key: cord-007798-9ht7cqhu authors: Smith, Silas W. title: Drugs and pharmaceuticals: management of intoxication and antidotes date: 2010-02-25 journal: Molecular, Clinical and Environmental Toxicology DOI: 10.1007/978-3-7643-8338-1_12 sha: doc_id: 7798 cord_uid: 9ht7cqhu The treatment of patients poisoned with drugs and pharmaceuticals can be quite challenging. Diverse exposure circumstances, varied clinical presentations, unique patient-specific factors, and inconsistent diagnostic and therapeutic infrastructure support, coupled with relatively few definitive antidotes, may complicate evaluation and management. The historical approach to poisoned patients (patient arousal, toxin elimination, and toxin identification) has given way to rigorous attention to the fundamental aspects of basic life suppport — airway management, oxygenation and ventilation, circulatory competence, thermoregulation, and substrate availability. Selected patients may benefit from methods to alter toxin pharmacokinetics to minimize systemic, target organ, or tissue compartment exposure (either by decreasing absorption or increasing elimination). These may include syrup of ipecac, orogastric lavage, activated single- or multi-dose charcoal, whole bowel irrigation, endoscopy and surgery, urinary alkalinization, saline diuresis, or extracorporeal methods (hemodialysis, charcoal hemoperfusion, continuous venovenous hemofiltration, and exchange transfusion). Pharmaceutical adjuncts and antidotes may be useful in toxicant-induced hyperthermias. In the context of analgesic, anti-inflammatory, anticholinergic, anticonvulsant, antihyperglycemic, antimicrobial, antineoplastic, cardiovascular, opioid, or sedative-hypnotic agents overdose, N-acetylcysteine, physostigmine, l-carnitine, dextrose, octreotide, pyridoxine, dexrazoxane, leucovorin, glucarpidase, atropine, calcium, digoxin-specific antibody fragments, glucagon, high-dose insulin euglycemia therapy, lipid emulsion, magnesium, sodium bicarbonate, naloxone, and flumazenil are specifically reviewed. In summary, patients generally benefit from aggressive support of vital functions, careful history and physical examination, specific laboratory analyses, a thoughtful consideration of the risks and benefits of decontamination and enhanced elimination, and the use of specific antidotes where warranted. Data supporting antidotes effectiveness vary considerably. Clinicians are encouraged to utilize consultation with regional poison centers or those with toxicology training to assist with diagnosis, management, and administration of antidotes, particularly in unfamiliar cases. The challenges to effective evaluation and management of a patient poisoned by drugs and pharmaceuticals are diverse. The circumstances surrounding exposure are often incompletely accessible. Poisoning signs or symptoms may be subtle or delayed. Patient-specific factors -pharmacogenetics and unique susceptibilities, drug-drug interactions, cultural or geographic practices, and underlying comorbidities -may complicate presentation, response to treatment, and outcome. Polypharmacy or mixed exposures may confuse the clinical presentation. Compared to the near-inexhaustible list of products and possible combinations, few specific antidotes exist. The toxicological profiles of newly intro-organ injury. Specific management of toxicant-induced hyperthermias follows later. Hypothermia may require active or passive rewarming techniques. Clinical hypoglycemia, which implies neuroglycopenia, must be rapidly reversed with administration of 0.5-1.0 g/kg of age-appropriate dextrose-containing solutions (D50 in adults, D25 in children, and D10 in neonates). Benzodiazepines (e.g., diazepam, midazolam, and lorazepam) are generally well tolerated and are first line agents for drug-and withdrawal-induced seizures and agitation. Persistent or refractory seizures should prompt consideration of empiric administration of pyridoxine and barbiturates (phenobarbital, pentobarbital), propofol, or ultimately, general anesthesia. Coincident endotracheal intubation may be required. Phenytoin and non-barbiturate anticonvulsants are typically ineffective or harmful in toxin-induced seizures [18, 19] . Altered mental status should also prompt parenteral administration of 100 mg thiamine hydrochloride. Alcohol-dependent patients without clinically apparent Wernicke's encephalopathy may require at least 200 mg of parenteral thiamine to improve neurological symptoms; overt Wernicke's encephalopathy necessitates a minimum of 500 mg thiamine hydrochloride three times daily for 2-3 days [20] . Naloxone use is considered in a separate section. Toxidromes (toxic syndromes) are characteristic signs and symptoms that correlate with exposure to certain xenobiotics. Identifying toxidromes suggests the etiology of the patient's condition and helps guide management. "Classic" class-effect toxidromes include anticholinergic, cholinergic, sedative-hypnotic, sedative-hypnotic withdrawal, opioid, and opioid withdrawal. These should be actively sought and managed if identified. While the patient is being stabilized, diagnostic investigations including a complete and thorough history and physical examination, laboratory analyses, and radiological studies may be undertaken to further characterize the exposure and effect. For significantly compromised patients, a typical "chemistry panel" (providing electrolytes, blood urea nitrogen, creatinine, and indirectly the anion gap), a complete blood count, arterial (or venous) blood gas, and lactate are reasonable studies. Urine or serum ketones may be required to determine the etiology of acidemia. Female patients benefit from an assessment of pregnancy status. It is useful to determine a serum acetaminophen concentration in suicidal patients or those with altered consciousness, as patients with significant acetaminophen poisoning may present without a toxidrome. Serum acetaminophen is detectable in 2-3% of patients without a reported history of ingestion; treatable concentrations are found slightly less frequently [21, 22] . Toxin-specific studies and other serum determinations are often not rapidly returned and should be obtained only if suggested by the history, physical examination, or bedside testing. Urine drug screening (UDS) is of minimal use in the acute management of intoxication. Results are not typically returned for hours; a reported "positive" substance may not be the proximate cause of the presenting condition (as the measured metabolites may persist in urine for days to weeks); and the UDS lacks sensitivity and specificity (particularly for opioids, benzodiazepines and other sedative-hypnotics, and amphetamines). Selected patients may benefit from methods to alter toxin pharmacokinetics -limiting exposure. A discussion of these modalities and their risks and benefits occurs in the following section. Ultimately, patients will require disposition depending of severity of presentation and anticipated sequelae, which may range from admission to intensive care units, cardiac monitoring (telemetry) units, ward beds, continued emergency department evaluation, to discharge. A psychiatric assessment and social assessment, when appropriate, should precede release from medical care. Appropriate and early consultation with medical toxicologists or regional poison centers may also assist with diagnosis and management. In the U.S., this has been simplified by a uniform telephone number (1.800.222.1222) for regional poison center consultation. The International Programme on Chemical Safety (IPCS) maintains a world directory of poison centers (http://www.who.int/ipcs/poisons/centre/directory/en/). Adjuncts to alter toxicant pharmacokinetics aim to minimize systemic exposure (either by decreasing absorption or increasing elimination) or to minimize exposure of a target organ or tissue compartment. In practice, this is achieved by expulsion or removal from the upper gastrointestinal tract (induced emesis, gastric lavage, or endoscopy); intraluminal binding to adsorptive materials (activated charcoal); or increasing intestinal transit time (cathartics and whole bowel irrigation). Endogenous elimination may be improved by more effective urinary clearance (urinary alkalinization and forced diuresis), improved hepatobiliary clearance, or "gut dialysis" with multiple-dose activated charcoal. Rarely, hepatic metabolism is altered to preclude ultimate toxicant formation (e.g., cimetidine to mitigate production of dapsone's methemoglobinemia inducing metabolite). Exogenous clearance utilizes hemodialysis, charcoal hemoperfusion, continuous renal replacement therapies, and exchange transfusion. All the adjuncts attempt to shift where a patient lies upon a particular dose-response curve (Fig. 2) . Drug recovery following gastrointestinal emptying techniques has been inconsistent; human studies attempting to demonstrate a survival benefit of any decontamination modality are inconclusive. Randomized trials in which a control group might not receive any decontamination could be considered unethical; volunteer studies using sublethal doses of xenobiotic cannot show mortality benefit. As might be anticipated from the fact that supportive care suffices for the majority of poisoned patients, a typical study of routine administration of charcoal following oral overdose of primarily benzodiazepines, acetaminophen, and selective serotonin reuptake inhibitors could not demonstrate benefit [16, 17, 23] . Past studies have suffered from significant exclusions. Recommendations are based both on theoretical grounds (animal and in vitro studies demonstrating lower peak serum concentration or faster serum Drugs and pharmaceuticals: management of intoxication and antidotes 403 clearance) and human studies with surrogate endpoints such as marker studies or area under the curve of plasma concentration versus time (AUC) improvement. Aggressive detoxification may be required for certain lethal toxins for which few antidotal options exist. Most gastric emptying techniques are thought to be relatively ineffective beyond 1 hour. These constraints diminish possible benefit. For example, the median time from ingestion to arrival at a health care facility is on the order of 2 hours, and only about 10% of patients can be lavaged within the idealized 1-hour time frame [24] . Although in ideal situations (patients presenting early to experienced health care providers with readily available ipecac syrup) pill retrieval averages 45-55%, ipecac's benefits can be completely negated when administration is delayed as briefly as 30 min [25] [26] [27] [28] . When orogastric lavage is performed by experienced providers within 5 min of ingestion, clinical manifestations of ingested xenobiotics have been prevented [29] . Practically, efficacy of tablet retrieval rates reduces to 45% in some cases and improvements in AUC vary from zero to 60% (averaging ~35%) [27, [30] [31] [32] . Similarly, restricting activated charcoal (AC) administration to patients presenting to health care within the first hour post ingestion would exclude up to 90% of poisoned patients from the potential benefits of AC when administered beyond an hour [24, 33] . Earlier administration of AC is more efficacious [34] . However, home and prehospital use of AC decreases the time to treatment, but has not improved clinical outcomes [35] . Drugs with opioid or anticholinergic properties that decrease peristalsis or particularly large ingestions, which independently decrease intestinal motility, may modify decision making in delayed presentations [36, 37] . Figure 2 . Adjuncts to alter toxicant pharmacokinetics attempt to shift where a patient lies upon a particular (idealized) dose-response curve. Risk will likely outweigh benefit if the patient begins at point A (negligible morbidity and mortality) and systemic exposure is reduced to B. This is the case for many drug poisonings which are managed effectively by supportive care alone. Decontamination might provide significant benefit if the patient lies upon the steep aspect of the curve [reduction from C to D -the same fixed amount as from A to B (although a percentage reduction could also be envisaged)]. With overwhelming overdose (point E), despite decontamination, benefit would be unlikely (point F). Independent of side effects, the efficacy of one modality over another or combination therapy is debated. Some studies rate ipecac syrup more efficacious than orogastric lavage, but most studies have found little or no difference, and neither has been shown to be more effective than spontaneous emesis [26, 27, 31, 38] . AC has demonstrated ~50% better reductions in AUC than ipecac, which may improve or worsen its efficacy [31, 39, 40] . Gastric lavage adds no benefit to AC, except for the most critically ill patients [34, 41, 42] . Compared directly, AC has better impact than lavage on AUC and clinical effect [29, 31, 43] . Data are equivocal regarding whole bowel irrigation's ability to function similar to multiple-dose AC (MDAC) as a medium for "gut dialysis" [44, 45] . Syrup of ipecac is obtained from a root extract of the Amazonian flowering plant Psychotria ipecacuanha [46] . Its active alkaloid components, cephaeline and emetine, induce emesis via local irritation and central stimulation of 5-hydroxytryptamine (serotonin) 5-HT 3 receptors [47] . Following appropriate dose (10 mL for infants, 15-20 mL for children under 12, and 30 mL otherwise), roughly 90% of patients have a first episode of emesis within 20 min [48, 49] . Patients average three episodes in 30 min [50] . However, since ipecac's removal from most homes, the median time to administration in the acute care setting is delayed on the order of an hour, with only one-third of patients successfully vomiting within the first hour post ingestion [51] . Indications for ipecac are limited. A routinely cited example is a patient known to have taken multiple lithium tablets, which do not bind AC and may not fit through a lavage tube, who presents early to health care [50] . The American Academy of Pediatrics no longer recommends ipecac syrup for home use; ipecac use does not impact outcomes or decrease utilization of emergency services [52, 53] . Ipecac may or may not have a role in other rare ingestions that mandate gastrointestinal decontamination, but are not amenable to orogastric lavage, AC, whole bowel irrigation, or an antidote; the patient must present alert and early (<60 min post ingestion) to medical care [50] . Unsurprisingly, ipecac's most common side effect is persistent emesis. As many as eight emetic episodes occurring more than 60 min after ipecac administration have been reported [54] . This impairs administration of oral therapeutic agents, as induced emesis can last up to several hours [55] . Prolonged vomiting associated with induced sedation or absent airway reflexes increases the risk of aspiration bronchospasm, pneumonitis, and pneumonia [28, 50] . Other life-threatening side effects have been reported, including bradycardia, CNS depression, Mallory-Weiss esophageal tears, pneumomediastinum, pneumoretroperitoneum, and intracranial hemorrhage [50] . Emesis of caustics reexposes damaged esophageal mucosa to the caustic agent. Analogous pulmonary aspiration concerns accompany induced emesis of hydrocarbons. Orogastric lavage is performed via a large bore orogastric tube (adults, [36] [37] [38] [39] [40] French; children, 24-28 French) with fenestrae large enough to accommodate whole tablets [32] . Serial 500-mL aliquots (100-250 mL in pediatric patients) of normal saline or lactated Ringer's solution are administered and suctioned until retrieved liquid is clear. Orogastric lavage can be expected to have its best risk-to-benefit ratio when patients present early enough to have a significant gastric burden, and when severe toxicological effects are manifest or expected to become manifest [32, 42] . Because advancement of stomach contents does occur despite proper left lateral decubitus positioning [26] , AC (see below) is sometimes provided prior to crystalline lavage [32, 43] . Introduction of a large, relatively rigid tube requires a cooperative patient with a protected airway (typically an endotracheal tube if the patient is ill enough to warrant gastric lavage). Orogastric lavage risks hypoxia, dysrhythmia, laryngospasm, hypothermia, gastrointestinal or pharyngeal traumatic laceration or perforation, fluid and electrolyte abnormalities, and vomiting with subsequent aspiration pneumonia [32, 56, 57] . AC is a convoluted macromolecule created via pyrolysis of carbonaceous material and subsequently "activated" with steam to further increase surface area [58] . The multiple pores of various size on the surface of each macromolecule of AC account for its high adsorptive affinity for a multitude of xenobiotics -particularly chemical species that are nonionized, aromatic, and/or branched [34, 53, 59] . Maximal xenobiotic binding occurs in 10-25 min [60] . AC decreases AUC by as much as 60%, seems to improve clinical outcomes for critically ill patients, and may benefit in certain poisonings such as acetaminophen [31, 40, 61, 62] . It also increases the rate of endogenous clearance of drugs with long half-lives and some degree of entero-enteric or enterohepatic circulation [59, 63, 64] . Those findings suggested the use of MDAC as a "gut dialysis" for toxins with slow pharmacokinetics [65, 66] . A meta-analysis of volunteer studies demonstrated increased clearance of xenobiotics with longer half-lives, but not necessarily improved clinical outcome [67, 68] . MDAC has enhanced amitriptyline, carbamazepine, dapsone, dextromethorphan, phenobarbital, phenytoin, quinine, and theophylline elimination, although without definitive clinical benefit in controlled trials [63, 64, 69] . Two studies provided conflicting results for survival benefit of MDAC for yellow oleander poisoning [70, 71] . The fraction of unbound xenobiotic decreases as the charcoal-to-toxin