Cancer Clinical Trials in the Genomic Era - PowerPoint PPT Presentation

About This Presentation
Title:

Cancer Clinical Trials in the Genomic Era

Description:

Predictive Biomarkers Estrogen receptor over-expression in breast cancer Anti-estrogens, aromatase inhibitors HER2 amplification in breast cancer Trastuzumab, ... – PowerPoint PPT presentation

Number of Views:199
Avg rating:3.0/5.0
Slides: 138
Provided by: rsi9
Learn more at: https://brb.nci.nih.gov
Category:

less

Transcript and Presenter's Notes

Title: Cancer Clinical Trials in the Genomic Era


1
Cancer Clinical Trials in the Genomic Era
  • Richard Simon, D.Sc.
  • Chief, Biometric Research Branch
  • National Cancer Institute
  • http//brb.nci.nih.gov

2
  • Prognostic biomarkers
  • Measured before treatment to indicate long-term
    outcome for patients untreated or receiving
    standard treatment
  • May reflect both disease aggressiveness and
    effect of standard treatment
  • Used to determine who needs more intensive
    treatment
  • Predictive biomarkers
  • Measured before treatment to identify who will
    benefit from a particular treatment

3
  • Endpoint
  • Measured before, during and after treatment to
    monitor pace of disease and treatment effect
  • Pharmacodynamic (phase 0-1)
  • Does drug hit target
  • Intermediate response (phase 2)
  • Does drug have anti-tumor effect
  • Surrogate for clinical outcome (phase 3)

4
Prognostic Predictive Biomarkers
  • Single gene or protein measurement
  • Scalar index or classifier that summarizes
    contributions of multiple genes

5
Prognostic Predictive Biomarkersin Genomic
Oncology
  • Many cancer treatments benefit only a minority of
    patients to whom they are administered
  • Being able to predict which patients are likely
    to benefit can
  • Help patients get an effective treatment
  • Help control medical costs
  • Improve the success rate of clinical drug
    development

6
Validation Fitness for Intended Use
7
Biomarker Validity
  • Analytical validity
  • Measures what its supposed to
  • Reproducible and robust
  • Clinical validity (correlation)
  • It correlates with something clinically
  • Medical utility
  • Actionable resulting in patient benefit

8
Clinical Utility
  • Biomarker informs action that benefits patient by
    improving treatment decisions
  • Identify patients who have very good prognosis on
    standard treatment and do not require more
    intensive regimens
  • Identify patients who are likely or unlikely to
    benefit from a specific regimen

9
ObjectiveUse biomarkers to
  • Develop effective treatments
  • Know who needs these treatments and who benefits
    from them

10
Prognostic markers
  • There is an enormous published literature on
    prognostic markers in cancer.
  • Very few prognostic markers (factors) are
    recommended for measurement by ASCO, are approved
    by FDA or are reimbursed for by payers. Very few
    play a role in treatment decisions.

11
Prognostic Biomarkers Can be Therapeutically
Relevant
  • lt10 of node negative ER breast cancer patients
    require or benefit from the cytotoxic
    chemotherapy that they receive

12
OncotypeDx Recurrence Score
  • Intended use
  • Patients with node negative estrogen receptor
    positive breast cancer who are going to receive
    an anti-estrogen drug following local
    surgery/radiotherapy
  • Identify patients who have such good prognosis
    that they are unlikely to derive much benefit
    from adjuvant chemotherapy

13
  • Selected patients relevant for the intended use
  • Analyzed the data to see if the recurrence score
    identified a subset with such good prognosis that
    the absolute benefit of chemotherapy would at
    best be very small in absolute terms
  • Used an analytically validated test

14
(No Transcript)
15
Major problems with prognostic studies of gene
expression signatures
  • Inadequate focus on intended use
  • Reporting highly biased estimates of predictive
    value

16
Major problems with prognostic studies of gene
expression signatures
  • Inadequate focus on intended use
  • Cases selected based on availability of specimens
    rather than for relevance to intended use
  • Heterogeneous sample of patients with mixed
    stages and treatments. Attempt to disentangle
    effects using regression modeling
  • Too a great a focus on which marker is prognostic
    or independently prognostic, not whether the
    marker is effective for intended use

