lecture III: drug screening & discovery Flashcards

1
Q

Historical drug discovery

A

Natural medicines, previous drugs were known, then refined.
→biological effect was discovered before target validation

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

Historical drug discovery steps

A
  1. Biologically (clinically) observed agent
  2. Bioassay
  3. Biological hit
  4. Lead optimization
  5. Preclinical & clinical studies
  6. Drug
  7. Medically validated target
  8. Better Drugs → high success rate!
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Target-based drug discovery

A

“Genes-to-drugs”

  • genome sequencing would reveal new targets
  • main issue: many targets are not biologically validated
  • drugs can be developed, but they don’t work in vivo
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Target-based drug discovery steps

A
  1. Molecular genetics (target selection)
  2. Target-based HTS assay
  3. Target hit
  4. Lead optimization
  5. Preclinical and clinical studies
  6. Drug → high failure rate :(
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Phenotype-based drug discovery

A

Return to biology first!

  • one specific protein is not targeted, but rather a certain phenotype
  • not really interested on how the drug works, but as long as you get the desired phenotype, then you go with it
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Phenotype-based drug discovery steps

A
  1. Human cell systems
  2. Biological hit
  3. Lead optimization
  4. Preclinical and clinical studies
  5. Drug
  6. Medically validated target
  7. Better drugs → high success rate!
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Cell-based assays

A
  1. Immortalized cell lines
    →cancerous, human embryonic kidney, HEK293 cells, HeLa
  2. Primary cell culture from tissue
  3. Induced pluripotent stem cells (iPSC)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Biosafety cabinet

A

Allows work in a sterile environment

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

Human iPSC

A

iPSC are derived from skin or blood cells that have been reprogrammed back into an embryonic-like pluripotent state that enables the development of an unlimited source of any type of human cell needed for therapeutic purposes.

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

Human iPSC types

A
  1. from healthy subjects
  2. from disease patients
  3. from diseased patient genetically reversed to healthy (isogenic)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

iPSC target-based screen read-out

A

target protein specific

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

iPSC phenotype-based screen read-out

A

increase/decrease of a phenotype

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

Zebrafish assay

A

A type of in vivo assay where Zebrafish can be used to assess the toxicity of drug candidates in early screening assays, sometimes in a high-throughput manner.

→can be target-based or phenotype-based
→advantage: whole system

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

High-throughput screening (HTS)

A

High-throughput screening (HTS) is a drug discovery process that allows automated testing of large numbers of chemical and/or biological compounds for a specific biological target, for example through binding assays.

→issue of false positive hits
→“fail fast, fail cheap”

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

Key elements for developing a HTS screening assay

A
  1. Quick, simple, robust, relevant
  2. Positive read-out
    →i.e. fluorescence increases upon drug binding
  3. Direct read-out
    →as little handling as possible
  4. Good dynamic range
    →good z factor
  5. Proper model: in vitro / in vivo
  6. Cost effective

→hopefully you can then identify a lead compound!

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

Examples of phenotype-based screens (lecture)

A
  1. ZIKA virus decreases CAS3 (responsible for apoptosis), but with addition of drug, CAS3 inhibition decreases.
    →read-out: CAS3 inhibition
    →not the best as it is NOT a positive read-out
  2. Lipid droplets in AD. ApoE4 expressing cells have more lipid droplets as compared to ApoE3.
    →read-out: number of lipid droplets per cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Consideration before a target-based screen

A
  1. Evidence that modification of the POI will cure the disease
  2. Evidence that the target is drugable
  3. Evidence of selectivity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Consideration before a phenotype-based screen

A
  1. Evidence that the phenotype will cure the disease
  2. Relevance of the phenotype for the disease
  3. Relevance of cell type and stimulus for the disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Considerations before a drug screen (!!)

A

Choose the disease and the clinical outcome you anticipate (cure or amelioration). Is there a target? Multiple? Which one do you select?

  1. Evidence that a modification will cure the disease
    →use KO mice, literature review, human mutations
  2. Evidence that the target is drugable
    →consider similar pharmacological compounds that may need optimization
  3. Evidence of selectivity
    →expression pattern of protein, unique phenotype/cellular pathway, localization
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

HTS: positive hit range

A

Positive hits are active in the range of 30 μM - 1 nM

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

NME

A

New Molecular Entity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

NME success order: first-in-class drug

A
  1. Phenotype-based screening (37%)
  2. Biologics
  3. Target-based screening
  4. Modified natural substances
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

NME success order: follower drug

A

1.Target-based screening (51%)
2. Biologics
3. Phenotype-based screening
4. Modified natural substances

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Best drug options: overall

A
  1. Synthetic small molecule
  2. Biologic
  3. Natural substance
  4. Natural product
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

Best drug options: phenotype-based screens

A
  1. Synthetic small molecule
  2. Natural product
  3. Natural substance
  4. Biologic
26
Q

Best drug options: target-based screens

A
  1. Biologic
  2. Synthetic small molecule
  3. Natural substance
  4. Natural product
27
Q

Library selection: Natural world

A

Natural sources have some form of biological activity
→serve as lead compound
→usually have complex structures
→EX: cocaine, digoxin, quinine, nicotine, muscarine, penicillin

  1. Plants
    →trees, flowers
  2. Microorganisms
    →fungi, bacteria
  3. Animals
    →frogs, snakes
  4. Biochemicals
    →neurotransmitters, hormones
  5. Marine chemistry
    →corals, bacteria, fish
28
Q

Library selection: Synthetic world

A

Synthetic libraries contain compounds synthesized by company where the starting point may have been a natural substance.

