• 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

1/167

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;

167 Cards in this Set

  • Front
  • Back

first synthetic drug

aspirin/acetyl salicylic acid (ASA)

first antibiotic

sulfanilomide

sulfanilomide side effect

kidney failure

thalidomide

- sedative used to alleviate morning sickness


- caused children to be born with no limbs, affected heart and congenital eye problems



phase 1 clinical study

tested on limited subjects

phase 2 clinical study

tested on small group out of target population

phase 3 clinical study

tested on larger population

phase 4 clinical study

assess risk benefits & confirm efficacy

3 types of toxicities

teratogenity


mutagenicity


reproductive toxicity

drug quality critera (5)

- purity


- stability


- sterility


- limits of potentially toxic impurities


- defined amount released at specific rate

orphan drugs

for a condition that affects less than 200,000 people (in the US)

Tubocurarine


- What does it do


- What is the effect on the body


- When is it used

- nAChR antagonist


- causes muscle paralysis


- surgery

pharmacology

study of drugs - what they are, how they work and what they do

2 branches of pharmacology

pharmacokinetics & pharmacodynamics

pharmacodynamics

- what the drug does to the body - effects elicited (desirable & undesirable)


- study of relationship between drug concentration & effects

pharmacokinetics

how the body absorbs & eliminates drug

receptors

- protein molecule found on cell surface or in the cell that receives chemical signals upon ligand binding

4 classes of receptors

- ligand gated ion channels


- enzyme linked receptors


- GPCR


- ligand activated transcription factors

ligand gated ion channels

reside on cell surface and act as gates to control ion passage into cell

enzyme linked receptor

reside on cell surface & cause intracellular enzymatic activity upon extracellular ligand binding

GPCR

reside on cells surface and couple with G proteins that do have enzyme activity to generate intracellular 2nd messengers

ligand activated transcription factors

receptor resides in the cell & stimulates transcription factors to promote DNA transcription to make mRNA & proteins

agonist

chemical that binds to a receptor and activates the receptor to get a cellular response

antagonist

chemical that binds to a receptor and blocks it

inverse agonist

agent that binds to a receptor and induces a pharmacological response opposite to that of an agonist

what are the variables in the equation


B= (Bmax x [D])/[D] + Kd

B - fraction of receptors bound


Bmax - maximal fraction of total receptors bound


D - drug concentration


Kd - equilibrium dissociation constant & drug concentration where 1/2 of Bmax is achieved

Emax

- maximum response achieved by agonist


- drug efficacy

ED50

- drug dose where you get 50% of Emax


- drug potency

what are the independent and dependent variables for quantal dose response curves?

x - dosage


y - % of individuals responding

what is potency and how is it expressed?

- how much drug you need to get an effect of a given magnitude


- expressed as ED50

what does a high ED50 mean?

drug is less potent

what is efficacy & how is it expressed?

- maximum effect that can be achieved with a drug


- expressed as Emax

what effect does a partial agonist have on efficacy?

it reduces efficacy and you need a higher dose to get your response

constitutive receptor activation

spontaneous conversion from inactive to active state

what 2 things does agonist binding to the receptor (related to receptor activation)

- conformational change from inactive to active


- stabilizes active state

what 2 things will spontaneous agonist dissociation do the receptor?

- conformational change from active to inactive


- stabilize inactive state

chemical antagonism

direct interaction of 2 drugs such that the effect of 1 or both drugs is lost

example of chemical antagonism

- protamine (acidic anti-coagulant) & heparin (basic coagulant)

physiological antagonism

indirect interaction of 2 drugs with opposing physiological effects

example of physiological antagonism

histamine + H1R --> vasodilation


epinephrine + beta adrenergic receptor --> vasoconstriction


* or any other 2 drugs with opposite effects

2 groups of antagonists

competitive & non-competitive

what do competitive antagonists do? how do you overcome the effect? what characteristic of the drug will they affect & how do you overcome it?

- they bind reversibly to the receptor


- increase agonist concentration to displace the antagonist


- affect drug potency so you need to increase your dose

what do non-competitive antagonists do? what characteristic of the drug will they affect?

