Shared Flashcard Set

Details

Intro
ex 3 - pharm
55
Medical
Professional
09/26/2011

Additional Medical Flashcards

 


 

Cards

Term
what is the most important limited factor in the movement of drugs in the body?
Definition
the lipid:water partition coefficient
Term
what happens to the solubilty of an amount of HA (a weak acid with pKa: 4.4) at pH 1.4?  7.4?
Definition
at 1.4, the majority will be in the non-ionized form (HA) and will therefore be more lipid soluble.  At 7.4, H+ and A- will be preferred and will be less soluble
Term
what is ion-trapping?
Definition

isolating a drug into a certain environment where its ionization status is favored.  this will prevent drugs from going back into cell compartments where they are no longer needed

 

fetal plasma is slightly more acidic than a mothers, therefore, ion trapping can happen across the placenta

Term
how is glucose usually transferred into muscles cells?
Definition
usually by facilitated diffusion which does not require energy, it simply goes down it conc gradient.  glucose is brought into a muscle cell by insulin-sensitive transported protein GLUT4
Term
SLC superfamily of transporters
Definition
includes organic anion transport proteins (OATP).  transports ionic and non-ionic and xenobiotics into cells.  relies on facilitated transport or secondary active sym or anti-porters
Term
ABC superfamily of transporters
Definition

includes p-glycoprotein and transports  xenobiotics out of cell against their gradients to keep cells safe.  relies on ATP hydrolysis to do this.

Term
difference between enterally and parentally administered drugs?
Definition
enterally is via alimentary system (mouth and rectal), parentally is injection
Term
why is absorption in small intestine always more successful than stomach absorption?
Definition
because there is 200 m2 of surface area in the duodenum and even it its present in the ionized form, it's still many time more effective than the stomach
Term
problems with rectal adminstration of drugs?
Definition
50% of drug will bypass liver, and absorption is irregular and incomplete, and it can irritate the rectal mucosa
Term
why choose parental adminstration of a drug?
Definition

patient is combatitive, you absolutely need the drug delivered in active for and cannot have the body perform that task for you., you need to deliver a large volume

 

IV administration delivers 100% bioavailability, but can cause infections and pain.  intramuscular and subQ can be delivered almost anywhere, but transfer of drug is limited to capillary permeability vs. the size of the drug

Term
why do drugs take long to reach interstitial fluid in the brain?
Definition

allthought capillaries in most endothelial tissue are highly permeable, the blood brain barrier is formed by tight juctions between capillary cells thus restricting the passage of substances to the CNS.

 

sinusoidal capillaries of the liver and spleen have larger openings that allow passage of red blood cells adn seum proteins

Term
what is the order of distribution of a drug starting from the blood?
Definition

it first quickly enteres vessel rich groups of organs (first phase), then muscles, skin, and fat (second phase).

 

a final redistribution of the drugs is possible following initial distribution, but this usually dignifies the begining of termination of effect (i.e., anesthesia, where maximum effect on brain has been reached, the effect was short lived, and not the drug is being redirected to start clearing it from the system)

Term
drug binding to inert plasma proteins
Definition

only free unbound drugs and produce effects, but some being plasma proteins specifically, but reversibly.  this takes these drugs out of bioavailability.

 

these extent of bound and unbound drugs is usually help pretty constant (known as steady state.)  is bound plasma-protein drug complexes began being eliminated from the system, circulating plasma-protein drug complexes will start dissociating to maintain the steady state.

 

albumin mainly sequesters acidic drugs

a-1 acid glycoprotein mainly sequesters basic drugs

Term
tissue reservoirs for drugs?
Definition

bones often can obsorb heavy metals and tetracycline and incorperate them into their crystal lattice.

 

fat hoards lipid soluble drugs

Term
biotransformation
Definition

many enzymes exist that transform drugs into metabolites.  these metabolites may be the active form of the drug, may make a hydrophobic molecule hydrophilic so that it can be excreted, and have been transformed into a percursor before becoming another metabolite

 

the liver is the primary organ for biotransformation bc:

-has large blood flow

-high conc of enzymes

-high conc of transporters including SLC type and ABC type and MRP2 type

Term
some major phase 1 metabolizing enzymes?  phase 2?
Definition

phase 1:  CYPS, FMOs, EHs

 

phase 2:  UGT (UDP-gluconyltransferase)

 

all are located in ER of the cell (often hepatocyte) and this is good because the ER is a distinct lipid-bilayer in the cell so when lipid soluble molecules enter the cytosol through the PM, they become embedded in the ER to be acted on by phase 1 enzymes.  once a phase one enzyme acts, another molecule like a phase 2  (UGT)  can modify the metabolite

