up visit a month later, his liver enzymes were normal (Gores et al, 2014).
    G) PENICILLIN G
    1) Penicillin G appears to displace amatoxin from plasma protein binding sites and possibly inhibit its uptake into hepatocytes.
    2) Experimental studies have shown that penicillin G reduced or inhibited the liver uptake of amatoxins and protected mice and rats against lethal doses of amatoxins (Floersheim, 1987).
    3) In a retrospective study, administration of penicillin was significantly more often associated with survival (Floersheim et al, 1982).
    4) Early treatment with high doses of penicillin G should be considered in doses of 300,000 to 1,000,000 units/kg/day in intravenous infusion (Floersheim, 1987; Floersheim et al, 1978).
    5) The true efficacy of penicillin G is difficult to assess, since it is often given in human cases as a part of a multi-drug therapy (Piering & Bratanow, 1990; Wright et al, 1992).
    H) TRANSPLANTATION OF LIVER
    1) Liver transplantation should be considered in poisonings with severe hepatic failure.
    a) A hepatic specialist should be contacted about the appropriateness of a liver transplant.
    b) PROGNOSTIC INDICATORS
    1) COAGULOPATHY: It appears that severity of coagulopathy is a reliable indicator of recovery or death in patients with Amanita phalloides poisoning (Enjalbert et al, 2002). In one study, a reliable tool for deciding emergency transplantation was a prothrombin index of lower than 10% (an approximate INR of 6), 4 days or more after ingestion (Escudie et al, 2007).
    a) STUDY: One study evaluated ways to identify early prognostic markers in patients with Amanita phalloides poisoning (n=27). Eight patients (30%) either died or received liver transplantation. The earliest significant predictor of a fatal outcome was an interval of less than 8 hours between the ingestion of mushrooms and the onset of diarrhea, but this was only 78% accurate. Overall, non-paracetamol and paracetamol King's College criteria were superior to Clichy's and Ganzert's criteria (accuracy of 100% compared to 85% and 85%, respectively). A reliable tool for deciding emergency transplantation was a prothrombin index of lower than 10% (an approximate INR of 6), 4 days or more after ingestion. No additional prognostic information were provided by encephalopathy and creatinine; therefore, it was proposed that encephalopathy should not be an absolute prerequisite for deciding transplantation (Escudie et al, 2007)
    1) In this study the following criteria were used (Escudie et al, 2007):
    a) Clichy's criteria: Combination of decrease in factor V below 30% normal in patients > 30 years or below 20% normal in patients < 30 years AND grade 3 to 4 encephalopathy (85% accurate in this study).
    b) Ganzert's criteria: decrease in prothrombin index at or below 25% of normal at any time between 3 to 10 days after ingestion in association with a serum creatinine of 106 micromol/L or more during the same time period (85% accurate in this study).
    c) King's college paracetamol criteria: Arterial pH < 7.3 or arterial lactate above 3 mmol/L after adequate resuscitation OR the combination of serum creatinine above 300 micromol/L AND INR above 6.5 AND encephalopathy grade 3 or more concurrently (100% accurate in this study).
    d) King's college non-paracetamol criteria: EITHER prothrombin time over 100 sec (INR over 7), OR AT LEAST 3 of the following: PT over 50 sec (INR over 3.5), serum bilirubin over 300 micromol/L, age less than 10 or greater than 40 years, an interval between jaundice and encephalopathy over 7 days (100% accurate in this study).
    2) Another study reported criteria of severe hepatic failure and poor prognosis were hepatic encephalopathy, serum total bilirubin level > 25 mg/dL, marked jaundice, oliguria or anuria, bleeding, refractory hypoglycemia, and prothrombin time of 100 seconds or longer (Lim et al, 2000).
    3) In a retrospective review of 144 amatoxin poisonings, fatal outcomes were associated with low mean arterial pressure, encephalopathy, mucosal hemorrhage, oliguria-anuria, hypoglycemia, or thrombocytopenia during hospitalization. Low sodium values and elevated urea, AST, ALT, total bilirubin, LDH, prothrombin time, INR, and activated partial thromboplastin time (APTT) values were associated with poor outcome. Fourteen patients with acute hepatic failure died (Trabulus & Altiparmak, 2011).
    4) Jaeger et al (1992) considered the poor prognosis factors to be: peak prothrombin time greater than 100 seconds, factor V less than 10 percent, lactic acidosis, gastrointestinal bleeding, and age less than 12 years (Jaeger et al, 1992). Galler et al (1992) also included hypoglycemia and a serum bilirubin level greater than 25 mg per deciliter (Galler et al, 1992).
    5) Factors not considered useful are the duration of the latency period, the peak of aminotransferases, or analysis for amanitins (Jaeger et al, 1992).
    c) PARTIAL LIVER TRANSPLANT
    1) Partial liver transplant may be an option in patients who do not have multiorgan failure, especially children and young adults. A complete regeneration of native liver after partial liver transplantation has been reported (Escudie et al, 2007).
    a) CASE REPORT: After ingesting Amanita phalloides mushrooms, a 23-year-old woman who did not have multiorgan failure, underwent right hepatectomy and was transplanted with a right lobe. Despite a complicated postoperative period, including invasive aspergillosis, acute respiratory distress syndrome, and small bowel perforation necessitating surgical repair, she had a progressive regeneration of her native liver. Her immunosuppression was gradually tapered (Escudie et al, 2007).
    d) CASE REPORTS
    1) Kinetic studies would indicate that the new liver would NOT be at risk from circulating amatoxins four days after ingestion (Jaeger et al, 1993).
    2) In a group of four patients treated with orthoptic liver transplantation, common clinical findings included grade I or II encephalopathy, gastrointestinal hemorrhage, peak prothrombin time greater than 50 seconds and platelets under 90 thousand. Metabolic acidosis was present in 3 and transient hypoglycemia in 2 patients (Daya et al, 1989).
    3) Through 1989, 9 patients have been treated this way. One was a 3-year-old girl who was comatose and on a respirator; the other eight were adults. All patients have survived (Duffy, 1985).
    4) After ingesting Amanita phalloides mushrooms, two patients with fulminant hepatic failure and encephalopathy (grade 2 and 3, respectively) underwent orthotropic liver transplantation. One patient had no postoperative problem and was discharged with clinical improvement. The second patient's condition improved in early postoperative period; however, his condition worsened again. Trucut liver biopsy revealed massive hemorrhagic necrosis, similar to the explanted liver. He died 10 days after transplantation. Both livers (naive liver and transplanted liver) showed similar macroscopic and microscopic findings. The authors speculated that amanita phalloides toxins could be deposited in tissues other than liver causing recurrence of mushroom poisoning after the transplantation. In addition, other toxic molecules (nonphalloidin and nonamanitins) may have caused a fatal outcome (Kucuk et al, 2005).
    5) Three members of a family (parents and a 15-year-old son) with Amanita phalloides poisoning underwent orthotopic liver transplantation. Although the father and son survived, the mother had 3 episodes of cardiac arrest and did not respond to intensive pharmacological treatment and cardiac massage (Pawlowska et al, 2002).
    6) Organ rejection presents the usual problems in these patients (Kern et al, 1992).
    e) INTRAPORTAL HEPATOCYTE TRANSPLANTATION
    1) After ingesting amanita phalloides, a 64-year-old woman with a history of hypertension and chronic heart failure repeatedly refused to undergo an orthotropic liver transplantation. As hepatic coma progressed to Level III, she was treated with intraportal transplantation of hepatocytes (HcTx; 8 x 10(9) hepatocytes including approx. 5 x 10(9) viable cells infused through the portal vein catheter over a period of 30 hrs). She was also given fresh frozen plasma, prothrombin complex, antithrombin III, IV steroids and cyclosporine A. Ammonia levels immediately before HcTx and 8 hours after the last infusion were 126 micromoles/L (peak) and 45 micromoles/L, respectively. One day after the completion of HcTx, bilirubin levels peaked, but decreased continuously during the next several days. She was extubated 7 days after HcTx, and was transferred from ICU. An abdominal ultrasound scan 2 months after HcTx revealed a normal liver and portal blood flow. Immunosuppressive therapy was discontinued 3 months after HcTx; recovery was complete without concomitant extrahepatic organ damage (Schneider et al, 2006).
    a) The hepatocytes were obtained by the following method: cadaveric livers were used to obtain the blood group matched human hepatocytes. The hepatocytes were cryopreserved and immediately before application, the cell suspensions were thawed rapidly and tested for viability by trypan blue exclusion test. The final transplant had the average viability of 62% (Schneider et al, 2006).
    I) HEPATIC FAILURE
    1) MOLECULAR ABSORBENT REGENERATING SYSTEM (MARS): MARS is a method for removing albumin-bound and water-soluble toxins from blood in patients with liver failure and hepatic encephalopathy (Kantola et al, 2009; Covic et al, 2003; Shi et al, 2002). It employs an albumin-impregnated highly permeable dialyzer with albumin-containing dialysate recycled in a closed loop with a charcoal cartridge, an anion exchange resin absorber, and a conventional hemodialysis membrane. It has been used in a small number of patients with fulminant hepatic failure secondary to cyclopeptide mushroom ingestion. The use of MARS in 7 children and adolescents with fulminant hepatic failure from cyclopeptide mushroom ingestion was associated with rapid improvements in serum transaminase, ammonia and bilirubin levels and improvement in prothrombin time. In addition, the degree of encephalopathy improved in all but one patient, and 5 of the 7 survived (Covic et al, 2003; Shi et al, 2002).
    a) In a retrospective study, the effects of MARS and therapeutic plasma exchange (TPE) therapies were evaluated in 9 patients (age range, 22 to 51 years old) with Amanita phalloides-induced fulminant liver failure. All patients were also treated with standard treatments, including gastric lavage, naso-duodenal tube with continual aspiration, multiple doses of activated charcoal, penicillin G, and vitamin K. Seven patients received 12 MARS treatments (time from intoxication to MARS: 52 to 138 hours). Seven patients received 12 TPE treatments (time from intoxication to TPE: 68 to 122 hours). Five of these patients received both TPE and MARS. Overall, 6 (66.7%) patients survived. Although both TPE and MARS removed toxins and improved liver functions, a single session of TPE produced a greater improvement in liver function as compared with MARS therapy. Patients with severe liver failure and renal failure had worse outcomes (Zhang et al, 2014)
    b) CASE SERIES: Ten patients presented with vomiting, stomach cramps, and diarrhea within 2 days (median 18 hours, range 14 to 36 hours) of ingesting 2 to 4 Amanita phalloides mushrooms. Within 4 days (range, 26 to 78 hours) of mushroom ingestions, all patients were treated with MARS albumin dialysis (a median of 3 MARS sessions (range, 1 to 7 sessions) lasting for 15.4 hours) and standard supportive care (eg, fluid resuscitation, NAC, oral activated charcoal, silibinin in 7 patients and penicillin G in 4 patients). Nine patients recovered completely (days in hospital, 3 to 14 days). One patient with necrotic liver, encephalopathy, severe coagulopathy, and acute renal failure underwent liver transplantation 9 days postingestion. After the surgery, he experienced persistent acute renal failure (requiring hemodialysis), rhabdomyolysis, and heparin-induced thrombocytopenia. He recovered and was discharged home after 128 days of hospitalization (Kantola et al, 2009).
    c) CASE SERIES: One retrospective study compared the outcomes of 6 patients (age range, 16 to 61 years) with Amanita Phalloides poisoning treated with either MARS (n=3; three 6-hour sessions per patient) plus optimal intensive care (OIC) or OIC alone (n=3). All 3 patients in the MARS plus OIC group had statistically significant reductions in ammonia (p value 0.011), ALT (p less than 0.01) and prothrombin time (p value 0.004). However, 2 of these patients had a significant rebound in bilirubin a day after MARS therapy. Five patients (3 from OIC group and 2 from MARS plus OIC group) who ingested 150 to 250 grams of mushrooms died within 9 days of mushroom ingestion. Mortality was 67% in the MARS group compared to 100% in the OIC group. One patient who ingested 50 grams of mushrooms and was treated with MARS plus OIC recovered and was discharged with good liver function (Sorodoc et al, 2010).
    d) CASE SERIES: In a series of 6 patients with fulminant hepatic failure secondary to cyclopeptide mushroom ingestion, use of MARS was associated with improvement in serum transaminase, bilirubin, ammonia and creatinine levels and improved prothrombin time. Two patients had sustained improvement in native liver function, two had orthotopic liver transplantation and survived, and two died (Faybik et al, 2003).
    e) CASE REPORT/PREGNANCY: A 27-year-old woman who was 20 weeks pregnant was admitted for amanita poisoning and developed hepatic encephalopathy. Despite aggressive supportive care and extracorporeal detoxification by hemodialysis plus hemoperfusion (performed daily), the patient's clinical status (hepatic and neuro function) did not improve. Uterine contraction was noted on day 5, with threatened abortion anticipated. The patient then underwent three courses of albumin dialysis using MARS intermittently starting from the 8th day of hospitalization. Liver function was noted to immediately improve, as well as clinical features; uterine contraction ceased. The patient was discharged with normal liver function and fetal evaluation appeared normal. Upon follow-up, a healthy baby was born at 36 weeks and had normal development at 7 months (Wu & Wang, 2004).
    f) CASE REPORT/PEDIATRIC: An 11-year-old boy with amanita phalloides poisoning developed fulminant liver failure and CNS depression, and was placed on a waiting list for emergent liver transplant. During this period, the patient was started on continuous albumin dialysis with MARS (Terakilin AG combined with BM25, Baxter); no neurological changes were observed after the first session. On day two of therapy the parameters for the MARS sessions were modified (ie, increased blood, albumin and dialysate flows), and the patient had a dramatic improvement in neurological function approximately 1 hour after the session was completed; preparation for emergent transplant was temporarily halted. Despite a presumed rebound deterioration in brain function, the patient had a third session and the patient continued to make neurological and clinical improvement on days 4 through 7. On day 24, the patient was discharged to home with normal liver function and intact neurological functioning; 12 month follow-up revealed no clinical or developmental deficits (Rubik et al, 2004).
    g) CASE REPORTS: MARS was reportedly used in another amanita phalloides poisoning with a favorable outcome (Catalina et al, 2003). MARS appears to be a promising bridging technique until the patient's liver can spontaneously recover or until liver transplantation can occur.
    2) FRACTIONATED PLASMA SEPARATION AND ADSORPTION SYSTEM (PROMETHEUS; FPSA; PROM): In one case series, 8 patients with hepatic failure secondary to Amanita phalloides poisoning underwent 21 FPSA procedures (1 to 4 each; mean durations, 6.5 hours). All patients also received fluid replacement, penicillin G, and N-acetylcysteine. Silymarin was given to one patient. Following the first FPSA treatment, improvement of the biochemical parameters was observed. Six patients recovered completely. One patient with grade 3 encephalopathy underwent liver transplantation. There were no major adverse effects during the procedures. Another patient with grade 4 encephalopathy died (Vardar et al, 2010).
    3) EXPERIMENTAL: NASOBILIARY DRAINAGE
    a) CASE REPORT: An 18-year-old man unintentionally ingested 11 amanita bisporigera mushrooms, and developed hepatotoxicity. Laboratory findings approximately 20 hours after exposure included: aspartate aminotransferase (AST) 42 Units/L, total bilirubin 0.9 mg/dL, total protein 5.1 g/dL and ammonia level of 62 micromol/L. Due to concern of developing severe hepatotoxicity, the patient was transferred to a higher level of care. Severe liver dysfunction developed within 72 hours (AST 2459 U/L, ALT 3649, U/L, INR of 5.96, ammonia of 67 micromol/L) and aggressive treatment including possible liver transplantation. The patient underwent an upper gastrointestinal endoscopy with nasobiliary drain placement to interrupt the enterohepatic circulation of amatoxins. The total amount of bile was 240 mL. Total amatoxin (including alpha-amatoxin {1.81 mg} and beta-amatoxin {2.22 mg} as determined by high performance liquid chromatography) excreted from the bile was 4.03 mg over 3 days. Charcoal hemoperfusion was also performed to enhance elimination. The patient recovered completely with normal liver enzymes and was discharged to home on day 8 (Madhok et al, 2006).
    J) CARDIOGENIC SHOCK
    1) INTRA-AORTIC BALLOON COUNTERPULSATION: In patients with mushroom poisoning and cardiogenic shock, intra-aortic balloon counterpulsation may restore tissue perfusion (Aygul et al, 2010).
    a) CASE REPORT: A 24-year-old woman presented with abdominal pain, nausea, vomiting, and weakness 6 hours after ingesting Amanita phalloides mushrooms (unknown quantity). Despite supportive therapy, including 4 sessions of hemodialysis, her condition did not improve. Her condition deteriorated very quickly and she developed multiorgan failure, including liver, renal, and cardiac failure. At this time, she was orthopneic, cyanotic, somnolent, tachycardiac (130 beats/min), and hypotensive (BP 70/50 mmHg). An ECG revealed sinus tachycardia with non-specific ST-T wave changes in anterior leads. Laboratory results revealed a prolonged prothrombin time (INR 5.19) and elevated liver enzymes and serum creatinine. Borderline cardiomegaly with bilateral pleural effusions at the costophrenic angles was observed in a chest x-ray. An echocardiogram showed a global left ventricular hypokinesia with left ventricular ejection fraction (EF) of 24%, end-diastolic diameter of 6.2 cm, and systolic pulmonary artery pressure of 50 mmHg. At this time, an intra-aortic balloon counterpulsation catheter was inserted and a marked improvement was noted within 1 hour. In addition, she was treated with 4 units of fresh-frozen plasma and a peritoneal dialysis catheter was inserted. Her condition continued to improve and both intra-aortic balloon counterpulsation and peritoneal dialysis were removed on day 5. She was discharged on day 12 (Aygul et al, 2010).
    K) EXPERIMENTAL THERAPY
    1) THIOCTIC ACID: The use of thioctic acid as a specific treatment has NOT been subjected to well controlled studies to assess its value. The clinical efficacy of this agent has NOT been proven.
    a) Experimentally, thioctic acid was totally ineffective as an antidote against amatoxins in mice or dogs (Floersheim, 1987). There seems to be little reason to continue the use of thioctic acid in Amanita intoxication.
    b) For those who still wish to consider it, administer thioctic acid (alpha-lipoic acid) 50 to 150 mg every 6 hours in intravenous glucose (Roldan & Lloret, 1986) Becker et al, 1976, Finestone et al, 1972). It must be protected from light (wrap intravenous bottle and tubing in aluminum foil).
    c) Thioctic acid is NOT commercially available for use in humans. The technical material may be available through chemical supply companies.
    2) CIMETIDINE: Based on clinical similarity to other liver toxic agents, cimetidine, a cytochrome P450 inhibitor, has been tested as a possible antidote to amanitin poisoning, since amanitins are believed to be converted to toxic metabolites via hepatic cytochrome P450 system (Lim et al, 2000).
    3) An animal experiment described by Schneider et al (1987) is presented:
    a) Three groups of 10 mice each were given 0.5 mg/kg of amanitin and either 1) saline before and after, 2) cimetidine both before and after, or 3) saline before and cimetidine 6 hours after.
    b) Group 1 mice were injected intraperitoneally with saline 1 milliliter, then 60 minutes later with 2.5 mg/kg of phallodin.
    c) Group 2 mice were injected intraperitoneally with 120 mg/kg of cimetidine, then 60 minutes later with 2.5 mg/kg phallodin. The cimetidine pretreatment group had a statistically worse survival rate than the control group (Schneider et al, 1991).
    d) Two mice in group 2 died due to reaction to the 120 mg/kg intraperitoneal dose. The surviving 18 mice appeared to have decreased fatty changes compared to the unprotected group.
    e) No comparison was made to other forms of therapy. The actual mechanism that produced this reduction is unknown.
    f) If the 120 mg/kg dose used in the experiment were applied to a 70 kg human, this would be 8,400 mg, well above the 800 to 1200 mg therapeutic dose.
    g) Much more work needs to be done before cimetidine can be recommended as a standard therapy.
    4) BASTIEN TECHNIQUE: This treatment method has NOT been clinically evaluated and is NOT considered scientifically sound or efficacious by most authorities.
    a) In 1957, Dr Bastien proposed a treatment where a patient is given intravenous vitamin C (ascorbic acid) 3 grams/day, oral nifuroxazide 1,200 mg/day, and dihydrostreptomycin 1500 mg/day.
    b) The three drugs are given for 3 days during which carrot broth is the only source of nutrition. He had treated himself and other patients using this technique (Laing, 1984).
    c) Some poison centers in France use the technique combined with fluid, electrolytes, and penicillin. It can NOT recommend this technique at this time.
    5) COMBINATION THERAPIES: One study reported the use of cimetidine, penicillin, and ascorbic acid in combination. Groups of mice were given 0.6 mg/kg of alpha amanitin intraperitoneally (Schneider et al, 1987).
    a) Four hours later they were given 1 milliliter of solutions containing the following combination therapies.
    b) All were tested versus controls who received no treatment. Seven day survival is indicated below. Caution should be used when interpreting these results.
    c) Similar to previous studies, the dose of cimetidine was approximately 28 times the usual single therapeutic human dose.
    THERAPY7-DAY SURVIVAL
    Cimetidine 120 mg/kg IP36.8%
    Cimetidine 120 mg/kg IP plus47.5%
    Penicillin 250 mg/kg IP47.5%
    Cimetidine 120 mg/kg plus42.1%
    Ascorbic acid 600 mg/kg IP42.1%
    Cimetidine plus penicillin plus ascorbic acid (as above)73.7%
    Saline31.6%

