Pharmacology-Unit 1 Flashcards
pharmacology
study of drug interactions (chemical substances) with biological systems
pharmacotherapy
selection of right drug/dose to interact with right target to produce a therapeutic effect
what are the therapeutic effects of pharmacotherapy?
prevention, diagnosis, treatment, cure of a disease
pharmacokinetic and pharmacodynamic principles allow what determination?
relationship btw. Dose of drug, plasma concentration (Cp) and clinical effect from that Cp
drug effects are directly correlated for what in clinical use?
Cp (plasma concentration)
graphs of what determine pharmacokinetics?
Cp vs. time
what is the MEC?
minimum effective concnentration
what can MEC be determined for?
for the therapeutic/desired response and adverse responses
onset of effect
time to reach MEC
duration of action
time above MEC
therapeutic window
difference in Cp btw. Desired and adverse response MEC
what is the goal of pharmacotherapy?
multiple doses are administered to maintain plasma concentrations at steady state in therapeutic window to produce responses with minimum toxicity
steady state
contition where rate in=rate out (elimination)
time to steady state
attained in 4-5 half lives when using maintenance doses at constant intervals
steady sate concentration
average Cp after SS is achieved
fluctuations in steady state Cp
the related number of half lives in dosing intervals (time btw. Doses)
dosage regimens are designed to ensure what?
steady state drug level is maintained in the therapeutic window
how do dosage regimens maintain a therapeutic window?
by balancing rate of elimination with prescribed rate of administration
how do you select drug and dose?
by looking at: pharmacodynamics–disease targets–drug regulation
how do you select a route of administration?
look at pharmacokinetics: absorbtion–distribution
how do you select drug duration?
base on disease pathophysiology
how do you select dosage frequency?
look at pharmacokinetics: metabolism–excretion
What should you see on a prescription?
drug, dosage, frequency, route, duration
pharmacodynamics is what?
what the drug does to the body, the mechanism of action
what does pharmocodynamics allow you to identify?
drug target and the therapeutic category of the specific drug
what are common drug targets?
intracellular receptors, enzymes, membrane transport proteins
do drugs have unique actions in the body?
NO! They enhance/blodk normal physiology of the organ systems
pharmacokinetics is what?
what the body does to the drug
what does pharmacokinetics provide a physician with?
information on absorption, distribution, elimination of drugs needed for dosing regimens
bioavailibilty (F)
how much of the dose of the drug reaches it’s target
time to peak effect (Tmax or Cmax)
how fast does the drug reach its target
volume of distribution (Vd)
what dose (mg) is needed to obtain desired Cp
absorption
passage of drug from administration site to blood
distribution
movement of drug from bloodstream to tissues
what must be considered when looking at distribution?
drug protein binding, passage across BBB/placenta, selecticve accumulation affecting efficacy/toxicity
route of administration
site of application of drug into/on the patient
what is a systemic effect?
absorbed into bloodstream for distribution ot sites of action in body
what is a topical effect?
remain at the site of application for local action
what is half life? (t1/2)
how long a drug stays at its target (duration of action)
what is elimination?
elimination of drug activity following administration. Can be done via metabolic or excratory paths
CL
clearance
describe a path of elimination/excretion
drug is absorbed into bloodstream, is distributed to site of action and organs of elimination (liver-metabolism, kidney-excretion)
what is the rate of elimination?
the length of time the drug remains in the bloodstream to exert clinical effects
therapeutic uses derive from?
mechanism of action–>Drug Target
how do drugs act?
via enhancement or blockade of physiological paths to alter an abnormal pahtophysiological disease state
are adverse reactions predictable from drug target mechanisms?
YES! Via mechanism of action
adverse reactions at a non-target system
side effects are seen at therapeutic doses, are dose-dependent and predictable
adverse reactions at target system
extension effects seen at higher than therapeutic doses (dose dependent and predictable)
adverse reactions not at drug target
idiosyncratic reactions (peculiar to individual). Less predictable/common
immunoligic reaction
allergic reaction
metabolic reaction
in origin (hepatotoxicity or blood dyscrasias)
what should you know about specific drugs/categories?
most common and severe effects
physiology of absorbtion
pharmacokinetic processes involving passage of drugs across membranes
what influences drug membrane passage?
