Pharmacology Flashcards

1
Q

what is potency? how is it measured?

A

the amount of drug necessary to produce an effect of a given magnitude

measured by EC50 (decreased EC50 = more potent)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

what is efficacy? how is it measured?

A

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!!

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

describe a competitive antagonist. what effect does it have on log-dose response curve?

A

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)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

what type of inhibitor is represented by the purple curve?

A

non-competitive inhibitor

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

what type of inhibitor is represented by the green curve?

A

competitive inhibitor

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

describe a noncompetitive antagonist. what effect does it have on log-dose response curve?

A

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)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

describe an uncompetitive antagonist. what effect does it have on log-dose response curve?

A

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)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

where do all agonists bind?

A

orthosteric site

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

when does EC50 = Kd?

A

for binding, especially for antagonists

(Kd= equilibrium dissociation constant)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

what are the two important properties of a partial agonist?

A
  1. does not yield max biological response
  2. can act as competitive inhibitor of a full agonist
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

describe the concept of spare receptors

A

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)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

when does EC50 NOT EQUAL Kd?

A

when spare receptors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

in a biological system, how can you distinguish from competitive inhibitor versus other antagonist given spare receptors?

A

if increase [noncompetitive antagonist], will eventually see a decrease in Emax response to full agonist

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

under what two conditions are constitutive receptor activity observed?

A
  1. tumors
  2. experimental artificial expression systems
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

what is ED50?

A

median effective dose, dose at which half of individuals get desired drug effect

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

what is LD50?

A

median lethal dose that produces death for 50% of experimental animals in the study

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

how to calculate therapeutic index

A

TI = toxic ED50/beneficial ED50

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

what do inverse agonists do?

A

reverse the constitutive activity of receptors and exert the opposite pharmacological effect of receptor agonists

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

what is warfarin? what is it’s therapeutic index?

A

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

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

what is diazepam? what is its therapeutic index?

A

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

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

drugs ending in …azopam or …azolam are _________ and act by ________

A

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

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

describe the features of the Na/K ATPase pump

A

makes inside of cell slightly negative

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

which direction do sodium and potassium leak channels flow?

A

Na+ leaks into cell –> depolarizes (more pos)

K+ leak leaves cell –> hyperpolarizes (more neg)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

what is equilibrium potential?

A

membrane potential that is just sufficient to oppose the chemical potential for a specific ion

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

what graded potential(s) exist(s) in neurons?

A

EPSP (excitatory post synaptic potential)

IPSP (inhibitory post synaptic potential)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

what graded potential(s) exist(s) at the neuromuscular junction?

A

end plate potential

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

what is the mechanism of EPSPs?

A

ionotropic glutamate receptors, conduct Na+ or Na+/Ca2+ (mostly monovalent cation channels)

depolarize membrane

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

what is the mechanism of IPSPs?

A

GABA receptors, conduct Cl-

in developing neuron, depolarize

in mature neuron, hyperpolarize

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

describe the developmental switch in GABAnergic neurons

A

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

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

what is the mechanism of the end plate potential (EPP)?

A

acetylcholine receptors, conducts Na+ or Na+/Ca2+, depolarizes membrane

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

what channels are responsible for absolute refractory period?

A

Na+ channel inactivation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

what channels are responsible for the relative refractory period?

A

incomplete Na+ channel recovery and residual K+ opening

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

name two major differences between neuronal, muscle, and cardiac action potentials

A

speed: nerve/skeletal are fast, cardiac is slow

diff ion channels: cardiac also uses Ca2+

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

describe the components of an action potential

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

how can cooperativity occur for receptors? (3 ways)

A
  1. binding of one agonist molecule influences the binding of another (rarely happens for receptors)
  2. binding of each molecule is independent, but you need more than one molecule bound to activate
  3. the relationship between binding and response is cooperative (multiple/ranges of active states are possible)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

describe the constrained subunits model for receptors (Monod-Wyman-Changeux)

A

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

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

describe the sequential activation model for receptors (Koshland-Nemthy-Filmer)

A

receptors undergo sequential changes to a multitude of different possible conformations

different ligands induce specific/different conformational states

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

name an example of a receptor that follows the constrained subunit model

A

nicotinic Ach receptor

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

name two examples of receptors that follow the sequential changes/activation model

A
  1. ionotropic glutamate receptors
  2. GPCRs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

what is primary active transport?

A

ATP-mediated transport

(ATPases)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

what is secondary active transport?

A

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)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

what are the two types of secondary active transporters?

A

symporters (co-transport)

antiporters (counter-transport)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

what are some examples of voltage-gated channels?

A

the Na+, K+, Ca2+, and Cl- channels that mediate electrical signaling

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

what are some examples of ligand-gated channels that are activated by extracellular ligands?

A
  1. Ach-gated channels
  2. GABA-gated channels
  3. ATP gated-channels
  4. exogenous substance-gated channels (i.e. capsaicin)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

what are some examples intracellular ligands that activate ligand-gated channels?

A
  1. IP3
  2. Ca2+
  3. cAMP/cGMP
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

what do leak channels do?

A

mediate passive movement of ions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

what are leak channels for water called?

A

aquaporins

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

what are the two categories of ATPases and what are examples of each?

A
  1. V-type: proton pumps
  2. P-type: Na/K ATPases, Ca-ATPases, H/K ATPases
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

describe how a uniporter works

A

uniporters enable facilitated diffusion by moving a solute down a concentration gradient

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

how many subunits does an inward rectifier channel have?

A

4

it’s a tetramer!

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

what are the 2 types of subunits of KATP channels?

A
  1. channel-forming subunits
  2. regulatory subunits (sulfonylurea receptors)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

describe why sulfonylureas can be used to treat diabetes

A

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)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

does ATP activate or inhibit KATP channels?

A

inhibit! presence of ATP causes them to close

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

when ATP is present, will KATP channels in smooth muscle cause vasoconstriction or vasodilation?

A

vasoconstriction

ATP inactivates KATP channels –> Ca2+ channels open –> vaosconstriction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

what does minoxidil treat and what’s its mech of action?

A

anti-hypertensive

activates KATP channels, causes membrane hyperpolarization and closure of Ca2+ channels

(therefore causes vasodilation!)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

what are the two domains in a voltage-gated K+ channel?

A
  1. voltage sensing domain (in particular the S4 helix)
  2. pore domain (S5-S6 helices)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

how does the S4 helix of the voltage gated K+ channel determine whether the channel is open or closed?

