pharmacology Flashcards
what 3 things are considered when considering a drug’s effects?
where the effect(s) are produced
the drug’s target
response produced after interaction with target
how would the effects of cocaine be described when considering the 3 main points (location of effect, target, response produced)?
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)
what is the key thing that allows a drug to produce an effect?
drug ‘binds’ to target
either enhances activation (“stimulation”) or prevents activation (block effect from being produced)
what are the 4 classes of drug target proteins? give an example of a drug that targets each one.
receptors (e.g. nicotine)
enzymes (e.g. aspirin)
ion channels (e.g. local anaesthetic)
transport proteins (e.g. Prozac)
how does aspirin act on enzymes (target protein) to bring about an effect?
aspirin binds to the enzyme cyclooxygenase
blocks production of prostaglandins
how does local anaesthetic act on ion channels (target protein) to bring about an effect?
block sodium ion channels
prevents nerve conduction
how does Prozac act on transport proteins (target protein) to bring about an effect?
block serotonin carrier proteins
prevents serotonin being removed from the synapse
how does nicotine act on receptors (target proteins) to bring about an effect?
binds to and activates nicotinic acetylcholine receptor
what characteristic must a drug display to be an effective therapeutic agent?
high degree of selectivity for particular drug target
why is it difficult to produce drugs with complete selectivity? what effect does this have?
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 does drug dose relate to drug selectivity?
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
what is an example of how drug dose affects selectivity?
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
what are the 4 types of interaction between drugs and receptors?
electrostatic
hydrophobic
covalent bonds
stereospecific
what types of bonds are involved in electrostatic interactions between drugs and receptors?
(most common interaction)
includes hydrogen bonds and van del Waal’s forces
when are hydrophobic interactions between drugs and receptors important?
in use of lipid soluble drugs
why are covalent bond interactions the least common type of interaction between drugs and receptors?
irreversible
why is stereospecific interaction between drugs and receptors important?
many drugs exist as stereoisomers and interact stereospecifically with receptors
what is pharmacology?
study of how a drug interacts with living organisms and how this influences physiological function
what does the term ‘pharmacodynamics’ refer to?
what the drug does to the body
what does the term ‘pharmacokinetics’ refer to?
what the body does to the drug
how can the basic reaction between a drug and its receptor be represented?
equilibrium between drug and receptor, drug-receptor complex
for a specific concentration of the drug, a specific number of drug receptor complexes are formed
what happens to the equilibrium between and drug and receptor and the drug-receptor complexes when the concentration increases?
more drug molecules available to bind to free receptors
equilibrium shifts to the right
what happens to the equilibrium between and drug and receptor and the drug-receptor complexes when the concentration decreases?
more receptors become available due to lower drug concentration
shifts equilibrium to the left
what are the 2 categories of drug in terms of interaction with receptors?
agonist
antagonists
what is the difference between agonist and antagonists?
both bind to receptors, but only agonists bind and activate receptors
what are the 2 key properties of agonists?
affinity
efficacy
what does the affinity of a drug determine?
strength of binding of drug to receptor, the strength of each drug-receptor complex
therefore affinity is strongly linked to receptor occupancy
how is affinity linked to receptor occupancy?
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
what is efficacy?
ability of an individual drug molecule to produce an effect once bound to a receptor
what are the 3 types of drug with reference to efficacy?
antagonists
partial agonists
full agonists
how does an antagonist work with reference to efficacy?
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 does a partial agonist work with reference to efficacy?
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 does a full agonist work with reference to efficacy?
drug has affinity for the receptor and maximal efficacy
produces the maximal response expected from receptor when bound to it
what is potency?
concentration or dose of a drug required to produce a defined effect
what is the standard measure of potency?
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)
what is the difference between EC50 and ED50?
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 is potency related to dose?
the lower the dosage required to produce a particular effect, the more potent the drug is (often compared using ED50)
what is the relationship between efficacy and potency?
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
what is the clinical relevance of the difference between potency and efficacy?
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)
what are the 4 major pharmacokinetic factors?
absorption
distribution
metabolism
excretion
how is absorption defined with regard to pharmacokinetics?
passage of a drug from site of administration into plasma
what is bioavailability (with regard to absorption in pharmacokinetics)?
fraction of the initial dose that gains access to the systemic circulation
what is the difference between absorption and bioavailability?