17
  • Goodness of fit is not a proper measure of
    predictive accuracy
  • Odds ratios and hazards ratios are not proper
    measures of prediction accuracy
  • Statistical significance of regression
    coefficients are not proper measures of
    predictive value

18
Goodness of Fit vs Prediction Accuracy
  • For pgtn problems, fit of a model to the same data
    used to develop it is no evidence of prediction
    accuracy for independent data

19
(No Transcript)
20
Validation of Prognostic Model
  • Completely independent validation dataset
  • Splitting dataset into training and testing sets
  • Evaluate 1 completely specified model on test set
  • Cross-validation

21
Leave-one-out Cross Validation for Classifier of
Two Classes
  • Full dataset P1,2,,n
  • Omit case 1
  • V11 T12,3,,n
  • Develop classifier using training set T1
  • Classify cases in V1 and count whether
    classification is correct or not
  • Repeat for case 2,3,
  • Total number of mis-classified cases

22
Complete cross Validation
  • Cross-validation simulates the process of
    separately developing a model on one set of data
    and predicting for a test set of data not used in
    developing the model
  • All aspects of the model development process must
    be repeated for each loop of the cross-validation
  • Feature selection
  • Tuning parameter optimization

23
Cross Validation
  • The cross-validated estimate of misclassification
    error is an estimate of the prediction error for
    the model fit applying the specified algorithm to
    full dataset

24
Prediction on Simulated Null DataSimon et al. J
Nat Cancer Inst 9514, 2003
  • Generation of Gene Expression Profiles
  • 20 specimens (Pi is the expression profile for
    specimen i)
  • Log-ratio measurements on 6000 genes
  • Pi MVN(0, I6000)
  • Can we distinguish between the first 10
    specimens (Class 1) and the last 10 (Class 2)?
  • Prediction Method
  • Compound covariate predictor built from the
    log-ratios of the 10 most differentially
    expressed genes.

25
(No Transcript)
26
Cross-validation Estimate of Prediction Error
27
(No Transcript)
28
  • Partition data set D into K equal parts
    D1,D2,...,DK
  • First training set T1D-D1
  • Develop completely specified prognostic model M1
    using only data T1
  • eg
  • Using M1, compute prognostic score for cases in
    D1
  • Develop model M2 using only T2 and then score
    cases in D2

29
  • Repeat for ... TK -gt MK -gt DK
  • Group patients into 2 or more risk groups based
    on their cross-validated scores
  • Calculate Kaplan-Meier survival curve for each
    risk-group

30
(No Transcript)
31
  • To evaluate significance, the log-rank test
    cannot be used for cross-validated Kaplan-Meier
    curves because the survival times are not
    independent

32
  • Statistical significance can be properly
    evaluated by approximating the null distribution
    of the cross-validated log-rank statistic
  • Permute the survival times and repeat the entire
    cross-validation procedure to generate new
    cross-validated K-M curves for low risk and high
    risk groups
  • Compute log-rank statistic for the curves
  • Repeat for many sets of permutations

33
Predictive Biomarkers
  • Cancers of a primary site often represent a
    heterogeneous group of diverse molecular entities
    which vary fundamentally with regard to
  • the oncogenic mutations that cause them
  • their responsiveness to specific drugs

34
  • In most positive phase III clinical trials
    comparing a new treatment to control, most of the
    patients treated with the new treatment did not
    benefit.
  • Adjuvant breast cancer 70 long-term
    disease-free survival on control. 80
    disease-free survival on new treatment. 70 of
    patients dont need the new treatment. Of the
    remaining 30, only 1/3rd benefit.

