• 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

1/106

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;

106 Cards in this Set

  • Front
  • Back
define clearance
volume of plasma that would have to be completely freed of drug to account for the elimination
how can you determine the total systemic clearance from the clearance of individual organs?
clearance by various organs is additive

CLliver + CLkidney + CLother = CLsystemic
what is the advantage of using clearance?
constant for first-order excretion
what are the units for clearance?
mL/min (volume/time)
what is an extraction ratio?
ER = (CA - CV) / CA

CA = arteriolar concentration of drug
CV = venule concentration of drug
how can you calculate clearance from extraction ratio?
CL = Q x ER

CL = clearance
Q = blood flow
ER = extraction ratio
what is the equation for renal clearance of a drug?

what is the disadvantage of this equation?
CLkidney = [urine] x (VOL/time) / [plasma]

not useful clinically
describe inulin
exogenous
freely filtered
not reabsorbed
CL = GFR
what is indicated if CLin > CLdrug?

give an example
drug is reabsorbed

glucose
what is indicated if CLin < CL drug?

give an example
drug is secreted

PAH
define dosing rate
denotes the amount of drug reaching the systemic circulation per amount of time

how much is being given, how often
assuming complete bioavailability (f=1), when will steady state for a drug be achieved?
when the rate of drug elimination equals the rate of drug administration
what is the equation for steady state?
f x Dosing Rate = CL x [drug]ss

f = bioavailability (1 for complete bioavailability)
CL = clearance
[drug]ss = concentration of drug at steady state
what term is synonymous with steady state?
plateau
what is the equation for Dosing Rate?
f x Dose / Dosing Interval
important equation at steady state, substituting dosing rate into steady state equation
f x Dose / dosing interval = CL x [drug]ss

f = bioavailability (1 for complete bioavailability)
CL = clearance
[drug]ss = concentration of drug at steady state
when is it difficult to achieve steady state quickly?
when the drug of interest has a long half-life
how is steady state achieved quickly for those drugs that have a long half-life?
give one/two priming or loading doses

the priming or loading dose(s) are large doses to get the plasma concentration up to steady state and then maintenance dosing is started
how do you calculate the priming dose for a drug?
f x loading dose = Vd x [drug]plasma

f = bioavailability (1 for complete bioavailability)
Vd = volume of distribution
[drug]plasma = desired plasma concentration of the drug
what phrase is synonymous with fluctuation of drug concentrations?
peak-trough difference
what is the equation for calculating the drug level fluctuation?
ln [M] / [Mo] = -ke x t

[M] = trough concentration
[Mo] = peak concentration
ke = elimination constant = CL / Vd
t = time between doses
CL = clearance
Vd = volume of distribution
for a drug, how do you calculate the time it takes to fluctuate 50% of peak concentration? 25%? 10%?
t1/2 = 0.693 / ke (trough conc. is 50% that of peak)

t1/4 = 0.29 / ke (trough conc. is 75% that of peak)

t1/10 = 0.11 / ke (trough conc. is 90% that of peak)
what is a normal value for glomerular filtration rate (GFR)?
125 mL/min
how does impaired liver function affect drug elimination?
cannot accurately predict which drugs will have prolonged elimination half-lives

cannot accurately predict the extent to which the elimination t1/2 for a drug will be prolonged
how do you account for liver function impairment in dosing medications?
start with low doses and work the dosage up until therapeutic results are achieved

switch to a drug with solely renal excretion
how does renal impairment affect drug elimination of renally-excreted drugs?
can predict (somewhat) accurately which drugs will have prolonged elimination half-lives

can accurately predict the extent to which the elimination half-life will be lengthened

elimination half-life for drugs excreted largely by the kidney correlates directly with GFR
what is creatinine used for?
endogenous substance used to measure GFR
where is creatine synthesized?
liver
how is creatine distributed to the brain and muscle?
via the blood as phosphocreatine (source of high energy phosphate in cardiac and skeletal muscle)
what is the function of creatine kinase?
convert phosphocreatine to creatine as phosphate is used for energy in cardiac and skeletal muscle
what happens to creatine after skeletal muscle has utilized its phosphate?
diffuses into serum, where it is rapidly converted to creatinine
how is creatinine eliminated?
primarily by the kidney, almost solely by glomerular filtration