    6) PICRORHIZA KURROA/KUTKIN: The roots of the Indian plant Picrorhiza kurroa contain an iridoid glycoside mixture that has been shown to be hepatoprotective against injury due to carbon tetrachloride and galactosamine (Ansari et al, 1988; (Visen et al, 1991; Visen et al, 1993).
    a) Kutkin is a mixture of the iridoid glycosides picroside 1 and kutkoside.
    b) When mice were given lethal doses of lyophilized Amanita phalloides, the protective effect of kutkin was comparable to that seen with silibinin (Floersheim et al, 1990).
    7) AUCUBIN: Aucubin is an iridoid glycoside obtained from the leaves of Aucuba japonica. It has low oral bioavailability (Chang & Yamara, 1993).
    a) Aucubin has shown to be protective against Amanita intoxication when tested in dogs and mice (Chang et al, 1984) Chang & Yun, 1985; (Chang & Yamaura, 1993).
    b) No human testing has been done.
    8) HYPERBARIC OXYGEN THERAPY (HBO): Was attempted in mice, in order to protect from hepatotoxicity, immediately following alpha-amanitin poisoning for 2 hr every 12 hr until sacrifice. Hepatotoxicity was concentration dependent, and no protection from HBO therapy was demonstrated at either high or low doses of amanitin (Thomas et al, 1997).