  1. Chemical synthesis
  2. Combinatorial synthesis
  3. Parallel synthesis
29
Q

Library selection: Virtual world

A
  1. Computer-aided drug design
30
Q

Natural compound library advantages

A
  1. Probably pre-selected/evolved for biological activity
  2. Serendipity
  3. Potent
  4. Cover large chemical diversity
31
Q

Natural compound library disadvantages

A
  1. Difficult to synthesize
  2. Difficult to produce, do extracts, reproducibility
  3. Bioavailability may be suboptimal
32
Q

Chemical space

A

The chemical space, a set of all possible molecular structures.

Beilstein database → 10^7

33
Q

3 phases of drug action

A
  1. Pharmaceutical phase
  2. Pharmacokinetic effect
  3. Pharmacodynamic effect
34
Q

What is the ideal administration of a drug

A

Orally! (oral bioavailability)

35
Q

Drug action: pharmaceutical phase

A
  • Dissolution of drug from pill
  • Survive gastric juices, survive food-degrading enzymes
  • Pass through gut cells (sufficient lipophilicity)
36
Q

Drug action: pharmacokinetic phase

A
  • Pass into the blood stream (sufficient hydrophilicity)
  • Distribution to tissue (every cell max. 20-30 μm away from capillary)
  • Pass between cells, through cells, transporters

→essentially, oral bioavailability!

37
Q

Drug action: pharmacodynamic phase

A
  • Drug interacts with molecular target, pharmacological effect
38
Q

Bioavailability

A

Bioavailability is a subcategory of absorption and is the fraction of an administered drug that reaches the systemic circulation.

39
Q

The Lipinski rule of 5

A

An indicator for the potential ability of a drug to be orally absorbed (bioavailability).
An orally active drug has no more than one violation of the following criteria:
1. MW≤ 500 kDa
2. HBD ≤ 5
(expressed as the sum of OH & NH groups)
3. HBA ≤ 10
(expressed as the sum of O & N atoms)
4. logP ≤ 5

40
Q

Partition coefficient

A

Measure of how much of the drug is in a hydrophilic vs. hydrophobic phase.

→ a measure of the drug’s lipophilicity
→ P = [drug in octanol] / [drug in aq. solution]

41
Q

Octanol

A

Hydrophobic organic solvent

42
Q

Oral bioavailability

A

Ability to cross hydrophobic cell membranes and be water soluble for distribution

43
Q

Which functional group do many drugs contain? What’s the purpose?

A

Amines.

→pH in different parts of the body (small intestine, stomach, blood) differ, thus you need a drug that can easily cross all of these different parts.
→amines have a pKa ranging from pH 6-8
→amines can be p+ or not depending on the environment’s pH (all parts of your body have
a pH within the amine’s pKa range, so drugs with amines are always switched between their different forms)

44
Q

Synthetic library characteristics

A

Fun! go read slide 28 :)

45
Q

Synthetic library: Parallel synthesis

A

Method to design a novel compound library.

Many wells starting with core structure and then to each well, you add a new reactant. In the second reaction, you switch up the new reactant.

→small scale
→each vial has one compound
→preferred method!

46
Q

Synthetic library: Combinatorial synthesis

A

Method to design a novel compound library.

Everything mixed in and see how it reacts with the core structure.

→small scale
→mixture of compounds in one vial
→obtain way more products

47
Q

Fragment-based lead discovery (FBLD)

A
  1. Screen for 1st ligand: Screening of low-molecular weight compounds for specific binding, then optimize.
  2. Screen for 2nd ligand: Repeat screen for second specific fragment, then optimize.
  3. Link: The two binding compounds are linked.
48
Q

Advantages of FBLD

A
  1. High level of diversity is possible
  2. Easier to synthesize
  3. Higher success probability
49
Q

Click chemistry

A

A variation of FBLD consisting of a class of biocompatible small molecule reactions commonly used in bioconjugation, allowing the joining of substrates of choice with specific biomolecules.

50
Q

How can you screen FBLD?

A
  1. NMR
  2. Bioassays
51
Q

Synthetic library advantages

A
  1. Characterized, can be synthesized
  2. Stratified for Lipinski’s rules of 5
    →higher likelihood for oral bioavailability
  3. Stratified for toxic compounds
52
Q

Synthetic library disadvantages

A
  1. Low serendipity
    →many compounds are variations on a single structural theme
  2. Limited chemical diversity
53
Q

Computer-aided drug design characteristics

A

Fun! go read slide 33 :)

54
Q

Computer-aided drug design method

A

Alphafold!!!

55
Q

Specificity

A

To cause one particular action in all biological systems

56
Q

Selectivity

A

To bind to one target, not to others

→less side effects

57
Q

Sensitivity

A

The lower the dose needed to cause an effect, the more sensitive the system is

58
Q

Structure-activity relationship: morphine & naloxone

A

Naloxone is a morphine antagonist. It was synthesized to possess the same pharmacophore as morphine, but with higher affinity, thus it will knock morphine out when introduced in our system.

59
Q

Pharmacophore of morphine

A
  1. Phenol-OH
  2. Aromatic ring
  3. Tertiary amine
60
Q

How can you test structure-activity relationships?

A
  • by systematically modifying functional groups to see their importance for biological effect
  • by resolving the structure of compound and target
  • by modelling the compound on theoretically derived Alpha fold structures
61
Q

Pharmacophore: spacial views
(draw it)

A
  1. 2D → binding groups
  2. 3D → angle between binding groups
  3. 3D → bonding type