• 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

1/29

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;

29 Cards in this Set

  • Front
  • Back

Describe a Phase I Study Design for Clinical Trials


Phase I: Researchers test a new ____ or ____ in a ____ group of people for the first time to evaluate its ____ , determine a safe ____ ____ , and identify ____ .

Phase I: Researchers test a new drug or treatment in a small group of people for the first time to evaluate its safety, determine a safe dosage range, and identify side effects.

Describe a Phase II Study Design for Clinical Trials


Phase II: The drug or treatment is given to a ____ group of people to see if it is ____ and to further evaluate its ____ .

Phase II: The drug or treatment is given to a larger group of people to see if it is effective and to further evaluate its safety.

Describe a Phase III Study Design for Clinical Trials


Phase III: The drug or treatment is given to ____ groups of people to confirm its ____ , monitor ____ , ____ it to commonly used treatments, and collect information that will allow the drug or treatment to be used ____ .

Phase III: The drug or treatment is given to large groups of people to confirm its effectiveness, monitor side effects, compare it to commonly used treatments, and collect information that will allow the drug or treatment to be used safely.

Describe a Phase IV Study Design for Clinical Trials


Phase IV: Studies are done after the drug or treatment has been ____ to gather information on the drug's effect in various ____ and any side effects associated with ____ - term use.

Phase IV: Studies are done after the drug or treatment has been marketed to gather information on the drug's effect in various populations and any side effects associated with long- term use.

Dose Escalation Design


What phase is this design under?


What does it determine?


What is monitored?


Type and level/grade of _____ specified in ____


Give an example of a dose escalation design.


Phase I


Determine maximum tolerated dose (MTD)
Monitor whether each patient experience a dose limiting toxicity (DLT)


Type and level/grade of toxicity specified in protocol


Traditional 3+3 Design

Single-arm design



What phase is this design under?


Usually how many patients?


What does it assess?

Phase II


Usually < 100 patients


Continued assessment of safety in addition to effectiveness

Multi-arm parallel design


What phase is this design under?


Name different types.


Which is referred to as the "control" group?


What does it assess?


What does parallel refer to?

Phase II


Two or more treatment arms/groups


Two-arm randomized controlled trial


Three-arm randomized controlled trial


Placebo or SOC (standard of care)


Continued assessment of safety in additional to main interest in efficacy


“Parallel” refers to the fact that each patient receives only one treatment and each treatment group proceed in parallel

Describe a Randomization design


What does it ensure?


Who does the randomization?

Two or more treatment arms/groups


Randomized Treatment Assignment


Ensures that patients are assigned to study arms without bias (systematic differences in baseline characteristics)


Independent, e.g. statistician, Centralized randomization for multi-site studies, Data coordinating center, pharmacist, Many computer programs

Block Randomization


For _____ number of patients, randomization does not guarantee ______ number of patients in treatment and control group

small, similar


Could do randomization in blocks, e.g., for 40 patients


a block size of 4


possible balanced combinations with 2 C (control) and 2 T (treatment) subjects are (TTCC, TCTC, TCCT, CTTC, CTCT, CCTT)


blocks are randomly chosen to determine the assignment of all 40


subjects,


e.g., one random block sequence: [TTCC / TCCT / CTTC / CTTC / TCCT /CCTT / TTCC / TCTC / CTCT / TCTC])


This ensures that there’s 20 subjects in both the control and treatment groups (equal-sized study groups)

Stratified Randomization


What is important to balance?


Examples of stratification factors?


Only feasible with ____ factors

It is important to balance baseline factors that may be associated with the response to treatment (prognostic factors)


• E.g. age or baseline severity/illness


Stratification factors – Stratified Randomization


Two factors: male, female & age (<18, >=18) -> 4 strata/groups


Separate randomization within each stratum


Multisite study: Randomization within each study site (standard of care maybe different by site)


Only feasible with a few factors; 2 or 3 especially for small trials


Common example: (a) stratified by study site (b) block randomization within site

Blinding


What are other sources of bias?


Describe single-blinding


Describe double-blinding


What is the most "rigorous" design?

There are other sources of bias during the study (after randomization), e.g.


Patients knowing they are on placebo may drop out early


For investigators, outcome assessment is more objective if s/he does not know which treatment group patient is on (e.g., subconscious bias)


Single-blinding means patient doesn’t which treatment s/he is on


Double-blinding means neither patient or clinician knows ...


Most “rigorous” design: Blinded, placebo-control

How should Unblinding Procedure be done?


What is the gold standard of unblinding?

Unblinding (breaking the code) should be done as specified I the protocol


Ideally/gold-standard is to do this at the end of the study / study close


This is not the same as when each patient complete his/her participation (e.g., 8 weeks of treatment for 100 patients recruited over


4 years)

Any premature unblinding should be


_______ an _______ accordingly (e.g., sponsor)


Stated in final study publication as potential source of _______

Documented an reported accordingly (e.g., sponsor)


Stated in final study publication as potential source of bias

Randomized Placebo-Controlled


Placebo group should have minimal ______/______(“______effect”)


Main use: Show drug/device (investigational product) have improved ______ over a ______ control


Also, design is very useful for understanding ______ and ______effects (commonly “____________”)


Design typically used for treatment administered for a ______ time period

minimal benefit/efficacy (“placebo effect”)


Show drug/device (investigational product) have improved efficacy over a placebo control


understanding safety and adverse effects (commonly “blinded randomized placebo-control”)


for treatment administered for a short time period

Describe Crossover Design


What is critical to avoid crossover effects?

