Pharmacology Flashcards
what is potency? how is it measured?
the amount of drug necessary to produce an effect of a given magnitude
measured by EC50 (decreased EC50 = more potent)
what is efficacy? how is it measured?
ability of a drug to elicit a response when it interacts with a receptor
measured by Emax
assumes all receptors are occupied by drug and no increase in response will be observed if more drugs added –> this is not true for spare receptors!!
describe a competitive antagonist. what effect does it have on log-dose response curve?
reversibly binds orthosteric site (same site as agonist), can be overcome by increasing [agonist] to outcompete, often has complimentary shape to receptor agonists
R shift on log-dose response curve (increase EC50, no change in efficacy)
what type of inhibitor is represented by the purple curve?
non-competitive inhibitor
what type of inhibitor is represented by the green curve?
competitive inhibitor
describe a noncompetitive antagonist. what effect does it have on log-dose response curve?
inhibition that cannot be overcome with increased [agonist]. can either be 1) antagonist irreversibly binds to agonist binding site (orthosteric site) OR 2) antagonist binds to another site (allosteric site) and prevents receptor activation.
Log-dose response curve: no change in EC50 (potency), decreased efficacy (Emax)
describe an uncompetitive antagonist. what effect does it have on log-dose response curve?
an antagonist that doesn’t antagonize unless receptor is activated (ie need formation of agonist-receptor complex)
Log-dose response curve: decrease efficacy (Emax) and EC50 (ie increase potency)
where do all agonists bind?
orthosteric site
when does EC50 = Kd?
for binding, especially for antagonists
(Kd= equilibrium dissociation constant)
what are the two important properties of a partial agonist?
- does not yield max biological response
- can act as competitive inhibitor of a full agonist
describe the concept of spare receptors
when responses are due to complex amplification system, low doses of antagonist will produce R shift on log-dose response curve. This is because even if only a small proportion of receptors bound to full agonist, will still be able to reach max response even though most receptors aren’t bound (ie unoccupied or spare receptors)
when does EC50 NOT EQUAL Kd?
when spare receptors
in a biological system, how can you distinguish from competitive inhibitor versus other antagonist given spare receptors?
if increase [noncompetitive antagonist], will eventually see a decrease in Emax response to full agonist
under what two conditions are constitutive receptor activity observed?
- tumors
- experimental artificial expression systems
what is ED50?
median effective dose, dose at which half of individuals get desired drug effect
what is LD50?
median lethal dose that produces death for 50% of experimental animals in the study
how to calculate therapeutic index
TI = toxic ED50/beneficial ED50
what do inverse agonists do?
reverse the constitutive activity of receptors and exert the opposite pharmacological effect of receptor agonists
what is warfarin? what is it’s therapeutic index?
blocks VCORC1 to inhibit synthesis of clotting factors, used to prevent clots after surgery and to tx atrial fibrillation
TI is very low at ~1 –> this means can be dangerous. sweet spot is 2-3x increase in clotting time to prevent stroke and excessive bleeding
what is diazepam? what is its therapeutic index?
allosteric activator of GABAA receptors to increase Cl- current by increasing frequency of channel openings
used to produce sedation, sleep, tx anxiety and muscle spasms
high TI: ~100
side effects: oversedation, dizziness, confusion, memory loss
drugs ending in …azopam or …azolam are _________ and act by ________
benzodiazepenes
bind to a site other than the site that binds the inhibitory transmitter GABA (“allo” in allosteric=other). This benzo binding increases the frequency of opening of GABAA-gated Cl- channels and enhances synaptic inhibition
describe the features of the Na/K ATPase pump
makes inside of cell slightly negative
which direction do sodium and potassium leak channels flow?
Na+ leaks into cell –> depolarizes (more pos)
K+ leak leaves cell –> hyperpolarizes (more neg)
what is equilibrium potential?
membrane potential that is just sufficient to oppose the chemical potential for a specific ion
what graded potential(s) exist(s) in neurons?
