pharmacology Flashcards

1
Q

what 3 things are considered when considering a drug’s effects?

A

where the effect(s) are produced

the drug’s target

response produced after interaction with target

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

how would the effects of cocaine be described when considering the 3 main points (location of effect, target, response produced)?

A

overall: euphoric ‘high’

where effect takes place
- dopaminergic neurons in nucleus accumbens in brain

target of drug
- dopamine reuptake protein on pre-synaptic terminal

response produced
- cocaine blocks dopamine reuptake protein, preventing quick removal of dopamine from synapse (therefore more available to bind to receptor and activate it)

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

what is the key thing that allows a drug to produce an effect?

A

drug ‘binds’ to target

either enhances activation (“stimulation”) or prevents activation (block effect from being produced)

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

what are the 4 classes of drug target proteins? give an example of a drug that targets each one.

A

receptors (e.g. nicotine)

enzymes (e.g. aspirin)

ion channels (e.g. local anaesthetic)

transport proteins (e.g. Prozac)

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

how does aspirin act on enzymes (target protein) to bring about an effect?

A

aspirin binds to the enzyme cyclooxygenase

blocks production of prostaglandins

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

how does local anaesthetic act on ion channels (target protein) to bring about an effect?

A

block sodium ion channels

prevents nerve conduction

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

how does Prozac act on transport proteins (target protein) to bring about an effect?

A

block serotonin carrier proteins

prevents serotonin being removed from the synapse

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

how does nicotine act on receptors (target proteins) to bring about an effect?

A

binds to and activates nicotinic acetylcholine receptor

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

what characteristic must a drug display to be an effective therapeutic agent?

A

high degree of selectivity for particular drug target

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

why is it difficult to produce drugs with complete selectivity? what effect does this have?

A

many drugs and chemicals are structurally quite similar - therefore they all have some degree of specificity for the receptors of the other chemicals

when producing a drug, there is a chance that the drug will interact with receptors that aren’t the intended one, producing side-effects

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

how does drug dose relate to drug selectivity?

A

at lower dose, drug will interact with only one target to produce the intended effect

as dose increases, effects become less specific as drug begins to interact with other drug targets

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

what is an example of how drug dose affects selectivity?

A

Pergolide (treatment of Parkinson’s)

low dose

  • target: dopamine D2 receptor
  • effect: anti-Parkinsonian

medium dose

  • target: serotonin receptor
  • effect: hallucinations

high dose

  • target: adrenergic receptor
  • effect: hypotension
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

what are the 4 types of interaction between drugs and receptors?

A

electrostatic

hydrophobic

covalent bonds

stereospecific

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

what types of bonds are involved in electrostatic interactions between drugs and receptors?

A

(most common interaction)

includes hydrogen bonds and van del Waal’s forces

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

when are hydrophobic interactions between drugs and receptors important?

A

in use of lipid soluble drugs

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

why are covalent bond interactions the least common type of interaction between drugs and receptors?

A

irreversible

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

why is stereospecific interaction between drugs and receptors important?

A

many drugs exist as stereoisomers and interact stereospecifically with receptors

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

what is pharmacology?

A

study of how a drug interacts with living organisms and how this influences physiological function

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

what does the term ‘pharmacodynamics’ refer to?

A

what the drug does to the body

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

what does the term ‘pharmacokinetics’ refer to?

A

what the body does to the drug

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

how can the basic reaction between a drug and its receptor be represented?

A

equilibrium between drug and receptor, drug-receptor complex

for a specific concentration of the drug, a specific number of drug receptor complexes are formed

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

what happens to the equilibrium between and drug and receptor and the drug-receptor complexes when the concentration increases?

A

more drug molecules available to bind to free receptors

equilibrium shifts to the right

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

what happens to the equilibrium between and drug and receptor and the drug-receptor complexes when the concentration decreases?

A

more receptors become available due to lower drug concentration

shifts equilibrium to the left

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

what are the 2 categories of drug in terms of interaction with receptors?

A

agonist

antagonists

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

what is the difference between agonist and antagonists?

A

both bind to receptors, but only agonists bind and activate receptors

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

what are the 2 key properties of agonists?

A

affinity

efficacy

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

what does the affinity of a drug determine?

A

strength of binding of drug to receptor, the strength of each drug-receptor complex

therefore affinity is strongly linked to receptor occupancy

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

how is affinity linked to receptor occupancy?

A

each individual drug receptor interaction is transient, with many interactions only lasting milliseconds - at any given moment a particular drug molecule might be bound to a receptor, or it may have unbound and may currently be free with the potential to bind another receptor

therefore if two drugs were added to the tissue (i.e. same number of receptors available), the drug with the higher affinity will form stronger drug receptor complexes

thus at any given moment, it is more likely that more of this drug will be bound to receptors

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

what is efficacy?

A

ability of an individual drug molecule to produce an effect once bound to a receptor

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

what are the 3 types of drug with reference to efficacy?

A

antagonists

partial agonists

full agonists

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

how does an antagonist work with reference to efficacy?

A

drug has affinity for the receptor but no efficacy

effectively ‘blocking’ a receptor when bound to it

prevents an agonist from binding to the receptor and inducing activation

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

how does a partial agonist work with reference to efficacy?

A

drug has affinity for the receptor and sub-maximal efficacy

when bound to the receptor, it can produce a partial response, but cannot induce the maximal response from that receptor

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

how does a full agonist work with reference to efficacy?

A

drug has affinity for the receptor and maximal efficacy

produces the maximal response expected from receptor when bound to it

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

what is potency?

A

concentration or dose of a drug required to produce a defined effect

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

what is the standard measure of potency?

A

determine the concentration or dose of a drug required to produce a 50% tissue response

called EC50 (half maximal effective concentration) or ED50 (half maximal effective dose)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

what is the difference between EC50 and ED50?

A

concentration that produced a 50% response is EC50

in some cases it is difficult to assess what a 50% response is (e.g. what does a 50% improvement in breathlessness look like?)

therefore dose of drug that produced the desired effect in 50% or the individuals tested is used (ED50)

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

how is potency related to dose?

A

the lower the dosage required to produce a particular effect, the more potent the drug is (often compared using ED50)

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

what is the relationship between efficacy and potency?

A

potency is related to dose, efficacy is not

highly potent drugs produce a large response at relatively low concentrations

highly efficacious drugs can produce a maximal response - effect not particularly related to drug concentration

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

what is the clinical relevance of the difference between potency and efficacy?

A

efficacy more important - drug should induce a maximal response

potency only gives dose needed to produce a response

if 2 drugs have equal efficacy, potency does not matter (maximal response with the less potent drug can be achieved by administering a slightly higher concentration)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

what are the 4 major pharmacokinetic factors?

A

absorption

distribution

metabolism

excretion

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

how is absorption defined with regard to pharmacokinetics?

A

passage of a drug from site of administration into plasma

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

what is bioavailability (with regard to absorption in pharmacokinetics)?

A

fraction of the initial dose that gains access to the systemic circulation

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

what is the difference between absorption and bioavailability?

A

absorption deals with the process for drug transfer into the systemic circulation

bioavailability deals with the outcome of drug transfer into the systemic circulation (i.e. how much)

determined by site of administration-

e. g. IV administration of drug
- absorption (process for drug passage): injecting dose into circulation
- bioavailability: 100% (hypothetical)

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

what are some common forms of drug administration?

A

IV

oral

inhalational

dermal (percutaneous)

intra-nasal

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

what are the 2 ways that drugs move around the body?

A

bulk flow transfer (i.e. in the bloodstream)

diffusional transfer (i.e. molecule by molecule across short distances)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

why is bioavailability likely to be less than 100% when drugs are not administered through an IV?

A

if IV is used, bulk flow transfer will deliver drug to intended site of action

other routes - in order for the drug to reach the bloodstream it is first going to need to diffuse across at least one lipid membrane

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

what are the 4 mechanisms by which chemicals diffuse across plasma membranes?

A

pinocytosis

diffusion across aqueous pores

diffuse across lipid membranes

carrier mediated transport

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

what is pinocytosis and and what is its relevancy in drug transport?

A

small part of cell membrane envelopes chemical molecule and forms a vesicle

chemical released on other side of membrane

e.g. insulin access to brain

but rarely used to transport drugs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

why is diffusion across an aqueous pores not a major route for drug transport across membranes?

A

most pores are less than 0.5nm in diameter

very few drugs are this small

therefore there is little movement of drugs across this aqueous route

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

how do most drugs move across membranes?

A

diffusion across lipid membrane down concentration gradient

carrier mediated transport (transmembrane protein binds drug molecules on one side of membrane, transfers them across to the other side - can be against concentration gradient using ATP)

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

why are the majority of drugs more water soluble than lipid soluble?

A

large proportion of drug molecules are given orally

need to be water soluble to dissolve in the aqueous environment of GI tract and be available for absorption

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

what feature of a drug affects its lipid solubility?

A

ionisation

drugs are either weak acids (e.g. aspirin) or bases (e.g. morphine) - donate/accept protons respectively when ionised

unionised form of drug retains more lipid solubility - more likely to diffuse across plasma membranes

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

what 2 things affect the ionisation of a drug?

A

dissociation constant (pKa) for that drug

pH in that particular part of the body

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

how does a drug’s dissociation constant (pKa) and the pH in a particular area of the body affect a drug’s ionisation when the drug is a weak acid?

A

if drug pKa and tissue pH are equal, drug will be equally dissociated between the two forms (i.e. 50% ionised and 50% unionised)

as pH decreases below pKa (more acidic tissue): unionised form starts to dominate (equilibrium shifts to the left)

as pH increases above pKa (more basic tissue): ionised form starts to dominate (equilibrium shifts to the right)

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

how does a drug’s dissociation constant (pKa) and the pH in a particular area of the body affect a drug’s ionisation when the drug is a weak base?

A

if drug pKa and tissue pH are equal, drug will be equally dissociated between the two forms (i.e. 50% ionised and 50% unionised)

as pH decreases below pKa (more acidic tissue): ionised form starts to dominate (equilibrium shifts to the right)

as pH increases above pKa (more basic tissue): unionised form starts to dominate (equilibrium shifts to the left)

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

what is ion trapping?

