• 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

1/124

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;

124 Cards in this Set

  • Front
  • Back
what are the ways in which drugs can be transported across membranes? (4) which of these require energy & which of these require a carrier?
passive diffusion
facilitated diffusion (carrier)
aqueous channels
active transport (energy & carrier)
what drives passive diffusion of a drug across a membrane? (2)
concentration gradient across membrane
lipid:water partition coefficient of the drug (greater cooefficient, high conc drug in membrane & faster diffusion)
what drives passive diffusion of ionic compound? why?
its pKa
pH gradient across the membrane

very small compounds can be carried in bulk water flow (molecular weight less than 200)


since they are not soluble in lipid
how are drugs passed across most capilliary endothelial membranes? with which exception & why
bulk flow through intercellular pores

tight junctions in CNS capillaries limit this intercellular diffusion
what defines facilitated diffusion?
carrier - mediated transport process that does not require energy (ie movement of substance is not against an electrochemical gradient)
when is facilitated diffusion useful?
when the rate of movement across the membrane by simple diffusion would otherwise be too slow
how does active transport differ from facilitated diffusion?
it is identical except that ATP powers the drug transport against a concentration gradient
what, generally speaking, is pharmacokinetics?
what the body does to the drug
what is the definition of drug absorption?
the rate at which a drug leaves site of administration & extent to which this occurs
what concept is more clinically useful than drug absorption?
bioavailability - proportion of administered dose reaching the systemic circulation
what does drug absorption ultimately depend on, regardless of site?
drug solubility
what are drug factors influencing absorption? (5)
ionization state
molecular weight
solubility (lipophilicity)
formulation (solution versus tablet)
concentration (increased conc absorbed more rapidly)
what patient factors influence drug absorption?
- local conditions at site of absorption
- pH local anaesthetic
- presence of food in gastro tract
- circulation to site of absorption

- surface area of absorbing surface
(determined by route of admin)
what features of a drug favour absorption in the GI tract? why?
non ionized & lipophilic

since most GI absorption occurs via passive process
why is the stomach a poor absorber of drugs?
thick mucous membrane
low surface area
increased electrical resistance
why does gastric emptying increase drug absorption?
intestine has a thin, low resistance membrane & a high surface area

so pushing drug from low absorbing stomach to high absorbing intestine increases drug absorption rate
how does enteric coating influence drug absorption in the GI tract? (2)
prevents drugs being destroyed by gastric contents

HOWEVER may also resist dissolution in the intestine -> low absorption
why is the sublingual route effective for GTN? (4) why would we think it wouldn't be so?
GTN - non ionic & high lipid soluble
- is very potent - need relatively few molecules for therapeutic effect
- venous drainage from mouth is to SVC, so limited 1st pass metabolism
- first pass metabolism would destroy GTN if swallowed as tab

the sublingual route has a low surface area which theoretically would decrease absorption
what are two negatives and two positives for using the rectal route of drug administration?
-ve:
- often irregular & incomplete absorption
- often irritates rectal mucosa

+ve
- ~50% absorption bypasses liver first pass
- useful when pt vomiting/unconscious
what kind of first pass metabolism are parenteral methods subject to?
lung
especially weak bases & those not ionized @ blood pH
how are SC & IM drugs absorbed?
simple diffusion along gradient
what factors limit SC/IM absorption (2)
SA of absorbing capillary membrane
solubility of substance in interstitial fluid
what are the positive features of IV route of admin (3)?
- circumvents absorption problems
- allows accuracy & immediacy not otherwise possible
- suitable for large volumes
what are the negative features of IV admin? (3)
can't go back
can't use for oily/insoluble drugs (WHY)
need a patent vein
what happens if we use an irritating drug via SC route?
severe pain, necrosis, slough
what features of SC route lead to sustained effect? how can this be prolonged?
constant & slow absorption

prolonged by adding vasoconstrictor or by using insoluble preparation
what human factor greatly increases absorption via IM route?
blood flow to area
how does presence of subcutaneous fat influence rate of absorption?
decreases, since poorly perfused
what characteristic of the lungs facilitates rapid absorption of volatile /atomized drugs?
large surface area
when is systemic absorption the goal of topical nasal therapy? when is it an unwanted side effect?
ADH therapy (desired)

