• 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

1/113

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;

113 Cards in this Set

  • Front
  • Back
What is definition of pharmacology?
Study of PK, PD, PT; study of action and reactions of drug therapies
What is pharmacokinetics?
Study conc. of drug in plasma during absorption, disturbution, metabolism, elimination
What is pharmacodynamics?
Study of physiologic mechanisms where drug produces intended therapuetic effect
What is pharmacotoxicology?
Study of physiologic mechanisms where drug produces toxic/adverse effect
What is pharmacogenetics? How does affect future drug treatments?
Study of genetic variations that cause differences in drug response among patients. Doctors may have to screen and adjust for these variations because pts can higher sensitivity to standard doses from genetics that decrease activity of enzyme that eliminates drug
Differences between gene "knockout, knockdown, & knockin"?
-out: breed homozygous animals where gene for receptor is absent or nonfunctional
-down: breed heterozygous with only limited suppression of function
-in: overexpress certain proteins of interest have been bred
What is difference between trade vs. brand name? What is the formulation of drug?
trade is patented trademark of drug; generic is common drug name but non-capitalized; formulation is to check for allergies or issuesof drug make-up
What are the three fundamental characteristics of drugs?
drugs only modify pre-existing functions, have multiple sites of action (for side effects/therapuetic uses), & dose is difference between drug vs. poison
What are the four FDA criteria for drug approval?
1. placebo controls
2. double-bind protocols
3. subjects randomly selected and appropriate for study
4. establishment of drug safety (acute vs. chronic toxicity)
What are four problems/controversies with new drug development?
animal models, genetic variation, uncontrolled variables, time and cost of drug development
What are the three main ethical issues related to drug tx & development?
automony (choice), beneficence (benefit patient), & justice (equal access)
What is the differences between phases I-IV of clinical trials?
I- low dose studies on healthy volunteers; II- sm group with dose response studies to establish efficacy; III- lg, multi-center dose response studies to check for eff. and toxicity; IV- post approval monitoring of drug on the general population
What will decrease a drug's availability to target tissue, also called barriers?
Protein binding and tissue deposits.
What is an absorption profile?
Change of concentration of drug in blood plasma over time. Should have concentration and time.
What three variables are present at peak drug concentration on the absorption profile?
Absorption, distrubution, and elimination.
What does MEC and MTC mean and why are they important?
MEC-minimal effective concentration. Minumum dose needed to see therapeutic effect.
MTC- minimum toxic concentration. Concentration when toxicity is seen.
What are the C max and T max? Where are these points located on the oral absorption profile?
C max= actual max "C"onc. of drug that peaks after administration; located at the peak of the curve from L side of graph (conc.)
T max= "t"ime takes after administration to reach peak max conc. in blood plasma; located at peak of curve down to designated time on graph
What is the onset time and duration of action?
Onset time-measure of time drug takes after admin. to work at MEC
Duration of action- elapsed time remains above MEC or how long produce therapuetic effect
Explain what is half-life (T 1/2) of a drug.
Measure of elapsed time it takes on absorption profile for drug to go from one conc. to 1/2 its conc.
dependant on drug absorption and usually is between MEC and MTC.
What is the AUC?
Area under the curve- total amt of drug enters into bld after administration, reflects PK of drug (characteristics of absorp profile), time drug appears till disappears
What is bioavailability and how is it measured?
% of total drug dosage that enters blood for available use; measured by AUC (mg)/dosage (mg) X 100, the remaining amt is loss % of drug
What are two reasons oral bioavail. of most drugs is less than 100%?
incomplete extent of absorption and first-pass elimination
How does route of administration influence bioavail. of a drug? What are alternative routes of administration to avoid first-pass metabolism?
(hepatic elimination)-the overall (oral) process of drug being absorbed/metabolized via gut wall and portal blood (liver, bile) before hits systemic circulation, which can decrease bioavail. of drug; alt routes: IV, subling, IM, transdermal, inhalation, SC
What is the onset time for IV route of administration and why?
No onset time because no delay in absorption in blood plasma due direct absorption; initial dilution/mixing in 1min-> distrubtion within 30min-> elimination begins after 30min
What happens during the initial mixing concentrations following bolus IV administration?
Bolus is not completely mixed with blood, with high initial conc. of drug and will come down as mix with blood/begin to dilute; can be toxic prior to mixing with blood with initial dose in first min. due to initial dose
Why are IV bolus doses contraindicated in most drugs? Give two example of these drugs.