ratio increases from 2.5:1 up to 50:1, although the yield curve levels off near 10:1 [59, 72] . In theory, the dose of AC administered to a poisoned patient would be ten times the mass of ingested xenobiotic, but those values are unknown in most clinical situations [73] . AC is practically dosed based on the patient's weight (1 g/kg), which can be divided into multiple smaller doses to be administered every 2-4 hours [59] . Although optimum dosing is unclear, MDAC is administered hourly, every 2 hours, or every 4 hours at a dose equivalent to 12.5 g/hour [66] . Pediatric charcoal doses are lower due to generally smaller ingestions and gut capacity. The total dose administered is the major determinant of efficacy particularly for larger overdoses, and can be administered continuously [74] . Emesis occurs in up to 12% of patients receiving AC; patients receiving AC via nasogastric tube or who vomited previously are at greater risk for emesis [75, 76] . Rarer complications include aspiration and intestinal obstruction or perforation [55, 59, 77, 78] . Aspirated AC may produce bronchiolitis obliterans, acute respiratory distress syndrome (ARDS), and death [79] . AC adheres to mucosa and obscures endoscopy; mineral acids and bases will not adhere to charcoal. AC poorly adsorbs short chain alcohols and metals such as iron, lead, and lithium [80] . AC administration requires an intact mental status or protected airway. Flavoring agents increase the palatability of AC for volunteers, but poisoned patients do not show increased compliance/tolerance with flavored AC [81] . Cathartics induce watery evacuation of bowel within a few hours. Hyperosmotic cathartic agents such as sorbitol are non-absorbed, osmotically active substances that draw water into the lumen, where increased intestinal volume and pressure promote peristalsis. So-called "saline" cathartic agents such as magnesium salts also directly stimulate smooth muscle to induce peristalsis [82] . Cathartics alone are not recommended for ingested poisons [83] . Cathartics have many adverse effects, including volume depletion, hypernatremia, hypermagnesemia, hyperphosphatemia, hypocalcemia, metabolic alkalosis, pain, nausea, emesis, and flatus [84, 85] . Sorbitol or laxatives are sometimes used in conjunction with the first dose of AC. While theoretically beneficial -minimizing the possible constipation of AC or promptly delivering AC to the duodenum, they do not increase the efficacy of AC [74, 86, 87] . Sorbitol is implicated in the fluid/electrolyte changes that occur with MDAC: hypermagnesemia, hypernatremia, and volume depletion [55, 84, 88] . Repetitive cathartic doses have been associated with rectal prolapse and death [89, 90] . Whole bowel irrigation (WBI) employs polyethylene glycol (PEG), a large, non-absorbable organic polymer and an electrolyte lavage solution (ELS) is-Drugs and pharmaceuticals: management of intoxication and antidotes 407 osmotic to serum. Large PEG-ELS volumes are introduced into the alimentary canal with less risk for fluid and electrolyte shifts caused by traditional cathartics. PEG-ELS provides non-viscous bulk for rapid transit of material in a normally functioning gastrointestinal tract. WBI should induce evacuation within 60 min, but requires 6 hours on average for complete effect. Reported improvements in AUC are modest given the more rapid absorption time for most pharmaceuticals [91] . However, reduction in AUC can be as high as 30% with poorly absorbed products or modified release preparations [92] . WBI might be considered for slowly absorbed significant ingestions such as iron, lead, and lithium, as well as modified-release preparations of β-adrenergic antagonists, bupropion, calcium-channel antagonists, carbamazepine, and theophylline [93] [94] [95] [96] . WBI is also employed to rid patients of enterally transported illicit substances which produce toxicity upon packet rupture or leakage (e.g., cocaine, heroin, and methamphetamine) [97] . Standard dosing protocols are 1.5-2 L/h (25 mL/kg per h) enterally until rectal effluent is clear [92] . At this point, intestinal contents are assumed to have been displaced, although this is not always true [91, 98] . Nasogastric tube placement is generally required to sustain compliance with the large volume requirements, and pretreatment with an antiemetic is prudent [98] . WBI may produce nausea, vomiting, cramping, and flatus. PEG-ELS for colonoscopy has precipitated colonic perforation [99] . Unintentional bronchial administration of PEG-ELS can produce acute lung injury [100] . Ileus, obstruction, perforation or threatened perforation should preclude WBI; a protected airway is required. Desorption of toxins from AC by PEG has been demonstrated in vitro and in vivo [93, 101] . Support for endoscopic therapy consists of limited case reports of retrieval in ingestions of cocaine packets, lead pellets, and medication such as sustained release calcium channel antagonists, clomipramine, iron, and meprobamate [102] [103] [104] [105] [106] . The procedure might be warranted for certain ingestions or cases of pharmacobezoar formation of toxic substances. Complications include perforation, aspiration, hemorrhage, and anesthetic-associated hemodynamic changes. When endoscopy fails, surgery may be required for definitive removal [107, 108] . Surgery may be required in patients with enterally transported illicit substances either due to failure of passage (with or without WBI), obstruction, or severe toxicity upon packet rupture or leakage [109, 110] . Weak acids in an alkaline environment exist predominantly in ionized form. Biological membranes are relatively impermeable to these charged molecules. Alkaline serum thus inhibits the diffusion of acidic toxins (low pK a ) across cellular membranes. Similarly, an alkaline urinary pH promotes renal sequestration (or "ion-trapping") of acidic species from the systemic circulation. The relative intolerance of biological systems to acidosis limits the effectiveness of converse urinary acidification (via ascorbic acid or diluted HCl solutions) for renal sequestration of weak bases. Critically ill patients may have reduced drug clearances due to decreased hepatic and renal perfusion, and thus interventions that increase clearance/ elimination have the potential to significantly reduce toxicity [111] . Alkalinization improves renal elimination of chlorpropamide, 2,4-dichlorophenoxyacetic acid, diflunisal, fluoride, mecoprop, methotrexate, phenobarbital, and salicylate [112] . Urine alkalinization is considered first line therapy in patients with moderate salicylism who do not meet hemodialysis indications. Dosing of 1-2 mEq/kg of 7.5-8.4% bicarbonate provided over 1-2 min is followed by "normal" bicarbonate infused at double the standard rate of i.v. fluid maintenance. The "normal" bicarbonate solution is prepared by adding three ampules of sodium bicarbonate (totaling 132-150 mEq) in 1 L 5% dextrose in water (D5W). The rate is titrated to maintain an alkaline urinary pH, without exceeding a serum pH of 7.55 [112] . Alkalemia decreases ionized calcium. Volume overload may occur, particularly in patients with congestive heart failure, acute renal failure, or end-stage liver disease. Bicarbonate treatment induces hypokalemia. As the proximal renal tubular cells conserve serum potassium by exchanging protons for urinary potassium, this defeats urinary alkalinization. Therefore, maintaining a normal serum potassium, with frequent monitoring and supplemental administration and/or inclusion in the bicarbonate solution, are important components of urine alkalinization. Saline diuresis is utilized to improve excretion and minimize toxicity of overdose of ions such as magnesium, calcium, and lithium in patients who do not meet hemodialysis indications [113] [114] [115] . Hypermagnesemia may occur with excessive antacid use, gargling or ingesting magnesium sulfate compounds, and iatrogenic error [113, 116] . Hypercalcemia can result from excess calcium (in antacid tablets) or vitamin D ingestion or parenteral administration [117, 118] . Renal lithium toxicity presumably results from cytotoxic accumulation of lithium entering via the apical epithelial sodium channel [119] . Ensuing nephrogenic diabetes insipidus, characterized by increased water and sodium diuresis, can result in dehydration, hyperchloremic metabolic acidosis, and renal tubular acidosis. In volume depletion, activation of the reninangiotensin-aldosterone axis leads to active resorption of sodium, and thus lithium, from the distal convoluted tubules. Therefore, adequate volume repletion with saline is prerequisite for effective renal elimination of lithium. Boluses of 0.9% sodium chloride are administered until the patient is clinically euvolemic. Saline infusion is then provided at 1.5-2 times a standard maintenance rate. Throughout treatment renal function, urine output, and electrolytes are monitored. Congestive heart failure, renal failure, or end-stage liver disease moderate volume administration and make saline diuresis less attractive than hemodialysis in significant ingestions. Loop diuretics such as furosemide inhibit sodium resorption in the proximal convoluted tubules, and would theoretically promote elimination of lithium as natriuretics. However, these effects are countered by the action of the renin-angiotensin-aldosterone axis on the distal convoluted tubules, and diuretics do not seem to improve outcomes in lithium overdose or radiographic contrast exposure [120, 121] . In hemodialysis (HD) the patient's blood is pumped through a circuit that includes a cartridge consisting of thousands of semi-permeable, membranelined capillary tubes. The blood traverses the cartridge counter-current to a circulating buffered salt solution (a.k.a. dialysate) before returning to the patient's venous circulation. Diffusible molecules flow down their electrochemical gradient from the serum to the dialysate. Hemoperfusion (HP) employs a similar circuit, but the cartridge is enveloped with AC (rather than a circulating dialysate) to adsorb xenobiotics regardless of plasma protein binding, and leave serum electrolytes largely unchanged. Continuous arteriovenous or venovenous hemofiltration (CAVH or CVVH) employ lower pressures and flow rates than HD over longer sessions for patients unable to tolerate HD or to remove xenobiotics with slow tissue redistribution [122, 123] . Peritoneal dialysis (PD) is ineffective in poisoning management, given its inherently slow kinetics and the availability of HD [124] . Extracorporeal therapies may be warranted when criteria are met for both the xenobiotic and the patient [125] . Favorable dialyzable toxin properties include low volume of distribution (V d ), relatively low molecular weight, and poor serum protein binding (or binding that worsens in overdose, as is the case for salicylate and valproate) [126] . Patient characteristics suggesting extracorporeal therapy include signs or symptoms of significant end organ toxicity; impaired elimination secondary to baseline comorbidities or critical illness-induced hypoperfusion; inability to tolerate or refractory to antidotal strategies (such as bicarbonate or saline); inadequate response to supportive care measures; concurrent electrolyte derangements (e.g., metformin-associated lactic acidosis); or serum drug concentrations historically associated with severe outcome [127] . Traditionally, charcoal HP was used for xenobiotics significantly bound to plasma proteins, but its use is declining while (high-flux membrane) HD increases. Methanol, ethylene glycol, salicylates, lithium, halides, theophylline, and metformin-associated lactic acidosis are commonly treated with dialysis [125] . HD is used for valproate and carbamazepine poisoning; however, in the absence of high-flux dialysis membranes, the characteristics of charcoal HP may more appropriately address the larger V d and protein binding [128] . Common side effects of extracorporeal elimination include hypotension, bleeding, and infection. Enhanced clearance of therapeutic medications and antidotes (e.g., antibiotics, fomepizole, N-acetylcysteine, water-soluble vitamins) may occur. The need for dialysis must be anticipated early; several hours of preparation time may be required to secure vascular access, equipment, and personnel. Exchange transfusion is a total blood volume exchange administered in small aliquots. Serial frequent phlebotomy of a small amount of circulating blood occurs with simultaneous transfusion of equivalent donor blood. This process is repeated until two to four vascular volumes have been exchanged. While the procedure is very rarely used for toxin removal, exchange transfusion is more familiar to clinicians treating severe hemolytic diseases of the newborn, hyperbilirubinemia without hemolysis, and sickle cell crisis. Exchange transfusion removes xenobiotics that are large or bound to plasma proteins, such as thyroxine, iron, or theophylline [129, 130] . For lifethreatening ingestions, exchange transfusion is a viable option for neonates and infants whose immature vasculature cannot tolerate extracorporeal elimination modalities or in institutions lacking pediatric dialysis capacity. Exchange transfusion has been successfully employed in pediatric iron, isoniazid, phenobarbital, salicylate, theophylline, and vincristine overdose [129] [130] [131] [132] [133] [134] . It has also been suggested for refractory drug-induced methemoglobinemia [135] . Whole blood exchange was utilized in an adult with a 50fold cyclosporine dosing error [136] . Anticipated complications arise from vascular access, bleeding, hypoglycemia, hypotension, and blood product administration (immune-mediated reactions, blood incompatibility, and infections). Several hyperthermic syndromes are caused by xenobiotics. These are generally spectrum disorders, whose features may overlap with other conditions such as CNS infection, agitated delirium, and sepsis. Malignant hyperthermia (MH) occurs in patients with an autosomal-dominant defect in genes encoding the skeletal muscle ryanodine receptor (RyR-1) or the voltage-gated calcium channel (Cav1.1) who are exposed to volatile anesthetics or depolarizing muscle relaxants (succinylcholine) [137] . Hypomagnesemia may increase the probability and possibly severity of an MH event [138] . The subsequent rapid increase in myoplasmic calcium concentration increases muscle metabolism and heat production and produces muscle contractures and hyperthermia. Neuroleptic malignant syndrome (NMS) is characterized by high fever, autonomic instability, altered mental status, and muscle rigidity. Potent antipsychotics (neuroleptics) such as haloperidol and other medications (metoclopramide, droperidol, and promethazine) with significant dopamine antagonism, as well as abrupt cessation of dopaminergic agents such as those used in Parkinsonism, can precipitate this life-threatening syndrome [139] . NMS typically develops over several days and is characterized by ''lead-pipe'' rigidity [139] . Drugs that impair serotonin breakdown or re-uptake, those that act as serotonin precursors or enhance its release, or those that are serotonin agonists may lead to serotonin syndrome. Like NMS, serotonin syndrome is a spectrum disorder for which various signs and symptoms have been proposed to establish diagnosis (e.g., Sternbach and Hunter criteria) [140, 141] . In its most severe form it consists of high fever, autonomic instability, altered mental status, and may have associated diaphoresis, shivering, tremor, diarrhea, or spontaneous clonus. In serotonin syndrome, onset of symptoms is usually rapid, with 60% of patients with the serotonin syndrome presenting within 6 hours of drug exposure, and tremor and hyperreflexia predominant in the lower extremities may be a prominent feature [142] . Sympathomimetic-associated hyperthermia, seen with acute intoxication with cocaine, amphetamines, substituted amphetamines, and phencyclidine, may be clinically indistinguishable from serotonin syndrome [143] . Additionally, the agitated delirium engendered by these agents may be difficult to distinguish from that induced by hyperthermia itself. Patients with anticholinergic-associated hyperthermia will generally present with a compatible "toxidrome" -agitation; mydriasis; dry, hot, and erythematous skin; hypoactive bowel sounds; and urinary retention. While rare, thyrotoxicosis factitia, the ingestion of excess thyroid hormones due to inadvertent intake (pharmaceutical or food contamination), misuse (dieting), or significant intentional ingestion may produce hyperthermia [144, 145] . Hyperthermia may accompany toxicity with agents that uncouple oxidative phosphorylation (e.g., salicylates, dinitrophenol, pentachlorophenol) [146] . Multiple medications can also complicate or contribute to environmental hyperthermia. Several reviews and epidemiological data from major heat waves have demonstrated that anticholinergics, antiepileptics, antihistamines, antihypertensives in general and diuretics in particular, antipsychotics, and others contribute to excess morbidity and mortality [147, 148] . Conversely, exogenous heat stress can increase mortality from specific xenobiotics. In an urban setting at ambient temperatures above 31.1°C, the mean daily number of fatal cocaine overdoses increased markedly [149] . Regardless of the cause for the hyperthermic syndrome, cessation of any possible offending or contributing agents and rapid cooling is critical. The degree of hyperthermia produced correlates with death and neurotoxicity in animal models, and temperature normalizing intervention is critically impor-tant in attenuating CNS injury and mortality [150] . Studies from the Chicago and France heat waves show that this is rarely done in a timely manner (if at all) in cases of environmental hyperthermia, with devastating results [147, 148] . The benefits of rapid cooling by ice water immersion were demonstrated over 80 years