L2: Lead optimisation Flashcards

1
Q

what is the goal of lead optimisation? [4]

A

improve properties of lead compounds by:
- improving efficacy
- improving potency
- reduce adverse effects and toxicity profile
- is the NCR suitable for the intended route of administration? @stability, absorption an distribution
- improving ADME: onset and duration of action

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

key criteria in creating a LEAD series [7]

A
  1. binding/functional potency in primary assay: IC50< 100nM
  2. potency in secondary assay [cell proliferation assay]: GI50<500nM [has higher threshold for secondary assay]
  3. fulfills Lipinski’s rule of 5
  4. in vitro ADME liabilities [half likfe>60mins]
  5. synthesis in less than 10 steps
  6. multiple points of modification to allow for improving and creating LEAD series
  7. patentable
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

steps to LEAD optimisation

A
  1. chemical modifications: alter/remove functional groups using chemical synthesis and test the activity of analog [altered molecule] –> then infer role of the groups in binding and function
  2. bioisosteric replacement: substitute atoms or groups of atoms in parent molecule to produce compounds w broadly similar biological properties to the parent but have structural diversity. eg. have diff size and structure to stick into diff parts of pocket, have diff charges to interact w target
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

give an example of bioisosteric replacement

A

resveratrol. its a compound found in red food, but need to consume large amts to see beneficial effects observed in mice –> optimise it by replacing linker with phenol ring substitution–> formed compound 13G that became the new lead molecule

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

how can we improve drug absorption? [4]

A
  1. increasing H20 solubility: impt for intravenous administration and dissolution in GI tract
  2. acid base properties: affects solubility in GI tract. physiological range is pKa 1.5-8
  3. lipophilicity: influences ability of small molecules to pass through lipid bilayer
  4. molecular size: smaller -> BETTER! shld be<500Daltons
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

formula for absorption:

A

solubility x permeability

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

which is the most convenient and cost-effective drug administration method?

A

oral administration. patient can consume at their own convenience.

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

does form of drug affect rate of dissolution?

A

yes. tablet, capsule, suspension and solution etc. solution is fastest.

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

what limits drug absorption?

A
  1. Drug: lipophilicity and extent of ionisation of drug
  2. External factors: what food is ingested with drug
  3. Clearance: may pass through portal vein and enter liver –> metabolised
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

how to ensure stability of drug?

A

observed at different pH and temperatures

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

state an example in which stability and solubility plays a part in drug absorption.

A

gemcitabine. active drug impairs DNA replication and induces apoptosis. but we create a prodrug version that is not soluble in low pH [in stomach]. prodrug promotes oral-mediated absorption of gemcitabine w less toxicity. at higher pH [in intestine], lower hydrolysis of drug and increased solubility of drug

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

describe what area under curve represents

A

the total amt of drug in circulation. plot drug conc in plasma against time

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

absorption affects bioavailability. T/F?

A

True

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

formula for bioavailabity

A

[AUC of test x dose of i.v] / [AUC of i/v x dose of test] x 100%

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

how are drugs distributed in the body?

A

via blood stream. diff drug conc are attained in diff tissues/organs. a drug may be preferentially distributed to target organ or not at all.

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

recap: how are drug cleared from the body?

A

via metabolism in liver [for more lipophilic drugs to be converted in polar metabolites] or excretion in kidneys [drug remains unchanged]

17
Q

drug is cleared only through liver and kidneys. T/F?

A

False. can be by perspiration etc

18
Q

recap: describe the process of metabolism.

A

Phase 1: functionalisation by CYP450 enzymes –> these enzyme changes the functional grps through monooxygenase and drug loses activity eg. Paclitaxel converted to inactive metabolite

Phase 2: conjugation
- addition of highly polar conjugates to allow for rapid excretion
eg. SN-38 [active metabolite of irinotecan] inactivated by UGTs which adds glucuronic acid

19
Q

how do we observe/know ADME properties of HIT/LEADS?

A

conduct in vitro ADME assays
1. eg. using microsome metabolism
process: add animal/human liver microsome and lead drug candidate into plate and incubate –> analyse % of parent drug remaining at diff timepoints using LC/MS, compare with control [addition of a substance that we know wont metabolise as quickly]

  1. PAMPA assay [parallel artificial memb permeability assay]
    Process: drug put in donor well and have to pass through lipid memb into receiver well. measures amt of drug in donor/receiver well after certain timepoint
20
Q

after a round of lead optimisation, what shld be done?

A

perform secondary assays in vitro studies on cells isolated from disease tissue grown in monoculture and in vivo.
-we want to do as many expts to find the BEST drug possible
- types of assays: eg. gene expression, proliferation, motility for anti-cancer drug–> assay depends on function of drug
- good to conduct expts in 3D models aka organoid spheroids

21
Q

why do we have to do in vivo expts in animal based models?

A
  • required to evaluate efficacy and toxicity of drug
  • can validate in vivo biomarkers for drug efficacy
  • allow for in vivo evaluation of PK/PD in normal and disease animal models
  • can analyse histology, tissue sample [RNA/DNA/protein]+ in vivo imaging of disease progression
22
Q

human chimera mice alws have lower metabolism than human chimera mice. T/F?

A

False, may not alws be the case

23
Q

why is lead optimisation an iterative process?

A

starts with synthesis/chemical modifications –> test in vivo and in vitro [check for PK/PD, metabolism, potency etc] –> restarts cycle if not optimal