IMC 09: Drug Discovery and Development Flashcards

1
Q

Pharmacodynamics

What is target inhibition?

A

blocking the activity of the biological binding partner for the drug/inhibitor

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

Pharmacodynamics

What are on-target effects?

A

pharmacologic effects caused by a drug interacting with its intended biological target

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

Pharmacodynamics

What are off-target effects?

A

pharmacologic effects caused by a drug, but not due to binding of its intended biological target

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

How are most new drugs produced (the processes)?

A

from high-throughput screening and subsequent optimization

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

What is high-throughput screening?

A

vast libraries of chemical compounds can be screened in high-throughput assays to identify inhibitors that may have therapeutic benefit

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

What is an assay?

A

analysis of the biochemical/biological potency of a chemical species

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

What are targeted assays/screens?

A

seek agents that bind to a specific biological target (protein) to alter its function

  • typically done in a biochemical (cell-free) setting
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What are phenotypic assays/screens?

A

seek agents that induce a particular biological effect in a model system

  • may be in a cellular assay, animal model, or other biological setting
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What are the steps of the drug discovery and development process?

A
  • target validation
  • hit identification and optimization
  • clinical candidate
  • clinical testing – phase I-III
  • FDA approval
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What is target validation?

A

identify promising strategies for drug intervention

  • will a drug against this target have the desired therapeutic effect
  • may include identifying a single protein target, targeting a process or pathway, using a disease model, etc.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What is hit-to-lead development?

A

transform compounds from screening ‘hits’ into promising candidates for clinical testing (leads)

  • role of medicinal chemists
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What is a hit compound?

A

chemical compound that shows promising activity in a high-throughput screen, but has not been chemical optimized using medicinal chemistry

  • represent starting points for medicinal chemistry studies
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What is hit expansion?

A

helps identify functional groups on a hit compound that could be modified to improve activity

  • modify molecule in different directions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What is a lead compound?

A

chemically optimized compound that has high affinity for its target and displays favourable activity in cells and pharmacokinetic models

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

What is a clinical candidate?

A

compound ready for clinical testing, with optimized efficacy and safety testing in animal models

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

What are structure-activity relationship (SAR) studies?

A

how changes to the chemical structure of a molecule affect its activity

  • can also introduce modifications that alter other properties of a drug molecule: decrease undesired binding to other proteins, increase metabolic stability, decrease toxicity, improve solubility, improve membrane permeability
  • balancing drug potency with other parameters such as metabolic stability, selectivity, and biological activity can be challenging
  • studies entail several rounds of modifications on multiple sites of a compound
17
Q

Rotatable Bonds

A

18
Q

Identify the strategies that can be used to improve the activity and efficacy of drug molecules.

A

structural modifications

  • hydrophobic expansion
  • isosteric replacement

in silico modelling and co-crystal structures

19
Q

What is hydrophobic expansion?

A

increasing hydrophobic attachment (chain length) to increase compound’s activity

  • enzyme active sites are generally hydrophobic compared to the surrounding environment
  • medicinal chemists can probe for hydrophobic interactions by increasing alkyl chain length – this also changes physicochemical properties of the drug molecule including solubility and logP
20
Q

What is isosteric replacement?

A

switching out different isosteres can alter drug activity and change physicochemical properties

21
Q

What is an isostere?

A

chemical group that can approximate the physical and chemical properties of another chemical group

  • isostere is a more specific term than mimetic – mimetic may refer to the entire drug molecule, whereas isostere would refer to a specific group
22
Q

What is a classical isostere?

A

two comparable groups with the same valency (number of attachment points)

23
Q

What is a bioisostere?

A

chemical group that can replace another chemical group without (significantly) affecting the biological activity of the drug

  • are typically less than exact structural mimetics, and are often more alike in biological rather than physical properties – therefore an effective bioisostere for one biochemical application may not translate to another setting, necessitating the careful selection and tailoring of an isostere for a specific circumstance
24
Q

In Silico Modelling and Co-Crystal Structures

What are protein-ligand docking strategies?

A

aim to position chemical compounds into a binding site on a protein and evaluate the strength of interaction (computer program)

  • designed compounds within hit-to-lead development stages can also be docked prior to their synthesis
25
Q

In Silico Modelling and Co-Crystal Structures

What is in silico screening?

A

massive libraries of chemical structures (> 1 million compounds) can be quickly screened for interactions with protein crystal structures, using several different software options

  • hit compounds from in silico screening campaigns must be validated in biochemical and cell-based assays
26
Q

In Silico Modelling and Co-Crystal Structures

What can protein crystal structures show?

A

what part of the drug molecule sticks out of the active site (hydrophobic)

  • one part of active site is exposed to water, solvent, etc. in cell – where most polar or charged part of inhibitor will be
  • changing hydrophobic group exposed to water (ie. neutral at physiological pH) to hydrophilic group (ie. charged at physiological pH) increases activity – better interaction
27
Q

What is topological polar surface area (TPSA)?

A

calculated term representing the sum of the exposed areas of HBDs and HBAs of a molecule

  • generally increases proportionally with molecular weight – therefore TPSA and 500 g/mol cut off by Lipinksi’s RO5 represent similar measurements
  • highly related to HBD and HBA count
28
Q

What is the Easson-Stedman 3-point attachment hypothesis?

A

if a chiral compound interacts with 3 sites on a protein, the more potent enantiomer will be involved in three intermolecular interactions whereas the less potent enantiomer will only interact with a maximum of two sites

  • if you have two enantiomers with 3 attachment points to a protein, one enantiomer/isomer will be more potent than the other – cannot fit the mirror image on the same interaction points
29
Q

How can a chiral molecule have the same activity as its chiral isomer (breaks the Easson-Stedman 3-point attachment hypothesis)?

A

only if it makes 3 attachment points

  • if it only makes 2, can have the same interaction with its mirror image