Therapeutic Concepts Flashcards

Week 6 - Monday (30th September 2024)

1
Q

What are the key drivers of drug discovery and development?

A
  • Scientific curiosity and advancement
  • Need for new treatments
  • Profitability
  • Socioeconomic aspects
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Scientific curiosity and advancement

A
  • Understanding disease biology
  • Identifying drug targets
  • Advances in screening, genomics, and molecular modelling
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Need for New Treatments

A
  • Unmet Medical Needs
  • Improving Standards of Care
  • Addressing resistance to current therapies
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Profitability

A
  • Blockbuster potential
  • Patents and exclusivity
  • Pricing and reimbursement environment
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Socioeconomic aspects

A
  • Global needs
  • Neglected diseases
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Types of macromolecules

A
  • Proteins
  • Nucleic acids
  • Polysaccharides
  • Lipids
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Importance of 3D structures in drug design

A
  • Information about the molecular basis of disease
  • Binding sites
  • Protein interactions
  • Target flexibility
  • Ligand binding and unbinding kinetics
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Techniques using 3D structures of macromolecules

A
  • Molecular docking
  • Structure-based drug design

Use 3D information to optimise drug-target interactions and enhance specificity

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

Disease-associated proteins and pathways

A
  • Cancer is caused by mutations in oncogenes or tumour suppressor genes that regulate cell growth and division.
  • Alzheimer’s Disease is linked to the accumulation of beta-amyloid plaques.
  • Imatinib: Targets the Bcr-Abl fusion protein involved in the growth of leukaemia cells and is highly effective in treating chronic myeloid leukaemia (CML).
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Nucleic acid targets

A
  • DNA-targeting drugs work by damaging DNA
  • mRNA-targeting drugs work by blocking the production of specific proteins involved in disease
  • Doxorubicin (ionically) binds to the DNA backbone and disrupts cell replication
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Membrane receptors and cell surface proteins

A

Tiotropium is a bronchodilator that inhibits the interaction between acetylcholine and the muscarinic acetylcholine receptor

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

Needs and opportunities for new therapeutic agents

A
  • Advances in biotechnology
  • Existing drugs may have side effects/toxicity
  • Need for personalised medicine due to genetics/lifestyle factors
  • Investments in R&D drive innovation
  • Greater understanding of disease mechanisms
  • Drug resistance
  • Improved screening methods
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Socio-economic factors

A
  • Developed countries: This market can afford new drugs and companies can recover the cost of DD—disease examples: migraine, depression, obesity etc.
  • Developing countries: Less R&D carried out due to lack of funding
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Therapeutic effect (definition)

A

Therapeutic effect is the intended, positive impact a drug has on the body, it is an endpoint clinicians want to achieve by prescribing a particular medication.

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

Concepts influencing therapeutic efficacy

A
  • Efficacy: Tested in clinical trials
  • Selectivity: Selective drugs are typically more effective and less toxic than non-selective drugs
  • Pharmacokinetics (ADME): How the drug is absorbed, distributed, metabolised, and excreted by the body
  • Pharmacodynamics: The molecular, biochemical, and physiological effects of the drug + mechanism of action
  • Toxicity: Acute toxicity can be life-threatening, while chronic toxicity can lead to serious health problems, such as organ damage
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Target identification and validation

A
  • Identification: using genomic, proteomic, and bioinformatics data
  • Validation: genetic screens, expression profiling, and functional assays
  • Assess druggability: evaluate target binding sites, ligand interactions, assay feasibility
  • Prioritisation: based on relevance, tractability, and novelty.
17
Q

Hit and lead compound identification

A
  • High-throughput screening
  • Natural products
  • Computational virtual screening
  • Structure-based DD
  • De novo DD
  • Fragment-based approaches
  • In silico screening
18
Q

High-throughput screening

A

Automated biochemical or cell-based assays to test large libraries of compounds

19
Q

Natural products

A

Screening substances from microbes, plants, and animals

20
Q

Computational virtual screening

A

Docking virtual libraries against a target structure to select candidates. Assessing ADMET early provides data on absorption, distribution, metabolism, excretion, and toxicity.

21
Q

Structure-based drug design

A

Utilises detailed 3D structural data on the target, often obtained by X-ray crystallography or cryo-EM, allowing for the rational design of complementarily-shaped small molecules to target binding pockets.

22
Q

De novo drug design

A

Employs computational modelling to create novel compounds predicted to bind with high affinity

23
Q

Fragment-based approaches

A

Screen low molecular weight fragments that bind weekly on their own but can be linked or grown into potent leads

24
Q

In Silico screening

A

Virtual docking of molecular libraries against target structures

25
Q

Lead optimisation

A
  • Structure-activity relationship: understanding how modifications to a molecule affect its potency and selectivity
  • ADME optimisation: improving the drug’s bioavailability, stability, and half-life, while minimizing toxicity
26
Q

Preclinical studies

A
  • In vivo and in vitro
  • Key studies: cell viability, enzyme kinetics, toxicology, pharmacokinetics, and bioavailability
27
Q

Phase I clinical trials

A
  • Safety and pharmacokinetics testing in healthy volunteers (20-100 people).
  • ADMEs are characterised to inform appropriate dosing.
  • Many drugs fail at this stage due to toxicity or poor pharmacokinetics.
  • Typical duration: 1-2 years.
28
Q

Phase II clinical trials

A
  • Efficacy testing in patients (100-500 people)
  • Short-term side effects and dosing/timings are assessed
  • Does the drug have pharmacological activity in real patients vs. healthy volunteers
  • Typical duration: 2-3 years
29
Q

Phase III clinical trials

A
  • Large-scale trials (1000-5000 patients)
  • Confirm efficacy and monitor safety by comparing the drug to placebo or standard of care
  • Frequently double-blinded, randomised, multiple centres/locations
  • Failure can occur due to a lack of efficacy or safety issues
  • Typical duration: 2-4 years
30
Q

Post-Approval Monitoring and Pharmacovigilance

A
  • Post-release monitoring for adverse effects.
  • Pharmacovigilance to detect rare side effects that may not have been apparent in clinical trials.