• 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

1/173

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;

173 Cards in this Set

  • Front
  • Back
what is pharmacokinetics?
term used to describe the study of the changes in concentration of a drug during the processes of absorption, distribution, metalosim, or biotransformation, and elimination from body
what system delivers drug to various tissues?
vascular system
once in the blood the drug can go where?
-either remain w/in vascular system and body water
-bound to proteins
-cross membranes to enter tissues
where does the unbound drug go?
enters organs, muscles, fat, and site of activity (receptors)
the transfer of drug to various sites depends on what?
-intrinsic properties of the agent
-molecular size
-degree of ionization
-lipid solubility
-protein binding
what does uptake of drug depend on?
-amt of blood flow to the tissue and the concentration gradient of the drug across membranes
the smaller the molecular size of an agent, the ______it crosses lipid barriers?
better
molecules with molecular weights greater than ___-___do not cross cell membrane?
100-200
transport across a membrane can occur how?
actively or passively
______transport does not require energy and involves transfer of a drug from ana area of high concentration to an area of lower concentration?
passive
______transport mechanisms are generally faster and require energy?
active transport
T or F most drugs are salts of either weak acids or weak bases.
true
the noncharged ionized/nonionized form is lipophilic?
nonionized
the charged ionized/nonionized form is water soluble?
ionized
because the nonionzied molecules are lipid solube they can diffuse across cell membranes such as ?
-blood-brain
-gastric
-placental
ionized molecules are are usually unable to penetrate lipid cell membranes because why?
of their low lipid solubility
the higher the degree of ionization the more/less access the drug has across tissues such as the GI tract, blood-brain, and placental barriers and liver hepatocytes?
less
how are ionized drugs excreted?
via the renal system
whether acidic or basic the degree of ionization of an agent at particular site is determined by what two factors?
1-dissociation constant (pKa)
2-ph gradient across teh membrane
what is pKa?
the negative log of the equilibrium constant for the dissociation of the acid or base
acids are proton____-?
donors
bases are proton ______--?
acceptors
weak base
pH>pKa=?
un-ionized form predominates
weak base
ph=pKa?
un-ionized equal to ionized
weak base
pH < pKa=?
ionized form predominates
weak acid
pH>pKa?
ionized form predominates
weak acid pH=pKa=?
Unionized equal to ionized
weak acid
pH<pKa?
unionized form predominates
Ion trapping has several anesthesia related applications what are they?
-influences on oral absorption of drugs
-maternal-fetal transfer
-central nervous system toxicity of LA
the pH of the fetus is more acidic or basic than that of the mother?
acidic
T or F lidocaine crosses the placenta easily
true
changes in what have been theorized to influence a drug's clincial effect?
protein binding
what is teh most abundant plasma protein?
albumin
what usually binds to albumin?
acidic compounds
what proteins favor binding to basic drugs?
AAG and B-globulin
what bind cyclosporine?
lipoproteins
what binds corticosteroids?
transcortin
high protein binding prevetns the drug from leaving where, to enter into tissue, which results in high ______concentrations?
blood, plasma
the degree of protein binding for a drug is proportional to what?
it's lipid solubility
the bond btwn drug and protein is strong/weak?
weak
b/c the bond btwn drug and protein is weak they can dissociate when?
1-when the plasma conc. of teh drug declines
2-or a second drugs that binds the same protein is introduced
displaced free drug becomes available for what?
biotransformation and elimination
protein binding is expressed in what terms?
in percentage of total drug bound
warfarin is approx ____% bound to plasma proteins
98
when hypoalbuminemia exists decreased albumin result in what?
the availability of a greater amount of free drug
a 5% increase in teh amt of unbound drug in the body produces at most a ___% increase in pharmacologically active unbound drug at the site of action
5%
when the amt of unbound drug in plasma increase what happens to the rate of elimination?
rate of elimination increases
after ___half lives teh unbound concentration returns to its previous steady state value?
four
when plasma proteins are lower than normal total drug concentrations are ________, but unbound concentrations are _______?
lowered, not effected
what determines how much of the drug is delivered to the systemic circulation?
the route of administration
when the entire amt amt of drug given is delivered the drug is said to what?
have 100% bioavailability
what are the routes of drug administration?
