• 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

1/242

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;

242 Cards in this Set

  • Front
  • Back
Define pharmacodynamics
Study of the actions and mechanism(s) of action of drug in living systems.
Define pharmacokinetcs
Study of the absorption, distribution, localization
Describe phase 1 of clinical trials of a drug (Who, Why, By Whom)
Who - Normal volunteers, (special pop.s)
Why - Safety, bio effects, metabolism, kinetics, drug interactions
By Whom - Clinical pharmacologists
Describe phase 2 of clinical trials of a drug (Who, Why, By Whom)
Who - Selected patients (with illness)
Why - Therapeutic efficacy, dose range, kinetics, metab.
By Whom - Clinical pharmacologists, clinical investigators
Describe phase 3 of clinical trials of a drug (Who, Why, By Whom)
Who - Large samples of selected patients
Why - Safety and efficacy
By Whom - Clinical investigators
Differentiate between "within-patient" trials and "between-patient" trials
Within patient trials have two drugs (or one and a placebo) with two groups of patients that swap over after a washout period. Between-patient trials have the two groups stay the same throughout.
Describe Schedule I drugs and give examples
High potential for abuse, no currently acceptable medical use, lack of accepted safety for use under medical supervision.

Heroin, Marijuana, LSD
Describe Schedule II drugs and give examples
High potential for abuse, currently accepted medical use, abuse may lead to severe psychological or physical dependence

Morphine, Cocaine, Methamphetamines
Describe Schedule III drugs and give examples
Potential for abuse less than I or II, currently accepted medical use, abuse may lead to dependence

Amphetamine, most barbituates, PCP
Describe Schedule IV drugs and give examples
Low relative potential for abuse, currently accepted medical use, possible dependence with abuse

Barbital, chloral hydrate, paraldehyde
Describe Schedule V drugs and give examples
You know this

Mixtures having small amounts of codeine or opium
Define absorption
Movement of drug into the blood from site of application
Define distribution
Movement of drug from blood to its site of action or storage
What is Ficks Law (equation)
J = -DA ⌂C/⌂X
What is the Henderson-Hasselbach Equation
pH = pKa + log[A-]/[HA]
What is pKa
The pH at which [A-] = [HA] (50% dissociated)
What does polarity do to absorption (membrane crossing)
Decreases absorption
Where are weak acids absorbed to the greatest degree
Small intestine (mostly due to surface area)
Given hematocrit and blood concentration of a drug, what equation would be used to determine the plasma concentration of a drug
Total concentration in blood = (fraction of blood that is serum x serum water content x concentration of drug in plasma) + (fraction of blood that is cells x cell water content x concentration of drug in cells)

[Cb] = ([serum] x 1 x [Cp]) + ([cells] x 0.7 x [Ci])
What is the water content of serum for the purposes of our equations
100%
What is the water content of cells for the purposes of our equations
70%
Phase 1 of biotransformation involves what
"Functionalization":
Oxidation, Hydroxylation, Dealkylation, Deamination, Hydrolysis

