Drug Development (Block 1) Flashcards

1
Q

Drug research

A

Testing 10,000 compounds

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

Preclinicals

A

<250 compounds

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

Clinical trials

A

Usually one lead compound

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

Drug discovery

A

First five years

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

Changes in drug discovery

A

Moving on from basic research to high throughput screening and on further to machine learning

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

High Throughput Screening (HTS)

A

Allows for testing thousands of compounds against single protein; introduced in 1990s

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

Machine learning

A

Brings together all the basic research to put everything into a network to see things from a different angle

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

Discovery timeline order

A

1) target identification and validation
2) Hit identification and lead identification
3) lead optimisation
4) nonclinical development

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

Target identification followed by

A

Identifying compounds that will hit that target via bioassay developments

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

Hit identification and lead identification

A

HTS and in silico screening

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

Between which stages does compound selection take place?

A

Lead optimisation and nonclinical development

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

Why does this process need to be completed?

A

To meet specific requirements to present information to regulatory authorities

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

Regulatory authorities in Europe and USA

A

Europe -> EMA
USA -> FDA

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

Clinical Trial Authorisation (CTA)

A

Drug companies contact EMA to apply for CTA
1) Justify the compound exhibits pharma activity to meet unmet need
2) Support product being reasonably safe from humans
3) Justify exposing humans to reasonable risks when used in limited, early-stage clinical studies
4) Manufacturing route

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

Animal pharmacology and toxicology studies

A

data to demonstrate drug efficacy and safety for initial testing in humans

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

Manufacturing information

A

Data to support that the adequate, consistent and stable batches of drug will be used

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Clinical protocols and investigator information

A

Detailed protocols, information of clinical investigator(s), information on process for obtaining ethics approval

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

G x P criteria

A

Standards at which studies must be performed

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Why are G x P criteria needed?

A

To ensure everyone adheres to the same standards that address risk control measures, standard quality and equality, and public safety.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Which sections are regulated by G x P?

A

Preclinical -> GLP (Good Lab Practice)
Clinical -? GCP (good clinical practice)
Manufacturing -> GMP (Good manufacturing practice)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Goals of preclinical studies to support CTA

A

Understand exposure related to efficacy
Identify organ toxicities and reversibility
Understanding of therapeutic index (efficacy vs toxicity)
Identify safe starting dose
Guide dosing regimens and escalation schemes
IND/IMPD will continue to be updated during clinical development with new significant information (eg. toxicology, clinical data etc)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

ADME

A

Absorption
Distribution
Metabolism
Excretion

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Types of DMPK studies -Absorption

A

Pharmacokinetic/toxicokinetic
Bioanalytical method development and validation
Permeability studies
Transporter assays

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Types of DMPK studies -Distribution

A

Protein binding
Quantitative whole body autoradiography (QWBA)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

Types of DMPK studies -Metabolism

A

In vitro cross-species metabolism
Metabolite identification
CYP450 metabolism
Transporter assays

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

Types of DMPK studies - Excretion

A

Mass balance
Transporter assays

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Pivotal toxicology studies

A

Shows whether anything is happening in other (non-target) organs
Multiple doses in 2 animal species - rodent and non-rodent
Doses must exceed intended human exposure
Prior to FTiH generally conduct a 28 day study
Recovery group included to see if toxicology is reversible
Study is under GLP conditions, ideally with drug manufactured to GMP

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

No Observed Adverse Affect Level (NOAEL)

A

Study designed to explore ascending does groups and establish a dose response curve

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Toxicokinetics (TK)

A

Critical to understand exposures associated NOAEL and MTD
Will be used in the calculation of starting dose for human clinical trials
Do not want to exceed exposures associated effect levels in humans
Interspecies differences in protein binding are taken into account

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What is meant by the term GLP?

A

Good Laboratory Practice is a set of guidelines to ensure quality and integrity of non-clinical lab studies

31
Q

Phase 1

A

1st time new investigational medicinal product is tested in humans
Usually healthy volunteers
Sometimes patients with advanced disease (eg. cancer)
Small numbers of subjects (20-80)
Aim is to evaluate safety, tolerability and pharmacokinetics
Clinical Pharmacology studies to explore ADME
These are called Phase I studies, but can span clinical development

32
Q

What is clinical pharmacology?