A

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

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

describe N-type and C-type inactivation of K+ voltage gated channels

A

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 well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

how many subunits does the twin pore channel have?

A

2

(it is a dimer!)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

what is the mechanism of action for a local anesthetic like lidocaine?

A

blocks Na+ voltage-gated channels, preferenitally binds to inactivated state of the channel

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

describe use-dependent or frequency-dependent block

A

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

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

what channel do neurotoxins like tetrodotoxin, saxitoxin, and brevotoxin block?

A

Na+ voltage gated channels

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

where are L-type voltage-gated Ca2+ channels active?

A

the heart

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

where are P/Q, R, and N-type Ca2+ voltage gated channels active?

A

the nervous system

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

where are T-type Ca2+ voltage gated channels active?

A

both the heart and the nervous system

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

what does it mean for a channel to be high voltage activated?

which Ca2+ voltage-gated channels are high voltage activated?

A

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

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

what does it mean for a channel to be low voltage activated?

which Ca2+ voltage-gated channels are low voltage activated?

A

it means that the channel just needs a little depolarization nudge to be activated (easily activated)

includes T-type channels

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

what are some diseases that Ca2+ channel blockers are used to treat?

A

hypertension, angina, arrhythmia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

how to Na+ channel blockers affect heart rhythm?

A

Na+ channels block initial upstroke, therefore slowing conductance of AP through heart

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

how do Ca2+ channel blockers affect heart rhythm?

A

don’t affect rate of rise, but do affect plateau phase (make it easier to repolarize the heart, shortens AP)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

how do K+ channel blockers affect heart rhythm?

A

K+ channel blockers impair repolarization, therefore lengthen AP

(need to find a balance with these between fixing arrhythmia and causing long QT syndrome)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

what happens to heart rhythm when the hERG K+ channel is inactiaved?

A

there is a lenghtening of action potential due to longer repolarization time

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

why did people unexpectedly die after taking the antihistamine terfenadine?

A

terfenadine has an off-target effect of blocking hERG channels and causing arrhythmia (long QT)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

describe epithelial cells

A

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)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

what role do TJs play in the epithelial barrier?

A

polarize the cell by separating membrane into two compartments (apical and basolateral), asymmetrical distribution of membrane proteins

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

what is the difference between the cellular pathway and the paracellular pathway in epithelial cells?

A

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

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

what is the difference between a tight and leaky epithelium?

A

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)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

describe the three types of vascular endothelial cells. which are the most important for drug transport?

A

**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

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

drugs are designed to be ____ soluble

A

lipid

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

in which form do lipid soluble drugs diffuse across the cell membrane?

A

uncharged (they are weak acids and bases, but uncharged form is what crosses)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

describe the concept of ion trapping across the gastric cell membrane

A

drugs can only get into lumen when uncharged, then rapidly convert back to charged form inside lumen so essentially “trapped”

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

what is the henderson hasselbach equation?

A

gross.

but also it’s log[HA]/[A-] = pKa - pH

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

describe ABC transporters

A

mediate active transport, use ATP, contain nucleotide binding domain (NBD) to bind ATP, most often for efflux, some can mediate anticancer drug resistance

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

what is p-glycoprotein?

A

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

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

what is an SLC transporter?

A

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

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

what is the role of neurotransmitter transporters at the synaptic cleft?

A

after neurotransmitters activate receptor on post-synaptic membrane, get taken back up into presynaptic terminal via transport to shut down signaling

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

SLC6A2

A

norepinephrine transporter

clears norepi from synapse

target of cocaine, ADHD drugs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

SLC6A3

A

dopamine transporter

clears dopamine from synapse

target of cocaine, amphetamines

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

SLC6A4

A

serotonin transporter

clears serotonin from synapse

target of antidepressants (SSRIs)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

what is transcytosis?

A

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

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

what is vectorial transport?

A

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

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

what is drug absorption?

A

transfer of drug from its site of administration to systemic circulation

most drugs need this to happen to reach their target

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
93
Q

name three factors that impact drug absorption

A

route of administration

bioavailability

bioequivalence

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

what is the main criteria for drug administration?

A

bioavailability

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

what are the fastest routes of drug administration? what are the slowest?

A

intravenous, intraosseous (in pediatrics), endotracheal

intramuscular, subcutaneous, oral (varies widely), transdermal

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
96
Q

list advantages and disadvantages for oral drug administration (brief)

A

advantages: easy, expensive, safe, preferred by pts
disadvantages: slow time to effect, consciousness required, requires functional gut, bioavailability can be limited

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

what are advantages and disadvantages of rectal drug administration

A

advantages: easy, good absorption, no first pass metabolism, good for infants and children
disadvantages: not preferred by pts

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

what are advantages and disadvantages of intravenous drug administration?

A

advantages- rapid onset, dependable, 100% bioavailability!!!!!

disadvantages- requires IV cannula, expensive, labor intensive, pain, risk of infection, bleeding

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

what are advantages and disadvantages of IM/Subcutaneous drug administration?

A

advantages: relatively fast onset, no first pass metabolism
disadvantages: absorption can be unpredictable, pain, risk of bruising/bleeding

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

what are some advantages and disadvantages of transdermal drug administration?

A

advantages: easy, non-invasive, high pt satisfaction
disadvantage: slow time to effect, most drugs not absorbed through skin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

what are some advantages and disadvantages of inhalation drug administration?

A

advantages: rapid absorption, limited systemic delivery
disadvantages: effectiveness depends on pt technique

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

name some factors to consider when choosing route of drug administration

A

clinical urgency, type and properties of drug including bioavailability, condition of pt including age and mental status, pt preference

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

define bioavailability

A

fraction of administered dose of unchanged drug that reaches systemic circulation

ie how well a drug is absorbed, usually with respect to oral administration

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

what is the bioavailability of IV drugs?

A

100%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

what is bioequivalence

A

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

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
106
Q

what is pharmaceutical equivalence?

A

contain same active ingredients, are identical in strength or concentration, dosage form, and route of administration

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

name some factors affecting oral absorption of drugs

A

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

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

why is ibuprofen largely absorbed from the intestine

A

surface area in intestine far outweighs the driver for absorption

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
109
Q

what is first pass metabolism?

A

for oral drugs, metabolism in either intestinal wall or liver before the drug can reach systemic circulation reduces bioavailability

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
110
Q

what receptor is largely responsible for 1st pass metabolism?

A

CYP3A

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

what are some drugs with low bioavailability? which are NOT administered orally?