the initial high dose side-effects they produce,
e.g. isoproterenolol can cause cardiac dysrrhythmias; thiopentol can induce respiratory depression/arrest.
What drug is one of the exceptions to IV bolus contraindication principle and why?
phenytoin- bolus doses for control of seizures; highly bound to plasma binding proteins so initial conc. are buffered by this binding
What two things are mainly affected during the absorption profile of tetracycline when given with dairy vs. tetracycline alone? Why?
C max and AUC would be reduced; related to absorption issues from calcium in dairy product that causes precipitate in gut that prevents absorption of drug
What is bioequivalence? What are the two main criteria?
guideline that requires that two preparations of the same drug (trade vs. generic) have identical AUC and C max values; usually suggests that drugs will have same biological effects
What are the four mechanisms of drug absorption (permeation)?
1. Diffuse passively thru aqueous channels in between cells (tight junctions)
2. Through lipid cell membranes
3. If drug has appropriate characteristics, may be moved by carriers into and out of cell
4. Bind to cell surface receptors inside cell by engulfing into IC space and expelling into EC space
via cell membrane
What is plateau principle and how can it be affected by the frequency of administration?
When drug given over equal periods of time to reach plateau conc. in a steady state (goal bet. MEC & MTC); rate of absorption becomes = to rate of metabolism and elimination (the balance of PK); Very important to follow the appropriate frequency for therapeutic effect and try to prevent potential of toxic dosage or blood levels will be all over place
What is chronopharmacology?
study of drug effects that are timed to body's biological rhythm
Discuss the drugs cisplatin, "statins", and glucocorticoids on chronopharmacology.
cisplatin-chemo drug given at pm R/T when UO is maximal to reduce renal tox.
"statins"-given in evening when rates of cholesterol synthesis in liver are highest
gluco- given in am when cortisol production naturally peaks making more alert and dec. some side effects
How does drug formulations affect absorption in tablets, coated tabs, and capsules? What are examples?
to regulate absorption by changing formulations;
tabs- asa, compressed powder that quickly breaks in stomach
coated tabs- triaminic, coated that passed stomach and dissolves slowly in intestine
caps- Darvon, gelatin covered to dissolve in either stomach or intestine
What happens to absorption when coated tabs are chewed/crushed vs. swallowed?
Get different absorption which goes above MTC when change the way drug taken (chewed- more quickly vs. swallow- slows absorption to intestines of EC tabs)
What variables of an absorption profile (conc. vs time) would be most profoundly effected by a drug that is administered in oil vs. saline vehicle?
Onset time because prolonged absorption and dissolves slowly with oil vs. rapidly absorped with NSS
What are three drug vehicles?
NSS, oil, epinephrine (localized vasoconstriction for slow absorption and prolonged local effects
What is difference between drug-antibody conjugates and liposomes with examples?
conjugates- e.g. smart drugs that chemically link to antibody to help to target specfic cancer cells
liposomes- artificial membrane vehicles that encapsulate drug (amp B to reduce renal tox)
What is molecular weight vs. lipid solubility?
mol wgt- larger size of molecule wgt more difficult/slower rate absorption (types of insulin)
lipid solubility- more soluble faster onset of action and rate of absorption (barbituates)
What is difference between strong and weak acids?
strong- HCL, H+ ion donor released ions to solution to make more acidic, lower pH
weak- asa, can release or pick up H+ (reversible)
What is difference between HA vs. A- for acid-base characteristics?
HA= unionized, lipid soluble of drug
A- = ionized, lipid insoluble
What is pKa?
equilibrium constant defined as pH (H+ concentration) at which the HA (unionized)concentration is equal to the A- (ionized) concentration; pH to keep it neutral/balanced; low pH=high H+
How will weak acid respond in different environments (acidic, basic, pKa)?
pKa=balanced, equal
Acidic- forces into unionized state, more readily soluble-more H+ ions
Alkaline- forces into ionized state, less soluble-less H+ ions
Because moves from higher to lesser conc.
Describe method of how can find out how weak acid responds in different environments.
1. look at pKa and pH; acid & acid= non-ionized; base & base= non-ionized
2. anything else is ionized
3. must decide if pH is more acidic or basic compared to your pKa
At what point in absorption profile does drug metabolism and elimination dominate blood level of drug?
Can be measured at half life (T 1/2), which is rate of how drug disappears
What will happen to the absorption of a weak acid across walls of stomach when also given will proton pump inhibitor to treat ulcer?
The absorption of weak acid will be decreased related to the proton pump inhibitor making the stomach acid more basic.
What happens with lipid solubility and onset times of various (3) barbiturates?
thiopental: highly lipid soluble; onset seconds to minutes (induce quick anesthesia)