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)
what are some common forms of drug administration?
IV
oral
inhalational
dermal (percutaneous)
intra-nasal
what are the 2 ways that drugs move around the body?
bulk flow transfer (i.e. in the bloodstream)
diffusional transfer (i.e. molecule by molecule across short distances)
why is bioavailability likely to be less than 100% when drugs are not administered through an IV?
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
what are the 4 mechanisms by which chemicals diffuse across plasma membranes?
pinocytosis
diffusion across aqueous pores
diffuse across lipid membranes
carrier mediated transport
what is pinocytosis and and what is its relevancy in drug transport?
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
why is diffusion across an aqueous pores not a major route for drug transport across membranes?
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 do most drugs move across membranes?
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)
why are the majority of drugs more water soluble than lipid soluble?
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
what feature of a drug affects its lipid solubility?
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
what 2 things affect the ionisation of a drug?
dissociation constant (pKa) for that drug
pH in that particular part of the body
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?
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 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?
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)
what is ion trapping?
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 is ion trapping overcome?
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
what are the 4 places where the most important carrier systems for drug action are found?
renal tubule
biliary tract
blood brain barrier
gastrointestinal tract
what are carrier systems in the renal tubule, biliary tract, blood brain barrier and GI tract responsible for?
(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)
what is distribution?
when a drug is absorbed it is then distributed to various tissues where they eventually take effect
what are the 4 factors that influence tissue distribution?
regional blood flow
plasma protein binding
capillary permeability
tissue localisation
how does regional blood flow affect tissue distribution?
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 does plasma protein binding affect tissue distribution?
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
what 3 factors affect the amount of drug that binds to plasma protein?
free drug concentration
affinity for the protein binding sites
plasma protein concentration
why are plasma proteins never saturated with drugs?
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
what factor affects differences in the extent of plasma protein binding?
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
what are the 4 different types of capillary structure?
continuous
blood brain barrier
fenestrated
discontinuous
how does a continuous capillary structure affect capillary permeability and therefore drug distribution?
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
what does a continuous capillary structure look like?
endothelial cells aligned in single file with small gap junctions between the cells
what does the blood brain barrier’s capillary structure look like?
continuous structure, but with tight junctions between endothelial cells
what are the 4 different types of capillary structure?
continuous
blood brain barrier
fenestrated
discontinuous
how does a continuous capillary structure affect capillary permeability and therefore drug distribution?
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
what does a continuous capillary structure look like?
endothelial cells aligned in single file with small gap junctions between the cells
what does the blood brain barrier’s capillary structure look like?
continuous structure, but with tight junctions between endothelial cells
why is the brain the most difficult tissue in the body for drugs to gain access to?
blood brain barrier’s capillary structure
how are less lipid soluble drugs transported into the tissue?
if very small, can pass through gap junctions
otherwise, via carrier proteins
what is an example of a tissue with a discontinuous capillary structure?
liver
how does a fenestrated capillary structure allow the kidney to carry out its function?
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
what does a discontinuous capillary structure look like?
big gaps between capillary endothelial cells
what does a fenestrated capillary structure look like?
fenestrations - circular windows within endothelial cells that allow for passage of small molecular weight substances (including some drugs)
what is an example of a tissue with a fenestrated capillary structure?
kidney glomerulus
how does a discontinuous capillary structure allow the liver to carry out its function?
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 does tissue localisation affect drug distribution?
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
what are the 3 main mechanisms of action of phase 1 of drug metabolism?
oxidation (produces electrophiles)
reduction (produces nucleophiles)
hydrolysis (produces nucleophiles)
what happens during oxidation in phase 1 of drug metabolism?
(most common mechanism)
all oxidation reactions start with a hydroxylation step using the cytochrome P450 system to incorporate oxygen into non-activated hydrocarbons
what enzymes within the liver are responsible for drug metabolism?
mostly cytochrome P450
what are the the 2 kinds of drug metabolism that act together to decrease lipid solubility?
phase 1 – introduce a reactive group to the drug
phase 2 – add a conjugate to the reactive group
what are the 3 main mechanisms of action of phase 1 of drug metabolism?
oxidation
reduction
hydrolysis
what happens in oxidation in phase 1 of drug metabolism?
