Drug discovery Flashcards

1
Q

give examples of where drug discovery and release has gone wrong:

A
  1. elixir sulfanilamide - contained diethylene glycol which killed 107 people
    2 sulfathiazole tablets - had sedative phenobarbital in it which caused 300 deaths
  2. thalidomide - sleeping pill which caused severe birth defect of arms and legs in 10000-20000 babies
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

what are the phases of drug discovery?

A
  1. target selection
  2. preclinical research - animal testing
  3. clinical research:
    - phase 1 = tests on healthy humans
    - phase 2 = patient entry
    - phase 3 = proving to regulatory authorities e.g. FDA
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

what is target selection?

A
  1. functional proteins identified from bioscience research
  2. lead finding: automated screens against libraries
    - when the biochemical target has been identified, the lead compounds must be identified
    - cloning of human target protein is used and an assay is developed to measure the functional activity of the target protein
    - predict toxicology
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

how are animals used in preclinical research?

A

rodents (rat) and non-rodents (beagles) are given 3 dose groups:
- low = no toxicology
- intermediate = reversible toxicology
- high = toxicology expected to be seen in target organ

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

what is clinical pathology vs pathology?

A

clinical pathology: haematology/clinical chemistry (lab results)

pathology: large organ toxicity

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

what are the goals of non-clinical safety evaluations?

A

toxicity:
- on-target = drug goes to wanted target,
- off-target = drug causes unwanted side effects

toxicokinetics: relate toxicity to exposure
- increase in dose should cause linear increase in toxicity effects
- preliminary toxicological testing to eliminate genotoxicity

identify max non-toxic dose/minimum effective dose

pharmacokinetic testing (ADME)

feasibility of large scale synthesis and purification

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

what is safety pharmacology?

A

the investigation of potential undesirable effects of drugs on physiological functions:
- test CVS (in vitro and in vivo)
- test CNS (rodent tests)
- test respiratory system (rodent tests)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

what preclinical conclusions must be made before starting clinical human trials?

A
  • evidence of pharmacological activity
  • maximum non-toxic dose
  • identify possible adverse effects on target organs
  • show relationship of effects to dose and exposure
  • differences observed in different species
  • evaluation of risk in humans
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

what is phase 1 of clinical trials?

A

IS IT SAFE?
- referred to as First in Man
- aim: test the drug is safe in humans and check for dangerous effects on CVS, respiratory, hepatic or renal functions
- test tolerability - does the drug produce unpleasant symptoms such as nausea?
- test pharmacokinetic properties - is the drug well absorbed? what is the time course of the plasma concentration?

performed on a group of 20-80 healthy volunteers - first cohort receives low dosage which is then adjusted for the following cohort

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

what is an example of phase 1 trials going wrong?

A

BIA 10-2474:
- fatty acid hydrolase inhibitor to treat anxiety disorder of Parkinson’s and for chronic pain of MS, cancer, hypertension and obesity
- interacts with endocannabinoid system
- phase 1 trials led to serious adverse effects in 5 participants - 1 person died after having 50mg repeat dose due to lack of metabolism of the drug in the brain
- 1.25mg would have been enough for saturation, which should have been identified in preclinical trials

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

what is phase 2 of clinical trials?

A

PROOF OF CONCEPT - how much should be given to be effective?
- performed on 100-300 patients to test for efficacy in clinical situations and establish dosage
- covers several clinical disorders such as anaemia and rheumatoid arthritis to identify therapeutic indications for the new compound and dose needed

made up of phase 2a and phase 2b

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

what is phase 2a of clinical trials?

A

conducted in healthy volunteers or patients to determine pharmacodynamics and biological activity:
- proof of efficacy
- studies on the mechanism of action of the drug
- exploring a range of doses
- pilot studies

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

what is phase 2b of clinical trials?

A

definitively finding the dose range and efficacy in patients:
- used as pivotal trials if the drug is intended to treat life-threatening or severely debilitating illnesses
- definite dose finding studies

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

what is phase 3 of clinical trials?

A
  • over 1000 patients
  • need definitive results via: double-blind, randomised trials
  • compare new drug with commonly used alternatives
  • multinational trials
  • application for marketing is made

includes phase 3a and 3b

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

what is phase 3a in clinical trials?

A
  • study designed to get statistically significant evidence of efficacy and safety for NDAs approval
  • pivotal studies: provide evidence for a drug marketing approval via randomised, double-blind, placebo trials
  • long-term safety studies for registration
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

what is phase 3b in clinical trials?

A

study started prior to approval with intention to support publications, claims or to prepare launch
- not intended for regulatory submissions

17
Q

what is phase 4 in clinical trials?

A
  • post marketing surveillance to detect rare or long-term adverse effects from the use of the drugs in a clinical setting with thousands of patients
  • can lead to limitation of drug use by certain patients, or even complete withdrawal of the drug
18
Q

what are some drugs which have been withdrawn from the market?