35
Predictive Biomarkers
  • Estrogen receptor over-expression in breast
    cancer
  • Anti-estrogens, aromatase inhibitors
  • HER2 amplification in breast cancer
  • Trastuzumab, Lapatinib
  • OncotypeDx gene expression recurrence score in N
    ER breast cancer
  • Low score -gt not responsive to chemotherapy
  • KRAS in colorectal cancer
  • WT KRAS cetuximab or panitumumab
  • EGFR mutation in NSCLC
  • EGFR inhibitor
  • V600E mutation in BRAF of melanoma
  • vemurafenib
  • ALK translocation in NSCLC
  • crizotinib

36
(No Transcript)
37
(No Transcript)
38
(No Transcript)
39
Standard Paradigm of Phase III Clinical Trials
  • Broad eligibility
  • Base primary analysis on ITT eligible population
  • Dont size for subset analysis, allocate alpha
    for subset analysis or trust subset analysis
  • Only believe subset analysis if overall treatment
    effect is significant and interaction is
    significant

40
Standard Paradigm Sometimes Leads to
  • Treating many patients with few benefiting
  • Small average treatment effects
  • Problematic for health care economics
  • Inconsistency in results among studies
  • False negative studies

41
The standard approach to designing phase III
clinical trials is based on two assumptions
  • Qualitative treatment by subset interactions are
    unlikely
  • Costs of over-treatment are less than costs
    of under-treatment

42
Subset Analysis
  • In the past generally used as secondary analyses
  • Numerous subsets examined
  • No control of type I error
  • Trial not sized for subset analysis

43
  • Neither conventional approaches to subset
    analysis nor the broad eligibility paradigm are
    adequate for genomic based oncology clinical
    trials
  • We need a prospective approach that includes
  • Preserving study-wise type I error
  • Sizing the study for the primary analysis that
    includes any subset analysis
  • If there are multiple subsets, replacing subset
    analysis with development and internal unbiased
    evaluation of an indication classifier

44
  • Although the randomized clinical trial remains of
    fundamental importance for predictive genomic
    medicine, some of the conventional wisdom of how
    to design and analyze rcts requires
    re-examination
  • The concept of doing an rct of thousands of
    patients to answer a single question about
    average treatment effect for a target population
    presumed homogeneous with regard to the direction
    of treatment efficacy in many cases no longer has
    an adequate scientific basis

45
  • How can we develop new drugs in a manner more
    consistent with modern tumor biology and obtain
    reliable information about what regimens work for
    what kinds of patients?

46
Development is Most Efficient When the Scientific
Basis for the Clinical Trial is Strong
  • Having an important molecular target
  • Having a drug that is deliverable at a dose and
    schedule that can effectively inhibit the target
  • Having a pre-treatment assay that can identify
    the patients for whom the molecular target is
    driving progression of disease

47
When the Biology is Clear the Development Path is
Straightforward
  • Develop a classifier that identifies the patients
    likely (or unlikely) to benefit from the new drug
  • Develop an analytically validated test
  • Measures what it should accurately and
    reproducibly
  • Design a focused clinical trial to evaluate
    effectiveness of the new treatment in test
    patients

48
Using phase II data, develop predictor of
response to new drug
Targeted (Enrichment) Design
49
Predictive Biomarkers
  • Estrogen receptor over-expression in breast
    cancer
  • Anti-estrogens, aromatase inhibitors
  • HER2 amplification in breast cancer
  • Trastuzumab, Lapatinib
  • OncotypeDx gene expression recurrence score in
    breast cancer
  • Low score for ER node - -gt no chemotherapy
  • KRAS in colorectal cancer
  • WT KRAS cetuximab or panitumumab
  • EGFR mutation in NSCLC
  • EGFR inhibitor
  • V600E mutation in BRAF of melanoma
  • vemurafenib
  • ALK translocation in NSCLC
  • crizotinib

50
Evaluating the Efficiency of Targeted Design
  • Simon R and Maitnourim A. Evaluating the
    efficiency of targeted designs for randomized
    clinical trials. Clinical Cancer Research
    106759-63, 2004 Correction and supplement
    123229, 2006
  • Maitnourim A and Simon R. On the efficiency of
    targeted clinical trials. Statistics in Medicine
    24329-339, 2005.