pentabarbital: moderately lipid soluble; onset minutes to hours (can induce sedation and hypnosis)
phenobarbital: low lipid solubility; onset hours to days (anti-sz med)
What is the difference between B vs. BH+ in acid-base characteristics?
B is the unionized, lipid soluble form of the drug

BH+ is the ionized, lipid insoluble form of the drug

Base neutralizes acids by being H+ acceptor
Lidocaine is a weak base; how would this characteristic affect drug absorption and fetal exposure to this drug during gestation?
Lidocaine would accumulate in the fetus and potentially become toxic

(see flashcard on seperate sheet for drawing)
What are (4) biological variables that influence the rates and extent of drug absorption?
1. GI contents
2. Presence of intestinal flora
3. GI functions
4. Availability of transport proteins
What would happen to the absorption of L-DOPA if it were administered orally prior to the ingestion of a meal that contained proteins?
Would decrease absorption because proteins from food would take up or compete for the a. a. transport proteins.
What GI functions (3) can affect absorption?
vomiting and diarrhea can alter drug absorption;
stress can reduce blood flow to the G-I tract and reduce drug absorption
How long does it take to reach steady state or plateau after a drug is administered?
4-5 half lives. Thats why we give scheduled doses. Frequency of drug admin is very important to main steady state.
Give an example of why intestinal flora affects drug absorption.
due to bacterial metabolism, the bioavailability of digoxin is 60-80% while the combination of digoxin and some antibiotics increases the bioavailability of digoxin to near 100%; this effect results in an increase the incidence of digoxin side-effects;
Give an example of why GI contents affect drug absorption.
dietary fibers bind to digoxin and reduce its absorption; some antibiotics when ingested with dairy products form calcium precipitates that reduces drug absorption;
How long does it take to reach steady state or plateau after a drug is administered?
4-5 half lives. Thats why we give scheduled doses. Frequency of drug admin is very important to main steady state.
What are four things that can affect distribution?
plasma binding proteins, blood flow, tissue deposits, barriers
Discuss the importance of the barriers in relation to distribution (BBB, blood testes, placental).
BBB-no space for drugs to pass thru capillary so they must diffuse thru the membrane; increase permeability occurs with inflamm/infections and young/elderly

everywhere else (testes and placental) have a nonbrain capillary with space between endothelial cells.
If you had to give a pregnant woman an anti-coagulant, would you give heparin or coumadin and why?
Heparins with MWs > 1000 do not pass placental membranes
While warfarin which has a MW of less than 500 passes placental membranes and is a fetal toxin;
What does polarity and lipid solubility of drug have to do with distribution of drug to fetus?
Insoluble are highly polar drugs- do not pass placenta membranes (e.g. succ.)

lipid soluble so more unionized so readily absorbed and becomes toxic in a fetus (e.g.- thiopental)
Name the two forms of plasma binding proteins and the differences between them.
Bound- “DP” large biologically inactive proteins; stays contained in plasma, unable to leave walls of capillaries

free- biologically active to circulate thru blood plasma and able to leave to go to target tissues
A drug that circulates in the blood in its highly bound form to plasma binding proteins wld be expected to have shorter biological half life than drugs that circulate in their unbound form?
Disappears more slowly in blood because it is bind (inactive) and harder to eliminate until converts into free form, longer half life so strongly disagree
What happens when ASA (50% B) and warfarin (99% B)are given together in relation to being both bound drugs?
ASA knocks warfarin off of its binding state making it more free form to be absorbed in blood, which increase toxic effects of increasing bleeding
What can malnutrition, liver, or kidney diseases do to drug responses in variations of PBP's?
reduce blood levels of PBP’s and increase drug toxicities because would increase the free form
What is biological importance of plasma binding proteins?
Initial concentration (displacement) will increase (free form) which can be toxic, but after period of time it will change its concentration and equal out with metabolism and elimination, Timing is Key!

Helps with interpretation of measured drug concentrations
Name the (4) major tissue deposits and the role.
(1) adipose tissue = 15-50% of body wt.
(2) muscle
(3) bone/teeth
(4) placenta/mammary tissue, etc.