A

Providing scientific basis for rational safe and effective prescribing

33
Q

PK-PD

A

The relationship between dose & therapeutic effect
The relationship between dose & toxicity
Exploring the causes of interindividual variability

34
Q

Cmax

A

Peak clinical conc in plasma

35
Q

Single Ascending Dose study (SAD)

A

First Time in Human Study (FTiH) is generally a single ascending dose study (SAD)

Cohort 6-8 active drug; 2-4 placebo

Double- blind

Emerging data is continually reviewed by a safety monitoring committee

Doses can be either escalated or de-escalated

36
Q

Multiple Ascending Dose (MAD) study

A

Cohort design, placebo controlled
Aim is to dose long enough to achieve steady state
i.e. concentrations remain constant – reached equilibrium
Generally achieved in 4-5 half-lives
Main endpoint is safety, tolerability and pharmacokinetics
Will determine what dose(s) can be studied in Phase 2

37
Q

Absorption and bioavailability

A

Not all of a drug that gets administered will enter the systemic circulation; during disintegration, dissolution, and transport some of the active compound may be lost.

38
Q

Bioavailability is a function of

A

Fabs – fraction absorbed
FG – fraction escaping gut metabolism
FH – fraction escaping hepatic metabolism

39
Q

Bioavailability (definition)

A

The fraction of drug that reached then circulation

40
Q

Absolute bioavailability

A

assessment of systemic drug availability of extravascular dose compared with IV dose

41
Q

Relative bioavailability

A

compares bioavailability of one formulation with another

42
Q

Comparative bioavailability study

A

Two or more formulations of the same drug are compared, are usually designed as crossover studies. Each patient acts as his or her own control. This allows for the direct comparison of treatments.

43
Q

Food effect studies

A

Initial studies are done in the fasted state and then food effect studies follow.
Study design:
Statistically sized cross-over design
PK of drug assessed following drug administration under fasted and fed conditions

44
Q

Distribution - tissue penetration studies

A

studying whether the drug actually reaches target site in sufficient concentration; clinical studies to understand relationship between plasma concentrations and concentrations at site of action may be required

45
Q

Metabolism

A

Most early healthy volunteer studies exclude co-medications
CYP450 enzymes are the main metabolising enzyme system
If drug is a substrate, inhibitor or inducer of a common pathway (eg. CYP3A4), the impact on exposure when dosed with other agents may need to be considered

46
Q

What could be the impact on exposure of Drug A (CYP3A4 substrate) when dosed with Drug B (strong CYP3A4 inhibitor)?

A

CYP450 liability is assessed non-clinically as part of DMPK package
However understanding this doesn’t predict the impact on clinical exposure
Clinical DDI study is conducted:
New Drug is dosed alone or in combination with a known inhibitor
Cross-over design
Endpoint is PK
Compare AUC and Cmax following drug administered alone or in combinatation
Dosing recommendations will be based on whether impact is clinically relevant

47
Q

What type of study is done to investigate drug excretion?

A

Mass balance study

48
Q

Aim of mass balance study

A

understand full clearance mechanisms of drug and metabolites

49
Q

Mass balance study (details)

A

Single dose of radio-labelled (C14) drug: plasma, urine and faeces collected
Radiolabelled drug and concentrations of parent and metabolite(s) measured
Assesses the major routes of clearance:
may influence whether a renal and hepatic impairment study is required
Proportion of parent drug converted to metabolite(s)
Metabolite identification (eg. detection of unique human metabolites, active metabolites)

50
Q

Renal and hepatic impairment studies

A

liver and kidney are major routes of elimination -> impairment = significant change in exposure
Assessment of influence renal and hepatic impairment on exposure can lead to dosage adjustment warnings on the label
Until these studies are done, patient population will need to be restricted
Requirement for studies will depend on major route(s) of elimination

51
Q

‘special’ populations

A

Elderly
Risk of altered exposure
Obese
Ethnic groups – if the main programme is conducted in the West, specific PK study may be required to justify dose in certain ethnic groups (eg. Chinese, Japanese)

52
Q

Why is obesity specifically included?