A

weak bases poorly absorbed from stomach due to low pH causing high drug ionisation - ions are “trapped” in the stomach

weak acids can be absorbed from the stomach (low pH allows them to stay unionised) but when they become ionised due to higher physiological pH ions may become “trapped” in the blood

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

how is ion trapping overcome?

A

transport proteins

weak bases become “trapped” in the stomach - however, when the small intestine is reached, many transport proteins will allow absorption from GI tract

weak acids become “trapped” in blood - however, most tissues have transport proteins that could move the ionised drug out of the bloodstream into the tissue

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

what are the 4 places where the most important carrier systems for drug action are found?

A

renal tubule

biliary tract

blood brain barrier

gastrointestinal tract

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

what are carrier systems in the renal tubule, biliary tract, blood brain barrier and GI tract responsible for?

A

(overcoming ion trapping)

drug access to the bloodstream (absorption from GI tract)

drug access to certain tissues (absorption across blood brain barrier)

excretion of drugs from the body (excretion from the kidney of the GI tract)

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

what is distribution?

A

when a drug is absorbed it is then distributed to various tissues where they eventually take effect

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

what are the 4 factors that influence tissue distribution?

A

regional blood flow

plasma protein binding

capillary permeability

tissue localisation

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

how does regional blood flow affect tissue distribution?

A

different tissues receive different amounts of cardiac output

therefore more drug will be distributed to those tissues that receive most blood flow

distribution can change depending on circumstances:

  • during exercise there is more blood flow to muscles
  • after a large meal the stomach and intestines will receive more blood flow
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

how does plasma protein binding affect tissue distribution?

A

drugs often bind to plasma proteins (e.g. albumin) when they reach systemic circulation

only free drug is available to diffuse out of the blood and access tissues - bound drugs cannot leave the blood until they dissociate from the protein

free drug + protein binding site ↔ drug-protein binding site

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

what 3 factors affect the amount of drug that binds to plasma protein?

A

free drug concentration

affinity for the protein binding sites

plasma protein concentration

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

why are plasma proteins never saturated with drugs?

A

e. g. albumin
- albumin blood concentration: 0.6mmol/l
- each molecule has 2 binding sites
- therefore binding capacity: 1.2mmol/l

drug plasma concentration required for clinical effect is generally less than 1.2mmol/l

therefore plasma proteins are never saturated with drugs

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

what factor affects differences in the extent of plasma protein binding?

A

mostly due to particular affinity for protein binding sites for a particular drug

e.g. acidic drugs bind particularly well to albumin, therefore tend to be more heavily plasma protein bound

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

what are the 4 different types of capillary structure?

A

continuous

blood brain barrier

fenestrated

discontinuous

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

how does a continuous capillary structure affect capillary permeability and therefore drug distribution?

A

most capillaries in the body have the continuous structure

if drugs are very lipid soluble then they can diffuse across the endothelial cell and access the tissue

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

what does a continuous capillary structure look like?

A

endothelial cells aligned in single file with small gap junctions between the cells

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

what does the blood brain barrier’s capillary structure look like?

A

continuous structure, but with tight junctions between endothelial cells

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

what are the 4 different types of capillary structure?

A

continuous

blood brain barrier

fenestrated

discontinuous

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

how does a continuous capillary structure affect capillary permeability and therefore drug distribution?

A

most capillaries in the body have the continuous structure

if drugs are very lipid soluble then they can diffuse across the endothelial cell and access the tissue

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

what does a continuous capillary structure look like?

A

endothelial cells aligned in single file with small gap junctions between the cells

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

what does the blood brain barrier’s capillary structure look like?

A

continuous structure, but with tight junctions between endothelial cells

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

why is the brain the most difficult tissue in the body for drugs to gain access to?

A

blood brain barrier’s capillary structure

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

how are less lipid soluble drugs transported into the tissue?

A

if very small, can pass through gap junctions

otherwise, via carrier proteins

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

what is an example of a tissue with a discontinuous capillary structure?

A

liver

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

how does a fenestrated capillary structure allow the kidney to carry out its function?

A

kidney is a key tissue involved in excretion of chemicals (including many drugs)

fenestrations allow for some small drugs to pass from blood to kidney tubules which will enhance excretion of these drugs

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

what does a discontinuous capillary structure look like?

A

big gaps between capillary endothelial cells

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

what does a fenestrated capillary structure look like?

A

fenestrations - circular windows within endothelial cells that allow for passage of small molecular weight substances (including some drugs)

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

what is an example of a tissue with a fenestrated capillary structure?

A

kidney glomerulus

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

how does a discontinuous capillary structure allow the liver to carry out its function?

A

kidney is a key tissue involved in excretion of chemicals (including many drugs)

fenestrations allow for some small drugs to pass from blood to kidney tubules which will enhance excretion of these drugs

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

how does tissue localisation affect drug distribution?

A

both lipid and water soluble drugs diffuse out of the blood into the brain down a concentration gradient, establishing an equilibrium between the blood and brain

however, equilibrium position is different in lipid and water soluble drugs -

lipid soluble: equilibrium more heavily weighted towards retention in the brain as brain has higher fat content

water soluble: equilibrium more heavily weighted towards retention in the plasma as blood has the higher water content

therefore a larger proportion of the lipid soluble drug will be ‘localised’ in the brain as compared with the water soluble drug

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

what are the 3 main mechanisms of action of phase 1 of drug metabolism?

A

oxidation (produces electrophiles)

reduction (produces nucleophiles)

hydrolysis (produces nucleophiles)

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

what happens during oxidation in phase 1 of drug metabolism?

A

(most common mechanism)

all oxidation reactions start with a hydroxylation step using the cytochrome P450 system to incorporate oxygen into non-activated hydrocarbons

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

what enzymes within the liver are responsible for drug metabolism?

A

mostly cytochrome P450

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

what are the the 2 kinds of drug metabolism that act together to decrease lipid solubility?

A

phase 1 – introduce a reactive group to the drug

phase 2 – add a conjugate to the reactive group

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

what are the 3 main mechanisms of action of phase 1 of drug metabolism?

A

oxidation

reduction

hydrolysis

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

what happens in oxidation in phase 1 of drug metabolism?

A

(most common mechanism)

all oxidation reactions start with a hydroxylation step using the cytochrome P450 system to incorporate oxygen into non-activated hydrocarbons

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

what is the end result of phase 1 of drug metabolism?

A

incorporation of certain functional groups (-OH, -COOH, -SH, NH2) or unmasking existing groups in the parent drug

produces metabolites with functional groups that serve as a point of attack for conjugating systems of phase 2

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

what are pro-drugs?

A

phase 1 of drug metabolism often produces pharmacologically active metabolites

sometimes the parent drug has no activity of its own - will only produce an effect once it has been metabolized to the respective metabolite (i.e. pro-drugs)

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

what enzymes are used in phase 2 of drug metabolism?

A

transferases

transfer the substituent group onto the phase 1 metabolite

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

what occurs in phase 2 of drug metabolism?

A

attachment of a substituent group to functional groups

produces metabolites that are more water soluble and are usually inactive to facilitate excretion in the urine or bile

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

what are the common phase 2 conjugates that complement the usual phase 1 metabolites?

A

for electrophiles:
- glutathione conjugation

for nucleophiles:

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

what is first pass (pre-systemic) hepatic metabolism?

A

orally administered drugs are mostly absorbed from the small intestine and enter the hepatic portal blood supply - therefore pass through the liver before reaching systemic circulation

drug is heavily metabolised so not much active drug will reach systemic circulation

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

what are the 3 major routes for drug excretion via the kidney?

A

glomerular filtration

active tubular secretion (or reabsorption)

passive diffusion across tubular epithelium

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

what is the problem with the usual solution for first pass (pre-systemic) hepatic metabolism

A

extent of first pass metabolism varies amongst individuals, therefore the amount of drug reaching the systemic circulation also varies

therefore drug effects and side effects are difficult to predict

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

what are the 2 most important routes of excretion?

A

via kidney (in urine)

via liver (in bile)

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

what are some other routes of excretion (excluding the kidney and liver)?

A

via lungs

in breast milk

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

what are the 3 major routes for drug excretion via the kidney?

A

glomerular filtration

active tubular secretion (or reabsorption)

passive diffusion across tubular epithelium

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

why does excretion of different drugs vary so much?

A

extent to which drug use each of the 3 major routes for drug excretion in the kidney varies between drugs

also impacted by the rate of metabolism

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

why does urine pH affect the process of excretion by passive diffusion in the kidney?

A

urine pH varies from 4.5 to 8

acidic drugs are better reabsorbed at lower pH

basic drugs are better reabsorbed at higher pH

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

why is active tubular secretion the most important method of excretion via kidney?

A

only 20% of renal plasma is filtered at the glomerulus, the rest passes onto the blood supply to proximal tubule - more drug delivered to proximal tubule than glomerulus

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

what is the process of excretion by active tubular secretion in the kidney?

A

depends on transporters

2 active transport carrier systems within the proximal tubule capillary endothelial cells (one for acidic drugs, one for basic drugs)

both systems can transport drugs against a concentration gradient

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

why does drug metabolism affect the process of excretion by passive diffusion in the kidney?

A

phase 2 metabolites tend to be more water soluble than the parent drug

therefore less well reabsorbed

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

why does urine pH affect the process of excretion by passive diffusion in the kidney?

A

urine pH varies from 4.5 to 8

acidic drugs are better reabsorbed at lower pH

basic drugs are better reabsorbed at higher pH

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

what is the process of excretion by passive diffusion in the kidney?

A

leads to reabsorption from kidney tubule

as glomerular filtrate moves through the kidney most of the water filtered is reabsorbed

if drugs are more lipid soluble they will also be reabsorbed, diffusing across the tubule back into the blood

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

how are drugs excreted via the bile?

A

hepatocytes transport some drugs from plasma to bile
- mostly via transporters (similar to those in the kidney)

very effective at removing phase 2 glucuronide metabolites

drugs transported to bile are excreted into intestines, eliminated in faeces

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

how can enterohepatic recycling prolong drug effect?

A

glucuronide metabolite is transported into bile

metabolite is excreted into the small intestine and is hydrolysed by gut bacteria - releases glucuronide conjugate

loss of glucuronide conjugate increases molecule’s lipid solubility

increased lipid solubility allows greater reabsorption from small intestine back into the hepatic portal blood system for return to liver

molecule returns to the liver - a proportion will be re-metabolised, but a proportion may escape into the systemic circulation to continue to have effects on the body

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

what is the primary mechanism of action of metformin?

