Pharmacokinetics and pharmacodynamics Flashcards
what is pharmacology?
the study of the effects of drugs
what is pharmacokinetics?
how the body affects the drug; Absorption, Distribution, Metabolism and Excretion (ADME)
what is pharmacodynamics?
how the drug affects the body
what is the role of zinc fingers in cytosolic/nuclear receptors?
can recognise discrete regions of DNA
what is potency?
measure of how well a drug works
what is EC50?
concentration (M) that gives half of the maximal response
what is an agonist?
a compound that binds to a receptor and activates it
what are types of normal response curves?
linear (normal scale) and sigmoidal (log scale)
what is a full vs partial agonist?
- full agonist can get a maximal response
- partial agonist cannot get maximal response when binding to receptor
what is Emax?
maximal response of a drug
what is intrinsic activity/efficacy? how is it calculated?
- the relative ability of a drug-receptor complex to produce a maximum functional response
- Emax of partial agonist ÷ Emax of full agonist
what is affinity?
how well a drug binds to a receptor
what is potency?
- a measure of drug activity expressed in terms of the amount required to produce an effect of given intensity
- highly potent drug evokes a given response at low concentrations
- lower potent drug evokes same response only at higher concentrations
what is an antagonist?
- a compound that reduces the effect of an agonist
- does not activate receptors
- affinity of the antagonist for the receptor affects its activity
what is a competitive antagonist? what is its effect on the dose-response curve?
- bind to receptors at same binding site as the endogenous ligand or agonist, but without activating the receptor
- competes for same binding site with agonist
- sufficient concentrations of an antagonist will displace the agonist from the binding sites
- dose-response curve shifts to the right; more agonist is required to elicit the same response; decreased potency
what are the types of competitive antagonists?
- reversible (surmountable)
- irreversible (insurmountable)
what are reversible (surmountable) competitive antagonists?
- bind via noncovalent intermolecular forces
- will eventually dissociate from the receptor, freeing it to be bound again
what are irreversible (insurmountable) competitive antagonists?
- bind via covalent intermolecular forces
- there is not enough free energy to break covalent bones in the local environment, so the bond is essentially permanent, so the complex will never dissociate
- receptor remains permanently antagonised until it is ubiquinated and destroyed
what is a non-competitive antagonist? what effect does it have on the dose-response curve?
- type of insurmountable antagonist that binds to an allosteric (other) site of the receptor, or by irreversibly binding to the active site of the receptor
- reduce maximum response that can be attained by any amount of agonist
- effects cannot be negated, no matter how much agonist is present
- shift right and down of the dose-response curve
what is the agonist and antagonist of the mAChR receptor?
muscarinic ACh receptor
- agonist: muscarine
- antagonist: atropine
what is the agonist and antagonist of the nAChR receptor?
nicotinic ACh receptor
- agonist: nicotine
- antagonist: curare
what is the agonist for the histamine receptor? what are its effects?
agonist: histamine
- contraction of the ileum
- acid secretion from parietal cells
what is the antagonist for the histamine receptor? what are its effects?
antagonist: mepyramine
- reversed contraction of the ileum
- no effect on acid secretion
what is efficacy?
how well a ligand activates the receptor
what shows affinity?
shown by both antagonists and agonists
what shows efficacy?
- only agonists show efficacy
- antagonists show zero efficacy
what are factors governing drug action?
- affinity
- efficacy
- number of receptors at tissue
- signal amplification
how does the number of receptors at the tissue affect drug action?
- the less number of receptors at a tissue will result in more drug being required to illicit the same effect and vice versa
- in most cases it is possible to still get maximum response despite a reduction in receptor numbers meaning you only need a small number of receptors to get a full response
- example of drug that will inactivate a receptor is: bromoacetyl alprenolol menthane (BAAM) which is an
irreversible B-adrenoceptor antagonist - an equation called the Furchgott equation is used to calculate how many receptors are available
- receptor reserve/Spare receptors:
• many tissues have holds for a full agonist, this reserve can be large or small depending on the tissue
• there is no receptor reserve for a partial agonist
what is the Furchgott equation? what does it calculate?
used to calculate how many receptors are available in a tissue
what is signal amplification? how does it affect drug action?
- when a ligand binds to a receptor it sets off a signalling cascade
- signal amplification determines how powerful the response will be
- determined by the type of tissue the receptor is based in
what is allosteric modulation?
- substance binds to a receptor to change the receptor’s response to stimulus
- bind to allosteric site
- affinity modulation: change in EC50
- efficacy modulation: change in Emax
- modulation can be positive-allosteric or negative-orthosteric
what is an example of an allosteric ligand?
benzodiazepine
what is an inverse agonist?
a drug that binds to the same receptor as an agonist but induces a pharmacological response opposite to that of the agonist; <0% efficacy
what is a neutral antagonist?
has no activity in the absence of an agonist or inverse agonist but can block the activity of either; 0% efficacy
what is tolerance?