A

PK variability due to alterations in Vd and Clearance. Vd could be overestimated for hydrophilic drugs due to poor penetration into adipose tissue. Obese patients have increased organ mass which may lead to increased renal blood flow thus increasing renal clearance. Those with gastric bypass have loss of surface area for drug absorption.

53
Q

Why are the elderly specifically included?

A

physiologic alterations include reduced gastric acid secretions affecting absorption, reduced renal and hepatic blood flow which slows the rate of clearance, and reduced serum protein concentrations which increases free drug concentrations.

54
Q

Pharmacokinetic variability

A

Most clinical pharmacology studies are conducted in healthy volunteers, however, it is important to understand PK in target patient population bc weight/body surface area, renal or hepatic function, illness severity, clinical indication, age, concomitant medications, and ethnicity can have an impact

55
Q

PK sampling - phase one (healthy volunteer)

A

Intensive PK samples can be taken in Phase 1
Easy to calculate Cmax, AUC, clearance, volume of distribution etc.
PK parameters calculated for each individual

56
Q

PK sampling - phase 2 and 3

A

Generally only sparse PK samples collected
Impossible to calculate PK parameters if individual data used in isolation; used in combination with phase 1 information

57
Q

Population pharmacokinetics

A

all data is analysed together
allows exploration of mean exposure and variability sources
use simulations to predict appropriate dosing regimen for majority of individuals

58
Q

Phase 2

A

Main aim is proof of concept– some form of clinical efficacy
Endpoint may be different to that explored in Phase 3
Generally in patients with condition under study (or related condition)
Further safety and PK evaluation
Can be comparator studies or single arm
May explore dose range
May be open label or blinded

59
Q

End of phase 2 meeting

A

Review of data and agreement for design of Phase III study
Sponsor looks for regulatory agreement for:
Endpoints and timing of endpoints of trial (eg. mortality, clinical cure)
Size
Patient population
Comparator and blinding
Dose
Duration
Paediatric plan

60
Q

Regulatory agreement

A

Clinical trial application; includes protocol, investigators brochure, IMPD, scientific advice, informed consent and patient information leaflet, investigational product labelling and GMP-related documents, and ethics

61
Q

Phase 3

A

Pivotal studies designed and executed to provide statistically significant evidence of efficacy and safety
Generally randomised controlled trial (RCT) against standard of care or placebo
Parallel study design
Sometimes adaptive design can be used
Often two identical independent studies required
Nearly always double blind

62
Q

Types of phase 3 trials

A

Superiority
Non-inferiority
Equivalence

63
Q

Superiority trial

A

To determine a clinically relevant difference between two treatments. i.e. want to show new treatment is statistically significantly better than existing therapy or placebo

64
Q

Non-inferiority trial

A

To determine whether new treatment is not inferior to another established treatment

65
Q

Equivalence trial

A

To determine whether a new treatment is neither worse nor better than another established treatment
More often used for approval of generics, or formulation changes

66
Q

Regulatory review

A

Submission documents reviewed by team of technical experts
EU – centralised procedure
Regulatory defence
Additional analysis may be required
Regulatory inspections
Process could take up to 2 years (longer in some countries)

67
Q

Biggest reason for drug failure

68
Q

Second biggest reason for drug failure

69
Q

Label and patient information leaflet

A

Core information is included in the summary of product characteristics (SPC) or label
Patient information leaflet are also produced written in clear, easy-to-understand language for general public and patients

70
Q

Post-approval

A

Post-marketing commitments
Pharmacovigilance
Paediatic investigation plan

71
Q

Post-marketing commitments

A

studies required by regulatory authorities but not included in original package
May remove some restrictions on the label

72
Q

Pharmacovigilance

A

science and activities relating to the detection, assessment, understanding and prevention of adverse effects or any other medicine-related problem
Up to approval, evidence of safety and efficacy is limited to data from clinical trials, where patients are selected carefully and followed up under controlled conditions
Safety of medicines is monitored throughout their use in healthcare practice

73
Q

Paediatric investigation plan

A

drugs are first approved for adults and aren’t always tested for children so there is a government push to bring in a focus on it; in the EU it’s mandatory unless there’s a specific waiver for conditions that don’t exist in paediatric patients

74
Q

Development in paediatric patients

A

children aren’t just small adults; process are different and must be accounted for