A

morphine, demerol, lidocaine, nitroglycerin = not administered orally

propranolol- oral administration bc wide therapeutic window

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
112
Q

what effect does grapefruit juice have on bioavailability? how does it achieve this?

A

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 well did you know this?
1
Not at all
2
3
4
5
Perfectly
113
Q

how do generic drugs differ from brand named drugs?

A

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

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
114
Q

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?

A

should have same Cmax peak at same time point and same AUC (area under the curve) which represents how much drug the body gets

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

what is drug distribution?

A

reversible transfer of drug between systemic circulation and extravascular (interstitial) fluids and tissues

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

what does perfusion-limited tissue distribution mean?

A

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)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

what does permeability-limited tissue distribution mean?

A

certain compartments have restricted access, for ex blood brain barrier (BBB), placenta

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

how does drug accumulation in tissues impact volume of distribution (Vd)?

A

if more drug accumulates in tissues, higher Vd

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

what is the mechanism of termination of thiopental?

A

redistribution, when removed from plasma and redistributed from brain to other tissues, that’s why it’s so short lasting

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
120
Q

describe the blood brain barrier (BBB)

A

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)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
121
Q

what can p-gp KO studies tell us?

A

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 well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

how can we leverage the BBB to increase drug delivery? (two ways)

A
  1. inhibit p-gp
  2. utilize blood-CSF interfact to bypass BBB with interthecal lumbar puncture
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

what type of endothelial cells are the choroid plexus capillaries?

A

fenestrated

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

albumin binds ______

A

drugs that are weak acids

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

alpha-acid glycoprotein binds _______

A

drugs that are weak bases

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

how does protein binding impact drug activity?

A

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

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

what is hypoalbuminemia?

A

disease affecting protein binding of drugs, affects free drug concentration without affecting total plasma concentration

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

name an example of a drug that can compete for protein binding to cause drug displacement

A

sodium valproate (displaces phenytoin from albumin and increases free phenytoin concentration)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

List the basic signaling processes used by pharmacological receptors (from the fastest to the slowest)

A
  1. Ligand-gated channels (fastest)
  2. G-protein coupled receptors
  3. Transmembrane enzymes (RTKs and related molecules)
  4. Cytoplasmic and nuclear receptors (slowest)
130
Q

name the 4 excitatory receptors

A
  1. Ach-nicotinic receptors
  2. Serotonin 5HT3 receptors
  3. P2X ATP receptors
  4. Glutamate (AMPA/kainite receptors and NMDA receptors)
131
Q

name the 2 inhibitory receptors

A
  1. GABA-A receptors
  2. Glycine receptors
132
Q

name the 4 CYS-loop receptors

A
  1. Ach (nicotinic)
  2. 5-HT3 (serotonin)
  3. GABA-A
  4. glycine
133
Q

name the non-cys loop receptors (2)

A
  1. glutamate
  2. P2X ATP receptors
134
Q

what does Ondansetron (Zofran) do?

A

competitive inh at 5HT3 receptors- treat nausea and vomiting

135
Q

what channels have benzodiazepine binding sites?

A

GABA-A

136
Q

what effect does penicillin have on GABA-A channels?

what negative physiologic effect does it cause?

A

they are open channel blockers

causes seizures!

137
Q

which neurotransmitter works only on ligand-gated channels? (hint: all other NTs also work on GPCRs)

A

glycine!

138
Q

what is the mech of action of Strychnine?

A

it’s a competitve inhibitor at glycine receptors (prevents an inhibitory effect)

causes spasticity

139
Q

what is the mech of action of tetanus toxin?

A

blocks glycine release (so it won’t bind to receptors), causes spasticity

140
Q

what is hyperekplexia?

A

familial startle disease

alpha subunits of glycine receptors are mutated, channel can’t bind glycine and therefore won’t open…no inhibitory effect in spinal cord :(

141
Q

what are the types of channels in the glutamate channel family?

A

NMDA, AMPA, Kainate

142
Q

what to nicotinic Ach receptors do?

A
  1. Initiate action potential in skeletal muscle and ganglionic transmission in peripheral NS
  2. Perform excitatory func in CNS
143
Q

what does the 5-HT3 serotonin receptor do?

A

it’s an excitatory receptor in CNS

causes emesis and GI activity

144
Q

is GABA-A excitatory or inhibitory?

What kind of channel does is it a ligand for?

A

inhibitory in CNS

opens ligand-gated Cl- channel

145
Q

is glycine inhibitory or excitatory?

where does it act?

A

inhibitory, in spinal cord

146
Q

what is the normal role of the NMDA receptor?

what disease can it cause when it’s letting too much calcium into the cell?

A
  1. Important in learning & memory (w/ normal to high Ca2+ levels)
  2. can cause Alzheimer’s when too much Ca2+ in cell
147
Q

what does memantine treat? what’s it’s mech of action?

A

open channel blocker of NMDA receptors, tx for Alzheimer’s

148
Q

List the three main types of signaling used by G-protein coupled receptors (GPCRs) from the fastest onset to the slowest

A
  1. Milliseconds: direct membrane effects (i.e. activation/inhibition of ion channels) by B/Y subunits of G-protein
  2. Seconds: second messenger synthesis (or block of synthesis by inh alpha subunits)
  3. Minutes: phosphorylation via protein kinase A after cAMP activation or protein kinase C after DAG activation
149
Q

what is the scaffold protein for RTKs when the ligand is a growth factor?

A

growth receptor bound protein 2

150
Q

EGF (and other growth factors) trigger which downstream signaling pathway?

A

Ras/MAPK

hint: mitogen-activated protein kinase

mitogen = GF!

151
Q

what is the scaffold protein for RTKs when insulin is the ligand?

A

IRS (insulin receptor substrate)

152
Q

what signaling cascade does insulin RTK trigger?

A

PI3K/AKT/mTOR

(metabolic)

153
Q

what signaling cascade do cytokines trigger?

A

JAK/STAT

154
Q

what are the main differences between RTKs for insulin and growth factors vs RTKs for cytokines?

(2 differences)

A
  1. for cytokine receptors, the TM domain does not have kinase activity (JAK serves as the kinase)
  2. for cytokine receptors, there is not a separate scaffold protein (JAK acts as the scaffold protein that recruits STAT)
155
Q

how to glucocorticoids work?

A

steroid binds receptor –> inhibitory HSP90 dissociates from glucocorticoid receptor –> ligand-bound receptors dimerize and enter nucleus, where they bind to DNA and trigger ∆s in gene TC –> causes inhibition of inflammatory cells & synthesis/release of inflammatory mediators

156
Q

how are GPCRs desensitized via GRKs?