A

primary effect:
metformin activates AMPK in hepatocyte mitochondria

inhibits ATP production

blocks gluconeogenesis and subsequent glucose output

secondary effect:
blocks adenylate cyclase which promotes fat oxidation

both help to restore insulin sensitivity

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

what is the drug target of metformin?

A

5′-AMP-activated protein kinase (AMPK) (enzyme)

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

what is the primary site of action of metformin?

A

hepatocyte mitochondria

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

what are the side effects of metformin?

A

GI side effects (20-30% of patients)

  • abdominal pain
  • decreased appetite
  • diarrhoea
  • vomiting

particularly evident when very high doses are given - slow increase in dose may improve tolerability

high doses may produce high lactic acid levels

(low risk) low blood glucose

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

why may metformin accumulate the in the liver and the GI tract to produce therapeutic and side effects respectively?

A

highly polar structure, requires organic cation transporter-1 (OCT-1) to access tissues

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

when is metformin most effective?

A

in the presence of endogenous insulin

i.e. with some residual functioning pancreatic islet cells

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

what is an example of a DPP-4 inhibitor?

A

sitagliptin

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

what is the primary mechanism of action of DPP-4 inhibitors?

A

primary effect:
inhibiting DPP-4 action
(enzyme is present in vascular endothelium and can metabolise incretins in plasma)

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

what are the functions of incretins (e.g. GLP-1)?

A

secreted by enteroendocrine cells

help stimulate the production of insulin when it is needed (e.g. after eating)

reduce the production of glucagon by the liver when it is not needed (e.g. during digestion)

slow down digestion and decrease appetite

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

what is the drug target of DPP-4 inhibitors?

A

DPP-4 (enzyme)

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

what is the primary site of action of DPP-4 inhibitors?

A

vascular endothelium

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

what are the side effects of DPP-4 inhibitors?

A

upper respiratory tract infections (5% of patients)

flu-like symptoms (e.g. headache, runny nose, sore throat)

serious allergic reactions (skin rash, increased upper respiratory tract infections)/ avoid in patients with pancreatitis (less common)



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

what is a benefit of DPP-4 inhibitors as opposed to other anti-diabetic drugs?

A

do not cause weight gain

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

what is required for DPP-4 inhibitor effectiveness?

A

some residual pancreatic beta-cell activity

since they act mainly by augmenting insulin secretion

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

what is an example of a sulphonylurea?

A

gliclazide

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

what is the primary mechanism of action of sulphonylureas?

A

inhibit ATP-sensitive potassium (KATP) channel on the pancreatic beta cell (controls beta cell membrane potential)

inhibition causes depolarisation

depolarisation stimulates Ca2+ influx and subsequent insulin vesicle exocytosis

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

what is the drug target of sulphonylureas?

A

ATP-sensitive potassium channel (ion channel)

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

what is the primary site of action of sulphonylureas?

A

pancreatic beta cell



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

what are the side effects of sulphonylureas?

A

weight gain (likely)

hypoglycaemia (2nd most common)

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

what is required for sulphonylurea effectiveness?

A

some residual pancreatic beta-cell activity

since they act mainly by augmenting insulin secretion

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
130
Q

how is the weight gain caused by sulphonylureas mitigated?

A

concurrent administration with metformin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
131
Q

when especially should the risk of hypoglycaemia associated with sulphonylureas be discussed with the patient?

A

when concomitant glucose-lowering drugs are prescribed

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

what is an example of a sodium-glucose co-transporter (SGLT2) inhibitor?

A

dapaglifozin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
133
Q

what is the primary mechanism of action of SGLT2 inhibitors?

A

reversibly inhibits sodium-glucose co-transporter 2 (SGLT2) in the renal proximal convoluted tubule

reduces glucose reabsorption, increases urinary glucose excretion

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
134
Q

what is the drug target of SGLT2 inhibitors?

A

SGLT2 (transport protein)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

what is the primary site of action of SGLT2 inhibitors?

A

proximal convoluted tubule

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
136
Q

what are the side effects of SGLT2 inhibitors?

A

uro-genital infections due to increased glucose load (5% of patients)

slight decrease in bone formation

can worsen diabetic ketoacidosis (stop immediately)



How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

what are some added benefits of SGLT2 inhibitors?

A

weight loss

reduction in BP

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

what is required for SGLT2 inhibitors effectiveness?

A

depends on normal renal function

less effective in patients with renal impairment

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
139
Q

what drugs can be used in the treatment of diabetes?

A

metformin

DPP-4 inhibitors

SGLT2 inhibitors

sulphonylureas

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
140
Q

what drugs can be used in the treatment of epilepsy?

A

lamotrigine

sodium valproate

diazepam

levetirecetam

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

what is the primary mechanism of action of lamotrigine?

A

blocks voltage gated Na+ channels, preventing Na+ influx

prevents depolarisation of glutamatergic neurones and reduces glutamate excitotoxicity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
142
Q

what is the drug target of lamotrigine?

A

voltage gated Na+ channels

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

what are the main (common) side effects of lamotrigine?

A

rash

drowsiness

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
144
Q

what are the main (more uncommon but serious) side effects of lamotrigine?

A

Steven-Johnson’s syndrome

suicidal thoughts

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
145
Q

how is lamotrigine used in treatment of allergic reactions?

A

lamotrigine introduced gradually - important in reducing frequency and severity of allergic skin reactions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
146
Q

what is the primary mechanism of action of sodium valproate?

A

inhibits GABA transaminase (prevents GABA breakdown)

directly increases GABA concentrations pre-synaptically in the synapse

indirectly prolongs GABA in the synapse (extraneuronal metabolism of GABA is slowed, slows GABA removal from synapse)

increased GABA availability

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
147
Q

what is the drug target of sodium valproate?

A

GABA transaminase (inhibitory presynaptic terminal)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
148
Q

what are the main (common) side effects of sodium valproate?

A

stomach pain

diarrhoea

drowsiness

weight gain

hair loss

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
149
Q

what are the main (serious) side effects of sodium valproate?

A

hepatotoxicity

teratogenicity

pancreatitis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
150
Q

how does sodium valproate interact with other drugs?

A

broad CYP (cytochrome P450) enzyme inhibitor

increases serum concentration of many co-administered drugs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
151
Q

what is the primary mechanism of action of diazepam?

A

increases Cl- influx in response to GABA binding at GABA A receptor

increased Cl- influx associated with hyperpolarisation of excitatory neurones

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
152
Q

what is the drug target of diazepam?

A

benzodiazepine site on the GABA A receptor

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
153
Q

what are the main (common) side effects of diazepam?

A

drowsiness

respiratory depression (if IV or at high dose)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
154
Q

what are the main (more uncommon but serious) side effects of diazepam?

A

haemolytic anaemia

jaundice

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
155
Q

why is diazepam not used for long term suppression of seizures?

A

development of tolerance

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
156
Q

why is diazepam

a Schedule 4 controlled drug?

A

addiction prone individuals more likely to become dependent on diazepam

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
157
Q

what is the primary mechanism of action of levetiracetam?

A

inhibition of synaptic vesicle protein SV2A in excitatory presynaptic terminal, prevents vesicle exocytosis

reduction in glutamate secretion reduces glutamate excitotoxicity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
158
Q

why is levetiracetam favourable in terms of lack of drug–drug interactions?

A

metabolism of levetiracetam has no effect on cytochrome P450 enzyme system

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
159
Q

what drugs are used in treatment of depression?

A

sertraline

citalopram

fluoxetine

venlafaxine

mirtazapine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
160
Q

what is the primary mechanism of action of sertraline?

A

serotonin reuptake inhibition causes accumulation

serotonin in CNS aids regulation of mood, personality, and wakefulness

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
161
Q

what is the drug target of sertraline?

A

serotonin transporter

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
162
Q

what are the main side effects of sertraline?

A

GI effects (nausea, diarrhoea)

sexual dysfunction

anxiety

insomnia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
163
Q

how does sertraline interact with CYP2D6 at high doses (150mg)?

A

partial inhibition

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
164
Q

how does sertraline interact with dopamine transporters?

A

mild inhibition

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
165
Q

how is sertraline discontinued?

A

gradually decreased

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
166
Q

what is the primary mechanism of action of citalopram?

A

serotonin reuptake inhibition causes accumulation

serotonin in CNS aids regulation of mood, personality, and wakefulness

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
167
Q

what is the drug target of citalopram?

A

serotonin transporter

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
168
Q

what are the main side effects of citalopram?

A

GI effects (nausea, diarrhoea)

sexual dysfunction

anxiety

insomnia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
169
Q

how does citalopram interact with muscarinic and histamine (H1) receptors?

A

mild antagonism

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
170
Q

how is citalopram discontinued?

A

gradually decreased

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
171
Q

how is citalopram metabolised?

A

CYP2C19

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
172
Q

what is the primary mechanism of action of fluoxetine?

A

serotonin reuptake inhibition causes accumulation

serotonin in CNS aids regulation of mood, personality, and wakefulness

173
Q

what is the drug target of fluoxetine?

A

serotonin transporter

174
Q

what are the main side effects of fluoxetine?

A

GI effects (nausea, diarrhoea)

sexual dysfunction

anxiety

insomnia

175
Q

which receptors does fluoxetine antagonise?

A

mild antagonism of 5HT2A and 5HT2C receptors

176
Q

which receptors does fluoxetine inhibit?

A

complete inhibition of CYP2D6

significant inhibition of CYP2C19

177
Q

what does inhibition of CYP2C19 and CYP2D6 by fluoxetine contraindicate?

A

caution with warfarin

178
Q

what is the primary mechanism of action of venlafaxine?

A

potent inhibitor of serotonin reuptake than norepinephrine reuptake

(noradrenaline in CNS implicated in emotional regulation and cognition)

179
Q

what are the drug targets of venlafaxine?

A

serotonin transporter

noradrenaline transporter

180
Q

what are the main side effects of venlafaxine?

A

GI effects (nausea, diarrhoea)

sexual dysfunction

anxiety

insomnia

(at higher doses) hypertension

181
Q

how is venlafaxine discontinued?

A

gradually decreased

182
Q

what is the primary mechanism of action of mirtazapine?

