• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/41

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

41 Cards in this Set

  • Front
  • Back
  • 3rd side (hint)

What is the typical development time of a drug from identification of a lead to approval for sales?

8-15 years.

What are the 4 major time phases of drug development? How many years (on avg.) per?

1. Pre-clinical R&D (3-7 yr)


2. Clinical trials (4-8 yr)


3. New drug application review (1-2 yr)


4. Post-marketing surveillance

What happens during pre-clinical R&D? (5 stages)

1. Disease is studied (how it works, mechanisms, etc)


2. Development of a bio-assay


3. Identification of a lead compound


4. Chemical synthesis to improve the lead and identify a viable drug candidate


5. Toxicity testing in animal models

What happens during post-marketing surveillance? (2 main things)

- Adverse reaction reporting (agencies look @ these reports to determine risk factor, validity, etc)


- Market surveys

Define IND.

Investigational New Drug

What 3 factors does Health Canada look at when considering an IND for the market?

- Manufactered under GMP


- Knowledge of the pharmacokinetics of the drug (ADME)


- Safety and efficacy (as shown in clinical trials)

What is involved in phase 1 clinical trials? What is the time frame?

Involves healthy patients (~20) and determines safety.


- 1-2 months

What is involved in phase 2 clinical trials? What is the time frame?

Involves small (<50), highly controlled group of patients; determines efficacy.


- 3 months - 1 year


What is involved in phase 3 clinical trials? What is the time frame?

Involves large, diverse group of patients; determines efficacy AND safety.


- 3-8 years

What is the approximate success rate of compounds in phase 1 clinical trials?

2/3 pass safety test (or ~600 out of 1000)

What is the approximate success rate of compounds in phase 2 clinical trials?

2/3 pass efficacy test (or ~400 out of 1000)

What is the approximate success rate of compounds in phase 3 clinical trials?

1/10 pass safety/efficacy test (or ~50 out of 1000)

What is the overall success rate of compounds undergoing the clinical trials?

50/1000 or 5% success rate

What is the most expensive step in drug development?

Clinical trials (increases with each phase; phase 3 is highest)

What are the 4 important guidelines for 'weeding out' unsuccessful compounds (prior to clinical trials)?

- Apply ADME test


- Apply Lipinski rules for ''drug-like molecules''


- Remove compounds with suspected toxicity


- Use computer-based screens

Why aren't all adverse side effects discovered in clinical trials?

- Genetic diversity in a large population makes it impossible to test against all these adverse effects!

What type of warning can be found on drugs with severe side effects?

Black box warning

What are the 6 steps of the drug discovery process?

1. Target identification


2. HTS (High Throughput Screening)


3. Active-to-Hit (AtH)


4. Hit-to-Lead (HtL)


5. Lead Optimisation (LO)


6. Candidate Drug (CD)

What 5 biological mechanisms are used in the top 50 drugs (by worldwide sales)? List IN ORDER.

1. Enzyme inhibitors (38%)


2. Receptor Antagonists (24%)


3. Receptor Agonists (12%)


4. Biologicals (10%)


5. Ion Channel Modulators (8%)

What is the mechanism of Lipitor? What does it treat? Who manufactures it?

- HMG CoA inhibitor (enzyme inhibitor)


- Cholesterol


- Pfizer

What is the mechanism of Nexium? What does it treat?

- Proton-pump inhibitor (enzyme inhibitor)


- Anti-ulcer


- AstraZeneca

What is the mechanism of Plavix? What does it treat?

- Anti-platelet (receptor antagonist)


- Thrombosis (blood clotting)


- BMS/Sanofi

What is the mechanism of Norvasc? What does it treat?

- Calcium channel blocker (ion channel modulator


- Hypertension


- Pfizer

What does a receptor agonist do?

Binds to a receptor to increase a desired function.

What does a receptor antagonist do?

Binds to a receptor to decrease an undesired function.

What is HTS?

High Throughput Screening


- Industrial process using robotics, data processing, etc, to conduct millions of tests to rapidly identify a lead compound

Describe radioligand binding assays.

- Beads with receptors/antibodies that will bind to compatible molecule


- If a molecule is able to bind to receptor, the bead activates and light is produced

What is SPA?

Scintillation Proximity Assay


- First homogenous HTS tech


What are the advantages of SPA?

- ~30k compounds/day in 384 microtitres


- Easy to automate and no significant v of aq. waste

What are the disadvantages of SPA?

- Radioactive (safety headaches)


- Long read times (>30min/plate)


- Susceptible to quench artifacts


- Not applicable to all targets

What is FLIPR?

Fluorescent Imaging Plate Reader


- Real time imaging of 96 & 384 well plates

What is FLIPR used for?

- HTS calcium flux assays


- Ion channel screening

How does FLIPR work?

- Cells are loaded w/ fluorescent dye sensitive to Ca2+ (fluorine-3)


- Addition of receptor agonist stimulates Ca2+ release and results in increases fluorescence


- Whole plate read simultaneously using cameras at base of microtitre plates


-

What is a colorimetric HTS?

Uses single compounds bound to beads.


These compounds would only bind a dye if they are also able to bind a receptor of interest.


So, in the end have a bunch of beads with only some stained.

Where are compounds isolated from? Name 4 sources.

- Plants


- Microorganisms (i.e. bacteria)


- Marine organisms (i.e. sponges, molluscs)


- Synthetic efforts by academic + industrial organic chemists

What are combinatorial libraries? What is one pro and one con?

- Using combinatorial chemistry, combine a bunch of different compounds in class A (i.e. a bunch of amines) with a bunch of different compounds in class B (i.e. carboxylic acids).
These undergo same reaction to prouduce a compound (i.e. a bunch of different amides).



Pro: Large libraries can be generated



Con: Little diversity;


Difficult separation of active compounds found

List 3 advantages of NP based libraries.

- High diversity of chemical structures in each extract


- Active compound has a unique structure


- Compounds are likely bioavailable

List 3 disadvantages of NP based libraries.

- Extremely complex mixtures


- Small amounts of active ingredient


- Difficult to identify active component

How is a "diversity oriented" library created?

- Take a biological NP structure and modify it (i.e. via alkylation, oxidation, etc)


- This creates hundreds of other compounds

What is ethnopharmacology?

Study of the use of plant-derived materials in traditional societies for maintaining/improving health.

___% of the world uses ethnopharmacology, and ___% of new cancer drugs + antibiotics are NPs inspired by ethnopharamacology.

80% of the world uses ethnopharmacology, and ~50% of new cancer drugs + antibiotics are NPs inspired by ethnopharamacology.