Might use p450, might not. *Oxidation* in microsomal cytochrom p450 *MOST IMPORTANT*
Phase 2 of biotransformation involves what
Conjugation
Which variant(s) of p450 is involved in ~90% of drug metabolism
2C9, 2D6, 3A4
List three cytochrome P450-independent oxidations
Microsomal Flavin-Containing Monooxygenase (FMO)
Alcohol dehydrogenase (ADH)
Xanthine oxidase
The local anesthetic procaine is degraded by what
Hydrolysis by plasma cholinesterase
What is a more suitable antiarrhythmic drug than procaine? why?
Procainamide, protected from degradation by plasma cholinesterase
List the six specific types of conjugation in phase II reactions
Glucuronidation
Sulfate conjugation
Glutathione conjugation
Glycine conjugation
Acetylation
Methylation
Describe the Endogenous Reactant, Transferase (location), and types of substrates for the conjugation reaction "glucuronidation"
Endogenous reactant - UDP Glucuronic acid
Transferase (location) - UDP-glucuronyl transferase (microsomes)
Types of substrates - Phenols, alcohols, carboxylic acids, hydroxylamines, sulfonamides
Describe the Endogenous Reactant, Transferase (location), and types of substrates for the conjugation reaction "Sulfate conjugation"
Endogenous reactant - Acetyl-CoA
Transferase (location) - N-Acetyl transferase (cytosol)
Types of substrates - Amines
Describe the Endogenous Reactant, Transferase (location), and types of substrates for the conjugation reaction "Glutathione conjugation"
Endogenous reactant - Glutathione
Transferase (location) - GSH-S-transferase (cytosol, microsomes)
Types of substrates - Epoxides, arene oxides, nitro groups, hydroxylamines
Describe the Endogenous Reactant, Transferase (location), and types of substrates for the conjugation reaction "Glycine conjugation"
Endogenous reactant - Glycine
Transferase (location) - Acetyle-CoA glycine transferase (mitochondria)
Types of substrates - Acetyl-CoA derivatives of carboxylic acids
Describe the Endogenous Reactant, Transferase (location), and types of substrates for the conjugation reaction "Acetylation"
Endogenous reactant - Phosphoadenosyl phosphosulfate
Transferase (location) - Sulfotransferase (cytosol)
Types of substrates - Phenols, alcohols, aromatic amines
Describe the Endogenous Reactant, Transferase (location), and types of substrates for the conjugation reaction "Methylation"
Endogenous reactant - S-Adenosylmethionine
Transferase (location) - Transmethylases (cytosol)
Types of substrates - Catecholamines, phenols, amines, histamine
Conjugation of catecholamines, phenols, and amines is carried out via what type of conjugation
Methylation
Conjugation of alcohols are carried out via what type(s) of conjugation
Glucuronidation or sulfate conjugation
Conjugation of amines are carried out via what type(s) of conjugation
Acetylation or methylation. (Sulfate conjugation if amines are aromatic)
Enzyme induction does what
Increases rate of hepatic biotransformation of drug
Increases the rate of production of metabolites
Increases hepatic drug clearance
Decreases serum halflife
Decreases serum and total and free drug concentrations
Decreases pharmacological effects if the metabolites are inactive
What does cimetidine do?
Antagonizes histamine at the H2 receptor
Also inhibits P450 and decreases hepatic blood flow = decreased drug metabolism
What is cimetidine used for?
Treatment of peptic ulcers and other states in which acidity should be minimized
What is a possible major mechanism for drug-drug interactions
Inhibition of P450
What are some drugs that cimetidine might have an effect on metabolism of
*Diazepam*
Warfarin, benzodiazepines, phenytoin, morphine
Inhibits P450 = decreased metabolism of other drugs
Disulfram is also known as
Antabuse
Inhibition of microsomal enzymes can cause what
Decreases the rate of hepatic biotransformation of drug
Decreases the rate of production of metabolites
Decreases total clearance
Increases serum half-life
Increases serum and total free drug concentrations
Increases pharmacological effects if the metabolites are inactive
What is first pass metabolism and what are some drugs of note that undergo considerable first pass metabolism
Metabolism of orally administered drugs as they pass through the portal system

Glyceryl, trinitrate, morphine, propanolol, isoproterenol, meperidine, pentazocine

Aspirin, levodopa, lidocaine, metoprolol, morphine
Can phase II reactions be induced by drugs? Exceptions?
No, except for glucuronidation
Can P450 reactions be induced?
Yes
Can reactions catalyzed by nonmicrosomal enzymes be induced
No
Nonsynthetic reactions (Phase I and P450) catalyzed by nonmicrosomal enzymes are most commonly of what type? What about those catalyzed by microsomal enzymes
Nonmicrosomal - Mostly hydrolyses

Microsomal - Mostly oxidations
A deficiency in the ability to detoxify chloramphenicol could lead to
aplastic anemia
List some factors influencing biotransformation
Age, Pathology, Genetics, Enzyme induction, Co-administration, Environmental agents, Route of administration
How does age influence biotransformation
1. Low activity of a particular hepatic enzyme in neonates

2. Decreased hepatic enzyme activity in the elderly
How might some pathologies influence biotransformation
1. Decreased hepatic enzyme activity in infectious hepatitis patients.