(hence it can be used as an estimate of GFR)
what is the equation for estimating the renal clearance of creatinine in males?
(140-age)(weight) / (72)(SrCrss)

age in years
weight in kg
SrCrss = serum creatinine in mg/dL (must be at steady state)
what is the equation for estimating the renal clearance of creatinine in females?
(0.85)(140-age)(weight) / (72)(SrCrss)

age in years
weight in kg
SrCrss = serum creatinine in mg/dL (must be at steady state)
what are the two important factors to consider when using the equations which estimate creatinine clearance via plasma creatinine concentration?
1) serum creatinine must be at steady state

2) the weight used reflects normal muscle mass, so ideal body weight must be used for obese individuals
how do you calculate ideal body weight for males?
= 50 + (2.3)(height in inches > 60)
how do you calculate ideal body weight for females?
= 45 + (2.3)( height in inches > 60)
what two actions can be taken to avoid toxicity and to save money?
decrease dose

increase interval
how does fraction of drug excreted by the kidney affect the half life?
drugs that are eliminated primarily by the liver will not have a significant change in half-life even with removal of the kidney

drugs that are eliminated primarily by the kidneys will have a very significant change with damage to or removal of the kidneys

drugs with partial excretion via kidney and partial excretion via liver will change according to what fraction is excreted via kidney when kidney is lost or destroyed
why is there greater variation in response to many drugs in the elderly than in younger adults?
the threshold for side effects is often lowered in elderly patients
what is the pharmacodynamic phenomena that must be considered in the elderly?
for many drugs, a given concentration at an effector site produces a greater pharmacologic response in elderly persons than younger persons

more sensitive to drug's effects
what are the five pharmacokinetic phenomena that must be considered in the elderly?
1) absorption of drugs by GI tract is not usually reduced in elderly

2) serum albumin levels are lower, therefore binding of drugs is reduced and free fraction is increased

3) decline in lean body mass and increase in adipose tissue mass in relation to total body weight in elderly affects drug distribution

4) renal clearance declines in proportion to reduced glomerular filtration

5) changes in liver clearance are difficult to predict
how does age affect the drug distribution of a hydrophobic drug?
as a patient's age increases, muscle is replaced by fat

a hydrophobic drug will distribute into the extra fat very well, so the volume of distribution increases
how does age affect the volume of distribution of a hydrophilic drug?
as a patient's age increases, muscle is replaced by fat

a hydrophilic drug will avoid the fat, therefore the volume of distribution will be decreased in elderly patients
what is the best measuring tool for determining the ability of an elderly patient to eliminate a drug?
clearance

NOT half-life
how is dose-response data usually expressed?
plotting drug effect vs. logarithm of dose
an increase in potency accounts for what type of shift in a drug effect vs. logarithm of dose curve?
right shift
a decrease in potency accounts for what type of shift in a drug effect vs. logarithm of dose curve?
left shift
to what does potency relate?
affinity of drug for receptor
an increase in efficacy accounts for what type of change in a drug effect vs. logarithm of dose curve?
expansion up
a decrease in efficacy accounts for what type of change in a drug effect vs. logarithm of dose curve?
compression down
to what does efficacy relate?
ability of drug to stimulate receptor

sometimes termed "intrinsic activity" of the drug
what type of antagonists are the most common
competitive
what is a competitive antagonist?
an antagonist that binds reversibly at the receptor site, inhibiting the natural ligand from binding
how is competitive inhibition overcome?
increasing the concentration of the agonist, ultimately achiving the same effect
what is the effect of a competitive inhibitor on potency? on efficacy?
potency - decreases

efficacy - unchanged
what is a noncompetitive antagonist?
antagonist that prevents the agonist from producing its effect at a given receptor site by blockading agonist binding or blockading events downstream of receptor stimulation
what is the effect of a noncompetitive inhibitor on potency? on efficacy?
potency - unchanged

efficacy - decreases
describe the change in a Lineweaver-Burk plot for a noncompetitive antagonist
shifts left

x-intercept decreases (more negative)

y-intercept increases

**causes a decrease in max effect**
describe the change in a Lineweaver-Burk plot for a competitive antagonist
line doesn't shift (rotates CCW)

x-intercept increases (less negative)