ago [151] . A large review concluded that cooling methods based on evaporative heat loss are less efficient than immersion in ice water in dissipating heat [152] . Additional studies demonstrate that cooling rates of up to 0.15-0.20°C/min can be achieved with immersion, two to three times that of evaporation [153, 154] . Regardless of the method used, effectiveness should be repeatedly assessed. Sedation with benzodiazepines and rigorous supportive care are necessary adjuncts in significant cases. This is primarily accomplished with titrated doses of benzodiazepines to inhibit muscle rigidity and control agitation. Animal models have demonstrated the benefit of benzodiazepines in prolonging survival, preventing seizure, and attenuating agitation in the toxicological hyperthermias [155, 156] . Phenytoin is ineffective in animal models [157] . Phenothiazines and butyrophenones, while reported, may have delayed onset and compromise mental status, lower seizure threshold, impair heat dissipation, and worsen hypotension [143] . Neuromuscular paralysis may be required to limit further heat generation in cases of NMS, serotonin syndrome, and sympathomimetic-associated hyperthermia. As the pathophysiology of MH is beyond the neuromuscular junction, paralytics are unlikely to provide benefit. Rapid i.v. administration of dantrolene, a direct-acting skeletal muscle relaxant, is the only drug proven effective for prevention and treatment of MH. Dantrolene disrupts the pathogenic excitation-contraction coupling by acting at RyR-1 to suppress depolarizationinduced sarcoplasmic reticulum calcium release and normalize the voltage dependence of contractile activation [158] . Reversal of increased myotube sensitivity may also play a role [159] . Intravenous 2-3 mg/kg dantrolene is repeated until symptoms are controlled or 10 mg/kg (or more) has been administered. Following initial treatment, 1-2 mg/kg i.v. or per os is given every 6 hours for 24-72 hours to prevent recurrence. Dantrolene is packaged in vials containing 20 mg dantrolene sodium; thus, multiple vials are needed for treatment of adult patients. A large review of NMS cases did not suggest a beneficial role for dantrolene, although one case-controlled analysis found benefit [160, 161] . Bromocriptine, a dopamine agonist, has been used (off-label) to treat NMS at doses ranging from 5 to 20 mg every 6 hours [143] . Common side effects include hypotension, dyskinesia, erythromelalgia, and hallucinations. Cyproheptadine, developed as an antihistamine, additionally antagonizes 5-HT 2 receptors. Cyproheptadine for serotonin syndrome (off-label) is initially used in a dose range of 4-12 mg, followed by 2 mg every 2 hours for persistent symptoms; upon symptom control, 8 mg maintenance dosing is provided every 6 hours [142] . The tablet form necessitates administration orally or crushed via nasogastric tube. N-Acetylcysteine (NAC) provides an effective means of prevention and treatment of acetaminophen (N-acetyl-p-aminophenol, APAP; paracetamol)induced hepatotoxicity. NAC is also employed to preclude radiographic contrast-induced nephropathy [162] . The ultimate toxicant of APAP, N-acetyl-pbenzoquinone imine (NAPQI) generated primarily by CYP2E1 and CYP3A4, depletes glutathione (GSH), binds intracellular components, and, through an incompletely understood process, produces hepatic injury, centrilobular necrosis, or hepatic failure [163, 164] . NAC works by multiple mechanisms. It augments APAP sulfation to a nontoxic metabolite, it acts as a glutathione precursor or glutathione substitute to detoxify NAPQI, and possibly reverses NAPQI oxidation [165, 166] . NAC provides substantial benefit even in cases of delayed presentation following overdose [167] . Extra-hepatic benefits of NAC include improving cardiac index and systemic mean oxygen delivery despite decreasing systemic vascular resistance [168] . In a range of hepatic disorders, NAC improved baseline oxygen delivery, oxygen consumption, and dye clearance in a majority of patients [169] . Liver blood flow and cardiac index improved in septic shock patients provided NAC [170] . Only L-NAC is beneficial. Animal experiments demonstrate that the L-isomer, derived from physiological L-cysteine, prevents hepatotoxicity and provides prolonged elevations of hepatic glutathione [171] . Nonphysiological D-NAC cannot increase glutathione stores or prevent hepatotoxicity, despite increasing acetaminophen sulfation [172] . According to Rumack [163] , the oral NAC dosing strategy was reached by estimating the absorption and turnover rate of glutathione at 6 mg/kg per h and an FDA safety factor of 3, to yield 70 mg/kg every 4 hours [6 (mg/kg per h) × 4 (h) × 3 (safety factor) = 72 ≈ 70 mg/kg every 4 h]. There were several assumptions as to "normal" hepatic glutathione levels and APAP to NAPQI conversion. A 140 mg/kg loading dose was added to provide an early high hepatic dose. The 72-hour duration of oral therapy was based on previous observations of multiple patients with prolonged APAP half-lives and a desire to implement a protocol that would accommodate those with half-lives longer than 12 hours (anticipating disappearance after five half-lives). While many have suggested that the 72-hour oral course is excessive, particularly after APAP has disappeared from the serum, the optimal duration of therapy is unclear. Studies assessing a shortened or "patient-tailored" approach have been small or methodologically limited [173, 174] . The Rumack-Matthew nomogram guides initiation of NAC therapy in single acute ingestions. The "treatment line" is anchored at an APAP serum concentration of either 200 μg/mL ("200 line") or 150 μg/mL ("150 line") at 4 hours post ingestion and decreased by 50% every 4 hours. The slope of the treatment line does not reflect APAP kinetics. The "150 line" is utilized in all patients in the U.S. and Australia; in the U.K. and elsewhere the "200 line" is employed, with a "100 line" modification for an array of individuals deemed at "high-risk": ethanol tolerant, those at risk for glutathione depletion (malnutrition, HIV, eating disorders, cystic fibrosis), pregnancy, and those prescribed enzyme-inducing drugs (carbamazepine, phenytoin, phenobarbitone rifampacin, isoniazid, etc.) [165, 175] . The U.S. multicenter study substantiated the safety and efficacy of its approach [176] . Proponents of the "150 line" point to the fact that 3.45-12.9% of patients above the "150 line" but below the "200 line" developed biochemical hepatotoxicity (aspartate aminotransferase, AST >1000 IU/L at any time during their course) in the U.S. multicenter trial and that patient deaths have occurred in untreated patients "between the lines" [177, 178] . In patients presenting near 8 hours after ingestion, or if a level is not available before 8 hours post ingestion, NAC is begun while awaiting APAP results and then continued or stopped once the results are available and have been plotted on the nomogram. If the time of ingestion is unknown or more than 24 hours has passed, NAC is administered. When APAP concentration and transaminase results are obtained, if transaminases are elevated or if measurable APAP exists, a full course of treatment is provided. With normal aminotransferases and without detectable APAP, treatment is not required. Concentrations obtained less than 4 hours post ingestion are not useful except to completely exclude ingestion (i.e., it is useful only if the APAP concentration is undetectable). Ongoing absorption may place individuals above the line at 4 hours, or metabolism or charcoal administration may result in a patient falling below the nomogram at 4 hours. In cases of chronic ingestion (>7.5 g/day in adult), laboratory evaluation and treatment are provided as for an unknown time of ingestion. With elevated transaminases or measurable APAP, NAC is provided. Oral NAC is cheap and familiar to clinicians. It has minimal side effects (other than vomiting and odor) and is preferred in patients with bronchospastic disease. Its use can become problematic in cases where oral delivery is compromised, e.g., in patients with depressed mental status, significant vomiting, or impaired gastric motility. Use of an anti-emetic is encouraged. Intravenous NAC appears to be similarly efficacious to oral NAC and eliminates many delivery issues. It has a much shorter therapy course (21 hours), expediting medical and psychiatric disposition. It avoids first pass metabolism in cases where the liver is not the only target or interest, such as those with cerebral edema or pregnancy. While i.v. NAC is slightly more expensive, total hospital charges may be less due to decreased treatment time. Histamine-mediated anaphylactoid reactions are more commonly seen with rapid i.v. loading and in patients with lower APAP levels [179] . Mild reactions have been treated by slowing the infusion rate and providing i.v. diphenhydramine, although this might alter NAC and APAP kinetics. Dosing complexity -150 mg/kg in 200 mL of 5% dextrose over 1 hour, followed by 50 mg/kg in 500 mL of 5% dextrose over 4 hours (12.5 mg/kg per h), and then 100 mg/kg in 1000 mL of 5% dextrose over 16 hours (6.25 mg/kg per h) -yields frequent administration errors [180] . The supplied 20% solution was too concentrated for children, and dilution according to adult guidelines resulted in excess free water, and cases of hyponatremia and seizures [181] . The current U.S. package prescribing information (http://www.acetadote.net/PI_Acetadote_Revised_Apr09.pdf) and dosage calculator website (http://www.acetadote.net/dosecalc.shtml) provide dosing and administration guidelines in patients of less than 40 kg. In a study limited by different comparison groups, data acquisition methodology, treatment location and several other factors, 20-hour only i.v. NAC was favored in patients with early presentation (<12 hours), whereas late presentation favored oral 72-hour NAC [182] . However, continuous i.v. infusion in delayed presentations with APAP-induced fulminant hepatic failure showed clear benefit in a prospective study [167] . Whatever the route, prior to cessation of NAC therapy, negative APAP concentrations and normal transaminases must be ensured, particularly in cases of massive ingestion; hepatotoxicity may follow premature cessation of therapy [183, 184] . The 16-hour maintenance dose is continued in patients receiving i.v. NAC until APAP is undetectable and transaminases are normal (or at baseline). Experimental evidence and human case reports demonstrate both delayed absorption, delayed increase following initial decline, and "crossing the nomogram" with extended-relief, opioid-or anticholinergic-containing APAP products, or co-ingestants [185, 186] . In cases of hepatic failure, i.v. NAC is continued until resolution, transplant, or death. Historically, physostigmine (eserine), a reversible carbamate inhibitor derived from the seed (Calabar bean) of the vine Physostigma venenosum Balfour, was used in the ancient trial by ordeal [187] . Medicinal use of physostigmine was first reported in 1864 to reverse severe atropine poisoning [188] . Naturally available (-)-physostigmine is over 100 times more effective in inhibiting acetylcholinesterase and butylcholinesterase in tissue, erythrocytes, and serum in humans and animal models than its stereoisomer [189, 190] . This activity depends upon interactions within the hydrophobic pocket of the acetylcholinesterase active center, which is distinct from the catalytic site [191] . Additionally, physostigmine binds nicotinic receptors close to, but distinct from, the acetylcholine binding site on the α-subunit [192] . At low doses, physostigmine functions as an ineffective nicotinic receptor agonist, while at higher doses it produces marked channel blockade. Physostigmine's nonspecific analeptic properties [8] are no longer considered useful in overdose, given the clear benefits of supportive care. Indiscriminate use of physostigmine and an incomplete understanding of the pathophysiology of tricyclic antidepressant (TCA) poisoning was associated with bradydysrhythmias including asystole, seizure, and several deaths [193, 194] . In animal models, physostigmine is ineffective in attenuating TCAinduced seizures [195] . It failed to abolish dysrhythmias, decreased blood pressure, and at high doses enhanced TCA toxicity [196] . Physostigmine is currently recommended as a diagnostic and therapeutic agent for antimuscarinic poisoning [197] . Patients should have clear peripheral or central manifestations of the anticholinergic toxidrome. As a tertiary amine, physostigmine can cross the blood-brain barrier to reverse the central effects. An ECG should exclude sodium or potassium channel blockade (QRS or QT prolongation). Excessive physostigmine will produce a cholinergic syndrome, with muscarinic and nicotinic effects. As the adverse effects of bradycardia and bronchorrhea can produce significant morbidity, continuous cardiac monitoring and immediate access to atropine are recommended during physostigmine administration. Physostigmine, 1-2 mg in adults and 0.02 mg/kg (maximum 1.0 mg) in children is infused slowly over at least 5 min [198] . Repeat doses every 10-15 min can be provided if an adequate response does not occur and adverse effects are absent. Re-bolusing may be required in the setting of antimuscarinics with a prolonged duration of action. The anticonvulsants include carbamazepine, ethosuximide, felbamate, gabapentin, lacosamide, lamotrigine, levetiracetam, oxcarbazepine, phenobarbital, phenytoin, pregabalin, primidone, tiagabine, topiramate, valproic acid (VPA), vigabatrin, and zonisamide. These drugs enjoy widespread approved and offlabel use for additional conditions, e.g., fibromyalgia (pregabalin); neuropathy and neuropathic pain (carbamazepine, gabapentin, lamotrigine, levetiracetam, and pregabalin); panic disorder (tiagabine); migraine prophylaxis and treatment of obesity, ethanol dependence, and depression (topiramate); and bipolar disorder (carbamazepine, lamotrigine, and VPA). Treatment of anticonvulsant overdose is largely supportive, with particular attention to the CNS-depressant and cardiovascular effects of some of these agents. L-(R)-Carnitine exists as the sole specific antidote in this class for significant VPA (di-n-dipropylacetic acid, 2-propylpentanoic acid) poisoning. Patients with drug-associated mitochondrial toxicity (particularly from nucleoside analogs) and anthracycline cardiotoxicity might also benefit from its administration [199, 200] . The anticonvulsant properties of VPA derive from its ability to increase γ-aminobutyric acid (GABA) availability via inhibition of GABA transaminase and succinic semialdehyde dehydrogenase, to attenuate N-methyl-Daspartate (NMDA)-type glutamate receptor excitatory effects, and to slow the rate of recovery from sodium channel inactivation [201] [202] [203] . Additionally, VPA appears to affect inositol levels similar to lithium. Therapeutic concen-trations are 50-100 mg/L. Potentially toxic concentrations are greater than 120 mg/L. Oral absorption of VPA is excellent [204] . Peak plasma concentrations are generally seen in 1-4 hours, although this may be markedly delayed by overdose, enteric coating, or meals [205] . Manifestations of significant VPA toxicity include CNS effects (lethargy, seizure, coma, cerebral edema), respiratory depression, metabolic derangement (hypernatremia, hyperammonemia, hypocalcemia, metabolic acidosis, carnitine deficiency), gastrointestinal effects (nausea, vomiting, and abdominal pain), pancytopenia, pancreatitis, and hepatotoxicity [206, 207] . Valproate toxicity is seen both in intentional acute overdose and in those on chronic therapy, either without adequate carnitine supplementation or on complex regimes. Cells attempt to metabolize the VPA that is not directly excreted or glucuronidated in a manner similar to other fatty acids (Fig. 3) . Thus, VPA is conjugated with coenzyme A (CoA). Carnitine enters via an ATP-dependent transporter. VPA is then transferred to carnitine, the normal mechanism for fatty acids entry into the mitochondrion. However, VPA-carnitine both inhibits the carnitine transporter and also diffuses out of the cell to be lost via renal excretion [208] . Renal resorption of carnitine is also impaired [209] . These factors contribute to intracellular carnitine depletion. Once VPA-carnitine is shuttled into the mitochondrion, it is reattached to CoA. It then undergoes β-oxidation, in an attempt to generate 2-carbon molecules for entry into the Krebs cycle. The 2-en-VPA-CoA product is neurotoxic with a prolonged half-life. The terminal 3-keto-VPA product traps CoA, leading to its mitochondrial depletion. Decreased mitochondrial CoA yields decreased ATP production, diminishing usable cellular energy currency and further limiting carnitine entry into the cell (via an ATP-dependent carnitine transporter). Once carnitine is depleted, normal fatty acid metabolism cannot occur [206] . Fatty acid build up is thought to underlie the Reye's-like steatohepatitis, which can be seen in toxicity [210] . CoA is also needed to make N-acetylglutamate, an activator of carbamoylphosphate synthetase I (CPS I), a critical enzyme in the urea cycle. When its effectiveness is limited due to inadequate activator, ammonia cannot be incorporated, and consequently, its concentrations increase. Furthermore, as CoA is depleted, β-oxidation shifts to omega (ω), or terminal carbon oxidation. This creates (among others) the hepatotoxic 4-en-VPA product. 