Term
what do you call the phase 1 modification of a molecule to an active metabolite?
Definition
usually phase 1 metabolism of a molecule leads to inactivation of a molecule through hydrolysis of an ester or amide linkage.  a molecule becomes a PRODRUG if the metabolism results in the bioactivation of the drug.
Term
main purpose of CYP family?
Definition
Phase 1 microsomal drug oxidations.  can do this because they carry a heme group
Term
main purpose of UGT family
Definition

a transferase (like all phase 2s) which adds a functional group to a metabolite (usually leading to hydrophilicity and are excrete in urine and feces)

 

urudine diphsophate-glucosyntransferases (UGT) is the most common of the phase  2 enzymes and has highest concentration in GI tract and liver

 

addes glucuronic acid leading to excretion.  normal flora in ileum can cleave off this acid and cause the substrate metabolite to go back into circulation.

 

can lead to drug-drug interactions bc UGTs are inducible and can be inhibited

Term
what do GSTs and SULTs do?
Definition

phase two enzymes

 

SULT makes a metabolite a reactive electrophile

 

GST addes a glutathione to reactive electrophiles therefore protecting it from reaction

 

Glutathione (usually stored as GSH) can cause oxidative damage to a cell if its conc falls too low.  Falling levels of GSH has been associated with cancer, AIDS, spsis, trauma, and burns 

Term
first pass metabolism?
Definition

a certain percentage of absorbed drugs taken orally will end up in the liver during the first pass, become metabolized, and not be biologically available.  This will happen a few more times until all the agents have been eliminated.

 

IV drug administration ensures 100% bioavailability

Term
explain one method of hastening the treatment of drug-poisoning.
Definition

acidify or make alkaline a patient's urine.  The more drug you can ionize, the fast it will clear.

 

ie. alkalation with sodium bicarbonate to rid of weak acids

acidification with ammonium chloride to rid of weak bases

Term
explain renal excretion of drugs
Definition

only drugs that are not bound to plasma proteins can be filtered with water by the glomerulus.  High plasma protein binding means low filtration and excretion of the drug.  as more unbound drugs are  passively diffused into the proximal tubule for elimination, more bound drugs unbind to maintain the steady state.

 

nonionized drugs are capabably of being reabsorbed if reabsorption of Na ions is causing a gradient.  The nonionized drugs will be carried back to circulation in the water.

Term
how to genetics explain some people's intolerance to certain drugs?
Definition

their phenotype is such that they have pharmacogenetic differences in the expression of xenobiotic metabolizing enzymes.  the difference has to appear in >1% of the population

 

often this is the case of SNPs

Term
properties of a good drug?
Definition
can reach target, exert effect, be metabolized and excreted
Term
factors affecting drug's abilty to exert desired effect?
Definition
structural features, solubilities, chirality, charge, size, try of bond it can form (weak? irreverslble?)
Term
what affects extend of drug action?
Definition

-a drug interacting with receptors that serve functions common to most cells - widespread effects

 

-a drug working on receptors that are only unique to a few cell types

 

-a drug interacting with a receptor that serve a vital function may be difficult to dangerous to use

Term
what can lead to supersensitivity of a receptor?
Definition

constant and chronic administration of a receptor antagoist

 

the denervation of a preganglionic fiber that indices postganglionic neuronal supersensitivity

Term

What is:

EC50

Emax

Kd

Bmax

Definition

EC50=conc at which 50% of the maximal response is seens

Emax=the dose at which no futher increase in response can be seen

Kd=conc of free drug at which 50% of all binding sites are filled 

Bmax=the total conc of binding sites

Term
intrinsic affinity
Definition
that ability of a drug bound to its receptor to activate downstream effector mechanism
Term
what is efficacy
Definition
a quantitative measure of the effects of a drug at 
Term
what is potenency?
Definition
a measure of drug concentration needed to ellicit an effect.  A more potent drug will require a lower conc of itself to elicit the same effect as another drug
Term
a physiological antagonist
Definition

an agonist and an antagonist acting at two independent sites and inducing independent, but opposite, effects

 

i.e. insulin lowers blood glucose by stimulating cells to take up glucose, glucoagon increases blood glucose by promoting hepatic glycogenolysis and gluconeogenesis

Term
define the therapeutic indexes
Definition
the LD50/ED50.  drugs are often defined by the width of their therpeutic window.  in a narrow window, sligh changes in systemic conc can lead to signifcan changes in pharmacodynamic response
Term
what is the formula for drug clearance?
Definition

CL= rate of elimination

       drug conc in fluid measured

Term
how do you calculate the volume distribution, Vd?
Definition

Vd= dose/Cp  <--conc in plasma.