51
  • Relative efficiency of targeted design depends on
  • proportion of patients test positive
  • specificity of treatment effect for test positive
    patients
  • When less than half of patients are test positive
    and the drug has minimal benefit for test
    negative patients, the targeted design requires
    dramatically fewer randomized patients than the
    standard design in which the marker is not used

52
Two Clinical Trial Designs
  • Standard design
  • Randomized comparison of new drug E to control C
    without the test for screening patients
  • Targeted design
  • Test patients
  • Randomize only test patients
  • Treatment effect D in test patients
  • Treatment effect D- in test patients
  • Proportion of patients test is p
  • Size each design to have power 0.9 and
    significance level 0.05

53
RandRat nuntargeted/ntargeted
  • If D-0, RandRat 1/ p2
  • if p0.5, RandRat4
  • If D- D/2, RandRat 4/(p 1)2
  • if p0.5, RandRat16/91.77

54
Comparing T vs C on Survival or DFS5 2-sided
Significance and 90 Power
Reduction in Hazard Number of Events Required
25 509
30 332
35 227
40 162
45 118
50 88
55
  • Hazard ratio 0.60 for test patients
  • 40 reduction in hazard
  • Hazard ratio 1.0 for test patients
  • 0 reduction in hazard
  • 33 of patients test positive
  • Hazard ratio for unselected population is
  • 0.330.60 0.671 0.87
  • 13 reduction in hazard

56
  • To have 90 power for detecting 40 reduction in
    hazard within a biomarker positive subset
  • Number of events within subset 162
  • To have 90 power for detecting 13 reduction in
    hazard overall
  • Number of events 2172

57
TrastuzumabHerceptin
  • Metastatic breast cancer
  • 234 randomized patients per arm
  • 90 power for 13.5 improvement in 1-year
    survival over 67 baseline at 2-sided .05 level
  • If benefit were limited to the 25 test
    patients, overall improvement in survival would
    have been 3.375
  • 4025 patients/arm would have been required

58
Web Based Software for Planning Clinical Trials
of Treatments with a Candidate Predictive
Biomarker
  • http//brb.nci.nih.gov

59
Regulatory Pathway for Test
  • Companion diagnostic test with intended use of
    identifying patients who have disease subtype for
    which the drug is proven effective

60
Implications for Early Phase Studies
  • Need to design and size early phase studies to
    discover an effective predictive biomarker for
    identifying the correct target population
  • Need to establish an analytically validated test
    for measuring the predictive marker in the phase
    III pivotal studies

61
When the drug is specific for one target and the
biology is well understood
  • May need to evaluate several candidate tests
  • e.g. protein expression of target or
    amplification of gene
  • Phase II trials sized for adequate numbers of
    test positive patients and to determine
    appropriate cut-point of positivity

62
When the drug has several targets or the biology
is not well understood
  • Should biologically characterize tumors for all
    patients on phase II studies with regard to
    candidate targets and response moderators
  • Phase II trials sized for evaluating candidates
  • Opportunity for sequential and adaptive designs
    to improve efficiency

63
Empirical screening of expression profiles or
mutations to develop predictive marker
  • Should re-think whether to develop the drug
  • Larger sample size required
  • Dobbin, Zhao, Simon, Clinical Ca Res 14108,
    2008.
  • Use of archived samples from previous negative
    phase III trial
  • Use of large disease specific panel of
    molecularly characterized human tumor cell lines
    to identify predictive marker

64
(No Transcript)
65
Stratification DesignInteraction Design
66
Develop prospective analysis plan for evaluation
of treatment effect and how it relates to
biomarker
  • Defined analysis plan that protects type I error
    and permits adequately powered evaluation in test
    patients
  • http//brb.nci.nih.gov
  • Trial sized for defined analysis plan
  • Test negative patients should be adequately
    protected using interim futility analysis

67
Fallback Analysis Plan
  • Test average treatment effect at reduced level p0
  • If significant claim broad effectiveness
  • If overall effect is not significant, test
    treatment effect in marker subset at level
    .05-p0
  • If significant claim effectiveness for marker
    subset
  • Claim of significance for marker subset should
    not require either
  • Overall significance
  • Significant interaction

68
Sample size for Analysis Plan
  • To have 90 power for detecting uniform 33
    reduction in overall hazard at 1 two-sided
    level requires 370 events.
  • If 33 of patients are positive, then when there
    are 370 total events there will be approximately
    123 events in positive patients
  • 123 events provides 90 power for detecting a 45
    reduction in hazard at a 4 two-sided
    significance level.

