• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/912

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

912 Cards in this Set

  • Front
  • Back
When are peak plasma concentrations reached with acetaminophen?
1 hour
When is complete absorption of acetaminophen achieved?
4 hours
What are the primary metabolic pathways for APAP?
Conjugation with glucuronide (40-67%)
Sulfate (20-46%)
What is the cytotoxic metabolite of APAP?
NAPQI
How is NAPQI typically dealt with by the body?
Combines with glutathione and other thiol containing compounds
How does APAP cause liver damage?
NAPQI covalently binds to critical cell proteins in the liver, it initially occurs in hepatic zone III - centrilobular region
What are the clinical effects of severe APAP toxicity?
Severe fulminant liver failure which causes multi organ failure, systemic inflammatory response syndrome, hypotension, cerebral edema, death
How is NAC effective in APAP OD?
-NAC enhances sulfation
-NAC serves as a glutathione precursor
-NAC is a glutathione substitute
-NAC may reduce systemic toxicity (scavenger or free radicles, decreasing edema)
What are the clinical stages of APAP toxicity?
1 - pre-injury (0-12 hours) - nausea, vomiting, anorexia, may be completely asymptomatic (elevated APAP)
2- liver injury (8-36hours)- nausea, vomiting, RUQ tenderness (transaminitis)
3 - maximum liver injury (2-4 days)- liver failure (encephalopathy, coagulopathy, hemorrhage, acidosis)
4 - recovery (>4days) - none
Which liver enzyme rises and peaks first?
AST (ALT, PT and bilirubin rise and peak shortly after AST)
What are signs and symptoms consistent with fulminant liver failure?
metabolic acidosis
coagulopathy
hepatic encephalopathy
What is the definition of an acute ingestion of APAP?
A single ingestion occurring within a 4-hour period (all other ingestions are considered chronic)
What amount of APAP must be consumed before significant liver toxicity is evident?
Significantly >150mg/kg
OR
>7.5g
When does the risk of hepatoxicity after APAP ingestion increase?
8 hours or longer after ingestion
What should be done for patients at risk of toxicity whose serum APAP cannot be obtained prior to 6-8 hours after ingestion?
Start NAC
When can the APAP treatment nomogram not be used?
-prior to 4 hours after ingestion
-chronic ingestion
-APAP concentration cannot be obtained
-Evaluation of patient >24h post ingestion
-Time of ingestion is unknown
What increases the risk of hepatotoxicity from chronic ingestion?
-increased total dose of APAP
-longer duration over which it has been ingested in supra therapeutic concentrations
What are possible higher risk factors for liver injury in patients taking therapeutic doses of APAP?
chronic INH and chronic ETOH (increased CYP2E1 activity)
prolonged fasting (malnourished, AIDS, prolonged vomiting)
-children with febrile illness
What is considered a significant elevation in AST in chronic APAP toxicity?
AST >/= 50 IU
What is the APAP serum concentration after a typical therapeutic dose?
peaks below 30ug/L and less than 10ug/L at 4 hours
Which patients is chronic APAP toxicity should be treated with NAC?
AST >/= 50IU
[APAP] higher than expected
Does acetaminophen cross the placenta?
Yes
What is the approach to a pregnant female with APAP OD?
-identical to that in any patient.
acute OD - plot on nomogram and treat as needed
chronic - treat if AST >/= 50IU or serum APAP is above expected
Does activated charcoal bind APAP?
YEs
What is the risk of liver injury in patients with APAP OD treated within 6-8 hours? What is the risk of death?
<4%
the risk of death approaches 0
What is the rate of liver injury in patients treated within 8-24 hours post ingestion?
~30%
What route of NAC delivery should be used in patients with evidence of liver failure?
IV (use of this route results in less hypotension, less cerebral edema, and death)
What proportion of patients treated with IV NAC develop anaphylactoid reactions?
2-6%
What is the downside to PO NAC?
50% of patients vomit (potentially delaying timely antidotal delivery)
What should be done if a patient receiving PO NAC vomits?
Any dose that is vomited within 1 hour of administration should be repeated
What are the lengths of the various NAC protocols?
IV NAC - 21 hours
PO NAC - 72 hours
What are the end points of the NAC protocol?
-predetermined length of the protocol
AND
AST concentration normal and serum APAP <10micrograms/mL
What are clinical predictors of severe hepatic failure?
pH <7.3 or Cr >3.3 and INR >5 and grade III or IV encephalopathy
APACHE II >20
Lactate >3.5mmol/L prior to resuscitation
What clinical picture can salicylate toxicity cause?
Metabolic acidosis
seizure
hyperthermia
pulmonary edema
cerebral edema
renal failure
death
How long does it take for salicylic acid to be absorbed and for peak levels to occur?
Absorbed - 30min
Peak levels - 2-4 hours
How do salicylates cause toxicity?
By uncoupling mitochondrial oxidative phosphorylation
this results in increased pyruvate and lactic acid thereby increasing the metabolic rate, temperature and O2 consumption which results in hypoglycaemia
inefficiency of anaerobic metabolism then results in hyperthermia
Is the majority of ASA in the blood ionized or nonionized?
At physiological pH, ASA is primarily ionized but as pH decreases more particles become un-ionized markedly increasing the movement of salicylate into the tissues and CNS (only non-ionized particles can cross the cell membrane)
What are reasons that patients with salicylate toxicity develop hypokalemia?
-vomiting (stimulation of the CTX)
-increased renal excretion of Na+, HCO3-. and K+ in compensation of respiratory alkalosis
-salicylate induced increased permeability of renal tubules
-intracellular accumulation of Na+ and H20
-inhibition of active transport
What are RF for the development of pulmonary edema in salicylate toxicity?
-age >30
-cigarette smoking
-chronic salicylate ingestion
-metabolic acidosis
-neurologic symptoms
-serum salicylate >40mg/dL
What is the most likely cause of an altered sensorium in salicylate toxicity?
-cerebral edema
How do salicylates cause toxicity?
-they stimulate the respiratory centre, leading to hyperventilation and respiratory alkalosis
-they are weak acids and impair renal function which leads to the accumulation of inorganic acids
-they interfere with Kreb's cycle and uncouple oxidative phosphorylation -> producing lactate and heat
-they induce fatty acid metabolism and generate ketone bodies resulting in a wide anion gap metabolic acidosis
What do patients with salicylate ingestion that develop cerebral or pulmonary edema require?
-immediate dialysis
What happens in chronic salicylate ingestion?
-decreased albumin binding which results in increased free salicylate
-this free salicylate enters the cell causing significant clinical illness with a relatively low serum salicylate
What is a toxic dose of ASA? What is a potentially lethal dose of ASA?
toxic 200-300mg/kg
potentially lethal 500mg/kg
Is there a nomogram for ASA?
Yes, it should not be used to determine prognosis or treatment
What should be done if a patient with salicylate toxicity requires intubation?
This patient requires dialysis
What are 3 main objectives in the treatment of salicylate toxicity?
-prevent further absorption
-correct fluid deficits/acid-base abnormalities
-increase excretion
What is the target urine output in salicylate toxicity? What is the risk of too much fluid?
2-3mL/kg/hr
excessive fluid can worsen cerebral and pulmonary edema
When is urine alkalization advisable in salicylate toxicity?
salicylate level >35mg/dL
significant acid-base disturbance or increased salicylate levels
How does urine alkalization work?
Alkaline urine traps the salicylate ion and increases excretion
What is the dose of NaHCO3?
1-2 mEq/kg over 1-2 hours
What must be done prior to urine alkalization?
correct K+ depletion
When is HD advisable in Salicylate toxicity?
Absolute serum level
-acute salicylate toxicity >7.2mmol/L
-chronic salicylate toxicity >4.4mmol/L
-rising ASA level
-clinical deterioration despite adequate supportive care

Manifestations of salicylism in vital end organs
-AMS
-ET intubation
-coma
-pulmonary edema
-severe acid-base imbalance

Unable to metabolize and eliminate drug
-renal or hepatic failure
What is the order when you want to alkalinize the urine?
bolus 1-2 mEq/kg of hypertonic sodium bicarb then IV infusion of 3 amps NaHCO3 (132mEq) in 1L of D5W at 1.5-2X maintenance
When should alkalinization be considered
tinnitus
CNS symptoms
all patients with a salicylate concentration greater that 30-40mg/dL
What can be considered in small children with salicylate toxicity instead of HD?
exchange transfusion
What is ASA metabolized to?
ASA-> salicylic acid-> glycine conjugation, glucuronyl transferase, hydrolysis (direct renal excretion)
Which NSAIDs are available in OTC and prescription strength?
ibuprofen
naproxen
How is the therapeutic anti-inflammatory effect of NSAIDs achieved?
Inhibition of COX and blockade of prostaglandin production
Where is COX-1 concentrated?
-platelets
-vascular endothelial cells
-gastric mucosal cells
-renal collecting tubules
When is COX-2 expressed?
-it is inducible, therefore it is only expressed in response to certain inflammatory stimuli
Why do NSAIDs frequently cause GI symptoms?
It becomes trapped in gastric mucosal cells
What happens to the NSAIDs after they are absorbed?
They become highly bound to albumin
What are the main clinical findings of NSAID OD?
Largely asymptomatic
+/- minor CNS or GI distrubance
At what dose of ibuprofen does symptomatic OD occur?
100mg/kg
When is renal dysfunction seen with ibuprofen OD?
With massive acute OD associated with hypotension and hypovolemia. Typically this is reversible
Which NSAID in OD is associated with a relatively high incidence of seizures?
Mefanamic acid
Should plasma NSAID concentrations be measured?
No, they are not useful, but screening for APAP should be done
At what level of ibuprofen ingestion, should children be evaluated?
>300mg/kg
Which NSAID OD should be managed more aggressively
Pyrazolone
Fenamate
How long should you observe patient with non-trivial NSAID ODs
4 hours post ingestion and until symptoms are noted to be mild or improving
What are the 3 main groups of anticholinergics?
-antimuscarinics
-neuromuscular blockers
-ganglionic blockers
What plants have anticholinergic activity?
Atropa belladonna -> atropine
Datura stramonium (jimson weed) -> scopolamine
What are current day uses for belladonna alkaloids?
-mydriatics (pupillary dilators)
-anti-spasmodics
-decreased gastric secretions
-prevent motion sickness
-treat asthma
-treat bradycardia
-dry airway secretions and block the vagal response to laryngoscopy
Which agents are central anti-muscarinics? what are they used for?
Benztropine
Trihexylphenidyl
-2nd line antiparkinsons agents and to counteract the effects of neuroleptics
What are other pharmacologic agents that possess anticholinergic activity
TCA
phenothiazines
anti-histamines
carbamazepine
cyclobenzaprine
Where are muscarinic receptors found?
-smooth muscles in the eye, intestinal tract and urinary bladder
-they regulate sweat, salivary and mucosal gland activity
What does generalized inhibition of muscarinic receptors by atropine cause?
Tachycardia
Pupillary dilatation
loss of accommodation
inability to sweat
drying on mucosal surfaces
GI paralysis
urinary retention
What does muscarinic inhibition in the CNS cause?
stimulation
seizures
coma
choreoathetosis
memory impairment
perceptual and cognitive dysfunction
What are the least sensitive organs to antimuscarinic drugs?
the bladder
the GI tract
What CNS manifestations of anticholinergic poisoning do children typically exhibit?
CNS stimulant effects
seizures preceded by CNS irritability or depression
What do patients with anticholinergic toxicity die of?
-elevated temperature (due to increased motor activity and impaired heat exchange)
What are manifestations of chronic anticholinergic poisoning?
-organic mental symptoms with lack of significant peripheral anticholinergic signs
What are 2 likely settings for chronic anticholinergic toxicity?
1) elderly patients taking anticholinergic drugs for Parkinsonism or other chronic diseases
2)the psychiatric patient receiving neuroleptic therapy and started on another anticholinergic drug
How can you differentiate patients with sympathetic overload from those with anticholinergic effects?
the presence or absence of diaphoresis
What are findings suggestive of phencyclidine poisoning?
-nystagmus
-diaphoresis
-small pupils
-extreme aggitation
What is often a distinguishing feature of serotonin syndrome?
aggitation
tremor in the lower extremities
What can be a distinguishing feature of NMS?
rigidity is a prominent physical exam feature
What agents are suggested by antimuscarinic effects and ECG abnormalities?
cardiotoxic agents with antimuscarinic side effects (TCA, carbamazepine, phenothiazines)
How should agitation be controlled in the patient with anticholinergic poisoning?
BZD
How should hyperthermia be treated in patients with anticholinergic toxicity?
Ice water or evaporative cooling (antipyretics, cooling blankets may be ineffective and there is no clear role for dantrolene)
How should seizures be managed in a patient with anticholinergic toxicity?
BZD (no role for phenytoin)
What does physostigmine do?
It is an acetylcholinersterase inhibitor that blocks the degradation of ACh which accumulates and overcomes the effect of ACh receptor blockade
What are the risks of physostigmine?
It can precipitate cholinergic excess causing seizures, muscle weakness, bradycardia, bronchoconstriction, lacrimation, salivation, bronchorrhea, vomiting and diarrhea
What can occur if physostigmine is given to patients with TCA OD?
asystole
(therefore TCA OD is a contraindication to physostigmine)
How is physostigmine given?
1-2 mg over 5 min (0.02 mg/kg, max 0.5mg) for children
Can be repeated in 10-15 minutes
How long after ingestion should you monitor patients with anticholinergic ingestion?
4 hours
How long should you observe patients who have ingested datura stramonium?
8 hours
When do most serious complications from TCA occur?
Within 30-60 minutes.
Why is absorption prolonged in overdose with TCAs?
The anticholinergic effect limits gastrointestinal motility and absorption.
Which TCAs have active metabolites?
All tertiary amines and amoxapine (a secondary amine_
What are the major pharmacodynamic effects of TCAs?
-sodium channel blockade
-alpha receptor antagonism
-anticholinergic
-antihistaminic
-inhibit GABA receptors
-impair serotonin and norepinephrine re-uptake
-impair K+ efflux
What is the mechanism of cardiovascular collapse in TCA OD?
-lactic acidosis which impairs myocardial sodium conduction
What is the predictive value of altered LOC for TCA induced seizures?
Mental status changes do not predict the occurrence of seizures and 23% of patients are awake and alert immediately before a seizure.
Which TC have the highest incidence of seizures? Of cardiac toxicity?
Amoxapine (seizures)
Maprotiline (cardiac and seizures)
What is the toxic dose of TCAs?
10mg/kg or 1000mg
What are ECG changes associated with TCA poisoning?
Wide QRS
Prolonged QT
Rightward deviation of the terminal 40msec QRS axis (R-wave in aVR >3mm)
Brugada-like pattern
What is the value of quantitative TCA concentrations in the clinical setting?
They have little value because they are not available in a timely manner and do not accurately predict the degree of toxicity
Which patients with TCA intox should be intubated?
Decreased LOC
Rapid Deterioration
Any patient who will be receiving gastric lavage
By which mechanism does NAHCO3 help in TCA OD
-provides an alkaline pH as well as a sodium load and hypertonicity that increases conductance through myocardial sodium channels.
What is the usefulness of various vasopressors in TCA induced hypotension?
Dopamin and NE should be used to treat refractory hypotension
Dobutamine can be used for inotropic support
Vasopressin has also been used in refractory hypotension
Which antiarrythmic are contraindicated in TCA poisoning?
Phenytoin (shown to prolong episodes of vtach)
Type Ia (quinidine, disopyramide, procainamide)
Type Ic (flecanide, propafenone, moricizine)
How long does an asymptomatic patient need to be monitored after a TCA OD
At least 6 hours
What are signs that a TCA poisoned patient should be admitted to the ICU?
-Ventilatory insufficiency
-Desaturation
-QRS >100msec
-sinus tachycardia >120bpm
-dysrhythmias
-hypotension
-decreased LOC
-seizures
-abnormal or inactive bowel sounds
Define serotonin syndrome
A constellation of signs and symptoms manifesting as autonomic, neuromuscular and mental status changes.
List manifestations of SSRI poisoning?
Gastrointestinal: emesis, abdo pain, nausea

