1/34
Looks like no tags are added yet.
Name | Mastery | Learn | Test | Matching | Spaced |
---|
No study sessions yet.
what is lead optimisation?
refining chemical structure of a confirmed hit to improve its drug like characteristics
what does lead optimisation involve?
series of analogue testing
testing the series to determine the structure activity relationship → assess potency, bioavailability, stability, selectivity etc
what is analogue synthesis?
work with all the information you have (e.g. x ray crystallography) to produce further active compounds
develop the pharmacophore
how was ziprasidone 5-HT2 antagonist developed?
known ligand for serotonin receptors taken, pharmacophore developed → screening showed poor in vivo efficacy → was optimised to ziprasidone
how many optimisation cycles are required?
might have to go through a number of cycles
what are some things to keep in mind to aid lead optimisation? (6)
start with a good lead → not always the most potent/active but rather has a good balance of MW, logP, potency, activity etc
chemistry is important → tractability, diversification, steps involved and how easy it is to make analogue
consider logP
insertion of heteroaromatic groups will increase polarity :. better drug like properties
biosiosteres (= functional groups that behave similarly within biological system)
structural simplification → don’t want unnecessarily complex structures, also reduces MW
what is lipinski’s rule of 5?
MW less than 500 Da
not more than 5 H bond donors
not more than 10 H bond acceptors
LogP value less than 5
how useful are lipinski’s rules?
for assessing bioavailability of oral drugs in humans :. measure of drug likeness
however, many commercial drugs break these rules e.g. antibacterials (because they target bacteria, not human cells)
good for considering final drug product, however there are better rules available for hit to lead activities stage
why is H bond donors and accceptors important for Lipinski’s rule of 5?
too many H bond donors and acceptors make desolvation (partitioning into membrane after dissolving in water) difficult :. affects absorption across gut membrane
donors have more influence than acceptors
why is cLogP important for Lipinski’s rule of 5?
lipophilic compounds have poor aqueous solubility :. poor absorption
lipophilic compounds have more interactions with CYP enzyme system
if >5 = poor absorption because compounds do not partition out of membrane into aqueous cellular environment
why is molecular weight important for Lipinski’s rule of 5?
if >500, absorption may be affected since larger molecules are harder to cross membranes :. limited diffusion
opportunity of modification is very limited
selectivity issues and higher conc of drug required due to absorption issues :. off target toxicity many be observed
give an example of where balancing lipophilicity was important for lead optimisation
olanzapine (antipsychotic) → developed from clozapine
clozapine = higher logP value (3.7) :. was not very water soluble
olanzapine altered its structure so it made the phenyl ring into a thiophene ring which improves polarity :. improves solubility
due to this, bioavailability went from 55% in clozapine to ~100% in olanzapine
daily dosing regimen decreased due to more exposure to drug
why are lipophilic drugs often known as being promiscuous?
promiscuous = many off target side effects
lipophilicity can often increase affinity to target but also increase off target interactions
hydrophobic interactions are less specific than polar interactions
what can you do to try reduce the promiscuity of lipophilic drugs?
maintain logP less than 3
how is drug safety considered in lead optimisation?
before clinical studies in humans, it is necessary to demonstrate safety in vitro and in vivo
what assumptions about drug safety are used in in vitro and in vivo safety testing?
in vitro assays predict in vivo effects
effects of chemicals in laboratory animals apply to humans (extrapolate effects)
use of high doses in animals is valid for predicting possible toxicity in humans
these assumptions are broadly true but we cannot be certain that a chemical will show no toxic effects in humans
what are the 5 sources of toxicity?
mechanism based pharmacology (toxicity linked to target activation)
formation of reactive metabolites
activation of other receptors e.g. hERG
interactions with other substances
idiosyncratic (= an unpredictable, characteristic, adverse reaction of an individual,) toxicity
how can toxicities be avoided?