Local anaesthesia (not desired)
which of the epidermis vs dermis acts as a barrier to topical absorption of drugs to skin? when is this clinically relevant
epidermis

dermis freely permeable to solutes

relevant when skin barrier has been disrupted (abraded, burned, denuded skin)
what is inunction?
suspending skin drugs in oily vehicle to increase absorption
how are eye drugs usually absorbed & when is this clinically important?
through the cornea
corneal infection/trauma results in rapid absorption
what is bioequivalence?
same active ingredients
identical in strength/concentration, dosage form & route of admin
same rate & extent of bioavailability of active ingredient
where would a drug with Vd = 3L be distributed? how about Vd = 16L? Vd >46L?
3L - plasma only (plasma vol 3L)
16L - extracellular H20 (3L plasma plus 10-13L interstitial fluid)
>46 likely sequestered in depot as body only contains 40-46L fluid
what kinds of organs receive most of a drug during the first few minutes after absorption?
heart, liver, kidney, brain & other well perfused organs
how long does delivery of a drug to muscle, most viscera & fat take after absorption?
several minutes to hours before steady state
what factors, as well as blood flow, determine rate of distribution? (4)
- highly permeable capillary endothelium means distro is rapid into interstitium, except in brain

- lipid insoluble drugs can't permeate membranes easily -> limited distro

- drug binding to plasma proteins eg albumin acid drugs limits access to cellular sites of action

- accumulation in tissues in higher conc than expected due to pH gradients, binding to intracellular contituents or partitioning into lipid
what lesson does thiopental teach us about distribution?
successive doses of thiopental accumulate in fat & can become thiopental reservoirs.

Thiopental, on first dose, has a rapid onset of anaesthetic but also of termination, since it doesn't actually bind to anything in the brain. Thiopental diffuses into other tissues eg muscle, so plasma conc falls.

Fat reservoirs can maintain plasma concentration and therefore brain concentration at/above anaesthetic threshhold -> can become long acting
how does the CNS pose problems to drug distribution?
tight junctions predominate & aqueous bulk flow is therefore severely restricted -> less drugs can get in

highly lipid soluble drugs can get into CNS

but strongly ionized agents are usually unable to enter CNS from circulation
what plasma proteins bind acidic drugs? basic drugs?
albumin - acidic

a1-acid glycoprotein - basic
what normal drug processing activities are prevented by plasma protein binding? (2)
- limits concentration of drug at tissues & locus of action (only unbound drug is in equilibrium across membranes)

- glomerular filtration (conc of free drug in plasma not immediately changed)
what renal drug processing function is not limited by binding to plasma protein & why?
renal tubular secretion/biotransformation since these processes lower free drug concentration, leading to dissociation of drug - protein complex
what kinds of molecules do drugs normally bind to in tissues & is tissue binding of drugs generally reversible/irreversible?
proteins/phospholipids/nucleoproteins

generally reversible
why is fat a stable drug reservoir?
relatively low blood flow
how does bone act as a drug reservoir?
tetracycline antibiotics & heavy metals can adsorb onto the bone crystal surface & be incorporated into the lattice, thus bone can become a reservoir for slow release of toxic agents for a long time after exposure has ceased
what is the major transcellular reservoir?
GI tract
what is redistribution of a drug?
when drug effect terminates because the drug has been redistributed from its site of action to other tissues/sites

esp when a highly lipid soluble drug acting on CNS/CVS administered rapidly IV/inhaled
how do drugs cross the placenta?
simple diffusion esp lipid soluble, non ionized drugs
when is the volume of distribution much larger than the animal itself?
when a drug binds preferentially to tissues at the expense of plasma
what size volume of distribution would an ionized molecule trapped in plasma have?
small
what is the usefullness of volume of distribution?
hypothetical measure of how well a drug is removed from the plasma and distributed to the tissues
how would the volume of distribution distinguish between drugs that are not highly bound to plasma but don't enter the cell vs those which enter the cell freely?
Vd in non cell entering would be volume of extracellular fluid (0.2L/kg) whereas those entering cells freely would be total body water volume (0.55L/kg)
how does the one compartment model limit our evaluation of the volume of distribution?
drugs will not be distributed to all the tissues evenly, this only looks at whether drug is in plasma or not
how does volume of distribution relate dose and concentration
a low volume of distribution means the concentration > dose ie H20 soluble, highly bound to plasma protein

a high vol of distribution means dose>concentration ie lipid soluble, highlhy bound to tissues
give example of 3 drugs with low volume distribution
warfarin
amoxycillin
gentamicin
give example of 4 drugs with high volume of distribution
diazepam
digoxin
amitryptiline
chloroquin
what is the formula for calculating volume of distribution?
Vd = dose (amount of drug in body)
____________

