FinalExamReview-YamalTopics Flashcards

1
Q
  1. Describe different types of studies and be able to rank in terms of strength of evidence
A

Rank in terms of strength of evidence (strong to weak)

1) Clinical Trial
2) Cohort Study
3) Case-control study
4) Cross-sectional study
5) case series

Descriptions:
Clinical Trial - A prospective study comparing the effects and value of intervention(s) against a control in human beings.

Cohort Study - Study where one or more samples (cohorts) are followed up prospectively and subsequent status evaluations with respect to a disease or outcome are conducted. In this type of study participants are identified based on their exposure characteristics (risk factors) associated with the disease or outcome of interest.

Case-Control Study - Study that compares patients who have a disease or outcome of interest (cases) with patients who do not( controls, and looks back retrospectively to compare how frequently the exposure to a risk factor is present in each group to determine the relationship between the risk factor and the disease.

Cross-sectional Study -

Case Series - Article that describes individual cases. (Advantages - can ID trends/disease; Disadvantage - lack of generalizability)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q
  1. Be able to identify different types of bias including their definitions
A

Information Bias - Misclassification of information/data collected

Confounding -the association between the predictor and outcome is distorted by extraneous factors (confounders)

Publication Bias - Studies with significant results are more likely to be published than studies without significant results

Selection Bias - A difference in characteristics of participants selected for a study and the population from which the participants were selected.

Recall Bias - Bias that occurs when participants are asked to recall events in the past that they may recall differently depending on current health status.

Ascertainment Bias - Loss to follow up, impacts perceived effect of treatment if participants are not loss to follow up at random.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q
  1. Define Phase I-IV trials
A

Phase I: Goal is to determine which dose of the drug is safe and most likely to show benefit.

Phase II: Goal is to identify agents with potential efficacy (NOT designed to definitively test efficacy). Discard agents without promise. Provide rationale for continuing the experiment.

Phase III: Goal is to define the “best” treatment which has implication of changing current practice

Phase IV: Goal is to monitor adverse effects, long-term morbidity and mortality after treatment is applied to large number of patients and follow up for a long period of time

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

Describe a Phase I (Clinical Pharmacology and Toxicity) clinical trial.

A

Primary Concern: Safety
Participants: Healthy volunteers or patients who have already tried and failed to improve on existing standard therapies.
Goal: Estimate tolerability and characterize pharmacokinetics and pharmacodynamics. (Max. Tolerable Dose etc)
n is typically 15-30 participants
Clinical Considerations: Route, schedule and starting dose, Plan for escalation, patient selection, extent of toxicity
Statistical Considerations: Number of patients per dose, Well-defined escalation theme, stopping rule, decision rule for recommended dose

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q
  1. Be able to explain how the eligibility criteria can affect generalizability and differences in results between phases of studies.
A

Goals:

  • setting certain eligibility criteria can increase the likelihood of detecting an effect
  • minimizing the possibility of adverse effects (exclude pregnant women, exclude people without adequate liver/renal function)

Advantage:
Heterogenous groups let us study the mechanism in action

Impact:

  • Many studies are performed in academic medical centers which may not be representative of general population
  • underrepresentation of minorities
  • underrepresentation of women
  • participants may be different from non-participants in many significant ways (health, disease, racial or sex, etc)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q
  1. Identify differences in outcomes and how they would be analyzed
A

Ideal Outcome:

  • valid and reliable
  • easy to observe
  • free of measurement error and other bias
  • ideally complete observation in all subjects
  • capable of being observed independent of treatment assignment
  • clinically relevant

Types:

  • continuous
  • categorical (attractive for descriptive purposes, but we lose power and can lose information, difficult to generalize as we saw in the studies we read on COVID treatments)
  • dichotomous
  • count, ordinal
  • nominal
  • time to event
  • composite or global
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q
  1. Be able to explain properties of good surrogate outcomes and what their limitations are
A

Surrogate - A biomarker that is intended to substitute for a clinical endpoint; expected to PREDICT the effect of an intervention on the clinical outcome.