- the diminishing effect of a drug resulting from repeated administration at a given dose
- seen with continuous, repeated high concentration of drug over time
what is desensitisation of receptors?
- uncoupled - receptor can’t interact with G-protein
- receptor is internalised in vesicle of cell
- receptor becomes degraded
what is specificity?
- the ability of a protein’s binding site to bind specific ligands
- the fewer ligands a protein can bind, the greater its specificity
- describes the strength of binding between a given protein and ligand
what is selectivity?
- how a ligand may bind more preferentially to one receptor than another
- defined with respect to the binding of ligands to a substrate forming a complex
what are examples of selectivity in beta-adrenoceptors?
- isoprenaline is a non-selective B-adrenoceptor agonist so it activates both B1 (heart) & B2 (lungs) receptors
- whereas salbutamol is a selective B2-adrenoceptor agonist, but at very high concentrations it loses its specificity so will activate both B1 and B2
what catalyses the conversion of membrane phospholipid to arachidonic acid?
phospholipase A2
what is the action of phospholipase A2?
converts membrane phospholipid to arachidonic acid
what catalyses the conversion of arachidonic acid to prostaglandin H2?
cyclooxygenase
what is the action of cyclooxygenase?
converts arachidonic acid to prostaglandin H2
what are examples of NSAIDs?
include aspirin and ibuprofen but there are about 50
what are the actions of NSAIDs?
• analgesic
• anti-pyretic (reduces fever)
• anti-inflammatory
• they inhibit the enzyme
cyclooxygenase (COX) which is responsible for the breakdown of arachidonic acid to prostaglandin H2 (PGH2)
• NSAIDs work by preventing arachidonic acid from reaching the active site of the COX enzyme and thus from being broken down into prostaglandin H2 - this is achieved by competitive inhibition
what is PGH2 acted on? what does this generate?
PGH2 is acted on by specific synthases to generate prostanoids: • Prostaglandin D2 (PGD2) • Prostaglandin E2 (PGE2) • Prostaglandin I2 (PGI2) • Thromboxane A2 (TXA2)
what are some examples of prostanoids?
- Prostaglandin D2 (PGD2)
- Prostaglandin E2 (PGE2)
- Prostaglandin I2 (PGI2)
- Thromboxane A2 (TXA2)
what produces the different prostanoids?
- Prostaglandin D2 - mast cells
- Prostaglandin I2 - vascular endothelial cells
- Thromboxane A2 - platelets
- Prostaglandin E2 - macrophages (most widely found and mediates largest number of effects in the body)
what is the action of aspirin?
- prevents arachidonic acid from reaching the active site of the COX enzyme and thus from being broken down into prostaglandin H2 (competitive inhibition)
- irreversibly blocks the active site resulting in irreversible inactivation of the COX enzyme
what are the two isoforms of COX?
- COX-1: found normally and widely around the body (normal physiology)
- COX-2: this is induced and found mainly in inflammation only
what are examples of selective and non-selective drugs for COX?
- aspirin is non-selective so acts on both COX-1 and COX-2
* whereas celecoxib for example is COX-2 selective
what are examples of ACE inhibitors?
captopril and enalapril
what are actions of ACE inhibitors?
- anti-hypertensives
- work by inhibiting ACE thereby preventing the conversion of angiotensin I to
angiotensin II meaning there is less angiotensin II so less bind to angiotensin receptors (AT1) resulting in reduced vasoconstriction and thus
hypertension as well as less aldosterone release further reducing hypertension
what is the mechanism of action of ACE inhibitors?
- Captopril - mimic dipeptides; His and Leu
- Enalapril - mimic tripeptides; Phe, His and Leu
- both captopril and enalapril work by binding to ACE active site thereby meaning it is unable to convert angiotensin I to angiotensin II
what are examples of beta-lactam antibiotics?
penicillins, amoxicillin and cephalosporins
what is the action of beta-lactam antibiotics?
work by inhibiting cell wall biosynthesis of peptidoglycan bacterial cell walls by inhibiting the activity of certain enzymes
what is the enzymatic inactivation of drugs?
- most drugs are excreted by the kidneys but lipophilic drugs are not effectively
removed as they are passively absorbed because they can diffuse through cell membranes easily - in these cases Cytochrome P450 is required to introduce a hydroxyl (OH) group into the drug to enable it to be excreted more easily by the kidneys
- note: some drugs act to either inhibit or induce CYP450s to either make drugs remain in the system for longer or to be excreted quickly (e.g. quite toxic drugs)
what are examples of proton pump inhibitors?
omeprazole, lansoprazole, pantoprazole and rabeprazole
what are the actions of proton pump inhibitors?