concentration in plasma
how does volume of distribution affect our calculation of drug loading doses?
if you know the desired drug concentration in plasma & the volume of distro (in L/kg) & the pts weight, you can change the equation around to:

dose = Vd x target concentration
how does Vd affect our calculation of top up doses if the drug is present but the concentration too low?
loading dose = (target conc - measured conc) x Vd
why is drug biotransformation necessary? (2)
because most active drugs are non polar and polar drugs are more easily excreted

also because duration of action would be very long if left polar vs changed into (usually less active) metabolite
what are the two main categories of drug biotransformation and what distinguishes them?
phase I = NONSYNTHETIC - drugs are oxidised/reduced to a more polar form

phase II = SYNTHETIC
polar group is conjugated to a drug

drugs undergoing phase II conjugation may have already undergone phase I transformation (& vice versa)
where does drug biotransformation mainly occur? where else may it occur (5)
liver

also GI, lungs, skin, kidneys, brain
give 4 examples of drugs which are more extensively metabolised in the intestine than the liver
clonazepam
chlorproazine
cyclospine
midazolam (50% intestinal metabolism)
what in the lower gut is capable of many biotransformation reactions?
microbes
give an example of a drug that can be metabolised by gastric acid
penicillin
give an example of a drug that can be metabolised by digestive enzymes
polypeptides eg insulin
give an example of a drug that can be metabolised by enzymes in intestinal wall
sympathomimetic catechoamines
where are biotransformations catalysed at the subcellular level (4)
ER, mitochondria, cytsol, lysossomes

occasionally nuc envelope/plasma membrane
what are microsomes and which kind are full of oxidative drug metabolism enzymes?
microsomes are vesicles formed from leftover endoplasmic reticulum

the ones formed from smooth ER contain the enzymes
?? how much detail do we need on the P450 system ??
...
what are the two microsomal enzymes that are key in microsomal biotransformation? which is more important? why?
NADPH-cytochrome P450 reductase

cytochrome P450 which is more important - it is more abundant in the liver, so P450 heme reduction is a rate limiting step in hepatic drug oxidation
why is P450 so named?
in its reduced (ferrous) form it binds CO to give a complex that maximally absorbs light at 450nm
what is required for microsomal drug oxidation? (4)
p450
p450 reductase
NADPH
molecular oxygen
what is the common structural feature of drugs metabolised in the p450 system?
high lipid solubility
how do P450s compare with many other enzymes in terms of speed?
sluggish & reactions are slow
?? HOW MUCH DETAIL DO WE NEED TO KNOW FOR PHASE I reactions??
???
what is the significance of CYP3A4?
is the P450 isoform responsible for metabolising >50% prescription drugs metabolised by the liver
?? how much detail about enzyme inducers & inhibitors?
...
what is required for increased P450 synthesis?
increased transcription/translation (mediated by cell receptors for the inducer) & increased synthesis of heme (prosthetic cofactor)
what is a cruciferous vegetable?
spinach/kale/tatsoi etc
induce CYP1A p450 genes
what is the common mechanism of inhibiting p450 enzymes?
binding or interact with the P450 heme iron -> inactivate enzyme/competitively inhibit binding etc
what distinguishes phase II reactions?
they are synthetic

vs nonsynthetic phase I reactions
are phase I or phase II reactions generally faster?
phase II
what are the general characteristics of conjugates formed by phase II reactions?
polar, readily excreted, often inactive
what are the most dominant phase II enzymes?
UGTs

uridine 5'-diphosphate - glucuronosyl transferases
?? need to know metabolism of paracetamol to hepatotoxic metabolites in detail?
katzung fig 4-5
what percentage of paracetamol metabolism is phase II vs phase I? why is this important
95% phase II

the remaining 5% phase I becomes important at very high doses as P450 pathway becomes increasingly important
what hepatic compound is necessary for safe phase I metabolism of paracetamol?
GSH (glutathione)