4-en-VPA additionally inhibits CPS I, further preventing nitrogen elimination and contributing to hyperammonemia. L-Carnitine (levocarnitine) supplementation has been recommended to reverse the adverse metabolic effects of VPA in cases of VPA-induced hepatotoxicity, VPA overdose, and primary carnitine-transporter defects [211, 212] . Hyperammonemia and serum and muscle carnitine deficiency are well described in patients chronically taking VPA [213] [214] [215] . Several studies and case reports demonstrate that carnitine supplementation reverses clinical symptoms, hypocarnitinemia, hyperammonemia, and VPA half-life prolongation in patients with toxicity due to chronic administration [216] [217] [218] . In patients with acute VPA overdose, limited clinical and laboratory data derived from case reports also suggest that reversal of metabolic derangements and improvement in clinical symptoms occurs when carnitine is provided [219] [220] [221] . A single large retrospective analysis showed a significant survival benefit with i.v. carnitine supplementation (with VPA cessation) in patients with valproate-induced hepatotoxicity [222] . L-Carnitine dosing for cases of overdose is not currently evidence based. An oral or i.v. dose of 100 mg/kg per day, divided and given every 6 hours (maximum daily dose 3 g), is provided to those patients with acute overdose and [201, 202, 204, 212, 482] . asymptomatic hyperammonemia or hepatotoxicity in the absence of CNS depression or metabolic derangement [211] . Symptomatic patients with hyperammonemia or symptomatic hepatotoxicity should receive 100 mg/kg L-carnitine i.v. over 30 min (maximum 6 g), followed by 15 mg/kg every 4 hours over 10-30 min until clinical improvement occurs [211, 223] . Others have supplemented at the higher dosing strategy when VPA concentrations exceed 450 mg/L [224] . In addition, given the decrease in protein binding that occurs, hemodialysis or hemoperfusion is recommended for patients with VPA concentrations exceeding 850-1000 mg/L or with severe clinical symptoms [202] . L-Carnitine is generally well tolerated. Side effects associated with carnitine supplementation are nausea, abdominal discomfort, dose-related diarrhea, and fishy body odor [223] . A small retrospective chart review found no adverse effects or allergic reactions in VPA overdose patients administered carnitine [225] . The current L-carnitine package inserts have no warnings or contraindications, but note that seizures have been reported to occur in patients, with or without pre-existing seizure activity, who received either oral or i.v. L-carnitine [226] . Up to 600 mg/kg per day for 5 days has been provided without complications [227] . The D-isomer and the racemate (D,L-carnitine) are contraindicated. Historic use of racemic D,L-carnitine was associated with myasthenialike syndromes and cardiac dysrhythmias, which disappeared after L-carnitine administration [228] . D-Carnitine also competitively depletes cardiac and skeletal muscles and kidneys of L-carnitine [229] . Dextrose Dextrose (D-glucose) is indicated to rapidly reverse organic or toxin-induced hypoglycemia (e.g., from sulfonylureas, insulin, ethanol, salicylates, β-adrenergic antagonists, quinolines, pentamidine, ritodrine, and disopyramide) [230, 231] . Hypoglycemia onset may be significantly delayed with certain agents (e.g., long-acting insulin or sulfonylureas). Limited CNS glycogen stores (in astrocytes) and the inability to acutely use free fatty acids make the CNS particularly vulnerable to hypoglycemia [232] . Patients (and providers) may be unaware of hypoglycemia in the absence of objective testing; both the counter-regulatory autonomic response and overt neurological deficit may be absent [233, 234] . Additionally, significant neuroglycopenia and hypoglycemia-associated delirium (particularly in salicylism) may occur despite a "normal" peripheral blood glucose [235] . A wide range of clinical presentations have been described, including diaphoresis, nausea, tachycardia, tremor, hypothermia, focal neurological deficits, and CNS agitation, confusion, or depression. These are generally reversible upon prompt treatment. Untreated hypoglycemia may result in seizure, coma, and death. Hypoglycemic seizures increase cerebral metabolic rate, contribute to ATP depletion, and produce irre-versible brain damage [236, 237] . For these reasons, when bedside testing is unavailable, a risk-benefit calculation has generally favored empiric dextrose administration in the absence of a very clear alternative history or explanation for altered mental status. Following a determination of absolute or relative hypoglycemia, 0.5-1.0 g/kg i.v of age-appropriate dextrose containing solutions should be provided immediately -D50W (50 g/100 mL) in adults, D25W (25 g/100 mL) in children, and D10W (10 g/100 mL) in neonates. Frequent re-evaluation of response to therapy is required. Glucose uptake and distribution, hyperglycemia-induced insulin secretion in those with a competent pancreas, and ongoing toxin exposure may cause recurrent hypoglycemia and necessitate repeat dosing. Feeding, which provides significantly more calories than each 50 mL ampule of D50W (85 kcal according to one manufacturer [238] ), should be commenced as soon as practicable. While D10W "maintenance" solutions may be subsequently required, at an infusion rate of 100 mL/h, this concentration only provides 34 kcal per hour. Continuous infusion of more concentrated solutions (e.g., D20W) requires a central venous catheter for administration. Only the D-isomer is clinically useful. Most glucose transporters (GLUTs) and the specific transporter required for facilitated diffusion of glucose across the blood-brain barrier, GLUT1 (SLC2A1), have a high affinity for D-glucose and negligible affinity for L-glucose [236, 239] . D-Glucose is also generally favored over other D-glucose epimers such as D-mannose or D-galactose. D50W is hypertonic and may cause phlebitis or thrombosis at the site of injection. Extravasations of solutions containing as low as 10% dextrose have caused significant tissue injury and necrosis, particularly in young children [240] . Pseudoagglutination of red blood cells may occur if concentrated dextrose solutions without electrolytes are administered simultaneously with blood through the same infusion set [238] . Hypertonic dextrose administration may also induce generally clinically irrelevant hypophosphatemia [241] . Octreotide acetate, a synthetic somatostatin analogue, is now favored in cases of refractory hypoglycemia due to sulfonylureas or quinine. It is FDA approved for treatment of acromegaly, carcinoid tumors, and vasoactive intestinal peptide tumors [242] . It is a more potent inhibitor of insulin secretion than the natural hormone [242] . In pancreatic β-islet cells, ATP generated from glucose uptake and subsequent metabolism normally induces closure of the ATP-dependent potassium channel by binding to its pore subunit (Fig. 4) . Sulfonylureas similarly induce channel closure after binding to a regulatory (SUR1) subunit. Increased intracellular potassium triggers calcium entry through voltage-dependent calcium channels, leading to increased cytosolic calcium and insulin exocytosis [243, 244] . Additionally, ATP contributes to insulin vesicles movement and provides a substrate for protein kinase A (PKA)-mediated phosphorylation. Octreotide binds to the somatostatin receptor (primarily SSTR 5 ) [243] . The subsequent effects continue to be explored and include inhibitory calcium channel effects, inhibition of adenylyl cyclase, and dephosphorylation of specific proteins required for movement and/or docking of vesicles [243, 245, 246] . Octreotide effectively suppresses endogenous insulin release in controlled studies in diabetics and in cases of sulfonylurea overdose, but does not (and would not be expected to have) an effect on exogenously administered insulin [247] [248] [249] . Several factors support octreotide usage following failure of initial dextrose administration and feeding. Bolused dextrose may produce hyperglycemia and thus subsequently stimulate an exaggerated insulin response, particularly when Figure 4 . Pancreatic β-islet cell mechanisms of insulin release and octreotide action (see text for details). Enzymes and substances: AC, adenylyl cyclase; ATP, adenosine triphosphate; cAMP, cyclic adenosine monophosphate; GLUT2, glucose transporter 2; PKA, protein kinase A; SFU, sulfonylurea; SSTR, somatostatin receptor. Symbols: ⊕, agonism or co-factor; ⊗, antagonism. Data used can be found in [243] [244] [245] [246] . sulfonylureas persist. This contributes to recurrent (sometimes more significant) hypoglycemia. A vicious cycle of serum glucose concentrations is described in case reports and controlled trials following dextrose administration after sulfonylurea exposure [249] [250] [251] . Additionally, as has been demonstrated, classic neuroglycopenic symptoms may not be present, and patients may need to be admitted during periods when circadian sleep patterns would complicate assessment. Octreotide administration also obviates the concern of excess water administration in pediatric patients receiving i.v. dextrose solutions. Relatively few trials are available to judge the efficacy of octreotide for sulfonylurea-induced hypoglycemia. In one study, glipizide was used to induce induced hypoglycemia (50 mg/dL) in eight healthy volunteers, who were then resuscitated with dextrose infusion, diazoxide, or octreotide [251] . Dextrose requirements were markedly less in patients provided octreotide and hypoglycemic events were markedly attenuated after all therapies were stopped. One retrospective chart review of nine patients demonstrated that octreotide significantly reduced the number of recurrent hypoglycemic events and dextrose requirement [252] . One prospective randomized controlled trial in 40 poisoned patients, despite a failure to control for carbohydrate intake and having an unusual dosing strategy (a single octreotide 75 μg dose subcutaneously), demonstrated consistently higher glucose values for the duration for which octreotide would be expected to be effective (6-8 hours) [253] . Controlled animal studies with 25-100 μg octreotide demonstrated a similar decrease in hypoglycemic events [254] . The remainder of human clinical experience of the effectiveness of octreotide in sulfonylurea overdose comes from abstracts, case reports, and case series (e.g., [249, 250, [255] [256] [257] ). Pediatric experience in sulfonylurea overdose comes only in the form of limited abstracts and case reports in children aged 12 months to 17 years [248, [258] [259] [260] . However, octreotide has been used for prolonged periods to treat persistent hyperinsulinemic hypoglycemia of infancy [261, 262] . Two human studies examined the effectiveness of octreotide in quinineinduced hypoglycemia. In one study of nine healthy volunteers, 50 μg/hour octreotide as a continuous i.v. infusion abolished quinine-induced insulin release [263] . The authors reported resolution of hypoglycemia in an additional patient being treated with quinine for Plasmodium falciparum malaria. A subsequent study in eight patients with P. falciparum malaria confirmed octreotide suppression of quinine-induced hyperinsulinemia [264] . Optimal dosing of octreotide has not been definitively determined. Initial doses of 40-100 μg subcutaneously in adults have been reported, although 50 μg every 6-8 hours is commonly provided [256, 265] . In children, an initial dose of 1.0-1.25 μg/kg is used, although up to 2.5 μg/kg (or more) has been reported [258, 260] . Peak serum concentrations are achieved within 30 min after subcutaneous administration and within 4 min after a short (3 min) i.v. infusion [266] . The elimination half-life (by either route of administration) is approximately 1.5 hours. In patients with severe renal impairment (which may have contributed to sulfonylurea-induced hypoglycemia in the first place), the plasma clearance is reduced by 50% [266] . The subcutaneous route is recommended due to longer duration of effect, as i.v. administration has resulted in treatment failure [267] . Side effects are generally minimal. Octreotide does inhibit gallbladder contractility and decreases bile secretion in normal volunteers [242] . When octreotide has been used to reverse sulfonylurea-induced hypoglycemia, bradycardia, hypokalemia, anaphylactoid reaction, and hypertension and apnea have been reported [257, 259] . Other adverse events include nausea, abdominal cramps, diarrhea, fat malabsorption and flatulence [268] . Octreotide also suppresses glucagon release, although hypoglycemia has been a concern only in patients on long-term therapy for organic hyperinsulinemia [269] . Glucagon is not generally recommended to correct hypoglycemia. Glycogen stores are frequently depleted by the time toxin-induced hypoglycemia manifests; glucagon's half-life (less than 20 min) is inadequate given the prolonged duration of the effect of sulfonylureas; and glucagon may exacerbate hyperinsulinemia [258] . Diazoxide, an antihypertensive agent, which reduces insulin release by opening the ATP-dependent potassium channel, is now of historical interest due to associated hypotension, reflex tachycardia, nausea and vomiting, and fluid retention [243, 265] . Since its introduction in 1952, isoniazid (INH, isonicotinic hydrazide, pyridine-4-carbohydrazide) has remained a mainstay for treatment and prophylaxis of mycobacterial infections [270] . The adult single tablet, 300 mg daily dose (4.3 mg/kg in a 70 kg individual) targets a peak plasma concentration of 3-5 μg/mL [271] . Acute INH toxicity may occur following ingestion of 20 mg/kg INH; it is common above 35-40 mg/kg [272] . The relatively narrow therapeutic window poses a significant risk for those with suicidal intent and for those who ingest extra pills to "catch up" after a brief period of incomplete compliance [273] . Historically, death rates of 21% were reported [274] . Seizures refractory to typical therapy, severe metabolic lactic acidosis, and coma may occur as early as 30 min post ingestion due to the rapid and nearly complete absorption of INH from the gastrointestinal tract. Seizures may occur at lower doses in those with pre-existing susceptibility. Associated respiratory failure, hypotension, and rhabdomyolysis may ensue. In patients provided 2.1-3.9 g (64-83 mg/kg) INH due to medication error, all experienced nausea or vomiting, vertigo, and coma within 30 min to 6 hours after ingestion [275] . Abnormal generalized discharges as sharp and slow waves were seen on EEG in all patients. Chronic INH toxicity may present with nausea, vomiting, hepatitis, hemolytic anemia, and neurological findings (restlessness, neuropathy, cerebellar findings, and psychosis). The acute clinical effects are a product of the multiple biochemical actions of INH, which lead to pyridoxine depletion and subsequent neuronal hyperexcitability (Fig. 5) [272, [276] [277] [278] . INH hydrazones inhibit pyridoxine phosphokinase, which activates pyridoxine. INH hydrazines and hydrazides inactivate active pyridoxal 5-phosphate. INH metabolites also complex with pyridoxal 5-phosphate, leading to increased urinary elimination. Glutamic acid decarboxylase (GAD) and GABA transaminase (GABA-T) both require pyridoxal 5-phosphate as a co-factor. Inhibition of GAD exceeds that of GABA-T [279] . The resulting GABA depletion and loss of neuronal inhibition is thought to underlie seizure activity. Metabolic acidosis may be profound -survival has been reported with a pH of 6.49 [280] . Seizure-associated lactate generation is substantial; INH-induced metabolic acidosis does not develop in paralyzed dogs (despite EEG evidence of seizure) [281] . Importantly, merely correcting the acidosis (e.g., by bicarbonate) does not prevent additional seizures or terminate INH toxicity [281, 282] . INH also impairs lactate conversion to pyruvate (Fig. 5) . Increased metabolism of fatty acids due to impaired glucose metabolism with hyperglycemia and ketonuria has been reported [272, 283] . INH also impairs cellular reduction-oxidation capacity via competitive inhibition of NAD [284, 285] . Pyridoxine deficiency also appears to play a role in INH-induced mental status changes (coma and lethargy) [275, 282, 286] . Appropriately dosed pyridoxine (vitamin B6) has been the mainstay of antidotal therapy for INH intoxication since the early reports of benefit versus his- Figure 5 . Mechanisms of isoniazid (INH) toxicity (see text for details). Enzymes (italicized): GABA-T, GABA transaminase; GAD, glutamic acid decarboxylase; GOT, glutamic-oxaloacetic transaminase; LDH, lactate dehydrogenase; PPK, pyridoxine phosphokinase; and SR, serine racemase. Substances: GABA, γ-aminobutyric acid; and SSA, succinic semi-aldehyde. Symbols: ⊕, agonism or co-factor; ⊗, antagonism. Data used can be located in [272, [276] [277] [278] 483] . Drugs and pharmaceuticals: management of intoxication and antidotes 425 torical controls [282] . Exogenous vitamin B6 provides the necessary precursor for the co-factor for GABA regeneration. Clinical experience with pyridoxine comes from case series, case reports, and animal data [273, 275, 281, 282, [287] [288] [289] . Clinical trials are absent due to ethical considerations. Vitamin B6 (as pyridoxine hydrochloride) is provided on a gram per gram basis for each gram of INH ingested, to a maximum of 5 g or 70 mg/kg (the empiric dose in ingestions of unknown quantity) [272, 282, 287] . A repeat dose can be provided if necessary. Due to the large amount of pyridoxine required, inadequate stocking and depletion of institutional and entire regional supplies have been widely reported [287, 290, 291] . In the convulsing patient, pyridoxine is administered i.v. at 0.5 g/min (5 g maximum) until seizure termination, with the remainder over 4-6 hours. Pediatric dosing should not exceed 70 mg/kg (5 g maximum). Large doses of pyridoxine have been safely administered; however, sensory neuropathy may occur with massive acute doses (>100 g) or chronic large daily doses [292] . Co-administration of benzodiazepines is synergistic in controlling seizures [288, 289] . Massive INH ingestion may require additional sedative hypnotics or anesthetic agents to suppress seizures [293] . INH is dialyzable, and hemodialysis has been used successfully in cases refractory to antidotal treatment, in those with extremely