EPSP (excitatory post synaptic potential)
IPSP (inhibitory post synaptic potential)
what graded potential(s) exist(s) at the neuromuscular junction?
end plate potential
what is the mechanism of EPSPs?
ionotropic glutamate receptors, conduct Na+ or Na+/Ca2+ (mostly monovalent cation channels)
depolarize membrane
what is the mechanism of IPSPs?
GABA receptors, conduct Cl-
in developing neuron, depolarize
in mature neuron, hyperpolarize
describe the developmental switch in GABAnergic neurons
in developing neuron: active Na-K-2Cl transporter so high intracellular Cl-. GABA receptor removes Cl- from cell, depolarizing membrane
in mature neuron: Na-K-2Cl transporter inactive but active K-Cl transporter which exports Cl-, so low intracellular Cl-. GABA receptor brings Cl- in to hyperpolarize the cell
what is the mechanism of the end plate potential (EPP)?
acetylcholine receptors, conducts Na+ or Na+/Ca2+, depolarizes membrane
what channels are responsible for absolute refractory period?
Na+ channel inactivation
what channels are responsible for the relative refractory period?
incomplete Na+ channel recovery and residual K+ opening
name two major differences between neuronal, muscle, and cardiac action potentials
speed: nerve/skeletal are fast, cardiac is slow
diff ion channels: cardiac also uses Ca2+
describe the components of an action potential
how can cooperativity occur for receptors? (3 ways)
- binding of one agonist molecule influences the binding of another (rarely happens for receptors)
- binding of each molecule is independent, but you need more than one molecule bound to activate
- the relationship between binding and response is cooperative (multiple/ranges of active states are possible)
describe the constrained subunits model for receptors (Monod-Wyman-Changeux)
there are only two states of receptors: active and inactive
agonists can only bind to the active conformation, and shift the equilibrium toward the active conformation
describe the sequential activation model for receptors (Koshland-Nemthy-Filmer)
receptors undergo sequential changes to a multitude of different possible conformations
different ligands induce specific/different conformational states
name an example of a receptor that follows the constrained subunit model
nicotinic Ach receptor
name two examples of receptors that follow the sequential changes/activation model
- ionotropic glutamate receptors
- GPCRs
what is primary active transport?
ATP-mediated transport
(ATPases)
what is secondary active transport?
coupled transport driven by potential energy stored in a concentration gradient
(move one ion down its conc gradient and the other against its conc gradient)
what are the two types of secondary active transporters?
symporters (co-transport)
antiporters (counter-transport)
what are some examples of voltage-gated channels?
the Na+, K+, Ca2+, and Cl- channels that mediate electrical signaling
what are some examples of ligand-gated channels that are activated by extracellular ligands?
- Ach-gated channels
- GABA-gated channels
- ATP gated-channels
- exogenous substance-gated channels (i.e. capsaicin)
what are some examples intracellular ligands that activate ligand-gated channels?
- IP3
- Ca2+
- cAMP/cGMP
what do leak channels do?
mediate passive movement of ions
what are leak channels for water called?
aquaporins
what are the two categories of ATPases and what are examples of each?
- V-type: proton pumps
- P-type: Na/K ATPases, Ca-ATPases, H/K ATPases
describe how a uniporter works
uniporters enable facilitated diffusion by moving a solute down a concentration gradient
how many subunits does an inward rectifier channel have?
4
it’s a tetramer!
what are the 2 types of subunits of KATP channels?
- channel-forming subunits
- regulatory subunits (sulfonylurea receptors)
describe why sulfonylureas can be used to treat diabetes
sulfonlyureas inactivate the KATP channel –> Vm is depolarized –> voltage-gated Ca2+ channel opens –> insulin is released from pancreatic B cells
(triggers insulin release without glucose stimulation)
does ATP activate or inhibit KATP channels?
inhibit! presence of ATP causes them to close
when ATP is present, will KATP channels in smooth muscle cause vasoconstriction or vasodilation?
vasoconstriction
ATP inactivates KATP channels –> Ca2+ channels open –> vaosconstriction
what does minoxidil treat and what’s its mech of action?
anti-hypertensive
activates KATP channels, causes membrane hyperpolarization and closure of Ca2+ channels
(therefore causes vasodilation!)
what are the two domains in a voltage-gated K+ channel?
