L1: Target selection; Lead Identification Flashcards

1
Q

Describe the 4 stages of Drug development process

A
  1. Recombinant systems: protein-drug interaction
  2. Cell systems: interaction in natural cellular environment
  3. in vivo: interaction in complex biological system
  4. clinic: interaction in human system under controlled pathological environment
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

state the 3 types of NEW drugs

A
  1. new drug for NOVEL use [new approach or disease]
  2. new drug that is a new generation of present drug
  3. new drug that is another variation of known drug [me-too drug]–> similar to present drugs but different enough to be patented
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

what are the different approaches to target selection?

A
  1. genomic approach
  2. proteomics approach
  3. RNAi screens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

what is genomic approach in target selection? give one example

A

analysing entire genetic information of organism in healthy vs disease state –> see how genome correlate to phenotype

eg. chronic myelogenous leukemia (CML): chr translocation that forms fusion gene BCR-Abl-> constitutive activated Abl kinase

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

what is proteomics approach in target selection? give one example

A

separation and characterisation of proteins in organism: compare protein expression in healthy vs disease state

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

problem with using genomic and proteomics approach in target selection?

A

just bc gene/protein is expressed differently does not mean it cause disease ==> NEED TO VALIDATE using RNAi screeen

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

how can RNAi screen validate target selection?

A

specific indiv genes are knocked down to find genes that regulate key disease processes ==> identify potential targets by function

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

limitations of RNAi screens

A

limited to studies in vitro [cell lines]

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

how do you validate your targets?

A

use in vitro assays and in vivo assays [BOTH REQUIRED]
In vitro: engineer cell lines with gain/loss of function using siRNA or shRNAi –> see ROLE of target
In vivo: usually use transgenic model and conduct loss/gain in function expts
- loss: use shRNA on tumour growing mouse
- gain: over express gene in normal mice

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

what is a HIT?

A

a compound that INTERACTS with chosen target at given conc [usually in micromolar range]

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

what is a LEAD? [3]

A

A compound with drug-like properties and initial structure-activity relationship and a promising IP position

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

what is a preclinical development candidate? [3]

A

a New chemical entity with optimised pharmacological and pharmacokinetic properties and secure IP position

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

methods of LEAD identification

A
  1. rational drug design
  2. high-throughput screening (HTS): cell-free or cell-based
  3. Fragment-based screening (FBS)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

elab on rational drug design

A

first identify potential HITS using interaction between drug and target –> require coordination between structural biology and organic chem and design drug based on pharmacophore of endogenous ligand/substrate

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

elab on high-throughput screening. what it is and methods.

A

uses either cell-free or cell-based assays. ideally on isolated target molecule aka cell-free. target-specific effects are measured quantitatively by reporter assay eg. fluorescence, luminescence, cell shape

Cell free:
- specific target is bound to antibody on plate + substrate added
- drug added afterwards to see if reaction is inhibited

Cell-based:
- cell+drug and look for reporter gene reaction –> problem: dont know the exact point in pathway that drug is acting on + difficult to do SAR without knowing exact molecular target and mode of drug interaction

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

give one example of small molecule rational drug design

A

discovery of drug for chronic myelogenous leukemia. found out that myristoylation of N terminus of c-ABL causes structural change in c-ABL–> autoinhibition of -ABL.

BCR-ABL lacks the N-terminus myristoylation site resulting in constitutive active BCR-ABL

17
Q

what is robust screening assay? formula?

A

used to assess quality of HTS assays. measured using z’. z’ ranges between 0-1, the higher the value of z’, the more robust the assay [>0.6 is acceptable].

18
Q

what does higher z’ score mean?

A

more robust the assay–> allows for wider range of identifying new drugs

19
Q

state the pros of rational design [1]

A

it is a focused analysis of drug candidates

20
Q

state the cons of the rational drug design [3]

A
  1. done on a limited set of candidates –> info gathered on a few drugs only
  2. not predictive of favourable pharmacological properties-> identified HITS may not be real HITs
  3. requires extensive knowledge of target
21
Q

state the pros of high throughput screening [2]

A
  1. real data that can identify competitive or allosteric inhibitors from 1 screen [from cell-free assay]
  2. eliminates compounds that are unstable or cannot access cellular target
22
Q

state the cons of HTS [2]

A
  1. requires screening of many thousands of candidates [more costly and time consuming]
  2. labour intensive
23
Q

describe fragment based screening

A

instead of identifying structures that bind really well to target, we find only those that bind decently –> can collate info on how diff interactions contributes to binding –> use structured based design and interaction to design drug with explored structures

24
Q

give one example of drug made with fragment based screening

A

novel stat3 inhibitor.

25
what are the common strategies in creating a compound library? [3]
1. acquisition from external vendor 2. generation form chemical library synthesis [random or focused libraries] 3. generation from medicinal chemistry efforts [targeted synthesis or combinatorial synthesis]
26
what should a good compound library be like? [2]
1. large and diverse 2. containing only lead-like or drug-like compounds that are: - non reactive - no known toxic moieties - follow Lipinski's rule of 5 - aqueous soluble
27
examples of compound libraries
1. FDA-approved drugs 2. natural product libraries
28
what is Lipinski's rule of 5? [4]
1. drug shld have fewer than 5 H bond donors 2. shld have fewer than 10 H bond acceptors 3. molecular weight of <500 Daltons 4. a partitioning coefficient (logP) of less than 5
29
how can lipophilicity be measured?
usually using logP, which is the log of conc of drug dissolved in lipophilic organic phase/octanol vs polar aqueous phase/water
30
high logP of >5 means ?
drug is highly soluble in lipids/organic solvents --> insoluble in water.
31
ideal logP at pH 7.4 is ____
between 1-3 as we want moderate interaction w both water and lipids
32
criteria for HIT to become a LEAD [3]
1. validate HITs with a primary assay. find those that have >75% inhibition of target 2. evaluate hits over a large drug dose conc range 3. HIT with the best inhibition at the lowest conc is the LEAD
33
drugs in compound libraries should be:
LEAD like or FDA approved drugs w the following properties: 1. not reactive 2. follow Lipinski's rule of 5 3. aqueous soluble 4. no known toxic moieties
34
when does a HIT compound become a LEAD?
1. validate HITs via primary assays to choose those that "make the cut" eg. ~75% inhibition 2. evaluate chosen HITs from ^ and compare activity over diff drug dose conc range ==> the one w highest activity aka best inhibitor at lowest dose becomes the LEAD
35
combination index values and its meaning
CI=1 additive effect CI>1 antagonistic CI<1 synergistic