A

antagonises central presynaptic alpha-2-adrenergic receptors - causes increased serotonin and norepinephrine release

antagonises central 5HT2 receptors - leaves 5HT1 receptors unopposed, causing anti-depressant effects

183
Q

what are the drug targets of mirtazapine?

A

alpha-2 receptor

5-HT2 receptor

(high affinity for H1 receptor causes sedation, low affinity for 5-HT3 receptor has anti-emetic effect)

184
Q

what are the main side effects of mirtazapine?

A

weight gain

sedation

(low probability of sexual dysfunction)

185
Q

how does mirtazapine affect REM sleep behaviour disorder?

A

causes exacerbation, suppresses REM sleep

beneficial impact on sleep continuity and duration

(sedation due to histamine H1 receptor blocking)

186
Q

what drugs can be used in the treatment of hypertension?

A

angiotensin converting enzyme (ACE) inhibitors

calcium channel blockers

thiazide or thiazide-like diuretics

angiotensin receptor blockers

187
Q

what are some examples of angiotensin converting enzyme (ACE) inhibitors?

A

ramipril (pro-drug)

lisinopril

perindopril

188
Q

what is the primary mechanism of action of angiotensin converting enzyme (ACE) inhibitors?

A

inhibit angiotensin converting enzyme

prevent conversion of angiotensin I to angiotensin II

189
Q

what is the drug target of angiotensin converting enzyme (ACE) inhibitors?

A

angiotensin converting enzyme (ACE)

190
Q

what are the main side effects of angiotensin converting enzyme (ACE) inhibitors?

A

cough

hypotension

hyperkalaemia (care with K+ supplements or K+-sparing diuretics)

foetal injury (avoid in pregnant women)

renal failure (in patients with renal artery stenosis)

urticaria/angioedema

191
Q

what must be monitored regularly when taking angiotensin converting enzyme (ACE) inhibitors?

A

eGFR

serum potassium

192
Q

what kind of drugs are most angiotensin converting enzyme (ACE) inhibitors (not lisinopril)?

A

pro-drugs

require hepatic activation to generate active metabolites required for therapeutic effects

193
Q

what are some examples of calcium channel blockers?

A

amlodipine

felodipine

194
Q

what is the primary mechanism of action of calcium channel blockers?

A

block L-type calcium channels (predominantly on vascular smooth muscle)

causes decreased in calcium influx, with downstream inhibition of myosin light chain kinase and prevention of cross-bridge formation (i.e. muscular contraction)

resultant vasodilation reduces peripheral resistance

reduced cardiac contraction and cardiac output

195
Q

what is the drug target of calcium channel blockers?

A

L-type calcium channel

196
Q

what are the main side effects of calcium channel blockers?

A

ankle oedema

constipation

palpitations

flushing/headaches

197
Q

what kind of calcium channel blockers demonstrate a higher degree of vascular selectivity?

A

dihydropyridine type

198
Q

what are some examples of thiazide or thiazide-like diuretics?

A

bendro-flumethiazide (thiazide)

indapamide (thiazide-like diuretic)

199
Q

what is the primary mechanism of action of thiazide or thiazide-like diuretics?

A

block Na+, Cl- co-transporter in early apical DCT

Na+ and Cl- reabsorption is inhibited

osmolarity of tubular fluid increases, decreasing the osmotic gradient for water reabsorption in the collecting duct

(decreased blood volume, decreased venous return, decreased cardiac output)

200
Q

what is the drug target of thiazide or thiazide-like diuretics?

A

sodium/chloride cotransporter

201
Q

what are the main side effects of thiazide or thiazide-like diuretics?

A

hypokalaemia

hyponatremia

metabolic alkalosis (increased hydrogen ion excretion)

hypercalcemia

hyperglycaemia (hyperpolarised pancreatic beta cells)

hyperuricemia

202
Q

how long do the diuretic effects of thiazide and thiazide-like diuretics last?

A

lose diuretic effects within 1-2 weeks of treatment

203
Q

thiazide and thiazide-like diuretics lose their diuretic effect within 1-2 weeks - what allows continuing anti-hypertensive action?

A

vasodilating properties (more pronounced for thiazide-like diuretics)

204
Q

what are some examples of angiotensin receptor blockers?

A

losartan

irbesartan

candesartan

205
Q

what is the primary mechanism of action of angiotensin receptor blockers?

A

these agents act as insurmountable (i.e. non-competitive) antagonists at AT1 receptor (found on kidneys and on the vasculature)

206
Q

what is the drug target of angiotensin receptor blockers?

A

angiotensin receptor

207
Q

what are the main side effects of angiotensin receptor blockers?

A

hypotension

hyperkalaemia (care with K+ supplements or K+-sparing diuretics)

foetal injury (avoid in pregnant women)

renal failure (in patients with renal artery stenosis)

208
Q

how does the efficacy of angiotensin receptor blockers compare with angiotensin converting enzyme (ACE) inhibitors?

A

angiotensin receptor blockers are not as effective anti-hypertensive agents as ACE inhibitors

ACE inhibitors are cheaper

209
Q

what kind of drugs are losartan and candesartan (angiotensin receptor blockers)?

A

pro-drugs

require hepatic activation to generate active metabolites required for therapeutic effects

210
Q

what drugs can be used in the treatment of asthma?

A

salbutamol

fluticasone

mometasone

budesonide

montelukast

211
Q

what is the primary mechanism of action of salbutamol?

A

β2 receptor agonist on airway smooth muscle cells

activation reduces Ca2+ entry - prevents smooth muscle contraction, prevents constriction

(adrenaline binds to β2 adrenergic receptor to cause bronchodilation)

212
Q

what is the drug target of salbutamol?

A

beta 2 (β2) adrenergic receptor

213
Q

what are the main side effects of salbutamol?

A

palpitations/agitation

tachycardia/arrhythmias

hypokalaemia (at higher doses)

214
Q

what is salbutamol an example of?

A

short acting beta agonist (SABA)

215
Q

what is the half life of salbutamol?

A

2.5 - 5 hrs

216
Q

how does salbutamol produce cardiac side effects?

A

beta 2 selectivity is not absolute

beta 1 is also activated (causing cardiac side effects)

217
Q

how does salbutamol produce hypokalaemia as a side effect?

A

via effects on sodium/potassium ATPase

218
Q

how can hypokalaemia as a result of salbutamol administration be exacerbated?

A

co-administration with corticosteroids

219
Q

what are the primary mechanisms of action of fluticasone?

A

(multiple actions on many different cell types)

directly decreases number of inflammatory cells (eosinophils, monocytes, mast cells, macrophages, dendritic cells etc.) and number of cytokines they produce

affects IL-5 to prevent growth, differentiation, and activation of eosinophils

220
Q

what is the drug target of fluticasone?

A

glucocorticoid receptor

221
Q

what are the main local side effects of fluticasone?

A

sore throat

hoarse voice

opportunistic oral infections

222
Q

what are the main systemic side effects of fluticasone?

A

growth retardation in children

hyperglycaemia

decreased bone mineral density

immunosuppression

effects on mood

(etc.)

223
Q

how does fluticasone’s affinity for the glucocorticoid receptor compare to that of cortisol?

A

greater affinity

224
Q

why is any systemic delivery of fluticasone via the inhaled route predominantly through the pulmonary vasculature?

A

oral bioavailability <1%

225
Q

what are the primary mechanisms of action of mometasone?

A

(multiple actions on many different cell types)

directly decreases number of inflammatory cells (eosinophils, monocytes, mast cells, macrophages, dendritic cells etc.) and number of cytokines they produce

226
Q

what are the main local side effects of mometasone?

A

sore throat

hoarse voice

opportunistic oral infections

227
Q

what are the main systemic side effects of mometasone?

A

growth retardation in children

hyperglycaemia

decreased bone mineral density

immunosuppression

effects on mood

(etc.)

228
Q

how does mometasone’s affinity for the glucocorticoid receptor compare to that of cortisol?

A

greater affinity

229
Q

why is any systemic delivery of mometasone via the inhaled route predominantly through the pulmonary vasculature?

A

oral bioavailability <1%

230
Q

what are the primary mechanisms of action of budesonide?

A

(multiple actions on many different cell types)

directly decreases number of inflammatory cells (eosinophils, monocytes, mast cells, macrophages, dendritic cells etc.) and number of cytokines they produce

231
Q

what is the drug target of mometasone?

A

glucocorticoid receptor

232
Q

what is the drug target of budesonide?

A

glucocorticoid receptor

233
Q

what are the main local side effects of budesonide?

A

sore throat

hoarse voice

opportunistic oral infections

234
Q

what are the main systemic side effects of budesonide?

A

growth retardation in children

hyperglycaemia

decreased bone mineral density

immunosuppression

effects on mood

(etc.)

235
Q

how does the potency of budesonide compare to that of fluticasone and mometasone?

A

less potent

236
Q

why does inhaled budesonide result in some systemic absorption through the GI tract?

A

oral bioavailability >10%

237
Q

what is the primary mechanism of action of montelukast?

A

antagonises CysLT1 leukotriene receptor on eosinophils, mast cells and airway smooth muscle cells

decreases eosinophil migration, bronchoconstriction, inflammation induced oedema

238
Q

what is the drug target of montelukast?

A

CysLT1 leukotriene receptor

239
Q

what are the main mild side effects of montelukast?

A

diarrhoea

fever

headaches

nausea or vomiting

240
Q

what are the main serious side effects of montelukast?

A

mood changes

anaphylaxis

241
Q

when should montelukast be administered for prophylaxis of exercise-induced bronchoconstriction?

A

at least 2 hours before initiating exercise

242
Q

what drugs are used in treatment of GORD/peptic ulcer disease?

A

NSAIDS

PPIs

histamine (H2) receptor antagonists

paracetamol (aka acetaminophen)

243
Q

what are some examples of NSAIDS?

A

ibuprofen

naproxen

diclofenac

244
Q

what is the primary mechanism of action of NSAIDS?

A

inhibit cyclo-oxygenase enzyme (COX) - rate limiting step for production of all prostanoids (prostaglandins, thromboxanes) from parent arachidonic acid

prostanoids act through many prostanoid receptors, produces highly complex array of actions

245
Q

how do the effects of NSAIDS vary depending on the type of COX inhibition?