A

GPCRs are phosphorylated on the cytoplasmic tail of receptor by GRKs (G-prot receptor kinases)

phosphorylated site binds B-arrestin, arrests signaling, which causes desensitization and receptor internalization

157
Q

what happens when you give repetitive doses of a competitive antagonist?

A
  1. Continual use leads to upregulation of receptors
  2. Discontinuation can cause rebound effects (esp with GPCRs)
158
Q

do allosteric activators increase or decrease likelihood of receptor desensitization?

A

decrease

159
Q

what are biased ligands and how do they differ from classical (balanced) ligands?

A
  1. Classical balanced signaling ligands (like endogenous substances) elicit their effects globally…
  2. Biased ligands can direct their effects toward one mechanism (such as GPCR signaling) vs another (arrestins)
160
Q

Why do we need drug metabolizing enzymes?

A

to rid the body of xenobiotics (which are sometimes cometabolized with endogenous agents), to make drugs more polar so they can be made inactive and excreted

161
Q

What family of proteins carry out the majority of Phase I metabolizing reactions?

A

the CYPS (cytochrome p450 monooxygenases). (They carry out 80% of phase I rxns)

162
Q

Which CYP is the most prevalent in Phase I rxns?

A

CYP3A4- (carries out about 60% of Phase I rxns)

163
Q

What is the purpose of Phase I drug metabolism reactions?

A

To provide a reactive site on the molecule where Phase II rxns can made modifications

164
Q

What is the most common rxn scheme for drug metabolism?

A

Phase I oxidation by CYP3A4, Phase II glucuronidation

165
Q

What is Midazolam, and how is it fully metabolized? (include which CYP is involved)

A

Mizadolam (Versed) is a benzodiazepine- a sedative used prior to surgery/procedures. Allosteric activator of GABA channels.

It undergoes Phase I metabolism by CYP3A4, and then Phase II glucuronidation.

166
Q

All benzodiazepines are metabolized by _____ in Phase II.

A

Glucuronidation

167
Q

What is a common structural element of all benzodiazepines?

A

a 7 membered diazepine ring, containing 2 N atoms

168
Q

Oxidations & hydrations constitute Phase ____ metabolism reactions.

A

Phase I

169
Q

What is a cosubstrate, and why is it important?

A

In a Phase I CYP-dependent metabolism reaction, a cosubstrate is a second enzyme besides CYPs that is needed for the reaction to occur.

In Phase I reactions, this enzyme is NADPH cytochrome p450 reductase- it supplies electrons and protons to activate oxygen, creating a reactive oxygen species needed for the rxn to finish successfully.

170
Q

Where are the CYPs and cosubstrate located within the cell and within the body?

A

On the cytoplasmic side of the smooth ER, in the liver and intestine

171
Q

Describe aliphatic hydroxylations. Give a substrate example.

A

Phase I rxn where CYP will add an OH group to an aliphatic carbon. Ex. Midazolam

172
Q

Describe aromatic hydroxylations. Give a substrate example.

A

Phase I rxn where a CYP will add an OH group to a carbon in a ring. Ex. Propanolol

173
Q

Describe propanolol, its therapeutic intent, its mechanism of action, and which CYP it is metabolized by.

A

-lol= beta blocker

anti-hypertensive, anti-arrhythmic, reduces HR and cardiac contraction

Blocks beta1 adrenoreceptors which mediate HR and cardiac contraction

Metabolized by CYP2C19

174
Q

Describe S-oxidations. Give an example of a substrate.

A

Phase I rxn where a CYP adds an O to an S atom on a drug. Ex. Cimetidine (Tagamet)

175
Q

Describe Cimetidine- it’s therapeutic effect, mechanism of action, and any other special properties.

A

Cimetidine (Tagamet) suppresses acid secretion. It’s used to treat heartburn, acid reflux, and ulcerative GI condtions.

Mechanism of action: competitive inhibitor of histamine @ H2 receptors (normally histamine, released from paracrine cells as a result of gastrin signaling, will bind to parietal cell H2 receptors causing HCl secretion)

Cimetidine is also an inhibitor of all the CYPs (except CYP2E1).

176
Q

Describe how drug interactions may affect CYP metabolism.

A

One drug substrate for a CYP may act as a competitive inhibitor of another, if the first drug has such high affinity for the CYP that it is unable to let go.

177
Q

Describe N-oxidations. Give an example of a substrate.

A

Phase I rxns where CYP adds an O group to an N atom on the drug. Ex. Diphenhydramine (Benadryl)

178
Q

Describe diphenhydramine (Benadryl)- it’s therapeutic effect, & mechanism of action

A

Used to treat mild allergic reactions.

Mechanism of action: competitive inhibitor of histamine @H1 receptors. (Normally, histamine released from mast cells binds to H1 receptors and causes mild allergies)

179
Q

Describe N-dealkylation, and list common substrates.

A

CYP removes an alkyl (-CH2) group from an N. Ex. methylxanthines (caffeine, theophylline), diazepam (Valium)

180
Q

At low doses, how do caffeine & theophylline act? At high doses, how do they act?

A

At low doses, caffeine & theophylline (methylxanthines) are competitive inhibitors of adenosine receptors. They will inhibit cell function.

At high doses, they inhibit phosphodiesterases, increasing cAMP.

181
Q

What is theophylline used to treat?

A

It’s a last resort drug to treat asthma & COPD

182
Q

What is diazepam (valium) used to treat? Describe its metabolism.

A

Diazepam is a benzodiazepine used to treat seizures, anxiety, muscle spasms, etc.

It’s metabolized by CYP3A4 and CYP2C19 into 2 active metabolites, 1 being very long acting. Then, it is glucuronidated in Phase II metabolism.

183
Q

Describe O-dealkylation. List common substrates.

A

Phase I rxn where CYP removes an alkyl group from O. Ex. Codeine, hydrocodone

184
Q

What are the two CYP-independent Phase I oxidation rxns? (list, no need to describe)

A

Dehydrogenase rxns (ethanol breakdown), and oxidative deamination

185
Q

Describe ethanol metabolism.

A

Ethanol ——> acetaldehyde ——> acetate.

The enzyme in the first step is alcohol dehydrogenase (found in liver and stomach), and it uses an NAD+ and produces an NADH.

The enzyme in the second step is aldehyde dehydrogenase, and it also uses an NAD+ and produces an NADH.

Acetaldehyde buildup causes nausea, headache, dizziness, facial flush.

186
Q

What substrate builds up and causes toxicity in chronic alcoholics?