-enteral (involves GI tract)
-parenteral (subq, IM, IV, intrathecally, epidremally)
-pulmonary
-topical
what is the most common and convenient method for administration of a drug?
oral route
what are disadvantages of oral route?
-emotions, physical activity and food intake change GI environ.
drugs administered orally have lower or higher bioavailability?
lower
what is a significant factor in absorption of drug taken orally?
the length of time drug remains in stomach
what is pH of stomach?
1.5-2.5
drugs that are highly acidic like __________tend to remain nonionized and are highly absorbed?
barbiturates
what type of drugs that remain intact in the stomach and pass through are highly absorbed in small intesting?
basic drugs
what is alkaline environment of small intestine?
7-8
what is presystemic elimination?
the elimination of a drug by the GI system before the drug reaches teh systemic circulation
what are the 3 mechasims that cause presystemic elimination?
1-stomach acid hydrolyzes the drug
2-enzymes in GI wall deactivate the drug
3-liver biotransforms ingested drug before it reaches the effect site
the liver activity that biotransforms the drug before it reaches the effect site is called what effect?
first-pass hepatic effect
drugs absorbed from GI tract after original ingestion enter what?
the portal venous blood and pass through the liver first
in the liver what can occur to the drug before it has a chance to enter systemic circulation?
extensive hepatic extraction and metabolism
what is bioavailability of IV route of drug administration?
100%
what is bioavailability of IM route of administration?
75-100%
what is bioavailability of subq route?
75-100
what is bioavailability of oral route?
5-100
what is bioavailability of rectal route?
30-100%
what is bioavailability of inhalation route?
5-100%
what is bioavailabilty of sublingual route?
60-100%
what is bioavailability of intrathecal route?
low (intentionally)
what is bioavailablity of topical route?
80-100%
what route is the most rapid and allows for titration of doses?
IV
what route allows for less first pass effect than oral?
rectal
what route has a lack of first-pass effect and is absorbed directly into systemic cirucation?
sublingual
what are some examples of drugs that undergo substantial first-pass elimination?
-ASA
-glyceryl trinitrate
-isosorbide dinitrate
-levodopa
-lidocaine
-metoprolol
-morphine
-propranolol
-verapamil
-salbutamol
P-glycoprotein transporter is part of a larger family of effluc transporters found where?
intestine
liver
renal proximal tubular cells
capillary endothelial cells
comprising the BBB
what two routes of administration of drugs bypass the presystemic, portal system first-pass effect and are delivered rapidly to the superior vena cava for transport ?
sublingual and buccal
what route of administration is teh ideal route for prevention from emesis caused by irritation of GI mucosa by the drug?
rectal route
drugs placed in the _________rectum are abosorbed into the portal system via what vein?
proximal, suprior hemorrhoidal vein
agents placed in the distal rectum do or do not undergo presystemic elimination?
do NOT and therefore have more predictable circulatory levels
parentareral administration refers to administration by what?
injection
with both IM and subq inj. systemic absorption of drug is dependent on what?
capillar blood flow to the area
lipid solubility of agent
T or F the lungs provide a lg. surface area for drugs administered by inhalatio.
true
what route is used for administratoin of sustained release agent
transdermal
what characteristcs must a drug have to be administered via transdermal route?
-water and lipid soluble
-have MW < 1000
-dose requirements < 10mg in 24 hr
-pH of 5-9
-area it's applied must be relatively thin epidermis w/ sufficien blood supply
what is bioavailability?
the extent to which a drug reaches its effect site after its introduction into the circulatory system
what factors play a role in the bioavailability of agents?
-lipid solubility
-solubility in aqueous and organic solvents
-MW
-pH
-pKa
-blood flow
the environment into which a drug is introduced also has impact so what are factors of pt's environment that may affect bioavailability?