  • strongly associated with definitive outcome
  • part of the causal pathway
  • yields the same inference as the definitive outcome
  • responsive to the treatment
  • short latency

Prentice Criteria:

  • proposed outcome predictive of true (clinical) outcome
  • proposed surrogate must fully capture effect of intervention on (clinical) outcome

Examples:

1) Treatment of hypertension:
- Clinical outcome - Myocardial Infarction
- Surrogate - Change in blood pressure
2) Treatment of MS
- clinical outcome - relapse lesions
- surrogate - MRI changes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q
  1. Identify issues with incorporating baseline measures of the outcome into change from baseline as the outcome and be able to discuss alternative approaches.
A
  • if the baseline is related with the outcome can induce spurious correlation
  • percent changes is dependent on what the baseline value is (we can have the same percent increase, but magnitude of change is very different)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q
  1. Identify the various types of control groups and their roles in different types of trials.
A

Placebo - same look and delivery method as treatment, but lacks an active ingredient.

No Treatment - control group has no treatment

Standard of care - control group receives the standard of care that is currently in practice.

Another treatment - not necessarily considered “standard of care”

Historical Control (new treatment is used and compared to historical outcomes from database)

Prospective registries as controls

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q
  1. Describe the randomization process and different types of randomization procedures
A
  • independent central unit should be responsible for developing randomization process (biostatistician for single center or data coordinating center for multicenter)
  • Best if investigators are blind to randomization
  • Sequenced and sealed envelopes that have assignment
  • randomization list kept in pharmacy for double-blind drug studies
  • web-based randomization - capture basic eligibility data, gives randomization assignment
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q
  1. Describe the role of blinding and types of blinding
A

Single Blinded:
- A trial in which the participant is unaware of which treatment group they have been assigned, but the investigator is aware of the assignment.

Advantage: design is similar to unblinded, simple to carry out

Disadvantage - investigators van add bias b/c they are aware of treatment assignment. Investigator may intentionally or unintentionally un-blind the participants.

Double Blinded:
- A trial in which neither the participant nor the investigators following the participants know the identity of intervention assignment.

Advantages:

  • reduce risk of bias
  • account for placebo effect

Disadvantage:

  • must manufacture placebo to match treatment
  • periodic sampling drug content
  • lab test such as checking serum level may be helpful but must be confidential

Triple Blinded:
- patients, investigators and data monitoring committee are all blinded

  • disadvantage: DMC’s ability to monitor safety and efficacy can be hampered and this can be counterproductive
  • improvement: DMC is blinded at first but code can be broken upon request.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q
  1. Describe why it’s reasonable to plan for testing non-inferiority and then superiority, but not the other way around.
A

Non-inferiority is the gate keeper. Similar to the ANOVA, if ANOVA is significant then we go in and look in more detail.

Non-inferiority first because if you get significant result it is ok to go ahead and look at the superiority hypothesis.

Power - Superiority requires a very large power in order to detect significance. Sample size for non-inferiority trial is not required to be as large as for superiority.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q
  1. Show how an estimated sample size can incorporate an expected loss to follow-up rate.
A

Example: Assume a 15% loss to follow up that is assumed to be at random-

N*=N/(1-.15)

Example: Assume 15% loss to follow up and assume not at random thus additional sensitivity analysis may be required -

N*=N/((1-.15)^2)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q
  1. Show what the risk is of not considering the clustering effect in sample size estimation for cluster randomized trials
A

If cluster effect is not accounted for, then variance is underestimated, standard error looks smaller than it actually is and the likelihood of committing a type 1 error increases.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q
  1. Explain and show what is the most efficient design in terms of allocation (balanced vs unbalanced) between two study groups. What is the effect of unbalanced designs?
A

Most efficient is balanced allocation.