A

NADH. NADH is produced in both steps of ethanol metabolism. NADH buildup causes metabolic disorders, an increase in reactive oxygen species, and liver destruction.

187
Q

Describe Phase I hydrolysis rxns. What are the enzymes involved, and what are common substrates?

A

Addition of H2O, catalyzed by esterases and hydrolases.

Ex. acetylcholine, succinylcholine

188
Q

What are the reactions of Phase II metabolism? (list, don’t describe). Which is the most common?

A

SAGGMeth: sulfation, acetylation, glutathione, glucuronidation, methylation.

Glucuronidation is the most common.

189
Q

Describe sulfation. What are common substrates?

A

Sulfation is the addition of a sulfate group to an OH. Occurs via sulfotransferases (found in the cytosol.) These require PAPS (ADP-Sulfate) to function.

Ex. Albuterol, Acetaminophen. Acetaminophen doesn’t require Phase I metabolism bc it already has a free OH. Sulfation will metabolize it into a nontoxic sulfate.

190
Q

What is special about acetylation?

A

Unlike all other metabolism reactions, acetylation makes drugs less polar (more lipid-soluble) but still inactive.

191
Q

Describe glucuronidation. What are common substrates?

A

Glucuronidation is the addition of a glucuronide molecule to the drug. It occurs via UDP Glucuronyl Transferase (UGT), found in the smooth ER. It requires UDP glucuronic acid as a substrate along with the drug.

Ex. Lorazepam (Ativan). It already has an OH group so therefore doesn’t need Phase I metabolism. UGT will turn it into lorazepam glucuronide.

Ex. Acetaminophen. In addition to sulfation, glucuronidation is another way to inactive acetaminophen.

192
Q

Why is lorazepam (Ativan) a good drug for the elderly?

A

Lorazepam is a useful benzo for the elderly because glucuronidation does not decline in the elderly. Other benzos require CYPs in Phase I, and CYP function declines substantially in the elderly.

193
Q

Describe glutathione. What is an example substrate?

A

Glutathione reactions protect against electrophiles. The transfer of an electrophile to an HS site on glutathione is catalyzed by Glutathione-S transferase (GST).

ex. Benzoapyrene (epoxide) can be neutralized by glutathione reactions.

194
Q

What is special about tumor cells and glutathione reactions?

A

Some tumors express high levels of glutathione S-transferase (GST), causing tumor cells to proliferate and be resistant to cancer chemo drugs.

195
Q

What are some substrates of CYP3A4?

A

Most drugs are metabolized by CYP3A4. These include Diazepam, Midazolam (-azolam benzos), oral contraceptive, lidocaine, etc.

196
Q

What makes CYP3A4 have so many substrates?

A

It has a very large active site and accepts a large number of ligands (shape does not matter).

197
Q

What are 2 inhibitors of CYP3A4?

A

Ketoconazole (-conazole) and erythromycin. These are 2 very different looking molecules- another example of how shape doesnt matter with CYP3A4

198
Q

Which CYP is most commonly involved in genetic polymorphisms?

A

CYP2D6

199
Q

What is a prodrug? Give 2 examples.

A

A prodrug is a drug that needs to be metabolized to be activated.

Ex. Codeine (analgesic, CYP2D6 will metabolize it to morphine), clopidogrel (anti-platelet drug, CYP2C19 will metabolize it to be active)

200
Q

What is an example of a drug that is already active but metabolized into other active intermediates?

A

Diazepam- metabolized by CYP3A4 and CYP2C19 into 2 other active intermediates, resulting in long lasting effects

201
Q

Describe how acetaminophen can be metabolized into a toxic intermediate. What are the steps taken after this intermediate is produced?

A

At low doses, acetaminophen is metabolized by sulfation and glucuronidation into nontoxic metabolites.

At high doses, CYP2E1 and CYP3A4 will metabolize it into a toxic electrophile called NAPQI.

After this, there are 2 choices: glutathione can rescue the molecule by neutralizing NAPQI, or, if glutathione is low, cell death will occur.

202
Q

How can we address the issue of acetaminophen toxicity if glutathione is low?

A

Glutathione cannot be adminstered because it does not cross cell membranes well. Instead, we use N-acetylcysteine (NAC). Its structurally similar to glutathione and can act in the same way.

203
Q

List the 3 CYPs most relevant to polymorphisms.

A

CYP2D6, CYP2C19, CYP2C9

204
Q

Describe the genetic variations seen in CYP2D6. Which populations are most affected?

A

Poor metabolizers: 2 mutant CYP2D6 alleles. Little to no enzyme function. Will need to lower drug dose to avoid toxicity. (8-10% of Caucasians)

Rapid/Ultrarapid metabolizers: duplification/amplification of functional CYP2D6 alleles. Will need to increase drug dose so it can reach therapeutic levels. (30% of North Africans)

205
Q

What are some CYP2D6 substrates?

A

Beta- blockers, and lots of CNS drugs- ex. opioids (codeine, hydrocodone), antidepressants, amphetamines.

206
Q

Describe metoprolol & its mechanism of action. Describe how metoprolol dosing may need to be adjusted based on individual polymorphisms.

A

Metoprolol is a beta blocker (-lol). It blocks B1 adrenoreceptors, making it an anti hypertensive, anti arrhythmic, anti anginal drug.

It is metabolized by CYP2D6. Poor metabolizers will require lower doses to avoid toxicity. Rapid/Ultrarapid metabolizers will require higher doses to achieve therapeutic levels.

207
Q

What is the difference between metoprolol and propanolol?

A

They are both beta blockers (-lol). Metoprolol acts to block B1 adrenoreceptors, and propanlol acts to block B1 and B2 adrenoreceptors. B2 agonists relax the airways, making metoprolol the better choice for patients with asthma and COPD.

(also, metoprolol is metabolized by CYP2D6 while propanlol is metabolized by CYP2C19)

208
Q

Describe how the dosing of codeine may need to be adjusted to account for individual polymorphisms.

A

Codeine is a prodrug, metabolized by CYP2D6 into active morphine.

Poor metabolizers: drug should be avoided because it’s not useful/therapeutic.

Rapid/Ultrarapid metabolizers: require lower doses (to avoid morphine toxicity)

209
Q

What is the common polymorphism seen in CYP2C19? What population is the most affected?

A

Poor metabolizer polymorphisms- 15-25% of the Asian population

210
Q

What are important substrates of CYP2C19?