Cardiovascular: sinus tach, hypertension, trigeminy and junctional rhythm

CNS: drowsiness, tremor, euphoria, headache
What are factors that help distinguish serotonin syndrome from neuroleptic malignant syndrome?
-history of precipitating medication (SSRI vs neuroleptic
-SS is more rapid in onset and more often exhibits clonus
How long after ingestion does it take to reach peak plasma concentrations of TCA?
2-4hours (therapeutic dose)
longer with OD because of anticholinergic effects delay GI motility
Describe the pharmacokinetics of TCAs?
highly lipophilic
extensively bound to plasm proteins
large VD
metabolized predominantly by the liver
What are the major pharmacodynamic effects of cyclic antidepressants?
Sodium channel blockade
alpha blockade
inhibition of biogenic amine re-uptake
muscarinic receptor blockade
histamine receptor blockade
potassium efflux blockade
indirect GABA antagonism
What phase of myocardial depolarization is prolonged in TCA OD?
Phase 0 (sodium conductance is blocked through fast sodium channels)
What is the result of the blocked fast sodium channels in TCA OD?
-Decreased conduction with a prolonged QRS complex (>100msec)
--ve inotropic effects (secondary to impaired excitation-contraction coupling)
What is the result of alpha adrenoceptor blockade?
decreased preload and afterload (decreased SVR, widened PP, decreased pupil size)
What does 5-HT and NE reuptake inhibition in the CNS cause?
Delirium
Seizures
What does K efflux blockade in TCA OD do?
prolongs phase 3 of the myocardial action potential resulting in increased QT interval duration
What does GABA inhibition cause?
Increased CNS excitation and seizures
Which patients should be considered to have cyclic antidepressant poisoning until proven otherwise?
-QRS >100msec
-Rightward deviation of the terminal 40msec QRS axis (R-wave in aVR >3mm or R/S ratio in aVR >0.7)
What are the PNS effects of cyclic antidepressants?
tachycardia, hyperthermia, mydriasis, anhydrosis, red skin, decreased BS, ileus, urinary retention, distended bladder (anticholinergic effects)
reflex tachycardia and miosis/mid-range pupils (alpha blockade)
Should tox screens for TCAs be done?
Neither tox or lab tests are useful in clinical decision-making.
Why may ventilatory support be required in TCA OD?
To avoid respiratory acidosis, because acidosis inhibits conductance through fast sodium channels
When should bicarb be administered in TCA OD?
-QRS >100msec
-symptomatic patient with hypotension or dysrhythmia
-acidemia
-IV sodium bicarbonate
How is NaHCO3 administered?
IV boluses of 1-2 mEq/kg until the QRS narrows or until serum pH increases to 7.50-7.53
If symptoms of TCA OD are refractory to sodium bicarb what can be done?
-hypertonic saline
-dopamine, NE
Which type of antidysrhythmic agents are contraindicated in wide complex tachycardia from TCA OD?
Type Ia and Type Ic agents because they inhibit fast sodium channels)
What else can be considered for polyventricular tachycardia?
Transvenous pacemaker for overdrive pacing
What should be used for agitation and seizures in TCA OD?
BZD
What else can be used for TCA associated seizures?
phenobarbital
propofol
After how many hours of observation can patients be discharged after suspected TCA OD?
6 hours if the following do not develop:
-ventilatory insufficiency
-QRS >100msec
-sinus tachycardia (>120bpm)
-dysrhythmia
-hypotension
-decreased LOC
-seizures
-abnormal or inactive BS
How are SSRI's metabolized?
predominantly by the liver
How long should you wait to start MAOI therapy after discontinuing SSRIs? why?
5 weeks
To avoid precipitating serotonin syndrome
What organ system are most affected by excessive serotonin?
GI tract
CVS system
CNS
What specific symptoms can SSRI OD cause?
sedation
-aggitation
-tremor
-hyper-reflexia
-tachycardia
-bradycardia
-nausea and vomiting
-abdo pain
-facial flushing
-dizziness
Which SSRI requires a prolonged observation period (beyond the routine 6 hours observation period) and why?
Citalopram
Seizures and QT prolongation followed by torsades with cardiac arrest have been observed as long as 8 hours after OD
WHat are Sternbach's suggested diagnostic criteria for serotonin syndrome?
-adding a serotonergic agent to patient's meds or increasing the dose of a patient's serotonergic agent
-at least 3 of the following symptoms
agitation
ataxia
diaphoresis
diarrhea
hyperreflexia
hyperthermia
mental status changes
myoclonus
shivering
tremor
-a neuroleptic has not been started or increased
-other aetiologies (infection, metabolic derangement and withdrawal) have been ruled out
What are helpful clues to differentiating serotonin syndrome from neuroleptic malignant syndrome?
-a history of precipitating medication use
-more rapid onset of symptoms
-the presence of clonus
Is HD an option for SSRI ingestion?
no, SSRIs have large volumes of distribution and are highly bound to plasma proteins
What medications have been proposed to treat the neurologic complications of SSRI OD and serotonin syndrome?
cyproheptadine
methysergide
chlorpromazine
propranolol
(none are considered proven therapy)
How long should patients with SSRI OD be observed and how?
-6 hours (except citalopram and escitalopram which should be observed for up to 12 hours)
-with cardiac monitoring
What are the potential complications of serotonin syndrome?
-This entity can be fatal
-vtach
-hypotension
-coma
-hyperthermia
-rhabdomyolysis
What is the most significant toxic effect of bupropion?
seizure activity
(which may occur after OD or when the maximal daily dose is exceeded)
How should bupropion-induced seizures be treated?
BZD (+/- phenobarb load for recurrent seizures)
WHat is the recommended observation period of immediate-release bupropion? extended-release bupropion?
8 hours
12 hours
What is the mechanism of action for bupropion?
-inhibits dopamine re-uptake
-enhances dopaminergic neurotransmission
What is the mechanism of action of Venlafaxine and its active metabolite O-desmethylvenlafaxine?
It inhibits the reuptake of serotonin to a greater extent than NE and dopamine
What are clinical features of venlafaxine OD?
Common:
somnolence
sinus tachycardia

Rare:
seizures
hypotension
prolonged QRS
prolonged OTc
rhabdomyolysis
hepatic failure
renal failure
What is the mechanism of action of mirtazipine?
-blocks pre-synaptic alpha 2 receptors and increases release of 5HT and NE
What are the effects of monoamine oxidase inhibitors?
-inhibit MAO
-MAOI effect on indirect symptahomimetics such as amphetamine and methamphetamine and potential for enhanced CNS and PNS toxicity
-depletion of NE stores
-inhibitor of vit-B6 containing enzymes
What are the 3 presentations of MAOI toxicity?
MAOI OD
MAOI food or beverage interaction
MAOI drug interaction
What are the 4 phases of MAOI OD?
1) asymptomatic or latent
2) neuromuscular and cardiovascular excitation with sympathetic hyperactivity
3) CNS depression and CV collapse with hypotension + bradycardia
4) secondary complications
What is a lethal dose of MAOI OD?
2mg/kg
What is the onset of sympathetic signs and symptoms with MAOI food or MAOI beverage interaction
minutes to hours
What are signs and symptoms of MAOI drug interactions?
sympathetic storm or serotonin syndrome
What are some typical drugs that are incompatible with MAOIs?
-Mixed-acting/indirect-acting sympathomimetics
-methylxanthines
-antidepressants
-opioids
-other drugs that can cause serotonin syndrome
What is the management of hypertension in MAOI tox?
-mild hypertension does not require treatment
-HTN with signs of impending or ongoing target organ damage -> should be treated with sodium nitroprusside or phentolamine
What is the dosing of phentolamine?
5mg bolus or 0.02-0.1 mg/kg
What is the management of bradycardia?
should not be treated unless associated with severe hypotension then IV atropine given until resolved then possibly pacemaker
What can be used to ttx life threatening hyperthermia in MAOI toxicity?
Dantrolene 2.5mg/kg IV
How long can it take to manifest the symptoms of MAOI OD?
up to 24 hours
What are the symptoms of SSRI discontinuation syndrome?
insomnia
nausea
HA
sensory disturbances
hyperarousal
anxiety
agitation
tachycardia
tremor
What is the definition of asphyxiation?
A condition of severely deficient supply of oxygen to the body
What happens when the Fi02 falls from 0.21 to 0.15
Autonomic stimulation
(tachycardia, tachypnea and dyspnea)
cerebral hypoxia (ataxia, dizziness, incoordination, confusion)
NB: dyspnea is not an early finding
What is the management of exposure to asphyxiants?
-removal from exposure
-supportive care
-administration of supplemental oxygen
What is the definition of an asphyxiant?
Any gas that displaces sufficient oxygen from the breathable air
How do irritant gases result in disease?
-they dissolve in the respiratory tract mucus and alter the air-lung interface by invoking an irritant or inflammatory response
How are pulmonary irritants grouped?
highly water soluble - impact mucous membranes of the eyes and upper airways

Poorly water soluble - do not readily irritate the mucous membranes and may have pleasant odors, prolonged breathing allows the gas to reach the alveoli