Increased variance, increased required sample size to achieve the same power

(Or if sample size is constant-> increased variance and decreased power)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q
  1. Describe what Cohen’s effect size is and general guidelines for magnitude of effect sizes.
A
Cohen's Effect Size:
-   based on behavioral science data
-    essentially the same as a z-score for two sample test with s-pooled as SE 
-   D=(mu1-mu2)/s-pooled
General Rules:
0.2<=D<0.5 (Small Effect)
0.5<=D<0.8 (Medium Effect)
D>=0.8(Large Effect)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q
  1. Describe what the differences are between the (1) Haybittle & Peto, (2) Pocock, and (3) O’Brien and Fleming sequential boundaries are with respect to the likelihood of stopping early and for the final look.
A

(1) Haybittle & Peto - constant until last time point and then increased change to reject null
(2) Pocock - constant throughout
(3) O’Brien and Fleming Increasing probability of rejecting null at every look

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q
  1. What is the advantage of alpha spending designs over group sequential designs?
A

Group sequential designs are cumbersome and alpha spending designs are less cumbersome.

  • Group sequential designs can have restrictive monitoring times that require equal increments of information (# patients) and sometimes causes administrative difficulties
  • Alpha spending are more flexible than group sequential designs so the idea is to spend(distribute) the total probability of false positive risk (Type I error) as a continuous function of the information time (alpha spending function)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q
  1. In a 3+3 design, how is the maximum tolerated dose determined?
A

3+3 Design:

  • Treat 3 participants at dose level k
    • If no DLT, escalate to dose level K+1
    • If 2+ DLTs, de-escalate to dose level K-1
    • If 1 DLT, treat 3 additional participants at dose level K
      - (Now 6 total participants) If 1 DLT, escalate to dose level K+1
      - (Now 6 total participants) If 2 DLT, de-escalate to dose level K-1

MTD is the highest dose where 0 or 1 DLT observed

NOTE: DOSE LEVELS MUST BE PRE-SPECIFIED

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q
  1. What are some strengths and limitations of the 3+3 design?
A
Limitations:
- Ignores dosage history other than the previous 3 patient cohort
-Imprecise and inaccurate MTD estimation
-low probability of selecting true MTD
-high variability of MTD estimates
-Dangerous outcomes
Can improve with 5+5 or CRM

Strengths?
- simple design, small, doesn’t require statistician.

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

Define Maximum Tolerated Dose.

A
  • Every person has a unique Maximum Tolerated Dose before reaching a Dose Limiting Toxicity
  • Ratios/Proportions of people at each dosage level for which that dose is their MTD
  • We look for the MTD that has a ratio of some target percentage (say 25%)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q
  1. Describe the intention-to-treat principle.
A

all participants randomized and all events, as defined in the protocol, should be accounted for in the primary analysis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q
  1. Describe the possible effect of dichotomization of a time to event outcome on the power of a trial.
A

When we dichotomize a time to event outcome, we lose information and thus we lose power in a trial.

24
Q
  1. Describe the possible effect of the dichotomization of a continuous outcome on the power of a trial.
A

When we dichotomize a continuous outcome in a trial we are losing information and thus we lose power in a trial.

25
Q
  1. Definition of a clinical trial
A

A prospective study comparing the effects and value of intervention(s) against a control in human beings.

26
Q
  1. Identify how multiple comparisons can affect type I error and be able to show how a sample size calculation can incorporate a Bonferroni correction.
A

Multiple Comparisons:
When we perform multiple comparisons without making adjustments, we increase the probability of identifying at least one outcome as significant that should not be. That is, the probability of committing a type I error increases if we do not make adjustments.

We can use a Bonferroni adjustment by dividing our desired alpha by the number of tests we wish to complete prior to completing them. We will then use the new generated threshold to complete all tests, including sample size calculations.

