AR - Drug Discovery & Development I Flashcards
(13 cards)
Q1: What are the four main stages in therapeutic drug development? (4)
- Discovery & Development – Identify potential drug compounds and evaluate their basic properties.
- Preclinical Research – Lab and animal testing to assess mechanism of action, efficacy, PK, and toxicity.
- Clinical Research – Human testing in Phases I–III to evaluate safety and effectiveness.
- Regulatory Review & Post-Marketing Surveillance – Agencies (FDA/MHRA/EMA) assess drug for approval and continue monitoring after market launch.
Q4: What is LADMET and what does it represent in drug development? (6)
- Liberation – Drug release from dosage form.
- Absorption – Entry into bloodstream.
- Distribution – Movement to tissues/organs.
- Metabolism – Chemical breakdown (mainly in liver).
- Excretion – Elimination via urine/feces.
- Toxicity – Potential harmful effects
Q3: What are key research questions addressed during drug development? (8)
- ADME – How is the drug absorbed, distributed, metabolised, and excreted?
- Therapeutic benefit and proposed mechanism of action.
- Optimal dosage and administration route.
- Adverse effects and toxicity profile.
- Population-specific responses (gender, race, ethnicity).
- Drug-drug interactions.
- Comparative effectiveness vs. other therapies.
- Pharmacokinetics and pharmacodynamics (PK/PD) models
Q2: How are new drug candidates typically discovered? (4)
- New insights into disease mechanisms guide rational design.
- High-throughput screening of compound libraries for activity.
- Repositioning existing drugs with unexpected benefits.
- Emerging technologies such as gene editing or targeted delivery tools.
Q5: What must be demonstrated during preclinical studies before human trials? (6)
- Mechanism of Action (MOA)
- Efficacy in disease-relevant animal models
- Pharmacodynamics – effect of drug on body
- Pharmacokinetics – what the body does to the drug
- Toxicity – adverse effects, LD50, organ damage
- Compliance with GLP (Good Laboratory Practices)
Q6: What practices are required under GLP for preclinical research? (8)
- Clear study protocols and objectives
- Qualified personnel
- Standardised facilities and equipment
- Defined SOPs (standard operating procedures)
- Accurate study reports and documentation
- Quality assurance systems
- Ethical animal care practices
- Use of both in vitro and in vivo models
Q7: Why are animal models essential in biomedical research? (6)
- Animals share >95% of genes with humans (e.g., mice).
- Mimic human physiology and organ systems.
- Suffer from similar diseases (e.g., cancer, asthma, infections).
- Essential for developing vaccines, antibiotics, and surgeries.
- Enable study of PK, efficacy, and long-term toxicity.
- Ethically regulated, often used with in vitro/computer models.
Q8: What are major breakthroughs made possible through animal studies? (5)
- Herceptin (breast cancer) – developed from mouse antibodies.
- HAART for HIV – enabled by animal testing.
- Insulin therapy – tested in rabbits and dogs.
- Tamoxifen – reduced breast cancer mortality by 30%.
- Vaccines – polio, HPV, meningitis, etc.
Q10: What happens in Phases I–III of clinical research? (6)
- Phase I – Test safety, dosage, and PK in 20–100 healthy volunteers.
- Phase II – Assess efficacy and side effects in 100–500 patients.
- Phase III – Confirm results in 1,000–5,000 patients; compare with existing therapies.
- All phases follow GCP (Good Clinical Practice).
- Trials include controls, randomisation, and blinding to reduce bias.
- All data used for New Drug Application (NDA) submission.
Q9: What must occur before moving a drug to clinical trials? (4)
- Submission of IND (Investigational New Drug) application.
- Design of clinical protocols (phases, dosing, endpoints).
- Define eligibility criteria and sample sizes.
- Approval from regulatory authorities (FDA, MHRA, EMA).
Q12: Why do many drug candidates fail or slow down in development? (7)
- Poor pharmacokinetics (e.g. low bioavailability).
- Toxicity issues identified in preclinical or clinical stages.
- Lack of efficacy in target population.
- Market-related concerns (cost, competition).
- Synthetic complexity – hard to manufacture.
- Ambiguous toxicology findings.
- Target has limited therapeutic window or disease is too complex.
Q11: What information is included in a New Drug Application (NDA)? (5)
- Full clinical trial data from Phases I–III.
- Reports on toxicity, pharmacokinetics, and efficacy.
- Proposed labeling and usage instructions.
- Safety updates and drug abuse potential.
- Patent, compliance, and manufacturing details.
Q13: What was the TGN1412 disaster and what did it teach us? (3)
- TGN1412 – anti-CD28 superagonist tested in humans in 2006.
- Caused cytokine storm in all 6 volunteers at Northwick Park, UK.
- Highlighted the critical gap between animal models and human immune response – led to updated regulations for first-in-human trials.