- they are all activated in acidic (i.e. stomach) environments
- they act to inhibit acid secretion
- when prostaglandin E2 (PGE2) released from chromaffin cells, it binds to EP3 receptors on parietal cells, this causes a reduction in the concentration of H+ and thus acidity of the stomach by reducing the activity of the H+/K+ ATPase pump - PGE2
inhibits parietal cells - when histamine released from histaminocytes, binds to H2 receptors on parietal cells, this causes the H+/K+ ATPase pump to increase activity
resulting in a higher concentration of H+ being pumped into the gastric lumen thereby increasing acidity - histamine activates parietal cells - PPIs act to irreversibly inactivate the proton pump (H+/K+ ATPase) resulting in a marked reduction in acidity of the stomach, thereby helping with reflux etc.
what is the action of diuretics?
- act to inhibit symporters
- the increased excretion of water is useful for the treatment of hypertension and heart failure since the removal of water from the blood will reduce its volume and thus blood pressure
what is the action of furosemide?
furosemide can inhibit the NKCC2 pump on the thick ascending part of the loop of Henle thereby reducing the amount of Na+,Cl- and K+ ions able to enter the medullary interstitium thereby reducing hyperosomolarity meaning
that less water will diffuse out of the collecting ducts into the blood, resulting in more water loss in the urine and thus dehydration
what is the action of thiazides?
act to inhibit the Na/Cl cotransporter on the distal tubule of the nephron; this results in increased water loss
what neurotransmitters are subject to uptake?
dopamine, noradrenaline, serotonin and GABA
what do uptake inhibitors do?
cause an increased concentration of neurotransmitter in the synapse by preventing their re-uptake
what is the action of imipramine?
tri-cyclic anti-depressant that mostly inhibits the reuptake of noradrenaline (majority) and serotonin
what is the action of cocaine?
inhibits reuptake of dopamine
what is the action of tiagabin?
inhibits the reuptake of GABA (the inhibitory transmitter in CNS) thus useful for the treatment of panic disorders
what are examples of local anaesthetics?
lidocaine and procaine
what is the action of local anaesthetics?
- these local anaesthetics work by interrupting axonal neurotransmission in the sensory nerves
- they do this by blocking voltage dependent sodium channels thereby preventing the neurones from depolarising, meaning the threshold isn’t met and thus no action potential is developed to be propagated
- this rests in pain relief since pain isn’t transmitted to the brain to be perceived
- local anaesthetics can diffuse through mucus membranes easily thus
sometimes can act on muscles too
what is a drug?
any compound that is administered with an intended therapeutic effect
what are the three phases of plasma level?
- uptake into the plasma
- distribution from the plasma
- elimination from the plasma
what is the rate of diffusion like for a dissolved drug (in the plasma)?
- rate of diffusion is directly proportional to the concentration gradient - it is a first order process
- an increase in concentration will result in a higher rate of diffusion from plasma to tissue
- rate of diffusion is directly proportional to temperature (most drugs won’t work outside normal physiological temperature)
- rate is influenced by chemical reactions between the drug and solute (i.e. plasma)
what is a first order reaction?
rate is directly proportional to the concentration of drug (rate ∝ [drug])
what is a second order reaction?
rate is directly proportional to the square of the concentration of the drug (rate ∝ [drug]^2)
what is a third order reaction?
rate is directly proportional to the cube of the
concentration of the drug (rate ∝ [drug]^3)
what is a zero order reaction?
rate is unrelated to the concentration of the drug
rate ∝ [drug]0
what order process is diffusion?
first order
how is diffusion an exponential function?
where the rate of reaction is governed by one of the components involved in the reaction whose quantity or
magnitude is changing
what are features of plasma?
• the plasma is the fluid fraction or aqueous solution that remains when cells are
removed from the blood
• proteins (including antibodies) are found in the plasma as a result of their polar amino acid side chains
• the pH of the interstitium is reflected in the plasma
• plasma has a slightly higher pH than interstitium due to diffusion gradient
what does the pharmacokinetic theory consider the body as?
pharmacokinetic theory considers the body as 3 main compartments divided by tissue lipid rich barriers
what are the three main compartments of the body?
- plasma (5 litres)
- interstitial (15 litres)
- intracellular (45 litres)
what is the cellular tissue divided into?
- vessel rich viscera: muscle tissue
- vessel poor: fat stores/subcutaneous tissue
what are the 5 ways a drug can move from its site of administration to its target?