high plasma INH concentrations, and in patients with renal failure [283, 293] . Pyridoxine also appears to rapidly reverse the impaired consciousness seen in INH overdose [282, 286] . The CNS excitatory neurotransmitters include glutamate and D-serine, which with glutamate is a co-agonist of the NMDA receptor [278] . Examination of the metabolic pathways affected by pyridoxal 5-phosphate depletion (Fig. 5) suggests that inadequate stores of these neurotransmitters (due to inadequate co-factors for glutamic-oxaloacetic transaminase and serine racemase) might be contributory, in addition to general substrate or catecholamine deficiency. Pyridoxine therapy is also recommended for poisoning through other hydrazines or hydrazine precursors (e.g., Gyrometra mushrooms, monomethylhydrazine, and unsymmetrical dimethylhydrazine fuel). Pyridoxine is effective in treating the chronic INH-associated neuropathy, particularly in patients with renal failure. Doses of 10-50 mg pyridoxine/day have typically been used in the chronic setting [271] . Pyridoxine has no effect in prevention or treatment of INH-associated hepatic injury. Antineoplastic agents are used for the treatment of a variety of benign and malignant neoplasms. Some antineoplastic agents (such as the antifolates) have an expanded spectrum that includes use in rheumatology, dermatology, and obstetrics and gynecology. Toxicity may be due to the agent itself or delivery of the agent to an unintended target (e.g., extravasation). Several antidotes are used in a prophylactic fashion or on chronic basis. Amifostine (WR-2721) -which is dephosphorylated by alkaline phosphatase to an activated, protective thiol form -is approved to decrease toxicity associated with radiotherapy and renal injury associated with cisplatin [294] . It has also been used to reduce chemotherapy-induced neutropenia; genitourinary injury associated with cyclophosphamide; and transfusion requirements, gastrointestinal and hepatic toxicity in pediatric patients [295, 296] . Cyclophosphamide and ifosfamide induce bladder toxicity (hemorrhagic cystitis) via their metabolite acrolein. Mesna (2-mercaptoethane sulfonate), a thiol agent that complexes with and inactivates acrolein, is provided orally or i.v. as prophylaxis [294] . Diethyldithiocarbamate (DDTC), the major metabolite of disulfiram, is an investigational agent for prevention of neuropathy from cisplatin and its analogs; it increased nephrotoxicity in one study [297] . Granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), erythropoietin (hemopoietin) and its derivatives, oprelvekin (recombinant interleukin-11), and other stimulating factors are employed as adjuvants to reconstitute various hematopoietic lines damaged by chemotherapy and radiation [298, 299] . Palifermin (recombinant truncated human keratinocyte growth factor) is used to prevent severe mucositis in patients receiving stem-cell transplantation with a total body irradiation conditioning regimen [294] . The remaining section focuses on antineoplastic antidotes used in the acute setting. A dreaded complication of administration of vesicant chemotherapeutic agents is extravasation. Risk factors for extravasation include small, fragile, or sclerosed veins, obesity, comorbid conditions (diabetes, circulatory disorders, impaired sensory perception), use of rigid i.v. catheters, and clinicians' lack of knowledge and skills [300] . Redness, burning pain, and swelling may portend later blistering, ulceration, and necrosis. Dexrazoxane is U.S. FDA approved for treatment of extravasation resulting from i.v. anthracycline chemotherapy, to diminish tissue damage and the need for surgical excision of necrotic tissue [301] . Clinical efficacy data comes from two simultaneously reported openlabel, single-arm, prospective multicenter studies in which only 1 out of 54 patients with biopsy-proven extravasation required surgical debridement [302] . Additional instances of successful dexrazoxane treatment of anthracycline extravasation are provided as case reports ( [303] and others). Dexrazoxane is provided once daily for 3 consecutive days, with the first infusion initiated as soon as possible. Daily doses are as follows: day 1, 1000 mg/m 2 (maximum 2000 mg); day 2, 1000 mg/m 2 (maximum 2000 mg); day 3, 500 mg/m 2 (maximum 1000 mg) [301] . The dose is reduced by 50% in patients with creatinine clearance of less than 40 mL/min. In mice, efficacy rapidly decreased when dexrazoxane was provided beyond 6 hours after extravasation [304] . Dexrazoxane's mechanism of action appears to involve reversible inhibition of topoisomerase II and inhibition by its metabolite, an ethylenediamintetraacetic acid (EDTA) analogue, of free radical formation via iron removal from the iron-doxorubicin complex [305] . Topoisomerase II-independent effects have also been described [306] . In contrast, some authors have encouraged the nonconcurrent, off-label use of topical dimethyl sulfoxide (DMSO) for anthracycline extravasation because of the risk of infection, neutropenia, and thrombocytopenia associated with dexrazoxane [307] . Dexrazoxane is also used prophylactically to limit anthracycline-associated cardiomyopathy [294] . In 1950, methotrexate (MTX) joined the oncological armamentarium for leukemia [308] . MTX treatment of solid cancers was reported in 1956, and it gained FDA approval for psoriasis in 1971 [309, 310] . MTX is now used intramuscularly, intrathecally, i.v., and orally for a range of dermatological, rheumatological, obstetric, and gynecological conditions. The dose ranges from 7.5-30 mg orally once weekly for psoriasis or rheumatoid arthritis to 8-12 g/m 2 or more for osteosarcoma, leukemia, and lymphoma [311] [312] [313] . MTX poisoning may result from intentional overdose; unintentional ingestion, prescription, dispensing, administration, and patient errors; or renal insufficiency leading to persistent MTX in patients receiving high-dose chemotherapy regimens [314, 315] . MTX antagonizes folate metabolism (and rapidly proliferating cells) via multiple mechanisms. Dihydrofolate reductase inhibition by MTX and its polyglutamated metabolites ensures that neither dihydrofolate nor active tetrahydrofolate can be generated from folate, nor can existing dihydrofolate be recycled. Thymidylate synthase inhibition compromises thymidine synthesis. Purine ring synthesis is impaired by inhibition of the participating enzymes amidophospho-ribosyltransferase (PPAT) and 5-aminoimidazole-4-carboxamide ribonucleotide transformylase (AICART) [316, 317] . Maintenance of brisk urinary elimination with i.v. hydration and urinary alkalinization are standard therapies for patients receiving MTX. MTX is ten times more soluble in alkalinized urine (i.e., pH 7.5) than at pH 5.5 [318] . Folate (folic acid) is an ineffective therapy for MTX poisoning. While folate will inhibit renal resorption of MTX, persistent dihydrofolate reductase inhibition by MTX inhibits folate's activation. Leucovorin (folinic acid, 5-formyltetrahydrofolic acid, citrovorum factor) sustains the folate cycle by bypassing the blocked dihydrofolate reductase pathways. Addition of leucovorin "rescue" permitted the administration of very-high-dose MTX chemotherapy [319] . However, in patients receiving MTX chemotherapy, 24-hour MTX concentrations greater than 1 × 10 -5 M (10 μmol/L), 48-hour concentrations greater than 1 × 10 -6 M (1 μmol/L), or 72-hour concentrations greater than 1 × 10 -7 M (0.1 μmol/L, 100 nM), or those with evidence of renal dysfunction are considered at high risk for toxicity [320] . In the setting of MTX persistence or toxicity, leucovorin i.v. doses are increased to 100 mg/m 2 or 1000 mg/m 2 every 6 hours according to established nomograms; doses and as high as 10 g/day have been used [319, 321] . Leucovorin therapy continues until MTX concentration are less than 0.5 × 10 -7 -1.0 × 10 -7 M (0.05-0.1 μmol/L, 50-100 nM) [319] . However, adequate leucovorin concentrations cannot be achieved for competitive reversal of MTX toxicity when MTX concentrations are persistently above 10-100 μmol/L; other antidotal strategies are then considered [313] . Treatment of patients ingesting MTX should not be delayed pending MTX concentrations. Inhibition of DNA synthesis is nearly complete when MTX plasma concentrations are greater than 1 × 10 -8 M (0.01 μmol/L, 10 nmol/L) [322] . Therefore, leucovorin is provided until MTX concentrations are less than 1 × 10 -8 M in patients receiving MTX for non-oncological indications or in patients not receiving MTX therapeutically [311] . Only leucovorin's S-form [levoleucovorin, (6S)-leucovorin] is active and rapidly metabolized to usable, reduced folates; the inactive isomer is slowly eliminated by renal excretion during i.v. administration [323] . Leucovorin was available in the U.S. only as a racemate until 2008, when levoleucovorin received FDA approval. Levoleucovorin at one-half of the usual racemic dose (as it is entirely active) appears to provide similar rescue therapy in high-dose MTX chemotherapy [324] . Oral rescue is not routinely recommended as leucovorin's bioavailability is poor above 40 mg due to saturation of active intestinal transport [323] . The calcium content of leucovorin (0.004 mEq calcium/mg leucovorin) mandates that infusion should not exceed 160 mg/min. Intrathecal administration of leucovorin is contraindicated, as death may result [325] . Glucarpidase (carboxypeptidase G 2 , CPDG 2 ) is undergoing evaluation as an additional antidote for MTX toxicity. U.S. or European marketing approval for glucarpidase has not been granted at the time of writing. Competitive and complete reversal of MTX toxicity by leucovorin may not be possible at MTX concentrations above 100 μmol/L (and perhaps even lower) [313, 326, 327] . Patients with systemic MTX toxicity (significant mucositis, gastrointestinal distress, myelosuppression, hepatitis, or neurotoxicity), persistent serum MTX, and renal impairment following high-dose MTX have been considered for glucarpidase therapy in addition to leucovorin. Recommendations for glucarpidase above certain MTX concentrations have varied by malignancy, degree of renal impairment, initial MTX dose, and serum MTX concentration (e.g., Clinical Trials NCT00424645, NCT00481559, and [313, [328] [329] [330] ). Purification of "carboxypeptidase G", a pseudomonad zinc-dependent enzyme capable of MTX cleavage, was reported in 1967 [331] . Its antidotal potential was suggested in 1972. In mice injected with lethal MTX doses, carboxypeptidase G 1 rapidly decreased MTX concentrations and improved survival [332] . CPDG 1 selectively eliminated systemic MTX in patients treated with high dosages targeting CNS malignancy, and rescued a patient receiving MTX with renal failure in 1978 [333, 334] . After the original enzyme source of CPDG 1 was lost, a revived recombinant CPDG 2 product demonstrated success in both i.v. and intrathecal rescue of MTX overdose in non-human primates [335] [336] [337] . Successful use in multiple case reports and human trials in adult and pediatric patients with i.v. and intrathecal MTX overdose emerged [313, 315, 328, 329, [337] [338] [339] . Glucarpidase is a dimerized protein with two domains -a zinc-dependent catalytic domain that removes C-terminal glutamate residues of folate and folate analogues and a β-sheet interaction site [340] . Glucarpidase splits MTX and its 7-hydroxy-MTX metabolite into inactive 4-{[2,4-diamino-6-(pteridinyl)methyl]-methylamino}-benzoic acid (DAMPA) and hydroxy-DAMPA plus glutamate [341, 342] . MTX serum concentrations decline by 71-99% within minutes after glucarpidase [313, 315, 326, 330, 343, 344] . Intracellular, intraluminal (gastrointestinal tract) and intracerebral MTX is unaffected, creating the potential for rebound concentrations and persistent cytotoxicity [317, 328, 332, [345] [346] [347] . Leucovorin therapy must continue after carboxypeptidase administration. DAMPA's poor urinary solubility also requires ongoing alkalinization and saline diuresis to prevent renal precipitation [315, 348] . Anti-glucarpidase antibodies have been detected in patients receiving glucarpidase, although patients have been successfully treated with additional doses of glucarpidase for persistently elevated MTX concentrations [313, 326, 328, 337, 339, 342, 345] . HPLC must be used to determine actual MTX concentrations after glucarpidase as both MTX metabolites, 7-hydroxy-MTX and DAMPA, interfere with immunoassay techniques [349] . Glucarpidase has an affinity for MTX approximately 10-to 15-fold higher than it does for leucovorin; however, its affinity for folate and 5-methyltetrahydrofolate are similar [350, 351] . Glucarpidase eliminates active levo-(6S)-leucovorin about 50% faster than nonphysiological dextro-(6R)-leucovorin [348] . A study to address the clinical consequence is ongoing. Because of the stereoselective destruction of active leucovorin and its metabolites, many protocols attempt to separate leucovorin administration from glucarpidase administration by 2-4 hours. Administration of glucarpidase more proximate to leucovorin administration, and which antidote to provide should glucarpidase become available at a leucovorin dosing interval, requires a thoughtful benefit-risk assessment. Countryspecific information on obtaining glucarpidase, institutional review board protocol, and consent issues have been made available online (www.btgplc.com/ BTGPipeline/273/Voraxaze.html; and www.fda.gov/cder/cancer/singleIND. htm). Cardiovascular pharmaceuticals comprise a wide variety of agents including anti-dysrhythmics, β-adrenergic antagonists (β-blockers, BBs), angiotensin antagonists, calcium channel antagonists (CCBs), cardioactive glycosides, and imidazoline derivatives. Overdose of these agents alone or in combination can generate potentially lethal combinations of impaired conduction, dysrhythmia, vasodilatation, and negative inotropy. Management of severe cases may necessitate diagnostic adjuncts such as echocardiography and right heart catheterization (Swan-Ganz measurements). In cases refractory to routine supportive care, vigorous gastrointestinal decontamination, and pharmacological intervention, aggressive measures including cardiac pacing, intra-aortic balloon counter-pulsation, or extracorporeal circulation (cardiopulmonary bypass) may be required until toxin elimination can be achieved [352] . Cardiac pacing may improve heart rate without increasing cardiac output if inotropy is compromised. Use of naloxone in the management of overdose of clonidine and angiotensin receptor antagonists and angiotensin converting enzyme inhibitors is provided in the opioid antagonists section. Strategies to mitigate the anticoagulant toxicity of vitamin K antagonists (i.e., coumadin) including exogenous oral or i.v. vitamin K, fresh frozen plasma, prothrombin concentrates, and recombinant factor VII are detailed in the 2008 American College of Chest Physicians Evidence-Based Clinical Practice Guidelines [353] . The Guidelines also address protamine sulfate for reversal of heparin anticoagulation and use of nonheparin anticoagulants for treatment and prevention of heparininduced thrombocytopenia [354, 355] . Atropine (D,L-hyoscyamine) is familiar to clinicians due to its use in several advanced cardiac life support (ACLS) algorithms [356] . Atropine is a centralacting, competitive antagonist of muscarinic acetylcholine receptors (M 1 -M 5 ) [357] . It is used to counteract bradycardia from BBs, CCBs, cardioactive glycosides, and clonidine. Atropine increases basal heart rate; it does not affect the basal force of contraction [357] . Positive chronotropy alone may not produce systemic benefit in severe poisoning, and conduction system poisoning may limit responsiveness to atropine [358] . For symptomatic bradycardia, atropine 0.5-1.0 mg (pediatric dose: 0.02 mg/kg) i.v. is provided every 2-3 min to a maximum dose of 3 mg. Paradoxical parasympathetic response may occur during slow infusions or doses less then 0.5 mg (0.1 mg minimum in children) [356] . In slowing gastrointestinal motility, atropine may impair decontamination with WBI or AC. CCBs antagonize L-type calcium channels, slowing entry of calcium ions during myocyte depolarization; however, intracellular calcium release is not directly affected. This disrupts calcium-mediated excitation-contraction coupling, action potential generation and conduction, and vascular smooth muscle tone [359] . Exogenous i.v. calcium is indicated in cases of CCB and BB toxi-city [352] . In animal models, calcium salts reverse CCB-induced deficits in contractility, blood pressure, and cardiac output [352] . Multiple uncontrolled cases reports document the effectiveness of calcium salts; however, interpretation of effectiveness is complicated by the co-administration of other modalities. Some authors advocate aggressive high-dose calcium therapy, providing large amounts of calcium without apparent ill effect [358] . This approach does carry a risk of death from hypercalcemia [reported concentration, 32.3 mg/dL (8.07 mmol/L) after 38 g calcium] [360] . Others recommend a bolus dose followed by continuous infusion to maintain physiological calcium levels [361] . Peripheral administration as calcium gluconate decreases the risk of extravasation and tissue necrosis. A standard container of 10 mL of 10% calcium gluconate provide 4.65 mEq (93 mg) elemental Ca 2+ ; 10 mL of 10% calcium chloride (1 g total CaCl 2 ) yields 13.6 mEq elemental Ca 2+ [362] . A suggested approach is to initially administer a 0.6 mL/kg (0.28 mEq/kg) bolus of 10% calcium gluconate (0.2 mL/kg 10% CaCl 2 ) over 5-10 min [359, 361] . Empirically, this is roughly one vial (1 g) of 10% CaCl 2 or three vials (3 g) of 10% calcium gluconate i.v. The bolus may be repeated several times. Due to bolus dissipation, most patients are placed on an infusion of 10% calcium gluconate at 0.6-1.5 mL/kg per hour (0.28-0.7 mEq/kg per hour) or 0.2-0.5 mL/kg per hour [359, 361] . Serum