y-intercept is unchanged

**causes no change in max effect**
what are the dependent and independent variables on a Lineweaver-Burk plot?
dependent - 1/effect

independent - 1/Dose
what are the x- and y-intercepts on a Lineweaver-Burk plot?
x-intercept = - 1/KD

y-intercept = 1/Max Effect

KD - drug concentration at which half of the drug is bound
what is a partial agonist?
molecule which binds to a receptor and produces a lower maximal response than a full agonist

binds and produces a response, but not the strongest response
in a log dose v. response curve, what is the effect of a partial agonist?
compresses curve down

efficacy is decreased;
potency can be greater, equal or less than a full agonist
how are partial agonists explained?
receptors are in equilibrium between an active (Ra) and an inactive (Ri) form

drugs alter the equilibrium rather than stimulating receptors

partial agonists have a weaker affinity (preferential binding) for Ra than do full agonists, so it produces a weaker biological response than full agonists
how does a full agonist affect receptor equilibrium?
has a strong preferential binding for Ra (active form), which drives the equilibrium toward the active form (Le Chatelier's principle), and leads to a stong biological response
what is the chemical equation for the explanation of full and partial agonists?
D:Ri <-> Ri <--> Ra <-> Ra:D

D - drug
Ri - inactive form of receptor
Ra - active form of receptor
how does a partial agonist affect receptor equilibrium?
has a slight/weak preferential binding for Ra (active form), which drives the equilibrium toward the active form (Le Chatelier's principle) only slightly/weakly, and leads to a weak biological response
how does a blocker affect receptor equilibrium?
binds to either Ra (active) or Ri (inactive) with similar affinity which leads to no change in the system's equilibrium

antagonism when a full agonist is present
what happens when a drug binds preferentially to the inactive form of a receptor?
may further inhibit the system, but will be difficult to see since Ri is already the dominant form
for receptor equilibrium, give the equation and tell what is the dominant form under equilibrium conditions (no drugs present)
D:Ri <-> Ri <--> Ra <-> D:Ra

D = drug
Ri = inactive form of receptor
Ra = active form of receptor

inactive form of receptor is dominant when no drugs are present, but equilibrium is changed with addition of drugs
what type of antagonist is a partial agonist considered to be?
competitive antagonist

decreased potency of full agonist when partial agonist is added, but efficacy remains the same
what are "spare rceptors"?
if the maximal activity of a drug can be reached before all of the receptors are saturated, some receptors remain empty and these receptors are known as spare receptors
if there are 100 receptors for a drug and 70 of them are bound at maximal drug effect, how many spare receptors are there?
30 spare receptors

spare receptors = total receptors - receptors bound at maximal effect
how are spare receptors usually exemplified experimentally?
for low-moderate concentrations of antagonist, appears as competitive inhibition (same efficacy, decreased potency), but once the antagonist concentration reaches a point where it saturates enough receptors that there aren't enough to reach maximal effect, the agonist loses efficacy
what is the advantage of spare receptors?
increased sensitivity to low concentrations of receptor agonist
if a drug's concentration is sufficient to bind 25% of available receptors, and there are three intracellular effector molecules, what will be the percent of Emax when there are four receptors?
33%

though there are more receptors than effectors (one spare receptor), only 1/4 of the receptors is bound to agonist and binds the effector molecule

1/3 effectors working = 33% of Emax
if a drug's concentration is sufficient to bind 25% of available receptors, and there are three intracellular effector molecules, what will be the percent of Emax when there are sixteen receptors?
100%

4/16 receptors are bound to agonist, but only three receptors can bind to effector molecules since there are only three effector molecules (16 receptors - 3 effectors = 13 spare receptors)

0.25 x 16 = 4 receptors bound by agonist
if a drug's concentration is sufficient to bind 25% of available receptors, and there are three intracellular effector molecules, what will be the percent of Emax when there are twenty receptors?
0.25 x 20 = 5 receptors bound by agonist

only 3 effector molecules

therefore, 100% Emax with 17 spare receptors
if a drug's concentration is sufficient to bind 25% of available receptors, and there are three intracellular effector molecules, what will be the percent of Emax when there are eight receptors?
0.25 x 8 = 2 receptors bound by agonist