A

anti-inflammatory (and most analgesic and antipyretic actions) related to COX-2 inhibition

unwanted effects mostly a result of COX-1 inhibition

246
Q

what is the drug target of NSAIDS?

A

cyclo-oxygenase (COX) enzyme

247
Q

what are the common side effects of NSAIDS?

A

gastric irritation

ulceration

bleeding

248
Q

what are the more extreme side effects of NSAIDS?

A

perforation

reduced creatinine clearance, possible nephritis

bronchoconstriction in susceptible individuals (contraindicated in asthma)

skin rashes & other allergies, dizziness, tinnitus.

249
Q

what are the side effects of NSAIDS following prolonged used/in patients with pre-existing cardiovascular risk?

A

adverse cardiovascular effects (hypertension, stroke, MI)

250
Q

what is prolonged analgesic abuse over a period of years associated with?

A

chronic renal failure

251
Q

what rare condition in children has aspirin been linked with?

A

post-viral encephalitis (Reye’s syndrome)

252
Q

how can NSAIDS be used?

A

analgesics - mild to moderate pain relief (e.g. musculoskeletal pain, headache, dysmenorrhoea)

antipyretics - reduce fever

anti-inflammatory drugs - chronic control of inflammatory diseases (e.g. rheumatoid arthritis, osteoarthritis)

anti-aggregatory agent (aspirin only) - inhibit platelet aggregation in patients at risk of stroke or MI

253
Q

what are some examples of PPIs?

A

omeprazole

lansoprazole

254
Q

what is the primary mechanism of action of PPIs?

A

irreversible inhibitors of H+/K+ ATPase in gastric parietal cells

weak bases, so accumulate in acid environment of parietal cell canaliculi (concentrates and prolongs duration of action)

(omeprazole plasma half-life approx. 1 h but single daily dose affects acid secretion for 2-3 days)

PPIs inhibit basal and stimulated gastric acid secretion by >90%

255
Q

what is the drug target of PPIs?

A

H+/K+ ATPase (‘proton pump’)

256
Q

what are the main side effects of PPIs?

A

headache

diarrhoea

bloating

abdominal pain

rashes

257
Q

symptoms of which condition may be masked by use of PPIs?

A

gastric cancer

258
Q

omeprazole inhibits cytochrome P2C19 - what other drugs are affected by this?

A

reduced function in antiplatelet medication e.g. clopidogrel, when platelet function is monitored

259
Q

how are PPIs administered?

A

generally given orally

degrade rapidly at low pH so administered as capsules containing enteric-coated granules

260
Q

how do PPIs affect H+ ion transport in parietal cells?

A

PPIs are pro-drugs - converted into 2 reactive species at low pH

react with sulphydryl groups in H+/K+ ATPase responsible for transporting H+ ions out of the parietal cells

261
Q

what are some examples of histamine (H2) antagonists?

A

ranitidine

cimetidine

262
Q

what is the primary mechanism of action of histamine (H2) antagonists?

A

structural analogues of histamine are competitive antagonists of H2 histamine receptors

inhibit stimulatory action of histamine released from enterochromaffin-like (ECL) cells on gastric parietal cells surface

inhibit gastric acid secretion
(histamine receptors increase acid production via cAMP dependent activation of H+/K+ ATPase - less acid production means less corrosive environment, less irritation of stomach wall due to damaged mucosal barrier)

263
Q

what is the drug target of histamine (H2) antagonists?

A

histamine H2 receptors

264
Q

what are the main side effects of histamine (H2) antagonists?

A

diarrhoea

dizziness

muscle pains

transient rashes

265
Q

why is cimetidine (histamine H2 antagonist) contraindicated with some drugs?

A

inhibits cytochrome P450

may retard the metabolism and potentiate the effects of a range of drugs

266
Q

what are some drug categories contraindicated by cimetidine?

A

various - includes oral anticoagulants and TCAs

267
Q

why does ranitidine only have 50% bioavailability?

A

undergoes first pass metabolism

268
Q

why is twice daily dosing of ranitidine effective?

A

ranitidine plasma half-life 2-3 h - well tolerated

269
Q

when do histamine (H2) antagonists not necessarily need to be on prescription?

A

low dose over-the-counter formulations available from pharmacies for short term use without prescription

270
Q

what is the primary mechanism of action of paracetamol in GORD?

A

mechanism of action unclear

(current hypothesis)
both central and peripheral actions possibly involving interactions with a COX-3 isoform (inhibition of prostaglandin synthesis), cannabinoid receptors or the endogenous opioids

interactions at 5HT or adenosine receptors also proposed

271
Q

where are the effects of paracetamol mostly restricted to?

A

nervous tissue

272
Q

what is the drug target of paracetamol?

A

not yet well defined - COX-3 isoform?

273
Q

what are some side effects of paracetamol?

A

few side effects at therapeutic doses

no gastric irritation but in overdose serious hepatotoxicity may occur

occasional allergic skin reactions

274
Q

why is paracetamol not an NSAID?

A

little anti-inflammatory activity

275
Q

what are the broad therapeutic effects of paracetamol?

A

good analgesic for mild-to-moderate pain

some antipyretic activity

276
Q

what is step 1 of the 7 step process of prescribing?

A

identify the patient’s problem

277
Q

what is step 2 of the 7 step process of prescribing?

A

specify the therapeutic objective

278
Q

what is step 3 of the 7 step process of prescribing?

A

select a drug on the basis of comparative efficacy, safety, cost and suitability

279
Q

what is step 4 of the 7 step process of prescribing?

A

discuss choice of medication with patient (and carer) and make a shared decision about treatment

280
Q

what is step 5 of the 7 step process of prescribing?

A

write a correct prescription

281
Q

what is step 6 of the 7 step process of prescribing?

A

counsel the patient on appropriate use of the medicine

282
Q

what is step 7 of the 7 step process of prescribing?

A

make appropriate arrangements for follow up

283
Q

BMI: 31

1st BP: 144/92 mmHg
2nd BP: 148/91 mmHg

HbA1c: 65mmol/mol

LDL: 5.18 mmol/L (200 mg/dL)

HDL: 0.8 mmol/L (30mg/dL)

triglycerides: 6.53 mmol/L (252 mg/dL)

glycosuria, no ketones

no polydipsia, polyuria or weight loss

what is the patient’s problem?

A

fasting glucose > 7
HbA1c > 48mmol/L (6.5%)
- diabetes

blood pressure high
- hypertensive

HDL < 50mg/dl
LDL > 100mg/dl, triglycerides > 150mg/dl
- dyslipidaemia

BMI 30-34.9
- obesity

overall, metabolic syndrome?

284
Q

fasting glucose > 7
HbA1c > 48mmol/L (6.5%)
- diabetes

blood pressure high
- hypertensive

HDL < 50mg/dl
LDL > 100mg/dl, triglycerides > 150mg/dl
- dyslipidaemia

BMI 30-34.9
- obesity

overall, metabolic syndrome?

what are the therapeutic objectives for this patient?

A

lose weight – reduced adiposity should help reduce insulin resistance linked to diabetes

reduce blood pressure – reduce CVD risk

improve lipid profile – reduce CVD risk

reduce blood glucose – reduce complications (microvascular and (atherosclerotic) macrovascular) associated with prolonged elevated blood glucose

285
Q

what should be done when HbA1c rises to 48 mmol/mol (6.5%) on lifestyle intervention? (adult with T2DM, can take metformin)

A

offer standard release metformin (500 mg per day, oral)

support person to aim for 48 mmol/mol

286
Q

what is the first intensification (when HbA1c rises to 58 mmol/mol (7.5%)) for adults with T2DM that can take metformin?

A

consider dual therapy with:

  • DPP-4 inhibitors e.g. sitagliptin (100 mg per day, oral)
  • SGLT2 inhibitors e.g. dapaglifozin (5 mg per day, oral)
  • sulphonylureas e.g. gliclazide (40-80 mg per day)
  • pioglitazone (15-45 mg per day)

support person to aim for 53 mmol/mol (7.0%)

287
Q

what is the second intensification (when HbA1c rises to 58 mmol/mol (7.5%)) for adults with T2DM that can take metformin?

A

consider triple therapy with:

  • metformin, a DPP-4i and an SU
  • metformin, pioglitazone and an SU - metformin, pioglitazone or an SU, and an SGLT-2i

or insulin-based treatment

support person to aim for 53 mmol/mol (7.0%)

288
Q

what are the benefits of metformin treatment?

A

decreases glucose production by liver

increases insulin sensitivity of body tissues

anorexiant effect (appetite suppression, thereby reducing caloric intake)

289
Q

expression of the organic cation transporter 1 (OCT-1) is highest in: liver hepatocytes (highest expression), small intestinal enterocytes and the renal proximal tubules.

why is this relevant to the pharmacokinetics of orally-administered metformin?

A

relation to absorption
- shows that metformin has a mechanism to leave small intestines and access the hepatic portal vein

relation to distribution

  • shows that metformin has a mechanism to access the liver
  • shows that metformin can access the kidney tubules
290
Q

what is a side effect of pioglitazone?

A

increased incidence of heart failure

291
Q

why may renal impairment cause problems for diabetic patients on metformin?

A

90% of standard dose (‘unchanged’,
active) of oral metformin excreted
via kidney

292
Q

how should metformin administration be adjusted with an eGFR of >60?

A

no adjustment needed

293
Q

how should metformin administration be adjusted with an eGFR of 45-60?

A

monitor eGFR in 3-6 months

294
Q

how should metformin administration be adjusted with an eGFR of 30-45?

A

do not initiate metformin

if already taking metformin, consider 50% dose decrease

295
Q

how should metformin administration be adjusted with an eGFR of <30?

A

contraindicated

296
Q

how is depression screened for in primary care?

A

patient health questionnaire 9 (PHQ 9)

297
Q

how is a PHQ-9 score related to depression severity?

A

0-4 - none/minimal

5-9 - mild

10-14 - moderate

15-19 - moderately severe

20-27 - severe

298
Q

hypertension - losartan (angiotensin 2 receptor blocker) but BP still high (141/91 mmHg)

erythromycin for chronic prostatitis

uneasy

low mood

low self esteem

difficulty getting to sleep

inability to think clearly

strained personal relationship

job performance affected?

PHQ-9 score - 14

what is the patient’s problem?