27
Q
  1. Describe the primary goals of Phase 1 drug trials.
A

Primary Concern: Safety
Participants: Healthy volunteers or patients who have already tried and failed to improve on existing standard therapies.
Goal: Estimate tolerability and characterize pharmacokinetics and pharmacodynamics. (Max. Tolerable Dose etc)
n is typically 15-30 participants
Clinical Considerations: Route, schedule and starting dose, Plan for escalation, patient selection, extent of toxicity
Statistical Considerations: Number of patients per dose, Well-defined escalation theme, stopping rule, decision rule for recommended dose

28
Q
  1. Describe what is meant by the efficacy/toxicity trade-off.
A

As dosage increases, in general, it is expected that efficacy will increase but so will toxicity. Thus there is a tradeoff between efficacy and toxicity as dosage increases and an “ideal” point at which a desirable efficacy is reached without overstepping a tolerable toxicity threshold.

29
Q
  1. What are some strengths and limitations of the Continuous Reassessment Model (CRM)?
A

CRM is Bayesian Based approach (uses prior distribution and observed outcome to determine posterior distribution)

Strengths:
-Relatively Precise estimation of MTD

Limitations:

  • More complex than 3+3
  • Requires Statistician and modeling
  • Requires assumption about dose/toxicity relationship
30
Q
  1. Write down the null and alternative hypotheses in a futility trial.
A

(Reversed from other trials)

Null:
New Treatment Mean >= Control Treatment Mean + Delta

Alternative:
New Treatment Mean < Control Treatment Mean + Delta

Notes:
Fail to Reject Null: Continue with New Treatment
Reject Null: Abandon New Treatment
Specifics of Null and Alternative can vary slightly depending on definition of “superior” direction (outcome definition is survival time vs number of adverse events, etc.)

31
Q
  1. Describe what the differences are in Type I and Type II errors between conventional designs and futility designs.
A

Conventional:
Type I Error (Alpha) - Probability of rejecting null (concluding intervention is effective) when the intervention in reality is NOT effective.

Type II Error (1-Beta) - Probability of failing to reject the null (concluding the intervention is NOT effective) when the intervention in reality IS effective.

Futility Trial:
Type I Error (Alpha) - Probability of rejecting null (concluding new treatment is futile) when the intervention in reality is effective.

Type II Error (1-Beta) - Probability of failing to reject the null (concluding the intervention is NOT futile) when the intervention in reality is NOT effective.

32
Q
  1. In cluster randomized trials, assume there is a positive correlation in the primary outcome for individuals within a cluster. Describe the impact on the alpha level if you ignore the design effect and analyze as if they are independent.
A

(NOTE: Alpha level here is described as the true probability of type I error)

If there is a positive correlation in the primary outcome for individuals within a cluster (Positive intraclass correlation) variation within the cluster is reduced, and if it is not adjusted for then the test statistic is artificially inflated and thus the probability of rejecting the null is increased, however the rejection of the null hypothesis in this case may be erroneous.

33
Q
  1. What are some specific features of futility designs that typically result in a smaller sample size compared to conventional efficacy designs?
A

Futility Design Features:

  • One Sided Test of Hypothesis (Or historical controls)
  • Looking to detect Larger differences
  • We care more about Type II error than type I error
  • We can power lower as result of error focus

Result: Less sample size required

34
Q
  1. Based on a figure of point estimates and confidence intervals, be able to explain whether one would conclude superiority, noninferiority, inconclusive, or inferior treatment.
A

“superior” - the entire confidence interval of the new treatment lies on the “superior” side of the null (no difference in treatment effect of new and old treatment).

“non-inferior” - the confidence interval of the new treatment covers the null (no difference) but NOT the null plus delta

“Inconclusive” - the confidence interval of the new treatment covers the null AND the null plus delta.

“inconclusive” - the upper bound of the new treatment is below the null, but the confidence interval covers the null plus delta delta.