- bind irreversibly to the receptor at a different site than that for the agonist


- affect drug efficacy

what is drug desensitization

diminished response because of continuous exposure to the drug

receptor mediated desensitization - 2 forms

- loss of receptor function because of a change in receptor conformation


- reduction of receptor number usually due to feedback effects of the agonist (ex: phosphorylation & internalization)

non-receptor mediated desensitization - 3 forms

- reduction of receptor coupled signalling components (ex: 2nd messenger depletion)


- increased metabolic degradation


- physiological adaptation

3 types of adverse drug effects

- dose dependent


- toxic reaction


- allergic reactions

how do dose dependent side effects work?

get worse side effects with increased dose

why do you get toxic reactions?

because of too much drug activity (also dose dependent)

cyclosporine - function, adverse effects & side effects

- immunosuppressant (ex: organ transplants)


- side effects: kidney damage, higher risk for infections


- allergic effect: rash, hives, itching, breathing difficulties

what determines drug toxicity?

the dose

what is therapeutic index/window?

a measure of drug safety that considers the range of therapeutic agent amounts associated with adverse effects, therapeutic effects and no effect in a population

what is the equation for therapeutic index?

Toxic ED50/beneficial ED50

what is a drug with a narrow therapeutic window?

warfarin

what does warfarin do

prevents blood from clotting

what is a drug with a large therapeutic window?

aspirin

what are the 4 outcomes of patient response to drugs with respect to benefit & toxicity?

- benefit + toxicity


- no benefit + no toxicity


- benefit + no toxicity


- no benefit + toxicity

what are the 3 components of drug administration?

absorption, distribution, elimination

what is drug absorption?

movement from the point of administration

what is drug distribution?

movement of the drug from the blood to tissues in the body & other sites

what are 2 routes for drug administration?

- systemic & local

what route of administration has a systemic effect via the digestive tract?

enteral

what route of administration would an injection have? what type of effect does it have?

- parenteral


- systemic


what is first pass effect? why does it happen?

- a phenomenon of drug metabolism where the concentration of a drug is greatly reduced before it reaches the systemic circulation


- happens because the drug is absorbed into the small intestine & liver where some of it gets digested

what is bioavailability?

the amount of drug that is available for activity in the target tissue

this route of administration bypasses the first pass effect (100% bioavailability)

parenteral

what route of administration has a local affect?

topical

what route will an inhalational drug take?

topical

what 5 physiochemical factors affect oral drug absorption?

- concentration difference across cell membrane


- size


- polarity


- ionization state


- environmental pH

when an oral drug is absorbed it can go to 1 of 2 places, what are the 2 places?

- it can be eliminated in the feces


- it can go to the systemic circulation

what 3 physiological factors affect drug absorption? hint: they differ among people

- GI motility


- metabolism


- changes in GI pH

what is volume of distribution (Vd)?

theoretical volume that the administered drug amount would have to occupy (if uniformly distributed), to provide the same concentration as it currently is in blood plasma

what does a high Vd infer?

retention in volumes outside of plasma

what does a low Vd infer?

retention in plasma

lipophilic drugs are poorly excreted by what 2 areas & why?

- kidney & liver


- they bind to plasma proteins (that inhibit glomerular filtration) & then are reabsorbed to go into lipid rich tissues

________________ increases polarity, ionization & water solubility of drugs

metabolism

what are prodrugs?

a drug that is administered in an inactive form and gets converted to its active form through metabolism

what are 2 sites for drug metabolism?

liver & intestine

what is the rate limiting step of metabolism

phase 1 where reactive groups are either added or unmasked

what family of genes contribute to phase 1 metabolism? what is the most common form?

- CYP gene superfamily


- CYP P450 3A4

what does CYP 3A4 do to felodipine? how can you reverse this with food?

- inhibits it (first pass metabolism)


- co-administration with grapefruit juice

what is felodipine? what is it used for?

- Ca channel antagonist


- lowering blood pressure (relaxes smooth muscle)

what prodrug does CYP 3A4 metabolize to give fexofenadine?

terfenadine

what happens if terfenadine isn't metabolized & accumulates in plasma?

inhibition of many K channels & potential cardiac toxicity

how do cyclosporine & rifampicin work?

cyclosporine is an immunosuppressant used to prevent organ rejection & rifampicin is an antibiotic that induces expression of CYP 3A4 so that some of the cyclosporine is inactivated

5 interindividual differences in drug metabolism:

- diet


- environment


- age


- disease


- genetic factors

what are the 4 phenotypic groups for drug metabolism & which is the most common?

- poor metabolizers


- intermediate metabolizers


- extensive metabolizers (most common; 50-90%)


- ultrarapid metabolizers

what 2 organs contribute to drug excretion

liver & kidney

drugs to be excreted must be (3 things):

- lipophilic


- low polarity


- low ionization

how does first order rate of reaction affect rate of drug elimination?

the rate of elimination decreases as the amount of drug decreases

what is clearance? what is the equation?