Intermediate water soluble - produce clinical syndromes that are composite of the other gases
What is the management of exposure to pulmonary irritants?
-signs of upper airway dysfunction mandate direct visualization of the larynx
-bronchospasm may respond to inhaled beta agonists
What additional therapy can be offered to patients exposed to chlorine or hydrogen chloride?
Inhaled 2% sodium bicarb
What is the disposition of patients exposed to pulmonary irritants?
-those exposed to water soluble agents can be discharged if asymptomatic or improve with symptomatic therapy
-those exposed to intermediate or poorly water soluble agents should be observed for increased dyspnea over several hours even if they are initially asymptomatic
What is smoke inhalation?
It is a variant of irritant injury in which heated particulate and adsorbed toxins injure normal mucosa similar to other irritant gases
What do patients that are exposed to filtered or distant smoke inhale?
CO
cyanide and metabolic poisons
(these patients do not inhale smoke)
What laboratory findings suggest cyanide poisoning?
metaboic acidosis
serum lactate >10mmol/L
Should corticosteroids be given to patients with smoke inhalation?
No, they are not indicated and amy be harmful in patients with cutaneous burns
What are worrisome clinical findings in smoke inhalation?
hoarseness
respiratory distress
What are identifiers of substantial exposure to smoke?
closed-space exposure
carbonaceous sputum
What industries is cyanide important in?
metallurgic
photographic
In what occupations do most hydrogen sulfide poisonings occur?
petroleum refinery
sewage storage
What is the odor of hydrogen sulphide?
rotten eggs
Why is the odor not a reliable predictor of hydrogen sulphide exposure?
Odor becomes unnoticeable with extremely high concentrations or prolonged exposures (olfactory fatigue)
How is cyanide toxic to tissues?
It inhibits oxidative metabolism by binding to complex IV of the electron transport chain within mitochondria
What are the toxic effects of hydrogen sulphide poisoning?
pulmonary irritant
cellular poison
How is hydrogen sulphide different from cyanide?
hydrogen sulfide spontaneously dissociates from the mitochondria rapidly, allowing many patients to survive after brief exposure
Which organs manifest dysfunction in cyanide poisoning?
the heart and CNS (coma, seizures, dysrhythmia and CV collapse)
What happens to venous blood in cyanide/HS poisoning?
the oxygen content remains high (tissues cannot extract oxygen) "arterialization"
What symptoms in a fire victim suggest cyanide poisoning?
CV collapse, ventricular dysrhythmia and seizures
What is the goal of therapy in treatment of hydrogen cyanide exposure?
To reactivate the cytochrome oxidase system by providing an alternative binding site for cyanide ion
What does the cyanide antidote kit produce?
A high affinity source of ferric ions (Fe3+) for cyanide to bind to
What therapy should be administered by paramedics and during mass poisoning events for hydrogen cyanide poisoning?
sodium thiosulfate in combination with oxygen and sodium bicarbonate
What are the first 2 components of the HC kit and what is the goal of these therapies?
inhaled amyl nitrite and IV sodium nitrite
the goal is methemoglobin formation -> cyanomethemoglobin
What is the purpose of sodium thiosulfate?
It increases the availability of sulfur donor to increase the rate of rhodonase function (cyanide + cyanomethnemoglobin are converted to thiocyanate by sulfur transferase/rhodonase
What is recommended in fire victims with possible simultaneous CO and CN poisoning?
The use of sodium thiosulfate alone (CO and MetHgb reduce O2 delivery to tissues)
What is the 2nd antidote for CN poisoning?
Hydroxycobalamin (vit B12)
it takes advantage of the high affinity of cobalt and CN
What lab tests does hydroxycobalamin interfere with?
Carboxyhemoglobin and lactate
What is another therapy for CN tox (that is as of yet unproven)?
HBOT (superoxygenates plasma and tissues permitting higher levels of methemoglobinemia)
What is the treatment of HS toxicity?
removal from exposure
standard resuscitation techniques
+/- nitrite portion of the cyanide antidote to form MetHgb
How is CO generated?
Through incomplete combustion of virtually all carbon-containing products
What are the various mechanisms of CO poisoning?
decreased oxygen carrying capacity
CO binding to myoglobin -> rhabdomyolysis
inhibits the final cytochrome complex involved in mitochondrial oxidative phosphorylation
Should cherry red skin colour be used to aid in the diagnosis of CO or CN poisoning?
No, it is a post-mortem finding
What are the delayed neurologic sequelae of CO exposure?
Neurologic syndromes (focal deficits and seizures)
psychiatric and cognitive findings (apathy, memory deficits)
What are RF that predict the development of delayed neurologic sequelae?
Age
Loss of consciousness
Why is the calculated oxygen saturation on an ABG normal in the setting of CO poisoning?
Because the P02 is a measure of dissolved oxygen in the blood which remains normal even in the presence of substantial CO poisoning
What is the purpose of 02 therapy in CO exposure?
-It decreases the half life of COHb from 5 hours to 1 hour
-a sufficient P02 with HBOT can be achieved to sustain life in the absence of adequately functioning Hgb
What is the benefit of HBOT?
HBOT is associated with a reduction in the rate of delayed neurologic sequelae from 12% to 1%
What are indications for HBOT in CO exposure?
1. Evidence of end-organ damage regardless of COHb level (neuro abnormality, cardiovascular instability)
2. Persistent symptoms after treatment with 100% O2
3. COHb >/=25%
4. COHb >/= 15% in pregnant women and children
How long should you treat mildly CO poisoned patients?
~6hours
How does cocaine work?
It causes release of dopamine, epi, NE and serotonin
it inhibits the re-uptake of these stimulatory neurotransmitters from synaptic clefts
It is a LA and results in Na+ channel blockade
What happens when you mix EtOH and cocaine
May form cocaethylene which may potentiate the drug's stimulatory effects
What compound is identified by the urine cocaine tox screen?
benzoyl ecgonine
What can happen when yo give naloxone to someone who has been speed balling?
You may precipitate the underlying cocaine intoxication
What are the rapidly fatal complications of cocaine intoxication?
-hyperthermia
-hypertensive emergencies
-cardiac dysrhythmias
What are cardiovascular sequelae of cocaine intox?
Aortic dissection
pulmonary edema
MI
infarction
intracranial hemorrhage
strokes
infarction of the anterior spinal artery
How long after cocaine use can the urine tox remain positive?
3 days
When is a urine drug screen beneficial?
To document possible abuse or neglect in a child with suggested exposure
To confirm cocaine as an unknown substance in body packers
To differentiate paranoia from drug-induced or psychiatric causes
What are the large categories to consider in agitated delirium?
Metabolic
Structural lesions of the CNS
Endocrine disease
Infection
Tox
Heat stroke
Post-ictal state
What are the potential toxicologic causes of agitated delirium?
sympathomimetic/stimulant ( cocaine, amphetamine, caffeine, PCP/ketamine)
anticholinergic
serotonin syndrome
sedative hypnotic withdrawal
What agents should be avoided in acutely intoxicated patients with sympathomimetic toxidrome?
Phenothiazines (droperidol and haloperidol)
What is the best way to cool a patient?
Ice water
Wet sheets with large fans
packing the entire body in ice
How is HTN managed in cocaine intox?
-BZD
-phentolamine IV 1-5 mg
-avoidance of beta antagonists
How should narrow complex dysrhythmias be treated in cocaine into?
BZD
How should wide complex dysrhythmias be treated in cocaine into?
-sodium bicarb 1-2 mEq/kg
-maybe lidocaine (if unresponsive to NaHCO3)
What are possible causes of stimulant-iduced chest pain?
PTX
pneumomediastinum
pneumopericardium
AD
pulmonary infarction
infection
FB aspiration
endocarditis
pericarditis
ischemia/infarction
coronary stent thrombosis
What should be given to patients with ECG criterial for MI with persistent CP and HTN and a clear history of cocaine intox?
phentolamine 1mg IV (slowly)
Why are beta antagonists (including labetalol) contraindicated during acute cocaine intoxication?
because they may worsen coronary vasoconstriction
Can TPA be given in acute cocaine related MI?
Yes, the same contraindications apply though cardiac cath is preferable
What are "body packers"
Those who ingest cocaine (or other illicit substances) that have been wrapped tightly into condoms or other latex products and sometimes coated in wax
What is the single absolute criteria for surgical removal of body packs?
leaking or poorly secured packets - patient becomes symptomatic
How long should you monitor patients with cocaine-relatd CP?
12 hours
If everything is negative they can go home
What criteria should body packers meet before they are discharged?
-3 packet free stools
-a reliable pack count consistent with ingestion
-a negative contrast radiographic study
What is the mechanism of action of amphetamines?
They enhance the release of catecholamines from pre-synaptic nerve terminals
WHat are patients with amphetamine intoxication at risk of?
hyperthermia
HTN emergency
dysrhythmia
myocardial ischemia
increased potassium
What is a "body stuffer"?
An individual who attempts to conceal evidence of cocaine possession by swallowing the drug while pursued by law enforcement officials
How long should you monitor patients with cocaine-relatd CP?
12 hours
If everything is negative they can go home
What is the mechanism of action of amphetamines?
They enhance the release of catecholamines from pre-synaptic nerve terminals
WHat are patients with amphetamine intoxication at risk of?
hyperthermia
HTN emergency
dysrhythmia
myocardial ischemia
increased potassium
What is a "body stuffer"?
An individual who attempts to conceal evidence of cocaine possession by swallowing the drug while pursued by law enforcement officials
How long should you monitor patients with cocaine-relatd CP?
12 hours
If everything is negative they can go home
What is the mechanism of action of amphetamines?
They enhance the release of catecholamines from pre-synaptic nerve terminals
WHat are patients with amphetamine intoxication at risk of?
hyperthermia
HTN emergency
dysrhythmia
myocardial ischemia
increased potassium
What is the main difference between amphetamine and cocaine?
the toxicity of amphetamines tends to be longer
What is the treatment for MDMA associated hyponatremia in the absence of seizures or other neurologic events?
Fluid restriction
What is different about methamphetamine ("crystal meth" "crank")?
The duration can be significantly longer
What do crystal meth labs contain?
anhydrous ammonia
HCl
NaOH
ether
ephedrine
What is the main difference between amphetamine and cocaine?
the toxicity of amphetamines tends to be longer
What specific life threatening emergency can MDMA precipitate
hyponatremia
What is the treatment for MDMA associated hyponatremia in the absence of seizures or other neurologic events?
Fluid restriction
What is different about methamphetamine ("crystal meth" "crank")?
The duration can be significantly longer
What do crystal meth labs contain?
anhydrous ammonia
HCl
NaOH
ether
ephedrine
What cardiovascular drugs account for the majority of fatalities?
digitalis
propranolol
verapamil
What are the effects of digitalis at therapeutic doses?
-Increasing the force of myocardial contraction to increase cardiac output
-decrease AV conduction to slow the ventricular rate in afib
What is the biochemical basis for the effect of digitalis on conduction?
-inhibition of Na+ and K+ ATPase which increases intracellular Na+ and Ca++ and extracellular K+
Is digitalis toxicity characterized by brady and tachydysrhythmias?
Both, which can also alternate in the same patient
What are the effects of digitalis on Purkinje fibbers?
1) decreases resting potential slowed phase 0 depolarization and conduction velocity
2) decreased action potential duration
3) enhanced automaticity
What is the most common ECG manifestation of digitalis toxicity?
PVCs
What happens to the ventricles at toxic extremes of dig toxicity?
Dangerous sensitivity to mechanical and electrical stimuli
How is digoxin eliminated? How is digitoxin eliminated?
Digoxin is primarily excreted in the urine.
Digitoxin is metabolized in the liver
Describe the pharmacokinetics of digitalis and how that affects how we treat intoxication?
There is significant protein binding and a large volume of distribution which suggests that HD, hemoperfusion and exchange transfusion are ineffective
-the long half lives suggest that temporizing measures (pacemakers and atropine) might cost time, money and lives especially since Fab fragments are available
What factors are associated with increased risk of digitalis toxicity?
Increased sensitivity to digoxin
underlying Heart disease
Electrolyte imbalance
hypoxia

Increased digoxin levels
renal failure
drug interactions
dehydration

Cardiotoxic co-ingestants
BB, CCB, TCA
When should you consider chronic digitalis intoxication?
In any patient on maintenance therapy who develops consistent symptoms, especially with new conduction disturbances or dysrhythmias
What are the main differences of chronic vs acute digitalis intox?
Chronic intox has a higher mortality, normal to low K+, more commonly exhibit ventricular dysrhythmias, usually in elderly people, often need Fab, patients often have underlying heart disease

