• 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

1/68

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;

68 Cards in this Set

  • Front
  • Back
Simvastatin (Zocor)
inhibits HMG-CoA reductase

SE: muscle breakdown
Niacin (Niacor)
inhibits diacylglycerol acyl-transferase 2

SE: flushing/rashes
Clofibrate (N/A)
PPAR-alpha agonist

SE: myopathy, arrhythmias
Fenofibrate (tricor)
PPAR-alpha agonist

SE: myopathy, arrhythmias
colestipol (colestid)
bind bile acids

SE: GI effects
Ezetimibe (Zetia)
inhibits cholesterol transporter NPC1L1

SE: GI effects

used with statins as last resort Tx
Betahistine (Serc)
activates HI receptors

use: vertigo
SE: headache/nausea
Dimenhydrinate (Dramamine)
inhibits H1 receptors

use: allergy generally
SE: sedation
Diphenhydramine (Benadryl)
inhibits H1 receptors

use: allergy generally
SE: sedation
Hydroxyzine (Atarax)
inhibits H1 receptors

use: allergy generally
SE: sedation
Bromopheniramine (Dimatapp)
inhibits H1 receptors

use: allergy generally
SE: sedation
Fexofenadine (Allegra)
inhibits H1 receptors

use: allergy generally
SE: sedation
Bicarbonates (Alka-Seltzer)
neutralizes gastric acid

use: GERD, s/a
SE: metabolic alkalosis
aluminum hydroxides (Gaviscon)
neutralizes gastric acid

use: GERD, s/a
SE: GI
Ranitidine (Zantac)
blocks H2-receptors

use: GERD, peptic ulcer, h/b
SE: GI
Famotidine (Pepcid)
blocks H2-receptors

use: GERD, peptic ulcer, h/b
SE: GI
Omeprazol (Prilosec)
blocks H/K ATPase pump

use: GERD, peptic ulcer, h/b
SE: GI
Pantoprazole (Protium)
blocks H/K ATPase pump

use: GERD, peptic ulcer, h/b
SE: GI
Sucralfate (Carafate)
forms protective barrier

use: ulcers, GI bleeding
SE: GI
Misoprostol (Cytotec)
prostaglandin receptor agonist

use: NSAID-induced ulcers
SE: GI
Bismuth subsalicylate (Pepto Bismol)
forms protective barrier

use: ulcers, diarrhea
SE: Reye's syndrome
weak bases are ionized, T/F?
True, thus more polar and more water soluble
weak acids are ionized, T/F?
False. Weak acids are not ionized and so are less water soluble when they are protonated
RNH3+ <-> RNH2 + H+
Which is more water soluble?
RNH3+; it is a protonated weak base and it is charged; RNH2 is an unprotonated weak base that is uncharged and more lipid soluble
RCOOH <-> RCOO- + H+
Which is more water soluble?
RCOO- because it is an unprotonated weak acid and charged; RCOOH is a protonated weak acid that is uncharged and thsu more lipid soluble
permeation
movement of drug molecules into biologic environment
-aqueous diffusion
-lipid diffusion
-transport by special carriers
-endocytosis/pinocytosis
the smaller the EC50 the greater the potency of the drug, T/F
T
What is the Kd?
the concentration of the drug required to bind 50% of the receptor sites; measures affinity of drug for its binding site on receptor
The smaller the Kd the greater the affinity of the drug for its receptor; T/F?
T
What is EC50?
in graded dose response curves, the concentration or dose required that produces 50% of max
competitive antagonist
binds the receptor in a reversible way without activating the effector system of that receptor (curve will shift to the right-more drug needed to reach max); Effect can be overcome by adding more agonist; competitive antagonists increase the ED50 (agonist appears less potent)
irreversible antagonist
causes a downward shift of the max with no shift to the curve on the dose axis unless spare receptors are present (reduces max)
physiological antagonist
drug that binds to a different receptor producing an effect opposite to that produced by drug its antagonizing
what if you find that the EC50 of a drug is smaller than the Kd?
you have spare receptors
Pharmacokinetics is the study of the actions of the body on drugs.

What are the 4 main concepts of pharmacokinetics?
MADE
M: metabolism
A: absorbtion
D: distribution
E: excretion
What is bioavailibitliy?

how is it measured?
bioavailability is the measure of drug in the systemic circulation.

measured by area under the curve (AUC) of blood concentration of drug over time.
What is first pass metabolism? (aka presystemic elimination)
The elimination of drug that occures before it reaches systemic circulation. (Eg. oral meds in gut/liver)
What are the 5 general ways drugs pass cell membranes? (2 passive and 2 active)
Passive
1) simple diffusion
2) facilitated diffusion (through specific/nonspecific transporters)

Active
1) pumps
2) pinocytosis
What form of the drug penetrates cell membranes best (protonated/unprotonated)?