- volume of plasma the drug is removed from per unit time


- CL = rate of elimination/drug concentration

the point where rate of administration = rate of elimination is known as

steady state

what 2 ways can you achieve steady state?

- multiple dosing


- continuous IV infusion

3 types of signalling

- endocrine


- paracrine


- autocrine

endocrine signalling

hormone is secreted into blood and travels to distant target cells

paracrine signalling

signalling happens close to site of secretion

autocrine signalling

cell releases a ligand where the receptor is on the same cell

the release of NE into the bloodstream by the adrenal medulla is an example of _________ signalling

endocrine

induced fit model

interaction between receptor and ligand causes a conformational change in the receptor to enhance its affinity for the drug

what type of receptor is involved in fast synaptic transmission such as that seen in the CNS

ligand gated receptors

what are the 3 superfamilies of GPCRs?

- A - rhodopsin-like


- B - glucagon/VIP/calcitonin


- C - metabotropic glu & chemosensor

there are 3 components of GPCR signalling - receptor, g protein & effector. what is the GTPase and why?

the receptor is the GTPase because it binds and hydrolyzes GTP when activated

which g protein inhibits the production of cAMP?

Gi

Gs ______ cAMP levels

increases

the ____ g protein increases DAG & IP3 2nd messenger levels


Gq

the mAChR is coupled to what g protein? what 2nd messenger gets released & why?

- Gq


- Ca gets released because of IP3 going to the endoplasmic reticulum where Ca is stored

what is the physiological effect of Ca release?

muscle contraction (cardiac and/or smooth)

what is the effector enzyme for Gs?

adenyl cyclase

what 2 effects can cAMP have?

- activation of PKA


- cAMP response element binding protein (CREB) activation & changes in gene transcription

example of homologous desensitization: phosphorylation of ________ causes desensitization of GPCRs by ____________

- beta arrestins


- binding to the GPCR and promoting internalization (either degradation or recycling)

activation of a GPCR causing down-regulation of another GPCR is an example of _______________ _____________ (2 words)

heterologous desensitization

what does bordetella pertussis do to g proteins & cAMP levels?

inactivates the Gi protein and you get increased cAMP levels

these 2 toxins cause the Gs protein to stay activated

pasteurella multocida & vibrio cholera


what does ligand binding do to


- tyrosine kinase receptors with intrinsic catalytic activity?


- tyrosine phosphatase receptors with instrinsic catalytic activity?

- receptor dimerization, autophosphorylation & signal propagation


- dephosphorylation, dimer separates & signal stops

this 2nd messenger causes relaxation of smooth muscle by reducing Ca levels:

cGMP

receptor serine/threonine kinases work through associated proteins (ex: cytokinase) and are what kind of receptor?

tyrosine kinase linked receptor

what receptors do steroids work on? how?

- intracellular receptors


- by moving through the bilayer and binding, an associated chaperone is lost from the receptor and the complex goes to the nucleus to initiate transcription

angiotensin converting enzyme is used for the treatment of ________

hypertension

what do ACE inhibitors do? what does this do to blood pressure?

- prevent ACE from turning angiotensin I to angiotensin II


- lowers blood pressure

what does renin do?

converts angiotensinogen from the liver to angiotensin I

how does viagra increase cGMP? (2 ways)

- increases NO that binds to guanylate cyclase which makes cGMP from GTP


- inhibits phosphodiesterase in the penis


where does the afferent NS go?

to the CNS

where does the efferent NS go?

away from the CNS

what are the 2 branches of the efferent NS?

somatic & autonomic

the _______ nervous system is voluntary

somatic

the _________ nervous system deals with skeletal muscle & motor neurons

somatic

which efferent NS pathway synapses onto a ganglion before going to target tissue?

autonomic

3 types of muscle cells

- cardiac


- skeletal


- smooth

when the sympathetic NS is activated, there is increased blood flow to __________ & decreased blood flow from ______________.

- increased to skeletal muscles


- decreased from gut

constricted pupils, relaxed muscles & a low heart rate are characteristic of the ________________ nervous system being activated

parasympathetic

this autonomic nervous system innervates cardiac, exocrine and smooth muscle & has long preganglionic axons near the target tissue

parasympathetic

sympathetic NS axons are shorter and terminate on ganglia in the spinal cord. the postganglionic neurons go to what 4 places & what NT is released? what type of receptors get activated?

- sweat glands - ACh (nicotinic receptor)


- exocrine glands - NE (alpha & beta)


- kidney - NE (alpha 1 & beta)


- adrenal medulla - NE & adrenaline to the blood (alpha & beta)

the somatic NS releases ACh where? what kind of receptor gets activated?