Acute into has K+ that's normal to high and more commonly exhibit bradycardia and AV block
Which digoxin level correlates with tissue toxicity and when it it achieved?
Steady-state
6-8 hours after a dose or overdose
What are typical K+ levels in digitalis toxicity?
Acute -> hyperkalemia
Chronic-> low serum and total body
What organ systems are affected in digitalis toxicity?
Cardiac -> dysrhythmias
CNS
visual distrubances
gastrointestinal disturbances
What is the antidote for digitalis toxicity?
digibind or digifab
How is hyperkalemia treatment in acute digitalis toxicity?
Serum K+ >5mEq/L warrants consideration of digitalis antibody if not immediately available.
IV glucose, insulin, NaHCO3
recent studies also suggest that Ca++ can be safely given
What should be done about K+ in chronic dig toxicity?
-raising the serum K+ to 3.5-4mEq/L is an important early treatment - K+ can be administered orally or IV
-Mg++ should also be replaced (except in those with renal failure)
What should be considered if you plan on cardioverting a patient with dig intoxication?
Cardioversion may cause asystole and lower energy settings may be less hazardous
Which drugs are believed to be the safest of antidysrhythmic drugs in the setting of digitalis intoxication?
phenytoin
lidocaine
Though digoxin immune Fab are the preferred therapy for dysrhythmias
What is the response rate to Dig Fab fragment?
90% in chronic or acute poisoning
What are expected reactions to Fab?
~1% hypersensitivity- erythema, urticaria, facial edema
-hypokalemia
-exacerbation of CHF
-increased ventricular rate with afib
What is the primary indication for Fab fragment in acute dig toxicity?
hyperkalmia K+>5.5mEq/L or EKG changes
What is the empirical dosing of digibind or digifab and when should it be given?
10 vials over 30min - acute
4-6 vials - chronic life-threatening symptoms with appropriate history and no time to assess dig levels at 1 hour or steady-state
What is the dose of digibind in cardiac arrest?
20 vials
What are 2 other approaches to determine the dosage of Digibind or DigiFab?
-Calculate based on ingested dose
-base the dosage on the steady-state dig or digitoxin level (after 6-8 hours)
What happens to dig levels after Fab fragment administration?
It will remain elevated because both bound and unbound drug is measured.
Which children are at greatest risk of dig toxicity?
Those on chronic digitalis therapy for heart therapy
What accounts for the most paediatric digitalis intoxications and death?
Dosage calculation and administration errors
What are the signs and symptoms of digitalis intoxication in children?
-Obtundation and vomiting are more common than in adults
-Children may be asymptomatic at higher levels than adults
-bradydysrhythmias and blocks are most common
Who requires admission for dig toxicity?
-symptomatic patients
-all patients treated with Fab
What beta blocking agent is most dangerous in OD?
Propranolol
What are beta 2 effects?
Vascular smooth muscle relaxation
Liver (glycogenolysis, gluconeogenesis)
bronchodilation
adipose tissue
uterine smooth muscle relaxation
When is the peak effect of normal-release beta blocker preparations observed?
1-4hours
Why is the bioavailability of PO vs IV beta blockers different?
First pass hepatic metabolism
What are the most common signs of Beta blocker OD?
-bradycardia
-hypotension
-unconsciousness
Why is propranolol much more toxic than other beta blockers?
Lipophilic nature and membrane stabilizing abilities allow it to penetrate the CNS
What is unique about labetalol?
It also blocks alpha adrenergic receptors
What is a main difference between dig and beta blocker OD?
Beta blocker has a more rapid onset
Why might multi dose activated charcoal be of use in Beta blocker OD?
Because these drugs undergo enterohepatic circulation
How is the order for whole bowel irrigation written?
Polyethylene glycol orally or via NG tube @ 1-2L/hour or 20cc/kg until clear effluent PR.
What is the first treatment of Beta blocker OD?
atropine, glucagon and crystalloid boluses
How is glucagon dosed?
5-10mg IV bolus followed by an infusion of 2-5mg/hour (diluted in D5W)
What are side effects of glucagon?
Nausea and vomiting
Hyperglycemia
Hypokalemia
Allergic reaction
What drugs can be used in Beta blocker OD with persistent hypotension post fluid bolus?
Isoprenaline
dopamine
epinephrine
Importantly - do not timidly titrate catecholamine infusions, much higher doses may be required than we are clinically used to
Should calcium be given in beta blocker OD?
Hyper/hypocalcemia can inhibit the action of glucagon therefore calcium should be given cautiously (infusion is safer than bolus)
What antidysrhythmic should be used in beta blocker toxicity with vtach and a pulse?
Lidocaine
(Class Ia and Ic agents should be avoided)
Should HD or HP be considered for Beta blocker toxicity?
It can be considered for certain Beta Blocker OD but given that Be blockers do not destroy tissues, if circulation can be supported, complete recovery can be expected
What is much more common in paediatric beta blocker OD?
Symptomatic hypoglycemia
What are RF for hypoglycaemia in Beta blocker OD?
Young age
Fasting state
Diabetes
How long after an ingestion should patients become symptomatic?`
normal release -> 6 hours
What is the treatment of beta blocker poisoning?
1) glucagon, atropine, fluids
2) infusion of glucagon, insulin-glucose, catecholamines, phosphodiesterase inhibitors, pacing, arterial line, swan gantz, consider HD
What are clinical applications of calcium channel blockers?
Angina pectoris
HTN
SVT
Hypertrophic Cardiomyopathy
Migraine Prophylaxis
How do calcium channel antagonists work?
They block slow calcium channels in the myocardium and vascular smooth muscle leading to coronary and peripheral vasodilation
Which CCA has the deadliest profile?
Verapamil (severe myocardial depression and peripheral vasodilation
What is the onset of action for CCA toxicity?
30-60min
What are the manifestations of Beta Blocker OD?
Bradycardia
Hypotension
Unconsciousness
Respiratory Arrest
Hypoglycemia
Seizures
Bronchospasm
VT or VF
Hyperkalemia
Hepatotoxicity
Should atropine be used in CCA OD?
It can be tried but often the effect is short lived
What are second line agents for bradycardia and hypotension in CCA OD?
cardiac pacing
isoproterenol
What is a reasonable dose of calcium in CCA OD?
6g
(10-20mL CaCl over 5-10 minutes then 5-10mL/hr)
children: 10-30mg/kg
What catecholamines have been reported to have success in CCA toxicity?
isoproterenol
dopamine
What other therapy (other than atropine, calcium and catecholamines) can be done for CCA OD?
insulin euglycemia
How do you write the order for insulin-euglycemia?
-insulin bolus 1U/kg, then insulin infusion 0.5U/kg/hr
-glucose 10-30g/hr (using 5 or 10% dextrose solution)
What is specific about nifedipine in children?
It is a medication that can kill a child with ingestion of a single tablet
What should be done in a patient who received nitrates after PDI type V ingestions (viagra/cialis etc)
IV fluids
If this is not enough then dopamine 5ug/kg/min
What do nitrites cause?
vasodilatation
methemoglobin formation
WHat are "poppers"
Inhaled alkyl nitrite in the hope of prolonging sexual pleasure
Who is particularly susceptible to the oxidative stress of nitrite exposure?
G6P-deficiency
At what levels of methemoglobin should you treat?
30%
How is methemoglobinemia treated?
methylene blue 1-2 mg IV over 5 minutes
What products are used in the clandestine production of methamphetamine?
Ammonium hydroxide
Anhydrous Lithium
Metallic Lithium
In what populations do we see caustic exposures?
intentional suicidal ingestions
Pediatric and elderly (accidental ingestion)
What pills are most likely to stick and therefore cause pill esophagitis?
Doxycycline
Tetracyclines
KCl
ASA
What factors influence the extent of injury from a caustic exposure?
Type of agent
Concentration
Volume
Viscosity
Duration of Contact
pH
Presence or absence of food in the stomach
What do acidic compounds cause?
coagulation necrosis
Why are acids less likely to cause esophageal or pharyngeal injury?
Squamous epithelial cells are somewhat resistant to coagulation necrosis
What does alkaline contact cause?
liquefaction necrosis
fat saponification
protein disruption
Why are alkali ingestions typically larger?
Alkalis are colourless, odorless and unlike acids, do to cause immediate pain on contact
What are the 4 steps of caustic ingestion?
1- necrosis
2- vascular thrombosis
3 - injured tissue begins to slough off
4 - granulation tissue forms
How are caustic injuries categorized?
1st/2nd/3rd degree, similar to thermal burns
What characterizes 1st/2nd and 3rd degree caustic burns?
1- edema and hyperemia
2a - non-circumferential
2b - near circumferential
(2nd degree is characterized by superficial ulvers, whitish membranes, exudate and friability)
3 - transmural involvement
What are the emergent issues in caustic ingestions?
airway edema
esophageal/gastric perforation
What clinical signs/symptoms are predictive of distal injury in caustic ingestions?
Prolonged drooling and dysphagia
What should be done immediately for hydrofluoric acid exposures?
Immediate cardiac monitoring (assess for prolonged QTc, torsades, ventricular dysrhythmia)
What is empirical intervention for life-threatening HF acid exposures with dysrhythmia?
High dose IV calcium
When is endoscopy contraindicated in caustic ingestions?
-possible or known perforation
->24 hours post ingestion (more hazardous because of wound softening)
What are the 4 categories of caustic ingestion based on endoscopy?
1) no esophageal/gastric injury
2) gastric injury
3) linear burns of esophagus
4) circumferential burns
When can water/mild dilution be considered?
Within the first few minutes after ingestion (in alert patients who are not vomiting and can tolerate liquids)
1-2 cups of milk or water can be given
When is surgical intervention required in caustic ingestions?
Free air
Peritonitis
Increasing chest or abdominal pain
hypotension
What should be done immediately for ocular alkali exposure?
Aggressive lavage with 2LNS per eye
What should be done in povidone-iondine ingestion?
Gastric irrigation with starch or milk (this converts the iodine to the much less toxic iodide)
What is the danger of phenol/formaldehyde ingestion?
They are protoplasmic poisons and cause protein denaturation and coagulation necrosis
What should be done in patients who ingest concentrated H2O2 and why?
Radiographic evaluation for the presence of gas in the chest/abdo/portal system because gas emboli can potentially be treated with HBOT
What is the treatment of ingested batteries?
Those lodged in the airway or esophagus require removal, while those that are gastric or intestinal are treated with watchful waiting.
What household products may contain methanol?
Antifreeze
Windshield washer fluid
Carburator fluid
Glass Cleaners
Illicit Alcohol Production
Formalin
Embalming fluid
How long does it take for Methanol to be absorbed?
30-60min
What is the smallest lethal dose of methanol in an adult?
15 mL of 40% methanol
What are the toxic metabolites of Methanol?
Methanol (alcohol dehydrogenase) -> formaldehyde (aldehyde dehydrogenase) formic acid
How is formic acid degraded?
Through a folate dependent pathway it is degraded to carbon dioxide and water
What are the main complications of methanol poisoning?
Optic neuropathy
Putaminal necrosis
What is the latency period b/w methanol ingestion and onset of visual or metabolic disturbances?
12-24 hours
(though may even be longer if concomitant EtOH ingestion)
What are early symptoms of methanol poisoning?
decreased MS, confusion, ataxia
What is the most common visual complaint in methanol poisoning?
central scotoma
What correlates with prognosis in methanol ingestion?
Degree of acidosis at the time of presentation and initiation of treatment within 8hours of exposure
Does the presence of a normal AG r/o methanol poisoning?
No
Apart from a wide AG what is another classic lab finding in Methanol toxicity?
Elevated osmolol gap
HOw do you calculate serum osmolality?
2Na+BUN+glucose+EtOH
What is the "normal" osmolal gap?
Often cited as <10mOsm/kg however there is considerable variability
Why should caution be used in ruling out a toxic alcohol ingestion with a normal osmolal gap?
-calculated osmalality may vary among laboratories
-in a later presentation there may be little or no osmolal gap because only the parent compound is osmotically active
-toxic levels of methanol or ethylene glycol may be present with a gap of 10mOsm/kg
What substances can cause elevated osmolal gaps?
Methanol
Ethylene glycol
Isopropanol
Ethanol
Acetone
Propylene glycol
mannitol
Glycerol
Ethyl Ether
What is a "double gap"?
Osmolal gap and anion gap
What substances may cause a "double gap"?
Ethanol
Methanol
Ethylene Glycol
Propylene Glycol
DKA
AKA
multiorgan failure
acetonitrile
chronic renal failure
What is an particular characteristic associated with methanol?
Ocular complaints
What is an particular characteristic of ethylene glycol?
Calcium oxalate crystaluria
What are sources of ethylene glycol?
Antifreeze
Airplane de-icing
Hydraulic brake fluid
Industrial solvents
How long does it take to reach peak blood levels of ethylene glycol after ingestion?
1-4 hours
What are the reported toxic and lethal doses of ethylene glycol?
0.2 mL/kg and 1.4 mL/kg
What is the metabolic pathway of ethylene glycol?
ethylene glycol (alcohol dehydrogenase) ->glycoaldehyde ->(aldehyde dehydrogenase) -> glycolic acid -> glycoxylic acid -> oxalic acid
What are the four stages of ethylene glycol ingestion?
1) acute neurologic - similar to inebriation
2) cardiopulmonary - HTN, tachycardia, ARDS, hypocalcemia
3) Renal Stages - ATN
4) Delayed Neurologic Sequelae - cranial neuropathy
Does ethylene glycol cause inebriation?
It causes the same degree of inebriation as EtOH
Does the absence of crystalluria r/o ethylene glycol ingestion?
No, it is a hallmark, less than 1/2 of patients have this finding
What shapes may the crystals in crystalluria take on?
Envelope-shaped
Needle - shaped
Any shape
Is urinary fluorescence useful in ethylene glycol toxicity?
No, a positive may be a false positive and a negative may be a false negative
What ECG changes can patients with ethylene glycol poisoning display?
QT prolongation secondary to hypocalcemia
Is Activated charcoal useful in toxic alcohol ingestion?
No
DO patients with inhalational exposure to Methanol need to go to the hospital?
Not necessarily
What are the treatment goals in patients with methanol or ethylene glycol toxicity?
-correction of metabolic acidosis with bicarbonate
-Alcohol dehydogenase blockade, inhibiting metabolism of methanol or ethylene glycol
-removal of the parent alcohol and its metabolites by HD
What should be considered when administering NaHCO3 to a patient with ethylene glycol ingestion?
The potential to worsen hypocalcemia
What are ways of blocking Alcoohol Dehydrogenase?
Antizol (fomepizole)
EtOH
At what level of methanol or ethylene glycol should alcohol dehydrogenase be blocked?
20mg/dL
What is the target blood Ethanol level to block ADH?
100-150mg/dL
What is the dose of fomepizole?
15mg/kg followed by 10mg/kg q12 hours x 4 and then re-assess
What are general indications for HD in a patient with toxic alcohol ingestion?
-metabolic acidosis
-renal compromise
-visual symptoms
-deterioration despite intensive care
-electrolyte imbalances unresponsive to conventional therapy
-blood level >50mg/dL
What else should be given to patients with methanol toxicity?
folinic acid
What else should be given to patients with ethylene glycol toxicity?
pyridoxine
thiamine
Does isopropanol cause CNS depression?
It causes twice the CNS depression of EtOH
What is the metabolism of isopropanol?
isopropanol (alcohol dehydrogenase)-> acetone
What is a potentially lethal dose of isopropanol
2-4cc/kg
What symptoms predominate with isopropanol ingestion?
CNS and GI
What is a marker of severe poisoning in isopropanol ingestion?
hypotension
Do patients with isopropanol ingestion have a wide AG?
No, acetone is uncharged
What is "pseudo renal failure"?
Isolated false elevation of Cr with normal BUN because of interference of acetone and acetoacetone in the calorimetric determination of Cr
What is a distinguishing feature of isopropanol ingestion?
Ketosis without acidosis (from acetone)
What can be done for isopropanol ingestion?
Dialysis
What are indications for HD in isopropanol ingestion?
hypotension despite treatment and coma
What are the 4 types of hydrocarbon exposures that present to the ED?
1) accidental ingestion involving children
2) intentional inhalational abuse of volatile HCs
3) accidental inhalational and dermal exposure to HC
4) massive oral ingestion
What do hydrocarbons contain?
Hydrogen and carbon
What are the 2 main categories of HC?
Aliphatic - straight chain
Aromatic - those containing a benzene ring
What target organs are affected by HC toxicity?
The lungs
The CNS
The heart
What characteristics determine the potential for acute toxicity of HCs?
Viscosity - substances with low viscosity have higher toxicity
Volatility - high volatility allows a substance to displace O2
Surface Tension - low surface tension allows a substance to disperse easily
Chemical side chains - increase the potential for toxicity
What is the primary organ of HC toxicity?
the lung
How does pulmonary toxicity occur in HC poisoning?
By aspiration
How do HCs affect the lung?
-penetrate the lower airways and cause bronchospasm and inflammation
-displace O2 in the alveolar space
-direct injury to alveoli and capillaries
-inhibit surfactant function leading to alveolar instability and collapse
What CNS symptoms do HCs cause?
Euphoria
Disinhibition
Confusion
Obtundation
What CNS abnormalities does chronic HC exposure cause?
-peripheral neuropathy
-cerebellar degeneration
-neuropsychiatric disorders
-chronic encephalopathy
-dementia
What cardiac abnormalities are caused by HC?
They may cause sudden cardiac death secondary to sensitization to endogenous and exogenous catecholamines
What are signs of significant HC exposure?
tachypnea, tachycardia, wheezing, hypoxemia
What poisonings may mimic the symptoms of HC aspiration?
organophosphate
salicylate
paraquat
Should routine GI decontamination of HC be done?
routine gastric lavage or emesis should be avoided (HC is much more toxic to the lung) There are some exceptions to this
In what situations post HC exposure is full decon indicated?
C- Camphor (seizures)
H - halogenated HC (dysrhythmia or hepatotoxicity)
A - aromatic HC (bone marrow suppression and CA)
M - Metals (arsenic, mercury, lead)
P - pesticides (cholinergic crisis, seizures, respiratory depression)
What meds should be avoided in HC intoxication?
Epinephrine + isoproterenol
How long should you observe asymptomatic HC ingestion?
6 hours
How long should patients with aspiration be observed
Minimum 24 hours
How long should you observe recreational HC exposure?
4-6 hrs
-offer drug addiction counseling
What are the most common iron exposures?
Ingestion of paediatric multivitamin formulations
What are normal serum iron levels?
50-150ug/dL
What is the total iron binding capacity (TIBC)?
A crude measure of the ability of serum proteins - including transferrin - to bind iron
What happens when iron levels rise following a significant iron OD?
Transferrin becomes saturated so that excess iron circulates as free iron - which is directly toxic to organs
What does the toxicity of an iron compound depend on?
The amount of elemental iron ingested
What is the risk stratification of iron ingestions?
<20mg/kg - no symptoms
20-60 - mild to moderate symptoms
>60mg/kg moderate to severe morbidity
What are the toxic effects of iron?
1-direct caustic injury to the gastrointestinal mucosa
2 - impairs cellular metabolism primarily of the heart, liver and CNS
What are the five stages of acute iron poisoning?
I - corrosive effects of iron on the gut - vomiting and diarrhea
II - apparent recovery - asymptomatic period
III - recurrence of GI symptoms, lethargy and coma, AG metabolic acidosis, leukocytosis, coagulopathy, renal failure, CV collapse
IV - fulminant hepatic failure - 2-5d (rare)
V - consequences of healing the injured gastric mucosa characterized by pyloric or proximal bowel scarring +/- obstruction
When does the serum iron level peak?
3-5 hours after ingestion except for sustained-release or enteric coated preparations
Why may levels measured late be deceptive?
Iron is rapidly cleared from the serum and deposited in the liver therefore levels may be deceptively low
Should TIBC be used as an indicator of free iron?
No, TIBC is a crude test and cases of serious toxicity have been reported even when TIBC exceeds the serum iron level
Should gastric decontamination be used in iron OD?
No, AC does not bind iron and lavage/ipecac do not remove tablets
What method of decon is recommended for iron OD?
>20mg/kg -> WBI
Peg-lyte solution oral or NG 20-40cc/kg/hour or 1.5-2L/hr until rectal effluent is clear and no radiographic evidence of pill fragments
When is WBI contraindicated?
Bowel obstruction, perforation or ileus
Can HD or HP be used for iron OD?
No, exchange transfusion has been recommended for severely symptomatic patients with iron >1000ug/dL
What does deferoxamine do?
It chelates iron to form the water soluble compound ferrioxamine which can be renal excreted or dialyzed
What does deferoxamine do to the urine?
turns it a "vin rose" color
Which patients should receive WBI?
-ingested >20mg/kg
-has pills visible on abdominal radiograph
When should serum iron levels be checked?
3-5 hours after ingestion and 6-8 hours after ingestion to confirm the level is decreasing
What is the management of a patient with peak serum iron levels of </=300ug/dL?
If asymptomatic d/c home after 6 hours of observation
Which patients require chelation for Iron OD?
serum iron >500ug/dL
-systemic signs of toxicity (AMS, shock, AG metabolic acidosis)
What tests are 100% specific for predicting serum iron >300ug/dL?
increased serum glucose and leukocyte count
What are potential sources of lead?
household paint
gasoline
fushing weights
pottery glaze
bullets in bone
bootleg whiskey ("moonshine")
What is the mechanism by which lead is toxic?
Lead binds sulfhydryl groups and other ligands and interferes with critical enzymes
What systems are most affected by lead toxicity?
hematopoietic
neurologic
renal
GI
What is the manifestation of hematopoietic toxicity in lead poisoning?
Anemia
What are neurologic signs of lead toxicity?
Wrist drop, foot drop
segmental demyelination and degeneration of motor axons
acute toxicity may result in cerebral edema
What are symptoms of acute lead exposure?
"lead colic" cramping abdominal pain, nausea, vomiting, constipation +/- diarrhea, fatigue, anemia, peripheral neuropathy, headache -> cerebral edema
What is the most important biomarker in lead toxicity?
Blood lead level (BLL)
What is a chronic toxic level of BLL in a child?
10ug/dL
What are the findings of lead toxicity on a peripheral smear?
basophilic stipling
What are "lead lines"?
Increased metaphyseal activity on wrist and knee radiographs that are characteristic of chronic lead exposure
What form of decontamination can be used for lead?
WBI
Which patients with lead expusure should be considered for chelation?
BLL>70ug/dL
signs suggestive of encephalopathy
which initial chelator is used for lead exposure?
BAL (british antilewisite)/dimercaprol
Which chelator is used after dimercaprol?
CaNa2EDTA (NOT Na EDTA)
What is the key to managing chronic lead toxicity?
Identify and reduction of sources of primary exposure
Should ingested lead FB be removed?
THey can be allowed to pass but if still there in 2 weeks should be removed
Where is arsenic found?
Smelters, electric power plants than burn arsenic and rich coal, wood preservative, glass production
What are the inorganic forms of arsenic and their properties?
As3+ - highly lipid soluble, 5-10x more toxic
As5+
Where does arsenic concentrate?
in the liver, kidneys, spleen, lungs and GI tract
Why is arsenic toxic?
It bind sulfhydryl groups inhibiting critical enzymes (especially those involved in glycolysis)
-disrupts oxidative phosphorylation
-arsine causes hemolysis
What are the manifestations of exposure to arsine gas?
Severe hemolysis and renal tubular injury
What are the predominant effects of exposure to arsenic salts?
acute GI effects
metallic/garlicky taste in mouth
later - encephalopathy, seizures, coma
What are the chronic effects of arsenic poisoning?
Mees lines on the nails
sensorimotor neuropathy
hyperkeratosis on the palms and soles
What is the best way to diagnose arsenic poisoning?
24 hour urine collection (abnormal is >50ug/L)
Which substance can cause false positive urinary arsenic?
Arsenobetaine found in seafood
What decontamination should be considered in arsenic poisoning?
-orogastric lavage and WBI
(AC does not adsorb)
-HD removes arsernic in ARF
-exchange or plasma exchange should be considered very early after exposure
What is the preferred chelator in arsenic poisoning?
IM dimercaprol
Is chelation useful for arsine?
No
What are the forms of mercury?
elemental, salts, organic
What industries use mercury in manufacturing?
-fluorescent lights
-batteries
-polyvinyl choloride
-latex paint
What is a common root of elemental mercury exposure and what can it cause?
Inhalational - severe pneumonitis and ARDS
Is ingestion of elemental mercury dangerous?
No, it is not absorbed by the GI trace, unless it becomes trapped in a diverticulae
What happens with ingestion of inorganic mercury? (mercurous or mercuric)
Significant GI and renal toxicity
What are the manifestations of organic mercury compounds?
Primarily through ingestion
Neurotoxicity: ataxia, tremor, dysarthria, tunnel vision
What is the timeline of symptoms of exposure to elemental, inorganic and organic mercury?
Elemental - inhalation - rapid
Inorganic salt ingestion - immediate
Organic - symptoms over weeks to months
What is the most helpful test for confirming exposure to mercury?
urine mercury
Is charcoal useful in mercury salt ingestion?
No, it adsorbs very little
When should BAL be used in mercury toxicity and when should it not be used?
It should be used in acute inorganic poisoning and it is contraindicated in patients poisoned with organic mercury
When do peak plasma concentrations occur in immediate release lithium ingestion? delayed-release?
immediate release 0.5-3 hours
delayed release - 6-12 hours
What are the kinetics of lithium distribution?
Two - compartment -> initially extracellular then redistributes to various tissues
What medications may increase lithium levels?
NSAID
diuretics
ACE Inhibitor
What problems are associated with chronic lithium therapy? and what are their manifestations?
Nephrogenic diabetes which manifests as dehydration and hyponatremia
-inhibits the synthesis of thyroid hormone as hypothyroidism
-leukocytosis (temporary)
What are the classifications of lithium poisoning?
-acute toxicity
-chronic toxicity
-acute on chronic
What is the definition of acute lithium toxicity?
An OD of lithium in a patient without any lithium body stores
what are the manifestations of acute Li+ toxicity?
GI symptoms
ECG changes - bradycardia, T wave flattening and QT prolongation
-delayed neurotoxicity
How does chronic Li+ toxicity occur?
Increased absorption
decreased renal elimination
What is the predominant presentation of chronic lithium toxicity?
tremor-> progressive drowsiness, hyper-reflexia, confusion, clonus, coma, seizures, EPS
What is SILENT syndrome?
Syndrome of irreversible lithium effectuated neurotoxicity
What is an acute on chronic lithium toxicity?
excess Li ingestion in a patient already taking Lithium
What are life-threatening syndromes that can be associated with Lithium
NMS
Serotonin syndrome
What are the cornerstones in management of Li+ poisoning
Selective GI decontamination
Enhanced renal and extracoporeal elimination
What form of GI decon is useful in Li and when?
WBI for sustained release Li
How do you maximize renal elimination of Li?
Fluid resuscitation with NaCl
IV NS at 2x maintenance - unless contraindicated
What is the most effective technique for elimination of Li?
HD
When is HD used in Li tox?
Clinical deterioration
-inadequate endogenous Li clearance
-inabiliyt to enhance renal elimination
->4.0mEq/L in acute
->2.5 mEq/L in chronic
What are the elements of a toxicology risk assessment?
Obtain all prescription bottles and containers
Inquire about OD history
Perform a pill count
How reliable is the pupillary examination in a poisoned patient?
The pupillary exam in the poisoned patient may give misleading information, it is especially less reliable in multiple drug ingestions
Which opioids do not produce miosis?
Propoxyphene and pentazocin
Which toxins give a shoe polish door?
nitrobenzene
Which toxins give a garlic odor?
Arsenic
DMSO
organophosphates
yellow phosphorus
selenium
tellurium
What toxins give rotten egg odor?
disulfiram
hydrogen sulfide
NAC
DMSA
What toxins give off pear odor?
chloral hydrate
paraldehyde
What toxins give off glue odor?
toluene
other solvents
Give circumstances where physostigmine is indicated?
Physostigmine is a naturally occurring acetylcholinesterase inhibitor It is indicad in anticholinergic poisoning syndrome (in the absence of TCA OD)
Mild temperature elevation
acute delirium with mumbling speech
typical "picking" movements
How do you give physostigmine?
It is given as 1-2 mg IV over 5 min (may be repeated in 10-15min)
What is Takutsubo cardiomyopathy? How does it present? How is it treated?
Takutsobo cardiomyopathy is a transient cardiac syndrome that results in left ventricular apical a kinesis and mimics ACS. It presents as chest pain, ST segment elevation and elevation of cardiac enzymes with no significant coronary stenosis on angiogram. Patients are treated as if they are having an ACS
What is the critical step in managing a patient with excited delirium?
Excited delerium is an extreme presentation of sympathomimetic excess. Patients are agitated hyperthermic, violent and possess "superhuman" strength.
It is critical to sedate and control hyperthermia aggressively. Also there is a need to treat the acidosis and hyperkalemia.
What is the first hallmark of a severe sedative overdose?
Depressed sensorium. Then the patient becomes increasingly obtunded, the deep tendon reflexes diminish and vital signs deteriorate.
When is a CT head indicated in a poisoned patient?
In a comatose patient if there is suspicion for stroke, infection or head trauma
Which receptors are involved in producing serotonin toxicity?
5HT1A
5HT2A
List 3 medications that can be used to alleviate serotonin toxicity? How are they given?
Cyproheptadine 4mg PO prn - liquid given through an NG tube
Methysergide
Benzodiazepines - IV
Activated charcoal may be considered PO
IV NaHCO3
Which SSRI give ECG abnormalities? What is the abnormality?
Citalopram
QTc prolongation, QRS widening and ventricular fibrillation
In which patients is it indicated to request a comprehensive drug screen?
Patients presenting with their first major psychotic episode
patients who are critically ill for unknown reasons when identification of an unsuspected toxin may change management
What is the value of a urine tox screen?
The urine tox screen has little value because
1)the lab does not screen for many substances, even commonly ingested agents that are capable of causing critical illness
2) the urine screen is often performed soon after the ingestion when the drug concentration is too low for a positive result. Even the drug responsible for life-threatening symptoms may be negative on the urine screen soon after ingestion.
3) drugs found on screening may not be those responsible for the initial symptoms, especially if the drugs are not quantified.
Which toxins are commonly associated with non cardiogenic pulmonary edema
Opioid
Salicylate
Which classes of toxins are radio-opaque?
Heavy metals
Phenothiazines
Potassium
Calcium
Chlorinated hydrocarbons (chloral hydrate)
What are two formulations of activated charcoal? Which one is no longer recommended?
Oral slurry
AC with added sorbitol