A

Diazepam and other -azepam (long-acting) benzodiazepines, omeprazole (-prazole: proton pump inhibitors), clopidogrel, propanolol

211
Q

Describe how diazepam metabolism is affected for CYP2C19 poor metabolizers.

A

Diazepam is metabolized by CYP3A4 and CYP2C19. It is metabolized into 2 long acting intermediates. For poor metabolizers, the half life of diazepam will be increased greatly.

212
Q

Describe the common variation seen in CYP2C9. Which population is most affected?

A

Poor metabolizers- 10-35% of Caucasian pop

213
Q

What are important substrates of CYP2C9?

A

Warfarin, NSAIDs (non-steroidal anti inflammatory drugs) including iburprofen & celecoxib

214
Q

How is warfarin metabolism affected in CYP2C9 poor metabolizers?

A

Warfarin is an anticoagulant often used post surgery. It has a very narrow TI. Poor metabolizers will show reduced warfarin clearance, changing (reducing) the number of days required to reach stable anticoagulation.

215
Q

What is the difference between ibuprofen and celecoxib (both NSAIDs)?

A

Ibuprofen and celecoxib are both pain relievers & anti-inflammatory. Ibuprofen inhibits both COX1 (housekeeping protein, used to maintain normal kidney and GI function) and COX2 (causes pain). Celecoxib inhibits only COX2. This makes it a better choice for post op pain, because it doesn’t affect the kidney or GI.

216
Q

How does NSAID dosing need to be adjusted for CYP2C9 poor metabolizers?

A

CYP2C9 poor metabolizers will require lower doses of NSAIDs

217
Q

Describe variations in the enzymes involved in ethanol metabolism. What accumulates as a result of these variations? What population is most affected?

A

Both increases in the amount of alcohol dehydrogenase (90% of Chinese pop) and decreases in the amount of aldehyde dehydrogenase (30-50% of Chinese/Japanese pop) will cause accumulation of acetaldehyde.

218
Q

What is the effect of mu and theta genotypes of glutathione S-transferase (GST)?

A

Glutathione is needed to neutralize evil electrophiles. Mu and theta genotypes have null alleles of GST. This leads ot an increased risk of cancer and increased toxicity of chemo drugs.

Mu genotype- 50% of Caucasian pop

Theta genotype- 60% of Chinese/Korean pop

219
Q

Explain how ethanol + acetaminophen interaction is toxic.

A

At high doses, acetaminophen is metabolized by CYP2E1 and CYP3A4 into the toxic electrophile NAPQI.

High doses of ethanol cause induction of CYP2E1, helping create NAPQI, and also reduced glutathione, preventing rescue from NAPQI.

220
Q

Charbroiled foods + environmental pollutants induce _____, causing epoxide formation.

A

CYP1A2

221
Q

What is Ginkgo used for? What are its effects on the CYPs?

A

Ginkgo is a herbal remedy used to enhance memory. It induces CYP3A4, CYP2C9, and CYP2C19.

222
Q

What is Echinacea? What effects does it have on the CYPs?

A

Echinacea is a herbal remedy used to enhance the immune system (ex. treat colds). It induces CYP3A4 and inhibits CYP1A2.

223
Q

What is St. Johns wort? What are its effects on the CYPs? How does it work to cause these effects?

A

St. Johns wort is a herbal remedy used to treat mild depression. It induces CYP3A4, CYP2C9, CYP2C19, and CYP2E1.

It goes straight to the nucleus and binds to receptors + coactivators, causing increased CYP gene transcription.

224
Q

what are the 3 processes for drug elimination through the kidneys?

A
  1. glomerular filtration
  2. tubular secretion
  3. passive tubular reabsorption
225
Q

will protein-bound drugs be secreted through the glomerulus? how about through the proximal tubule?

A

NO

only unbound drugs will be secreted

226
Q

excretion through the glomerulus is proportional to:

(2 things)

A
  1. glomerular filtration rate (GFR)
  2. concentration of free drug in plasma
227
Q

for which groups should we decrease doses of drugs that are exclusively renally-excreted?

A

old people

people with kidney damage/disease

228
Q

where does tubular secretion of drugs occur?

A

in the proximal tubule of the kidney

229
Q

what transporters are used for renal excretion of weak bases?

weak acids?

A

organic cation transporters (OCTs) for bases

organic anion transporters (OATs) for acids

230
Q

why/how does probenecid increase the half life of penicillin?

A

probenecid and penicillin use the same transporter to be secreted. probenecid will competitively bind to that transporter and prevent penicillin from being secreted, therefore increasing the amount of penicillin in the body and increasing its half life

231
Q

explain how tubular pH can be manipulated to promote or impair reabsorption

(hint: this is used for drug overdoses!)

A

acidifying tubule prevents reabsorption of weak bases by causing them to be protonated (this enhances excretion). example = meth

alkalinizing tubule prevents reabsorption of weak acids by causing them to be deprotonated. ex = phenobarbital/other barbituates

ionized drugs will not cross the membrane!

232
Q

how does the glomerulus achieve size sensitivity?

A

it has 3 layers of membranes, all with varying sizes of fenestrations/pores/slits.

the endothelial layer has medium-sized fenestrations

the basement membrane has larger pores

the podocyte layer has tiny filtration slits

233
Q

how does the glomerulus achieve charge sensitivity?

A

endothelial cells and podocytes are lined with negatively charged glycoproteins

(negatively charged particles such as albumin are not filtered)

234
Q

which transporter mediates secretion of conjugated metabolites like glucuronides, sulfates, and glutathione in the kidney?

A

MRP2

235
Q

what is hepatic clearance determined by?

A

hepatic blood flow and extraction ratio

(extraction ratio tells you liver’s metabolic capacity)

236
Q

in terms of hepatic clearance, what are high extraction drugs limited by?

A

hepatic blood flow

(they can be metabolized rapidly, so that won’t hold them up)

237
Q

in terms of hepatic clearance, what are low extraction drugs limited by?

A

liver’s metabolic capacity and fraction of free drug

(they are metabolized very slowly, so the liver is already overwhelmed trying to clear these drugs out. increased blood flow won’t make much of a difference since the rate is already slow)

238
Q

how are drugs handled through biliary excretion eliminated?

A

through feces

239
Q

what does the OATP1B1/SLCO1B1 transporter do?

what happens when you have genetic variants of this transporter?

A

it’s an organic anion transporter that transports statin into hepatocytes

when you have the genetic variant, you are predisposed to muscle toxicity

240
Q

what is enterohepatic circulation?

