44
Clinical Trial Design Developed by Center for Cancer Research, National Cancer Institute, NIH Endorsed by the CTN SIG Leadership Group

Rethinking Oncology Clinical Trial Designclinicaltrial.vc.ons.org/file_depot/0-10000000/0-10000/3367/folder/... · Clinical Trial Design Developed by Center for Cancer Research, National

Embed Size (px)

Citation preview

Clinical Trial Design

Developed by Center for Cancer Research, National Cancer Institute, NIH

Endorsed by the CTN SIG Leadership Group

OverviewClinical research is research conducted on human beings (or on material of human origin such as tissues, specimens, and cognitive phenomena) with the goal of generating useful knowledge about human health and illness. A clinical trial is one type of clinical research that seeks to answer a scientific or medical question about the safety or potential benefit of an intervention such as a medication, device, teaching concept, training method, or behavioral change.

This module will provide an overview of clinical trial design. At the conclusion of this module, the learner will be able to:• Describe five types of clinical trials.• Discuss the objectives, endpoints and standard design for Phase I, II,

and III clinical trials.

Types of Clinical Trials

• Natural History • Prevention• Screening and Early Detection• Diagnostic• Quality-of-life and supportive care• Intervention/Treatment

Natural History Trials

A prospective study to determine the natural course of cancer when:

• Left untreated

• Treated with standard therapy

Prevention Trials:Chemoprevention

• Evaluate the effectiveness of ways to reduce the risk of cancer

• Enroll healthy people at high risk for developing cancer

• 2 types of trials:• Action studies – “do something” • Agent studies – “take something”

Screening and Early-Detection Trials• Assess new means of detecting cancer

earlier in asymptomatic people

• Tools:• Tissue sampling/procurement• Laboratory tests, including genetic testing• Imaging tests• Physical exams• History, including family hx (pedigree)

Diagnostic Trials• Develop better tests or procedures to

identify a suspected cancer earlier or more accurately

• Tools:• Imaging tests• Laboratory correlative studies/tumor

marker

Supportive Care/QOL Trials• Evaluate improvements in comfort of and

quality of life (QOL) for people who have cancer

• Seek better therapies or psychosocial interventions for subjects

• Focus on subjects AND families or caregivers

Imaging Trials

Imaging clinical trials differ from drug treatment trials in that the scientific question being asked is aimed at understanding if or how a specific imaging test can best be used to screen, diagnose, direct the treatment of, or monitor the response to a therapy for a disease.

Types of Imaging Trials• Screening for cancer

• determine if a person has any suspicious areas or abnormalities that might be cancerous.

• Diagnosis/staging• used to find out where a cancer is located in the body, if it has

spread, and how much is present• Guiding cancer treatments

• used to make cancer treatments less invasive by narrowly focusing treatments on the tumors

• Determining if a treatment is working• used to see if a tumor is shrinking or if the tumor has changed and

is using less of the body's resources than before treatment • Monitoring for cancer recurrence

• used to see if a previously treated cancer has returned or if the cancer is spreading to other locations

Treatment Clinical Trials• Test new treatments, new combination of

drugs, or new approaches to surgery or radiation therapy

• 4 Phases

Phase I TrialTo determine the appropriate dose for further evaluation

Phase II TrialTo determine whether an agent has activity against a specific cancer type

Phase III TrialTo determine whether a treatment is effective

Phase IV TrialPost FDA approval, various goals

Phases of Clinical Trials

Phase I: Primary Goal(s)Evaluate Toxicity:• Define dose limiting

toxicity (DLT)• Define maximum

tolerated dose (MTD)• Begin development of

side-effect profile

May provide early evidence of response, but NOT primary aim

Evaluate Pharmacokinetics PKs): ADMEHow the drug(s) is:• Absorbed• Distributed• Metabolized• Excreted

Phase I: Additional Goal(s)

• Also used to:

• Evaluate new treatment schedule

• Evaluate new drug combination strategy

• Evaluate new multi-modality regimen

Phase I: Patient Population

• 15 – 30 (< 100) subjects

• Usually many cancer types (e.g. solid tumors)

• Refractory to standard therapy

• No remaining standard therapy

• Adequate organ function

• Adequate performance status

Phase I: Standard Design

• Open label, non-randomized, dose escalation

• Low starting dose• 1/10th the lethal dose (LD10) in the most sensitive

species tested = dose at which 10% of the animals die• Unlikely to cause serious toxicity• Pediatric dose starts at 80% of adult MTD

• 3-6 patients per cohort

• Increase dose gradually• Most common scheme is a Modified Fibonacci

Classic Modified Fibonacci Dose Escalation Scheme

% Increase Above Preceding Dose:Level 1: Starting doseLevel 2: 100% increase from Level 1Level 3: 67% increase from Level 2Level 4: 50% increase from Level 3Level 5: 40% increase from Level 4Levels 6+: 33% increase from Level 5+

3 + 3 Phase 1 Study Design Schematic

Alternate DesignsIn addition to the 3 + 3 standard design, we are beginning to see alternate designs:

• “Accelerated” design: • E.g.: 1 subject enrolled per dose level until one grade 2

adverse event that is related to the investigational agent(s) is seen. Then the design returns to the 3 + 3 design as per previous schematic.