is depleted faster than it can be regenerated with time, leading to accumulation of reactive toxic metabolite
which enzymes do charcoal broiled foods & cruciferous vegetables induce?
CYP1A
what does grapefruit juice inhibit?
CYP3A metabolism of coadministered drug substrates
what affect does cigarette smoke have on enzyme induction?
increases in some drugs
which drugs are enzyme inducing? (5)
various sedative hypnotics
antipsychotics
anticonvulsants
rifampicin (anti TB)
insecticides
how do sedatives and certain antipsychotics affect warfarin levles?
routine use of these may lead to considerably higher doses of warfarin required & cessation of these may lead to reduced anticoagulant metabolism & bleeding
what is the inductive explanation for drug tolerance?
an inducer may enhance not only the metabolism of other drugs but its own metabolism
apart from liver, what other system disease can impair drug metabolism & how?
cardiac - by preventing blood flow to liver of very rapidly metabolised drugs

thyroid (esp hypo)

bacterial/viral infections -> inflamm mediators/cytokines & NO release
what is drug clearance?
the factor that predicts rate of elimination in relation to drug concentration
what is the formula for clearance?
C = rate of elimination/concentration
how is drug clearance additive?
drug elimination may involve processes occuring in kidney, lung, liver & other organs. total systemic clearance equals CL kidney + CLliver + CLother
what are the two main sites of drug elimination?
kidneys & liver
what represents renal clearance?
clearance of unchanged drug in urine
how are drugs cleared in liver? (3)
biotransformation to metabolites

excretion of unchanged drug into bile

both
how does rate of drug elimination compare to concentration?
directly proportional

clearance is constant over concentration range
what is first order elimination? how can it be calculated?
when rate of elimination = clearance x concentration

estimated by calculating the area under the curve of time/concentration profile after a dose?
how can clearance be related to area under the curve?
clearance = dose/AUC
how does first order elimination clearance relate to capacity-limited (or saturable/non linear/ Michaelis-Mentin elimination)?
first order elimination is not saturable & the rate of drug elimination is directly proportional to concentration

capacity limited elimination means that clearance will vary depending on the concentration of drug achieved & is saturable
which are the three drugs for which clearance has no real meaning & why?
aspirin, ethanol, phenytoin

concentration will keep rising as long as dosing continues (if dosing rate exceeds elimination capacity, steady state cannot be achieved)
what is the equation to relate rate of elimination in capacity limited elimination?
rate of elimination = Vmax x C
________

Km + C

where V max is max elimination capacity
Km is drug concentration where rate of elimination is 50% of Vmax
how does flow dependent elimination relate to capacity limited elimination?
flow dependent - some drugs cleared so readily by organ that most of drug is eliminated on first pass ie blood flow determines rate of elimination

capacity limited drug elimination has no relevance to blood flow & can be saturated by drug concentration
what is the formula for half life in a single compartment?
t1/2 = log2 x Vd / CL

nb log2 approx equal to 0.7
when does the "true" half life exceed that from the half life formula?
in multicompartment models (most drugs)
what is the accumulation factor? what does the accumulation factor equal for a drug given every half life?
an index of accumulation

accumulation is inversely proportional to the fraction of the dose lost in each dosing interval

ie AF = 1/1- fraction remaining

so for dose every half life

AF = 1/0.5 = 2
what are the 3 primary processes of pharmacokinetics?
absorption
distribution
elimination
what is volume of distribution?
the measure of apparent space in the body available to contain the drug
what links pharmacokinetics & pharmacodynamics?
concentration
what is steady state?
pseudo zero order elimination
elimination rate almost independent of concentration

average total amount of drug in the body does not change over multiple dosing cycles
what is first pass effect?
elimination of drug occuring after administration but before it enters systemic circulation
how do we calculate loading dose?
target conc (mg/L) x vol distribution (L/kg)
how do we calculate IV maintenance dose?
Infusion rate
= target steady state conc x clearance rate

divided by salt & molar correction factor
how do we calculate oral maintenance dose?
avg target steady state x clearance x dosage interval

divided by

bioavailability x salt corr x molar corr
when do ionizable forms of drugs cross membranes best?
when they are in their non ionized form
when would a weak acid be in its best form to cross the lipid membrane? a weak base?
weak acid - non ionized at more acid pH

weak base - non ionized at more alkaline pH
how can we use pH to trap drugs in the urine for excretion?
by adjusting urine pH to make sure drug is in ionized state & hence can't be reabsorbed from the tubule
give a clinical example of how we could possibly use urine to excrete a drug?
methamphetamine, a weak base, is freely filtered at the glomerulus but can be rapidly reabsorbed at the renal tubule

ammonium chloride acidifies the urine which converts a larger fraction to charged form, poorly absorbed & thus rapidly eliminated

however there is a risk of renal damage with forced diuresis & urinary pH manipulation