- voltage sensing domain (in particular the S4 helix)
- pore domain (S5-S6 helices)
how does the S4 helix of the voltage gated K+ channel determine whether the channel is open or closed?
S4 is a partially charged voltage sensor–when the membrane is depolarized, the S4 portion is pushed in an outward direction, opening the channel
when the membrane is hyperpolarized, the S4 portion is pulled pack into the cell, which triggers a conformational change & closes pore
describe N-type and C-type inactivation of K+ voltage gated channels
N type inactivation: the NTD of one of the channel helices plugs the inner mouth of the pore shut (like a ball and chain)
C type inactivation: collapse of the channel
how many subunits does the twin pore channel have?
2
(it is a dimer!)
what is the mechanism of action for a local anesthetic like lidocaine?
blocks Na+ voltage-gated channels, preferenitally binds to inactivated state of the channel
describe use-dependent or frequency-dependent block
means that the degree of channel block is proportional to the rate of nerve stimulation (degree of block is enhanced during repetitive stimulation)
important for fast-firing neurons
what channel do neurotoxins like tetrodotoxin, saxitoxin, and brevotoxin block?
Na+ voltage gated channels
where are L-type voltage-gated Ca2+ channels active?
the heart
where are P/Q, R, and N-type Ca2+ voltage gated channels active?
the nervous system
where are T-type Ca2+ voltage gated channels active?
both the heart and the nervous system
what does it mean for a channel to be high voltage activated?
which Ca2+ voltage-gated channels are high voltage activated?
it means that the cell needs to be HIGHLY depolarized for the channels to be activated
include L, P/Q, N, and R-type channels
what does it mean for a channel to be low voltage activated?
which Ca2+ voltage-gated channels are low voltage activated?
it means that the channel just needs a little depolarization nudge to be activated (easily activated)
includes T-type channels
what are some diseases that Ca2+ channel blockers are used to treat?
hypertension, angina, arrhythmia
how to Na+ channel blockers affect heart rhythm?
Na+ channels block initial upstroke, therefore slowing conductance of AP through heart
how do Ca2+ channel blockers affect heart rhythm?
don’t affect rate of rise, but do affect plateau phase (make it easier to repolarize the heart, shortens AP)
how do K+ channel blockers affect heart rhythm?
K+ channel blockers impair repolarization, therefore lengthen AP
(need to find a balance with these between fixing arrhythmia and causing long QT syndrome)
what happens to heart rhythm when the hERG K+ channel is inactiaved?
there is a lenghtening of action potential due to longer repolarization time
why did people unexpectedly die after taking the antihistamine terfenadine?
terfenadine has an off-target effect of blocking hERG channels and causing arrhythmia (long QT)
describe epithelial cells
monolayer sheets, polarized cells: apical and basolateral aspects. often have brush border morphology @ apical side
imp for drugs in intestinal lining (absorption), renal tubules (elimination), intrahepatic biliary (elmination)
what role do TJs play in the epithelial barrier?
polarize the cell by separating membrane into two compartments (apical and basolateral), asymmetrical distribution of membrane proteins
what is the difference between the cellular pathway and the paracellular pathway in epithelial cells?
cellular- molecule goes through cell on both apical and basolateral sides using diff membrane proteins to travel between lumen and interstitium
paracelluluar- molecule goes between cells through TJs, solutes and water and dissolved drugs can do this
what is the difference between a tight and leaky epithelium?
leaky- bulk movement of fluid, low resistance to flow of charged ions so voltage doesn’t build up (ex. small intestine, proximal renal tubule)
tight- high resistance, resists flow of charged ions so voltage builds up, doesn’t let as much through (ex colon, renal collecting duct)
describe the three types of vascular endothelial cells. which are the most important for drug transport?