Enhanced Elimination

    A) DIURESIS
    1) Toxicokinetic studies indicate that significant amounts of amatoxins are eliminated in urine, especially during the 48 hours following ingestion (Vesconi et al, 1985; Jaeger et al, 1988).
    2) Thus, maintenance of a normal or slightly high urine output should be a goal or therapy especially during the first 48 hours.
    B) EXTRACORPOREAL ELIMINATION
    1) Toxicokinetic studies showed that amatoxins were present in serum only during the first 24 to 48 hours and at very low concentrations (in comparison with concentrations found in urine) (Vesconi et al, 1980; Vesconi et al, 1985; Jaeger et al, 1988). Because amatoxins are cleared rapidly from the plasma by the kidneys, extracorporeal elimination techniques may not clear significant amounts of toxin (O'Brien & Khuu, 1996).
    a) Thus, the different techniques of extracorporeal elimination given AFTER 25 to 48 hours (plasmapheresis, peritoneal dialysis, hemodialysis, and hemoperfusion) are generally NOT thought of as useful or indicated.
    b) One study described the use of hemoperfusion and hemodialysis in 2 patients with confirmed Amanita phalloides poisoning. Hemodialysis was started 23 hours after ingestion and hemoperfusion was begun later. A series of blood samples was taken to determine clearance of toxin by each method. No amatoxin was detected in serum 24 hours after the ingestions nor after treatment, and none was detected in HD/HP circuits. These methods did not appear to contribute to removal of amatoxins (Mullins & Horowitz, 2000).
    c) Another reported a series of 11 patients with severe Amanita poisoning. Intensive therapy was started on all patients 34 hours (range 20 to 54 hours) after ingestions. Extracorporeal elimination consisted of combined hemodialysis and hemoperfusion, in two 8 hour sessions for 10 of the patients and one session for one patient, with no clinical complications. Hospitalization period of 13 +/- 2 days was required by these patients (p < 0.01), and all were discharged asymptomatic (Sabeel et al, 1995; Sabeel et al, 1995a).
    d) Hemodialysis or hemoperfusion in order to remove amatoxins would be indicated if a patient with previous renal failure develops Amanita intoxication.
    1) Nine of 10 patients given hemodialysis died in one case series. This treatment was not thought to be responsible for the poor outcome (Paydas et al, 1990).
    2) HEMOPERFUSION: (Hemodetoxifier B-D) may be helpful to remove amatoxins in early stages or to support the patient during hepatic failure. Hemoperfusion would be most effective if initiated prior to 24 hours after ingestion. A major risk of hemoperfusion is thrombocytopenia with increased risk of bleeding (Gynp et al, 1999; (Feinfeld et al, 1994).
    a) Three patients with severe Amanita poisoning were started on charcoal hemoperfusions 36, 37, and 42 hours, respectively, after the ingestions. Two of the patients received hemodialysis in addition to hemoperfusion. All three patients recovered (Aji et al, 1995).
    b) Charcoal plasmaperfusion (CPP) for 3 hours every day for 3 and 5 days, respectively, and continuous venovenous hemofiltration (CVVH) for 72 and 100 hours, respectively, were used in 2 patients following severe amanita mushroom poisoning. The purpose of the 2 therapies was to eliminate via CVVH the small and medium molecules and by CPP the poisons linked to serum proteins. Both patients developed full recovery (Splendiani et al, 2000). Clearance of amitoxins by CPP and CVVH were not determined.
    3) HEMODIALYSIS: Should be instituted if renal failure occurs.
    4) HEMOFILTRATION: In one study, the efficacy of conventional and hemofiltration therapies in patients (n=58; mean age 38.03 +/- 15.96 years) with mushroom poisoning was evaluated, using demographic characteristics, symptoms in ED, and latent phase periods of patients. The latent phase was defined as the duration of time from ingesting the mushroom to the onset of symptoms. Two groups of patients were included, with group 1 (n=36) having a latent phase of 0 to 5 hours after ingesting mushrooms with early acting toxins and group 2 (n=22) with a latent phase of 6 to 24 hours after ingesting mushrooms with late acting toxins. Patients in group 1 had significantly lower AST, ALT, INR, and BUN values than group 2. Treatments included gastric lavage and activated charcoal (36 patients; 62%), conventional treatment (penicillin G and silibinin; 30 patients; 55.2%), hemofiltration (22 patients; 37.9%), and fluid with symptomatic treatment (all patients). A statistically significant difference was observed when treatments of patients were compared according to their latent phases. Patients who received hemofiltration with conventional treatment had significantly higher AST, ALT, and INR values than patients who only received supportive treatment. It was concluded that hemofiltration, in combination with conventional therapy, was effective in decreasing mortality of patients after exposure to mushrooms with late acting toxins (eg, A. phalloides, Gyromitra esculenta) (Colak et al, 2015).
    C) SURGICAL THERAPY
    1) CANNULATION OF THE COMMON BILE DUCT: Common bile duct cannulation may be of benefit in severe poisonings, to interrupt enterohepatic recirculation of the toxin (Duffy & Vergeer, 1986; Frank & Cummins, 1987).

Case Reports

    A) PEDIATRIC
    1) OTHER
    a) MUSHROOM POISON CASE REGISTRY
    1) MUSHROOM POISON CASE REGISTRY
    a) Mushroom poisoning cases may be reported to the North American Mycological Association's Mushroom Poisoning Case Registry. Reporting is voluntary and patient confidentiality is maintained.
    b) Forms may be obtained from the website and completed forms or questions may be sent to:
    c) Dr. Michael W. Beug, PO Box 116, Husum, WA 98623; phone: (509) 493-2237
    d) Alternatively, reports may be submitted online at www.sph.umich.edu/~kwcee/mpcr. The website also contains a list of volunteers from different regions of North America willing to assist in the identification of mushrooms.
    2) SPECIFIC AGENT
    a) AMANITA PHALLOIDES
    1) Woodle et al (1985) reported a 3-year-old girl who ingested Amanita phalloides. She developed abdominal pain, vomiting, diarrhea, and then hepatitis. Laboratory data showed a marked hepatitis (AST 16.648 Units/L; ALT 9.844 Units/L), and coagulation disorders (prothrombin time: 34.2 sec). Serum bilirubin was 23 mg/L and ammonia was 122 mcg/dL. Despite supportive treatment, the patient deteriorated rapidly and developed an encephalopathy (grade III). An orthotopic liver transplantation was performed on the fifth day. After transplantation, hepatic parameters improved rapidly. However, some neurologic abnormalities remained (Woodle et al, 1985).
    2) ORGAN DONATION: Langer et al (1997) reported a case of a 9-year-old boy who died on the 7th day from liver failure and intracranial hypertension following amanita phalloides ingestion. There was no evidence of renal, pulmonary or cardiac dysfunction. Consequently, his kidneys, heart, and corneas were transplanted (Langer et al, 1997).
    a) Both patients receiving kidney transplants, showed no signs of liver dysfunction or necrosis in the early phases. One of the two patients rejected the kidney, 4 days later, due to vascular thrombosis and the other patient survived the transplant procedure with good renal function. The patient, who received the heart, died due to complications of the transplant procedure. The patient did not show any signs of hepatic dysfunction.

Summary

    A) TOXICITY: Amatoxins are highly potent toxins and are amongst the most lethal poisons known. The LD50 of the dose is 0.1 mg/kg which is equivalent to an ingestion of 6 to 7 mg of mushroom or a cap by an average person. The weight of an average Amanita phalloides cap is 60 g and the concentration of alpha-amanitin is usually 0.5 to 1.5 mg/g of mushroom. One Amanita cap is potentially lethal. Fifteen to 20 Galerina caps and 30 Lepiotas can kill a healthy adult. Mortality is 20% to 30% untreated (generally higher in children), but can be reduced to 5% with appropriate, aggressive, and intensive supportive care.

Minimum Lethal Exposure

    A) FACTS CONCERNING THE TOXIN/MUSHROOM
    1) Amatoxin is one of the most lethal poisons known. The LD50 of the dose is 0.1 mg/kg which is equivalent to an ingestion of 6 to 7 mg of mushroom or a cap by an average person (Vesconi et al, 1985; Benjamin, 1995). The weight of an average Amanita phalloides cap is 60 g and the concentration of alpha-amanitin is usually 0.5 to 1.5 mg/g of mushroom (Benjamin, 1995).
    2) The lethal dose of fresh Galerina mushrooms is 100 to 150 g (approximately 10 to 20 mushrooms caps) (Benjamin, 1995).
    3) It has been suggested that about 30 Lepiotas might be fatal (Haines et al, 1985).
    4) One author found a concentration (dry weight) of 2560 mcg/g of Amanita phalloides and 3150 mcg/g in Lepiota brunneoincarnata (Piqueras, 1989).
    B) CASE REPORT
    1) A 58-year-old woman presented with an 8-hour history of diarrhea and vomiting a day after ingesting 6 boiled Amanita phalloides mushrooms. Despite aggressive treatment with IV silibinin 5 mg/kg 4 times daily, high-dose benzylpenicillin (1,000,000 International Units/kg/day), n-acetylcysteine, and IV fluids, she developed decompensated coagulopathic liver failure and later became encephalopathic. Five days postingestion, she died of fulminant liver failure (Lawton & Bhraonain, 2013).
    C) MORTALITY RATE
    1) A literature review suggests that the percentage of mortality has gradually decreased during the last decades, probably due to intensive care hospitalization with early rehydration of the patients (Costantino et al, 1978; Vesconi et al, 1985).
    2) Mortality is 50% if untreated and less than 5% with intensive supportive care. These mushrooms account for over 95% of the cases of fatal mushroom poisoning in the United States (Lampe, 1978).
    3) In a collaborative study of 205 cases of intoxications recorded throughout Europe from 1971 to 1980, the overall mortality was 22.4%. A significant difference between adults and children was observed. Mortality was 51.3% in children before 10 years of age, and 16.5% in the patients older than 10 years (Floersheim et al, 1982).
    4) Prognosis seems to be determined by the quantity of mushroom eaten (dose of toxins/kg of body weight).