69
(No Transcript)
70
(No Transcript)
71
(No Transcript)
72
Strong confidence in test Small r2 and large
r1 Weak confidence in test Small r2 and small
r1 p00 selected to control type I error rates
73
Bayesian Two-Stage DesignRCT With Single Binary
Marker
74
(No Transcript)
75
Adaptive Threshold Design
  • Randomized clinical trial of E vs C
  • Single candidate biomarker with K candidate
    cut-points
  • Entry not restricted by biomarker value
  • Adaptive in the sense that no pre-specified
    cut-point is provided. Eligibility is not changed
    during trial based on interim results

76
Final Analysis in Two Parts
  • Test global null hypothesis that treatment E is
    equivalent to C in efficacy for all biomarker
    values
  • If global null hypothesis is rejected, develop
    information about how effectiveness of E depends
    on biomarker value

77
(No Transcript)
78
Bootstrap Confidence Intervals for Threshold b
79
(No Transcript)
80
The confidence interval for the cut-point can be
used to inform treatment decisions for future
patients
81
(No Transcript)
82
(No Transcript)
83
Key Points
  • It can be beneficial not to define a cut-point
    for the biomarker prior to conducting the phase
    III clinical trial
  • The phase II database may be inadequate with
    regard to number of cases, lack of control group,
    different endpoint
  • The only thing that stands in the way of a more
    informative phase III trial is the aspirin
    paradigm that the ITT analysis of the eligible
    population is required to serve as a basis for
    approval

84
The Biology is Often Not So Clear
  • Cancer biology is complex and it is not always
    possible to have the right single predictive
    classifier identified with an appropriate
    cut-point by the time the phase 3 trial of a new
    drug is ready to start accrual

85
With a Small Number of Candidate
BiomarkersBiomarker Selection Design
  • Based on Adaptive Threshold Design
  • W Jiang, B Freidlin R Simon
  • JNCI 991036-43, 2007

85
86
Biomarker Selection Design
  • Have identified K candidate biomarkers B1 , , BK
    thought to be predictive of patients likely to
    benefit from T relative to C
  • Cut-points not necessarily established for each
    biomarker
  • Eligibility not restricted by candidate markers

87
Marker Selection Design
88
Designs When there are Many Candidate Markers and
Multi-marker Classifiers are of Interest
89
(No Transcript)
90
Adaptive Signature Design
91
  • The indication classifier is not a binary
    classifier of whether a patient has good
    prognosis or poor prognosis
  • It is a two sample classifier of whether the
    prognosis of a patient on E is better than the
    prognosis of the patient on C

92
  • The indication classifier maps the vector of
    candidate covariates into E,C indicating which
    treatment is predicted superior for that patient
  • The classifier need not use all the covariates
    but variable selection must be determined using
    only the training set
  • Variable selection may be based on selecting
    variables with apparent interactions with
    treatment, with cut-off for variable selection
    determined by cross-validation within training
    set for optimal classification
  • The indication classifier can be a probabilistic
    classifier

93
(No Transcript)
94
(No Transcript)
95
(No Transcript)
96
Treatment effect restricted to subset.10 of
patients sensitive, 400 patients.
Test Power
Overall .05 level test 46.7
Overall .04 level test 43.1
Sensitive subset .01 level test (performed only when overall .04 level test is negative) 42.2
Overall adaptive signature design 85.3
97
Overall treatment effect, no subset effect. 400
patients
Test Power
Overall .05 level test 74.2
Overall .04 level test 70.9
Sensitive subset .01 level test 1.0
Overall adaptive signature design 70.9
98
  • This approach can be used with any set of
    candidate predictors
  • The approach can also be used to identify the
    subset of patients who dont benefit from the new
    treatment when the overall ITT comparison is
    significant

99
Key Idea
  • Replace multiple significance testing by
    development of one indication classifier and
    obtain unbiased estimates of the properties of
    that classifier if used on future patients

100
(No Transcript)
101
(No Transcript)
102
  • At the conclusion of the trial randomly partition
    the patients into K approximately equally sized
    sets P1 , , PK
  • Let D-i denote the full dataset minus data for
    patients in Pi
  • Omit patients in P1
  • Apply the defined algorithm to analyze the data
    in D-1 to obtain a classifier M-1
  • Classify each patient j in P1 using model M-1
  • Record the treatment recommendation E or C