A

reabsorption of drug from bile once the bile reaches the gut lumen

241
Q

how are conjugated drugs liberated back into their native state to be reabsorbed during enterohepatic circulation?

A

intestinal bacterial B-glucuroindases liberate them

242
Q

if you have malignant hyperthermia susceptibility, which drugs should you NOT take?

A

volatile anesthetics

243
Q

glucose-6-phosphate dehydrogenase deficiency leads to acute hemolysis when you take which drugs?

A

anti-malarial drugs

sulfonamides

244
Q

what is succinylcholine normally used for?

what happens if you take it when you have pseudocholinesterase deficiency?

A

it’s a transient paralytic used during surgery/intubation

if you have pseudocholinesterase deficiency, it will cause persistent paralysis

245
Q

which CYP metabolizes warfarin?

A

CYP2C9

246
Q

what’s affected when you have VKORC1 variants?

A

warfarin efficacy changes

247
Q

are pharmacogenomic inheritance patterns traceable in families??

A

no

248
Q

which CYP2D6 alleles have normal levels of activity?

A

*1, *2, *4

249
Q

which CYP2D6 alleles have intermediate/poor levels of metabolic activity?

A

*3, *5, *10

250
Q

how do you get ultra-rapid metabolism of CYP2D6?

A

duplication/amplification of CYP2D6*1

251
Q

what’s the difference between a standard drug and a prodrug?

A

a standard drug is administered in its active form

a prodrug has to be activated through metabolism

252
Q

what happens when poor metabolizers take a standard drug?

A

they have decreased elimination rate and increased toxicity risk because they can’t effectively clear the drug

253
Q

what happens when poor metabolizers take a prodrug?

A

they have decreased activation and reduced effectiveness of the drug

254
Q

what happens when ultra-rapid metabolizers take a standard drug?

A

they have increased elimination rate and reduced effectiveness of drug

255
Q

what happens when ultra-rapid metabolizers take a prodrug?

A

they have increased activation and therefore increased toxicity risk

256
Q

what happens when intermediate metabolizers take a standard drug?

A

they have potential for increased toxicity (eliminate the drug a bit slower than normal)

257
Q

what happens when intermediate metabolizers take a prodrug?

A

they have possible reduced effectiveness (aren’t transformed to active drug as quickly)

258
Q

which CYP metabolizes codeine?

A

CYP2D6

259
Q

which CYP metabolizes clopidogrel?

A

CYP2C19

260
Q

what happens if you take clopidogrel and you have CYP2C19*2 or *3 alleles?

A

you won’t metabolize prodrug clopidogrel to its active form

therefore you won’t get anticoagulative effects, so you’re at higher risk of clotting, cardiovascular complications, death, MI, etc

261
Q

what happens when you have genetic variants for TMPT and need to take an anti-cancer drug?

how is this avoided?

A

you won’t break down and excrete thiopurines as effectively, putting you at risk for bone marrow toxicity

testing for variants is done to dose correctly and avoid toxicity

262
Q

what happens if you have CYP2C9*2 or *3 alleles and take warfarin?

A

you will have slowed elimination of warfarin and increased anticoagulative effects

this puts you at risk for bleeding

263
Q

how can we monitor anticoagulative activity of warfarin?

A

do a prothrombin assay and look at the INR

264
Q

what happens if you have an INR that is higher than the therapeutic window for warfarin?

A

you get increased bleeding

265
Q

usually we see germline variants in pharmacogenomics. where do we see an example of somatic pharmacogenomic variants?

A

cancer!

specifically, non small cell lung cancers with hyperactive EGFR are more sensitive to EGFR inhibitors like gefitinib and erlotinib

266
Q

What is the equation for apparent volume of distribution?

A
267
Q

What is the clearance? What is the equation for it? When is it constant, and when does it change?

A

Clearance is the volume of plasma cleared of a drug per unit time. In 1st order drugs, its constant. For zero order drugs, it changes (rate of elimination remains constant/plateaued).

268
Q

1st order drug clearance= constant ______ of drug eliminated per unit time, while zero order clearance= constant ______ of drug eliminated per unit time.

A

Fraction, amount

269
Q

What is the equation for loading dose?

A

LD= Vd*Css

270
Q

True/False: the patient’s weight affects the apparent volume of distribution and must be accounted for.

A

True

271
Q

What is the equation for maintenance dose?

A

Maintenance dose= CL*Css

272
Q

What is Css?

A

Steady state, desired plasma concentration of drug

273
Q

What is the equation for Css in a continuous infusion?

A

Css= infusion rate/CL

274
Q

What is the equation for Css for a multiple dose drug?

A

Css= dose/(dosing interval * CL)

275
Q

What is the equation for half life?

A

t(1/2)= 0.693Vd/CL

276
Q

How many half lives does it take to reach 90% steady state (considered good enough)?

A

3.3

277
Q

What is the equation for loading dose if bioavailability is not 100%?

A

F*LD= Vd*Css

278
Q

Describe what happens with parasympathetic innervation of the heart.

A

Acetylcholine is released onto muscarinic receptors. This causes an inhibitor, membrane delimited GPCR response through activating K+ channels. This slows the heart rate.

279
Q

What is the effect of multivalent cations (except Ca2+) and aminoglycoside antibiotics on skeletal muscle?

A

They are competitive inhibitors of voltage gated Ca2+ channels- blocking Ca2+ from entering- making it so vesicles can’t release content.

280
Q

What is the effect of botulinum toxin type A on skeletal muscle? What can it be used to treat?

A

Cleaves SNAP-25 of SNARE. This blocks secretion of Ach into the neuromuscular junction. It can be used to treat spasticity (blepharospasm- a disease of eye closure due to spastic eye muscle contraction)

281
Q

Describe what happens at the somatic neuromuscular junction to cause skeletal movement.

A

There is a vesicle full of ACh. It’s connected to the Ca2+ channels in the presynaptic plasma membrane via the Fusion Machine. SNARES on the fusion machine are responsible for priming the vesicles. SNAREs have Ca2+ sensors.

If there is an AP, it will propagate down the presynaptic membrane, and voltage gated Ca2+ channels will respond and open. Ca2+ will flow into the cell, binding Ca2+ sensors on the Fusion Machine, allowing vesicles to release ACh by exocytosis into the neuromuscular junction.

ACh will bind to nicotinic ACh receptors and cause Na+ influx, which produces the EPP. EPP will generate an action potential via voltage gated Na+ channels opening.

282
Q

What is the role of TTX?

A

At the skeletal neuromuscular junction, it blocks voltage gated Na+ channels from the outside, preventing initiation of AP in nerve and skeletal muscle.

