Drug discovery Flashcards

1
Q

What are the stages of drug discovery?

A
  • Disease identification
  • Target identification
  • Target validation
  • High throughput screening assay
  • HIT identification
  • Primary screening assay
  • Lead identification
  • Lead optimisation
  • Animal model
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What is the drug life cycle?

A

4 stages:
- Research and discovery
- Non clinical (good lab practice)
- Clinical (good clinical practice)
- Post approval

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

Describe the disease identification.

A
  • Identify a disease that requires treatment and how is it define?
  • What therapies already exist or problems with existing therapies?
  • What are the competitors doing?
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What is a disease?

A
  • Absence of health.
  • Health: a state of complete physical, mental and social well-being and not merely the absence of disease or infirmity.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What is Target identification?

A
  • Necessary to identify a specific receptor or enzyme involved in a disease.
  • Target should be specific to diseased tissue.
  • ## Some targets are not “drug-able”.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What is Target validation?

A
  • Obtain evidence that drugs acting on a specific target can modify the disease process.
  • Transgenic animals
  • Polymorphisms/ mutations in human disease
  • Use of antibodies
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What does the pre-clinical stage entail?

A
  • Hit compound: A compound from high-throughput screen with activity on the target.
  • Lead compound: The most promising hit compound selected for further work.
  • Lead optimisation: Screening of derivatives of lead compound for improved activity.
  • Candidate compound: Best compound selected for clinical development.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What is High throughput screening?

A
  • Necessary to have assay for drug-target combination.
  • Needs to be high throughput
  • Between 10,000-1 million compounds will be screened.
  • Used in drug discovery to identify hits from compound libraries that may become leads for medicinal chemistry optimisation.
  • Cheap, easy, quick and dirty.
  • Choose criterion for selection.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What are HTS libraries?

A
  • Drug companies store every compound that they synthesise.
  • Thus, each company has their own library
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What is HIT generation?

A
  • Most active compounds from screening are selected as hit compounds.
  • Chosen for unique chemical structure
  • Potential for derivative synthesis:
    1.Easy chemistry
    2. Few patents
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Give an example of a HTS assay.

A

HTRF: Homogenous time-resolved fluorescence.

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

Fluorescence resonance energy transfer (FRET).

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

What is a primary screening assay?

A
  • Hit derivatives are tested in primary screening assay.
  • Does not need to be high throughput.
  • Needs to be more clinically relevant.
  • Dose response curves generated.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What is lead selection/ identification?

A
  • Criteria are chosen for lead compound.
  • This is close to final drug.
  • Best drug from best series of compounds.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What is lead optimisation?

A
  • The lead compound is not a drug.
  • It is usually the most potent agent discovered.
  • To be an effective drug ADME-T factors must also be considered.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What is an animal model?

A
  • Drug is tested in animal model of disease.
  • Essential that animal model is good representative of disease process.
17
Q

What is toxicity?

A
  • Minimum toxic dose determined in mice to confirm suitability of drug.
  • Possibility of in vitro toxicology on human cells.
18
Q

What is bio-availability?

A
  • Drug is tested for oral activity.
  • If necessary pro-drug is synthesised.
  • Some drugs are very active and meet all criteria except oral activity.
  • % of oral drug that reaches the systemic circulation.
19
Q

What is candidate selection?

A
  • Best drug becomes a candidate compound.
  • Drug is passed over to Development group.
  • Back-up project is then initiated.
20
Q

What is rational drug design?

A
  • Understanding receptor-ligand interactions at molecular levels can be used to design new drugs.
  • Numerous techniques used.
21
Q

What is QSAR?

A
  • Quantitative structure-activity relationship
  • Requires data from a series of compounds
  • Relates features of molecules to activity
    1. Solubility
    2. Electron density
    3. Molecular volume
  • Used to predict potential derivatives
22
Q

Example of QSAR?

A
23
Q

Discuss QSAR parameters.

A
  • A measure of the potential contribution of its group to a particular property of the parent drug.
  • MIC: minimum inhibitory concentration against E. coli
  • pi, sm, sp, F, R, ES & MR are molecular parameters
  • Best models for R1, R6, R7 and R8 established.
24
Q

What are QSAR models?

A
  • Mathematical models that can be used to predict the physicochemical, biological and environmental fate properties of compounds from the knowledge of their chemical structure.
25
Q

What are other rational methods?

A
  • Used if no lead or hit compounds available.
  • Other software allows ligand prediction if there is a crystal structure of receptor/enzyme or of a homolog.
26
Q

What is Rational drug design?

A
  1. Swiss-pdb:
    - 3D protein analysis and homology model generation-based on a protein crystal structure.
  2. Virtual screening:
    - Docking small ligands into a known 3D protein structure.
    - Usually 10 million + compounds screened
    - Rapid and inexpensive
  3. Pharmacophore generation.
27
Q

What is a pharmacophore?

A
  • A theoretical object that summarises the chemical requirements for a compound in 3D.
  • It shows where the hydrophobic regions are, where the electron donor/acceptors are.
  • One can also include parameters to force the ligand into a specific volume.
  • Pharmacophores can be generated based on the pocket of the enzyme, or on the ligand.
28
Q

What are functional groups?

A
29
Q

What is Lipinski’s rule of five?

A
  • States that an orally active drug has no more than one violation of the following criteria:
    1. No more than 5 hydrogen bond donors (nitrogen or oxygen atoms with one or more hydrogen atoms)
    2. No more than 10 hydrogen bond acceptors (all nitrogen or oxygen atoms).
    3. A molecular mass ≤500 Da
    4. log P (octanol-water partition coefficient ) ≤5
30
Q

What is the Rule-of-three for fragments?

A
  • Criteria proposed to describe fragments suitable for biophysical screening:
    1. molecular mass <300 Da,
    2. ogP≤3,
    3. Hydrogen-bond acceptors ≤3
    4. Hydrogen-bond donors ≤3.