Maximum Tolerated Exposure

    A) No data are available about a maximum tolerated dose in humans.
    B) Aji et al (1995) reported 3 brothers, in phases 2 and 3 of cyclopeptide mushroom poisoning, who ate an estimated 80 g of Amanita phalloides. All 3 survived following supportive therapy and hemoperfusion and hemodialysis treatments (Aji et al, 1995).
    C) LEPIOTA SUBINCARNATA: A 43-year-old woman, with a medical history significant for hepatitis B carrier status, developed fulminant hepatic failure with pancreatitis, coagulopathy, profound metabolic acidosis, and renal insufficiency, 36 hours after ingesting approximately 170 g of sauteed Lepiota subincarnata mushrooms that she picked in the front yard of her home in Chicago. Following supportive care and a liver transplant, she was discharged home on day 12 (Mottram et al, 2010).

Serum Plasma Blood Concentrations

    7.5.2) TOXIC CONCENTRATIONS
    A) TOXIC CONCENTRATION LEVELS
    1) CONCENTRATION LEVEL
    a) SERUM AMATOXIN LEVELS were detectable in 65 percent of patients in one study (Vesconi et al, 1985) even as late as 30 hours postingestion. Concentrations were 0.5 to 24 nanograms/milliliter. Only 4 of 19 had levels greater than 3 nanograms/milliliter.
    b) In another study, serum amatoxin levels were detected in only 10 of 30 patients (Jaeger et al, 1988). Concentrations were 8 to 190 nanograms/milliliter for alpha amanitin (6 patients) and 16 to 162 nanograms/milliliter for beta amanitin (5 patients).
    1) In one case, only amatoxins were detected in serum 48 hours after the ingestion.
    c) Urinary amatoxin level of 37.3 mcg/L (measured approximately 4 days post-ingestion) confirmed a suspected diagnosis of Amanitas phalloides poisoning following ingestion of several mushrooms that had been previously frozen for 7 to 8 months (Himmelmann et al, 2001).
    2) ANIMAL DATA
    a) In animals fatally poisoned by oral amatoxins, amatoxin levels are always below 30 nanograms/milliliter even in the first hour of poisoning (Vesconi et al, 1985).

Toxicologic Mechanism

    A) AMATOXINS
    1) The amatoxins, particularly the amanitins, have been shown to interfere with both DNA and RNA transcription by interfering with RNA polymerase II (Parra et al, 1992). Cells with the highest rate of multiplication, such as the intestinal mucosa, are injured first (Piqueras, 1989).
    2) The liver and kidneys are next. Amanitins cause necrosis of hepatocytes and of the cells in the proximal tubules of the kidney (pp 1233-1234). However a direct toxicity of amanitin on kidney has NOT been confirmed in human poisoning (Constantino et al, 1978).
    3) Experimental studies in mice showed that amanitin conjugated with albumin was ten times more toxic than amanitin (Bonneti et al, 1976). However, this mechanism does NOT play a role in human toxicity because amanitin is NOT bound to serum albumin.
    B) PHALLOTOXINS
    1) It is most probable that the phallotoxins play no role in human poisoning (Wieland T, 1990). Phallotoxins are NOT absorbed from the gastrointestinal tract in the experimental animals investigated and probably NOT in the human.
    2) When given parenterally to laboratory animals, phallotoxins destroy the endoplasmic reticulum and mitochondria of the liver cells and induce a necrosis of the hepatocytes.
    3) Phalloidin binds to the actin F (filamentous polymer) of the plasma membranes, and hence increases the permeability of the plasma membranes of hepatocytes (Wieland T, 1990).

Physical Characteristics

    A) Amatoxins and phallotoxins isolated from methanolic extracts of the mushrooms are colorless, mainly crystalline compounds, soluble in water, methanol, and other polar organic solvents.
    B) Amatoxins: Amatoxins are bicyclic octapeptides. Nine amatoxins have been isolated (Piqueras, 1989): alpha, beta, gamma, and epsilon amanitins, amanullin, amanullinic acid, proamanullin and amanin. Amanitins are the most toxic compounds.
    1) The bicyclic structure and gamma hydroxyl group at the dihydroxy isoleucine portion are necessary for the toxicity.
    C) PHALLOTOXINS: Phallotoxins are bicyclic heptapeptides. Seven compounds have been isolated: phalloidin, phalloin, prophalloin, phallisin, phallacin, phallacidin, phallisacin.
    1) The bicyclic structure and a allo-positioned OH group at the pyrrolidine ring are necessary for the toxicity (Wieland & Faulstich, 1978).
    D) At least five virotoxins (monocyclic heptapeptides) have been isolated from Amanita virosa (Faulstich et al, 1980a).

Molecular Weight

    A) AMATOXINS contain 3 or 4 mol of water of crystallization. Molecular weights are 990, 373, and 974 g/mol for alpha, beta, and gamma amanitin, respectively.
    B) PHALLOTOXINS crystallize with 5 mol of water of crystallization. Molecular weights are 879, 863, and 895 mol wt for phalloidin, phalloin, and phallisin, respectively (Weigland & Faulstich, 1978).

Clinical Effects

    11.1.3) CANINE/DOG
    A) The symptoms of amanitin intoxication have been analyzed in beagle dogs. Early symptoms are hyperglycemia followed by hypoglycemia which caused death in most dogs after 1 to 2 days.
    1) If compensated with glucose, the dogs developed an acute liver dystrophy, causing death after 2 to 3 days.
    B) Severe hemorrhages in various organs were the main cause of death in some cases. A late symptom was kidney failure from which a few dogs died after the seventh day of the intoxication (Wieland & Faulstich, 1978).
    C) When phalloidin was tested in dogs, they showed pathological changes in the SGPT, SGOT, alkaline phosphatase, and total bilirubin.
    1) Histologically the liver parenchyma revealed hemorrhagic necrosis and peliosis-like changes with penetration of RBCs into hepatocytes.
    D) Ataxia, extreme listlessness, and petit mal seizures were seen in a golden retriever puppy. The animal died 6 hours after symptom onset and after treatment with lactated ringers and diazepam. The dog had eaten A. verna mushrooms (Liggett & Weiss, 1989).

Treatment

    11.2.1) SUMMARY
    A) GENERAL TREATMENT
    1) SUMMARY
    a) Begin treatment immediately.
    b) Keep animal warm and do not handle unnecessarily.
    c) Remove the patient and other animals from the source of contamination or remove dietary sources.
    2) Treatment should always be done on the advice and with the consultation of a veterinarian.
    3) Additional information regarding treatment of poisoned animals may be obtained from a Veterinary Toxicologist or the National Animal Poison Control Center.
    4) ASPCA ANIMAL POISON CONTROL CENTER
    a) ASPCA Animal Poison Control Center, 1717 S Philo Road, Suite 36 Urbana, IL 61802
    b) It is an emergency telephone service which provides toxicology information to veterinarians, animal owners, universities, extension personnel and poison center staff for a fee. A veterinary toxicologist is available for consultation.
    c) Contact information: (888) 426-4435 (hotline) or www.aspca.org (A fee may apply. Please inquire with the poison center). The agency will make follow-up calls as needed in critical cases at no extra charge.
    11.2.2) LIFE SUPPORT
    A) GENERAL
    1) MAINTAIN VITAL FUNCTIONS: Secure airway, supply oxygen, and begin supportive fluid therapy if necessary.
    11.2.4) DECONTAMINATION
    A) GASTRIC DECONTAMINATION
    1) GENERAL TREATMENT
    a) EMESIS/GASTRIC LAVAGE -
    1) CAUTION: Carefully examine patients with chemical exposure before inducing emesis. If signs of oral, pharyngeal, or esophageal irritation, a depressed gag reflex, or central nervous system excitation or depression are present, EMESIS SHOULD NOT BE INDUCED.
    2) HORSES OR CATTLE: DO NOT attempt to induce emesis in ruminants (cattle) or equids (horses).
    3) DOGS AND CATS
    a) IPECAC: If within 2 hours of exposure: induce emesis with 1 to 2 milliliters/kilogram syrup of ipecac per os.
    b) APOMORPHINE: Dogs may vomit more readily with 1 tablet (6 milligrams) apomorphine diluted in 3 to 5 milliliters water and instilled into the conjunctival sac or per os.
    1) Dogs may also be given apomorphine intravenously at 40 micrograms/kilogram, although this route may not be as effective.
    4) LAVAGE: In the absence of a gag reflex or if vomiting cannot be induced, place a cuffed endotracheal tube and begin gastric lavage.
    a) Pass large bore stomach tube and instill 5 to 10 milliliters/kilogram water or lavage solution, then aspirate. Repeat 10 times.
    b) ACTIVATED CHARCOAL/CATHARTIC -
    1) ACTIVATED CHARCOAL: Administer activated charcoal. Dose: 2 grams/kilogram per os or via stomach tube. Avoid aspiration by proper restraint, careful technique, and if necessary tracheal intubation.
    2) CATHARTIC: Administer a dose of a saline or sorbitol cathartic such as magnesium or sodium sulfate (sodium sulfate dose is 1 gram/kilogram). If access to these agents is limited, give 5 to 15 milliliters magnesium oxide (Milk of Magnesia) per os for dilution.
    3) ACTIVATED CHARCOAL/HORSES: Administer 0.5 to 1 kilogram of activated charcoal in up to 1 gallon warm water via nasogastric tube. Neonates: administer 250 grams (one-half pound) activated charcoal in up to 2 quarts water.
    4) ACTIVATED CHARCOAL/RUMINANTS: Administer 2 to 9 grams/ kilogram of activated charcoal in a slurry of 1 gram charcoal/3 to 5 milliliters warm water via stomach tube. Sheep may be given 0.5 kilogram charcoal in slurry.
    5) CATHARTICS/HORSES: Mineral oil is administered 30 minutes after activated charcoal. DOSE: 4 to 6 liters in adult horses and 1 to 4 liters in neonates or foals.
    a) Magnesium sulfate: 0.2 to 0.9 grams/kilogram (500 grams for adults).
    b) The sulfate laxatives are especially effective when given 30 to 45 minutes after mineral oil administration.
    c) Carbachol (lentin): administer 1 milligram to an adult.
    6) CATHARTICS/RUMINANTS & SWINE: Adult cattle: administer 500 grams sodium or magnesium sulfate. Other ruminants and swine: administer 1 to 2 grams/kilogram.
    a) The sulfate laxatives are especially effective when given 30 to 45 minutes after cathartic administration.
    b) Mineral oil: Do not administer within 30 minutes of activated charcoal. DOSE: small ruminants and swine, 60 to 200 milliliters; cattle, 0.5 to 1 gallon.
    c) Magnesium oxide: (Milk of Magnesia) Small ruminants, up to 0.25 gram/kilogram in 1 to 3 gallons warm water; adult cattle up to 1 gram/kilogram in 1 to 3 gallons warm water or 2 to 4 boluses MgOH per os.
    d) Give these solutions via stomach tube and monitor for aspiration.
    11.2.5) TREATMENT
    A) GENERAL TREATMENT
    1) MAINTAIN VITAL FUNCTIONS - as necessary.
    B) SEIZURES -
    1) SEIZURES/LARGE ANIMALS: May be controlled with diazepam.
    a) HORSES/DIAZEPAM: Neonates: 0.05 to 0.4 milligrams/kilogram; Adults: 25 to 50 milligrams. Give slowly intravenously to effect; repeat in 30 minutes if necessary.
    b) CATTLE, SHEEP AND SWINE/DIAZEPAM: 0.5 to 1.5 milligrams/kilogram intravenously to effect.
    2) SEIZURES/DOGS & CATS:
    a) DIAZEPAM: 0.5 to 2 milligrams/kilogram intravenous bolus; may repeat dose every ten minutes for four total doses. Give slowly over 1 to 2 minutes to effect.
    b) PHENOBARBITAL: 5 to 30 milligrams/kilogram over 5 to 10 minutes intravenously to effect.
    c) REFRACTORY SEIZURES: Consider anaesthesia or heavy sedation. Administer pentobarbital to DOGS & CATS at a dose of 3 to 15 milligrams/kilogram intravenously slowly to effect. May need to repeat in 4 to 8 hours. Be sure to protect the airway.
    C) FLUID/ELECTROLYTE BALANCE -
    1) Monitor blood glucose concentration every hour during the acute intoxication. Correct hyperglycemia or hypoglycemia as necessary.
    2) Begin electrolyte and fluid therapy with isotonic solutions as needed at maintenance doses (66 milliliters solution/kilogram body weight/day intravenously) or, in hypotensive patients, at high doses (up to shock dose 60 milliliters/kilogram/hour). Monitor for urine production and pulmonary edema.
    D) LABORATORY -
    1) This agent may cause hepatotoxicity. Monitoring liver function tests is suggested for patients with significant exposure.
    2) This agent may cause nephrotoxicity. Monitoring renal function tests and urinalysis is suggested for patients with significant exposure.
    E) MONITORING -
    1) Admit all symptomatic patients and begin treatment.
    2) Symptomatic patients must be monitored continuously. Refer to an emergency hospital or critical care clinic for 24 hour monitoring.
    F) FOLLOW-UP -
    1) Instruct the owner to return for a follow up appointment at which physical examination and appropriate laboratory tests will be repeated.