phosphate, calcium, and hydration status should be closely monitored. Calcium administration for hyperkalemia has been generally contraindicated in cases of cardioactive glycoside toxicity, out of concern for dysrhythmias or systolic arrest (also known as "stone heart") [363] . While more recent studies have challenged this assertion, it is advisable to withhold calcium until the definitive cardiac glycoside antidote, digoxinspecific Fab fragments, has been provided [364] . Digoxin and cardioactive glycosides inhibit the cardiac sodium-potassium ATPase. The subsequent accumulation of sodium in the cytoplasm dissipates the driving force for calcium expulsion via the sodium-calcium exchanger. Increased intracellular calcium enhances actin-myosin coupling, myocyte contraction, and inotropy. In overdose, the excess calcium may result in membrane hyperexcitability and delayed after-depolarizations. Increased vagal tone decreases conduction through the AV node. The combination of increased automaticity and vagotonicity may yield lethal ventricular escape rhythms. Digoxin-specific antibody fragments bind free digoxin in serum to decrease digoxin serum concentrations to undetectable levels within minutes [365] . Successful reversal of digoxin toxicity with digoxin-specific Fab was first reported in 1976 [366] . The results of a prospective multicenter study demonstrated significant effectiveness in reversing life-threatening digitalis toxicity, and more recent studies confirm ongoing Fab fragment utility [367, 368] . Digoxin-specific Fab were also shown to be effective in children [369] . Digoxin-specific Fab are produced from purified ovine-derived immunoglobulin G. Cleaving the Fc antibody portion significantly improves renal excretion of the complex, decreases immunogenicity, and facilitates diffusion of remaining free Fab into tissue [370] . Reflecting digoxin redistribution from target organs of toxicity, the initial response to digoxin-specific Fab was 19 min (0-60 min), and complete reversal of systemic toxicity occurred on average by 88 min (30-360 min) [367] . Indications for therapy include life-threatening or progressive dysrhythmia or shock; potassium greater than 5.0 mEq/L (acute poisoning); chronic poisoning with other end-organ manifestations such as altered mental status, significant gastrointestinal symptoms or renal impairment; or serum digoxin concentration >15 ng/mL or greater than 10 ng/mL beyond 6 hours after ingestion. Hyperkalemia is rapidly reversed by digoxin-specific Fab [365] . One vial neutralizes approximately 0.5 mg of digoxin (or digitoxin). Dosing is based either on amount ingested [number of vials = amount ingested (in mg) × 0.8 (oral bioavailability) / 0.5], or a serum concentration [number of vials = serum digoxin concentration (ng/mL) × patient weight (kg) / 100]. The number of vials is rounded up and administered i.v. over 30 min. Empiric therapy is 10-20 vials for adult or pediatric patients in acute poisoning or 3-6 vials (1-2 vials in children) in chronic poisoning. Partial reversal is recommended by some authors [371] , but is not common U.S. practice due in part to concern for recrudescent toxicity with inadequate therapy [370] . Following treatment, free digoxin concentrations may rebound upwards within 12-24 hours, most likely reflecting tissue redistribution into the vascular space [372] . This provides a measure of protection against development of significant congestive heart failure (CHF) in patients dependent upon digoxin for inotropy, although exacerbation of CHF may occur [370] . Clinically significant late rebound of digoxin concentrations and toxicity have occurred in patients with marked renal dysfunction [373] . Immunogenicity from repeat digoxin-specific Fab has generally not been significant, although allergic reactions have been infrequently reported with administration [374] . Digoxin-specific Fab has been used clinically or experimentally to treat poisoning by other cardiac glycosides -yellow oleander (Thevetia peruviana), Nerium oleander, Chan Su and "Love Stone" (extract of the Bufo bufo gargarizans toad) [375, 376] . Higher dosing may be required due to poor binding affinity. BBs competitively antagonize catecholamine effects at cardiac β-receptors, leading to decreased inotropy and slowed conduction through the AV node. Bradycardia, conduction delay, hypotension, and decreased cardiac output may accompany significant poisoning. BB interference with gluconeogenesis and glycogenolysis may lead to hypoglycemia, as well as blunt the catecholamine response that is important in its recognition. Glucagon, a 29-amino acid peptide hormone secreted by pancreatic α-cells, counteracts hypoglycemia and the actions of insulin; regulates gastrointestinal motility; and mediates the rate of renal filtration, urea excretion, and water resorption [377] . The current glucagon product is now produced in non-pathogenic E. coli by recombinant techniques [378] . Myocardial binding occurs at a distinct glucagon receptor (GCGR) coupled with the β-agonist binding site. Antidotal (off-label) use of glucagon thus bypasses β-receptor blockade to directly induce G-protein-mediated stimulation of adenylate cyclase to convert ATP to cAMP [379] . cAMP, in turn, activates protein kinase A (PKA), which promotes the phosphorylation and opening of dormant L-type calcium channels to improve calcium-dependent excitation-contraction coupling [361] . Another proposed mechanism is C-terminal cleavage of glucagon to miniglucagon, which has a direct effect on sarcoplasmic reticulum calcium stores via arachidonic acid [380] . In human volunteers evaluated by cardiac catheterization, glucagon increased heart rate, cardiac index, and mean atrial pressure, but not left ventricular end-diastolic pressure (EDP) or systemic vascular resistance (SVR) [381] . Clinical experience in overdose consists primarily of case reports [382, 383] . Due to the complex nature of overdose, glucagon is often used in combination with other agents in severe BB overdose. Additionally, several ex vivo experiments, controlled animal studies, and uncontrolled case reports have demonstrated that glucagon can be beneficial in CCB exposure [384] [385] [386] . The recommended initial bolus dose of glucagon is 50-150 μg/kg, which may be repeated after 3-5 min [359] . A continuous infusion corresponding to the total effective bolus reversal dose is then provided per hour (e.g., if clinical response was observed following administration of 2 mg, 3 mg, and finally 5 mg, the hourly infusion would be 10 mg/hour). The effects of glucagon administered i.v. begin within 1-3 min, peak at 5-7 min and last for approximately 15 min [381] . Nausea and vomiting are common and should be anticipated. This may complicate management of patients with depressed mental status or airway concerns. Flushing, transient hyperglycemia, and smooth muscle relaxation, and ileus may also occur. Since CCBs antagonize the L-type calcium channel in pancreatic islet cells, a subsequent decreased insulin production can produce hyperglycemia [361] . Animals poisoned by CCBs have impaired myocardial fatty acid uptake (leaving them dependent upon carbohydrate metabolism), impaired uptake of glucose, and myocardial insulin resistance [387, 388] . In humans, intracoronary verapamil increased glucose release and altered myocardial lactate use from consumption to release [389] . Decades ago, glucose-insulin-potassium (GIK) was proposed as adjuvant therapy for acute myocardial infarction, with the intent of suppressing uptake of free fatty acids, improve myocardial energy production, and stabilize intracellular potassium [390] . Randomized trials of GIK therapy in patients with acute myocardial infarction (AMI) have not shown benefit, although the insulin doses tend to be low (in general, ≤0.075 U/kg) [390] . Experience in the surgical literature in cases where much higher insulin doses have been used has been somewhat different [391] . Patients undergoing aortic valve replacement and coronary artery bypass who received high-dose insulin at 1 unit/kg per hour demonstrated more rapid lactate clearance, lower glucose, lower dobutamine requirements, a trend for improved cardiac indices, and potential anti-inflammatory benefit (lower C-reactive protein and free fatty acid levels) [392] . Insulin doses of 2.5 units/kg were tolerated without excess increase of insulin-induced potassium elimination [393] . In combination with dopamine, insulin 7 units/kg was used to significantly augment cardiac output and decrease systemic vascular resistance in post-coronary artery bypass graft (CABG) patients without generating excess in oxygen demand [394] . Additional benefits of high-dose insulin included overcoming insulin resistance, increased expression of glucose transporters, and improved turnover of sodium-potassium-ATPases [391] . The basis for high-dose insulin euglycemia therapy (HIET) (off-label) in overdose has been explored in a series of animal models of CCB and BB toxicity [387, 388, [395] [396] [397] . HIET increased myocardial lactate uptake and improved systolic and diastolic heart function. Insulin outperformed epinephrine and glucagon [395] [396] [397] . Multiple human cases of successful management of CCB overdose with HIET have been described [359, 398] . Because the beneficial cardiovascular effects of HIET are not seen for 15-60 min after initiation, it must be considered early, before patients become unsalvageable [359] . A proposed dosing scheme includes a bolus dose of regular insulin of 1.0 units/kg, followed by an infusion of 0.5-1.0 units/kg per hour, titrated upwards as necessary [359] . A dextrose bolus is also provided unless significant hyperglycemia exists, followed by an infusion of 0.5-1.0 g/kg per hour to maintain blood glucose between 100 and 250 mg/dL. Persistent physician reticence to utilizing the high-dose insulin out of concern for excess hypoglycemia presents an obstacle for implementation of adequate HIET [399] . This ignores a body of physiological data that demonstrate that the insulin transport follows saturation kinetics [400, 401] . Alternatively, it has also been demonstrated that insulin-stimulated glucose clearance reaches a maximum in both lean and obese subjects [402] . Taken together, this suggests that, from a therapeutic standpoint, since insulin effect via insulin receptors appears saturable, additional mechanisms must be at work. The effects of HIET may include counteracting CCB-mediated insulin impairment or shockinduced hyperglycemia, improving myocardial substrate utilization, and improving myocardial metabolism [359] . From an adverse-effects standpoint, once adequate and ongoing glucose has been provided, hypoglycemia should not present an excessive risk [398] , although frequent serum glucose and potassium evaluation are obvious components of HIET therapy. Due to the high dosing, insulin may persist after the infusion cessation and necessitate ongoing supplemental dextrose beyond insulin infusion. As hypokalemia is an intracellular result of shift, it is supplemented cautiously. During administration of local anesthetics, severe toxicity may result from systemic absorption or unintended intravascular administration. Loss of consciousness, dysrhythmia, cardiovascular collapse, seizures, and lactate-associated acidemia may rapidly ensue [403] . Furthermore, in animal models, for some of the local anesthetics (bupivacaine, levobupivacaine, and ropivacaine), treatment with "standard" advanced cardiac life support (ACLS) drugs such as epinephrine may precipitate ventricular dysrhythmia [404] . Following a serendipitous observation that pretreatment with a lipid emulsion altered the dose-response to bupivacaine-induced asystole, murine and canine studies provided evidence of survival benefit with lipids in bupivacaine toxicity [405, 406] . Case reports of successful resuscitation of patients severely affected by bupivacaine, levobupivacaine, mepivacaine, prilocaine and ropivacaine (alone or in combination) followed [403, [407] [408] [409] . Pediatric experience is limited to a case of successful resuscitation following lidocaine/ropivacaine toxicity from a posterior lumbar plexus block [410] . Lipid therapy has been successfully applied in human bupropion toxicity and combined quetiapine and sertraline overdose [411, 412] . Animal models have suggested a possible benefit in clomipramine, propranolol, thiopentone, and verapamil poisoning [413] [414] [415] [416] . An understanding of lipid's mechanism of action is incomplete. It may act as a "circulating lipid sink" in which excess lipophilic drug may dissolve; modulate intracellular processes; or provide an alternative myocardial energy supply [411] . Presumably due to central sympathetic activation, human volunteers given a 4-hour lipid emulsion (20%) infusion had increased systemic vascular resistance, blood pressure, muscle sympathetic nerve activity, and concentrations of insulin and aldosterone, without increased cardiac output [417] . Lipid emulsion increased inotropy in both spontaneously beating and paced isolated rat hearts poisoned with levobupivacaine [418] . Dosing guidelines for the off-label use of lipid emulsion in resuscitation are provisional, as optimal bolus and continuous infusion therapy and timing are still being explored. The Association of Anaesthetists of Great Britain and Ireland recommends an i.v. bolus of 1.5 mL/kg Intralipid ® (20%) over 1 min, which may be repeated twice at 5-min intervals if an adequate circulation has not been restored [419] . Following the initial bolus, an infusion is commenced at 0.25 mL/kg per min (which may be increased to 0.5 mL/kg per min in inadequate circulation). Propofol is an inadequate substitute [419, 420] . Ongoing lipid therapy may be required as recrudescence may occur [421] . Hyperamylasemia may be anticipated. Additional concerns include pancreatitis, allergic reactions, acute myocardial infarction, fat embolism, and altered coagulation [420] . In lapine and porcine models of asphyxial cardiac collapse (pulseless electrical activity or arrest), lipid emulsion was markedly ineffective [422, 423] . In vitro, lipid affinity for both bupivacaine and ropivacaine is also adversely affected by low pH (by a factor of 1.68 in a pH drop from 7.40 to 7.00) [424] . These data suggest that ventilatory status must be aggressively addressed early in toxicity. Due to their physicochemical characteristics and structure, many non-antiarrhythmic drugs are able to antagonize or alter expression of the myocardial potassium delayed rectifier channel (hERG, KCNH2, LQT2). With channel block, potassium efflux is compromised, and the repolarizing cardiac I Kr current is impaired. The surface ECG reflects this as QT prolongation. Age, female gender, comorbidities such as structural heart disease, electrolyte disturbances such as hypokalemia, and heart rate (bradycardia) may provide additional risk. Certain antibiotics, antihistamines, antipsychotics, antidepressants, and methadone are prone to induce QT prolongation. QT prolongation is associated with torsade de pointes, a polymorphic ventricular arrhythmia that can degenerate into ventricular fibrillation, cardiac arrest and sudden death [425] . If significant QT prolongation (QTc >500 ms) is detected, administration of 1-2 g magnesium sulfate i.v. (pediatric dose, 25-50 mg/kg) over 5 to 60 min (depending on urgency of presentation), followed by an infusion of 2-4 mg/min is suggested [426] . Rapid infusion may cause hypotension, and magnesium should be administered cautiously in renal failure. A second bolus can be provided 5-15 min later [427] . Magnesium sulfate i.v. is effective in arrhythmias occurring due to early or delayed depolarization-induced triggered activity [427] . Acceleration of the heart rate with isoproterenol or transvenous pacing (overdrive pacing) may be needed to preclude recurrence of torsade de pointes while correction of underlying risk factors (hypokalemia and hypocalcemia) ensues. Immediate non-synchronized defibrillation is required for unstable polymorphic ventricular tachycardia or ventricular fibrillation. Severe cardiovascular toxicity may result from blockade of cardiac sodium channels by tricyclic antidepressants (TCAs) -leading to conduction delays, dysrhythmias, and myocardial depression. TCAs adversely affect maximum upstroke velocity (V max ), which approximates the magnitude of sodium entry [428] . The sodium channel blockade displays rate dependence. At slow rates the TCA has time to disassociate, allowing channel recovery. At faster rates, block progressively worsens. Given the anticholinergic effects of TCAs that speed the heart rate, this is a significant concern. However, attempts to decrease heart rate with propranolol produced hypotension and lethality in canine studies [429, 430] . With progressive sodium channel block, ventricular impulse propagation becomes delayed. Sodium channel blockade manifests on the surface ECG as QRS widening. A QRS equal or greater than 100 ms is a significant predictor of seizure; a QRS ≥160 ms predicts ventricular dysrhythmia [431] . The right bundle branch has a relatively longer refractory period, and it is affected disproportionately by impaired intraventricular conduction delay. Rightward terminal axis shift or outright bundle branch block may be present [432] . These rightward terminal forces may also produce terminal R waves in leftwarddirected leads [433] . Acidemia secondary to hypoperfusion or seizure may generate progressively worsened block. In an acidemic environment, free TCA concentrations increase as binding to α-1 acid glycoprotein decreases, the TCA ionized fraction increases, and sodium channel blockade worsens [434] . Seizures are severe and consequential, leading to QRS widening and hypotension [435] . Administration of sodium bicarbonate improves V max and action potential amplitude by increasing extracellular pH and sodium concentration [428] . Consequentially, compromised myocardial inotropy, conduction aberrations, and dysrhythmia are reversed. Several animal studies have demonstrated these beneficial effects [429, 430] . Both the sodium and alkalemia induced by sodium bicarbonate improve cardiac performance [429] . The enhanced inotropy with sodium bicarbonate is independent of and additive to vasopressor treatment [436] . Hyperventilation-induced alkalinization similarly narrows the QRS [437] . Sodium bicarbonate outperformed hyperventilation in a swine model, although hypertonic saline was superior to both [438] . This approach has been reported clinically [439] . While sodium bicarbonate is recommended for QRS widening in TCA evidence-based consensus guidelines for out-ofhospital management, actual human studies are not as extensive as one might suspect [440, 441] . Initially, hypertonic sodium bicarbonate 1-2 mEq/kg i.v. is provided, preferably with continued ECG monitoring of the QRS. Institutions usually stock either an 8.4% solution (1 mEq/mL sodium and bicarbonate ions) or a 7.5% solution (0.892 mEq/mL sodium and bicarbonate ions). Rarely, a 5% solution may be encountered (0.595 mEq/mL). A "standard" 50-mL ampule of 8.4% or 7.5% solutions would deliver 50 or 44.6 mEq of NaHCO 3 . Several boluses may be required, either initially or as the bolus effect declines due to redistribution [429] . Ongoing alkalinization should be provided as discussed previously, with a goal of serum pH 7.55. If sodium bicarbonate administration is problematic due to fluid load, hyperventilation and/or hypertonic saline may be required [437, 439] . Due to mechanistic similarities, sodium bicarbonate has been recommended for QRS widening seen in poisoning by Vaughn-Williams Class IA and IC antidysrhythmics, cocaine, diphenhydramine, carbamazepine, and propoxy-phene [442] [443] [444] [445] . Treatment of bupropion-induced QRS widening with sodium bicarbonate has met with both success and failure [446] . Sodium bicarbonate has also been suggested to treat QRS widening from venlafaxine; similar effects seen with lamotrigine might also be amenable [447, 448] . Sodium bicarbonate therapy may have a role in Taxus species (yew berry) toxicity [449] . Treatment of amantadine-induced QRS widening with sodium bicarbonate may be complicated by concurrent profound hypokalemia [450] . Naloxone is a competitive opioid antagonist at all receptor subtypes [451] . It can prevent or reverse the effects of opioids, notably CNS and respiratory depression. Massive doses of naloxone (5.4 mg/kg with 4.0 mg/kg per hour infusion) have been administered safely in non-opioid tolerant individuals suffering from spinal cord injury [452] . However, indiscriminate use of naloxone in opioid-tolerant individuals can precipitate acute opioid withdrawal, with attendant acute lung injury, seizure, hypertension, or cardiac dysrhythmia [453] . These are likely associated with the abrupt, significant, and sustained increases in plasma catecholamine concentrations (epinephrine and norepinephrine) that accompany narcotic reversal, particularly in the setting of hypercapnia [454] . Withdrawal-induced vomiting may compromise the airway in patients with concomitant sedative-hypnotic ingestion. Precipitated withdrawal-associated agitation and violent behavior may require chemical restraint, leading to a vicious cycle of compromised CNS and cardiopulmonary function as naloxone wears off. Self-release and relapse following naloxone administration is also a concern in opioids with prolonged duration of effect (methadone, controlled-release oxycodone hydrochloride, etc.). Naloxone is no longer recommended as the initial resuscitation of newborns with respiratory depression in the delivery room; precipitation of acute neonatal opioid withdrawal may produce severe consequences [455] . Sudden cardiac arrest has occurred in preterm neonates given naloxone to reverse opioid overdose [456] . Naloxone is utilized in those individuals with clear evidence of the opioid toxidrome. Those with a respiratory rate ≤12 or hypopnea are likely to benefit [457] . The goal of therapy is titration to adequate ventilatory status without withdrawal. After normocapnia is achieved by supported ventilation, this can be done with i.v. administration of 0.04-0.05 mg initially (e.g., 1 mL of 0.4 mg naloxone in 10 mL diluent or 1 mg naloxone in 20 mL diluent). Due to rapid onset, effectiveness can be assessed, and if required, the dose can be titrated upwards incrementally to 0.4 mg, 2 mg, or even 10 mg. Patients without response to 10 mg naloxone are unlikely to have opioid-induced respiratory depression. Nonopioid-dependent adults are administered 0.4-2 mg i.v. Pediatric dosing for infants and children from birth to 5 years of age or less than 20 kg body weight is 0.1 mg/kg; children older than 5 years of age or weighing more than 20 kg are provided 2 mg [455] . For longer acting opioids, following adequate initial opioid antagonism, two-thirds of the initial naloxone reversal bolus is provided as a continuous i.v. infusion [458] . Naloxone can successfully antagonize buprenorphine overdose in children, although prolonged therapy and monitoring may be required [459] . Higher doses may be required due to reverse buprenorphine effects because of its high affinity for opioid receptors [460] . Naloxone has also been used to reverse clonidine toxicity, although this is not always the case [461] . It has been postulated that patients with higher hyperadrenergic tone (who have higher concentrations of endogenous opioids) or those in whom clonidine induces more endogenous opioid release may respond best to naloxone [462] . Mental status, blood pressure, and heart rate may respond differently. Naloxone has been employed in angiotensin converting enzyme inhibitor overdose. One author reported that a 1.6 mg bolus of naloxone followed by repeat 2 mg bolus reversed hypotension due to overdose with 500 mg captopril [463] . Naloxone has been ineffective in reversing hypotension in other cases complicated by co-ingestants [464] . The mechanism may relate to antagonism of endogenous opioids [465] . Co-administration of 0.2 mg/kg naloxone mitigated captopril-related decreases in systolic and diastolic blood pressure in healthy volunteers [465] . A placebo-controlled study of healthy men found that naloxone pretreatment with 10 mg followed by 2.46 mg/hour infusion precluded systolic blood pressure decrease induced by captopril (50 mg) [466] . Under different experimental conditions [naloxone, 0.4 mg bolus and a 2-hour continuous infusion (4.0 mg/hour), and captopril (25 mg)], no difference was observed [467] . Analogous to naloxone antagonism at opioid receptors, flumazenil competitively antagonizes benzodiazepine receptors -allosteric sites located at the macromolecular GABA A receptor complex, which regulate chloride ion flux within the associated ion channel [468] . Flumazenil reverses the sedative, psychomotor, and amnestic effects of benzodiazepines [469] . Flumazenil's effectiveness depends upon the number of benzodiazepine receptors that can be occupied according to the mass-action law, the affinity of a particular benzodiazepine for the receptor, and the free benzodiazepine concentration near the receptor [470] . In contrast, antagonism of benzodiazepine-induced respiratory depression is inconsistent, and acute tolerance may develop to large doses [471] [472] [473] . Flumazenil administration can reverse bispectral index (BIS) depression and permit earlier emergence from anesthesia in patients provided non-benzodiazepine anesthesia (propofol/remifentanil) [474] . Postulated mechanisms included intrinsic CNS stimulant activity or antagonism of endogenous benzodiazepine-like ligands (endozepines). Under certain experimental conditions, flumazenil may also demonstrate partial agonist or even inverse agonist activity [475, 476] . The appropriate utilization of flumazenil as an antidote in patients with benzodiazepine overdose is still a matter of debate. Patients who ingest benzodiazepines alone or in combination generally have acceptable outcomes with supportive care alone. Proponents argue that awakening is therapeutic and diagnostic, obviates requirements for investigatory procedures, and limits complications of sedation. Opponents point to the low risk of mortality with benzodiazepine ingestion, frequent co-ingestants for which flumazenil is ineffective or contraindicated, relapse, and risks of reversal. While flumazenil can be administered safely, indiscriminate flumazenil administration may produce an acute withdrawal syndrome in benzodiazepine-dependent patients, seizures, dysrhythmias, vomiting, and agitation [477] [478] [479] [480] . Flumazenil is not recommended in cases complicated by co-ingestants capable of inducing seizures or dysrhythmias (e.g., bupropion, carbamazepine, chloral hydrate, chlorinated hydrocarbons, chloroquine, cocaine, cyclic antidepressants, cyclosporine, isoniazid, lithium, methylxanthines, monoamine oxidase inhibitors, phenothiazines, and propoxyphene) [477, 479, 481] . As might be anticipated with an antidote of lesser half-life than many benzodiazepines, clinical condition may deteriorate following initial improvement, mandating ongoing monitoring. In one study, patients with primarily benzodiazepines ingestion remained awake for 72 ± 37 min following flumazenil; this was markedly decreased to 18 ± 7 min with co-ingestants [478] . This may be problematic in patients who, once aroused, demand release from medical care. After excluding co-ingestants of concern, vital sign abnormalities, and an aberrant ECG, and considering the risk-benefit ratio, flumazenil is administered slowly i.v., titrated to clinical effect (0.1 mg/min, max ≤1 mg) [481] . Offlabel continuous infusions of 0.3-0.5 mg/hour have been provided to preclude relapse. Patients poisoned by pharmaceuticals present many challenges to the treating clinicians. They generally benefit from aggressive support of vital functions, a careful history and physical examination, specific laboratory analyses, and a thoughtful consideration of the risks and benefits of decontamination and enhanced elimination. Data on the effectiveness of certain antidotes ranges from isolated case reports to robust clinical trials. Clinicians are encouraged to liberally utilize consultation with regional poison centers or those with toxicology training to assist with diagnosis, management, and administration of antidotes, particularly in unfamiliar cases. No outside funding or support was received. The author has no financial interest in any products mentioned or the companies that produce them. Use of trade names is for identification purposes only and does not constitute endorsement by the author, the NYU School of Medicine, the New York City Poison Control Center, or the New York City Department of Health and Mental Hygiene. Within the medical literature, pharmaceuticals, pharmaceutical combinations, and other products are used offlabel as antidotal therapies; off-label uses are referred to in this review. This is for discussion purposes only and does not constitute endorsement of off-label use by the author, the NYU School of Medicine, the New York City Poison Control Center, or the New York City Department of Health and Mental Hygiene. As medicine is an ever-changing science, readers are encouraged to confirm the information contained in this review -by consulting product and safety information sheets, regional Poison Centers, those with toxicological expertise, and other resources -particularly in the case of new or infrequently used drugs. Analeptic use in clinical toxicology: A historical appraisal Treatment of choice in barbiturate poisoning; series of twenty-nine cases of barbiturate poisoning treated with pentylenetetrazole (metrazol) and supportive therapy A study of the anti-barbiturate effects of bemegride Respiratory stimulant effects of ethamivan and picrotoxin The case against the use of respiratory stimulants A double blind comparison of doxapram, ethamivan and methylphenidate NAGD regimen for the coma of drug-related overdose Physostigmine in coma due to drug overdose Physostigmine for γ-hydroxybutyrate coma: Inefficacy, adverse events, and review The poisoned patient with altered consciousness. Controversies in the use of a 'coma cocktail Anonymous (1915) Practical pharmacology. XXIV Management of oil of citronella poisoning Emetics, cathartics, and gastric lavage Fullers earth; its absorptive power, and its antidotal value for alkaloids Management of acute childhood poisoning Therapeutic trends in the treatment of barbiturate poisoning. The Scandinavian method Patients with acute poisoning seen in a general medical unit (1960-71) Antagonism of imipramine poisoning by anticonvulsants in the rat Phenytoin prophylaxis of cardiotoxicity in experimental amitriptyline poisoning Wernicke's encephalopathy: New clinical settings and recent advances in diagnosis and management Acetaminophen and salicylate serum levels in patients with suicidal ingestion or altered mental status Value of rapid screening for acetaminophen in all patients with intentional drug overdose A randomized clinical trial of activated charcoal for the routine management of oral drug overdose How feasible is it to conform to the European guidelines on administration of activated charcoal within one hour of an overdose? Position statement: Ipecac syrup Gastric emptying procedures in the self-poisoned patient: Are we forcing gastric content beyond the pylorus? Residual gastric content after gastric lavage and ipecacuanhainduced emesis in self-poisoned patients: An endoscopic study Efficacy of ipecac during the first hour after drug ingestion in human volunteers Gastric decontamination performed 5 min after the ingestion of temazepam, verapamil and moclobemide: Charcoal is superior to lavage Gastric lavage for liquid poisons Efficacy of ipecac-induced emesis, orogastric lavage, and activated charcoal for acute drug overdose Position paper: Gastric lavage Efficacy of superactivated charcoal administered late (3 hours) after acetaminophen overdose Activated charcoal alone or after gastric lavage: A simulated large paracetamol intoxication Out-of-hospital administration of activated charcoal by emergency medical services Prolonged gastric emptying half-time and gastric hypomotility after drug overdose Effect of anticholinergic drugs on the efficacy of activated charcoal Efficacy of gastric emptying: Gastric lavage versus emesis induced with ipecac Superiority of activated charcoal alone compared with ipecac and activated charcoal in the treatment of acute toxic ingestions Management of acutely poisoned patients without gastric emptying Assessment of the efficacy of activated charcoal following gastric lavage in acute drug emergencies Gastric emptying in acute overdose: A prospective randomised controlled trial Comparison of three methods of gut decontamination in tricyclic antidepressant overdose Whole-bowel irrigation versus activated charcoal in sorbitol for the ingestion of modified-release pharmaceuticals Multiple-dose charcoal and whole-bowel irrigation do Drugs and pharmaceuticals: management of intoxication and antidotes 443 not increase clearance of absorbed salicylate Ipecacuanha: The South American vomiting root Toxicology of ipecac: A review Effectiveness of 15-mL versus 30-mL doses of syrup of ipecac in children Urinary excretion of ipecac alkaloids in human volunteers Position paper: Ipecac syrup Evaluation of the time frame for home ipecac syrup use when not kept in the home Poison treatment in the home Home syrup of ipecac use does not reduce emergency department use or improve outcome A comparison of ipecac syrup and apomorphine in the immediate treatment of ingestion of poisons The frequency of complications associated with the use of multiple-dose activated charcoal Esophageal laceration and charcoal mediastinum complicating gastric lavage Severe hypernatremia secondary to gastric lavage Advantage of high-surface-area charcoal for gastrointestinal decontamination in a human acetaminophen ingestion model Position paper: Single-dose activated charcoal Clinical pharmacokinetics of oral activated charcoal in acute intoxications Efficacy of activated charcoal administered more than four hours after acetaminophen overdose Activated charcoal reduces the need for N-acetylcysteine treatment after acetaminophen (paracetamol) overdose Acceleration of the body clearance of phenobarbital by oral activated charcoal Enhancement of theophylline clearance by oral activated charcoal Gastrointestinal clearance of drugs with activated charcoal European Association of Poisons Centres and Clinical Toxicologists (1999) Position statement and practice guidelines on the use of multi-dose activated charcoal in the treatment of acute poisoning Physicochemical characteristics of drugs and response to repeatdose activated charcoal Randomized study of the treatment of phenobarbital overdose with repeated doses of activated charcoal The treatment of tricyclic antidepressant overdose with repeated charcoal Multiple-dose activated charcoal for treatment of yellow oleander poisoning: A single-blind, randomised, placebo-controlled trial Multiple-dose activated charcoal in acute self-poisoning: A randomised controlled trial Effect of charcoal-drug ratio on antidotal efficacy of oral activated charcoal in man The effect of food and ice cream on the adsorption capacity of paracetamol to high surface activated charcoal: In vitro studies Role of repeated doses of oral activated charcoal in the treatment of acute intoxications Aspiration of activated charcoal and gastric contents Risk factors for emesis after therapeutic use of activated charcoal in acutely poisoned children Gastrointestinal tract perforation with charcoal peritoneum complicating orogastric intubation and lavage Gastrointestinal obstruction associated with multiple-dose activated charcoal Fatal pulmonary aspiration of oral