AC with added sorbitol is no longer recommended but cathartics have never been shown to be of benefit and repeated doses of sorbitol can cause dehydration
What agents do not adsorb to charcoal?
Ions (acids, alkalies, lithium, borate, bromide)
hydrocarbons
metals (iron)
ethanol
What is the proper decontamination for chemical gas exposure?
Exposure to a gas does not require decontamination because the patient and rescuers are not at risk once the patient is removed from the toxic environment. The exception is when the patient's skin or clothing is contaminated with a liquid that is evaporating.

If this is the case, remove all clothing as soon as possible. Skin should be irrigated with warm watery and with attention to skin folds and other areas that might be missed.
List medications containing APAP that are available over the counter?
benadryl
dayquil
dimetapp
dristan
excedrin
midol
nyquil
robitussin
sinutab
sudafed
vicks
What is the toxic dose of APAP?
150mg/kg
Describe the metabolism of acetaminophen?
Conjugation with glucuronide or sulfate or oxidation by CYP4502E1 to NAPQI which rapidly combines with glutathione to form non-toxic metabolites excreted in the urine
In which organs is APAP metabolized?
Primarily the liver but may also be metabolized by the renal CYP enzymes
What are the characteristic histology findings with APAP hepatotoxicity? Why?
Centrilobular necrosis (hepatic zone III) because this is where oxidative metabolism is concentrated. In severe toxicity, the entire liver parenchyma may be affected.
List the mechanism of action of NAC?
It is:
glutathione precursor
sulfur containing glutathione substitute
enhances APAP conjugation with sulfate
free- radical scavenger
antioxidant
Describe the clinical course of an APAP toxicity?
Stage 1 -preinjury - nausea, vomiting, anorexia (0-12)
Stage 2 - liver injury - nausea, vomining, RUQ pain (8-36 hours)
Stage 3 - maximal liver injury - liver failure (2-4 days)
Stage 4 - recovery (>4 days)
List the different types of APAP exposure and their management?
Acute and chronic.
How is APAP overdose managed in the pregnant patient?
Same as other individuals
What is the risk of liver injury if NAC is given within 8 hours?
<4%, close to 0
When do NAC reaction occur? How are they treated? WHat is the pathophysiological mechanism?
IV NAC - anaphylactoid section within the first 15-60 min
PO NAC - anaphylactoid reaction (less frequent than IV)

They are treated with antihistamines +/- slowing or stopping the infusion. You can also give glucocorticoids or epinephrine for more severe reactions
When can NAC be stopped?
21hours after IV admin
72 hours after PO admin
AND APAP <10ug/mL, AST normal
When do patients with APAP induced hepatotoxicity need to be referred to a herpetology centre?
Established hepatotoxicity and at risk of fulminant hepatic failure.
Describe the mechanism of action of ASA?
ASA is converted to salizylate. Salicylate stimulates the respiratory center and increases the sensitivity of the respiratory centre to pH and carbon dioxide partial pressure.
Toxicity results primarily from interference with aerobic metabolism by uncoupling of mitochondrial oxidative phosphorylation. Salicyate is also a potent neurotoxin
Contrast first-order vs zero-order kinetics
First order kinetics depend on the concentration of the drug, whereas zero order kinetics are independent of the drug concentration
At which serum concentration of ASA are zero order kinetics reached?
Serum concentrations grater than 30mg/dL
What is the mechanism of ASA induced hyperthermia?
Inefficiency of anaerobic metabolism results in less energy being used to create ATP and energy is released as heat causing hyperthermia
List 3 mechanisms contributing to the hypokalemia seen in ASA toxicity?
Vomiting secondary to stimulation of the CTZ
increased renal excretion of Na, HCO3 and K in response to respiratory alkalosis
Salicylate induced increased permeability of renal tubules
Intracellular accumulation of sodium and water
Inhibition of the active transport system
Which patients are more at risk of developing ASA induced NCPE?
Adults
->30year olds
-cigarette smokers
-chronic salicylate ingestions
-metabolic acidosis
-neurologic symptoms
-serum salicylate >40mg/dL