- simple diffusion
- facilitated diffusion
- active transport
- through extracellular spaces
- non-ionic diffusion
what are features of simple diffusion? what order process is it?
• movement of solutes from a region of their high
concentration to a region of
their low concentration through a lipid barrier
• first order process
what are features of facilitated diffusion?
• movement of solutes from a region of their high concentration to a region of their low concentration through protein channels • no energy required
what are features of active transport? what order reaction is it?
• movement of solutes from a region of their low concentration to a region of their high concentration using protein carriers • require energy • goes against the concentration gradient • first order reaction initially but when saturated it becomes zero order
what are features of movement through extracellular spaces?
- movement of solutes through pores in cell membrane
* protein typically cannot enter through these pores
what are features of non-ionic diffusion?
• the movement of ionic molecules (which find it difficult to diffuse across a lipid membrane) across a lipid membrane (such as that found in a cell) into the cell • ionised molecules are water soluble • un-ionised molecules are freely diffusable and lipid soluble • the mechanism of action of non-ionic diffusion is that an ionic molecule becomes less ionic and thus more non- ionic to enable it to cross the lipid membrane and enter the cell
what is the solubility of ionised and unionised molecules?
• ionised molecules are water soluble
• un-ionised molecules are
freely diffusable and lipid
soluble
what drug works by non-ionic diffusion?
aspirin
what affects non-ionic diffusion?
• the degree of ionisation of
weak acids and weak bases is influenced by pH
• non-ionic diffusion is enhanced if adjacent compartments have a pH difference
what is the effect of acids and bases when pH is increased?
weak acid: more ionised
weak base: less ionised
what is the effect on acids and bases when pH is decreased?
weak acid: less ionised
weak base: more ionised
what is bioavailability?
amount of drug taken up as a proportion of the amount administered
why does oral administration have the greatest variability?
has the greatest variability due to different factors involved; surface area of gut, diarrhoea (bioavailability is greatly decreased) and pH of
gut (alkaline at duodenum)
how do drugs move across the gut membrane?
simple diffusion of the lipid
soluble freely diffusible non ionised fraction
what are features of oral administration?
- has the greatest variability due to different factors involved; surface area of gut, diarrhoea (bioavailability is greatly decreased) and pH of
gut (alkaline at duodenum) - unpredictable
- movement across the gut membrane is simple diffusion of the lipid soluble freely diffusible non ionised fraction
- most tablets are either weak acids or weak bases
- water soluble tablets will not pass across the membranes of cells unless there is carrier mediated transport
what is the bioavailability of intramuscular administration?
close to 1
what is the bioavailability of IV administration?
should be 1
what are features of transcutaneous administration?
- such as a skin patch
- has a lower bioavailability than I.V.
what are types of routes of administration?
- oral
- intramuscular
- IV
- transcutaneous
- intrathecal (into CSF)
- sublingual
- inhalation
- topical
- rectal
is aspirin acidic or basic?
acidic
how is aspirin absorbed?
- aspirin is acidic
- in the stomach the acidic tablet dissolves but it then becomes less ionised due to the fact that a weak acid (such as aspirin) becomes less ionised in lower pH
- movement of the un-ionised aspirin across the gut is rapid
- in the plasma, aspirin is more ionised due to the higher pH
- gastric pH affects the amount of aspirin uptake
why is aspirin more ionised in the plasma?
due to higher pH
what affects aspirin uptake?
- in the plasma aspirin is more ionised due to the higher pH
* gastric pH affects the amount of aspirin uptake
why does aspirin become less ionised in the stomach?
- in the stomach the acidic tablet dissolves but it then becomes less ionised due to the fact that a weak acid (such as aspirin) becomes less ionised in lower pH
- movement of the un-ionised aspirin across the gut is rapid
how can gastric pH be increased?
- antacids act to increase pH e.g. treatment of ulcer
- omeprazole (PPI) and ranitidine (H2 blocker) acts to reduce acid secretion in stomach and thus increase pH
- ingesting alkali foods results in an increased pH
- a raised pH results in the reduced uptake of aspirin from the stomach and thus a reduction in bioavailability
what reduces aspirin uptake?
a raised pH results in the reduced uptake of aspirin from the stomach and thus a reduction in bioavailability
what is a drug distributed according to?
a drug is distributed in the plasma according to its chemical properties and
molecular size
what is found in the aqueous phase?
dissolved gases and small ionic molecules
where do lipophilic drugs adhere to?
hydrophobic areas of plasma proteins
what compounds are active in the three compartments of the body?