molecular size, lipid solubility, degree ionization, concentration gradient
molecular size
can be affected by drug binding to plasma proteins
lipid solubility
estimated by oil:water partition coefficient
degree ionization
affected tissue pH, influences lipid solubility
concentration gradient
created at site of administration
list the mechanisms of membrane passage
passive diffusion, carrier mediated diffusion, endocytosis/exocytosis
passsive diffusion is driven by what?
driven by concentration gradient
aqueous diffusion/filtration
limited capacity, size varies, drugs
lipid diffusion
favored for drugs w/ high lipid:water partition coefficient, pH dependent, unionized moiety crsossed down conc. gradient. Drugs 500-800mw
carrier mediated diffusion
specialized transporters regulating entry/exit. Some transport foreign chemicals including drugs
endo and exo cytosis
vesicles that bring things in (vit B12 and iron) or release (neurotransmitters)
what does the effect of a drug on Cp depend on?
rate (time from peak to peak) and extent (bioavailability) of transfer of drug from administration site to blood
what is bioavailability (extend of absorption, F, f[%])?
the fraction of unchanged drug reaching systemic circulation after administration
how is bioavailability determined?
compare area under curve (AUC, from Cp vs. time) folloiwng a single dose of drug between IV and oral routes
what information is readily available for most drugs on extent of absorption?
bioavailability of oral route
what does knowledge of bioavailablity of a drug allow for?
dosage adjusmtents when giving the drug by a different route
IV administration
fraction of dose reading general circulation (F)=100%
oral administration
F varies from 100-0%
what does oral administration F depend on?
survival of GI environment, ability to cross GI membrane (lipid solubility, size, ionization state), efficiency of drug in gut wall/liver, variation btw. Patients
what is first pass effect?
a drug’s metabolic efficeincy in the gut wall/liver
what are some other routes of drug administration?
systemic drug action via IM, SC, inhalation, sublingual
what is the F for other routes of drug administration?
~100% (75-100%) since tissue is non-destructive to drugs
what is the rate of absorption estimated as?
the peak Cp or time to attain peak Cp plasma levels
what is the rate of absorption for oral route?
affected by preparation (liquid or rapid disentigration=faster; enteric coas or sustained release=slower), this changes time and Cp max
what is the rate of absorption for parenteral routes?
determined by the route rather than drug charachteristics. Intravenous=inhalation>intramuscular>subQ>oral
how is equivalency of drug products for the oral route determined?
FDA requires that formulations are bioequivalent. Rate and extent of active ingredient must be similar (within set limits) to brand name. if there is a 90% conficence interval in man AUC and mean Cmax is w/in 80-125% of brand name
what general factors affect drug absorption?
solubility in bilogic fluids (aq environment)=must be hydro and lipo phylic to cross membranes/distribute self; rate of solid dissoution; concentration of drug at site of administration; circulation at site of absorption; area of absorbing surface
what are the most important variable to consider about routes of drug administration?
bioavailability and rate of onset
what is the most common route for systemic effects?
oral. Slow onset of action, variable bioavailability
is absorption greater in the stomach or small intestine?
small intestine due to surface area
when is a drug favorable for absorption in GI tract?
when it is non-ionized and lipophilic
how does gastric emptying time effect absorption?
increased motility increases emptying speed and increases speed of absorption. Food delays gastric emptying and absorption
what do enteric coatings do?
prevent GI irritation, destruction from gastric secretions until they reach the basic intestines
slow release preparations do what?
slow rate of dissolution allowing for 8 hr or longer uniform absorption
what are the advantages to slow release preparations?
decreased frequency of administration, compliance, effect is maintained, eleiminates peaks/troughs
what are the disadvantages to slow release preparations?
