Introduction to Clinical Trials Flashcards

1
Q

What are the three key features of an RCT broadly speaking?

A

Controlled
Unbiased
Adequately sized

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

List some different types of intervention that may be evaluated by a clinical trial

A
Drug therapy
Medical procedures
Educational intervention 
Screening Trial
Vaccine trial
Medical therapy
Complex interventions (e.g effects of educational programmes)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What is the Declaration of Helsinki?

A

International recommendation on the conduct of medical research on human participants to which all clinical trials should comply

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

What is the national ethical committee responsible for reviewing of trial protocols in the UK to ensure safety of future participants?

A

IRAS (Integrated Research Application System)

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

What is the role of the MHRA in the UK and FDA in the USA

A

Regulatory authorities approve trials of investigational medicinal products and medical devises to ensure trials have been conducted with sufficient quality and standards and further to licence products which are effective and safety has been properly evaluated

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

Outline the role of an Observational Trial

A

Non-experimental study where the individuals are observed and data collected with nothing being done to influence either the exposure or the course of events.

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

What is the major flaw of observational trials?

A

Can be subject to considerable bias if not designed well. Often have very different outcomes to RCTs

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

Outline the different phases of clinical trials

A

Phase I - Testing effect(s) of novel therapy on healthy sample
Phase II - Dose-response curve
Phase III - large sample size
Phase IV - post-marketing (long-term safety and interactions)

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

What is the most common trial design?

A

Parallel trial - New vs standard therapy

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

List some other trial designs other than parallel

A
Cross-over
Factorial
Equivalence
Bioequivalence 
Non-inferiority 
Superiority 
Exploratory
Confirmatory 
Cluster
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Outline the utility of single vs multi-centre trials

A

Single centre - used for Phase I and II trials

Multi-centre trials - good for large recruitment over short periods of time and increases generalisability of results

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

Outline three reasons why trial results may not represent the ‘truth’?

A

Biased in a predictable fashion
Contimainated with a confounding factor
Random chance

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

What are the most common sources of bias in trials?

A

Most commonly at selection and outcome measurement.

Also includes omission of data and loss to follow-up

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

How are confounding errors minimised?

A

Adequate planning and exploration of variables with sound scientific principles and reasoning.

Randomisation (e.g. stratified) to account for known dependent variables

Stratified analysis and regression analysis

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

How is the effect of random chance minimised in trials?

A

Large sample sizes, appropriate of results statistical analysis and meta-analysis

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

What are the advantages of uncontrolled trials?

A

Used to generate hypotheses and justifications of PIII trials

Can assess pharmacokinetic properties of novel agents

Might be the only study design possible due to ethical considerations

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

What are the disadvantages of uncontrolled trials?

A

Investigator bias

Susceptible to over-interpretation

Much less informative than RTCs

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

What is the purpose of the clinical trial protocol?

A
  1. Defines the clinical question
  2. Outlines how it is answered
  3. How will it be compliant to ICH-GCP, EUCTD, FDA/MHRA regulations
  4. What analyses should be reported
  5. How will results be interpreted?
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What is the purpose of the clinical trial protocol?

A
  1. Defines the clinical question
  2. Outlines how it is answered
  3. How will it be compliant to ICH-GCP, EUCTD, FDA/MHRA regulations
  4. What analyses should be reported
  5. How will results be interpreted?
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

How are study objectives stated differently in a priori (proscpective) versus retrospective research?

A

Prospective (a priori) = stated as a hypothesis

Retrospective research = stated as a question

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

What are primary endpoints in a trial?

A

Measure outcomes that answer the primary question of the trial

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

What are secondary endpoints?

A

Ask other questions relevant to the study

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

What are the main type of endpoints investigated in RCTs?

A
Binary (e.g. death of any cause)
Disease specific death
Occurrence of symptoms 
QOL outcomes
Utilisation of healthcare resources
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What qualities must endpoints have to be clinical relevant?

A

Clinically meaningful
Answer the main question
Can be practically assessed
Occur frequently enough within the time frame of the study to be powerful enough

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

Death as an endpoint can be impractical for what reasons?

A

May be rare in the timeframe of the study

Death can results from factors other than those under scrutiny in the trial

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

What are composite endpoints?