Range Of Toxicity

    11.3.2) MINIMAL TOXIC DOSE
    A) SPECIFIC TOXIN
    1) Toxicologic data about animal poisonings are only available from experimental studies. Susceptibility to the poison differs among the various animal species (Wieland & Faulstich, 1978).
    B) AMATOXINS -
    1) MOUSE - In the white mouse, the LD50 after IP administration is 0.3 milligram per kilogram and death occurs in 2 to 5 days.
    2) RAT - The rat is more resistant: LD50 is about 4 milligrams per kilogram after IP administration.
    3) DOG - Dogs are more sensitive with a LD50 of only 0.1 milligram per kilogram if administered intravenously (Wieland & Faulstich, 1978).
    C) PHALLOTOXINS -
    1) Phallotoxins are less toxic than amatoxins.
    2) MOUSE - In the white mouse, the LD50 is 2.5 milligrams per kilogram after IP administration.
    3) RAT - Rats are more susceptible to phallotoxins than mice (Wieland & Faulstich, 1978).

Continuing Care

    11.4.1) SUMMARY
    11.4.1.2) DECONTAMINATION/TREATMENT
    A) GENERAL TREATMENT
    1) SUMMARY
    a) Begin treatment immediately.
    b) Keep animal warm and do not handle unnecessarily.
    c) Remove the patient and other animals from the source of contamination or remove dietary sources.
    2) Treatment should always be done on the advice and with the consultation of a veterinarian.
    3) Additional information regarding treatment of poisoned animals may be obtained from a Veterinary Toxicologist or the National Animal Poison Control Center.
    4) ASPCA ANIMAL POISON CONTROL CENTER
    a) ASPCA Animal Poison Control Center, 1717 S Philo Road, Suite 36 Urbana, IL 61802
    b) It is an emergency telephone service which provides toxicology information to veterinarians, animal owners, universities, extension personnel and poison center staff for a fee. A veterinary toxicologist is available for consultation.
    c) Contact information: (888) 426-4435 (hotline) or www.aspca.org (A fee may apply. Please inquire with the poison center). The agency will make follow-up calls as needed in critical cases at no extra charge.
    11.4.2) DECONTAMINATION
    11.4.2.2) GASTRIC DECONTAMINATION
    A) GASTRIC DECONTAMINATION
    1) GENERAL TREATMENT
    a) EMESIS/GASTRIC LAVAGE -
    1) CAUTION: Carefully examine patients with chemical exposure before inducing emesis. If signs of oral, pharyngeal, or esophageal irritation, a depressed gag reflex, or central nervous system excitation or depression are present, EMESIS SHOULD NOT BE INDUCED.
    2) HORSES OR CATTLE: DO NOT attempt to induce emesis in ruminants (cattle) or equids (horses).
    3) DOGS AND CATS
    a) IPECAC: If within 2 hours of exposure: induce emesis with 1 to 2 milliliters/kilogram syrup of ipecac per os.
    b) APOMORPHINE: Dogs may vomit more readily with 1 tablet (6 milligrams) apomorphine diluted in 3 to 5 milliliters water and instilled into the conjunctival sac or per os.
    1) Dogs may also be given apomorphine intravenously at 40 micrograms/kilogram, although this route may not be as effective.
    4) LAVAGE: In the absence of a gag reflex or if vomiting cannot be induced, place a cuffed endotracheal tube and begin gastric lavage.
    a) Pass large bore stomach tube and instill 5 to 10 milliliters/kilogram water or lavage solution, then aspirate. Repeat 10 times.
    b) ACTIVATED CHARCOAL/CATHARTIC -
    1) ACTIVATED CHARCOAL: Administer activated charcoal. Dose: 2 grams/kilogram per os or via stomach tube. Avoid aspiration by proper restraint, careful technique, and if necessary tracheal intubation.
    2) CATHARTIC: Administer a dose of a saline or sorbitol cathartic such as magnesium or sodium sulfate (sodium sulfate dose is 1 gram/kilogram). If access to these agents is limited, give 5 to 15 milliliters magnesium oxide (Milk of Magnesia) per os for dilution.
    3) ACTIVATED CHARCOAL/HORSES: Administer 0.5 to 1 kilogram of activated charcoal in up to 1 gallon warm water via nasogastric tube. Neonates: administer 250 grams (one-half pound) activated charcoal in up to 2 quarts water.
    4) ACTIVATED CHARCOAL/RUMINANTS: Administer 2 to 9 grams/ kilogram of activated charcoal in a slurry of 1 gram charcoal/3 to 5 milliliters warm water via stomach tube. Sheep may be given 0.5 kilogram charcoal in slurry.
    5) CATHARTICS/HORSES: Mineral oil is administered 30 minutes after activated charcoal. DOSE: 4 to 6 liters in adult horses and 1 to 4 liters in neonates or foals.
    a) Magnesium sulfate: 0.2 to 0.9 grams/kilogram (500 grams for adults).
    b) The sulfate laxatives are especially effective when given 30 to 45 minutes after mineral oil administration.
    c) Carbachol (lentin): administer 1 milligram to an adult.
    6) CATHARTICS/RUMINANTS & SWINE: Adult cattle: administer 500 grams sodium or magnesium sulfate. Other ruminants and swine: administer 1 to 2 grams/kilogram.
    a) The sulfate laxatives are especially effective when given 30 to 45 minutes after cathartic administration.
    b) Mineral oil: Do not administer within 30 minutes of activated charcoal. DOSE: small ruminants and swine, 60 to 200 milliliters; cattle, 0.5 to 1 gallon.
    c) Magnesium oxide: (Milk of Magnesia) Small ruminants, up to 0.25 gram/kilogram in 1 to 3 gallons warm water; adult cattle up to 1 gram/kilogram in 1 to 3 gallons warm water or 2 to 4 boluses MgOH per os.
    d) Give these solutions via stomach tube and monitor for aspiration.

Kinetics

    11.5.1) ABSORPTION
    A) SPECIFIC TOXIN
    1) AMATOXINS -
    a) Kinetics of amatoxins have been studied in dogs after IV administration, and in mice after IP administration (Faulstich et al, 1985; pp 1233-1234).
    b) Animal species differ in their ability to absorb amatoxins from the gastrointestinal tract.
    c) In mice and rats, the poison is absorbed extremely slowly or NOT at all.
    d) In guinea pigs, cats, and dogs, doses of a few milligrams per kilogram cause death.
    2) PHALLOTOXINS -
    a) Phallotoxins are NOT absorbed from the gastrointestinal tract.
    11.5.2) DISTRIBUTION
    A) GENERAL
    1) PROTEIN BINDING - Amatoxins are NOT bound to albumin (Faulstich et al, 1985; Fiume et al, 1977).
    2) VOLUME OF DISTRIBUTION - In the dog at equilibrium, the volume of distribution is identical with the extracellular space; VD is about 200 mL/kg (Faulstich et al, 1985).
    11.5.3) METABOLISM
    A) GENERAL
    1) No metabolites of amatoxins could be detected after administration of radioactive amanitin (Jahn et al, 1980; Faulstich et al, 1985).
    11.5.4) ELIMINATION
    A) DOG
    1) After administration of labeled amatoxins in dogs, 83% to 89% is eliminated in urine and less than 10% is excreted in the bile.
    2) In dogs after IV administration, the serum half-life is 27 to 50 minutes and no amatoxin is detected in serum after 5 hours.
    B) RODENT
    1) In mice, the elimination of amatoxins in serum is rapid and no amatoxin is detected 4 hours after intraperitoneal administration.