2. Low levels of hepatic and nonhepatic enzymes present in uremic patients
How might genetics influence biotransformation
1. Variations of enzyme activity in the general population

2. Unusual variations of enzyme activity
A combination of an oral anticoagulant and a barbiturate might cause what
A decrease in the effectiveness of the anticoagulant
A combination of an oral contraceptive with what drugs might cause a decrease in its effectiveness
Barbiturates, Phenytoin, rifampicin
What polarity best suites a drug in order for it to be excreted in urine
Polar
Know the glucuronidation pathway associated with excretion of drugs via the gastrointestinal tract
Know it!
As a person ages, what happens to their total body water as a portion of body weight
Body water decreases
What happens to the fraction of a lipophilic drug in fatty tissue as we age
Increases
What happens to the capability of the liver to metabolize drugs as we age
Increases from birth to adulthood, decreases as we become elderly
What happens to renal drug elimination capability as we age
Increases from birth to adulthood, decreases as we become elderly
What is the equation used to adjust dosage for weight (generic)
Dose required = (average dose/70kg) x weight of individual (in kg)
Homologous desensitization means what
Desensitization to a drug acting on a specific receptor and not similar ones after exposure to the original type of receptor
Heterologous desensitization means what
Desensitization to a drug at multiple receptors after exposure to only one receptor
What is tachyphylaxis? Examples of drugs that have this?
Repeated administration of the same doe of a drug results in a reduced effect of the drug over time

Ephedrine, tyramine, Morphine, nitrites, Atropine
What are the objectives of pharmacogenetics
Identification of *genetically controlled variations in responses to drugs*
Study of the molecular basis for these conditions
Their clinical significance
Development of methods by which susceptible individuals can be recognized before drug is administered
Defective plasma cholinesterase might lead to what drug having what undesirable effect
Succinylcholine

Persistent apnea and paralysis
Deficient acetyltransferase might lead to what drug having what undesirable effect
Isoniazid

Increased toxicity
Deficient G6PD in erythrocytes might lead to what drug(s) having what undesirable effect
Primaquine, antimalarials, sulfonamide, probenecid, chloramphenicol

Drug-induced hemolytic anemia
Tetracycline antibiotics combined with metal ions (Al3+, Fe2+, etc.) causes what
Reduced absorption of the antibiotic due to formation of poorly soluble chelates
Digitalis combine with propantheline or other anticholinergics causes what
Reduced absorption due to reduced gut motility
Combinations of anticholinergics can result in what
Enhanced anticholinergic effects; heat stroke in hot and humid conditions; toxic psychosis; adynamic ileus
Combinations of CNS depressants can result in what
Enhanced CNS depression; drowsiness
Combinations of neuromuscular blockers can result in what
(ie conventional neuromuscular blockers and aminoglycoside antibiotics)
Enhanced neuromuscular blockade, prolonged apnea
Phenytoin combined with chloramphenicol or disulfiram can result in what
Increased serum phenytoin levels, possible intoxication
A possible cause of lethal digitalis intoxication can result from interactions with what other drugs
Diuretics causing hypokalemia
A toxic response occurs in what populations
All populations if dose is high enough
An idiosyncratic response occurs in what populations
Only genetically abnormal
Half-life generalized equation
t1/2 = 0.693/k
Regarding drug absorption and bioavailability, what equation describes the value of F (bioavailability)?
F = AUCoral / AUCiv

or, if the two doses are different
F = (AUC oral x dose iv) / (AUC iv x dose oral)
What is the most common cause of low bioavailability for a drug
Oral dosage forms of poorly water soluble drugs (PRM-5-5)
What is efficacy and what factors determine it
Ability of a drug to stimulate a response

Type of interaction with receptor (ie agonists vs partial agonist)
Characteristics of the effector system involved
Limitations on the amount that can be administered
What is potency and what factors determine it
Relative dose required to produce the same effect on a system

Affinity for its receptor
Efficiency of transduction step
Ability of the drug to reach its site of action
How is therapeutic index determined and what does it suggest for us
LD50/ED50