A
  1. sibutramine (appetite suppressant) - heart attack and stroke
  2. propoxyphene (opioid painkiller) - heart attack and stroke
  3. drotrecogin alpha (severe sepsis) - had no benefit
  4. rimonabant (obesity) - severe depression and suicide
  5. hydromorphone (narcotic painkiller) - high risk of accidental overdose with alcohol
19
Q

what is a small molecule drug?

A
  • low molecular weight
  • chemically synthesised
  • well-defined structure
  • not complex
20
Q

what is a biological molecule drug?

A
  • high molecular weight
  • derived from living organisms
  • large and complex structure
21
Q

what is a monoclonal antibody?

A
  • high molecular weight
  • derived from living organisms
  • complex
22
Q

what are biologics?

A
  • drugs which are derived from living organisms
23
Q

how have biologics contributed to new medicines?

A
  1. antibody-based: anti-VEGF
  2. vaccines: HPV-vaccine, COVID19-vaccine
  3. RNAi: Duchenne Muscular Dystrophy
  4. cell-based: CAR-T for children with acute lymphoblastic leukaemia
  5. gene therapy: adeno-associated virus causes haemophilia
    - plasmid is delivered to body so the body can produce its own clotting factor 9 enzyme
24
Q

what are biopharmaceuticals?

A

products in which the active substance is produced by, or extracted from, a biological source e.g. monoclonal antibodies to be a human protein
- off-target toxicology is rare

25
Q

what are the adverse reactions of biopharmaceuticals?

A
  • exaggerated pharmacology
  • anti-drug antibody responses - immune system fighting the biologic, causing accelerated clearance and metabolism to neutralise the pharmacological activity
  • on-target toxicity can be high - target can be hit so strongly that it causes adverse effects, so balance is critical
  • max dose: 10x max exposure in clinic
26
Q

what is the immunotoxicity of small molecule drugs and biopharmaceuticals?

A

small molecule: often unexpected, off-target reactions

biopharmaceuticals: cytokine storms

27
Q

what is the phase 1 dose?

A

minimum effective dose:
smallest possible dose that will bring about a response from the organism
- minimum anticipated biological effect

28
Q

what is CD28-superMAB (TGN1412)? what happened during its clinical trials?

A
  • immunomodulatory drug which was withdrawn from development after inducing severe inflammatory reactions in phase 1 trials
  • was intended to treat B cell chronic lymphocytic anaemia
  • strong agonist for CD28 receptors on T cells
  • preclinical profile was good - showed doses with no side effects
  • in phase 1, 6 volunteers went into possible as they developed fever
  • the drug caused a cytokine release syndrome (like an anaphylatic shock)
29
Q

what is immunotherapy?

A
  • form of cancer treatment that uses the immune system to attach cancer cells
30
Q

what are checkpoint inhibitors in immunotherapy?

A
  • causing a natural break in the immune system so that the immunity T cells recognise and attack tumours
31
Q

what is CAR-T cell therapy?

A
  • chimeric antigen receptor (CAR) T cell therapy is the genetic engineering of a patient’s own immune cells to make new protein to fight tumour cells
  • T cells are taken from a patient and their receptors are altered to recognise tumours
32
Q

how was CAR-T cell therapy developed?

A

1990 - cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) was found on T cell surface which could be blocked, leading to tumours vanishing

1999 - checkpoint inhibitors:
- programmed cell death protein 1 (PD1) on T cell dampens down the immune system
- programmed death-ligand 1 (PD-L1) protects cancers from T cells
- these proteins were how cancer evaded the immune system, so if these receptors are blocked, the immune system can detect and destroy the cancer

33
Q

what was the action of the anti-CTLA-4 antibody?

A
  • worked against the CTLA-4 receptor on T cells to treat malignant melanoma
  • CTLA-4 on T cells bind to B7 proteins which in turn prevent the T cells from activating and attacking tumour cells
  • the anti-CTLA-4 antibody blocks CTLA-4, so T cells can activate and attack cancer cells
34
Q

what is the major histocompatibility complex?

A

MHC is a set of cell surface proteins essential for the acquired immune system to recognise foreign molecules
- they bind to antigens derived from pathogens and display them on their surface for T cells to recognise

35
Q

what is pembrolizumab?

A
  • a drug which blocks PD-L1 to treat lung cancer
  • can eliminate, regress or stabilise cancer, with long lasting effects
  • only works on cancers with PD-L1 receptor
36
Q

how are chimeric antigen receptors made for CAR-T therapy?

A
  • CARs are produced by splicing together the gene for an antibody that recognises a tumour antigen, and the gene for a receptor that sits on surface of T cells
  • this new gene is inserted into the T cell, causing it to target the tumour
37
Q

What is the process in which CD3 cells are made to target cancers? (example of CAR-T)

A
  • CD3 cells with CD19 marker are extracted and modified with a plasmid (transfection)
  • cells are formulated into a new therapy and monoclonal antibody fuses with the receptor on T cells to attack cancers
  • cells are put back into the patient without rejection and destroy the cancer
38
Q

what are the limitations of CAR-T therapy for CD19 blood cancers?

A

CD19 is only common to the surface of a few blood cancers

can cause fatal immune reactions:
- cytokine-release syndrome
-B cell aplasia
- brain swelling
- neurotoxicities