General Bibliography

    1) Aji DY, Caliskan S, & Nayir A: Haemoperfusion in Amanita phalloides poisoning. J Trop Ped 1995; 41:371-374.
    2) Alaspaa AO, Kuisma MJ, Hoppu K, et al: Out-of-hospital administration of activated charcoal by emergency medical services. Ann Emerg Med 2005; 45:207-12.
    3) Altintepe L, Yazici R, Yazici M, et al: Temporary left ventricular dysfunction in mushroom poisoning: report of three cases. Ren Fail 2014; 36(8):1337-1339.
    4) Andres RY & Frei W: Amatoxin and anti-amatoxin for radioimmunoassay prepared by a novel chemical approach. Toxicon 1987.
    5) Atkinson SW, Young Y, & Trotter GA: Treatment with activated charcoal complicated by gastrointestinal obstruction requiring surgery. Br Med J 1992; 305:563.
    6) Aygul N , Duzenli MA , Ozdemir K , et al: A case report of an unusual complication of Amanita phalloides poisoning: Development of cardiogenic shock and its successful treatment with intra-aortic balloon counterpulsation. Toxicon 2010; 55(2-3):630-632.
    7) Bartoloni ST, Omer FB, & Giannini A: Amanita poisoning: A clinical-histopathological study of 64 cases of intoxication. Hepato-gastroenterol 1985; 32:229-231.
    8) Belliardo F, Massano G, & Accomo S: Amatoxins do not cross the placental barrier. Lancet 1983; 1:1381.
    9) Benjamin DR: Mushrooms: Poisons and Panaceas, WH Freeman & Co, New York, NY, 1995.
    10) Beuhler M, Lee DC, & Gerkin R: The meixner test in the detection of alpha-amanitin and false-positive reactions caused by psilocin and 5-substituted tryptamines. Ann Emerg Med 2004; 44:114-120.
    11) Bivins HG, Bivins R, & Lammer R: Mushroom ingestion. Ann Emerg Med 1985; 14:101-106.
    12) Bonneti E, Derenzini M, & Fiume L: Increased penetration of amanitine into hepatocytes when conjugated with albumin. Arch Toxicol 1976; 35:69-73.
    13) Boyer JC, Hernandez F, Estorc J, et al: Management of maternal Amanita phalloides poisoning during the first trimester of pregnancy: a case report and review of the literature. Clin Chem 2001; 47(5):971-974.
    14) Burton JR Jr, Ryan C, & Shaw-Stiffel TA: Liver transplantation in mushroom poisoning. J Clin Gastroenterol 2002; 35:276-280.
    15) Busi C, Fiume L, & Costantino D: Amanita toxins in gastroduodenal fluid of patients poisoned by the mushroom, Amanita phalloides. N Engl J Med 1979; 300:800.
    16) CDC: CDC: MMRW 1997; 46(22):489-492.
    17) Caccialanza G, Gandini C, & Ponci R: Direct simultaneous determination of alpha-amanitin, beta amanitin and phalloidine by high-performance liquid chromatography. J Pharmacol Biomed Anal 1985; 3:179-185.
    18) Cakir B, Kirbas I, Cevik B, et al: Mushroom poisoning in children: liver MDCT findings in three cases. Pediatr Radiol 2007; 37(9):908-911.
    19) Cappell MS & Hassan T: Gastrointestinal and hepatic effects of Amanita- phalloides ingestion. J Clin Gastroenterol 1992; 15:225-228.
    20) Caravati EM, Knight HH, & Linscott MS: Esophageal laceration and charcoal mediastinum complicating gastric lavage. J Emerg Med 2001; 20:273-276.
    21) Catalina MV, Nunez O, Ponferrada A, et al: Toxicidad hepatica por ingest de setas: curso clinico y nuevas perspectivas de tratamiento. Gastoenterol Hepatol 2003; 26(7):417-420.
    22) Chang IM & Yamaura Y: Aucubin: a new antidote for poisonous Amanita mushrooms. Phytother Res 1993; 7:53-56.
    23) Chang IM, Yun HS, & Kim YS: Aucubin, potential antidote for alpha-amanitin poisoning. Clin Tox 1984; 22:77-85.
    24) Chyka PA & Seger D: Position statement: single-dose activated charcoal. American Academy of Clinical Toxicology; European Association of Poisons Centres and Clinical Toxicologists. J Toxicol Clin Toxicol 1997; 35:721-741.
    25) Chyka PA, Seger D, Krenzelok EP, et al: Position paper: Single-dose activated charcoal. Clin Toxicol (Phila) 2005; 43(2):61-87.
    26) Chyka PA: Multiple-dose activated charcoal and enhancement of systemic drug clearance: summary of studies in animals and human volunteers. J Toxicol Clin Toxicol 1995; 33(5):399-405.
    27) Colak S, Kandis H, Afacan MA, et al: Assessment of patients who presented to the emergency department with mushroom poisoning. Hum Exp Toxicol 2015; 34(7):725-731.
    28) Conti M, Porzio R, & Malandrino S: Activity of silipide against ethanol-induced liver damage in rats. Planta Medica 1991; 57(S2):A115-A116.
    29) Costantino D, Falzi G, & Langer M: Amanita phalloides related nephropathy. Cont Nephrol 1978; 10:84-97.
    30) Covic A, Goldsmith DJA, Gusbeth-Tatomir P, et al: Successful use of Molecular Absorbent Regenerating System (MARS) dialysis for the treatment of fulminant hepatic failure in children accidentally poisoned by toxic mushroom ingestion. Liver Intl 2003; 23:21-27.
    31) Dagnone D, Matsui D, & Rieder MJ: Assessment of the palatability of vehicles for activated charcoal in pediatric volunteers. Pediatr Emerg Care 2002; 18:19-21.
    32) Daly FF, Fountain JS, Murray L, et al: Guidelines for the management of paracetamol poisoning in Australia and New Zealand--explanation and elaboration. A consensus statement from clinical toxicologists consulting to the Australasian poisons information centres. Med J Aust 2008; 188(5):296-301.
    33) Daya MR, Norton RL, & Field RD: Liver transplantation in Amanita phalloides poisoning. Vet Hum Toxicol 1989; 31:360.
    34) DeCarlo E, Milanesi A, Martini C, et al: Effects of amanita phalloides toxins on insulin release: in vivo and in vitro studies. Arch Toxicol 2003; 77:441-445.
    35) Donnelly M, Brouxhon S, & Schneider S: A rare cause of Amatoxin poisoning resulting in liver transplantation and death (abstract). Clin Toxicol 2000; 38:516-517.
    36) Duffy TJ & Vergeer P: Treatment of mushroom amanitin poisoning. West J Med 1986; 145:521.
    37) Duffy TJ: Liver transplantation for victims of Amanita mushroom poisoning. McIlvaniea 1985; 9:4-5.
    38) Elliot CG, Colby TV, & Kelly TM: Charcoal lung. Bronchiolitis obliterans after aspiration of activated charcoal. Chest 1989; 96:672-674.
    39) Enjalbert F, Rapior S, Nouguier-Soule J, et al: Treatment of amatoxin poisoning: 20-year retrospective analysis. J Toxicol Clin Toxicol 2002; 40(6):715-757.
    40) Escudie L, Francoz C, Vinel J, et al: Amanita phalloides poisoning: Reassessment of prognostic factors and indications for emergency liver transplantation. J Hepatol 2007; 46(3):466-473.
    41) Eyer F, Felgenhauer N, & Zilker T: Die Entwicklung eines toxischen megakolons bei knollenblatterpilzvergiftungen (Toxic megacolon, an unusual complication of amanita phalloides poisoning) . Dtsch Med Wochenschr 2004; 129:137-140.
    42) FDA: Poison treatment drug product for over-the-counter human use; tentative final monograph. FDA: Fed Register 1985; 50:2244-2262.
    43) Faulstich H, Jahn W, & Wieland T: Silybin inhibition of amatoxin uptake in the perfused rat liver. Arzneimittelforschung 1980a; 30:452-454.
    44) Faulstich H, Talas A, & Wellhoner HH: Toxicokinetics of labeled amatoxins in the dogs. Arch Toxicol 1985; 56:190-194.
    45) Faulstich H, Trischmann H, & Zobeley S: A radioimmunoassay for amanitin. FEBS Lett 1975; 56:312-315.
    46) Faybik P, Hetz H, Baker A, et al: Extracorporeal albumin dialysis in patients with Amanita phalloides poisoning. Liver Intl 2003; 23:28-33.
    47) Feinfeld DA, Mofenson HC, & Caraccio T: Poisoning by amatoxin-containing mushrooms in suburban - report of four cases. Clin Tox 1994; 32(6):715-721.
    48) Fiume I, Busi C & Campadelli-Fiume G et al: Production of antibodies to amanitins as the basis for their radioimmunoassay. Experimentia, 1975.
    49) Fiume L, Sperti S, & Montanaro L: Amanitins do not bind to serum albumin. Lancet 1977; 1:1111.
    50) Fiume L: Detection of molecular mechanism of action of amatoxins, in Faulstich H, Kommerell B & Wieland T (Eds): Amanita Toxins and Poisoning, Witzstrock Publishers, New York, 1980, pp 59-65.
    51) Floersheim GL, Bieri A, & Koenig R: Protection against Amanita phalloides by the iridoid glycoside mixture of Picrorhiza kurroa (kutkin). Agents Actions 1990; 29:386-387.
    52) Floersheim GL, Eberhard M, & Tschumi P: Effects of penicillin and silymarin on liver enzymes and blood clotting factors in dogs given a boiled preparation of Amanita phalloides. Toxicol Appl Pharmacol 1978; 46:455-462.
    53) Floersheim GL, Weber O, & Tschumi P: Die klinische knollenblatterpilzvergiftung (Amanita phalloides): prognostische faktoren und therapeutische massnahmen. Schweiz Med Wschr 1982; 112:1164-1177.
    54) Floersheim GL: Treatment of human amatoxin mushroom poisoning myths and advances in therapy. Med Toxicol 1987; 2:1-9.
    55) Frank IC & Cummins L: Case review. Amanita poisoning treated with endoscopic biliary diversion. J Emerg Nurs 1987; 13:132-134.
    56) French LK, Hendrickson RG, & Horowitz BZ: Amanita phalloides poisoning. Clin Toxicol (Phila) 2011; 49(2):128-129.
    57) Galler GW, Weisenberg E, & Brasitus TA: Mushroom poisoning: the role of orthotopic liver transplantation. J Clin Gastro 1992; 15(3):229-232.
    58) Garcia de la Fuente I, McLin VA, Rimensberger PC, et al: Amanita poisoning and liver transplantation: do we have the right decision criteria?. J Pediatr Gastroenterol Nutr 2011; 53(4):459-462.
    59) Garrouste C, Hemery M, Boudat AM, et al: Amanita phalloides poisoning-induced end-stage renal failure. Clin Nephrol 2009; 71(5):571-574.
    60) Gazzero RC & Goos RD: A case of mushroom poisoning involving Amanita virosa. McIlvainea 1991; 10:45-46.
    61) Genser AS & Marcus SM: Amanita mushroom poisoning, an "outbreak" of 10 cases (abstract). Vet Hum Toxicol 1987; 29:461.