Indication of safety factor

Higher therapeutic index = safer
Rank the membrane receptors and their relative speed of action
Channel-linked receptors (ionotropic) - milliseconds
G-protein coupled receptors (metabotropic) - seconds
Kinase-linked receptors - minutes
Receptors linked to gene transcription (nuclear receptors) - hours
What is the action of a Gs receptor
Activates Ca++ channels, activates adentylyl cyclase
(stimulatory)
What is the action of a Gi receptor
Activates K+ channels, inhibits andenylyl cyclase
(inhibitory)
What is the action of a Gq receptor
Activates Phospholipase C
Summarize if Phosphoinositide cycle
Ligand -> Gi/Go -> PL-C -> phosphatidylinositol -> IP3 & DAG -> Ca++ release and PKC activation -> Ca++ calmodulin and CDPKs and PKC substrates -> Biological response
Describe arachidonic acid's role in signal transduction
Go -> PLA2 -> Phosphoinositol -> Arachidonic acid -> HPETE, Leukotrienes, Prostaglandins and thromboxanes
Tyrosine kinase receptors are mostly associated with what
growth
What is the major visceral sensory relay cell group in the brain that receives inputs from all the major organs of the body
Nucleus Tractus Solitarius
What is the nucleus tractus solitarius
The major visceral sensory relay cell group in the brain that receives inputs from all the major organs of the body
Junctions using acetylcholine are known as
Cholinergic
Acetylcholine is the neurotransmitter mediator in what locations
Skeletal neuromuscular junction
All autonomic ganglia
All postganglionic parasympathetic junctions
Some postganglionic sympathetic junctions (sweat glands)
Select synapses in CNS
Junctions using norepinephrine are known as
Adrenergic
List some Nonadrenergic - Noncholinergic Transmission types
ATP; Vasoactive Intestinal Peptide; Neuropeptide Y; y-aminobutyric acid (GABA); dopamine
Afferent (Sensory) Nerves might use what transmitters?
Substance P, Somatostatin, VIP, Cholecystokinin, Enkephalins
A rapid response neurotransmitter from postganglionic parasympathetic neurons might be? Sympathetic?
Parasympathetic - ACh

Sympathetic - ATP
An intermediate response neurotransmitter from postganglionic parasympathetic neurons might be? Sympathetic?
Parasympathetic - NO

Sympathetic - NE
A slow response neurotransmitter from postganglionic parasympathetic neurons might be? Sympathetic?
Parasympathetic - VIP

Sympathetic - NPY
Radial muscles of the iris undergo contraction (mydriasis) when what receptor is stimulated
Alpha 1 adrenergic
Sphincter muscles of the iris undergo contraction (miosis) when what receptor is stimulated
M3, M2 muscarinic cholinergic
Ciliary muscles undergo contraction when stimulated by what? Relaxation?
Contraction - M3, M2 muscarinic cholinergic

Relaxation - Beta 2 adrenergic
Lacrimal glands secrete most when what receptor is stimulated
M3, M2 muscarinic cholinergic
Adrenergic stimulation causes what and is mediated primarily by what receptor in the heart? Cholinergic?
Adrenergic - Beta 1 - Increased heart rate and contractility

Cholinergic - M2 - Decreased rate and contractility (little effect on HIS Purkinje)
Describe autonomic stimulation on blood vessels
Adrenergic:
Alpha stimulation = constriction
Beta 2 = dilation in skeletal muscle arteries and all veins

Cholinergic:
Only has effect on arteries of erectile tissue and salivary glands *M3 = dilation*
Adrenergic stimulation of tracheal and bronchial smooth muscle causes what and is mediated by what receptor? Cholinergic?
Beta 2 - Relaxation

M2/M3 = Contraction
Adrenergic stimulation of bronchial glands causes what and is mediated by what receptor? Cholinergic?
Alpha 1 - Decreased secretion

Beta 2 - Increased secretion

M2 - Stimulation
The primary cholinergic receptor in the GI tract is what
M3
Renin secretion is stimulated by what mediator at what receptor
NE (maybe E) at Beta 1 receptor
Erection is mediated by what at what receptor? Ejaculation?
Erection - ACh at M3

Ejaculation - NE at alpha 1
Secretion of E and NE forom the adrenal medulla can be stimulation by what at what receptor
ACh at a nicotinic receptor
At the sympathetic terminal, autoreceptors may experience inhibition of NE release when what is stimulated? Heteroreceptors?
Autoreceptor inhibition of NE release by stimulation of alpha 2A receptor

Heteroreceptor inhibition of NE release by stimulation of the M2 receptor
Discuss activation of the M1, *M3*, M5 receptor
Gq
Activation of PLC - Increased IP3, DAG, Ca++, PKC
Depolarization - Increased EPsP
Activation of PLD2
Acitvation of PLA2 - Increased AA
Discuss the activation of *M2*, M4 receptor
Gi/Go
Inhibition of Ad Cyl - Decreased cAMP
Activation of K+ channels
Inhibition of voltage gated Ca++ channels
Hyperpolarization and inhibition
Atropine antagonizes what receptors
Both pre and post ganglionic muscarinic
Which receptors are sensitive to hexamethonium
Nicotinic I (can also be antagonized by curare)
Which receptors are sensitive to curare
Nicotinic II (neuromuscular)
List three agonists of a muscarinic receptor? Antagonists?
Agonists - ACh, Methacholine, Pilocarpine