“inferior” - the entire confidence interval of the new treatment is on the “inferior” side of the null plus delta

(NOTE: we can think of “null plus delta” could be number of bad outcomes if we are considering ‘plus delta’ to be a negative direction)

35
Q
  1. Be able to define what the margin of non-inferiority is. Also, be able to comment on the choice of this margin compared to an active control’s effect. For example, if an active control has a delta difference with placebo, in a non-inferiority trial for a new treatment compared to that active control, describe how the margin of non-inferiority may compare to this delta and the rationale.
A
  • maximum difference in responses that is considered to be clinically unacceptable (retain some % of active control’s effect)
  • smaller than differences observed in superiority trials of the active comparator

Aim:
New intervention is better than placebo and similar to active control

36
Q
  1. Describe what is meant by sub-group analyses.
A

a subset of participants in a clinical trial defined by a baseline characteristic (men/women; old/young; aspirin use (yes/no))

DO NOT DEFINE BY POST-BASELINE AS IT CAN BE AFFECTED BY TREATMENT

Always test for interaction with treatment (does the treatment work better in certain groups)

Consider linear model, test for interaction in the linear model and then if it is significant in the linear model then don’t look inside. If there is a significant interaction then we will examine treatment effect in each group.

Assessing an effect modifier.

37
Q
  1. What is the role of testing of interactions in sub-group analyses?
A

Always test for interaction with treatment (does the treatment work better in certain groups)

Consider linear model, test for interaction in the linear model and then if it is significant in the linear model then don’t look inside. If there is a significant interaction then we will examine treatment effect in each group.

38
Q
  1. Describe some ethical reasons for conducting interim analyses.
A

Detect benefit, safety, harm, or anything strongly indicating one of the treatments might be inferior or ineffective early

39
Q
  1. Describe administrative reasons for conducting interim analyses.
A
  • Ensure the study is being executed as planned.
  • ensure that only appropriate patients are enrolled
  • uncover the presence of unanticipated problems such as non-compliance
  • check on assumptions made in designing the trial such as patient accrual rate and sample size parameters
40
Q
  1. Describe economic reasons for conducting interim analyses.
A
  • Early stopping for negative results prevents wasting of resources, abandoning lost cause.
  • Allows for informed management decisions re: allocation of limited R&D funds
41
Q
  1. Describe the primary factors that go into the decision to terminate a study.
A

Decision to terminate:
*Risk-Benefit Ration - are the side effects/toxicities so severe that continuing is unethical?

External information - have other studies/information been conducted during this study that provide revelations that mean it would be unethical to continue?

42
Q
  1. Be able to define and give examples of the various missing data mechanisms: (1) missing completely at random; (2) missing at random; and (3) missing not at random.
A

(1) Missing Completely at Random (MCAR) :
* Some data are missing on Y (assumption)
* the probability of being missing is unrelated to the measurement Y or other variables X
* MCAR is best situation, can do complete case analysis and will just lose power but have valid results

(2) Missing At Random (MAR):
* Some data are missing on Y (assumption)
* the probability of being missing is unrelated to the measurement Y AFTER controlling for some other observed variable(s) X
* P(Y missing| X,Y)=P(Y missing|X)
* Almost the same as ignorable missingness, we don’t have to model missing data mechanism
* Weaker assumption than MCAR

(3) Missing Not at Random
* Some data are missing on Y (assumption)
* Probability of missing data is dependent on the value of the missing data
* Non-ignorable
* requires good prior knowledge about causes of missingness
* Must model missing data
* results may be very sensitive to choice of model
* sensitivity analysis is critical

43
Q
  1. Be able to reassign missing values in a table according to best-case and worst-case analyses.
A

Three Analyses:

  • Naive Analysis
    * ignore missing data
  • Best Case Analysis
    * assume all missing values in controls had bad outcome
    * all missing in experiment group had good outcome
  • Worst Case Analysis
    * assume all missing values in controls had good outcome
    * all missing in experiment group had bad outcome
44
Q
  1. Be able to explain the Last Observation Carried Forward method for missing data imputation and its main limitation.
A

LOCF:
*Select variables of interest that make records/patients “alike” (might be date within a single subject or multiple variables across subjects)
*sort records according to these variables
*find first missing values and use the cell value immediately prior to that record and use to impute the missing value
Example: Previous outcome of psychological evaluation carried forward for a patient