greater independent variability, dose dumping, toxicity
describe rectal route charachteristics
not rapid onset, variable bioavailability (greater than oral route). Used when vomiting, unconscious, post GI surgery, GI irritation, uncooperative pt. 50% bypasses liver (less 1st pass than oral), faster onset of action. Useful for lipid soluble and potent drugs (
describe systemic IV route charachteristics
direct route, fastest (
describe systemic intramuscular route charachteristics
close to IV route (~90%), aqueous are absorbed quickly (5-10min), affected by blood flow/activity factors, depot/oil suspended vehicles are slower and sustained, can have erratic/incomplete absorption, can cause pain/necrosis/contamination
describe systemic subcutaneous route charachteristics
close to IV route (~100%), slower/constant rate of absorption, rate can be altered (eg: insulin), local anasthetics/contraceptives, non-irritating and volume limited
describe systemic inhalation route charachteristics
fast (
describe systemic transdermal route charachteristics
patch to skin allows for 1st pass avoidance, allows for prolonged drug levels, must be potent drug (
describe local inhalation route charachteristics
aerosol/microparticles molecules in suspension, increases local topical effects reducing systemic effects (asthma), depends on size (
describe local topical route charachteristics
treats local conditions, skin/mucosal membranes, minimal systemic absorption
how do tight junctions affect drug distribution?
tissues with tight junctions (GI mucosa, BBB, placenta, renal tubules) require drugs to pass through lipid membranes, and into/out of the blood. Drugs that can’t pass through membranes (large, protein bound, charged, hydrophilic) can’t move btw. compartments
GI mucosa absorption
oral-negligable absorption
bbb/placenta absorption
limited distribution
renal tubule absorption
filtration at glomerulus, then reduced reabsorption of drug into blood, excretion via urine
how does pH affect drug distribution?
affects lipid solubility. HH equation and tissue pH and pKa let you predict conditions favoring passage or preventing passage.
describe what dissociation of a proton does to a weak acid or base drug?
dissociation of proton from acid=ionized drug, dissociation of proton from base=unionized drug
what is the significance of ionization for drug absorption?
nonionized forms are more readily absorbed, ionized don’t cross lipid membrane
state the conditions that form nonionized acids and bases
acids become nonionized in acid medium, bases in alkaline medium
what does the HH equation allow you to predict with drugs?
the pH at which the majority of the drug will be non-ionized
what is the rule of thumb for qualitative preditions regarding drug absorption?
note whether the drug is weak acid/base, and pH of the fluid it’s in relative to pKa of the drug. pH is lower than pKA, more protons, protonated base/acid will dominate. When pH is higher than pKa of drug fewer protons, unprotonated form of weak acid/base, absorption favored.
state HH weak acid equation
HA H+ + A- [non-ionized in acidic solutions, ph
state HH weak base equation
BH+ H+ + B- [non-ionized in basic solutions, ph>pKa]
what is ion trapping?
lipid barriers separate 2 aqueous phases at different pHs, only non-ionized drugs can cross, equilibration is same on both sides of membrane, on each side of membrane separate equilibriums occur btw. Ionized and un-ionized forms of drug. At equilibrium, unionized conc. of drug is same, but total concentrationo of drug is greater on side where ionization is greater. drugs are trapped where they ionize
where are acidic drugs trapped
basic solutions
where are basic drugs trapped
acidic solutions
what is the clinical significance of ion trapping?
alters urine pH to trap weak acids/bases and hasten excretion, potential to concentrate basic drugs, weak bases are concentrated in acidic stomach contents
can ionized drug molecules cross capilary walls?
YES! Through pores.
can lipid solubility affect the rate of passage of non-ionized drugs?
YES! 2 drugs may have similar pKa, but will absorb differently
how does protein binding affect drug absorption?
will influence distribution if the free drug is diffusible. Acidic drugs bind albumin, basic to alpha-1-glycoprotein. Reduces concentration of active drug, hinders metabolic degredation/excretion, prolongs drug action, decreases volume o distribution, decreases ability to enter CNS across BBB
displacement drug interactions
displacement of a 1st drug from a protein binding site by a second drug, results in increased levels of unbound 1st drug. Total levels of drugs are unchanged b/c administration rate is the same
when does drug displacement have clinical consequences?
drug has narrow therapeutic index, started in high doses, Vd is small, response occurs more rapidly than distribution
why do most drugs act as if they display single compartment kinetics?
they are absorbed into/eliminated from a central compartment and are rapidly distributed. Some drugs may go from a second compartment (tissue) and are slower (2 compartment kinetics)
Vd
size of compartment necessary to account for total amount of drug in body if it were present throughout body at same concentration found in plasma. Gives indication of extent a drug passes from plasma to extravascular tissue. Partition coefficient btw plasma and body.
determination of Vd
single dose of drug is administered in IV (Ab), plasma concentration at time 0 (C0) is determined and Vd is calculated. Ab/C0
distribution compartments
distribution may not be homogenous. Sites are speculative w/o comparison
what are the volumes of body compartments?
plasma=3-5L, extracellular water=12-15L, total body water=42L, other compartments >50L
higher values of Vd indicate what?
drugs are located outside the plasma (lipid soluble, tissue binding)
lower values of Vd indicate what?
drugs located inside plasma or ECF (bind plasma proteins, large size, low lipid solubility)
what are the values of Vd?