• “Intrapatient dose escalation”: • E.g.: once a dose level has been proven “safe” (not the

MTD) then subjects at lower levels are able to escalate to the “safe” level

• E.g.: once a subject is able to tolerate a dose level (no DLT) they are allowed to escalate to the next level using themselves as the own control

Phase I: Endpoints

• Dose Limiting Toxicity (DLT)• General DLT Criteria:

• ≥ Grade 3 non-heme toxicity• Grade 4 neutropenia lasting longer than 5 days• Grade 4 thrombocytopenia

• Typically the DLT is defined for the first course/cycle

• Maximum Tolerated Dose (MTD)• Highest dose level at which ≤1/6 patients develop a

DLT

Phase I: Limitations

• Questionable risks without benefits

• Initial patients may be treated at low (sub-therapeutic) doses

• Slow to complete trial (need to find fairly healthy advanced cancer patients)

• Toxicity may be influenced by extensive prior therapy

• Inter-patient variability

• MTD definition is imprecise

• Minimal data about cumulative toxicity since only the first cycle/course is taken into consideration for a DLT

Phase II: Primary Goals

Evaluate activity

Further safety (adverse events) evaluation at the MTD

Phase II: Patient Population

• ~100 subjects (100-300)

• More homogenous population that is deemed likely to respond based on:• phase I data• pre-clinical models, and/or • mechanisms of action

• Subject needs to have measurable disease

• May limit number of prior treatments

Phase II: Standard Design

Two-stage design with early stopping rule for efficacy or futility

Stage 1 (n=9)Single Agent – Single Dose

Inactive ActiveStage 2 (n=24)

ActiveInactive

≥3/24<3/24

0/9 ≥1/9

Phase II: Alternate Design…

Low dose vs. higher dose Placebo (Inactive) vs agent

Randomization (n=50)Randomization (n=50)

25 25

New Agent/ InterventionLow Dose

New Agent/ Intervention Higher Dose

25 25

New agent alone or

+ placebo

New agent + standard of

care

…Phase II: Alternate Design

Active agent administered over defined time frame, i.e., 16 weeks

Stable Disease?

No

Yes

RANDOMIZE

Active Agent

Placebo*

Randomized Discontinuation Design

Off study

* Patients with progressive disease on placebo can switch back to active agent.

Phase II: Endpoints

• Response (see response assessment module for more details)• Complete Response (CR)

• Partial Response (PR)

• Stable Disease (SD)

• Progressive Disease (PD)

• Additional safety data

Phase II: Limitations

• Lack of activity may not be valid

• Measurable disease required

• No internal control group

Phase III: Primary Goals

Efficacy compared to standard therapy • Activity demonstrated in Phase II study

Further evaluation of safety

Phase III: Patient Population

• Hundreds to thousands of subjects

• Single cancer type

• May be front-line therapy

• Well-defined eligibility criteria

• Internal control group (e.g., standard treatment, placebo)

• Multi-institutional participation necessary to reach targeted accrual goals

Phase III: Standard Design

• Randomized assignment of patients to treatment arms

• Equal distribution of known important prognostic factors to each arm (stratification)

Presenter
Presentation Notes
In phase III trials (and some phase II trials), participants are assigned to either the investigational or control group by chance, via a computer program or table of random numbers. This process, called randomization, gives each person the same chance of being assigned to either group. Randomization ensures that known or unknown factors do not influence the trial results so that the treatment/intervention being compared receive a fair test. If physicians or participants themselves chose the therapy participants would receive, assignments might be biased. Bias results from collecting the data in such a way that one answer to the research question is given undue favor over another. Physicians, for instance, might unconsciously assign younger participants with a more hopeful prognosis to the experimental group, thus making the new therapy seem more effective than it really is. Conversely, participants with a less hopeful prognosis might pick the experimental treatment, leading it to look less effective than it really is. Stratification is used in randomized trials when factors that can influence the intervention’s success are known. For example, participants in cancer prevention trials are separated or stratified by risk factors such as age since cancer risk increases with older age. Assignment of interventions within the stratified groups is then randomized. Stratification enables researchers to look in separate subgroups to see whether differences exist, and it ensures that the two groups are balanced for the known risk factors.