- proteins/large molecules are only active in the plasma compartment (5L)
- water soluble molecules are active in plasma and interstitial compartment
(5L + 15L) - lipid soluble molecules are only active in the intracellular fluid (45L)
what compartment are proteins/large molecules active in?
plasma compartment (5L)
what compartment are water soluble molecules active in?
plasma and interstitial compartments (5L + 15L)
what compartment are lipid soluble molecules active in?
intracellular fluid (45L)
what is the volume of distribution derived from?
derived value from steady state (where drug intake is in equilibrium with its
elimination) plasma level studies
what is the volume of distribution? how is it calculated?
- volume of distribution = total amount of drug in body ÷ concentration of drug in plasma
- it is the volume (litres) that the drug would occupy if it was distributed through all the compartments as if they were all plasma
what should be kept in mind when calculating the volume of distribution?
- the volume of distribution can appear to be larger than it actually is, this is because when calculating volume of distribution, concentration and volume is taken from the plasma (blood)
- however, when a drug is injected it can be taken up by organ systems (such as the liver, lungs and kidney etc.) this will mean its concentration in the blood will decrease
- thus when you calculate the volume of distribution after the drug has been partly taken up then the value you get can be much larger than it physically is
- drugs that are highly lipid soluble, enter the CNS and have a high volume of distribution
what is the volume of distribution of lipid soluble drugs?
drugs that are highly lipid soluble, enter the CNS and have a high volume of distribution
what drugs are found in the plasma?
- plasma expanders
- immunoglobulin
- warfarin
what drugs are found in the interstitial compartment?
- aspirin/ other NSAIDs
- antibiotics
- muscle relaxants
what drugs are found in the intracellular compartment?
- steroids
- local anaesthetics
- opioids
- CNS drugs
- paracetamol
- amiodarone (has a volume of distribution of 450L i.e. very easily taken up by tissue)
what is compartment modelling? what does it assume?
- assumes plasma is in equilibrium
- shows plasma concentration against time during the distribution of drug phase
- line of best fit to 1,2 or 3 compartment models of distribution
what is the 1 compartment model?
C = C0 e^-kt
what is the 2 compartment model?
C = C0 e^-kαt + C0 e^-kβt
what is the 3 compartment model?
C = C0 e^-kαt + C0 e^-kβt + C0 e^-kλt
what is the pattern of compartment modelling?
for every compartment you
add to the model you add
another “C0 e-kt”
what compartment model do lipid soluble drugs have?
most lipid soluble drugs have 3 compartment models - this suggests that the movement between the compartments is plasma > viscera > adipose
tissue
where is drug eliminated from?
plasma compartment
how are drugs eliminated?
- renal and/or hepatic elimination are the route for the vast majority of drugs
- in renal failure, a patient will not be able to eliminate for up to a week
what order process is elimination?
first order process
what are the two definitions of clearance?
- the volume of plasma that can be completely cleared of drug per unit time (mls minute^-1 (ml/min))
- the rate at which plasma drug is eliminated per unit plasma concentration (mls minute^-1 (ml/min))
what are features of clearance?
- the removal of drug from the plasma by either liver or kidney is clearance
- both definitions are measures of efficiency
- can influence the rate of elimination depending on plasma concentration of drug
how are units of clearance derived?
- the rate at which plasma drug is eliminated = mg per minute
- per unit plasma concentration = mg per ml
- total = mg per minute ➗ mg per ml
- cancel out the “mg”
- results in = mls minute^-1 (ml/min)
- thus the units of clearance are mls minute-1 (ml/min)
what are features of renal clearance? how are water soluble molecules processed?
• all of the factors affecting renal blood flow affect clearance most notably
blood pressure
• water soluble molecules which pass through the glomerular endothelial gap are eliminated by glomerular filtration
• larger water soluble molecules can be eliminated by active tubular secretion
how is renal clearance measured?
- the calculation assumes; rate of elimination = rate of appearance in urine
- plasma concentration measured during the clearance process is assumed to be constant
- clearance = rate of appearance in urine ➗ plasma concentration
- creatinine is used as a marker substance in kidney
what is used as a marker substance in the kidney?
creatinine
what is adult renal blood flow?
renal blood flow is 18% of cardiac output = 1L/min
what is the adult renal plasma flow?
renal plasma flow is 60% of blood flow = 600mls/min
what is the adult glomerular filtration rate?
glomerular filtration is 12% of renal blood flow = 130mls/min
what drugs are eliminated by renal clearance?
- many drugs are eliminated by the kidney either by glomerular filtration e.g digoxin and gentamicin or via active secretion e.g. penicillin, frusemide and thiazides
- many highly lipid soluble drugs metabolised to water soluble glucuronic acid conjugates are eliminated by active secretion e.g.
morphine 3 & 6 glucuronides - only free plasma is cleared
- highly protein bound drugs have little exposure to renal clearance
- most drugs eliminated by the kidney are water soluble and small molecules
what are most of the drugs eliminated by the kidney?
water soluble and small molecules
what is acute renal impairment often secondary to?
reduced pre-renal perfusion
what is chronic renal impairment caused by?
diabetes and hypertension
what will kidney damage result in?
reduced clearance and prolonged elimination of a large number of drugs
how does renal impairment affect drug clearance?