By taking up the drug they reduce drug that is in plasma that is available to target tissues
How does blood flow affect distribution of drug?
more blood=more drug

very rich group (VRG: brain, liver, heart); muscle group (MG: skeletal muscle); very poor group (VPG: fat, skin, bone, ligament, hair, teeth)
Biological importance of tissue deposits.
Serves as drug reservoir, alter drug dosage, sites of drug interactions, sites of drug toxicity
What is the difference between loading/induction/priming and maintenance doses?
loading or induction doses of a drug are given to saturate tissue deposits with a drug and to maintain MEC

Maintenance- maintain MEC after saturation of tissue deposits
Explain why the Vd for digoxin changes in the elderly and explain how this change might alter the toxic side-effects of this drug. Speculate on how anoxexia nervosa might alter the toxicity of digoxin in a patient.
Elderly has less muscle mass and dehydration which is decrease Vd because digoxin is usually stored in tissue deposits it wld make higher concentrations of drug in plasma which could lead to higher toxicity of drug

Same for anorexic patients (dehydration) and less tissue deposits
What is volume distribution and how is it measured?
total amount of a drug that is in body (mg) is divided by the concentration of the drug in the blood plasma (mg/l) an apparent volume distribution can be calculated (l).
What are the types of PBPs?
1. albumen-most abundant
2. globulins; sex steroid binding globulins
3. alpha
4. lipoproteins
What is the relationship betweed Vd. and PBPs?
Highly bound has less Vd- higher conc. in bld plasma

Weakly bound has high Vd- most goes to tissues so when bld taken, bld conc of drug looks more diluted in lg vol.
What are the phases of drug metabolism?
phases = Phase I: CYP-catalyzed oxidation or hydrolysis

Phase II: conjugation reactions
What are the differences between the (3) types of metabolic enyzmes?
CYP- Takes a drug and convert to a metabolite

1. catalyzed oxidation- Bind to drug and interact with other enyzmes, combine with O2 to form oxidated form of drug

2. hydrolysis- Genetic situations when taking local anesthetic and pt doesn’t have enzymes to break drug down
Uses water to break hydrogen bonds

3. Conguation reactions- Take a drug and join to certain molecules to for conjugation product which then becomes biological inactive and increases water which increase rate of elim.
What is difference between inducing and inhibiting enyzmes? Give examples.
Inhibiting agents are CYP inhibitiors (cimitidine,quinidine,verapamil,
grapefruit juice)

Inducing agents induce CYP (phenobarbital,phenytoin,rifampin,tobacco,troglitazone,St. John’s wort,Ethanol)
What is the importance of drug metabolism?
drug biotransformation: alterations in the chemical structure of drugs that leads to:
1 drug inactivation: most drugs

2 drug (pro-drug) activation: e.g. loratridine (Claritin) and naproxen

3 drug bioconversion; e.g. some anabolic steroids can be converted to estrogens

4 production of toxic drug metabolites (e.g. tylenol)
Explain relationship of acetaminophen and to depletion of glutathione in liver. How does OD tylenol result in liver failure-draw picture.
Basically nothing for glutathione to bind with anymore after tylenol –accum of covalent bonds
Chemical metabolite accumulates and cause bonds in liver proteins thus destroying leading to liver failure
Draw absorption profiles for theophylline and show how the Cmax and the T1/2 would differ between a non-smoker and smoker.
theophylline would be higher c max and longer duration of action to non-smoker vs. smoker with increased cyps causing faster elimination
What does sulbactam inhibit in relation to drug manipulation of metabolism?
Inhibits the area of resistance or enzyme (beta-lactamases) that bacteria begin to do to resist pcn form working thus pcn is able have therapeutic effect
What are some things that can affect metabolism?
Ethnic diversity, gender, individual genetic variability, induction and inhibition of metabolic enzymes, diseases/injuries, occupation, food/prep (char-broiled)
What is a heptan and what does it have to do with individual genetic variability of metabolism for PCN?
some drug metabolites are converted to heptans that
creates a foreign protein complex that induces allergic responses
The difference in CYPs inherited thus when pcn given body makes heptan which body thinks is foreign and causes allergic response
What is drug resistance?
Cancer will begin to destroy metabolic enyzmes causing to be metabolized out of body faster thus decreasing therapuetic response
Rate of renal elimination is determined how?
= GFR + secretion - reabsorption
Discuss the importance of renal clearance.
Volume of bld cleared of drug by kidney per unit of time- effects kidney function
One strategy for reducing side effects produced by OD of amphetamines would be to increase its rate of excretion. Amphetamine is a weak base and thus its excetion could be increased by:
Making urine more acidic
Draw absorption profile of pcn and indomethacin with/without probenecid
Secretion if from blood into nephron thus if inhibit secretion it will increase duration of action and half-life of pcn
What happens to the rate of elimination in ionized/insoluble weak acids/bases?
Ionized base/acid is increased in excretion in kidneys because insoluble form
What effects does renal diseases have on drug excretion?
alterations (reductions) in renal blood flow (RBF) or
glomerular filtration rates (GFR) increases drug toxicities by increasing the Cmax and T1/2 of drugs
Who would be more sensitive to liver toxicity from tylenol (ped or adult) and why?
Ped because of cyps and liver not fully developed yet to help conjugate the tylenol
What functions does conjugation enzymes have in the control of drug activity and elimination?
Rate of excretion increases and conjugates are inactive
The half life of a drug given to a neonate would likely be extended when compared to the half life in a healthy young adult.
Liver and kidney not fully matured to excrete so strongly agree
Is it safe to assume that all the genetic diversity in drug metabolism is the result in differences in CYP enzymes? What about succinylcholine; genetic variation?
CYP's are NOT only genetic variations; another example is succ. (lead to MH)
What is the measurement difference between absorption and dose-response curves?
Conc vs. time= absorption curve
Dose of drug vs. intensity of response that a drug produce = DR curve
What is the definitions of ED 50 and ED 100?
ED 50- a measure of drug at ½ of max effect (effective dose)
ED 100- lowest dose of drug that produces max effect; if 200x or 300x dose will not effect max effect but will increase toxicities
Explain the relationship of ED 50 vs. potency of drug?
Lower ED 50, higher the potency
What is intrinsic activity in relation to the target tissues?
Compare height of ED 100 (measure biological effect); measure of the magnitude or intensity of a biological responserelative to the maximal attainable response that can be produced in a target tissue (alpha=1.0).
Define the classifications of drugs based upon their intrinsic activity: agonist, partial agonist, antagonist
agonists: drugs with high intrinsic activities (close to 1.0)
partial agonists: drugs with mid-range intrinsic activities (alpha = ~0.5)
antagonists: drugs with little or no intrinsic activity; often called “blockers” (eg. Beta-blocker)
On a single figure, draw two dose-response curves; one for drug A and the second for drug B. Show how drug A would differ from drug B if drug A has a higher potency than drug B and drug B has a higher intrinsic activity than drug A.
see notecard
What is therapeutic index for drugs?
measures drug safety; LD50/ED50; the higher the value (distance) for the therapeutic index the safer the drug
LD- lethal dose 50, usually done in animals for toxic effects
Potency, intrinsic activity, and safety is what dose-response curve discusses
What is the difference between the types of drug target tissue interactions (2)?
structurally non-specific interactions= mainly physical presence in a target tissue is all that's necessary; e.g. osmotic laxative or diuretics