Children
-high serum salicylate level
-high anion gap
-decreased serum potassium
-low pco2
How many tablets of ASA are needed for a 70 kg patient to reach a toxic dose? A lethal dose?
Toxic - 175 80mg tabs
Lethal - 475 80mg tabs
How are seizures treated?
HD
What are indications for HD in ASA toxicity?
-coma/seizures
-renal/hepatic/pulmonary failure
-severe acid-base imbalance
-deterioration of condition
-serum salicylate level >100mg/dL in acute OD or >40 mg/dL in chronic OD
What is the role of activated charcoal in ASA OD?
There is little role for AC in salicylate OD, it can limit the absorption of salicylates if given within 1 hour but has not been shown to alter the clinical outcome or morbidity
Why is dextrose given to these patients?
Tissue glycolysis (due to increased metabolic rate, temperature, tissue CO2 production and O2 consumption) results in hypoglycaemia. Rarely patient may display hyperglycemia
How is urinary alkalization performed?
Administer 1-2mEq/kg sodium bicarbonate over 1-2hours and then follow the urine pH which should ideally be maintained at a pH of 7.5-8. This should not come at the cost of alkalemia
What are the goals of alkalization?
increase the fraction of salicylate ion in the urine, thereby trapping it there and resulting in its excretion
How is ASA toxicity treated in the pregnant patient? In the postpartum period?
ASA toxicity should be treated the same way. If the fetus is viable, the fetus should be delivered since salicylate poisoning may result in fetal demise. In infants and congenital salicylism exchange transfusion may be considered
What is the definition of an illusion?
A misperception of a real object (as opposed to a hallucination)
What are serotonin-like agents?
lysergic acid
tryptamines
What is the most common form of LSD?
A blotter which is placed sublingually or eaten whole
What is an example of a tryptamine?
Psilocybin and psilocin found in mushrooms
What receptors do serotonergic agents act on?
5-HT2A
What is the most common adverse reaction to psychedelics?
Acute panic reaction (paranoid delusions and fear of impending death)
What is the mainstay of treatment for serotonergic agents?
supportive
sedation
What are enactogens?
Hallucinogenic stimulants that are analogues of amphetamines, mescaline, N-substituted piperazines
What are complications of MDMA?
Rhabdomyolysis
Hyperthermia
Hyponatremia
What is the mechanism of action of enactogens like MDMA?
Release serotonin, dopamine and NE from presynaptic terminal and inhibit catecholamine reuptake
What are the mechanisms responsible for hyponatremia in MDMA/
-excess intake of free water
-MDMA mediated release of antidiuretic hormone
What are dissociative agents?
PCP
Ketamine
Dextromethorphan
What are the characteristics of dissociative agents?
Analgesic
Amnestic
Lack of respiratory or cardiovascular depression
What substances may cross react with the PCP urine assay?
Dextromethorphan
Chlorpromazine
Methadone
Ketamine
Diphenhydramine
What lab abnormality can be caused by chronic dextromethorphan use?
Hyperchloremia with negative anion gap (caused but bromide interference with the assay)
What is the main active ingredient in cannabinoids?
Delta-9-THC (tetrahydrocannabinol)
What are the most common effects from smoking marijuana?
Relaxation and euphoria
What is the difference between an opioid and opiate?
Opioid refers to all natural, synthetic and semi-synthetic agents with morphine-like actions.
Opiate refers only to natural agents
What does the term endorphin refer to ?
enkephalin, beta endorphin and dynorphin (any of the 3 endogenous opioid families)
What are the opioid receptors
mu
kappa
delta
What opioids can cause seizures?
propoxyphene
meperidine (its metabolite)
hypoxia (with OD of any opioid)
What is the toxidrome of opioid toxicity?
hypoxia from CNS and respiratory depression
Which opioids have serotonergic properties?
meperidine
dextromethorphan
Which opioids have may not produce miosis?
meperidine
propoxyphene
lomotil
pentazocin
What cardiovascular problems can opioids cause?
hypotension (histamine release)
bradycardia
propoxyphene may cause sodium channel blockade
What opioids can cause seizures?
propoxyphene
meperidine (its metabolite)
hypoxia (with OD of any opioid)
What is the toxidrome of opioid toxicity?
hypoxia from CNS and respiratory depression
Which opioids have serotonergic properties?
meperidine
dextromethorphan
Which opioids have may not produce miosis?
meperidine
propoxyphene
lomotil
pentazocin
What cardiovascular problems can opioids cause?
hypotension (histamine release)
bradycardia
propoxyphene may cause sodium channel blockade
What is "cotton fever"?
A benign fever caused by cotton used to strain drug and remove particulates
What are signs of opioid withdrawal?
CNS excitation
tachypnea
mydriasis
Is opioid withdrawal life-threatening?
No
What commonly used opioid is missed on urine screens?
fentanyl
What can be done to hasten the passage of packets from body packers?
WBI
WHat route of administration cannot be used for naloxone?
PO
What is the duration of action of naloxone?
1-2 hours
What is the dose for a naloxone infusion?
2/3 the effective initial dose/hr
What agent is used for opioid withdrawal? How does it work?
Clonidine
(a central alpha agonist)
It suppresses sympathetic hyperactivity
How long should patients be observed if they receive naloxone?
2 hours
What is special about patients who have ingested and OD of lomotil?
They should be observed even if asymptomatic
What is the mechanism of action of barbiturates?
They enhance the activity of GABA and depress the activity of all excitable cells.
Barbituates have a separate binding site on GABA channels
What are the effects of barbiturates?
Depressive effects
respiratory depression
decreased pulse and BP
How are barbiturates classified?
1) Ultra short acting (imm onset with IV)
2) Short acting (10-15 min onset)
3) intermediate acting (45-60 min onset
4) Long acting (onset 1hour)
How can elimination of phenobarbital be enhanced? Can this be used with other barbiturates?
Alkalinization (this increases the amount of drug present in ionized form minimizing tubular absorption)
No, this cannot be used with other barbituates
What are ultrashort acting barbiturates?
Methohexital
Thiopental
What is the life threat in severe barbiturate toxicity?
respiratory depression
Is hypoventilation always apparent in barbiturate intoxication?
No, respirations can be shallow and rapid. Pulse oximetry or capnography will detect respiratory compromise
What are symptoms of barb withdrawal?
Tremors
Hallucination
Seizures
Delirium
Do barb levels other than phenobarbital correlate with toxicity?
No, barbs have a high VD therefore serum levels do not accurately reflect CNS concentrations or correlate with clinical severity
What can occur to EEGs in the presence of barbs?
EEG may be silent as a result of barb OD therefore no patient should be declared brain dead if barbs are present in therapeutic levels
What GI decon can be considered in phenobarb OD?
MDAC 25g q 2hours
Why are BZD safer than other sedative hypnotics?
Cardiac effects and fatalities are rare
Respiratory depression is less pronounced than with barbs
drug drug reactions are uncommon
How do BZD work?
They enhance the inhibitory actions of GABA by binding to a BZD receptor. They decrease the ability of the nerve cell to initiate an action potential
Which BZDs have predictable IM absorption?
Lorazepam
Midazolam
Which BZD have erratic IM absorption?
Diazepam
Chlordiazepoxide
Where are BZD metabolized?
In the liver
Which BZDs have no active metabolites?
Oxazepam
Temazepam
Lorazepam
What are clinical features of BZD ingestion?
CNS depression
respiratory depression (large oral OD or IV admin)
What is the most common sign of BZD toxicity?
ataxia
What is a contraindication to flumazenil
Any possibility of TCA OD
What complications can result from flumazenil?
Seizures
Cardiac Dysrhythmias
When is flumazenil especially hazardous?
Pt habituated to BZD
When seizure-causing drugs (such as cocaine and TCA) have been ingested
What are indications for flumazenil use?
Isolated BZD OD in non-habituated user (accidental paediatric exposure)
Reversal of conscious sedation
Which patients are at risk of BZD withdrawal?
Continuous treatment for >4months
What is a potential date rape drug easily obtainable outside the US?
Flunitrazepam
What is buspirone?
A non-BZD agent used for generalized anxiety
Are zaleplon and zolpidem detected on urine drug screen?
no
Where do zolpidem and zaleplon act?
At a specific BZD receptor
What are the signs of chloral hydrate toxicity?
CNS and respiratory depression
Gastrointestinal irritation
CV instability an dysrhythmia
What is the treatment chloral hydrate induced ectopy?
propranolol
What are the only drugs found in non-prescription hypnotics?
Diphenhydramine
Doxylamine
What is the most frequent finding with diphenhydramine OD?
Impaired consciousness
What may occur with severe diphenhydramine toxicity?
Seizures
Rhabdomyolysis
What is a medical use for GHB
Narcolepsy
What is characteristic of GHB intox?
Periods of agitation alternating with periods of decreased LOC
In the absence of EtOH how long does it take for patients to awaken after GHB use? Otherwise, after what time interval should most patients with GHB ingestion awaken?
3-4 hours

8 hours
Why should intubation be considered with GHB?
High incidence of emesis
Is there a withdrawal syndrome with GHB?
YEs
How is GHB withdrawal treated?
High dose BZD
Barbiturates (if severe)
Is there delayed toxicity with GHB?
No
What are symptoms of GHB withdrawal?
Anxiety
Tremor
Insomnia
Delirium
Autonomic instability
What is the typical presentation of BZD OD?
Coma with normal vital signs
What is the starting dose of naloxone that should be given to a patient with opioid OD who is at risk of withdrawal?
0.04mg
In the context of caustic ingestion, which patients should not receive steroids?
Those at risk of mediastinitis
What are some of the subtle clinical signs that can help differentiate between beta blocker toxicity and CCB toxicity?
Beta Blockers are often associated with AMS, normal glucose (hypoglycaemia in children) and high degree AV blocks