    62) Ghannoum M & Gosselin S: Enhanced poison elimination in critical care. Adv Chronic Kidney Dis 2013; 20(1):94-101.
    63) Giannini L, Vannacci A, Missanelli A, et al: Amatoxin poisoning: a 15-year retrospective analysis and follow-up evaluation of 105 patients. Clin Toxicol (Phila) 2007; 45(5):539-542.
    64) Golej J, Boigner H, Burda G, et al: Severe respiratory failure following charcoal application in a toddler. Resuscitation 2001; 49:315-318.
    65) Gomez HF, Brent JA, & Munoz DC: Charcoal stercolith with intestinal perforation in a patient treated for amitriptyline ingestion. J Emerg Med 1994; 12:57-60.
    66) Gores KM, Hamieh TS, & Schmidt GA: Survival following investigational treatment of amanita mushroom poisoning: thistle or shamrock?. Chest 2014; 146(4):e126-e129.
    67) Graff GR, Stark J, & Berkenbosch JW: Chronic lung disease after activated charcoal aspiration. Pediatrics 2002; 109:959-961.
    68) Guenther Skokan E, Junkins EP, & Corneli HM: Taste test: children rate flavoring agents used with activated charcoal. Arch Pediatr Adolesc Med 2001; 155:683-686.
    69) Haines JH, Lichstein E, & Glickerman D: A fatal poisoning from an amatoxin containing Lepiota. Mycopathologia 1985; 93:15-17.
    70) Hanrahan JP & Gordon MA: Mushroom poisoning. JAMA 1984; 251:1057-1061.
    71) Harris CR & Filandrinos D: Accidental administration of activated charcoal into the lung: aspiration by proxy. Ann Emerg Med 1993; 22:1470-1473.
    72) Harsch HH: Aspiration of activated charcoal. N Engl J Med 1986; 314:318.
    73) Himmelmann A, Mang G, & Schnorf-Huber S: Lethal ingestion of stored Amanita phalloides mushrooms. Swiss Med Wkly 2001; 131:616-617.
    74) Homann J, Rawer P, & Bleyl H: Early detection of amatoxins in human mushroom poisoning. Arch Toxicol 1986; 59:190-191.
    75) Hruby K, Donner A, Csomos G, et al: Silibinin in the treatment of deathcap fungus poisoning. Hum Toxicol 1983a; 2:183.
    76) Hubenova A & Stankova E: Some recent problems in Amanita phalloides poisonings (abstract), EAPCCT, XV Congress, Istanbul, Turkey, 1992, pp 127.
    77) Hydzik P, Bielanski W, Ponka M, et al: Usefulness of 13C-methacetin breath test in liver function testing in Amanita phalloides poisoning; breast feeding woman case. Clinical toxicology 2008; 46(10):1077-1082.
    78) Ilkhanipour K, Yealy DM, & Krenzelok EP: The comparative efficacy of various multiple-dose activated charcoal regimens. Am J Emerg Med 1992; 10:298-300.
    79) Jaeger A, Jehl F, & Flesch F: Amatoxins kinetics in Amanita phalloides poisoning. Vet Hum Toxicol 1989; 31:360.
    80) Jaeger A, Jehl F, & Flesch F: Kinetics of amatoxins in human poisoning: therapeutic implications. Clin Toxicol 1993; 31:63-80.
    81) Jaeger A, Jehl F, & Flesch F: Toxicokinetics of alpha and beta amanitins in Amanita phalloides poisoning, Communication: XII Congress of EAPCC August 27-30, 1986, Bruxelles, Belgium, 1988.
    82) Jaeger A, Kopferschmitt J, & Flesch F: Liver transplantation for Amanita poisoning (abstract), EAPCCT, XV Congress, Istanbul, Turkey, 1992, pp 103.
    83) Jahn W, Faulstich H, & Wieland T: Pharmacokinetics of (3H)-methyl-dihydroxymethyl-amanitin in the isolated perfused rat liver, and the influence of several drugs, in Faulstich H, Kommerell B & Wieland T (Eds): Amanita Toxins and Poisoning, Witzstrock, Baden-Baden, Germany, 1980, pp 80-85.
    84) Jehl F, Gallion C, & Birckel P: Determination of alpha-amanitin and beta-amanitin in human biological fluids by high-performance liquid chromatography. Anal Biochem 1985; 149:35-42.
    85) Kaneko H, Tomomasa T, Inoue Y, et al: Amatoxin poisoning from ingestion of japanese Galerina mushrooms. Clin Toxicol 2001; 39:413-416.
    86) Kantola T, Kantola T, Koivusalo AM, et al: Early molecular adsorbents recirculating system treatment of Amanita mushroom poisoning. Ther Apher Dial 2009; 13(5):399-403.
    87) Kaufmann M, Muller A, & Paweletz N: Fetale schadigung bei einer knollenblatterpilzvergiftung der mutter in der Fruhschwangerschaft Geburtsh. Frauenheilk 1978; 38:122-124.
    88) Kelner MJ & Alexander NM: Endocrine hormone abnormalities in Amanita poisoning. Clin Toxicol 1987; 25:21-37.
    89) Kern C, Zilker T, & Charmann MV: Successful liver transplantation in a child after Amanita poisoning (abstract), EAPCCT, XV Congress, Istanbul, Turkey, 1992, pp 126.
    90) Kervegant M, de Haro L, Patat AM, et al: Phalloides syndrome poisoning after ingestion of lepiota mushrooms. Wilderness Environ Med 2013; 24(2):170-172.
    91) Krenova M, Pelclova D, & Navratil T: Survey of Amanita phalloides poisoning: clinical findings and follow-up evaluation. Hum Exp Toxicol 2007; 26(12):955-961.
    92) Kucuk HF, Karasu Z, Kilic M, et al: Liver failure in transplanted liver due to Amantia falloides. Transplantation Proceedings 2005; 37:2224-2226.
    93) Laing MD: A cure for mushroom poisoning. Sa Mediese Tydskrif 1984; 65:590.
    94) Lampe KF & McCann MA: Differential diagnosis of poisoning by North American mushrooms, with particular emphasis on Amanita phalloides-like intoxication. Ann Emerg Med 1987; 16:956-962.
    95) Lampe KF: Chapter 7 in Rumack BH and Salzman E: Mushroom Poisoning: Diagnosis and Treatment, CRC Press, West Palm Beach, FL, 1978.
    96) Langer M, Gridelli B, & Piccolo G: A liver transplant candidate (fulminant hepatic failure from amanita phalloides poisoning) as a multiorgan donor. Transplantation Proceedings 1997; 29:3343-3344.
    97) Lawton LD & Bhraonain SN: Accidental poisoning by death cap mushrooms: be careful what you eat. Wilderness Environ Med 2013; 24(2):168-170.
    98) Lheureux P, Bourgeois F, & Askenasi R: Conjugal Amanita poisoning (abstract), EAPCCT, XV Congress, Istanbul, Turkey, 1992, pp 108.
    99) Liggett AD & Weiss R: Liver necrosis caused by mushroom poisoning in dogs. J Vet Diagn Invest 1989; 1:267-269.
    100) Lim JG, Kim JH, & Lee CY: Amanita virosa induced toxic hepatitis: report of three cases. Yonsei Med J 2000; 41:416-421.
    101) Lincoff G & Mitchell DH: Toxic and hallucinogenic mushroom poisoning, Van Nostrand Reinhold Company, New York, NY, 1977.
    102) Madaus Inc.: Intravenous Milk Thistle (Silibinin-Legalon) for Hepatic Failure Induced by Amatoxin/Amanita Mushroom Poisoning. ClinicalTrials.gov: a service of the US National Institutes of Health. Bethesda, MD. 2009. Available from URL: http://clinicaltrials.gov/ct2/show/NCT00915681. As accessed 2010-01-13.
    103) Madhok M, Scalzo AJ, Blume CM, et al: Amanita bisporigera ingestion: mistaken identity, dose-related toxicity, and improvement despite severe hepatotoxicity. Pediatr Emerg Care 2006; 22(3):177-180.
    104) Marena C & Lampertico M: Preliminary clinical development of silipide: a new complex of silybin in toxic liver disorders. Planta Medica 1991; 57(S2):A124-A125.
    105) Maurer HH, Schmitt CJ, & Weber AA: Validated electrospray liquid chromatographic-mass spectrometric assay for the determination of the mushroom toxins a- and b-amanitin in urine after immunoaffinity extraction. J Chromatogr 2000; 748:125-135.
    106) Mauro LS, Nawarskas JJ, & Mauro VF: Misadventures with activated charcoal and recommendations for safe use. Ann Pharmacother 1994; 28(7-8):915-924.
    107) Meixner A: Amatoxin-Nachweis in Pilzen. Z Mycol 1979; 45:137-139.
    108) Meunier BC, Camus CM, & Houssin DP: Liver transplantation after severe poisoning due to amatoxin-containing Lepiota - report of three cases. Clin Tox 1995; 33(2):165-171.
    109) Mina B, Dym JP, & Kuepper F: Fatal inhalational anthrax with unknown source of exposure in a 61-year-old woman in New York City. J Am Med Assoc 2002; 287:858-862.
    110) Mizutani T, Naito H, & Oohashi N: Rectal ulcer with massive hemorrhage due to activated charcoal treatment in oral organophosphate poisoning. Hum Exp Toxicol 1991; 10:385-386.
    111) Montanini S, Sinardi D, & Pratico C: Use of acetylcysteine as the life-saving antidote in Amanita phalloides (death cap) poisoning. Arzneim-Forsch/Drug Res 1999; 49:1044-1047.
    112) Mottram AR, Lazio MP, & Bryant SM: Lepiota subincarnata J.E. Lange induced fulminant hepatic failure presenting with pancreatitis. J Med Toxicol 2010; 6(2):155-157.
    113) Mullins ME & Horowitz BZ: The futility of hemoperfusion and hemodialysis in Amanita phalloides poisoning. Vet Human Toxicol 2000; 42:90-91.
    114) Nagy I, Pogatsa-Murray G, & Zalanyi S Jr: Amanita poisoning during the second trimester of pregnancy. A case report and a review of the literature. Clin Investig 1994; 72:794-798.
    115) Nicholls DW, Hyne BEB, & Buchanan P: Death cap mushroom poisoning. NZ Med J 1995; 108:234.
    116) Nicholson FB & Korman MG: Death from Amanita poisoning. Aust NZ J Med 1997; 27:448-449.
    117) None Listed: Position paper: cathartics. J Toxicol Clin Toxicol 2004; 42(3):243-253.
    118) O'Brien BL & Khuu L: A fatal Sunday brunch: Amanita mushroom poisoning in a gulf coast family. Amer J Gastroenterol 1996; 91:581-583.
    119) Olesen LL: Amatoxin intoxication. Scand J Urol Nephrol 1990; 24:231-234.
    120) Omidynia E, Rashidpourai R, & Qaderi MT: Case report - Mycetismus in Hamadan, of West Iran. Southeast Asian J Trop Med Pub 1997; 28:438-439.
    121) Parish RC & Doering PL: Treatment of Amanita mushroom poisoning a review. Vet Hum Toxicol 1986; 28:318-322.
    122) Park GD, Radomski L, & Goldberg MJ: Effects of size and frequency of oral doses of charcoal on theophylline clearance. Clin Pharmacol Ther 1983; 34:663-666.
    123) Parra S, Garcia J, & Martinez P: Profile of alkaline phosphatase isoenzymes in ten patients poisoned by mushrooms of the genus lepiota. Dig Dis Sci 1992; 37:1495-1498.