A

indicative of moderate depression

major depressive disorder includes minor, moderate and severe depression

299
Q

hypertension - losartan (angiotensin 2 receptor blocker) but BP still high (141/91 mmHg)

erythromycin for chronic prostatitis

uneasy

low mood

low self esteem

difficulty getting to sleep

inability to think clearly

strained personal relationship

job performance affected?

PHQ-9 score - 14

likely moderate depression

what are the therapeutic objectives?

A

alleviate depressive symptoms:

  • improve mood
  • help with sleep difficulties
  • improve self-esteem
  • improve ability to think clearly

reduce the likely functional impairment the depressive symptoms have on daily life:

  • improve personal relationships
  • improve job performance
300
Q

the 3 most common SSRIs are sertraline, citalopram and fluoxetine - which is contraindicated with erythromycin?

A

citalopram

301
Q

why is citalopram contraindicated with erythromycin?

A

both erythromycin and citalopram prolong QT interval - most manufacturers advise avoiding 2+ drugs that do this

severe interaction

302
Q

what may be seen on an ECG if polymorphic ventricular tachycardia is occurring in the context of QT prolongation (e.g. interaction of citalopram and erythromycin)?

A

torsades de pointes

303
Q

what are some factors that predispose to QT prolongation?

A

increasing age

female sex

disease

metabolic disturbance (notably hypokalaemia)

304
Q

what is the optimal SSRI dose (normalised to fluoxetine) to cause 50% or greater reduction in depression rating after 8 weeks of treatment?

A

30 mg

305
Q

what is the link between dropouts due to adverse effects and 50% or greater reduction in depression rating after 8 weeks of treatment?

A

as SSRI dose increases, dropouts due to adverse effects increases

reduction in depression rating begins at 1.0 at 0 mg, peaks at 30 mg, returns to 1.0 by 80 mg

20-30 mg is optimal dose to manage adverse effects

306
Q

why does the plateau in therapeutic effect occur in anti-depressant action (based on action at serotonin transporter)?

A

limited receptor numbers

307
Q

why is caution required when switching from one antidepressant to another?

A

risk of drug interactions

risk of serotonin syndrome

withdrawal symptoms

relapse

308
Q

what is required before starting a new drug?

A

washout

high starting doses

309
Q

why may venlafaxine be preferred to mirtazapine in a patient with hypertension?

A

noradrenaline mediates the sympathetic nervous system effects on the heart

> 150 mg per day - adrenergic effects

> 300 mg per day - increased BP, increased HR

310
Q

rheumatoid arthritis for 9 years – awaiting total right knee arthroplasty

BP

  • initial: 149/93mmHg
  • subsequent: 147/92mmHg and 145/91mmHg

generally well

smokes (< 10 per day)

height: 167 cm
weight: 85kg

what is the patient’s problem? (check Q-risk score)

A

Q-risk = 14.9%

stage 1 hypertension

311
Q

Q-risk = 14.9%

stage 1 hypertension

what are the therapeutic objectives for this patient?

A

set reasonable blood pressure reduction goals (previous case reduce below 140/90 mmHg)

reduce cardiovascular risk associated with increased morbidity and mortality (modifiable risk factors = obesity and smoking)

no new medication that interferes with upcoming procedures/other treatment

312
Q

what is the Q-risk score?

A

shows risk of heart attack or stroke within 10 years

e.g. score of 12% - in a population of 100 all of whom have the same risk factors, 12 are likely to die of heart attack/stroke within 10 years

313
Q

what is the procedure if ambulatory or home blood pressure monitoring (ABPM or HBPM) shows a BP under 135/85 mmHg?

A

monitor at least every 5 years

314
Q

what is the procedure if ambulatory or home blood pressure monitoring (ABPM or HBPM) shows a BP between 135/85 - 149/94 mmHg? (stage 1 hypertension)

A

start drug treatment if there is:

target organ damage

CVD

renal disease

diabetes

10 year CVD risk >10%

315
Q

what is the procedure if ambulatory or home blood pressure monitoring (ABPM or HBPM) shows a BP over 150/95 mmHg? (stage 2 hypertension)

A

start drug treatment

316
Q

when is ambulatory or home blood pressure monitoring (ABPM or HBPM) offered?

A

clinic BP 140/90 mmHg or over

317
Q

what is clearance?

A

measure of body’s ability to eliminate a drug

clearance by means of various organs of elimination is additive

elimination of drug may occur as a result of processes that occur in the liver, kidney, and other organs

318
Q

what is elimination half life?

A

length of time required for drug concentration to decreasetohalf of itsstarting dose in the body

319
Q

what is time to peak plasma levels?

A

time required for a drug to reach peak concentration in plasma

the faster the absorption rate, the lower the time to peak plasma concentration

320
Q

why may amlodipine be better than felodipine considering BP tends to increase most rapidly in the morning after waking up?

A

felodipine has higher plasma clearance, shorter elimination half life and shorter time to peak plasma levels

therefore causes dose dependent decrease in systolic and diastolic BP along with reflex tachycardia

amlodipine has slow onset and longer half life - slow onset mitigates reflex tachycardia

321
Q

what is the mechanism of action of ACE inhibitors in the treatment of hypertension?

A

inhibits angiotensin converting enzyme (ACE) on pulmonary and renal endothelium

prevents conversion of angiotensin I to angiotensin II

causes vasodilation, less salt and water retention, less aldosterone secretion

322
Q

why may ACE inhibitors have a negative effect on eGFR?

A

angiotensin II causes efferent vasoconstriction (increased pressure for filtration) - helps maintain GFR when renal perfusion is low (renal artery stenosis, volume depletion, older people with congestive heart failure)

323
Q

why may ACE inhibitors have a negative effect on serum potassium?

A

hyperkalaemia

usually aldosterone increases Na+ reabsorption and water in distal convoluted tubule at the expense of K+ (increasing plasma volume and blood pressure) A

increasing the gene expression and availability of:

  • Na+ ion permease enzyme - more Na+ crosses from the lumen to the inside of the renal tubular cell
  • Na+/K+ATPase on the peritubular side of the renal tubular cell - transfer increased cytosolic Na+ into peritubular fluid (so lower intracellular electronegativity)
  • citrate synthase - more activity within the mitochondria for more ATP (for Na+/K+ATPase)

less aldosterone from the adrenal cortex means overall reduction Na+ and water reabsorption, allows K+ retention

324
Q

thiazide-like diuretics are excreted unchanged in urine - why is this a vital part of their therapeutic action?

A

diuretic needs to move from the blood

it is transported on basolateral side, uses a transporter on apical side

only then can it access the sodium chloride transporter on the apical side of distal tubule

325
Q

why do thiazides increase potassium excretion?

A

increase delivery from early distal tubule

326
Q

why does the diuretic effect of thiazides only work for 1-2 weeks?

A

kidney becomes tolerant to the diuretics due to rebound activation of renin angiotensin system

consequent increasing sodium reabsorption counteracts diuretic effect

327
Q

why do the antihypertensive effects of thiazides continue while the diuretic effect of thiazides only works for 1-2 weeks?

A

further vasodilating action (not as well understood)

328
Q

how often should BP and medication be checked once stabilised?

A

6-12 months

329
Q

how often should serum potassium and creatinine be checked once BP is stabilised?

A

yearly

330
Q

3 yr old

great difficulty in breathing

whistle like wheeze when exhaling, occasional coughing

slight temperature (38.2°C)

history and physical examination reveal nothing

has had mild breathlessness before, usually when she has an infection of some type

what is the patient’s problem?

A

first presentation of asthma

no allergic trigger, and most likely precipitated by the viral infection

hard to objectively diagnose asthma in children < 5 years old

no evidence that this is atopic at the moment, based on history

331
Q

first presentation of asthma

no allergic trigger, and most likely precipitated by the viral infection

what are the therapeutic objectives for this patient?

A

short term = relief
- relieve symptoms of breathlessness and expiratory wheeze during the acute asthma attack

long term = prevention

  • dampen/prevent late phase of the asthma attack
  • reduce the risk of further asthma attacks.
  • attempt to improve lung function
332
Q

what is the difference between the early phase and late phase of asthma?

A

early phase - bronchospasm
- immediate decrease in forced expiratory volume

some recovery in between

late phase - inflammation (up to 6 hours after early phase)
- further decrease in forced expiratory volume

333
Q

what is the first line treatment for asthma in under 5s?

A

SABA (salbutamol)

334
Q

what are the differences between oral and inhalation routes when taking salbutamol?

A

inhaled has local effects

oral has systemic effects (undergoes first pass metabolism etc.)

335
Q

how does acetylcholine cause bronchoconstriction?

A

post ganglionic PSNS nerve releases acetylcholine

binds to M2 and M3 muscarinic receptor on bronchial smooth muscle

causes bronchoconstriction

336
Q

why is a nebulizer the best method for delivering salbutamol in emergency situations?

A

many drug solutions

can deliver combinations

minimal patient co operation required

can deliver to all ages

concentration and dose can be modified

normal breathing pattern

337
Q

what are the 5 places where are inhaled drugs lost?

A

1 - exhalation

2 - lack of absorption from lungs

3 - mucociliary clearance (trapped by cilia or mucous)

4 - swallowed (down oesophagus rather than trachea)

5 - some absorbed across mucous membrane in oral cavity and pharynx

338
Q

due to drug loss and consequent difference between expected and actual dose, why is a spacer clinically useful (especially in children)?

A

inhalation indicator makes it easier to coordinate breathing in and pressing your puffer

increased space allows better delivery to lungs

339
Q

why are eosinophils dangerous in asthma?

A

asthma can be exacerbated by viral infection

common viral infections (e.g. rhinovirus) release mediators that specifically activate eosinophils

eosinophils can induce epithelial damage (e.g. due to release of major basic protein)

increased susceptibility to viral infections (and therefore increased likelihood of asthma exacerbation)

340
Q

why is the oral bioavailability (proportion of drugs reaching plasma via GI tract) of some inhaled drugs (e.g. fluticasone) low despite a significant proportion being swallowed?

A

first pass inactivation

341
Q

what is the mechanism of action of montelukast?

A

targets CysLT1 receptor (Cysteinyl leukotriene receptor 1)

located on eosinophils/mast cells/airway smooth muscle

decreases smooth muscle constriction, eosinophil migration and oedema

342
Q

why is the mechanism of action of montelukast useful in NSAID induced asthma?