A

Multiple outcomes measured

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

What stage of trials are composite endpoints more common?

A

Exploratory / Early-phase trials

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

How can composite endpoints be expressed in a trial?

A

Can be expressed as total numbers or in a hierarchical fashion (only counting the most serious endpoint)

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

What are the advantages of composite endpoints?

A
  1. Increases event rate therefore decreases participants required
  2. More comprehensive evaluation can be achieved including clinical, pathologic, radiographic and biochemical can be measured simultaneously
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Limitations of composite endpoints are:

A

Assumption that avoidance of any one endpoint is as clinically significant as any other

Assumption that each outcome related to disease progress are equally important

Inconsistent results if some outcomes improve and some deteriorate.

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

What are surrogate endpoints?

A

A lab measurement / physical sign used as a substitute for a clinically meaningful endpoint that directly measures how a patient feels, functions or survives

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

Give some common examples of surrogate endpoints

A

Pharmacokinetic measurements e.g. conc/time curves / active metabolites

In vitro measurements e.g. ABx concentration (mean) required to inhibit microbial growth.

Radiological appearance

Disease markers

Macroscopic appearance of pathology

32
Q

What are the advantages of surrogate endpoints?

A

Decrease sample size

Particularly used in phase II trials where changes in surrogate endpoints occur before adverse clinical events

33
Q

What are the limitations of surrogate endpoints?

A

Relationship between surrogate and clinical outcomes must be well understood otherwise it can produce misleading results

34
Q

What are the two main health-economic outcomes?

A
  1. QUALYs - measurement of the duration of an individuals life in the context of clinical significance
  2. Cost
35
Q

What measurement can be made from both QUALY and cost?

A

Cost-effectiveness ratio in terms of QOL

36
Q

What are the advantages of health-economic endpoints?

A

Creates a common variable with which to compare unlike outcomes such as sight-years gained, symptom free time etc.

37
Q

What are the limitations of health-economic outcomes?

A

Economic analyses not easily transferrable across countries since cost of care and access to resources vary massively.

Effects of acute conditions may not have an effect on long term impact of QOL if not fatal therefore economic analysis may be inappropriate in these scenarios

38
Q

Why is the process of patient selection so important? How do we define selection?

A

A set of eligibility criteria is essential to ensure that the results of the trial have external validity.

39
Q

What considerations need to be taken into account when reviewing eligibility criteria?

A

Source of patients - sample size and clinical outcome balanced against practical considerations

Investigators and centres - when estimating potential participants a screening log is useful

40
Q

What are eligibility criteria usually based on?

A

Patient characteristics
Diagnostic test criteria
Disease duration
Disease severity

Convention is to have 1-2 inclusion criteria and more exclusion criteria

41
Q

What is the general aim of eligibility criteria?

A

TO recruit the most number of patients without including patients with significant load of co-morbidity which may affect results of the trial

42
Q

IF specific diagnostic tests are required for patient selection when must these tests be done in relation to randomisation?

A

Before randomisation - stratification/minimisation techniques may be done to ensure balance across groups

43
Q

How does disease status effect patient selection?

A

May be necessary for a washout period pre-trial beginning

In trials it may be unethical to withold treatment and therefore more patients are recruited to the treatment in 1:2 ratio

44
Q

What ethical issues are considered with regards to patient selection?

A

Consider age ranges that reflect the general population

Special considerations

Consider the inconvenience of the trial (e.g. time off work, repeat visits, invasive tests e.g.) - properly counsel patients

45
Q

Why might patients not be selected?

A

Patients unavailable during the screening patients
Study personelle not available to screen patient
Drug/intervention not available
Procedure unavailable e.g. broken ECG machine
Patient lost to follow up

46
Q

If a large number of patients are excluded by the criteria what course of action may be indicated?

A

Review of criteria and potentially a protocol amendment and ethical review

47
Q

What is the definition of clinical trial bias?`

A

Systematic distortion of the estimated intervention effect away from the true value caused by inadequacies of design conduct or analysis / publication of a trial.

48
Q

List some common biases

A
Selection bias
Bias in study management 
Observer ascertainment 
Bias from exclusion post-randomisation 
Publication bias
49
Q

How is selection bias controlled?