2/3 effector molecules utilized

66% Emax with five spare receptors (8 receptors - 3 effector molecules = 5 spare receptors)
what are the independent and dependent variables in a quantal log dose-response curve?
independent - dose (mg)

dependent - number responding (NOT magnitude of effect)
what is ED50?
effective dose in 50% of subjects
what is LD50?
lethal dose in 50% of subjects

(NOT 50% of lethal dose)
how is therapeutic index calculated? how is it more commonly seen in clinical settings?
TI = LD50 / ED50

given in qualitative fashion (e.g. wide therapeutic window) because the numerical therapeutic index is not clinically useful
why are barbiturates dangerous?
a moderate dose puts a person to sleep (hypnosis) but just giving a little more can cause them to stop breathing
what are the major types of drug receptors?
ligand-gated ion channels

G-protein coupled receptors

transmembrane receptors with enzymatic cytosolic domains

steroid receptors
what is the most famous ligand-gated ion channel?
ligand-gated nicotinic acetylcholine receptor
how does the nicotinic acetylcholine receptor function?
the receptor forms a transmembrane channel, that permits the passage of Na from outside the cell in only when ACh is bound to the receptor; when this happens the cell is depolarized

(ligand-gated ion channel)
how does the GABA-subA receptor function?
receptor forms a transmembrane channel, that permits the passage of Cl from outside the cell in only when GABA is bound to the receptor; when this happens the cell is hyperpolarized

(ligand-gated ion channel)
what is the most common type of receptor?
G-protein coupled receptor
describe the G-protein in the resting state
alpha and beta:gamma subunits are associated with one another, and GDP is bound to the alpha subunit
what causes the exchange of GTP for GDP on the alpha subunit of a G protein?
binding of an extracellular ligand (agonist) to a Gprotein-coupled receptor
what happens to a G protein after GTP is exchanged for GDP on the alpha subunit?
the beta:gamma subunit dissociates from the alpha subunit and the alpha subunit (bound to GTP) diffuses to interact with effector proteins
how is GTP hydrolysed to GDP in G proteins?
alpha subunit has intrinsic GTPase activity, which leads to hydrolysis of GTP to GDP
what causes reassociation of the subunits of a G protein?
alpha subunit intrinsic GTPase activity hydrolyses GTP to GDP and the subunits reassociate, so that the cycle can start again
in the heart, what is the effect of epinephrine binding to its G protein-coupled receptor?
alpha subunit of G protein activates adenylyl cyclase, which catalyzes the conversion of ATP to cAMP, which activates PKA, which causes protein phosphorylation, which increases heart rate
in the vascular smooth muscle, what is the effect of epinephrine binding to its G protein-coupled receptor?
alpha subunit of G protein activates PLC, which catalyzes conversion of PIP2 to DAG and IP3

IP3 stimulates release of Ca from the ER

DAG activates PKC, which works with Ca to phosphorylate proteins, which causes vascular smooth muscle contraction, which increases total peripheral resistance
how are different alpha subunits of G protein-coupled receptors denoted?
different subscripts

Epi receptor in heart - alpha-subs
Epi receptor in vascular smooth muscle - alpha-subq
what are two important examples of tyrosine kinases?
insulin receptors

BCR-Abl receptor
how do tyrosine kinase receptors function?
after ligand-induced activation, these receptors dimerize and transphosphorylate tyrosine residues in the receptor and, often, on target cytosolic proteins
what is the cause of chronic myelogenous leukemia?
BCR-Abl, the tyrosine kinase, is always turned on
how do steroid receptors function?
small lipophilic molecules can diffuse through the plasma membrane and bind to intracellular transcription factors

ligand binding triggers a conformational change in the receptor and dissociation of a chaperone that leads to transport to nucleus

in nucleus, the ligand-receptor complex typically dimerizes and then binds to DNA, recruiting coactivators or corepressors

these complexes alter the rate of gene transcription, leading to a change (up or down) in cellular protein expression
where can steroid hormone receptors be located?
cytosol

nucleus
what is a chaperone?
repressor protein bound to a steroid hormone receptor that prohibits it from travelling to the nucleus until a conformational change is induced by the steroid hormone ligand
what happens to a steroid hormone:receptor complex in the nucleus?
typically dimerizes into a homodimer or a heterodimer
what is an example of a steroid hormone:receptor heterodimer?
thyroid hormone receptor with the retinoid X receptor