1) weak bases
2) weak acids
Penetrate cell walls best
1) weak bases : unprotonated
2) weak acids : protonated
At a LOW pH environment like the stomach, what type of drug (weak acid or weak base) is more able to absorb into cells?
At LOW ph : weak acids become protonated and are more able to absorb into cells

weak bases become protonated and are LESS able to be absorbed.
In a relatively neutral pH (like blood or the body's aqueous humor), which type of drug is able to be absorbed into cells more readily, weak acid or weak base?
WEAK ACID drugs are able to absorb more readily than weak base drugs in neutral pH becasue at pH 7:

weak acids become unprotonated (become uncharged)

weak bases become protonated (become charged)
Tissue distribution of a drug depends on what two factors?
1) blood perfusion of the tissue
2) the tissue/blood partition ratio (Kp). Kp= Ctissue/Cven-blood (at equilibrium)
Rank the following tissues in order of most partition ratio to least partition ractio: Fat, Kidney, Brain
MOST to LEAST partition ratio:
1) Kidney
2) Brain
3) Fat
What are 5 unique characteristics of CNS capillary endothelia that make it different than most capillary endothelia?
1) Tight intercellular junctions
2) Lacks fenestrations
3) minimal pinocytosis
4) drug metabolizing enzymes
5) endothelial drug transporters (efflux into the blood)
6) abundant mitochondria (power efflux pumps)
What is VOLUME OF DISTRIBUTION (Vd)?
Given a set quantity of drug:
the volume of fluid the drug would need to be dissolved in to match the concentration of the drug in given volume of plasma.
How do you experimentally measure (calculate) the Vd for a drug?
1) Inject someone with a measured dose of a drug
2) plot plasma concentratio of drug
3) extrapolate time-zero plasma concentration
4) divide the dose by time-zero pasma concentration
There are two phases of drug metabolism. what happens in phase 1?
Drugs are FUNCTIONALIZED, commonly by oxidaiton, reduction or hydrolysis
There are two phases of drug metabolism. What happens in phase 2?
The products of phase 1 are CONJUGATED, commonly with endogenous molecules like glucuronic acid.
What is metabolic inactivation?
converting a pharmacologically active, lipophilic drug to an inactive hydrophilic metabolite (conjugate) which is readily excreted.
volume of distribution
the ratio of the amount of a drug in the body to its concentration in the plasma or blood

Vd= amt of drug (body)/plasma drug concent.
What is metabolic bioactivation?
changing a "prodrug" into its pharmacoliogically active form through a metabolic reation
clearance
the ratio of the rate of elimination of a drug to its concentration in plasma or blood

CL=rate of el of drug/plasma drug concent.
half-life
time it takes for the amount or concentration of a drug to fall to 50% of an earlier measurement. For drugs eliminated by first order kinetics, this number is a constant regardless of concentration. Half life is not a constant and therefore not particularly useful for drugs eliminated by zero order kinetics (ethanol)

t1/2= 0.693xVd/CL
what is metabolic toxicity?

example?
Toxic products of drug metabolism.

eg. acetaminophen through CYP2E1 metabolism in liver
bioavailaibility
the fraction (or %) of the administered dose of a drug that reaches the systemic circulation.
AUC
the graphic area under a plot of drug concentration in plasma versus time after a single dose of a drug or during a single dosing interval; the AUC is imp for calculating the bioavailability of a drug given by any route other than IV
Peak and trough concentration
the max and min drug concentrations in plasma or blood measured during cycles of repeated dosing
minimum effective concentrations (MEC)
the plasma concentration below which a patient's response is too small for therapeutic benefit
first pass effect
the elimination of drug that occurs after administration but before it reaches the systemic circ
steady state
in pharmacokinetics the condition in which the average total amount of drug in the body does not change over multiple dosing intervals (rate of drug input equals the rate of elimination)
extraction
the fraction of adrug in the plasma that is removed by an organ as it passes through that organ
bioequivalence
the equivalence of blood concentrations of two preparations of the same drug measured over time; if the concentration time plots for hte two preparations are nearly superimposable (within certain statistical limits) teh preparations are said to be bioequivalent; one preparation may be safely substituted for the other. this parameter is used in determining the safety and efficacy of generic drugs
biodisposition
often used as a synonym for pharmacokinetics; the processes of drug absorption, distribution and elimination
What families of P450 enzyme (CYP gene) are responsible for a major portion of drug metabolism?
CYP1-3
What are three major human liver P450s that metabolize....
1) weakly acidic drugs
2) weakly basic drugs
3) practically everything else
1) CYP2C2 : weakly acidic drugs
2) CYP2D6 : weakly basic drugs
3) CYP3A4 : practically everything else
What are three major reaction types that occure during phase 1 metabolism?
1) oxidation at C, N, S ceneters
2) Reductive Rxns at C, N, S centers
3)Hydrolysis Rxns (of esters and amines)
What is enzyme induction?

example?
Inc. in steady-state concentrations of an enzyme due to stimulus.

eg (1) inc. in CYP2E1 concentration due to alcoholism (2) Rifampicin induction of P450 -> inc. metabolism (lowers efficacy) of oral contraceptives/warfarin
What is enzyme inhibition?

eg?
Inhibition is loss or dec. of drug-metabolizing enzyme due to stimulus

eg. grapefruit juice degrades CYP3A4, inhibiting metabolic activation of Felodipine (dihydropyridine Ca channel antagonist) preventing its uptake (higher plasma concentrations)