A

NSAIDs generally work by blocking the production of prostaglandins (both leukotrienes and prostaglandins derive from arachidonic acid)

if the leukotriene pathway is blocked, more prostaglandin will be formed to combat NSAID effect

343
Q

71-year-old man

osteoarthritis

  • prescribed diclofenac gel (1%)
  • regular paracetamol (500-1000mg orally every 6 hours when required) to help with the pain

bilateral knee pain diclofenac and paracetamol have reduced his pain, but he still has moderate pain when walking or carrying out strenuous activities such as gardening

pain is restricting his mobility and he has not been able to carry out the exercises that he has been advised to do

GP prescribes naproxen (250-500mg orally twice daily when required) as an additional analgesic

after taking the naproxen pain associated with his osteoarthritis is much improved

however, he has been suffering with intermittent upper abdominal pain for the last 2 weeks - described as a dull, gnawing ache, sometimes wakes him at night

what is the patient’s problem?

A

pain from osteoarthritis that limits daily activities

upper abdominal pain since starting naproxen

344
Q

pain from osteoarthritis that limits daily activities

upper abdominal pain since starting naproxen

what are the therapeutic objectives for this patient?

A

treat pain from osteoarthritis to ensure it is not interfering with daily living (or at least minimising the effect on activities)

treat new stomach pain, ideally without impacting on treatment of osteoarthritis pain

(these objectives are trade-offs)

345
Q

how does naproxen have an analgesic effect on joint pain?

A

target – COX enzymes (naproxen is non-selective i.e. inhibits COX1 & COX2)

location – peripheral nociceptive nerve endings (analgesia)

effect – COX produces prostaglandins that do not directly cause pain themselves, but sensitise peripheral nociceptors mediators (bradykinin and histamine) which causes pain (NSAIDs inhibit COX)

indirect effect on pain – PGs mediate inflammation therefore NSAIDs will reduce inflammation

346
Q

how can opiates reduce pain?

A

increased activity in descending inhibitory pathways

decreased excitation of transmission neuron

desensitise peripheral nociceptors

347
Q

how does naproxen have an adverse effect within the stomach?

A

target – COX I enzyme

location – gastric mucosal cells

effect – inhibited prostaglandin production means inhibition of prostaglandin mediated protection of gastric mucosa

348
Q

how do prostaglandins in gastric mucosal cells protect from acid?

A

increase bicarbonate release

increase mucous production

increase blood flow

349
Q

why would a patient be on oral naproxen and topical diclofenac? (topical drugs can cause systemic side effects)

A

GP mistake

communication failure

350
Q

what changes should be made to a prescription of naproxen and topical diclofenac?

A

3 options:
stop the gel

switch to ibuprofen

stop NSAIDs completely

351
Q

after an oesophago-gastro-duodenoscopy that leads to a diagnosis of peptic ulcer disase with no active bleeding and H. pylori negative, what treatment should be given?

A

stop NSAIDs where possible

offer full dose PPI therapy for 4-8 weeks
e.g. omeprazole 20 mg once daily

352
Q

why do PPIs help with peptic ulcer disease associated with NSAIDs?

A

NSAIDs leave stomach wall exposed to the effect of acid, which is causing pain

PPIs reduce the acid production

353
Q

what is the disconnect when comparing guidance for PPI treatment and what happens in practice?

A

treatment algorithm states that patients should be given 20mg omeprazole once a day for 4-8 weeks

data suggests that most patients prescribed PPIs are treated for at least 3 months (above the recommended treatment duration of 4-8 weeks) at twice the standard treatment dose i.e. 40mg versus 20mg.

nearly 50% are still on 40mg after 12 months

(data does include PPI use for indigestion or dyspepsia)

354
Q

when suffering with osteoporosis as well as peptic ulcer disease, why is a histamine H2 receptor antagonist prescribed (as opposed to PPIs)?

A

PPIs increase fracture risk (worsens chances if patient has osteoporosis)

(mechanism of action is unclear- main theory)
absorption of calcium salts is pH dependent

change in pH induced by PPIs causes reduction in absorption and decrease in calcium available for bone

355
Q

21 year old

single episode of collapse with jerking

lost consciousness, started convulsing before coming around

stressed and not much sleep

occasionally makes a quick jerk of her arms as she wakes up

similar episode 18 months ago

urea, electrolytes, calcium, glucose: normal

general and neurological exams: normal

EEG shows interictal epileptiform discharge (IED)

what is the patient’s problem?

A

generalised tonic-clonic seizures

EEG shows interictal epileptiform discharge (IED) - increased risk of seizure recurrence

356
Q

generalised tonic-clonic seizures

EEG shows interictal epileptiform discharge (IED) - increased risk of seizure recurrence

what are the therapeutic objectives for this patient?

A

eliminate seizures or reduce their frequency to the maximum degree possible

avoid the adverse effects associated with long-term treatment

aid patients in maintaining or restoring their usual psychosocial and vocational activities, and in maintaining a normal lifestyle

357
Q

how does epilepsy affect driving?

A

must document advice to contact DVLA that they cannot drive

358
Q

what are the different types of seizure and how are they differentiated?

A

absence

focal

generalised tonic-clonic

myoclonic

359
Q

what are the features of an absence seizure?

A

lose awareness of your surroundings for a short time

  • stare blankly into space
  • look like they’re “daydreaming”
  • flutter their eyes
  • make slight jerking movements of their body or limbs
360
Q

what are the features of a simple partial focal seizure?

A

strange feeling that’s hard to describe

“rising” feeling in your tummy – like the sensation in your stomach when on a fairground ride

a feeling that events have happened before (déjà vu)

unusual smells or tastes

tingling in your arms and legs

an intense feeling of fear or joy

stiffness or twitching in part of your body, such as an arm or hand

remain awake and aware

361
Q

how long does a typical absence seizure last?

A

up to 15 secs, will not remember

362
Q

what can simple partial seizures indicate?

A

sometimes referred to as warnings or auras - can be a sign that another type of seizure is about to happen

363
Q

what are the features of a complex partial focal seizure?

A

lose sense of awareness, make random body movements, such as:

  • smacking your lips
  • rubbing your hands
  • making random noises
  • moving your arms around
  • picking at clothes or fiddling with objects
  • chewing or swallowing

unresponsive to anyone, no memory of it

364
Q

what are the features of a tonic-clonic seizure?

A

2 stages

tonic stage – lose consciousness, body goes stiff, may fall to the floor

clonic stage – limbs jerk about, may lose control of bladder or bowel, may bite tongue or the inside of cheek, might have difficulty breathing

365
Q

how long does a typical tonic-clonic seizure last?

A

normally stops after a few minutes, some last longer

may have a headache, difficulty remembering what happened

may feel tired or confused afterwards

366
Q

what are the features of a myoclonic seizure?

A

some or all of your body suddenly twitches or jerks, like you’ve had an electric shock (often happen soon after waking up)

367
Q

how long does a typical myoclonic seizure last?

A

usually only a fraction of a second but many can occur in a short space of time

368
Q

what is the difference between a tonic and atonic seizure?

A

tonic - all your muscles suddenly become stiff (like the first stage of a tonic-clonic seizure), may lose balance and fall over

atonic - all muscles to suddenly relax, so you may fall to the ground - tend to be very brief, usually able to get up straight away

369
Q

what is the first line of treatment for a generalised tonic-clonic seizure in men and women with no child bearing potential?

A

sodium valproate

370
Q

what is the first line of treatment for a generalised tonic-clonic seizure in men and women with child bearing potential?

A

lamotrigine or carbamazepine

371
Q

what is the first line of treatment for an absence seizure in men and women with no child bearing potential?

A

ethosuximide or sodium valproate

372
Q

what is the first line of treatment for an absence seizure in men and women with child bearing potential?

A

ethosuximide

373
Q

what is the first line of treatment for a focal seizure in men and women with no child bearing potential?

A

lamotrigine or carbamazepine

374
Q

what is the first line of treatment for a focal seizure in men and women with child bearing potential?

A

carbamazepine or levetiracetam

375
Q

what is the first line of treatment for a myoclonic seizure in men and women with no child bearing potential?

A

sodium valproate

376
Q

what is the first line of treatment for a myoclonic seizure in men and women with child bearing potential?

A

levetiracetam or topiramate

377
Q

what is the first line of treatment for a tonic or atonic seizure in men and women with no child bearing potential?

A

sodium valproate

378
Q

what is the first line of treatment for a tonic or atonic seizure in men and women with child bearing potential?

A

sodium valproate (pregnancy prevention programme)

379
Q

why is there a difference in epilepsy treatment depending on childbearing potential?

A

sodium valproate: first line in absence, tonic-clonic, tonic/atonic, myoclonic

can cause:
- neural tube defects
- decreased IQ
- autism
after in utero exposure
380
Q

how does lamotrigine interact with the combined oral contraceptive pill?

A

drug-drug interaction

COC appears to have an impact on lamotrigine blood levels – coadministration leads to a reduced level of lamotrigine in the blood

381
Q

what are the effects of drug interactions between lamotrigine and the combined oral contraceptive pill?

A

reduced seizure control but no contraceptive failure

382
Q

how may the combined oral contraceptive decrease lamotrigine concentrations in the blood?

A

reduce lamotrigine absorption - less gets into the blood in the first place

enhance lamotrigine metabolism - more cleared from the blood

enhance lamotrigine excretion - enhance clearance from the blood

383
Q

how does lamotrigine affect blood ethinyl estradiol levels?

A

appears to have no effect

384
Q

what prescription change should be made to lamotrigine if taken with the combined oral contraceptive?

A

increase lamotrigine dose

385
Q

when on the combined oral contraceptive with lamotrigine, why may more seizures occur during the second and third week? why may someone feel drowsy in the fourth week?

A

oestrogen = “seizure promoting”
increased seizure frequency during oestrogen peak

COC taken over four weeks:

  • first three weeks: active drug
  • fourth week: nothing or ‘dummy’

fourth week - no COC affecting
liver enzymes (usually induces UDPGA):
- liver enzymes normalise
- lamotrigine levels increase

386
Q

how can side effects in the fourth week of a combined oral contraceptive cycle when taken with lamotrigine be addressed?

A

decrease lamotrigine in fourth week

387
Q

what are some side effects in the fourth week of a combined oral contraceptive cycle when taken with lamotrigine?