-age
-sex
-pathology
-pH
-blood flow
-temperature
what are compartment models used for?
prediction of serum concentrations and changes in drug concentrations in other tissues
a single compartment moel represents what through which homogenous distribution occurs?
entire body
a two-compartment model is used to simplify and explain what that can be extrapolated to more complex models?
pharmacokinetic
in the two-compartment model what are the names of the two compartments?
central compartment (vessel-rich grp)
peripheral compartment (vessel-poor group)
what composes teh vessel poor group?
muslce
fat
bone
(represents 90% of body mass)
the second compartment of two compartment model the peirpheral compartment receives how much of teh cardiac output?
25%
the central compartment receives what percent of the CO?
75%
drugs leave central compartment by what two phases?
-distribution into tissues
-metabolism and excretion
the drug enters what compartment for clearance from teh body?
central compartment
drugs cross lipid membranes by what?
-passive diffuional transfer
-carrier mediated transfer
absorption from the gut depends on what?
-GI motility
-GI pH
-particle size
-physiochemical interaction w/ gut contents
PH partition refers to the fact that weak acids tend to accumulate in compartments w/ relatively high or low pH?
high
the volume of distribution is a proportional expression that relates to what?
the amt of drug in the body to the serum concentration
(the apparent volume in which the drug is distributed after it has been intoduced into the system)
what is formula for volume of distribution?
Vd= dose of drug/plasma conc. of drug
a drug with large Vd has high or low conc. in the plasma?
low--very little drug would be available to the organs for elimination
what is Vd of drug affected by?
-physiochemical properties of drug
-lipid solubility
-plasma protein binding
-molecular size
the affinity of a drug for a specific macromolecular component of the cell and its intrisic activity are related to it's what?
chemical structure
relatively minor modification in the drug structure may result in major changes in what?
pharmacologic properties
what are stereoisomers?
molecules with the same chemical bonds but differnt configurations in their fixed spatial arrangements
what are enatiomers?
stereoisomers are non-superimposable mirror images of each other
what are 3 drug examples of select isomers w/ improved clinical properties?
1-cisatrcurium
2-levo-bupivacaine
3-ropivacaine
what does the y-axis on the plasma conc. curve represent?
plasma concentration
what does the x-axis on the plasma conc curve represent?
the time after the dose is injected
what is the first alpha phase of the plasma conc. curve?
distribution phase-represents initial dispersal of drug into the tissue compartments from the central compartment
what is the second phase of the plasma conc. curve?
a logarithmic plot of the slower eliminaton or beta phase of the curve
the plasma concn. curve is an example of what kind of curve?
biexponential decay curve
a steady state occurs when?
a stable plasma concentration of a drug is achieved
when is steady state typically reached?
with chronic administration ofa drug or
by continuous IV infusion
drug metabolism is synomous w/ what term?
drug biotransformation
what is metabolism?
enzyme-catalyzed change in chemical structure of agents and usually involves more than one pathway
what is the main organ for drug metabolism?
liver
the main organ for metabolism is liver but metabolism can also occur where?
-plasma
-lungs
-GI tract
-Kidneys
-heart
-brain
-skin
what is the goal of metabolism/
to change lipid-soluble agents into more water soluble forms so kidneys can eliminate them from the body
metabolism usually leads to transformation of active drugs to what?
inactive metabolites
water-solube drugs are mainly confined to what and do not enter the brain after acute dosing?
plasma
plasma half-life is directly proportional to what and inverserly proportional to what?
proportional to volume of distribution
inversely proportional to overall rate of clearance
what are some drugs that are cholineseterase catalyzed?