(most common mechanism)
all oxidation reactions start with a hydroxylation step using the cytochrome P450 system to incorporate oxygen into non-activated hydrocarbons
what is the end result of phase 1 of drug metabolism?
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
what are pro-drugs?
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)
what enzymes are used in phase 2 of drug metabolism?
transferases
transfer the substituent group onto the phase 1 metabolite
what occurs in phase 2 of drug metabolism?
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
what are the common phase 2 conjugates that complement the usual phase 1 metabolites?
for electrophiles:
- glutathione conjugation
for nucleophiles:
- glucoronidation
- acetylation
- sulfation
what is first pass (pre-systemic) hepatic metabolism?
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
what are the 3 major routes for drug excretion via the kidney?
glomerular filtration
active tubular secretion (or reabsorption)
passive diffusion across tubular epithelium
what is the problem with the usual solution for first pass (pre-systemic) hepatic metabolism
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
what are the 2 most important routes of excretion?
via kidney (in urine)
via liver (in bile)
what are some other routes of excretion (excluding the kidney and liver)?
via lungs
in breast milk
what are the 3 major routes for drug excretion via the kidney?
glomerular filtration
active tubular secretion (or reabsorption)
passive diffusion across tubular epithelium
why does excretion of different drugs vary so much?
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
why does urine pH affect the process of excretion by passive diffusion in the kidney?
urine pH varies from 4.5 to 8
acidic drugs are better reabsorbed at lower pH
basic drugs are better reabsorbed at higher pH
why is active tubular secretion the most important method of excretion via kidney?
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
what is the process of excretion by active tubular secretion in the kidney?
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
why does drug metabolism affect the process of excretion by passive diffusion in the kidney?
phase 2 metabolites tend to be more water soluble than the parent drug
therefore less well reabsorbed
why does urine pH affect the process of excretion by passive diffusion in the kidney?
urine pH varies from 4.5 to 8
acidic drugs are better reabsorbed at lower pH
basic drugs are better reabsorbed at higher pH
what is the process of excretion by passive diffusion in the kidney?
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 are drugs excreted via the bile?
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 can enterohepatic recycling prolong drug effect?
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
what is the primary mechanism of action of metformin?
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
what is the drug target of metformin?
5′-AMP-activated protein kinase (AMPK) (enzyme)
what is the primary site of action of metformin?
hepatocyte mitochondria
what are the side effects of metformin?
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
why may metformin accumulate the in the liver and the GI tract to produce therapeutic and side effects respectively?
highly polar structure, requires organic cation transporter-1 (OCT-1) to access tissues
when is metformin most effective?
in the presence of endogenous insulin
i.e. with some residual functioning pancreatic islet cells
what is an example of a DPP-4 inhibitor?
sitagliptin
what is the primary mechanism of action of DPP-4 inhibitors?
primary effect:
inhibiting DPP-4 action
(enzyme is present in vascular endothelium and can metabolise incretins in plasma)
what are the functions of incretins (e.g. GLP-1)?
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
what is the drug target of DPP-4 inhibitors?
DPP-4 (enzyme)
what is the primary site of action of DPP-4 inhibitors?
vascular endothelium
what are the side effects of DPP-4 inhibitors?
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)
what is a benefit of DPP-4 inhibitors as opposed to other anti-diabetic drugs?
do not cause weight gain
what is required for DPP-4 inhibitor effectiveness?
some residual pancreatic beta-cell activity
since they act mainly by augmenting insulin secretion
what is an example of a sulphonylurea?
gliclazide
what is the primary mechanism of action of sulphonylureas?
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
what is the drug target of sulphonylureas?
ATP-sensitive potassium channel (ion channel)
what is the primary site of action of sulphonylureas?
pancreatic beta cell
what are the side effects of sulphonylureas?
weight gain (likely)
hypoglycaemia (2nd most common)
what is required for sulphonylurea effectiveness?
some residual pancreatic beta-cell activity
since they act mainly by augmenting insulin secretion
how is the weight gain caused by sulphonylureas mitigated?
concurrent administration with metformin
when especially should the risk of hypoglycaemia associated with sulphonylureas be discussed with the patient?
when concomitant glucose-lowering drugs are prescribed
what is an example of a sodium-glucose co-transporter (SGLT2) inhibitor?
dapaglifozin
what is the primary mechanism of action of SGLT2 inhibitors?
reversibly inhibits sodium-glucose co-transporter 2 (SGLT2) in the renal proximal convoluted tubule
reduces glucose reabsorption, increases urinary glucose excretion
what is the drug target of SGLT2 inhibitors?