structurally specific interactions= chemical
structure of the drug is essential for its biological activity because the drug generally forms reversible chemical bonds with some component in a target tissue; e.g. receptors and drug receptor can fits like a “lock and key”
What happens to affinity and potency related to the lower the concentration of drug that binds to its receptor?
Higher the affinity (lower dose it takes to occupy/saturate receptor) and higher its potency on a dose-response curve (low dose to produce its effect)
affinity and potency both have inverse relationship to drug concentration or dose
A drug antagonist (low efficacy) can have a high affinity of a receptor?
strongly agree but there is NO cause and effect relationship so can be any make-up
Discuss agonist, partial agonist, and antagonist on efficacy.
Bind the receptor but doesn’t activate drug-antagonist (wrong key)-little or no effect
Bind the receptor changes shape slightly-prt agonist (valet or maid key)-mod. effective
Bind the receptor lock and key-agonist (house or car key)-highly effective
What is efficacy dependent on?
the ability of a drug to induce a conformational change in the shape of receptor that leads to S.T. and the biological response.
What is when receptor is in activated form, can produce sm observable effect in downstream mechanisms, even with absence of drug?
Constitutive receptor activity
Discuss allosteric binding in relation to benzos.
drugs that bind to the same receptor molecule but do not prevent binding of agonist, may enhance or inhibit action of agonist; benzo’s- bind non-compet. to ion channels activated by neurotransmitter GABA, enhancing net activating effect of GABA on channel conductance
What has a higher affinity for receptor inactive form, which reduces constitutive receptor activity and may produce a contrasting/opposite physiologic result?
Inverse agonist
What two different responses does epinephrine produce and why?
coronary vasodilation and renal vasoconstriction
because of different types of receptors that cause these different actions
What is an example of mechanism for allowing a target tissue to change its response to a drug?
the “pre-term” uterus responds to ritodrine while the “term” uterus does not respond to ritodrine
Some receptors differ at different periods of times
How does dopamine allow a drug produce an ordered sequence of responses?
Dopamine changes responses that are ordered by the desired doses and subset receptors
What are some examples of "up" regulation numbers of receptors?
some breast cancers (HER), hyperthyroidism (tachy/cardiac tissue), propranalol (rebound HTN)
What are some examples of "down" regulation numbers of receptors?
NIDDM (hyperinsulinemia/resistance), albuterol (tolerance)