45
Q
  1. What is the advantage of multiple imputation versus single imputation methods?
A

Multiple Imputation:

  • Repeat single random imputation m times to create m data sets with complete data
  • Run m analyses (one on each of the m datasets)
  • Pooling - consolidate the m results by calculating the mean and variance and CI of the parameter estimates

Advantages:
* takes into account the uncertainty in the imputation process

46
Q
  1. Be able to identify and/or explain what a SMART trial is.
A

Sequential Multiple Assignment Randomized Trial

  • Sequence of individually tailored decision rules
  • specifies if/how and/or when to alter intensity/type/dosage and delivery of treatment at specific points
  • at each stage look at responders/non-responders and can do further randomization into increased intervention
47
Q
  1. Explain what the difference between basket, umbrella, and platform trials.
A

Basket:

  • targeted therapy
  • evaluated on multiple diseases
  • common predictive biomarkers or other common predictive patient characteristics
  • used to predict if patient will respond to intervention under unifying criteria

Umbrella:

  • Multiple targeted interventions
  • single disease stratified into subgroups by biomarker or patient characteristic

Platform Trials

  • intervention arms that can be dropped or added
  • Also known as multi-arm/multi-stage (MAMS) design trials
  • several interventions against common control
  • can be perpetual
  • prespecified adaptive rules
  • flexibility of adding new interventions
48
Q

Describe a Phase III (Full-scale evaluation of Treatment) clinical trial.

A

Goals:
-define the “best” treatment which has implication of changing current practice
N is typically hundreds or thousands of patients

Types:
Comparative Trial (New/treatment is different than standard/placebo)
Equivalence Trial (new treatment may not be better, but is cheaper/less side-effects)

Therapeutic trial(disease is present and being treated)
Prevention Trial (prevention of progression, perhaps second malignancies in cancer trials)
Individual-based trial
Community/population based trials

49
Q

Phase IV (Postmarketing Serveillance) Trial

A

Goals: Monitor adverse effects, long-term morbidity and mortality after treatment is applied to large number of patients and follow up for a long period of time

50
Q

What is the relationship between a Pilot study and a Phase II study?

A

Pilot study and Phase II studies have overlapping definitions, a Phase II studies are pilot studies focused on either efficacy or optimal dosage.

51
Q

Define Dose Limiting Toxicity.

A

DLT:
This describes the side effect that limits the dose amount that can be safely given.

  • a serious or life-threatening side effect
  • can be reversible
  • definition depends on clinical setting

(Side effects that are defined in the study that tell us when to stop increasing the dose, or when a maximum tolerated dose is reached by a subject)

52
Q

What is an active control trial?

A

A trial in which an experimental intervention is compared with an accepted standard intervention.

Objectives:

1) Superior to active control OR
2) as good as active control

53
Q

How do we choose the margin of non-inferiority delta?

A

Margin of Non-Inferiority Delta:* maximum difference in responses that is considered to be clinically unacceptable (retain some % of active control’s effect)
* smaller than differences observed in superiority trials of the active comparator

Aim:
New intervention is better than placebo and similar to active control

54
Q

Describe the key statistical features of a Futility Design study.

A

Futility Design:
*Formulation of null and alternative hypothesis are reversed from the norm
(smaller sample size):
-Single Arm Study
-More liberal Alpha
- one sided hypothesis
*Historical controls +/- Calibration/masking controls (Single arm studies in which subjects in the study receive experimental agent except in calibration/masking controls)

55
Q

Define a noninferiority Trial

A

A trial with the primary objective of showing that the response to the investigational product is NOT CLINICALLY INFERIOR to a comparative agent.

56
Q

What is the relationship between a Pilot study and a Phase II study?

A

Pilot study and Phase II studies have overlapping definitions, a Phase II studies are pilot studies focused on either efficacy or optimal dosage.