**continuous- snug connections, have TJs, can be tight or leaky, BBB
**fenestrated- holes in cell cytoplasm, more leaky, most capillaries including gut and kidney
sinusoidal- intercellular gaps and incomplete basement membrane, incomplete barriers, whole cells can get through, in spleen and bone marrow
drugs are designed to be ____ soluble
lipid
in which form do lipid soluble drugs diffuse across the cell membrane?
uncharged (they are weak acids and bases, but uncharged form is what crosses)
describe the concept of ion trapping across the gastric cell membrane
drugs can only get into lumen when uncharged, then rapidly convert back to charged form inside lumen so essentially “trapped”
what is the henderson hasselbach equation?
gross.
but also it’s log[HA]/[A-] = pKa - pH
describe ABC transporters
mediate active transport, use ATP, contain nucleotide binding domain (NBD) to bind ATP, most often for efflux, some can mediate anticancer drug resistance
what is p-glycoprotein?
an ABC transporter, drug exporter, can pump out chemotherapy drugs, imp in both cancer resistance and normal physiology –> eliminates drugs from brain, reduce GI absorption, excrete drugs in bile and urine
what is an SLC transporter?
mediate secondary active transport (don’t use ATP directly but use gradient formed via ATP active transport) and facilitated diffusion (via alternating access and conformational change), can have coupled co-transport (Na and glucose)
ex. neurotransmitters
what is the role of neurotransmitter transporters at the synaptic cleft?
after neurotransmitters activate receptor on post-synaptic membrane, get taken back up into presynaptic terminal via transport to shut down signaling
SLC6A2
norepinephrine transporter
clears norepi from synapse
target of cocaine, ADHD drugs
SLC6A3
dopamine transporter
clears dopamine from synapse
target of cocaine, amphetamines
SLC6A4
serotonin transporter
clears serotonin from synapse
target of antidepressants (SSRIs)
what is transcytosis?
vesicle is formed on one side of the membrane to capture substance to be transported, next vesicle and its contents are shuttled inside the cell to reach an opposing membrane. cells can use vesicles to transport substances across PM either into or out of cell
what is vectorial transport?
net, directional transfer of a solute across epithelial or endothelial cells, imp for efficient transfer of nutrients across epithelial or endothelial barriers, drug absorption and elimination
transcytosis may also be used here
what is drug absorption?
transfer of drug from its site of administration to systemic circulation
most drugs need this to happen to reach their target
name three factors that impact drug absorption
route of administration
bioavailability
bioequivalence
what is the main criteria for drug administration?
bioavailability
what are the fastest routes of drug administration? what are the slowest?
intravenous, intraosseous (in pediatrics), endotracheal
intramuscular, subcutaneous, oral (varies widely), transdermal
list advantages and disadvantages for oral drug administration (brief)
advantages: easy, expensive, safe, preferred by pts
disadvantages: slow time to effect, consciousness required, requires functional gut, bioavailability can be limited
what are advantages and disadvantages of rectal drug administration
advantages: easy, good absorption, no first pass metabolism, good for infants and children
disadvantages: not preferred by pts
what are advantages and disadvantages of intravenous drug administration?
advantages- rapid onset, dependable, 100% bioavailability!!!!!
disadvantages- requires IV cannula, expensive, labor intensive, pain, risk of infection, bleeding
what are advantages and disadvantages of IM/Subcutaneous drug administration?
advantages: relatively fast onset, no first pass metabolism
disadvantages: absorption can be unpredictable, pain, risk of bruising/bleeding
what are some advantages and disadvantages of transdermal drug administration?
advantages: easy, non-invasive, high pt satisfaction
disadvantage: slow time to effect, most drugs not absorbed through skin
what are some advantages and disadvantages of inhalation drug administration?
advantages: rapid absorption, limited systemic delivery
disadvantages: effectiveness depends on pt technique
name some factors to consider when choosing route of drug administration
clinical urgency, type and properties of drug including bioavailability, condition of pt including age and mental status, pt preference
define bioavailability
fraction of administered dose of unchanged drug that reaches systemic circulation
ie how well a drug is absorbed, usually with respect to oral administration
what is the bioavailability of IV drugs?