- neuromuscular junction


- nicotinic AChR

what is the NT released at preganglionic sympathetic nerves? what receptor is activated?

- ACh


- nicotinic

what does the SNS do to the pupils?

causes dilation

what is the enzyme that catalyzes ACh synthesis? ACh breakdown?

- cholineacetyltransferase


- acetylcholinesterase

if the M3 AChR couples to Gq, what happens to Ca levels?

increase

what are 3 mAChR agonists?

- ACh


- muscarine


- pilocarpine


what are the side effects of stimulating a mAChR?

- GI disturbances (nausea/vomiting)


- CNS effects


- bradycardia


- salivation


- bronchoconstriction

what are side effects of nAChR overstimulation? all of these side effects apply only to the _____ nervous system

- tremor


- convulsions


- coma


- central

what type of drug gives increased ACh similar to exogenous cholinergic agonists? what are the 2 types?

- cholinesterase inhibitors


- reversible and irreversible

irreversible cholinesterase inhibitors are indirect acting agents on nicotinic ACh receptors. how do they work?

- malathion & sarin


- muscarinic side effects


- muscle twitches, paralysis, then death

what are 2 ways to inhibit irreversible AChE inhibitors?

- use muscarinic antagonist like atropine to block the receptor. It acts as a competitive antagonist


- use pralidoxime to break the covalent bond

what would atropine do in the PNS? in the SNS?

- PNS - block input & cause increase in heart rate, pupil constriction


- SNS - pupil dilation

what are 2 other uses of muscarinic antagonists?

- alleviate morning sickness (scopolamine)


- reduce bladder activity


side effects of muscarinic antagonists affect only the ________________ nervous system. what are some of those side effects?

- parasympathetic


- blurred vision, sedation, dilated pupils, dry mouth (decreased salivation), less GI motility (constipation)

at what 2 places do nicotinic antagonists work?

- receptors at autonomic ganglia


- receptors at NMJ

what are the 2 types of NMJ antagonists? how do they work?

- depolarizing non-competitive antagonists - form tight bond with receptor & activate it continuously until there is desensitization & muscle paralysis. these cannot be overcome with more ACh


- non-depolarizing competitive antagonists - can be overcome with more ACh or by inhibiting its breakdown

what type of antagonist is succinylcholine? tubocurare?

- succinylcholine is a non-competitive depolarizing nAChR antagonist


- tubocurare is a competitive non-depolarizing nAChR antagonist

sympathetic nerves release catecholamines - what 3 NTs are in that group?

NE, epinephrine & DA

NT feedbacks and acts at the presynaptic receptor to either inhibit or facilitate more release. what happens at alpha 2 adrenergic receptors? beta 2 adrenergic receptors?

- alpha 2 - inhibition of NT release


- beta 2 - more NT release

in what 3 ways do adrenergic agonists cause signalling?

- direct binding to postsynaptic receptor - NE release & signalling


- indirectly by preventing reuptake


- indirectly by promoting release of NE

what is an example of an alpha 1 agonist? what is it used for? what is a side effect?

- phenylephrine


- nasal congestion - constricts blood vessels, pupil dilation


- can cause blood pressure increase

what is an example of an alpha 2 agonist? what is it used for? how do these drugs work?

- clonidine, used for hypertension


- bind to alpha 2 autoreceptor on presynaptic nerves to prevent NE release (negative feedback)

what g protein is associated with alpha 2 agonists?

Gi

what is an example of a beta 1 agonist? what is it used for?

- dobutamine


- heart failure

how do beta 1 agonists work? what is a side effect?

- increase Ca influx & increase muscle contraction


- arrhythmias

how/where do beta 2 agonists work? what is an example?

- increase cAMP in the bronchial tissue which inhibits myosin light chain kinase from phosphorylating myosin for muscle contraction


- salbutamol

how does salbutamol work? what is it used for & what are the side effects?

- the decreased muscle contraction relaxes smooth muscle


- prevent bronchospasm in asthma & relax uterine muscles in premature labor


- high levels can cause non-specific activation of beta 1 cardiac receptors & increase heart rate

what is an example of a direct adrenergic agonist? what is it used for and what are side effects of its action?

- adrenaline


- anaphylaxis including bronchospasm, mucous membrane constriction & hypotension


- arrhythmias

what are 2 mechanisms for indirect adrenergic agonists? what is an example?

- inhibition of reuptake


- NE release


- cocaine

how does cocaine work? what are side effects?

- prevents reuptake of NE & DA in the synapse


- CNS stimulant, increases HR, BP & force of heart contractions, arrhythmias