- for patients with renal impairment, choose drugs that are eliminated by the liver instead
- hypoalbuniaemia means lipid soluble drugs in the plasma have highly freely diffusable fractions and greater effects
- elevated plasma creatinine and urea compete for lipid binding sites on protein and displace more lipid soluble free drug
- patients with renal impairment have unpredictable reactions to drugs due to oedema in compartments
what is the management of renal impairment?
- choose alternatives not eliminated by the kidney
- avoid nephrotoxic drugs
- make corrections based on plasma creatinine which estimated the percentage reduction in clearance and then alter dose
- measure plasma concentrations if there is a toxicity risk
what are features of hepatic clearance?
- has no equivalent to glomerular filtration
- all hepatic clearance involves active transport
- there is active secretion (if water soluble) from the liver into the bile duct
what is the hepatic blood flow?
hepatic blood flow is 24% of cardiac output (3/4 from portal vein and 1/4 from hepatic artery)
what is the hepatic extraction ratio?
the proportion of drug removed by one passage through the liver
what does a high HER mean?
can be so high that clearance is only limited by hepatic blood flow - perfusion limited
what does a low HER mean?
the process is slow and not efficient with a low amount
removed - diffusion limited
what can affect HER? how does HER affect the liver?
- liver enzymes can alter the HER
- if the liver is exposed to a low HER drug then it will produce more enzymes to enable it to increase clearance
- high HER drugs have little effect on the number of new enzymes produced
what are features of drugs with a high HER?
drugs with a high HER are described as having a high first pass metabolism and thus have limited uptake from oral administration since they will first go to the liver before it can reach its target but by
that time most of it will have been eliminated - ineffective
how are different drugs eliminated from the liver?
- some large water soluble molecules are secreted unchanged in bile
- most large lipid soluble drugs are metabolised to increase their water solubility so that they may be excreted more easily by the kidney
- pro-drugs are activated in the liver, whereby the pro-drug is cleaved into the active drug via metabolism e.g hydrocortisone is broken down into the active drug cortisol
where do most phase I reactions happen in the liver?
95% of phase I reactions are in liver smooth endoplasmic
reticulum
what is the aim of phase I reactions?
aim is to make the drug more hydrophilic so that it can be excreted by the kidneys - it does this by adding a hydroxyl group to the drug
what happens in phase I reactions?
- they introduce or expose hydroxyl (-OH) groups or other reactive sites that can be used for conjugation reactions (the Phase II reactions)
- introduces reactive group to drug - attack point for conjugation
- hydrophilic molecules usually do not reach the metabolising enzymes since they are excreted easily
- oxidation
- reduction: add hydrogen (saturate unsaturated bonds)
- hydrolysis: split amide (peptide bond (between a carboxyl (COOH) and amino (NH) group) O=C-NH) and ester (the H from a COOH
(carboxylic acid) is replaced by some sort of hydrocarbon) bonds
what happens in oxidation reactions in phase I reactions?
- hydroxylation (add -OH)
- dealkylation (remove -CH side chains)
- deamination (remove -NH)
- hydrogen removal
what happens in reduction in phase I reactions?
add hydrogen (saturate unsaturated bonds)
what happens in hydrolysis in phase I reactions?
split amide (peptide bond (between a carboxyl (COOH) and amino (NH) group) O=C-NH) and ester (the H from a COOH (carboxylic acid) is replaced by some sort of hydrocarbon) bonds
what is functionalisation in phase I reactions?
- introduces reactive group to drug
- includes adding or exposing; -OH,-SH,-NH2,-COOH
- the product of the reaction is usually more reactive
- there is a small increase in hydrophilicity
where do phase I reactions occur? what are they catalysed by?
- mainly occur in the liver
- mainly catalysed by cytochrome P450 enzymes
what is cytochrome P450?
- type of microsomal enzyme i.e. found on liver smooth
endoplasmic reticulum - involved in Phase I reactions
what is the action of cytochrome P450?
- uses heme group (Fe2+) to oxidise substances
- products of P450 enzymes are more water soluble
- requires energy and molecular oxygen
what are examples of drugs that inhibit cytochrome P450?
some drugs act to inhibit cytochrome P450 such as amiodarone and cimetidine resulting in drugs lasting longer/not being eliminated as fast
what is involved in phase II conjugation reactions?