 

therefore, if the concentration in plasma is very low, we make an apparent assumption that most of the dose is taken up into the extravascular tissues

 

operates under an assumption of first order kinetics where any drug that has been eliminated from the plasma per unit time depended on the conc of drug remaining in the plasma

Term
what is the half-life formula
Definition
half life = (0.693 x Vd)/CL
Term
what is the calculation of dosing rate?
Definition

rate of drug elimination must be equal to the rate of drug adminstration

 

dosing rate = CL x Css

 

by the 4-5th half life, steady state will be achieved

Term
what first effects bioavailability?
Definition

bioavailability is abbreviated by F (the fraction reaching systemic circulation). 

 

it is effected by inital dose and the amount of first pass hepatic metabolism

 

the efficiency of first pass hepatic clearance is determined by its intrinsic value, CL, and the flow rate of the blood to it, Q

 

if the extraction rate of the liver is .33, then the bioavailability is .67 right off the bat

 

increased blood flow will slow down the liver intrinsic metabolism

Term
when wanting to continue a patient on a medicine, what must one do prior to discharge
Definition

calculate the absolute and variable bioactivites of the drugs they were taking.

 

the formula is set up like is

 

F = 100 x AUCa x doseb

               AUCb x dose a

 

a and b can be brand vs. generic, or IV vs. oral

 

looking for bioequivalence

Term

explain saturable elimination

 

what does it assimilate graphically

Definition

it seems to follow michaelis-menten kinetics (ie. Km is equal to 50% of receptor binding, a definate Vmax exists)

 

it is risky to increase a dose in a saturation index is near.  even increasing it a small bit could lead to a large jump in plasma conc.  if dosing rate exceeds elimination capacity, toxicity can ensue

 

these are ZERO-ORDER kinetics and include ethanol, aspirin, and phenytoin

Term
recommendations for dosage increase
Definition

stay low and go slow

 

increase no more than 50%, and don't do it until 4-5 halflives

Term
what is a maintenance dose
Definition

an adjusted formula which allows for the state-state dose to factor in the rate of loss

 

dosing rate = CL x Css

                   F

 

(F doesn't matter for IV infusions)

Term
considerations to make if drug absorption seems to be lower than expected
Definition

-patient's compliance might be low

-patient could have organ impairments

-plasma protein binding could be fluxuating

     -if it (Cp) goes up, Vd does down and vice versa

-patient may be obese

-skeletal muscle mass differences

Term
what is an idiosynchratic drug response?
Definition

unpredictable, manifested as ultra sensitivity to a low dose, or extreme insensitivity to high doses

 

can result from genetic polymorphisms

Term
what are cumulative effects?
Definition
results of chronic exposures to therpeutic conc of drugs as well as low conc or other natural and synthetic material in environment
Term
some causes of hepatotoxicity
Definition
alcoholism, acetaminophen tox, viral infections, steatosis (fatty liver)
Term
some causes of nephrotoxicity
Definition
NSAIDS (sometimes in combination with acetaminophen), ACE inhibitors, cyclosporine
Term
what is an additive effect?
Definition
the compounding of two drugs.  each performs their single dose effect, but you will ultimately have two doses
Term
what is a synergistic/potentiation effect?
Definition
one drug magnifies the negative quality of another.  the resultant effect exceeds any of the individual drug's possible effects
Term
actions patients take that alter the absorption of drugs
Definition

alteration of gastic pH what they swallow their pill with.  ginger ale is acidic, milk is basic, this could have good or bad consequences

 

sequestration of drug tetracycline binds to antacids, so if they're taken at the same time, you're going to get poor absorption

 

alteration of gastic motility slowing it down slows down reabsorption

 

inhibition or induction of Pgp transporters  they are meant to keep out absorption 

Term

Strong inducers of CYP3A4 and Pgp: 

Definition
 Carbamazepine (antiepileptic drug)  Phenobarbital (antiepileptic drug)  Phenytoin (antiepileptic drug)  Rifampin (antibiotic)  St. John’s wort (herbal remedy) 
Term

Induction of CYP1A can?

Definition

 potentially lead to carcinogenicity - cigarette smoke and other  hydocarbons are inducers of this family

 

 

 

 

 

 

 

 

Term

Moderate-to-strong CYP3A4 inhibitors 

Definition

Cimetidine (reduces stomach acidity) Erythromycin and Clarithromycin (macrolide antibiotics) Ketoconazole and Itraconazole (antifungals) Ritonavir (protease inhibitor; one of the most potent CYP3A4 inhibitors) Atazanavir (protease inhibitor); also inhibits UGT1A1 Grapefruit juice 

Term
urinary acidifiers will have what effect on the clearance of drugs?
Definition
they will decrease the clearence of acid drugs and increase the clearance of basic drugs (this is because of ion trapping)
Supporting users have an ad free experience!