Antagonists - Atropine, Scopolamine, Methantheline
List three agonists for a Nicotinic I receptor? Antagonists?
Agonists - ACh, Nicotine, Succinylcholine

Antagonists - Hexamethonium, Mecamyline, Trimethaphan, Curare
List three agonists for a Nicotinic II receptor? Antagonists?
Agonist - ACh, Nicotine

Antagonists - Curare, Succinylcholine, Pancuronium
Describe the synthesis of Epinephrine
Tyrosine
DOPA
Dopamine
NE
E
Describe alpha 1 transduction
Gq
Activation of PLC; Increased IP3, DAG and Ca++
Activation of PLA2; Increased AA
Activation of Na+/H+ exchanger
Modulation of K+ channels
Increased MAPK signalling
Describe alpha 2 transduction
Gi/Go
Inhibition of AdCyc; decreased cAMP
Inhibition of PkA
Describe beta 1/2/3 stimulatory transduction
Gs
Activation of AdCyc; Increased cAMP
Activation of PkA
Activation of Ca++ channels
Describe beta 2/3 inhibitory transduction
Gi
Inhibition of AdCyc; decreased cAMP
Inhibition of PkA
Does Epi have any specificity for adrenergic receptors
No selectivity
Does Norepi have any specificity for adrenergic receptors
alpha 1 & 2 and beta 1, not beta 2
Does Isopenterenol have any specificity for adrenergic receptors
Beta 1 & 2, not alpha
Does Phenylephrine have any specificity for adrenergic receptors
Alpha 1
Does Clonidine have any specificity for adrenergic receptors
Alpha 1 & 2 (mostly 2)
Does Dobutamine have any specificity for adrenergic receptors
beta 1
agonist
Does Terbutaline have any specificity for adrenergic receptors
beta 2
agonist
Does Phentolamine have any specificity for adrenergic receptors
Alpha 1 & 2
antagonist
Does prazosin have any specificity for adrenergic receptors
alpha 1
antagonist
Does yohimbine have any specificity for adrenergic receptors
alpha 2
antagonist
Does Propanolol have any specificity for adrenergic receptors
Beta 1 & 2
antagonist
Does Metoprolol have any specificity for adrenergic receptors
Beta 1
antagonist
Does Butoxamine have any specificity for adrenergic receptors
Beta 2
antagonist
List four choline esters
Acetylcholine
Methacholine
Carbachol
Bethanechol
Which choline ester is selective for muscarinic receptors? What is it used for?
Bethanechol

Post-operative paralysis of gut and bladder
Which choline ester produces a large/strong response at nicotinic recptors
Carbachol
Name a Muscarinic agonist that is not a choline ester
Pilocarpine
Describe the properties of pilocarpine and what it is used for
Natural muscarinic alkaloid (not a choline ester)

Markedly stimulates secretion of sweat and saliva

Treats glaucoma
What are the side effects of muscarinic agonists
Salivation
Lacrimation
Nausea & Vomiting
Headache and visual disturbances
Bronchospasm
Hypotension
Diarrhea
Urination
Sweating
Pupil constriction
CNS activation
When a muscarinic agonist is applied directly to the eye, what happens
Miosis, spasm of accommodation and persistent fall in intraocular pressure
List, in ranked order, the susceptibility of muscarinic sites to muscarinic antagonists
Salivary, bronchial, and sweat secretion
Pupil, ciliary muscle, and cardiac vagus
Urinary bladder and GI tract
Gastric secretions
Does atropine cross the BBB
Yes, causes excitation and possible hallucinations
List the effects of the muscarinic antagonist atropine in the eye
Eye: Blocks sphincter of iris (mydriasis), paralyzed accommodation (cycloplegia), with glaucoma = increased intraocular pressure
List the effects of the muscarinic antagonist atropine in the Heart and blood pressure
Heart: Low dose = possible bradycardia. Higher [] = tachycardia via blockade of vagal effects in SA node
Blood pressure: No effect -> parasymp. tone
List the effects of the muscarinic antagonist atropine in Smooth Muscle, Glands, and Sphincters
Smooth muscle: Non-vascular become relaxed

Sphincters: Muscarinic and cholinergic stimulation of smooth muscles in sphincters is blocked

Glands: Inhibition of exocrine gland secretions
Does scopolamine cross the BBB
Yes -> Sedation
What is/are the use(s) of scopolamine
Sleep aide / sedative
Anti-motion sickness
Anti-parkinsons
"Truth Serum"
Compare/contrast the CNS effects of the muscarinic antagonists Atropine and Scopolamine
Scopolamine causes drowsiness, euphoria, amnesia, and fatigue whereas atropine stimulates medulla and higher cortical functions