SGLT2 (transport protein)
what is the primary site of action of SGLT2 inhibitors?
proximal convoluted tubule
what are the side effects of SGLT2 inhibitors?
uro-genital infections due to increased glucose load (5% of patients)
slight decrease in bone formation
can worsen diabetic ketoacidosis (stop immediately)
what are some added benefits of SGLT2 inhibitors?
weight loss
reduction in BP
what is required for SGLT2 inhibitors effectiveness?
depends on normal renal function
less effective in patients with renal impairment
what drugs can be used in the treatment of diabetes?
metformin
DPP-4 inhibitors
SGLT2 inhibitors
sulphonylureas
what drugs can be used in the treatment of epilepsy?
lamotrigine
sodium valproate
diazepam
levetirecetam
what is the primary mechanism of action of lamotrigine?
blocks voltage gated Na+ channels, preventing Na+ influx
prevents depolarisation of glutamatergic neurones and reduces glutamate excitotoxicity
what is the drug target of lamotrigine?
voltage gated Na+ channels
what are the main (common) side effects of lamotrigine?
rash
drowsiness
what are the main (more uncommon but serious) side effects of lamotrigine?
Steven-Johnson’s syndrome
suicidal thoughts
how is lamotrigine used in treatment of allergic reactions?
lamotrigine introduced gradually - important in reducing frequency and severity of allergic skin reactions
what is the primary mechanism of action of sodium valproate?
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
what is the drug target of sodium valproate?
GABA transaminase (inhibitory presynaptic terminal)
what are the main (common) side effects of sodium valproate?
stomach pain
diarrhoea
drowsiness
weight gain
hair loss
what are the main (serious) side effects of sodium valproate?
hepatotoxicity
teratogenicity
pancreatitis
how does sodium valproate interact with other drugs?
broad CYP (cytochrome P450) enzyme inhibitor
increases serum concentration of many co-administered drugs
what is the primary mechanism of action of diazepam?
increases Cl- influx in response to GABA binding at GABA A receptor
increased Cl- influx associated with hyperpolarisation of excitatory neurones
what is the drug target of diazepam?
benzodiazepine site on the GABA A receptor
what are the main (common) side effects of diazepam?
drowsiness
respiratory depression (if IV or at high dose)
what are the main (more uncommon but serious) side effects of diazepam?
haemolytic anaemia
jaundice
why is diazepam not used for long term suppression of seizures?
development of tolerance
why is diazepam
a Schedule 4 controlled drug?
addiction prone individuals more likely to become dependent on diazepam
what is the primary mechanism of action of levetiracetam?
inhibition of synaptic vesicle protein SV2A in excitatory presynaptic terminal, prevents vesicle exocytosis
reduction in glutamate secretion reduces glutamate excitotoxicity
why is levetiracetam favourable in terms of lack of drug–drug interactions?
metabolism of levetiracetam has no effect on cytochrome P450 enzyme system
what drugs are used in treatment of depression?
sertraline
citalopram
fluoxetine
venlafaxine
mirtazapine
what is the primary mechanism of action of sertraline?
serotonin reuptake inhibition causes accumulation
serotonin in CNS aids regulation of mood, personality, and wakefulness
what is the drug target of sertraline?
serotonin transporter
what are the main side effects of sertraline?
GI effects (nausea, diarrhoea)
sexual dysfunction
anxiety
insomnia
how does sertraline interact with CYP2D6 at high doses (150mg)?
partial inhibition
how does sertraline interact with dopamine transporters?
mild inhibition
how is sertraline discontinued?
gradually decreased
what is the primary mechanism of action of citalopram?
serotonin reuptake inhibition causes accumulation
serotonin in CNS aids regulation of mood, personality, and wakefulness
what is the drug target of citalopram?
serotonin transporter
what are the main side effects of citalopram?
GI effects (nausea, diarrhoea)
sexual dysfunction
anxiety
insomnia
how does citalopram interact with muscarinic and histamine (H1) receptors?
mild antagonism
how is citalopram discontinued?
gradually decreased
how is citalopram metabolised?
CYP2C19