L/kg
Vd allows you to determine what of a dose?
effect a dose will have on Cp. Dilution factor. Cp=D/Vd
necessary dose calculation
LD=Cp desired x Vd. The loading dose (LD) to fill the distribution volume to achieve steady state plasma
what is biotransformation/drug metabolism?
enzyme catalyzed chemical structure transformation
what are the general principles/charachteristics of drug metabolism?
enzymes have endogenous substrates and play role in metabolism, liver is the primary organ of metabolism (others can do it too), oxidation is the most frequent path (some by enzyme bound of smooth ER, some by cytosol enzymes), lipid soluble compounds are converted to water soluble then excreted
biotransformation phase 1
inserts/unmasks fundtional group into drug rendering it water soluble and able to be conjugated. Oxidation, reduction, hydrolysis
biotransformations phase 2
conjugation. Combine with pre-existing/metabolically inserted functional groups, forms polar conjugate excreted in urine.
what type of process is drug metabolism?
detoxifying. Forms readily excreted metabolytes (inactive). Drug metabolized to less active compound
what are other metabolic outcomes?
can become more active drug, inactive to active, toxic metabolyte
what are cytochrome p450-dependent oxidations?
part of oxidation system that includes NADPH, NADPH-cytochrome P450 reductase, molecular O2. Liver is richest source
list the charachteristics of CYP450
substrate must be lipid soluble, inducible, inhibitable, postnatal development variable, different isozymes
inducibility
increase in enzyme protein and drug metabolizing activity
postnatal development variable
early neonatal levels exist at 50-75% of adult, some are metabolized faster
isozymes of CYP450
CYP1-3
how are pateints classified regarding enzyme activity?
ultra rapid metabolizers (UM) or decreased/poor metabolizers (PM). Depends on ethnic groups.
amplichip cyp450 test
analyzes blood derived DNA and detects genetic polymorphisms in activity of CYP2 D6 and C19. these account for 25% of drug metabolism
what does clinical effect depend on?
whether metabolism is detoxifying or activating and whether polymorphism results in increase or decreased enzyme activity
PM CYP2D6 detoxifying
increased antipsychotic drug toxicity
UM CYP2D6 detoxifying
nonresponse to antidepressants
PM CYP2C19 activating
decreased PPI efficacy for peptic ulcer disease
PM CYP2D6 activating
insufficient analgesia with codine-dont metabolize to morphine
UM CYP2D6 activating
codeine intoxication-too rapid metabolism
differences in warfarin response may be due to genetic variation in?
vitamin K reductase (what it targets), CYP2C9 (metabolizes)
cytochrome P450-independent oxidations
several types, dehydroginases, oxidases
amine oxidases
located in outer membrane of mitochondria (monoamine oxidase), important for neurotransmitter metabolism
dehydrogenations
alcohol dehydrogenase, hepatic soluble fraction, several types, reached at 5 yr, aldehyde dehydrogenase
list the reductions of Phase 1
azo reduction (activates slfonamides), nitro reduction (diff. enzymes; toxic intermediate), carbonyl reduction
list hydrolysis of phase 1
esterases, amidases
esterases
adult values in 1st few months (used for pro-drugs). Hydrolyze exters to alcohol and acid, very reactive. Present plasma, liver, tissue.
amidases
liver and gut, amides to acids and amines
conjugations of phase 2
synthetic transformations. Drug or drug metabolite is condjugated to endogenous biochemical unit by a coenzyme. Done by transferases. Product is often water soluble and excreted.
glucuronidation
create conjugates that are excreted in urine, some in bile, some in feces. Groups that conjugate: aliphatic OH, aromatic OH, COOH, NH, SH. When doen by beta glucuronidase results in free drug
gucuronyl transferases
microsomal enzymes in liver, kidney, GI. Inducible.