-succs
-cocaine
-procaine
-chloroprocaine
-tetracaine
-neostigmine
-pyridostigmine
-edrophonium
what are some nonesterase dependent drugs that undergo phase I hydrolysis?
remifenatnil
atracurium
cisatracurium
esmolol
aspirin
what does zero order kinetics mean?
even at therapeutic levels they exceed teh body's ability to excrete or metabolize them
what are phase I reactions that occur during drug metabolism?
oxidation
reduction
hydrolysis
what are phase II reactions?
involve conjugate reactions in which drug metabolite is conjugated w/ endogenous substance such as glucuronic, sulfonic or acetic acid
what are oxidative reactions?
oxygen is introduced into the molecule or the oxidative state of a molecule is changed so that its relative oxygen content is increased
oxidative metabolim reactions are catalyzed by enzymes of what system?
cytochrome P-450 system
reduction involves gain or loss of electrons?
gain of electrons
what is hydrolysis?
addition of water to an ester or amide to break the bond and form two smaller molecules
what is the end result of phase I reactions?
a more polar compound that is easily excreted by the kidney
why are phase II reactions also referred to as synthetic reactions?
the body synthesizes a new compound by donating a functional grp usually derived from an endogenous acid
what are some of the intracellular sites that exist for drug metabolizing enzymes?
-endoplasmic reticulum
-mitochondria
-cytosol
-lysosomes
-plasma membrane
the cytochrome P-450 is also called what system because it includes both oxidation and reduction steps and has low substrate specificity?
mixed-function oxidase
where are some extrahepatic sites of P-450 system?
-kidneys
-lungs
-skin
-intestial mucosa
what is elimination half time?
the time necessary for the plasma content of the drug to drop to half of its prevailing concentration after a rapid bolus injectin
a drug is regarded as being fully emliminated when approx ___% has been eliminated from teh body?
95%
___half lives is considered sufficient time for the drug's action to be terminated and the agent eliminated from the body?
4 half lives
what is context sensitive half time?
the time to halving of the blood conc. after termination of drug administration by an infusion designed to maintain a constant conc.
how is clearance defined?
the volume of plasma completely cleared of drug by metabolism and excretion per unit of time
clearance is directly proportional to ?
dose
clearance is inversely related to ?
the agent's half life as well as its concentration in the central compartment
what are two main organs for clearance?
kidney and liver
formula for clearance is clearance=?
Q (blood flow) x E (extration ratio)
what are high clearance drugs?
drugs that have high conc ratio of 0.7 or greater rely heavily on perfusion to liver to be cleared
what are 3 examples of high clearance drugs?
verapamil
morphine
lidocaine
what are three drugs that have low hepatic extraction?
thiopental
diazepam
theophylline
the kidneys excrete water soluble or lipid solule molecules with great ease?
water-soluble
the excretion of drugs from kidneys involves what 3 processess?
1-passive glomerular filtration
2-active tubular secretion
3-some reabsorption
the amt of drug made available to renal tubule for elimination depends on what?
the amt of free, unbound drug and the glomerular filtration rate
lipid soluble agents depend on what to excrete them?
liver-(kidney does not excrete lipid soluble agents as efficiently as water soluble compounds)
what is an example of a lipid soluble drug that is almost completely reabsorbed?
thiopental
the pH of _____ can affect the elminiation of drugs via the kidney?
urine
_________(type of lab) is an indicator of renal function?
creatnine clearance
why is elimination of drugs via the kidney's altered in neonates?
because of poor renal function the first year of life
T or F gender differences account for some variability in the pharmaokinetics of many agents.
true
female pt's have __-___% greater sensitivity to muscle relaxat effects of vec, pan, and roc?
20-30%
who is more sensitive to propofol male or females?
males
who is more likely to experience recall under GA, males or females?
females
males require ____-___% more narcotics for pain mngt vs. females?
30-40%
temperature affects tissue metabolism and what?
blood flow
what is the most important pharmacokinetic parameter?
clearance
what is definition of pharmacogenetics?
the study of variations in human genes that are responsible for different responses to drug therapy
what is pharmacogenomics?
evolution of pharmacogentic reserach involves the identification of drug response markers at the level of disease, drug metabolism, or drug target
pharmacotherapy may be affected by 3 types of genetic variation, what are they?
1-variations in target proteins
2-enzyme metabolism
3-idiosyncratic effects
what are polymorphisms?
variations in the DNA sequences that occur in at least 1% of the population
most variations in the human genome occur in what type of enzyme genes?
drug metabolizing enzyme genes