Both work for antitremor in Parkisnsons
List five muscarinic antagonists
Atropine
Scopolamine
Ipratroprium
Tolterodine
Oxybutyin
Describe Ipratropium's absorption
Inhalation
Poorly absorbed
Does not cross BBB
Poorly crosses membranes
After MI, muscarinic antagonists can be used for what
Treating Sinus bradycardia
Sinoatrial arrest/block
What can muscarinic antagonists be used for in the respiratory system
Inhibition of secretions
Decrease vagal overactivity
Used in cold remedies
COPS
Describe the symptoms of belladonna alkaloid (ie atropine) toxicity
Dry mucous membranes, widely dilated unresponsive pupils, tachycardia, abnormal mental state, excessive heat/fever
How might the mental agitation brought on by overdosage of atropine be treated
Reassurance, Diazepam or chlordiazepoxide, Physostigmine (choline esterase inhibitor that crosses BBB)

Phenothiazines are contraindicated b/c they are anticholinergic
What occurs at what dosages of atropine
0.5mg - Slight salivary and sweating decrease
1.0mg - Accelerated heart, mild pupil dil.
2.0mg - Rapid HR, palpitation, dil. pupils, blurred vision
5.0mg - Disturbed speech, restlessness and fatigue, fever
10.0mg - All more severe
List the anticholinesterases and label them as reversible of irreversible
Reversible: Edrophonium, Neostigmine, Physostigmine, ?pyridostigmine?

Irreversible: Isoflurophate (DFP)
Describe the properties of edrophonium
Short acting, fast response
Inhibition is readily reversible
Acts mainly at neuromuscular junction
Describe the properties of Neostigmine, Physostigmine, and ?pyridostigmine?
Attaches to two sites of enzyme
Medium (2-8 hours) duration
More potent than edrophonium, but more side effects
Physostigmine will cross BBB
Treats myasthenia gravis
Describe the properties of Isoflurophate (DFP)
Almost irreversible
highly toxic, long duration
Relatively selective for neuromuscular junction
Doesn't cross BBB
What are the pharmacological actions of antcholinesterases
Eye: miosis and spasm of accommodation
Gut: Acts on Auerbach's plexus
Skeletal Muscle: Blocks AChE and edrophonium and neostigmine have direct stimulation
Heart: Bradycardia
Stimulates exocrine glands, bronchioles, and urinary tract
Possible respiratory paralysis (depression of respiratory centers in CNS)
What are the muscarinic manifestions of organophosphate (anticholinesterases) poisoning? How are they treated?
Treat with atropine
Ciliary spasm
Marked miosis
Bronchoconstriction
Increased bronchial secretion
Sweating
Salivation
Bradycardia
Hypotension
Incontinence
What are the nicotinic manifestions of organophosphate (anticholinesterases) poisoning? How are they treated?
Treat with pralidoxine to regenerate enzyme
Weakness
Muscle fasciculation
Muscular paralysis
What are the CNSmanifestions of organophosphate (anticholinesterases) poisoning?
Confusion
Ataxia
Convulsions
Coma
Respiratory depression
How is myasthenia gravis diagnosed and treated? Any drug contraindications?
Resversible cholinesterase inhibitors

Several, including muscle relaxants like curare
What are two treatments for glaucoma
ChE inhibitor - Demecarium