activated charcoal Activated charcoal and the absorption of ferrous sulfate in rats Treatment of acetaminophen ingestion with a superactivated charcoal-cola mixture Effects of emetic and cathartic agents on the gastrointestinal tract and the treatment of toxic ingestion Position statement: Cathartics Severe hypermagnesemia due to multiple-dose cathartic therapy Electrolyte disorders following oral sodium phosphate administration for bowel cleansing in elderly patients Sorbitol catharsis does not enhance efficacy of charcoal in a simulated acetaminophen overdose The effects of charcoal and sorbitol (alone and in combination) on plasma theophylline concentrations after a sustained-release formulation Hypernatremia due to repeated doses of charcoal-sorbitol Fatal poisoning from sodium phosphate enema. Case report and experimental study Rectal prolapse after oral cathartics Effect of whole bowel irrigation on the pharmacokinetics of an acetaminophen formulation and progression of radiopaque markers through the gastrointestinal tract Position statement: Whole bowel irrigation Activated charcoal alone and followed by whole-bowel irrigation in preventing the absorption of sustained-release drugs Use of whole bowel irrigation in an infant following iron overdose Whole-bowel irrigation as a treatment for acute lithium overdose Whole bowel irrigation in an acute oral lead intoxication Parachuting" meth: A novel delivery method for methamphetamine and delayed-onset toxicity from "body stuffing Drugs and pharmaceuticals: management of intoxication and antidotes 445 Efficiency of whole bowel irrigation with and without metoclopramide pretreatment Colonic perforation with volume laxatives Life-threatening respiratory failure following accidental infusion of polyethylene glycol electrolyte solution into the lung Theophylline desorption from activated charcoal caused by whole bowel irrigation solution Endoscopic removal of iron bezoar following acute overdose Endoscopic removal of slow release clomipramine bezoars in two cases of acute poisoning Bezoar formation requiring endoscopic removal after intentional overdose of extended-release nifedipine Endoscopic removal of a cocaine packet from the stomach Acute pediatric lead poisoning: Combined whole bowel irrigation, succimer therapy, and endoscopic removal of ingested lead pellets Gastrotomy -A surgical approach to iron overdose Radiopacity of clomipramine conglomerations and unsuccessful endoscopy: Report of 4 cases Prognosis of cocaine body-packers Surgical treatment in cocaine body packers and body pushers The altered pharmacokinetics and pharmacodynamics of drugs commonly used in critically ill patients Position Paper on urine alkalinization Acute hypermagnesemia: A rare complication of antacid administration after bone marrow transplantation Methods used to decrease lithium absorption or enhance elimination Hypercalcemic crisis in pregnancy associated with excessive ingestion of calcium carbonate antacid (milk-alkali syndrome): Successful treatment with hemodialysis Hypermagnesemia-induced fatality following epsom salt gargles Excessive calcium ingestion leading to milk-alkali syndrome Pamidronate treatment of hypercalcemia caused by vitamin D toxicity Lithium nephrotoxicity revisited Forced euvolemic ciuresis with mannitol and furosemide for prevention of contrast-induced nephropathy in patients with CKD undergoing coronary angiography: A randomized controlled trial Lithium poisoning: Pharmacokinetics and clearance during different therapeutic measures Treatment of life-threatening lithium toxicity with high-volume continuous venovenous hemofiltration Clinical pharmacokinetics during continuous haemofiltration Treatment of acute leukemia with amethopterin (4-amino, 10-methyl pteroyl glutamic acid) Principles and techniques applied to enhance elimination Successful treatment of valproic acid overdose with hemodialysis Hemodialysis in the treatment of lactic acidosis in diabetics treated by metformin: a study of metformin elimination Hemodialysis is as effective as hemoperfusion for drug removal in carbamazepine poisoning Severe iron intoxication treated with exchange transfusion Exchange transfusion in the treatment of severe theophylline poisoning Exchange transfusion in severe infant salicylism Acute poisoning with isoniazid treated by exchange transfusion Exchange transfusion treatment in a newborn with phenobarbital intoxication Vincristine overdose: Experience with 3 patients Drug-induced methaemoglobinaemia Successful whole blood exchange by apheresis in a patient with acute cyclosporine intoxication without long-term sequelae Therapeutic approaches to ion channel diseases Halothane modulation of skeletal muscle ryanodine receptors: Dependence on Ca 2+ , Mg 2+ , and ATP Medical complications of psychiatric treatment The Hunter serotonin toxicity criteria: Simple and accurate diagnostic decision rules for serotonin toxicity The serotonin syndrome The serotonin syndrome Toxin-induced hyperthermic syndromes A thyrotoxicosis outbreak due to dietary pills in Paris Thyrotoxicosis factitia in a post-aortocoronary bypass patient Pediatric fatality following ingestion of dinitrophenol: Postmortem identification of a "dietary supplement Shortand long-term outcomes of heatstroke following the 2003 heat wave in Lyon, France Near-fatal heat stroke during the 1995 heat wave in Chicago Ambient temperature and mortality from unintentional cocaine overdose Further studies of the role of hyperthermia in methamphetamine neurotoxicity Heat stroke: Clinical and chemical observations on 44 cases Cooling and hemodynamic management in heatstroke: Practical recommendations Whole-body cooling of hyperthermic runners: Comparison of two field therapies Emergency treatment of exertional heatstroke and comparison of whole body cooling techniques Diazepam and chlormethiazole attenuate the development of hyperthermia in an animal model of the serotonin syndrome Antagonism of cocaine, amphetamine, and methamphetamine toxicity Effects of diltiazem on hyperthermia induced seizures in the rat pup Dantrolene stabilizes domain interactions within the ryanodine receptor Enhanced excitation-coupled calcium entry in myotubes expressing malignant hyperthermia mutation R163C is attenuated by dantrolene Managing an effective treatment for neuroleptic malignant syndrome Drug treatment of the neuroleptic malignant syndrome Contrast medium-induced nephropathy: Strategies for prevention Acetaminophen hepatotoxicity: The first 35 years Acetaminophen bioactivation by human cytochrome P450 enzymes and animal microsomes Acetylcysteine for acetaminophen poisoning Mechanism of action of N-acetylcysteine in the protection against the hepatotoxicity of acetaminophen in rats in vivo Intravenous acetylcysteine in paracetamol induced fulminant hepatic failure: A prospective controlled trial Improvement by acetylcysteine of hemodynamics and oxygen transport in fulminant hepatic failure N-Acetylcysteine improves indocyanine green extraction and oxygen transport during hepatic dysfunction N-Acetylcysteine increases liver blood flow and improves liver function in septic shock patients: Results of a prospective, randomized, double-blind study Selective effects of N-acetylcysteine stereoisomers on hepatic glutathione and plasma sulfate in mice Dissociation of increased sulfation from sulfate replenishment and hepatoprotection in acetaminophen-poisoned mice by N-acetylcysteine stereoisomers A patient-tailored N-acetylcysteine protocol for acute acetaminophen intoxication A prospective evaluation of shortened course oral N-acetylcysteine for the treatment of acute acetaminophen poisoning Guidelines for the management of paracetamol poisoning in Australia and New Zealand -Explanation and elaboration Efficacy of oral N-acetylcysteine in the treatment of acetaminophen overdose. Analysis of the national multicenter study (1976 to 1985) Deaths from low dose paracetamol poisoning Acetaminophen misconceptions Risk factors and mechanisms of anaphylactoid reactions to acetylcysteine in acetaminophen overdose Frequency of medication errors with intravenous acetylcysteine for acetaminophen overdose Dilution of intravenous N-acetylcysteine as a cause of hyponatremia Comparison of the 20-hour intravenous and 72-hour oral acetylcysteine protocols for the treatment of acute acetaminophen poisoning Hepatotoxicity despite early administration of intravenous N-acetylcysteine for acute acetaminophen overdose Acetaminophen overdose with altered acetaminophen pharmacokinetics and hepatotoxicity associated with premature cessation of intravenous N-acetylcysteine therapy Pharmacokinetic effects of diphenhydramine or oxycodone in simulated acetaminophen overdose Tylenol extended relief overdose On the characters, actions, and therapeutic uses of the bean of Calabar The first use of physostigmine in the treatment of atropine poisoning. A translation of Kleinwachter's paper entitled 'Observations on the effect of Calabar bean extract as an antidote to atropine poisoning Comparative inhibitory effects of various physostigmine analogs against acetyl-and butyrylcholinesterases Inhibition of acetylcholinesterase from electric eel by (-)-and (+)-physostigmine and related compounds Accommodation of physostigmine and its analogues by acetylcholinesterase is dominated by hydrophobic interactions Unconventional ligands and modulators of nicotinic receptors Asystole complicating physostigmine treatment of tricyclic antidepressant overdose Assessing physostigmine's contraindication in cyclic antidepressant ingestions Mechanism of imipramine-induced seizures in amygdala-kindled rats Failure of physostigmine in intoxications with tricyclic antidepres-Drugs and pharmaceuticals: management of intoxication and antidotes 449 sants in rats A comparison of physostigmine and benzodiazepines for the treatment of anticholinergic poisoning Physostigmine: Is there a role for this antidote in pediatric poisonings? Detecting life-threatening lactic acidosis related to nucleosideanalog treatment of human immunodeficiency virus-infected patients, and treatment with L-carnitine Supplementation with L-carnitine does not reduce the efficacy of epirubicin treatment in breast cancer cells Science review: Carnitine in the treatment of valproic acid-induced toxicity -What is the evidence? Case files of the Children's Hospital of Michigan Regional Poison Control Center: The use of carnitine for the management of acute valproic acid toxicity Basic pharmacology of valproate: A review after 35 years of clinical use for the treatment of epilepsy Valproic acid metabolism and its effects on mitochondrial fatty acid oxidation: A review Delayed valproic acid toxicity: A retrospective case series Acute valproate poisoning: Pharmacokinetics, alteration in fatty acid metabolism, and changes during therapy Valproic acid toxicity: Overview and management Carnitine disposition before and during valproate therapy in patients with epilepsy Involvement of recognition and interaction of carnitine transporter in the decrease of L-carnitine concentration induced by pivalic acid and valproic acid Acute valproate-associated microvesicular steatosis: Could the [ 13 C]methionine breath test be useful to assess liver mitochondrial function? Carnitine as an antidote for acute valproate toxicity in children L-carnitine supplementation in childhood epilepsy: Current perspectives Serum and muscle carnitine levels in epileptic children receiving sodium valproate Vigabatrin, lamotrigine, topiramate and serum carnitine levels Carnitine deficiency and hyperammonemia in children receiving valproic acid with and without other anticonvulsant drugs The effect of carnitine supplementation in valproateinduced hyperammonaemia Diet-and valproate-induced transient hyperammonemia: Effect of L-carnitine Carnitine deficiency during valproic acid treatment Valproic acid overdose and L-carnitine therapy A case of valproateassociated hepatotoxicity treated with L-carnitine Valproate-associated hyperammonemic encephalopathy Effect of L-carnitine treatment for valproate-induced hepatotoxicity The role of carnitine supplementation during valproic acid therapy Valproic acid overdoses: A retrospective study comparing serum drug levels and the incidence of adverse outcomes L-Carnitine was safely administered in the setting of valproate toxicity CARNI-TOR ® (levocarnitine) Oral Solution (1 g per 10 mL multidose). CARNITOR ® SF (levocarnitine) Sugar-Free Oral Solution (1 g per 10 mL multidose) A case of hemoperfusion and L-carnitine management in valproic acid overdose EMG changes in chronically dialyzed uraemic subjects undergoing D,L-carnitine treatment Enhancement of activities relative to fatty acid oxidation in the liver of rats depleted of L-carnitine by D-carnitine and a γ-butyrobetaine hydroxylase inhibitor Drug-induced hypoglycemia. A review of 1418 cases Drug-induced disorders of glucose metabolism. Mechanisms and management Astrocytic glycogen influences axon function and survival during glucose deprivation in central white matter Brain glucose uptake and unawareness of hypoglycemia in patients with insulin-dependent diabetes mellitus Neuroglycopenia and adrenergic responses to hypoglycaemia: Insights from a local epidemic of serendipitous massive overdose of glibenclamide Reversal of salicylate-induced euglycemic delirium with dextrose Hypoglycemic seizures during transient hypoglycemia exacerbate hippocampal dysfunction The distribution of hypoglycemic brain damage Concentrated Dextrose for Intravenous Administration Docking studies show that D-glucose and quercetin slide through the transporter GLUT1 Peripheral intravenous infiltration necrosis The effect of 25 grams i.v. glucose on serum inorganic phosphate levels Sandostatin ® octreotide acetate Injection Prescribing Information Pharmacological agents that directly modulate insulin secretion Oral hypoglycemic agents Drugs and pharmaceuticals: management of intoxication and antidotes 451 Inhibition of insulin secretion as a new drug target in the treatment of metabolic disorders Molecular signaling of somatostatin receptors Effect of octreotide on insulin requirement, hepatic glucose production, growth hormone, glucagon and c-peptide levels in type 2 diabetic patients with chronic renal failure or normal renal function Octreotide for pediatric sulfonylurea poisoning A diabetic woman with worsening heart failure, hunger, and tremor Effectiveness of octreotide in a case of refractory sulfonylureainduced hypoglycemia Octreotide reverses hyperinsulinemia and prevents hypoglycemia induced by sulfonylurea overdoses Octreotide: An antidote for sulfonylureainduced hypoglycemia Comparison of octreotide and standard therapy versus standard therapy alone for the treatment of sulfonylurea-induced hypoglycemia The effectiveness of various doses of octreotide for sulfonylurea-induced hypoglycemia after overdose Successful treatment of sulfonylureainduced prolonged hypoglycemia with use of octreotide Octreotide for sulfonylurea-induced hypoglycemia following overdose Anaphylactoid reaction to octreotide Case files of the Medical Toxicology Fellowship Training Program at the Children's Hospital of Philadelphia: A pediatric exploratory sulfonylurea ingestion Bradycardia and hyperkalemia associated with octreotide administration Octreotide in children with hypoglycaemia due to sulfonylurea ingestion Laparoscopic pancreatectomy for persistent hyperinsulinemic hypoglycemia of infancy Octreotide therapy for persistent hyperinsulinemic hypoglycemia of infancy Effectiveness of SMS 201-995, a synthetic, long-acting somatostatin analogue, in treatment of quinine-induced hyperinsulinaemia Hypoglycaemia and counterregulatory hormone responses in severe falciparum malaria: Treatment with sandostatin Bench-to-bedside review: Antidotal treatment of sulfonylurea-induced hypoglycaemia with octreotide Somatostatin and somatostatin analogues: Pharmacokinetics and pharmacodynamic effects Glyburide sold as "Street Steroid" causes hypoglycemia complicated by inappropriate IV administration of octreotide Long-term non-surgical therapy of severe persistent congenital hyperinsulinism with glucagon Isoniazid-associated hepatitis. Report of an outbreak Therapeutic drug monitoring in the treatment of tuberculosis Pyridoxine in clinical toxicology: A review Acute isoniazid poisoning Accidental isoniazid poisoning -A report The origin and turnover of D-serine in brain Glutamatergic regulation of serine racemase via reversal of PIP2 inhibition The effect on GABA metabolism in brain of isonicotinic acid hydrazide and pyridoxine as a fuction of time after administration Profound acidosis caused by isoniazid ingestion Convulsions as the etiology of lactic acidosis in acute isoniazid toxicity in dogs Single high-dose pyridoxine treatment for isoniazid overdose Isoniazid self-poisoning Fitzpatrick's Dermatology in General Medicine Acute isoniazid poisoning in childhood Reversal of prolonged isoniazid-induced coma by pyridoxine Acute isoniazid neurotoxicity in an urban hospital Isoniazid overdose: Four case reports and review of the literature Evaluation of diazepam and pyridoxine as antidotes to isoniazid intoxication in rats and dogs Acute isoniazid toxicity and the need for adequate pyridoxine supplies Isoniazid toxicity: A survey of pyridoxine availability Sensory neuropathy from pyridoxine abuse. A new megavitamin syndrome Treatment of refractory seizures in massive isoniazid overdose clinical practice guideline update: Use of chemotherapy and radiation therapy protectants Cytoprotective effects of amifostine in the treatment of childhood malignancies Amifostine and glutathione prevent ifosfamide-and acrolein-induced hemorrhagic cystitis Drugs and pharmaceuticals: management of intoxication and antidotes 453 Randomized placebo-controlled multicenter evaluation of diethyldithiocarbamate for chemoprotection against cisplatin-induced toxicities Myeloid growth factors NEUMEGA ® [nu-meg