making a very potent compound
dose required will decrease
this concept does not apply to mechanism based since this is to due to target activation and more potent drugs will allow tighter binding to target
when can mechanism based pharmacology toxicity be a big problem?
for drugs designed for completely novel targets rather than new drugs for a known mechanism
usually not predictable from in vitro tests, can sometimes predict from animal models
give an example of mechanism based pharmacology toxicity
beta agonists (e.g. salbutamol) → activation of B2 receptors in the lunch causing dilation of airways
inhalation :. most of the drug stays in the lungs
if patient takes too many doses, the levels in systemic circulation rise :. can act on B2 receptors in the heart causing palpitations
give an example of toxicity caused by the formation of reactive metabolites
paracetamol
phase 1 oxidation causes production of NAPQI
NAPQI normally reacts with glutathione :. becomes more polar and excreted from urine
in toxicity, NAPQI reacts with proteins causing toxic effects→ hepatotoxicity
what type of electrophile is glutathione?
scavenger electrophile → reacts with reactive metabolites and removes them from the system
how can you avoid toxicities from occurring due to reactive metabolites?
avoid functional groups known to show reactive metabolites
‘Ames’ test to detect mutagenicity
how can you test for functional groups to show reactive metabolites?
look for binding to proteins or glutathione (detect by mass spec)
how does the Ames test work?
use a genetically modified bacterium that cannot synthesise histidine
expose bacteria to drug
if the bacteria mutates and regains the ability to produce histidine, they will form colonies on an agar plate
number of colonies is compared to a control group to determine if the substance causes an increase in mutations
can also be carried out in the presence of liver enzymes to simulate how the substance is metabolised in the body
describe how you can avoid activation of other receptors/enzymes i.e. off target toxicity
screen against other systems
before nomination to preclinical studies, the compound will be tested in many other assays in vitro to look for activity
potency is important to have a safe therapeutic window
if you can increase potency at the target receptor, a lower dose is needed :. lower risk of off target side effects
why is mimicking hERG interaction important during lead optimisation?
every drug is screen for this according to MRHA regulations
hERG = potassium cardiac channel
activation causes prolongation of QT wave (hyperpolarisation)
can lead to fatal arrhythmias (irregular heart heat)
what is the pharmacophore for hERG?
aromatic ring
alkyl spacer
basic centre
drugs are screened for activity based on this (all basic aromas)
give an example of a drug that activates hERG and how this activity was reduced
farnesyltransferase inhibitors
changes lipophilic aromatic ring to a polar one reduces hERG activity by >10x
why is the cytochrome P450 system important?
majority of top 200 drugs are primarily metabolised by CYP450 enzymes
:. compounds which inhibit/induce CYPs have potential to interact with many other drugs
give an example of a drug that causes hERG toxicity
terfenadine
now banned → was on the market for OTC hayfever treatment
found to cause life threatening cardiac arrythmias when co-administered with e.g. erythromycin
other drug e.g. erythromycin causes inhibition of CYP450 → prevents metabolism of terfenadine :. [drug] in blood increases :. can bind to hERG :. arrhythmia
which PK parameters are screened during lead optimisation? what are these useful for?
Cmax , Tmax , half life, volume of distribution, clearance
useful to optimising onset of action, duration of action and bioavailability
what are the concepts of being proactive vs reactive in selecting a pre-clinical candidate?
proactive: deal with issues before they arise
reactive: solve issues that arise in later stages of development
why is it harder to make modifications after selecting a pre-clinical candidate?
already carried out screening/testing earlier :. modifications would require repeats of these tests which isn’t preferable
what are the criteria for preclinical candidates?
preferred crystalline form identified
compound sufficiently stable to allow a shelf life of >2 yrs
scale up of lead compound to 100g demonstrated (ned this much in animal studies)
full PK and metabolite profiling in 2 species (1 lower species e.g rodents and 1 higher species e.g. primate/dog)
predicted human half life and dose
no toxicity in extended animal study