CCB - normal mental status, hyperglycaemia (precent insulin release from the pancreas), complete AV block
What is the clinical picture of clonidine OD?
Opioid toxidrome
Datura stramonium?
Jimson weed
anti-cholinergic
What is an indication of severe snake envenomation?
metallic taste in the mouth
What are less commonly known sources of CO?
Banked blood
methylene chloride (converted to CO by CYP 2E1 in the liver)
inhalational anesthetics
Which metabolite of cocaine is atherogenic?
benzylechinine
Can you use a VBG for co-oximetry?
Yes
What are the lesions on CT scan most commonly found in CO poisoning?
bilateral lesions of the globus pallidus
What % of patients with ethylene glycol ingestion have calcium oxalate crystals in the urine?
50%
What is the classic shape of a calcium oxalate crystal?
Envelope but they can be any other shape i.e.. needles
What can result in a negative CO co-oximetry?
hydroxycobalamin
Which patients may have a negative reaction to methylene blue?
Those with G6PD deficiency
What is the treatment for sulfhemoglobin?
Remove the offending agent
x-change transfusion
HBO?
how do sulfonylureas work?
They close potassium channels on pancreatic cells and bring the cell to depolarization sooner
Which patients do not have large glycogen stores?
children
alcoholics
cirrhotics
Can you become hypoglycaemic while fasting on a sulfonylurea?
No, you should not become hypoglycaemic, if this happens you should check a creatinine because sulfonylureas are renal cleared.
What makes an animal poisonous?
If it has various toxins distributed in its tissues
What makes an animal venomous?
If it possess specific glands for producing venom connected to an apparatus for delivering that venom to another animal
What are life-threatening injuries related to venomous animals?
Venomous snake bites
Black widow spider bites
Certain marine animal envenomations
Anaphylactic reactions to insect stings
What % of snakes species are venomous?
10-15%
Where do the majority of snake bites occur (anatomically)?
97% in the extremities
2/3 upper extremity
1/3 lower extremity
What is the difference between a legitimate and an illegitimate snake bite?
legitimate - occurring accidentally
illegitimate - occurring when attempting to handle or disturb a snake
What are the 5 venomous families of snakes?
colubriae
hydrophiidae
elapidae
viperidae
crotalidae
What snakes make up the crotalidae family?
pit vipers
What is the most prevalent snake in the US?
pit vipers (98% of all venomous snake bites)
What are the 3 main groups of pit vipers?
rattlesnakes
copperheads
moccasins
What snakes make up the elapidae family?
cobras
kraits
mambas
coral snakes
What characterizes a coral snake?
The nose of the coral snake is black
The red and yellow bands are adjacent on the coral snake (they are separated by black on king snakes)
What is a consistent way to identify pit vipers?
The presence of a heat sensitive pit midway between the eye and the nostril on both sides of the head
What are other characteristics used to identify venomous snakes?
-triangular shaped head
-the presence of an elliptic pupil
-the arrangement of sub caudal plates
-the tail structure
-the presence of fangs
What are the 2 venomous lizards?
the gila monster
the mexican beaded lizard
What are the 2 predominant clinical responses to snake venom?
-neurotoxic
-hematotoxic
in general which snake species venoms have predominantly systemic effects? local effects?
systemic - elapidae, hydrophiidae
local - colubridae, viperidae, crotalidae
What is a dry bite?
A bite where no venom is injected
What systemic problems can be caused by envenomation?
DIC
pulmonary edema
shock
anaphylaxis
How do pit viper envenomations cause death?
disruption of coagulation
increased capillary membrane permeability
How do coral snakes cause death?
Compounds bock neuromuscular transmission at the ACh receptor causing direct inhibition on cardiac and skeletal muscle therefore death occurs by respiratory failure
What is the best first aid for a snakebite?
rapid transportation to a medical facility
What can be done to limit the spread of venom?
Calm the victim
immobilize the bitten area
warp in an elastic bandage to collapse the lymphatics and superficial veins
What is the only proven therapy for snake envenomation?
antivenin
What lab tests should be done in a snake bite?
cbc
coagulation profile
fibrinogen
fibrin split products
SMA-7
type and crossmatch 4Units
What is the grading of snake envenomation?
Grade O - IV
0 minimal no evidence of envenomation
I - minimal envenomation
II moderate envenomation
III - severe envenomation
IV - very severe envenomation
Who is a candidate for antivenin?
Any victim of a venomous snake bite with moderate to severe envenomation
What precautions must be taken when administering antivenin?
prepare for possible anaphylaxis
What is fab AV
polyvalent antivenin less likely to produce allergic reactions
What is appropriate wound care for snake envenomations?
-cleansing
-immobilization
-elevation at or above the heart
-tetanus
-constricting band
How do arthropod fatalities mostly occur?
From an autopharmacologic response
What arthropods make up the hymenoptera family?
bees
wasps
hornets
yellow jackets
ants
In what time frame do most serious reactions to bee stings occur?
30 min
What are "killer bees"
More aggressive bees (they are not more)
What does a black widow spider look like?
Glossy black with bright red markings on the abdomen which may have an hourglass shape
How long should a patient receiving dpi be watched?
24hours
What ingredient of the black widow venom is most dangerous
neurotoxin
What are the clinical features of BW spider bite?
pinprick sensation with local swelling and redness
crampy pain especially in the chest or abdomen which may last several hours to days
What is the management of BW spider bites?
Ice pack
transport to hospital
cleanse wound
tetanus
symptomatic treatment with diazepam and analgesia
consider antivenin especially in paediatric, pregnant and elderly
The antivenin is derived from horse serum
What is the most distinctive feature of the brown recluse spider?
Violin shaped area on the cephalothorax
What are the most concerning effects of brown recluse spider bites?
local tissue destruction
What is the most common mimic of loxosceles (brown recluse spider)?
MRSA skin infection
What species of scorpions is dangerous?
centuroides
What is the predominant characteristic of centuroides toxin?
neurotoxin
Is there scorpion antivenin?
yes
What should you think of in a patient presenting with ascending paralysis?
Tick paralysis
What is the most toxic (coelenterate) jelly fish?
box jelly fish
Which venomous snake bites should be treated empirically if there is a safe anti venom?
elapidae (because the venom is neurotoxic)
What type of decon should be done for theophylline
AC - it acts as gut dialysis by drawing drug out of the plasma
What is the treatment for a hypotensive patient with theophylline OD?
B blocker why?
What toxicity presents with "snowfield vision"?
Formic acid
What does 50% ethylene glycol mean
50g/100mL
What is the simplified mnemonic for AG metabolic acidosis
K - ketones
U - uremia
L - lactate
T - toxins
S - salicylate
How are organophosphates absorbed?
Derma, GI, Respiratory
How do organophosphates work?
They inhibit the enzyme acetylcholinesterase which results in accumulation and prolonged effect of acetylcholine
What is the classical syndrome of muscarinic acetylcholinesterase inhibition
Salivation
Lacrimation
Urination
Defectaion
GI cramps
Emesis
What happens at the neuromuscular junction with excess acetylcholine?
Hyperstimulation of the muscles with secondary paralysis, the diaphragm becomes affected and respiratory arrest ensues
What is the timeline of organophosphate poisoning?
May be acute or chronic
What are mechanisms for early morbidity and mortality in acetylcholinesterase inhibitor poisoning?
Seizures
Pulmonary hypersecretion/bronchorrhea
Bronchospasm
What is the ultimate cause of morbidity and mortality in AChE Inhibition?
Skeletal muscle hyperactivation and subsequent paralysis from nicotinic hyperstimulation
What is "aging"?
The irreversible conformational change that occurs when an organophosphorus agent is bound to the cholinesterase enzyme for a prolonged time
On average, how long does aging take?
48 hours
How is exposure to cholinesterase inhibitors confirmed?
Plasma and erythrocyte cholinesterase levels.
What are the goals in management of AChI tox?
Decontamination
Supportive Care
Reversal of ACh excess at muscarinic sites
reversal of toxin binding on cholinesterase molecule
What protective clothing should be used by HCP in organophosphorus poisoning?
Universal precautions
Nitrile or butyl rubber gloves
eye shields
protective clothing
Which agent should be used for neuro muscarinic blockade in RSI?
Rocuronium (succinylcholine may have a prolonged duration)
Why do we treat with atropine?
It reverses the clinical effects of cholinergic excess at parasympathetic end organs and sweat glands
What is the endpoint for atropine administration?
Control of mucous membrane hyper secretion
Clear airways
What do oximes do?
They restore cholinesterase activity
What are indications for oxime therapy?
Respiratory depression/apnea
Fasciculation
Seizures
Arrhythmias
CV instability
Large amounts of atropine (> 2-4mg atropine)
What is the treatment for agitation, seizures, coma?
BZD after the airway has been secured
What are differences between nerve agents and organophosphates?
The nerve agents tend to age very quickly therefore reversal is very time sensitive. These agents do not require large doses of atropine but do require pralidoxime
What is IMS?
Intermediate syndrome that consists of proximal muscle and respiratory muscle weakness.
How are carbamates different from organophosphates?
-Much shorter duration
-carbamate-cholinesterase binding is reversible
Should oximes be used in the management of carbamate toxicity?
If unsure, you should use it
What are chlorinated hydrocarbons used for?
DDT - insecticides
Lindane - scabies
What are the predominant symptoms of chlorinated hydrocarbon pesticides?
Neurotoxicity - tremor, parethesia, seizure
Vent dysrhythmias (halogenated)
How should seizures be controlled in chlorinated hydrocarbon exposure?
BZD or barbs
What are substituted phenols?
insecticides
termiticides
wood preservative
weight reduction agents
How do substituted phenols work?
they uncouple oxidative phosphorylation
How do patients with phenol toxicity present?
Hypermetabolic
Hyperthermic
hypovolemic
renal and hepatic injury
rhadomyolysis
yellow staining of skin at the site of absorption
What is agent orange?
chlorphenoxy pesticide used during the Vietnam war
What is the target organ of chlorphenoxy poisoning?
The skeletal muscle
What is specific about paraquat and diquat?
They are extremely corrosive and patients may die of esophageal perforation prior to developing the characteristic pulmonary injury
What is the classic finding in paraquat?
progressive pulmonary injury over 1-3 weeks
What is the key to successful treatment of acute paraquat exposure?
Early decontamination
Consider gastric lavage or AC
Charcoal hemoperfusion vs HD and HP
What are pyrethrins?
Naturally occurring insecticides of the yellow chrysanthemum
What is the most likely route of exposure with pyrethrins?
inhalation
What are clinical manifestations of pyrethrins?
Allergic manifestations
Local irritation
Sodium channel and GABA mediated chloride channel effects -> neurologic signs and symptoms
What are the clinical effects of DEET?
Contact dermatitis -> skin blisters (chronic LFT and neuro abnormalities)
How does capsaicin work?
Depletes nerve terminals of substance P causing a local inflammatory response, swelling, exudation and pain
Where do most capsaicin exposures occur?
pepper spray
causing chemical conjunctivitis +/- keratitis (rarely inhalation causes pulmonary edema and possibly ARDS)
What is the treatment for pepper spray?
irrigation (eyes) and analgesics
What is one of the most commonly reported plant fatalities in the US?
water hemolck
What properties does datura stramonium have?
anticholinergic
jimson weed
In what population do most plant exposures occur?
children<6years
What can occur if capsaicin is inhaled?
Severe pulmonary exudation and ARDS
What can occur if capsaicin is inhaled?
severe pulmonary exudation and ARDS
What is the most common manifestation of diffenbachia ingestion?
inability to talk because of severe pain and swelling and sensation of biting glass, this is due to calcium oxalate crystals found in idioblasts
Are poinsettias poisonous?
ingestions are benign/minimally toxic but contact dermatitis is very common
What toxins does oleander contain?
cardiac glycosides
Is the digoxin level reliable in oleander exposure?
Qualitatively it is valuable but there may be variable cross reactivity therefore you cannot use a level to rule it out
What is the treatment for oleander ingestion?
MDAC
digi FAB -> usually larger doses are needed than in digoxin poisoning
What are 4 major types of problems associated with herbal product use?
-misidentification of an herbal plant and toxicity resulting from the substituted plant
-contamination with non-herbal toxic material
-direct toxicity or OD of herbal products
-drug-herbal interaction
What are the most common undeclared pharmaceuticals in herbal products?
-ephedrine
-chlorpheniramine
-methytestosterone
-phenacetin
Under what circumstances do mushroom exposures occur?
-Accidental ingestion by found children playing outdoors
-mistaken selection of poisonous mushroom while foraging for edible wild mushrooms
-abuse of certain mushrooms for their mind altering potential
which mushroom species is responsible for most deaths?
amanita
What is the onset of symptoms from mushrooms that have serious toxicity and potential for death?
>6hours post ingestion
Which mushroom groups are associated with CNS effects?
Ibotenic acid/muscimol (related to glutamic acid and GABA)
-psilocybin (related to LSD)
What is the treatment for cholinergic syndrome from a mushroom?
atropine
no role for 2PAM
What is the hallmark of coprine mushroom group ingestion?
disulfiram type reaction with EtOH
What groups of mushrooms cause late onset symptoms?
cyclopeptide
gyromitrin
orelline
What is the clinical presentation of cyclopeptide (amanita phylloides)
-6-24 GI symptoms
-hepatic toxicity followed by other end organ involvement over days-weeks
What are useful treatment for amanita poisoning?
MDAC
What are the effects of ingestion of gyromitra mushrooms?
excitatory CNS effects - HA, agitation, seizures
What is a possible therapy for gyromitrin ingestion?
pyridoxine
What is the toxicity associated with orelline?
renal toxicity which can progress to CRF
What type of dcon should be done for theophylline?
AC - it acts as gut dialysis by drawing drug out of the plasma
What is the treatment for a hypotensive patient with theophylline OD?
Beta blocker
What toxicity presents with "snowfield vision"
Formic acid
What does 50% ethylene glycol mean?
50g/100mL
What is the simplified mnemonic for AG metabolic acidosis?
K - ketones
U - uremia
L - lactate
T - toxins
S - salicylates
Why may patients with a wide anion gap metabolic acidosis from toxic alcohols not have an osmolar gap?
Only parent compounds are osmotically active, toxic metabolites (which cause a wide AG) are charged so they are accounted for by the calculation. Therefore once the patient has a wide AG they are so far along in the poisoning that they don't have an osmotic gap anymore
What are features of valproid acid OD?
hypernatremia
hypocalcemia
metabolic acidosis
hypocarnitemia
hyperammonemia
What drugs may cause renal tubular acidosis?
(A hypercholoremic non-AG metabolic acidosis)
topiramate
toluene
What mechanisms result in seizures?
Na+ channels
Ca++ channels
glutamate excess
GABA depletion
What is K2/spice?
a synthetic cannabinoid
What are bath salts? What are prominent features of bath salt ingestion?
synthetic amphetamines
psychotic halluciations
What are the effects of synthetic cannabinoids?
sympathomimetic and THC
What is alcoholic hallucinosis?
Visual hallucinations with a clear sensorium not DTs (which have AMS as a prominent feature)
HOw can you determine if an alcohol withdrawal tremor is real?
Hand and tongue tremor simultaneously
WHy can symptoms of dissociated ingestion resemble other toxidromes?
At higher concentrations these agents bind other agents therefore they may have effects that are anticholinergic, sympathomimetic or opioid
Which patients are at risk of respiratory depression from BZD?
Elderly
very young
those with OSA
What is a reasonable initial dose of flumazenil?
0.05-0.1mg followed by titration (0.5mg/vial)
What is the preferred BZD for cocaine?
midazolam (quick on/quick off)
What is the preferred BZD for alcohol?
longer onset of action but longer CNS duration
How do you calculate the drip rate of naloxone?
2/3 the dose that arouses x 10 injected into a 1L bag of NS, run at 100cc/hr
When is flumazenil indicated?
Acute BZD OD in a patient known not to be a BZD user
in what patients does flumazenil carry a risk of seizures?
-chronic BZD user
-patient who ingests TCA
Which opioid agonists may not produce characteristic miosis?
-propoxyphene
-pentazocine
What should be done in critical patients suspected of body stuffing or packing?
rectal
vaginal
abdo radiograph
Under what circumstances are bowel sounds increased or decreased?
ingestion of agents affecting the cholinergic nervous system
What are the signs of anticholinergic CNS poisoning?
mild temperature elevation
acute delirium
mumbling speech
"picking movements" of the fingers
What is a good way to distinguish between anticholinergic and sympathomimetic OD?
anticholinergic - dry, flushed skin
sympathomimetic - diaphoresis
Which agent should be used for opioid OD of agent with long half life (methadone)?
nalmefene
Which drugs may cause serotonin syndrome in interactions with others?
SSRI
SRI
MAOI
tryptophans
sympathomimetics
dextromethorphan
lithium
What are the features of serotonin syndrome?
AMS
Temperature
agitation
tremor
myoclonus
increased reflexes
ataxia
diaphoresis
shivering
diarrhea
Why is tox screen not useful?
-lab doesn't screen for many things
-if too soon, drug concentration may be too low in the urine
-drugs found may not be responsible for the symptoms
What toxins may cause non-cardiogenic pulmonary edema?
salicylates
opioids
When should AC be considered?
-<1hr post ingestion
-type/amt of medication that are truly toxic beyond required for supportive care (BB, CCB, TCA)
What agents do not adsorb to charcoal?
ions (acid, alkali, Li, borate, bromide)
hydrocarbon
metals
ETOH
When may WBI be useful?
-recent ingestion of lithium, iron, lead
-sustained release formulation of highly toxic agents
-body packers
How is gastric lavage performed?
place 30F or greater OG
lavage with large bore tube
What causes the majority of dermal injuries after chemical exposures?
Direct damage to the skin (as opposed to hyperthermic injury)
Describe the pathophysiology of acid injuries?
Acid injuries produce protein denaturation, coagulation necrosis and eschar formation which limits the extent of injury
By what mechanism does alkali produce injury?
Saponification and liquefactive necrosis
What is a hazardous material (HAZ-MAT)?
A substance that can cause physical injury or environmental damage if not handled properly
What are the two components of a contingency plan for HAZMAT situations?
Initiation of site plan
Evacuation
How are individuals decontaminated in a HAZMAT incident?
Remove clothing
Brush off any powder
Irrigate with copious amounts of H20
Collect H20
What are the minimum requirements for personal protective equipment in HAZMAT situations?
Chemical resistant clothing with hood
eye protection
2 pairs of gloves
Boots
Some form of respiratory protection
What are priorities in decontamination?
Contaminated wounds, then eyes, then mucous membranes, then skin, then hair
How is hydrotherapy done?
Large amount
Low pressure
Prolonged time
Should high pressure irrigation be used?
No, it can drive chemicals into skin or splatter into the eyes
What are adjuncts for decontamination of elemental metals and phenols?
Elemental metals -> mineral oil
Phenols -> polyethylene glycol
How long does hydrotherapy have to be continued to normalize pH in acid/alkali burns?
alkali - up to 12 hours
acid - up to 2 hours
How are ocular burns classified?
Grade I-IV
How long should ocular irrigation continue?
Until ocular pH is 7.4, but for severe burns prolonged irrigation is needed regardless of a normal ocular pH
Where is hydrofluoric acid used?
Petroleum industry
Glass etching
Removing rust
Cleaning cement and bricks
What part of HF results in the most damage?
The free fluoride ion which scavenges cations such as Ca++ and Mg++ resulting in hypocalcemia and hypomagnesemia, also inhibits Na+/K+/ATPase
What are the types of HF exposure?
-inhalation (often industrial)
-dermal
-ocular
What distinct characteristic does HF skin burn have?
The exposure causes progressive tissue destruction (over days)
What is the treatment for HF exposure?
-immediate irrigation
-if still severe pain then requires detox with Calcium salt formation
-remove blisters
-3.5g calcium gluconate in 150cc K-Y jelly
-deep painful burns require infiltrative therapy 0.5cc/cm2 of 10% calcium gluconate
-intraarterial catheter - 10xc of 10% calcium gluconate in 40-50cc of saline
What are systemic manifestations of fluoride toxicity?
Similar to hypocalcemia
-abdo pain
-muscle fasciculations
-nausea
-seizures
-dysrhythmia
How do you treat hypocalcemia secondary to HF toxicity?
IV 10% calcium gluconate
What are signs of systemic toxicity from formic acid?
Acidosis
Hemolysis
Hemoglobinuria
What has prompted an increase in anhydrous ammonia exposures?
Its use in methamphetamine production
What are the mechanisms of injury with anhydrous ammonia?
-freezes tissues
-chemical burns by liquefaction necrosis
What happens with inhalational injury due to anhydrous ammonia?
Proximal airway injury
What is the pH of cement?
10-14
What is the treatment for cement burn?
Hydrotherapy
(excision and grafting are usually necessary)
Where are phenols used?
agriculture
cosmetic
medical fields
What are signs of systemic phenol toxicity?
CNS - stimulation, lethargy, seizures
CVS - conduction disturbances
What is the most effective treatment to reduce the severity of phenol burns?
Polyethylene glycol
How can white phosphorus be identified on the skin?
UV light or copper sulfate solution
What is methemoglobin?
When the ferrous ion (Fe2+) becomes oxidized to ferric (Fe3+)
How is methemoglobinemia treatment?
Asymptomatic -> remove offending agent
Symptomatic 2cc/kg 1% methylene blue over 3-5 minutes or exchange transfusion
How long should you monitor a patient who has ingested hydrocarbons?
6 hours
if no symptoms and normal CXR may d/c home
What should be done immediately with hot tar exposure?
cool with cold H20
Why does tar adhere to the skin?
because it is enmeshed in the hair
What happens when elemental metals (Li+, Na+, K+) come into contact with H20?
a violent exothermic reaction producing heat, H2 gas and OH
What should be done with known elemental exposure?
No H20 lavage
Cover metal with mineral oil
Descrive the ED response to a chemical attack?
-triage outside the ED
-decon outside the ED
-PPE: full face respirator mask, self-contained breathing apparatus, impermeable suits
How are chemical attack agents classified?
-nerve agents
-vesicants
-choking agents
-cyanide
What are vesicants?
A class of drug that produces blisters at the site of contact
What are examples of vesicants?
Mustard gas
Lewisite
Phosgene
What are choking agents?
Chemicals that induce the sensation of choking and can produce upper airway damage and pulmonary edema
What are examples of choking agents?
Phosgene
Chlorine
Zinc containing smoke
How do nerve agents work?
Prevent acetylcholinesterase from hydrolyzing ACh
What are symptoms of muscarinic excess?
Diarrhea
Urination
Miosis
Bronchorrhea
Emesis
Lacrimation
Salivation
What are manifestations of nicotinic excess?
Muscle fasciculations
Weakness
WHy should succinylcholine be used with caution in nerve agent exposures?
Its duration of action will be significantly prolonged
What is the endpoint for atropine administration in nerve agent exposure?
Drying of bronchial secretions
What is given to treat seizures in nerve agent exposure?
BZD
What are the unique features of mustard gas?
Garlic or fishlike odor
does not cause pain immediately
What is the management of vesicant injury?
Remove from environment
Decon with H2) or 0.5% hypochlorite solution
How does cyanide work?
Binds and inactivates cytochrome oxidase part of cytochrome a3 on the electron transport chain
How does the Rumack-Matthew Nomogram work?
APAP concentrations must be measured between 4 and 24 hours after ingestion and plotted on the nomogram. Patients with APAP concentrations on or above the treatment line should be treated.
How do you evaluate ingestions of extended-release APAP preparations?
Peak plasma APAP occurs within 4 hours although some absorption can continue for up to 8 hours.
-use nomogram at 4 hours
-then get a 2nd level 4 hours after the 1st one and treat if above the nomogram line
What is the IV NAC protocol?
150mg/kg over 60min
50mg/kg over 4 hours
100mg/kg over 16hours