A

dizziness

drowsiness

diarrhoea

loss of balance

abnormal eye movement

trouble speaking

388
Q

between lorazepam, diazepam and midazolam, which is best to administer out of hospital?

A

midazolam

others require IV access

389
Q

how may pregnancy affect seizure frequency?

A

may increase

390
Q

why may pregnancy affect seizure frequency (if on lamotrigine)?

A

lamotrigine dose may be reduced due to fears of harming the unborn child

pregnancy may lead to changes in liver metabolism - lamotrigine levels may decrease (so increased seizure risk)

391
Q

how can increased seizure frequency be avoided in pregnancy (if on lamotrigine)?

A

check serum lamotrigine at the beginning of pregnancy and during the second and third trimester

dose could then be adjusted as described

392
Q

how can increased seizure frequency during week 2-3 of the combined oral contraceptive cycle be addressed if lamotrigine is well tolerated?

A

change oral contraceptive to long-term progesterone implant (e.g. Implanon/Nexplanon)

progesterone = “seizure inhibiting”

393
Q

what are the 2 ways in which variation in adverse effects of drugs can be considered?

A

differences in concentrations of drug reaching the tissues (i.e. factors affecting absorption, distribution, metabolism and excretion of the drug)

differences in response of the target tissues to the same degree of stimulation (i.e. factors such as variation in receptor sensitivity, number and distribution)

394
Q

responses to a drug vary between individuals - how can the reasons for this be subdivided?

A

absolute differences in dose administered

relative overdose or underdose

395
Q

how can an absolute difference in dose administered take place?

A

(deliberate or accidental)

error in prescription or dispensing

patient non-compliance

drug formulation

396
Q

what are the 5 factors that may cause a relative overdose or underdose of a drug to produce variable effects?

A

environmental exposure to chemicals, including other drugs

food intake

fluid intake

age

disease

397
Q

how can environmental exposure to chemicals, including other drugs, cause relative overdose or underdose of a drug to produce variable effects?

A

enzyme induction

enzyme inhibition

398
Q

how can food intake cause relative overdose or underdose of a drug to produce variable effects?

A

drugs may interact chemically with components of food; this may alter their absorption (e.g. bisphosphonates chelated by Ca, Mg, Fe so not absorbed with food)

foods delay gastric emptying and alter gastric pH

399
Q

how can fluid intake cause relative overdose or underdose of a drug to produce variable effects?

A

most drugs are better absorbed if taken with water e.g. may dissolve better

fluids may stimulate gastric emptying

400
Q

how may newborn infants’ bodies cause relative overdose or underdose of a drug to produce variable effects?

A

more body water than adults

poorer renal function, with immature tubular secretion

an immature blood brain barrier

lower capacity for drug metabolism

401
Q

how can changes in drug absorption in elderly people cause relative overdose or underdose of a drug to produce variable effects?

A

decreased absorptive surface of small intestine

altered gastric and gut motility

increased rate of gastric emptying

402
Q

how can changes in drug distribution in elderly people cause relative overdose or underdose of a drug to produce variable effects?

A

reduced lean body mass and body water, relative increase in fat

lipid soluble drugs have increased volume of distribution and decreased blood levels

water soluble drugs have decreased volume of distribution and increased blood levels

reduced plasma albumin, so fewer plasma protein binding sites

403
Q

how can changes in drug metabolism in elderly people cause relative overdose or underdose of a drug to produce variable effects?

A

splanchnic and hepatic blood flow decrease by 0.3 – 1.5%/year

liver size and hepatocyte number decrease

hepatic enzyme activity and induction capacity decrease

404
Q

how can changes in drug excretion in elderly people cause relative overdose or underdose of a drug to produce variable effects? (most important in drug handling in elderly patients)

A

reduced renal mass

reduced renal perfusion

reduced glomerular filtration rate

reduced tubular excretion

(steady decline is normal - situation worsened in renal disease)

405
Q

how can changes in organ sensitivity in elderly people cause relative overdose or underdose of a drug to produce variable effects?

A

elderly tend to be more sensitive to CNS active drugs

406
Q

what are the 5 aspects of ageing that affect relative overdose or underdose to produce variable effects?

A

drug absorption

drug distribution

drug metabolism

drug excretion

organ sensitivity

407
Q

what are the 6 aspects of disease that affect relative overdose or underdose to produce variable effects?

A

general nutritional status

GI disorders

congestive heart failure

kidney failure

liver failure

other acute or chronic disease states

408
Q

how can changes in general nutritional status (disease) cause relative overdose or underdose of a drug to produce variable effects?

A

unbalanced diets may lead to deficiency states and enzyme abnormalities

starvation – decreased plasma protein binding and metabolism

obesity – increased lipid fraction

409
Q

how can GI disorders (disease) cause relative overdose or underdose of a drug to produce variable effects?

A

e.g. achlorhydria, coeliac disease, Crohn’s disease

altered drug absorption

410
Q

how can congestive heart failure (disease) cause relative overdose or underdose of a drug to produce variable effects, especially in the elderly?

A

reduced splanchnic blood flow

intestinal mucosal oedema

reduced hepatic clearance

411
Q

how can kidney failure (disease) cause relative overdose or underdose of a drug to produce variable effects, especially in the elderly?

A

decreased drug excretion leading to toxicity

water overload leading to changes in drug concentrations in different body fluid compartments

412
Q

how can liver failure (disease) cause relative overdose or underdose of a drug to produce variable effects?

A

reduced metabolism

reduced first pass metabolism (hence increased bioavailability)

decreased biliary secretion and hence decreased removal

decreased albumin synthesis and hence reduced plasma protein binding

413
Q

how can other acute or chronic disease states (disease) cause relative overdose or underdose of a drug to produce variable effects?

A

varies

414
Q

72-year-old

atrial fibrillation which could not be converted back to sinus rhythm

suffered a small stroke, warfarin prescribed (‘thins’ blood) to prevent a recurrence

warfarin dose stabilized (checked by clotting time), but later develops a chest infection and is given clarithromycin for one week

effectively treats the infection, but now has a severe nosebleed, bruises very easily

how has this adverse drug reaction been caused (by what mechanism)?

A

increased external bleeding (nose bleed)

increased internal bleeding (bruising)

warfarin metabolised by cytochrome P450 pathway

Cyt P450 inhibited by clarithromycin, so increased plasma levels of warfarin

415
Q

72-year-old

atrial fibrillation which could not be converted back to sinus rhythm

suffered a small stroke, warfarin prescribed (‘thins’ blood) to prevent a recurrence

warfarin dose stabilized (checked by clotting time), but later develops a chest infection and is given clarithromycin for one week

effectively treats the infection, but now has a severe nosebleed, bruises very easily

what drugs might produce a similar effect to clarithromycin?

A

other antibiotics of same class (i.e. macrolides): e.g. erythromycin

some antibiotics of other classes: e.g. quinolones like ciprofloxacin

some systemic antifungal drugs: e.g. fluconazole

proton pump inhibitors (PPIs) - for peptic ulcers: e.g. omeprazole

some anti-HIV drugs, especially protease inhibitors

416
Q

what is INR?

A

International Normalized Ratio (INR) - standardized measure of blood clotting time

417
Q

how is INR calculated?

A

ratio of a patient’s prothrombin time (‘test’) to a control (‘normal’) sample, raised to the power of the ISI (International Sensitivity Index) value for the tissue factor used (usually 0.94 - 1.4)

(test PT / normal PT) ᶦˢᶦ

418
Q

what does a raised INR signify?

A

increased bleeding (decreased clotting)

bleeding risk increased

419
Q

what does a lowered INR signify?

A

less bleeding (increased clotting)

thrombosis risk increased

420
Q

what are the reference ranges for INR?

A

normal: 0.9-1.2

normal in atrial fibrillation: 2.0-3.0
(thrombosis risk increases <2.0)

> 4.0 serious bleeding risk increases

421
Q

why may self-medication with St. John’s Wort have an effect on warfarin requirement?

A

contains hypericin (inducer of cytochrome P450) (hyperforin also has this effect)

increases metabolism of warfarin, decreasing its concentration and making it less effective (less anticoagulant effect)

422
Q

what drugs may have similar adverse effects to St. John’s Wort when administered with warfarin?

A

fewer enzyme inducers than inhibitors but drugs that induce Cyt P450 include:

rifampicin: antibiotic used in TB therapy (onset/duration of induction can depend on half-life)

carbamazepine and phenytoin, as well as older and largely obsolete antiepileptic drugs phenobarbitone and primidone

griseofulvin (antifungal drug)

423
Q

what blood test should be done before increasing digoxin dose?

A

serum potassium

424
Q

why does serum potassium have to be checked before increasing digoxin dose?

A

digoxin binds to the Na+/K+ exchange ATPase on cardiac myocytes and nodal tissue

competes with K+ for its binding site

hypokalaemia means more digoxin binding (less competition by K+) - therefore, enhanced therapeutic and adverse effects of digoxin (heart ‘block’)

425
Q

how does hypokalaemia affect the effects of digoxin?

A

means more digoxin binding (less competition by K+)

therefore, enhanced therapeutic and adverse effects of digoxin (heart ‘block’)

426
Q

how does hyperkalaemia affect the effects of digoxin?

A

indication of possible kidney disease - possible reduced kidney function and therefore reduced digoxin clearance

more digoxin in system, enhanced therapeutic and adverse effects of digoxin

427
Q

how can you ensure that a dose of digoxin is correct?

A

measure plasma levels of digoxin directly – recognised target range for these

428
Q

why may confusion be caused in older patients taking temazepam (benzodiazepine)?

A

(pharmacokinetic) metabolism by liver of some drugs is reduced in older patients – so more temazepam
(pharmacodynamic) older patients may have increased sensitivity to drugs, even if metabolism is normal (may relate to changes in receptor number or function or other)

429
Q

why may INR increase as a consequence of malnutrition if a person is on warfarin?

A

metabolism of many drugs affected by malnutrition

warfarin: similar to a drug which inhibits cyt pathway (e.g. clarithromycin) – therefore, more warfarin

malnutrition also decreases plasma protein levels (warfarin is heavily plasma protein bound)

  • decreases oxidative metabolism (warfarin is metabolised by oxidation)
  • decreases GFR

(increased likelihood of adverse drug reaction)