Muscarinic agonist - Pilocarpine
If edrophonium is used for diagnosis of cholinergic vs myasthenic crisis what could happen
Cholinergic crisis could be worse for longer
What is a clinical use of edrophonium
Diagnosis of myasthenia gravis
What is a clinical use of tacrine or donepezil
Treatment of alzheimers (maybe)
What is a clinical use of physostigmine
Topical application
Wide angle glaucoma
What is a clinical use of neostigmine
Paralytic ileus and atony of bladder from surgery
Diagnosis of myasthenia gravis
Reversal of NM blockade
What is a clinical use of pyridostigmine
Treatment of myasthenia gravis
Pretreatment to reduce risk of nerve gas toxicity
Reversal of NM blockade
What are the pharmacological actions of nicotine at various sites
Autonomic ganglia - Stimulation (two phases)
Adrenergic nerve terminals - Direct stim. for release of NE
NMJ - Same as ganglia (less sensitive)
CNS - stimulation
CV system - Increased BP and cardiac performance
GI tract - Increased tone and motility
Exocrine glands- Increased salivation and bronchial secretions
What are some therapeutic uses of ganglionic antagonists
Hypertensive cardiovascular disease
Controlled hypotension
Autonomic hyperreflexia
List the organs and tissues that are primarily under parasympathetic control (cholinergic tone)
Mycardium - Atrium & SA node
Eye - Iris and Ciliary muscle
GI tract
Urinary bladder
Salivary Gland
Sweat Glands
List the organs and tissues that are primarily under sympathetic control (adrenergic tone)
Myocardium - Ventricles
Blood vessels
How does botulinum toxin cause paralysis
Blocks vesicular ACh release
How do nondepolarizing agents like curare function
Competitively binds to receptor binding site, block ability of ACh to open ligand gated ion channels
Effects of excessively high levels of nondepolarizing agents like curare can be reversed by what
Increasing levels of ACh by blocking function of AChE

Use - Neostigmine, edrophonium, pyridostygmine
List some nondepolarizing agents other than curare
Atracurium, Mivacurium, Pancuronium, Tubocurarine, Vecuronium
List the depolarizing neuromuscular blocker
Succinylcholine
What does succinylcholine do
Causes continuous depolarization at NMJ
Describe Phase I and Phase II blockade
Phase I - Prolonged depolarization makes unresponsive to stimulation
What is the effect of neostigmine on Tubocurarine, and Phase I and II of Succinylcholine
Tubocurarine - Antagonistic
Succinylcholine:
Phase I - Augmented
Phase II - Antagonistic
Neuromuscular blockers can be used for what
General Anesthesia - Endotracheal intubation, Muscle relaxants in surgery

Electroshock therapy:
Succinylcholine to prevent fracture - Control muscle spasms of convulsive disorders, Diagnosis of myasthenia gravis (possibly)
Nondepolarizer NMJ blockers can be enhanced by what
Certain anesthetics and certain antibiotics like streptomycin, neomycin, and polymyxins
Nendepolarizer NMJ blockers can be antagonized by what
Anticholinesterase agents like neostigmine
A combination of certain general anesthetics and succinylcholine can result in what
Malignant hyperthermia
Malignant hyperthermia can be treated with what
Dantrolene
Study Topic 9 - The Geriatrics
Study it!
Go over the potentially inappropriate medications in older adults (9-20)
Do eeeet
List the six main effects of adrenergic agonists
1) Excitation of smooth muscle & glands
2) ?Inhibition of smooth muscle and glands?
3) Stimulation of heart
4) Metabolic effects
5) Endocrine effects
6) CNS effects
What are the therapeutic uses of epinephrine based on which receptor it's effecting
Alpha - Combined with local anesthetics and prolongs duration of action and decreases toxicity by reducing absorption, Local hemostasis, mydriatic - decongestant, wide angle glaucoma, shock

Beta - Bronchial asthma, heart block, increase blood sugar and FFAs
What are some untoward effects of epinephrine
Tachycardia, ventricular arrythmias, hypertension leading to stroke, MI, interacts with halogenated hydrocarbons to produce arrythmias
What are the therapeutic uses of NE
Shock, pressor and inotropic support post cardiac surgery
What are some untoward effects of NE
Tissue sloughing, phlebitis, arrythmias, increased myocardial oxygen demand
Tolerance develops
Why does NE not have a great change in heart rate when administered
Reflex bradycardia from barrow receptor response
Does NE have much of an effect on metabolics
None to not as much as Epi
What receptors does isoproterenol act on
Selective for Beta 1 and beta 2
What are the therapeutic uses of isoproterenol
Shock, heart block, bronchial asthma
What are some untoward effects of isoproterenol
Cardiac stimulation, arrhythmias
After IV infusion of epinephrine, what will happen to systolic and diastolic blood pressure as well as heart rate, and peripheral resistance.
Systolic - Increased
Diastolic - No change/slight decrease
Heart rate - Increase
Peripheral resistance - Decrease
After IV infusion of norepinephrine, what will happen to systolic and diastolic blood pressure as well as heart rate, and peripheral resistance.
Systolic - Increase
Diastolic - Increase
Heart rate - No change/decrease
Peripheral resistance - Large increase
After IV infusion of isoproterenol, what will happen to systolic and diastolic blood pressure as well as heart rate, and peripheral resistance.
Systolic - No change/slight increase
Diastolic - Large decrease
Heart rate - Large increase
Peripheral resistance - Decrease
What receptor does phenylephrine act on
Directly acts on alpha-1 receptor
What are some therapeutic uses of phenylephrine
Nasal and conjunctival decongestant
Vasoconstrictor (can treat hypotension)
What receptor does clonidine act on
Alpha 2 selective- Will decrease release of NE
What is the therapeutic use of clonidine
Ainthypertensive
What is the mechanism of action for clonidine
Acts peripherally/directly on presynaptic alpha-2 receptor
Acts centrally at unkown site - Decreased sympathetic outflow from CNS = Decreased vasoconstriction and contractility etc.
What receptors does Terbutaline act on
Beta 2 selective
What are the therapeutic uses of terbutaline
Bronchial asthma