- also known as glucuronidation, used if drug is very hydrophobic
- essentially adding a glucuronic acid group (glucuronide) to the drug to make it more hydrophilic
- process forms covalent bonds
what enzymes are involved in phase II reactions?
- enzyme: glucuronosyltransferase (Uridine 5’-diphospho-
glucuronosyltransferase) (UGT) - microsomal enzyme, used in
phase II reactions, catalyses reaction - uridine diphospho-glucuronic acid (UDPGA) - essentially a co-
enzyme/donor compound required to conjugate glucuronic acid
what is the product of phase II reactions?
UDPGA + drug -> uridine diphosphate + drug-glucoronide
- catalysed by UGT
what happens to phase I water soluble metabolites and phase II glucuronides?
- phase I water soluble metabolites and phase 2 glucuronides enter the bile and then enter the gut via the cystic duct
- some phase I metabolites and phase 2 glucuronides also enter the blood and are actively excreted into the urine
- during cholestasis (stoppage of bile flow), increased amounts are excreted by the kidneys
- conversely biliary secretion may increase if renal function is reduced
how does the phase II glucuronide get metabolised in the enterohepatic circulation?
- large bowel flora tend to metabolise the glucuronic acid group on the phase 2 glucuronide
- this results in the liberation of the drug from the glucuronic acid
- the drug is then able to re-diffuse from the gut into the blood and then have a prolonged effect before going back to the liver to be re-conjugated and excreted again
- thus the enterohepatic circulation can prolong the action of some drugs
when does liver failure affect drug metabolism?
- isolated liver failure with no effects on other organs is unusual
- there is minimal effect on drug metabolism until at least 70% of functioning liver is lost
what is a disadvantage of prolonged duration of action of drug?
the prolonged duration of the action of a drug comes with the risk of drug accumulation and thus toxicity
how can liver failure lead to toxicity when metabolising drugs?
- the prolonged duration of the action of a drug comes with the risk of drug accumulation and thus toxicity
- hypoalbuminaemia means an increased free plasma levels of freely diffusable lipid soluble drugs
- pharmacodynamic (how drug affects body) alterations are often secondary to disease
- the nitrogen containing substances that are cleared by the liver contribute to toxicity
what are some drugs with active metabolites?
- Prednisone
- Isosorbide dinitrate
- Codeine
- Diamorphine
- L-dopa
- Cortisone
- Morphine
what are drugs that are given IV?
- Insulin
- Heparin
- Antibiotics
- Frusemide
- Anti-arrhythmics
- Sedation anaesthsia
why are IV infusions used?
- enables steady state plasma (where drug intake is in equilibrium with its elimination) levels to be maintained for as long as possible
- enables highly accurate drug delivery
- useful for drugs that are ineffective administered by other routes or those who cannot absorb oral medication
- is the quickest administration route
- guarantees 100% bioavailability (the gold standard)
what are advantages/disadvantages of oral administration?
- much less accurate drug delivery since bioavailability can be highly variable resulting in uncertainty or effectiveness of treatment
- however it does have excellent patient compliance with one tablet a day
what are advantages/disadvantages of IV infusion?
- requires constant monitoring of patency of IV access, replenishing of drug delivery and observation of response to therapy
- has the potential for serious calculation errors
- limited by the number of places a cannula can be inserted
what must be monitored in IV infusion?
patency of IV access, replenishing of drug delivery and observation of response to therapy
how are IV infusions dosed? what are the units?
- IV regimes lack standardisation of prescription
- units are mg hour-1 (mg/hour)
- drug dosage is based on body weight - problematic with extreme body weights
- monitoring necessary to look at therapeutic response
what are pharmacokinetic considerations for dosage of IV infusions?
- high volume of distribution means that there will be a small fraction in the plasma so it will take a long time to reach a steady state
- with high volume of distribution drugs, adjusting infusions rate takes ages to change the plasma concentration
- thus a ‘loading’ bolus dose is often recommended to speed up saturation of all the components
- steady state means that infusion dosage = rate of elimination from plasma
- increasing infusion rate aims to raise plasma level and maintain equilibrium
- if elimination becomes saturated then this will lead to the accumulation of the drug and thus toxicity
why is a loading bolus dose recommended for IV infusions?
- high volume of distribution means that there will be a small fraction in the plasma so it will take a long time to reach a steady state
- with high volume of distribution drugs, adjusting infusions rate takes ages to change the plasma concentration
- thus a ‘loading’ bolus dose is often recommended to speed up saturation of all the components
what does a steady state mean for IV infusions?
steady state means that infusion dosage = rate of elimination from plasma
what does increasing IV infusion rate aim to do?
increasing infusion rate aims to raise plasma level and maintain equilibrium
what happens if elimination becomes saturated in IV infusion?
if elimination becomes saturated then this will lead to the accumulation of the drug and thus toxicity
what is the ideal drug for infusions?