    124) Pastorello L & Tolentino D: Determination of alpha amanitin by high-performance liquid chromatography. J Chromatogr 1982; 233:398-403.
    125) Pawlowska J, Pawlak J, Kaminski A, et al: Liver transplantation in three family members after Amanita phalloides mushroom poisoning. Transplant Proc 2002; 34(8):3313-3314.
    126) Paydas S, Kocak R, & Erturk E: Poisoning due to amatoxin-containing Lepiota species. Br J Clin Pharmacol 1990; 44:450-453.
    127) Petkovska L, Naumovski DZ, Pereska Z, et al: Transaminase levels in amanita phalloides poisoning (abstract). J Toxicol - Clin Toxicol 2003; 41:512.
    128) Piering WF & Bratanow N: Role of the clinical laboratory in guiding treatment of Amanita virosa mushroom poisoning: report of two cases. Clin Chem 1990; 36:571-574.
    129) Pifferi G: Silipide: a new bioavailable complex of psilybin. Planta Med 1991; 57(suppl 2):A12.
    130) Piqueras J: Hepatotoxic mushroom poisoning: diagnosis and management. Mycopathologia 1989; 105:99-110.
    131) Pollack MM, Dunbar BS, & Holbrook PR: Aspiration of activated charcoal and gastric contents. Ann Emerg Med 1981; 10:528-529.
    132) Prescott LF, Illingworth RN, & Critchley JA: Intravenous N-acetylcysteine: the treatment of choice for paracetamol poisoning. Br Med J 1979; 2:1097.
    133) Product Information: ACETADOTE(R) IV injection, acetylcysteine IV injection. Cumberland Pharmacuticals,Inc, Nashville, TN, 2006.
    134) Ramirez P, Parrilla P, & Bueno FS: Fulminant hepatic failure after Lepiota mushroom poisoning. J Hepatol 1993; 19:51-54.
    135) Rau NR, Nagaraj MV, Prakash PS, et al: Fatal pulmonary aspiration of oral activated charcoal. Br Med J 1988; 297:918-919.
    136) Ray MJ, Padin DR, & Condie JD: Charcoal bezoar. Small-bowel obstruction secondary to amitriptyline overdose therapy. Dig Dis Sci 1988; 33:106-107.
    137) Rengstorff DS, Osorio RW, & Bonacini M: Recovery from severe hepatitis caused by mushroom poisoning without liver transplantation. Am Gastro Hepatol 2003; 1:392-396.
    138) Rivett MJ & Boon GPG: Mushroom (Amanita phalloides) poisoning in Ciskei. S Afr Med J 1988; 73:317.
    139) Roldan EJA & Lloret AP: Thioctic acid in Amanita poisoning (letter). Crit Care Med 1986; 14:753-754.
    140) Rubik J, Pietraszek-Jezierska E, Kaminski A, et al: Successful treatment of a child with fulminant liver failure and coma caused by amanita phalloides intoxication with albumin dialysis without liver transplantation. Pediatr Transplan 2004; 8:295-300.
    141) Sabeel AI, Kurkus J, & Lindholm T: Intensified dialysis treatment of ethylene glycol intoxication. Scand J Urol Nephrol 1995a; 29:125-129.
    142) Sabeel AI, Kurkus J, & Lindholm T: Intensive hemodialysis and hemoperfusion treatment of Amanita mushroom poisoning. Mycopathologia 1995; 131:107-114.
    143) Sanz P, Reig R, & Borras L: Disseminated intravascular coagulation and mesenteric venous thrombosis in fatal Amanita poisoning. Human Toxicol 1988; 7:199-201.
    144) Scalzo A, Blume C, & Weber J: Amanita bisporigera - dose related toxicity from intentional abuse (abstract). J Tox-Clin Tox 1998; 36:456.
    145) Schneider A, Attaran M, Meier PN, et al: Hepatocyte transplantation in an acute liver failure due to mushroom poisoning. Transplantation 2006; 82(8):1115-1116.
    146) Schneider SM, Borochovitz D, & Krenzelok EP: Cimetidine protection against alpha-amanitin hepatotoxicity in mice: a potential model for the treatment of Amanita phalloides poisoning. Ann Emerg Med 1987; 16:1136-1140.
    147) Schneider SM, Vanscoy G, & Michelson EA: Failure of cimetidine to affect phallodin toxicity. Vet Hum Toxicol 1991; 33:17-18.
    148) Schneider SM, Vanscoy GJ, & Michelson EA: Combination therapy with cimetidine, penicillin, and ascorbic acid for alpha amanitin toxicity in mice. Ann Emerg Med 1989; 18:482.
    149) Seeger R & Stijve T: Amanitin content and toxicity of Amanita verna Bull. Z Naturforsch 1979; 34:330-333.
    150) Serne EH, Toorians AW, & Gietema JA: Amanita phalloides, a potentially lethal mushroom: its clinical presentation and therapeutic options. Neth J Med 1996; 49:19-23.
    151) Shi Y, He J, Chen S, et al: MARS: optimistic therapy method in fulminant hepatic failure secondary to cytotoxic mushroom poisoning - a case report. Liver 2002; 22:78-80.
    152) Sorodoc L, Lionte C, Sorodoc V, et al: Is MARS system enough for A. phalloides-induced liver failure treatment?. Hum Exp Toxicol 2010; 29(10):823-832.
    153) Spiller HA & Rogers GC: Evaluation of administration of activated charcoal in the home. Pediatrics 2002; 108:E100.
    154) Splendiani G, Zazzaro D, & Di Pietrantonio P: Continuous renal replacement therapy and charcoal plasmaperfusion in treatment of amanita mushroom poisoning. Artificial Organs 2000; 24:305-308.
    155) Spoerke DG & Rumack BH: Handbook of Mushroom Poisoning - Diagnosis and Treatment, CRC Press, Boca Raton, FL, 1994.
    156) Stein CM, Wu PE, Scott JA, et al: Fulminant hepatic failure following ingestion of wild mushrooms. CMAJ 2015; 187(11):822-824.
    157) Strenge-Hesse A, Michael H, & Zilker TR: Amanita intoxication - treatment with silibinin plus penicillin vs. silibinin (abstract), EAPCCT XVI Internatl Congress, Vienna, Italy, 1999.
    158) Tanghe LJ: Spread of Amanita phalloides in North America. McIlvainea 1983; 6:4-8.
    159) Thakore S & Murphy N: The potential role of prehospital administration of activated charcoal. Emerg Med J 2002; 19:63-65.
    160) Thomas J, Tomaszewski C, & Gordon B: Effect of hyperbaric oxygen on alpha amanitin hepatotoxicity in mice (abstract). J Tox Clin Tox 1997; 35:557.
    161) Timar L & Czeizel AE: Birth weight and congenital anomalies following poisonous mushroom intoxication during pregnancy. Reprod Toxicol 1997; 11:861-866.
    162) Trabulus S & Altiparmak MR: Clinical features and outcome of patients with amatoxin-containing mushroom poisoning. Clin Toxicol (Phila) 2011; 49(4):303-310.
    163) Vale JA, Krenzelok EP, & Barceloux GD: Position statement and practice guidelines on the use of multi-dose activated charcoal in the treatment of acute poisoning. J Toxicol Clin Toxicol 1999; 37:731-751.
    164) Vale JA, Kulig K, American Academy of Clinical Toxicology, et al: Position paper: Gastric lavage. J Toxicol Clin Toxicol 2004; 42:933-943.
    165) Vale JA: Position Statement: gastric lavage. American Academy of Clinical Toxicology; European Association of Poisons Centres and Clinical Toxicologists. J Toxicol Clin Toxicol 1997; 35:711-719.
    166) Vanooteghem S, Arts J, Decock S, et al: Four patients with Amanita Phalloides poisoning. Acta Gastroenterol Belg 2014; 77(3):353-356.
    167) Vardar R, Gunsar F, Ersoz G, et al: Efficacy of fractionated plasma separation and adsorption system (Prometheus) for treatment of liver failure due to mushroom poisoning. Hepatogastroenterology 2010; 57(99-100):573-577.
    168) Velvaert J & Schlatter-Lanz I: Vorgemen bei vergiftungen mit amanita phalloides. Schweiz Arzte-Ztg 1982; 1483.
    169) Vesconi S, Langer M, & Costantine D: Mushroom poisoning and forced diuresis. Lancet 1980; 2:854-855.
    170) Vesconi S, Langer M, & Iapichino G: Therapy of cytotoxic mushroom intoxication. Crit Care Med 1985; 13:402-406.
    171) Visen PKS, Shukla B, & Patnaik GK: Hepatoprotectine acitivity of picroliv isolated from Picrorhiza kurroa against thioacetamide toxicity on rat hepatocytes. Phytochemistry 1991; 5:224-227.
    172) Visen PKS, Shukla B, & Patnaik GK: Prevention of galactosamine-induced hepatic damage by picroliv: study on bile flow and isolated hepatocytes (ex vivo). Planta Med 1993; 59:37-41.
    173) Wacker A, Riethmuller J, Zilker T, et al: Fetal risk through maternal Amanita phalloides poisoning at the end of pregnancy. Am J Perinatol 2009; 26(3):211-213.
    174) Wieland T & Faulstich H: Amatoxins, phallotoxins, phallolysin and antamanide: Bio. Active components of poisonous amanita mushrooms. Crc Crit Rev Biochem 1978; 5:185-260.
    175) Wieland T: Personal Communication. Max Planck Institut fur Med, 1990.
    176) Woodle ES, Moody RR, & Cox KL: Orthotopic liver transplantation in a patient with amanita poisoning. JAMA 1985; 253:69-70.
    177) Wright N, Malloch D, & Jalali S: Amanita poisoning: 2 case reports and discussion of management (abstract), EAPCCT, XV Congress, Istanbul, Turkey, 1992, pp 109.
    178) Wu BF & Wang MM: Molecular adsorbent recirculating system in dealing with maternal amanita poisoning during the second pregnancy trimester: a case report. Hepatobil Pancreat Dis Int 2004; 3(1):152-154.
    179) Yamada EG, Mohle-Boetani J, & Olson KR: Mushroom poisoning due to Amatoxin. West J Med 1998; 169:380-384.
    180) Yarze JC & Tulloss RE: Acute liver injury and renal failure due to poisonous mushroom (Amanita bisporigera) ingestion. Am J Gastroenterol 2012; 107(5):790-791.
    181) Zawadzki J, Jankowska I, & Moszczynska A: Hypouricemia due to increased tubular secretion of urate in children with Amanita phalloides poisoning. Nephron 1993; 65:375-380.
    182) Zevin S, Dempsey D, & Olson K: Amanita phalloides mushroom poisoning - Northern California, January 1997 (from MMWR, June 6, 1997). Clin Toxicol 1997; 35:461-463.
    183) Zhang J, Zhang Y, Peng Z, et al: Experience of Treatments of Amanita phalloides-Induced Fulminant Liver Failure with Molecular Adsorbent Recirculating System and Therapeutic Plasma Exchange. ASAIO J 2014; 60(4):407-412.
    184) Zilker T, Pfab R, & Kleber JJ: Liver transplantation (LTX) for Galerina marginata poisoning (abstract). J Toxicol-Clin Toxicol 1999; 37:417-418.