103
  • Repeat the above steps for all K loops of the
    cross-validation (develop classifier from scratch
    in each loop and classify omitted patients)
  • When cross-validation is completed, all patients
    have been classified once as what their optimal
    treatment is predicted to be

104
  • Let S denote the set of patients for whom
    treatment E is predicted optimal
  • Compare outcomes for patients in S who actually
    received E to those in S who actually received C
  • Compute Kaplan Meier curves of those receiving E
    and those receiving C
  • Let z standardized log-rank statistic

105
Test of Significance for Effectiveness of E vs C
  • Compute statistical significance of z by
    randomly permuting treatment labels and repeating
    the entire cross-validation procedure to obtain a
    new set S and a new logrank statistic z
  • Do this 1000 or more times to generate the
    permutation null distribution of treatment effect
    for the patients in S

106
  • The size of the E vs C treatment effect for the
    indicated population is (conservatively)
    estimated by the Kaplan Meier survival curves of
    E and of C in S

107
Cross-Validated Adaptive Signature Design
  • Define indication classifier development
    algorithm A
  • Apply algorithm to full dataset to develop
    indication classifier for use in future patients
    M(xA,P)
  • Using K fold cross validation
  • Classify patients in test sets based on
    classifiers developed in training sets e.g.
    yiM(xiA,P-i)
  • Si yi E
  • Compare E to C in S and estimate size of
    treatment effect
  • is an estimate of the size of the
    treatment effect
  • for future patients with M(xA,P)E

108
Cross-Validated Adaptive Signature Design
  • Approximate null distribution of
  • Permute treatment labels
  • Repeat complete cross-validation procedure
  • Generate permutation distribution of the
  • values for permuted data
  • Test null hypothesis that the treatment effect in
    classifier positive patients is null using as
    test statistic cross-validated estimate of
    treatment effect in positive patients

109
70 Response to E in Sensitive Patients25
Response to E Otherwise25 Response to C30
Patients Sensitive
ASD CV-ASD
Overall 0.05 Test 0.830 0.838
Overall 0.04 Test 0.794 0.808
Sensitive Subset 0.01 Test 0.306 0.723
Overall Power 0.825 0.918
110
25 Response to T 25 Response to CNo Subset
Effect
ASD CV-ASD
Overall 0.05 Test 0.047 0.056
Overall 0.04 Test 0.04 0.048
Sensitive Subset 0.01 Test 0.001 0
Overall Power 0.041 0.048
111
(No Transcript)
112
(No Transcript)
113
(No Transcript)
114
The Objectives of a Phase III Clinical Trial
  • Test the global null hypothesis that the new
    treatment E is uniformly ineffective relative to
    a control C for all patients while preserving the
    type I error of the study
  • If the global null hypothesis is rejected,
    develop an internally validated labeling
    indication for informing physicians in their
    decisions about which patients they treat with
    the drug.
  • Not a hypothesis testing problem

115
Prediction Based Clinical Trials
  • We can evaluate our methods for analysis of
    clinical trials in terms of their effect on
    patient outcome via informaing therapeutic
    decision making

116
  • Hence, alternative methods for analyzing RCTs
    can be evaluated in an unbiased manner with
    regard to their value to patients using the
    actual RCT data

117
(No Transcript)
118
Expected t Year DFS Using Indication Classifier
119
Expected t Year DFS With Conventional Analysis
120
Prediction Based Clinical Trials
  • The resampling approach provides an internally
    validated way of evaluating the effectiveness of
    indication classifiers for informing treatment
    selection to improve patient outcome

121
Prediction Based Clinical Trials
  • By switching from subset analysis to development
    of indication classifiers and by using
    re-sampling and careful prospective planning, we
    can more adequately evaluate new methods for
    analysis of clinical trials in terms of improving
    patient outcome by informing therapeutic decision
    making

122
  • By applying the classifier development algorithm
    to the full dataset D, an indication classifier
    is developed for informing how future patients
    should be treated
  • M(xA, D) for all x vectors.
  • The cross validation merely serves to
  • provide an estimate of the treatment effect for
    future patients with M(xA, D)E
  • and to provide a significance test of the null
    hypothesis that the treatment effect is zero

123
  • The stability of the indication classifier
    M(xA,D)can be evaluated by examining the
    consistency of classifications M(xiA, B) for
    bootstrap samples B from D.