It blocks the input to synapses (intial AP needed for Ca2+ influx) and the output ( the production of AP from the EPP).

283
Q

What are the -curoniums? What are they used for?

A

Rocuronium and vecuronium- nondepolarizing blockers. Competitive inhibitors at skeletal neuromuscular junction. Used to relax skeletal muscle during surgical procedures.

284
Q

What is Sux (succinylcholine) used for?

A

Used to relax skeletal muscles for intubation. It’s a depolarizing blocker because it binds to nicotinic receptors and depolarizes/activates the muscle before it blocks. Very fast acting.

285
Q

What is neostigmine?

A

A reversible inhibitor of acetycholinesterase, making it so ACh has to remain in the cleft longer, improving transmission when nicotinic and muscarinic receptors are impaired (ex. after surgery and to treat disease of muscle fatigue).

286
Q

Fight or flight is the ______ division of the ANS, while rest and digest is the ______ division.

A

Sympathetic, parasympathetic

287
Q

Where are preganglionic fibers located for sympathetic nerves?

A

Thoracic and lumbar regions of the spinal cord

288
Q

Where are preganglionic fibers located for the parasympathetic division of the ANS?

A

Cranial and sacral regions of CNS

289
Q

Muscarinic receptors are ______ (GPCRs/Ligand gated ion channels).

A

GPCR

290
Q

How is norepinephrine action terminated?

A

Reuptake by SLC into presynaptic membrane (SLC6A2)

291
Q

What is the therapeutic use of atropine? How does it act?

A

Atropine is used to increase heart rate during spinal anesthesia. It is a competitive inhibitor of Ach at muscarinic receptors.

292
Q

Describe what happens after ACh binds to a receptor in the parasymapthetic nerve terminal for cardiac muscle.

A

Parasympathetic= rest/digest.

ACh binds to muscarinic receptors on postsynaptic membrane, these are GPCRs. It acts via a membrane delimited process- beta gamma subunits of G protein break off and activate K+ channels, inhibiting cardiac muscle (reducing heart rate and contraction).

293
Q

How does Ach released by parasympathetic nerve endings constrict airways?

A

Parasympathetic= rest+ digest.

Ach binds to muscarinic receptors, causing the alpha subunit of the G protein to break off and activate Phospholipase C. The water soluble arm of this involves PIP2 synthesizing IP3, which releases Ca2+ from storage sites. This causes smooth muscle contracition- constriction of airways.

294
Q

What is the mechanism of action of ipratropium and tiotropium? What are they used for clinically?

A

Used to treat airway disorders (asthma, COPD). They are competitive inhibitors of ACh at muscarinic receptors- inhibiting the inhibiton of airway opening.

295
Q

How does norepinephrine released by sympathetic nerve endings onto alpha1 receptors constrict blood vessels?

A

NE binds to alpha1 adrenoreceptors (GPCR), which works through the Phospholipase C inhibitory pathway. Ca2+ release causes smooth muscle contraction via alpha1 adrenoreceptors.

296
Q

What is phenylephrine used for?

A

It’s an agonist of alpha1 adrenoreceptors. Can be used to relieve nasal congestion by constricting blood vessels in nasal passages, and increase blood pressure during surgery.

297
Q

What is the neurotransmitter that increases heart rate and contractile force in the sympathetic NS? How does it work?

A

Epinephrine or norepinephrine.

Binds to B1 receptors, which are GPCRs- causes alpha subunit to dissociate and stimulate adenylyl cyclase, causing cAMP formation. This leads to activated PKA and protein phosphorylation.

298
Q

Where are beta2 receptors located?

A

Smooth muscle- blood vessels and bronchioles

299
Q

What happens to smooth muscle when B2 agonists bind to their receptors?

A

Smooth muscle relaxation– happens via adenylyl cyclase activation and cAMP synthesis

300
Q

How does cAMP work in cardiac muscle?

A

increases Ca2+ entry and increases stores of Ca2+, causing contraction

301
Q

how does cAMP work in smooth muscle?

A

decreases cellular Ca2+ concentrations, making it so less is available for contration, causing relaxation. Increases calcium removal/reduces its entry

302
Q

What is the mecahnism of action of beta blockers like propanolol and metoprolol?

A

Competitive inhibitors of NE at beta1 receptors in the heart.

303
Q

What is the mechanism of action of albuterol (-erols)?

What are they used for?

A

Used to treat asthma and COPD.

They are agonists of B2 receptors, relaxing smooth muscle and opening airways.

304
Q

The control of blood vessel ____ is the major determinant of blood pressure.

A

Radius

305
Q

Describe sympathetic tone and the constriction/dilation of blood vessels.

A

Increasing the tonic firing of AP of the sympathetic nerve will cause an increase in the amount of norepi release onto A1 receptors of blood vessels, causing vasodilation.

Decreasing the firing of AP of the sympathetic nerve will cause a decrease in the amount of norepi released, causing vasoconstriction.

This is the major determinant of blood pressure- controlling radius of the vessel.

306
Q

Norepinephrine does not activate____ receptors.

A

B2

307
Q

______ can activate all catecholamine receptors(a1, a2, B1, B2)

A

Epinephrine

308
Q

B2 receptors on blood vessels are _____ (innervated/not innervated)

A

not innervated. NE cannot activate B2 receptors.

309
Q

What activates B2 receptors on blood vessels? What is the effect?

A

Epi released from adrenal medulla. Happens during emergency situations. Vasodilation

310
Q

What are the receptors on the heart, and what effects do their agonists have?

A

parasympathetic: muscarinic Ach receptors. Ach increases K+ conductance, slowing heart rate.

Sympathetic: B1. Epinephrine binds. Increases heart rate and cardiac contraction via cAMP/PKA pathway.

311
Q

What are the receptors on blood vessels? What effects do their agonists have?

A

Sympathetic: B2- not innervated. Epi from bloodstream causes vasodiation via cAMP increases during emergencies.

Sympathetic: A1- Norepi and epi are agonists. Bind and cause smooth muscle contraction via Ca2+ release from Phospholipase C activation.

312
Q

What are the receptors on the airways? What effects do their agonists have?

A

Parasympathetic: muscarinic. ACh constricts airways using smooth muscle contraction.

Sympathetic: B2. Receptor activation by -erol drugs causes airway smooth muscle relaxation

313
Q
A
314
Q
A
315
Q
A
316
Q
A
317
Q
A
318
Q
A
319
Q
A
320
Q
A
321
Q
A