100%
what is bioequivalence
related to the manufacturing of the drug, same rates (pharmacokinetics) and extents of bioavailability of active ingredient
(TLDR; same amt active ingredient delivered in same amt time)
a generic drug must have 8-%-125% bioequivalence of brand-name drug without identical formulation
what is pharmaceutical equivalence?
contain same active ingredients, are identical in strength or concentration, dosage form, and route of administration
name some factors affecting oral absorption of drugs
intrinsic chemical and physical properties, bioavailability (high efficient absorption), gastric acidity and digestive enzymes, gastric emptying time, relationship to food intake, drug metab by intestinal epithelium (CYP3A4), drug efflux from intestinal epithelial cells (p-gp), inhibitors of CYP3A4 and drug transporters
why is ibuprofen largely absorbed from the intestine
surface area in intestine far outweighs the driver for absorption
what is first pass metabolism?
for oral drugs, metabolism in either intestinal wall or liver before the drug can reach systemic circulation reduces bioavailability
what receptor is largely responsible for 1st pass metabolism?
CYP3A
what are some drugs with low bioavailability? which are NOT administered orally?
morphine, demerol, lidocaine, nitroglycerin = not administered orally
propranolol- oral administration bc wide therapeutic window
what effect does grapefruit juice have on bioavailability? how does it achieve this?
furanocoumarins in grapefruit juice (and pomegranate) INHIBIT CYP3A4 and p-gp –> this increases bioavailability for some meds by decreasing 1st pass metabolism, can cause toxic elevation of plasma drug concentration
how do generic drugs differ from brand named drugs?
A generic drug must conform to 80 to 125% of bioequivalence of the brand-name drug, but it must not be identically formulated.
3 criteria: pharmaceutical equivalence, bioequivalence, effective and safe
what should be the same if looking at a time-concentration curve for generic and brand name drug in order for generic drug to be manufactured?
should have same Cmax peak at same time point and same AUC (area under the curve) which represents how much drug the body gets
what is drug distribution?
reversible transfer of drug between systemic circulation and extravascular (interstitial) fluids and tissues
what does perfusion-limited tissue distribution mean?
initial rate of drug distribution depends on blood flow to tissue
first phase: brain, heart, liver, kidney
second phase: muscle, most organs, skin and fat (have moderate blood flow)
what does permeability-limited tissue distribution mean?
certain compartments have restricted access, for ex blood brain barrier (BBB), placenta
how does drug accumulation in tissues impact volume of distribution (Vd)?
if more drug accumulates in tissues, higher Vd
what is the mechanism of termination of thiopental?
redistribution, when removed from plasma and redistributed from brain to other tissues, that’s why it’s so short lasting
describe the blood brain barrier (BBB)
no fenestrations, limited extracellular space due to other cells and basement membrane, TJs
lots of efflux transporters to send things out of the brain (MRPs and p-gp)
what can p-gp KO studies tell us?
if KO increases drug penetration in BBB, we know it’s normally pumped out of CNS by p-gp
ex. quinidine gets pumped out, ritonavir does not
how can we leverage the BBB to increase drug delivery? (two ways)
- inhibit p-gp
- utilize blood-CSF interfact to bypass BBB with interthecal lumbar puncture
what type of endothelial cells are the choroid plexus capillaries?
fenestrated
albumin binds ______
drugs that are weak acids
alpha-acid glycoprotein binds _______
drugs that are weak bases
how does protein binding impact drug activity?
bound drug is inactive, free drug is active and can equilibrate across diff fluid spaces.
protein binding lowers effective concentration and slows distribution to extravascular sites bc less is metabolized, increased half life
what is hypoalbuminemia?
disease affecting protein binding of drugs, affects free drug concentration without affecting total plasma concentration
name an example of a drug that can compete for protein binding to cause drug displacement
sodium valproate (displaces phenytoin from albumin and increases free phenytoin concentration)