124
  • Although there may be less certainty about
    exactly which types of patient benefit from E
    relative to C, classification may be better than
    for standard clinical trials in which all
    patients are classified based on results of
    testing the single overall null hypothesis

125
  • This approach can also be used to identify the
    subset of patients who dont benefit from a new
    regimen C in cases where E is superior to C
    overall at the first stage of analysis. The
    patients in SC D S are not predicted to
    benefit from E. Survivals of E vs C can be
    examined for patients in that subset and a
    permutation based confidence interval for the
    hazard ratio calculated.

126
(No Transcript)
127
506 prostate cancer patients were randomly
allocated to one of four arms Placebo and 0.2 mg
of diethylstilbestrol (DES) were combined as
control arm C 1.0 mg DES, or 5.0 mg DES were
combined as T. The end-point was overall
survival (death from any cause).
  • Covariates Age, performance status (pf), tumor
    size (sz), stage/grade index (sg), serum acid
    phosphatase (ap)

Cova
128
Figure 1 Overall analysis. The value of the
log-rank statistic is 2.9 and the corresponding
p-value is 0.09. The new treatment thus shows no
benefit overall at the 0.05 level.
129
Figure 2 Cross-validated survival curves for
patients predicted to benefit from the new
treatment. log-rank statistic 10.0, permutation
p-value is .002
130
Figure 3 Survival curves for cases predicted not
to benefit from the new treatment. The value of
the log-rank statistic is 0.54.
131
(No Transcript)
132
Marker Strategy Design
133
Marker Strategy Design
  • Generally very inefficient because some (many)
    patients in both randomization groups receive the
    same treatment
  • Often poorly informative
  • Not measuring marker in control group means that
    merits of complex marker treatment strategies
    cannot be dissected

134
Validation of Predictive BiomarkerStratification
Design
135
Prospective-Retrospective Study
136
In some cases a trial with optimal structure for
evaluating a new biomarker will have been
previously performed and will have pre-treatment
tumor specimens archived
  • Under certain conditions, a focused analysis
    based on specimens from the previously conducted
    clinical trial can provide highly reliable
    evidence for the medical utility of a prognostic
    or predictive biomaker
  • In some cases, it may be the only way of
    obtaining high level evidence

137
Prospective-Retrospective Design
138
Conclusions of Simon, Paik, Hayes
  • Claims of medical utility for prognostic and
    predictive biomarkers based on analysis of
    archived tissues can have either a high or low
    level of evidence depending on several key
    factors.
  • These factors include the analytical validation
    of the assay, the nature of the study from which
    the specimens were archived, the number and
    condition of the specimens, and the development
    prior to assaying tissue of a focused written
    plan for analysis of a completely specified
    biomarker classifier.
  • Studies using archived tissues from prospective
    clinical trials, when conducted under ideal
    conditions and independently confirmed can
    provide the highest level of evidence.
  • Traditional analyses of prognostic or predictive
    factors, using non analytically validated assays
    on a convenience sample of tissues and conducted
    in an exploratory and unfocused manner provide a
    very low level of evidence for clinical utility.

139
Guidelines Proposed by Simon, Paik,
HayesProspective-retrospective design
  • Adequate archived tissue from an appropriately
    designed phase III clinical trial must be
    available on a sufficiently large number of
    patients that the appropriate biomarker analyses
    have adequate statistical power and that the
    patients included in the evaluation are clearly
    representative of the patients in the trial.
  • The test should be analytically validated for use
    with archived tissue.
  • Testing should be performed blinded to the
    clinical data.
  • The analysis plan for the biomarker evaluation
    should be completely specified in writing prior
    to the performance of the biomarker assays on
    archived tissue and should be focused on
    evaluation of a single completely defined
    classifier.
  • The results should be validated using specimens
    from a similar, but separate study involving
    archived tissues.

140
Acknowledgements
  • Kevin Dobbin
  • Boris Freidlin
  • Wenyu Jiang
  • Aboubakar Maitournam
  • Shigeyuki Matsui
  • Michael Radmacher
  • Jyothi Subramanian
  • Yingdong Zhao
Write a Comment
User Comments (0)
About PowerShow.com