- one with a small volume of distribution so it is easy to reach steady state and so that the plasma concentration is responsive to dose rate
- one which is broken down by tissue/plasma enzymes irrespective of liver and renal function
- one with an obvious and predictable dose to response relationship
- one with a low risk of toxicity and that is easy to determine the concentration of it in the plasma
what are features of IV insulin infusion?
- insulin is infused with 10% dextrose for managing a diabetic who is nil by mouth
- there is a danger of hypoglycaemia if the insulin is not given alongside dextrose since plasma glucose can change rapidly
what are features of vacomycin IV infusion?
- type of antibiotic
- requires plasma level monitoring of peak (sample hour after drug given) and trough (sample hour before next dose) level after three doses
what are features of amiodarone IV infusion?
• anti arrhythmic drug
• has problem of returning arrhythmia when the infusion is turned off
• is a high volume of distribution drug and thus often steady state is not
achieved after 48hrs of IV infusion
what are features of heparin IV infusion?
- for emergency anticoagulation if at high risk of thrombosis
- chemical reaction works in minutes
what are features of noradrenaline IV infusion?
- alpha-1 agonist
- used as a vasoconstrictor
- given for the treatment of septic shock
what are examples of continuous/pulsatile drug delivery?
- GnRH (pituitary) when given in continuos infusion results in a contraceptive
effect - pulsatile GnRH however is a physiological fertility treatment
- desensitisation occurs with many agonist drug infusions, perhaps pulsatile delivery could avoid this
what are features of pulsatile IV gentamicin?
intermittent/pulsatile IV gentamicin results in a higher peak level than steady state infusion and thus has higher bactericidal activity
what are features of patient controlled morphine?
patient controlled morphine analgesia only requires a 1/3 of the normal IV dose if given as intermittent IM injections, this is very beneficial since there is less respiratory depression than with IV infusion
what does the peripheral nervous system consist of? what neurotransmitters are used here?
- somatic (NMJ) = voluntary = acetylcholine (ACh)
* autonomic = involuntary = ACh and Noradrenaline (NAd)
what does the autonomic nervous system consist of? what neurotransmitters are used here?
- parasympathetic - ACh
* sympathetic - NAd
what are the types of cholinergic receptors? what kind of receptors are they?
- nicotinic (nAChR) - ion channel receptors
* muscarinic (mAChR) - G-protein coupled receptor
what are the effects of ACh in the parasympathetic system mediated by?
muscarinic ACh receptors (M1, M2, M3 - most common)
what are the effects of ACh in the sympathetic nervous system?
ACh mediates the release of adrenaline (Ad) and
noradrenaline (NAd)
what are the effects of ACh in the somatic nervous system?
nicotinic receptors (nAChR) mediate the response of ACh in the somatic system at the neuromuscular junction
what receptors are found at the neuromuscular junction?
nicotinic ACh receptors (nAChR)
what is the process of ACh production, release and reuptake?
- choline acetyl transferase enzyme is required to make ACh from acetyl CoA and choline (substrate) in the neurone
- ACh is then packaged into a vesicle ready to be released when the neurone is stimulated
- after ACh has been used it is broken down in the synaptic cleft by acetylcholinesterase AChE into choline and acetate
- choline is taken up into the neurone where it can be used to make more ACh
what enzyme catalyses ACh production?
choline acetyl transferase enzyme makes ACh from acetyl CoA and choline (substrate) in the neurone
what enzyme breaks down ACh in the synaptic cleft?
after ACh is used, it’s broken down in the synaptic cleft by acetylcholinesterase into choline and acetate
what is botulinum toxin? what does it do?
- lethal toxin
* inhibits ACh release into the neuromuscular junction resulting in paralysis
what is the mechanism of action of botulinum toxin?
- uses protease to degrade vesicle proteins
* thereby preventing vesicle fusion and thus the release of ACh into the synaptic cleft
what is the use of botulinum toxin?
cosmetic and spasticity
what is a competitive nAChR antagonist?
curare
what is curare?
- competitive nAChR antagonist
- nAChR found at the neuromuscular junction
- arrow poison known as tubocurarine
- acts as a neuromuscular blockade by binding to the 2
nicotinic binding sites on the nAChR thereby preventing the
binding of ACh
what is the effect of curare?
- results in a muscle relaxant effect
- can result in being conscious but aware of pain and paralysed
how can the effect of curare be reversed?
can reverse the block using sugammadex which is a selective relaxant binding agent that encapsulates the blocker thereby preventing it from binding