One hour after the protocol is complete, recheck the LFTs and APAP. If APAP is >66umol/L or LFTs elevated restart the NAC at 100mg/kg
What are side effects of PO and IV NAC?
PO - vomiting

IV - mild anaphylactoid reaction (rash, flushing, pruritus) (2-18%)
Severe anaphylactoid reaction <1%
What is the elimination of ASA?
Salicylate can be metabolized in the liver, however in overdose, hepatic metabolism becomes saturated and renal elimination of free salicylate becomes more important.
Describe the acid-base disturbance in ASA intox
-initial respiratory alkalosis (direct stimulation of the medulla)
-increased anion gap metabolic acidosis (interference with Krebs cycle, uncoupled oxidative phosphorylation, increased energy demand)
-mixed respiratory alkalosis and metabolic acidosis
-respiratory acidosis occurs when the patient becomes fatigued, suffers salicylate induced acute lung injury or blunted respiration due to co-ingestant) This is a poor prognostic marker and a preterminal event
What electrolyte disturbances are seen in ASA intox?
Hypokalemia
Hyponatremia
Low bicarb

(increased renal excretion of Na and HCO3 to compensate for the respiratory alkalosis)
What are the various degrees of salicylate toxicity?
Asymptomatic: occasional subjective but no objective manifestations
Mild: mild to moderate hyperpnea, tinnitus, sometimes lethargy
Moderate: severe hyperpnea, prominent neuro disturbance
Severe: severe hyperpnea, coma or semi coma, sometimes with convulsions
What are sources of salicylate exposure other than ASA?
Methyl salicylate: oil of wintergreen, tiger balm
Acetyl salicylate: Percodan, fiorinal, peptobismol
Describe chronic ASA intox
Primarily occurs in the elderly
insidious onset
significant illness with low serum salicylate concentration
Mortality = 25%
What are the goals of urine alkalinization?
Serum pH 7.45-7.55
urine pH 7.5-9.0
Which drugs exhibit primarily anticholinergic toxicity
Belladonna alkaloids (atropine, scopolamine, cyclopentolate, topicamide)
Antiparkinsonians (benztropine, trihexylphenidyl, procyclidine)
Prototypical H1 receptor blockers (diphenhydramine, chlorpheniramine, brompheniramine)
Phenothiazines - promethazine
What are the potential ECG findings in anticholinergic poisoning?
sinus tachycardia
What is the indication for physostigmine?
Severe, refractory pure anticholinergic intoxication
What is the toxic differential diagnosis of delirium?
Steroids
lithium
Salicylates
Anticholinergic
Sympathomimetics
PCP
Mushrooms containing muscimol/ibotenic acid
MOA
solvents
CO
Sedative hypnotic withdrawal
What specific presentation is associated with overdose of bupropion?
seizures
What specific presentation is associated with overdose of trazodone?
priapism
What medications are contraindicated in TCA overdose?
Physostigmine
Class Ia antidysrhythmics (quinidine, procainamide)
Class Ic antidysrhythmics (flecanide, propafenone)
Phenytoin
what are the Hunter criteria for serotonin toxicity
1. Serotonin agent administered in the past 5 weeks

Any of the following
-tremor and hyperreflexia
-spontaneous clonus
-muscle rigidity, temp >38, ocular or inducible clonus
-ocular clonus and aggitation or diaphoresis
-inducible clonus and aggitation or diaphoresis
What are drugs associated with serotonin toxicity
Amphetamines
Cocaine
Dextromethorphan
Citalopram
fluoxetine
L-tryptophan
sumatriptan
What is the treatment for NMS?
Bromocriptine
List foods and beverages associated with MAOI toxicity
Aged meats
fava beans
avocados
ginseng
chocolate
figs

ales
beers
wine
sherry
What is the digoxin effect?
Scooped ST segments
Prolonged PR
What are specific dysrhythmias associated with digitalis toxicity
Atrial fibrillation with slow ventricular rate
nonparoxysmal junctional tachycardia
atrial tachycardia with block
bidirectional vtach
What ar indications for administration of digiFab in adults?
Ventricular dysrhythmia
Symptomatic bradydysrhythmia unresponsive to atropine
K+>5mEq/L
Rapidly progressive rhythm disturbances or rising serum potassium
co-ingestion of other cardiotoxic drugs
ingestion of plant containing cardiac glycoside plus severe dysrhythmia
acute ingestion >10mg plus any of above factors
steady state >6ng/mL plus any of the above factors
What are the indications for administration of digiFab in children?
-ingestion of 0.1-0.3mg/kg or steady state >5ng/mL plus rapidly progressive symptoms, potentially life-threatening dysrhythmias, conduction blocks or K+>6
-co-ingestion of cardiotoxic drugs
-ingestion of plant containing cardiac glycosides plus severe dysrhythmia
What is the distribution and function of different beta receptors?
B1
heart - increase HR, contractility and conduction velocity
eye - stimulate aqueous humor production
kidney - stimulate plasma renin secretion

B2
smooth muscle - relaxation of bronchi, BV, intestine, uterus
skeletal muscle - glycogenolysis
liver - glycogenolysis
heart

B3
adipose tissue - lipolysis
What is a caustic substance?
A substance that causes both clinical and histologic damage on contact with tissue surfaces.
What are indications for endoscopy in caustic ingestions?
Indicated in all patients with intentional ingestions
patients with unintentional ingestions with stridor, pain, vomiting and/or drooling
What is crack dancing?
Transient choreoathetoid movement disorder after using cocaine
Why should haldol be avoided in a cocaine-intoxicated patient?
Haldol can limit cooling by impeding diaphoresis
Haldol may increase morbidity
May have associated dysrhythmic effects that can be additive or even synergistic with cocaine
What are admission criteria for cocaine related chest pain
persistent chest pain
ECG changes
elevated enzymes
dysrhythmias
CHF/cardiogenic shock
preexisting CAD
multiple risk factors for CAD
patient requires vasodilation
What are indications for fomepizole (or EtOH)?
strong history or witnessed ingestion of ethylene glycol or methanol containing products
methanol concentration >/=6mmol/L or ethylene glycol >/=3mmol/L
large osmol gap with suspicion of a toxic alcohol ingestion
significant unexplained anion gap acidosis
What are the benefits of fomepizole over ethanol?
No increased risk of CNS depression
No risk of respiratory depression from EtOH
No absolute ICU admission
Decreased risk of electrolyte imbalance
Avoid EtOh induced hypoglycaemia
No need for serial etOH measurements
NO risk of subtherapeutic concentrations
What are the different types of hallucinogens?
Serotonin like agents
LSD
Tryptamines (psilocybin)

Enactogens
Designer amphetamines (MDMA, ecstasy)
Mescaline
Nutmeg

Dissociative agents
PCP
Ketamine
Dextromethorphan

Plants
Marijuana
Salvia
Absinthe
Isoxazole mushrooms
What is the mechanism of action of MDMA?
Release of 5-HT, DA, NE and inhibition of catecholamine reuptake
What are complications associated with MDMA?
Hypovolemia
Hyponatremia
Rhabdomyolysis
Renal Failure
Hyperthermia
Serotonin syndrome
Dysrhythmias
Seizures
Hypertensive crisis in patients taking MAOI
What is dextromethorphan?
-Similar in structure to PCP and opioids
-found in OTC antitussive preparations
-clinical effects increased HR, increased BP, diaphoresis, euphoria, dysphoria, dissociative phenomenon, slurred speech, ataxia, rotary nystagmus
What is the pathophysiology of iron toxicity?
GI toxicity
-direct irritant and corrosive effect on GI mucosa
-N/V/diarrhea/GI bleeding

Cardiovascular
-potent vasodilator
-negative inotrope
-direct myocardial toxicity

Metabolic acidosis
-uncoupling of oxidative phosphorylation
-disruption of the electron transport chain
-free radical production
How do you calculate the elemental iron load?
(#tabs) x (mg of iron salt/tab) x (%elemental iron)
What is the side effect of deferoxamine?
hypotension which is rate dependent
What are sources of mercury?
Elemental
Spill from mercury containing devices
inhalational exposure
deliberate injection or ingestions
accidental ingestion

Salts
accidental disc battery ingestion
deliberate ingestion
laxative abuse

Organic
oral/dermal exposure to thimerosal
occupational/agricultural accidents
water/soil pollution
contaminated seafood consumption
paint
What is the pathophysiology of mercury toxicity?
binds sulhydryl groups and inhibits multiple enzymes
Differentiate huffing and bagging?
Huffing: inhaling HCs through a saturated cloth
Bagging: pouring HCS in a bag and then inhaling them deeply
What are the 4 mechanisms of inhaled toxins?
Pulmonary irritant - alter air lung interface by irritation or inflammation
simple asphyxiant - gas that displaces O2 and lowers FiO2
chemical asphyxiant - inhibits oxidative metabolism
Poor Hb-O2 carrying capacity (CO)
What are simple asphyxiant gases?
carbon dioxide
methane
nitrogen
nitrous oxide
helium
What are examples of highly water soluble gases
ammonia
chlorine
hydrogen chloride
hydrogen fluoride
What are 6 sources of cyanide
-ingestion of cyanide salts as suicide or homicide
-smoke inhalation during fires
-occupational exposure
-medicinal sources (nitroprusside)
-foot sources (plant pits)
-cyanogenic chemicals (nitriles)
Hoe does cyanide inhibit oxidative phosphorylation?
It binds to complex IV of the electron transport chain within the mitochondria
What is the dose of hydroxycobalamin?
5g IV
70mg/kg in children
What are RF for cyanide toxicity in patients receiving nitroprusside?
short term High dose therapy
long term therapy
renal failure
failure to protect the nitroprusside bottle from light (light increases the release of cyanide molecules)
What are potential sources of carbon monoxide?
structure fires
clogged vents for home heating units
gasoline powered generators indoors
What is the pathophysiology of CO intoxication?
CO binds Hb ~250times greater affinity than oxygen
CO shifts the oxyhemoglobin dissociation curve to the left (decreased offloading of O2)
CO binds to cytochrome oxidase in the mitochondria
CO binds to myoglobin in muscles
CO induces lipid peroxidation in the CNS
What are the fetal and neonatal concerns in CO intoxication?
Fetal Hb has a greater affinity for CO than adult hemoglobin
Fetal COHb levels are 10-155 greater than the mother's
Why can patients be severely toxic with lithium levels in the therapeutic rance?
because toxicity does not develop until lithium distributes into the cells
How does acute on chronic lithium toxicity present?
clinical presentation of both acute and chronic intoxication
What are predisposing factors for chronic lithium toxicity?
Dehydration
Hyponatremia
thermal stress
renal failure
Drug interactions (NSAID, ACE, ARB, diuretics, metronidazole, carbemazepine, antipsychotics, SSRI)