Patients randomized to a sequence of treatment


Example: 2 treatments (A=treatment and B=placebo), 2 periods


Patients randomized to the sequence AB or BA


Wash out period in between treatments (A and B) is very critical to avoid crossover effects


Note: also short treatment period

Limitations to Crossover Design


______ effect; Try to avoid by design with adequate ______


One must understand the disease; it should be ______ /condition and treatment doesn’t “cure” condition
Behavioral or educational intervention, physical training etc. – can’t undo or wash out



Advantages


Each subject “serving as own ______


More ______ (fewer patients needed) to estimate treatment efficacy compared to parallel design


All patients will get “______ ” intervention/treatment

Limitations


Carryover effect; Try to avoid by design with adequate washout


One must understand the disease; it should be stable/condition and treatment doesn’t “cure” condition
Behavioral or educational intervention, physical training etc. – can’t undo or wash out



Advantages


Each subject “serving as own control”


More efficient (fewer patients needed) to estimate treatment efficacy compared to parallel design


All patients will get “active” intervention/treatment

Describe Phase III


Sample size?


What is needed for management of data, data collection, randomization, interim analysis etc.


What type of duration?


What is challenging about this design?



Larger sample size, often multi-center/site e.g. > 300 people


Need data coordinating center for management of data, data collection, randomization, interim analysis etc.


Longer duration


Expensive – logistically most challenging

Phase IV


What type of surveillance?


Duration and type of study?


When is it conducted?


What type of study design is typically used?


What happens if AEs are discovered?

Postmarketing surveillance, safety surveillance (pharmacovigilance)


long-term, longitudinal studies


After FDA approval


Study design used typically observational, epidemiological methods, e.g., case-control, case series design; multivariate modeling (“uncontrolled”/no control – not randomized controlled trial)


Adverse effects discovered in Phase IV may lead to restricted drug use or being withdrawn from market (e.g., Vioxx)

Equivalence/Non-inferiority Studies


Treatment A is less ______ and have similar benefit (efficacy) compared to standard treatment B


We might tolerate a small reduction in ______ but we gain in having much less ______ side effects


Study null hypothesis: Treatment A is less efficacious than standard treatment B by a ______ amount Δ (alternative hypothesis is that A=B)


Δ is call the______

Treatment A is less toxic and have similar benefit (efficacy) compared to standard treatment B


We might tolerate a small reduction in efficacy but we gain in having much less adverse side effects


Study null hypothesis: Treatment A is less efficacious than standard treatment B by a small amount Δ (alternative hypothesis is that A=B)


Δ is call the non-inferiority margin

Bioequivalence Clinical Trials


A CT whose objective is to demonstrate that ___ or ____ formulations of the same drug have comparable _______


Bioavailability = ___ and ___ an active drug gets to the ___ (and hence available to tissue and target organ)

A CT whose objective is to is to demonstrate that 2 or more formulations of the same drug have comparable bioavailability


Bioavailability = extent and rate an active drug gets to the bloodstream (and hence available to tissue and target organ)

Describe Observational studies


Which concepts do and do not apply?

No intervention


All concepts apply, except randomization, blinding etc.

Studies that's not Clinical Trial


“_________” studies, e.g., to assess _________?
• When possible, incorporate _________and _________ (biosample selection, raters, blind biosample sample labeling etc.)

“Laboratory” studies, e.g., to assess mechanism?
• When possible, incorporate blinding and randomization (biosample


selection, raters, blind biosample sample labeling etc.)

Adaptive Designs


Adaptive designs in clinical trials may allow for changes in:


______/______criteria


Study ______, ______ and treatment ______


______ method


Inclusion/exclusion criteria


Study dose, end points and treatment durations


Analysis method

Adaptive Designs:


A ________ (GSD) is an adaptive/flexible design that allows for early _______ of a trial based on interim analysis for:


_______ , _______ , _______


Statistician work with _______ to plan


ahead these data _______ events

A group sequential design (GSD) is an adaptive/flexible design that allows for early stopping of a trial based on interim analysis for:


Safety, Efficacy, Futility


Statistician work with investigator to plan


ahead these data monitoring events

Data Safety and Monitoring Board (DSMC)


What do they play a role in?


What is the common misconception?

Reviewing baseline data, safety and toxicity data, effectiveness of randomization


Early study termination


Planned efficacy analysis


Etc., a DSMC charter is created prior to start


May make recommendation including early study termination, study protocol modification


Common misconception (still...) DSM Plan: “The PI will review all data...” (w/o independent DSMC)

Protocol Development: Main Elements of Protocol


The protocol sets the ______ by which study will be conducted



List the content

The protocol sets the standards by which study will be conducted


Objectives: primary, secondary etc. must be clearly defined


Background: Justification for pursuing the objectives; include biologic rationale, clinical observation, state-of-knowledge, response to “SOC” or preliminary data to justification statistical consideration and study design


Investigational product/drug information


Stage definition (oncology)
Eligibility criteria, selection of subjects
Stratification factors
Treatment plan: detailed description, table of dose, time, administration etc.
Treatment modification: e.g. under excess toxicity
Subject withdrawal
Study calendar
Efficacy assessment and endpoint definition: response, survival etc.


Recognized efficacy parameters for the given area; RECIST for solid tumors; CGI- Clinical Global Impression e.g. autism
Safety is the endpoint in some studies


Trial design
Statistical considerations: Statistical analysis plan, power analysis


Safety evaluation to protect subject: med hist., baseline data at


enrollment, AE reporting, clinical/lab tests – area-specific


Ethics


Data management


Quality control and quality assurance procedures


Appendix/Supplement

What goes into the amendments of the protocol?

Sponsor and IRB approval

Why is data management important?

Faulty data compromises studies

Study Reporting: CONSORT Standards


What does CONSORT stand for?


Why was it developed?


How many items on the checklist?

Consolidated Standards of Reporting Trials


Developed to alleviate the problems arising from inadequate reporting


of RCTs.


25-item checklist (first 8 ...)