Could stimulate heart in large doses
What receptors does dobutamine act on
Beta 1 selective
What are some therapeutic uses for dobutamine
Congestive heart failure, cardiopulmonary by-pass, inotropic support following cardiac surgery
What are some untoward effects possible with dobutamine
Cardiac arrhythmias, enhanced AV conduction (arrhythmia risk)
What is the mechanism of action for ephedrine
Mixed sympathomimetic amine
How do the pharmacological characteristics of ephedrine differ from epinephrine
Ephridine is:
Orally effective
Has a longer duration of action
Has a lower potency
Has a greater CNS (stimulation) activity
What are the therapeutic uses of ephredrine
Asthma, Hypotension, Decongestant, Mydriatic
Describe the mechanism of action of dopamine on adrenergic/sympathetics
Mixed/complex
Directly acting dopamine agonist in mesentary and kidney to release NE
No selectivity for alpha or beta receptors
What are the therapeutic uses of dopamine
Shock - due to cardiac stimulation (beta 1) and vasodilation of dopamine receptors in renal and mesenteric vascular beds
What are some therapeutic uses of amphetamine
Narcolepsy, obesity, ADD, parkinsonism, epilepsy

CNS stimulant
Go over tables 10-26-ish
10-26 + or -
Non selective adrenergic agonists will generally have what effects
Increased heart rate (unless baroreflex overcomes), blood pressure, vasoconstriction (dilation in skeletal muscle)

Note differences between NE and E
What are the effects of a non selective beta agonist
Decrease in PVR
Increase in CO
Tachyarrhythmias
Bronchodilation
What are the effects of a Beta 1 selective agonist
Increased contractility
Increased HR
Increased AV conduction
What are the effects of a Beta 2 selective agonist
Relaxation of bronchial smooth muscle
Relaxation of uterine smooth muscle
What are the effects of an alpha 1 selective agonist
Vasoconstriction (main)
What are the effects of an alpha 2 selective agonist
Decreased symp outflow from brain = decreased PVR and BP
Decreased nerve-evoked release of sympathetic transmitters
Decreased production of aqueous humor
What are the effects of some indirectly acting alpha agonists
CNS stimulation
Increased BP
myocardial stimulation
What are the effects of mixed acting adrenergic agonists like dopamine and ephedrine
Vasodilation (coronary, renal, mesenteric beds)
Increase in glomeruler filtration rate
Increased heart rate & contractility
Increased systolic BP
Similar to non-selective adrenergic agonists
CNS stim.
Haloalkylamines, like phenoxybenzamines, are irreversible non-selective alpha receptor antagonists. What therapeutic uses do they have?
Treatment of catecholamine excess such as pheochromocytoma

Treatment of peripheral vascular disease
Imidazoles, like Phentolamine and tolazoline, are non-selective alpha receptor antagonists. What therapeutic uses do they have?
Same as phenoxybenzamine:
Treatment of catecholamine excess such as pheochromocytoma

Treatment of peripheral vascular disease
Quinazolines, like prazosin and terazosin, are alpha-1 selective antagonists. What therapeutic uses do they have?
Primary hypertension
Benign prostatic hypertrophy
nonselective alpha blockers will have what pharmacological actions
Decrease in PVR and BP
Venodilation

Untoward:
Postural hypotension
Failure of ejaculation

*Possible cardiac stimulation due to enhanced release of NE via alpha2-receptor blockade
Alpha 1 selective blockers will have what pharmacological actions
Decreased PVR and BP
Relxation of smooth muscles in neck of urinary bladder and in prostate
The active ingredient in Mormon Tea (or Ma Huang) is a
Mixed acting adrenergic (Ephedrine)