Phase III: Other Considerations• Sample size chosen to detect difference• Intention-to-treat analysis may be used

• strategy for the analysis of randomized controlled trials that compares patients in the groups to which they were originally randomly assigned whether they complied with the treatment they were given

• Data Safety Monitoring Committee/Board (DSMC/DSMB) for interim analysis

Presenter
Presentation Notes
The number of participants to be included in a phase III trial (or sample size) is determined using statistical analysis. The sample size must be sufficiently large enough to detect a difference when the study is completed. “Intention to treat” is a strategy for the analysis of randomized controlled trials that compares patients in the groups to which they were originally randomly assigned. This applies regardless of whether they complied with the treatment they were given. An analysis that excludes noncompliant patients is no longer randomized and might cause serious bias. Noncompliant patients tend to have worse outcomes than compliant patients. It is therefore possible that excluding noncompliant patients from the analysis leaves those who may have a better outcome and destroys the unbiased comparison provided by randomization. Excluding noncompliant patients may also result in ignoring the influence of poor tolerability on the efficacy of a treatment. Care must therefore be taken to minimize missing data and to follow up those who withdraw form treatment. A Data & Safety Monitoring Committee (DSMC, sometimes called data & safety monitoring board or DSMB) is an independent group of experts who monitor unblinded safety and efficacy data while a trial is ongoing. The timing of DSMC meetings is typically based on the number of patients with endpoint data. DSMC meetings might be convened based on some threshold, for example occurrence of more than two serious adverse events (or another pre-determined number) or they may be regularly scheduled meetings in which data for multiple trials reaching a pre-determined boundary are reviewed. An example of a pre-determined boundary would be the DSMC will review the study after every 200 participants are enrolled. The DSMC can recommend making changes to the conduct of the trial, including recommending to terminate the trial early for safety concerns, for overwhelming treatment efficacy, or for demonstrated futility in being able to show a benefit of treatment. DSMCs have generally been established for large, randomized multisite studies that evaluate interventions intended to prolong life or reduce risk of a major adverse health outcome such as recurrence of cancer. The role of the DSMC is advisory to the sponsor and study leadership of the trial. The sponsor has ultimate responsibility for making decisions about the trial.

Randomized Study DesignsTwo Commonly Used Randomized Designs:• Parallel Group Design

• Subject randomized to 1 of 2 or more arms with each arm being a different treatment

• Crossover Design• Subject randomized to a sequence of 2 or more

treatments• Subject acts as own control• May “crossover” to other treatment for progressive

disease• May “crossover” to other treatment after treatment

course completed

Phase III: Endpoints

• Efficacy• Overall survival• Disease-free survival• Progression-free survival• Symptom control• Quality of life

Phase III: Limitations

• Difficult, complex, expensive to conduct

• Large number of patients required

• Incorporation of results into front-line therapy in community is often slow and incomplete

FDA Cancer Approval Endpoints

• Overall survival• Endpoints based on tumor assessments• Symptom endpoints (PROs)

The next few slides will define the endpoints.

Overall Survival

• Time from randomization until death• Intent-to-treat population

Endpoints: Tumor Assessments…• Disease-free survival

• Randomization until recurrence of tumor or death from any cause

• Adjuvant setting after definitive surgery or radiotherapy

• Large % of patients achieve CR after chemo• Objective response rate (ORR)

• Proportion of patients with reduction of tumor size of a predefine amount and for a minimum time period

• Measure from time of initial response until progression

• Sum of PRs + CRs• Use standardized criteria when possible

• Progression free survival (PFS)• Randomization until objective tumor progression or

death• Preferred regulatory endpoint• Assumes deaths are r/t progression

• Time to Progression (TTP)• Randomization until objective tumor progression,

excluding deaths• Time-to-treatment failure (TTF)

• Randomization to discontinuation of treatment for any reason (PD, toxicity, death, etc.)

• Not recommended for regulatory drug approval

…Endpoints: Tumor Assessments

Presenter
Presentation Notes
See Appendix 1 in Guidance Document

Endpoints: Symptom Assessment

• Time to progression of cancer symptoms• FDA Guidance: Patient-Reported Outcome

Measures: Use in Medical Product Development to Support Labeling Claims

• Tools/surveys• Issues:

• Missing data• Infrequent assessments

Phase IV TrialFollow-up investigation to further evaluate long-term safety and effectiveness of a recently approved drug:

• Further assess risk/benefit ratio• Further evaluation of efficacy • Further evaluation of toxicity• Further evaluation in other populations (e.g.,

elderly)• Facilitate integrate new treatment into primary

therapy• Costly to conduct so often not done

References• Adjei, A.A., Christian, M., Ivy, P. (2009). Novel

Designs and End Points for Phase II Clinical Trials. Clinical Cancer Research, 15(6), 1866-1872.

• Kummar, S., Gutierrez, M., Doroshow, J., Murgo, A.J. (2006). Drug development in oncology: classical cytotoxics and molecularly targeted agents. British Journal of Clinical Pharmacology, 62(1), 15-26.

Module Evaluation

The CTN SIG would greatly appreciate your feedback on this learning module. Please complete the evaluation form and fax to Elizabeth Ness at 301-496-9020.