A

Randomisation and concealment

50
Q

How is bias in study management controlled?

A

Standardised procedures, equipment and training of staff across sites

51
Q

How is observer ascertainement controlled?

A

Blinding

52
Q

How is bias from exclusion post-randomisation controlled?

A

Intention to treat analysis or worst case scenario analysis

53
Q

How is publication bias controlled?

A

Prospective registration of trials and publication of negative trials

54
Q

What is allocation concealment?

A

Decreases selection bias by shielding randomisation code before and until treatments are administered.

55
Q

What is blinding?

A

Decreases observer bias by shielding treatment after administration

56
Q

What is the most common cause of accidental unblinding?

A

Variation in side effect profile between treatment and placebo

57
Q

How may accidental unblinding due to side effect profile variation between treatment and placebo arms be controlled?

A

Three arm parallel trials

58
Q

What is intention to treat analysis?

A

All participants who are intended to be treated are included in analysis despite non-compliance (may or may not have missing data)

59
Q

What is worst case scenario analysis?

A

If dichotomous data. Assume drop-outs did badly and therefore if the analysis is still in favour of the intervention then must not be due to loss to follow-up

60
Q

What are the issues associated with worst case scenario analysis?

A

May underestimate the true magnitude of effect of loss to follow-up

If the results are negative then the data is uninterpretable

It ignores all data except the final analysis

61
Q

What is meant by per-protocol analysis?

A

Any missing data points are then not included in the final analysis

62
Q

What is publication bias?

A

Editors favour positive findings and therefore pre-registration of the trial with the agreement to publish whatever results are found is good practice

63
Q

List some common methods of randomisation?

A

Simple
Block
Stratified
Minimisation/adaptive

64
Q

What is the definition of simple randomisation?

A

Allocates participants without regard to those who have already been enrolled in trial

65
Q

What is block randomisation?

A

Periodically reinforces a balance in patients for each arm of the trial.

66
Q

When is forced balance most essential for a trial?

A
  1. When recruitment is slow
  2. When the types of patients reviewed over time changes
  3. When the trial may be stopped early
  4. Routine practice changes over the course of the trial.
67
Q

What is stratified randomisation?

A

Within each stratum block randomisation is used to allocate patients to each group.

Different strata account for a dependent variable and balance this between each arm of the trial.

68
Q

What is minimisation / adaptive randomisation?

A

Assigning patients to each arm in a way that adjusts the probability of being assigned to each arm depending on existing imbalances in the groups. Starts in a simple randomisation fashion

69
Q

When is minimisation a fvoured technique for randomisation?

A

In diseases w many prognostic factors and is becoming increasingly popular w computer / interactive voice response systems

70
Q

What five factors affect sample size required for a trial?

A
  1. Expected event rate in control arm
  2. Smallest treatment effect that is clinically worth measuring
  3. Determining the significance level of rejecting the null hypothesis
  4. Choosing the power of the study
  5. Study design
71
Q

How do we approximate the event rate in the control arm?

A

By retrospective research and review of previous studies/ registries

72
Q

How do you work out absolute treatment effect?

A

Event rate in control arm - event rate in treatment arm = absolute event rate

73
Q

How does one determine significance level for type 1 error?

A

Conventionally set at 95% (P-value =0.05) confidence however increasingly up to 97.5% (P-value = 0.025)

74
Q

What is the conventional type 2 error rate? Why is it higher than conventionally accepted type 1 error rate?

A

80% power = 20% type 2 error rate.

Conventionally less stringent than type 1 error rate as the burden of evidence required to change common practice to a new therapy is high.

75
Q

What variable directly affects the power of a study?

A

Number of participants

76
Q

What effect does the study design have on the power and sample size of a study?

A

Parallel studies require larger sample size.

77
Q

How is drop out rate accounted for?

A

Conventionally 1 year drop out rate is assumed to be 20% therefore requirement of sample size is increased by at least 20% from calculated minimum to adjust for this occurrence.

78
Q

Are negative result trials simply due to small sample sizes?

A

Yes and no. Be careful with assumptions for alpha, beta and event rates between the arms of a trial. Meta-analysis can be helpful in increasing power of results.