PowerPoint - Q-CROC

advertisement

How to Design High Impact Trials to Indentify Biomarkers

Janet Dancey, MD

Ontario Institute for Cancer Research

NCIC Clinical Trials Groups

2 nd Quebec Conference on Therapeutic Resistance

Montreal, November 5-6 th 2010

Potential Conflict of Interest

• Dr. Janet E. Dancey

– None

Objectives

• Types of biomarker studies

• Uses in clinical trials

• Methods and design issues

3

Why do biomarker studies?

• Biology

◦ Understand cancer

◦ Identify new targets for therapeutics

◦ Identify new markers of diagnosis, prognosis, prediction, monitoring

• Diagnosis

◦ To identify site of origin of an undifferentiated tumour,

◦ To identify second primary from metastases

• Prognosticate

◦ To predict outcome (risk of toxicity, relapse, progression)

• Predication

◦ To predict benefit/risk (or lack) from a specific treatment

• Monitor

◦ Identify cancer early, monitor response/progression

4

Why do biomarker studies?

• To understand cancer biology

• To improve treatments

• To change medical practice

5

Why is doing biomarker studies so difficult?

• Cancer models are not patients and people are not laboratory models

• ….and its not just biology

Things you don’t hear in the lab

By the way….

• My family has been in-bred for generations

• Those cancer cells in my flank had been in culture for decades

No visit, treatment, biopsy, imaging today, please….

• I’m not well

• the insurance won’t cover it

• the REB says you can’t

6

Sometimes it’s where the needle went

Intratumoral heterogeneity of carbonic anhydrase IX (CAIX)

Effect of distributional heterogeneity on the analysis of tumor hypoxia based on carbonic anhydrase IX

VV Iakovlev, M Pintilie, A Morrison, et al

Laboratory Investigation (2007) 87, 1206 –1217

a) Immunoperoxidase staining for CAIX in a single tissue section. Analysis of the entire section gave a value of 10.8% CAIX labeling.

The circles limit the analysis to 0.6 mm simulated tissue microarray (TMA) cores, and show a wide range in CAIX (for publication purpose only, the image was digitally enhanced to better visualize CAIX areas).

7

Sometimes it’s the timing pSer473-Akt antibody in human GI tumors and HT-29 colon cancer xenografts measured by immunohistochemical staining. surgically specimens biopsy specimens

Baker, A. F. et al. Clin Cancer Res 2005;11:4338-4340

A, patient tumor samples. 1 and 2 are two surgically resected specimens and 3 and 4 are two biopsy specimens.

B, HT-29 human tumor xenografts excised from scid mice and kept at room temperature for the times shown. Each section also includes in the upper right-hand quadrant an on-slide control of HT-29 colon cancer cells stained for pSer473-Akt.

8

Copyright ©2005 American Association for Cancer Research

Sometimes it’s how we measure it

9

Why are successful biomarker studies uncommon?

• Biological heterogeneity

◦ Cellular, tumour, patient

• Assay variability

◦ Within assay, between assays

• Specimen variability

• Effect size

A lot of “noise” that blur marker and outcome correlation and validation

10

‘Validation’

Feinstein

“Validation is one of those words — like health, normal, probability, and disease — that is constantly used and seldom defined.

We can ... simply say that, in data analysis, validation consists of efforts made to confirm the accuracy, precision, or effectiveness of the results.”

Feinstein, A. R. Multivariable Analysis: An Introduction (Yale University

Press, New Haven, 1996).

11

Biomarker Validation

• Biomarker – marker of biology;

◦ Scientific validation

• Assay – method/means of measurement;

◦ Technology/analytical validation

Laboratory

• Test - clinical context

◦ Clinical validation/qualification

• Clinical utility

◦ Value of using the test versus alternatives

Clinical Uptake

Multistep, multi-year, interative process requiring multidisciplinary collaboration 12

Trial Designs and Biomarkers

Trial Phase

0

I Metastatic

Purpose

Define dose

Select agents

Dose/schedule

II Metastatic Activity

III Metastatic Clinical benefit

III Adjuvant Clinical benefit

Biomarkers Modifications

Target modulation

PK

Target Modulation

PK

Toxicity

Activity

Normal Volunteers

Pre-surgical

Expanded cohorts to evaluate target , toxicity or screen activity

Predictive markers Randomized

Predictive markers Subset analyses

Predictive

Prognostic

Subset analyses

Type of marker depends on trial phase

13

Phase 1 Trials: Considerations

• Primary goal: To identify an appropriate dose/schedule for further evaluation

Small

• Design principles: patient numbers

◦ Maximize safety

◦ Minimize patients treated at biologically inactive doses

◦ Optimize efficiency

• Study population:

◦ Patients for whom no standard therapy

Heterogenous

Refractory

Tumours

Expect target modulation but not anti-tumour activity

14

1.0

Where/when do biomarkers play a role?

Target Versus Toxic Effects

Off Target Toxicity Target Effect in Tumour

Target Toxicity

Target Toxicity

Dose/Concentration/Exposure

15

Biomarker Studies in Phase 1 Trials

• MGMT activity after O 6 -benzylguanine

 Friedman H et al J Clin Oncol 16:3570-5, 1998; Spiro et al. Cancer Res

59:2402-10 1999; Dolan et al Clin Cancer Res 8:2519-23, 2002

• 20S proteosome inhibition after bortezomib

 Lightcap E et al. Clin Chem 46:673-683, 2000; Adams J, Oncologist 1:

9-16, 2002;

• DCE-MRI after PTK787/valatanib

 Galbraith S et al NMR in Biomed 15:132-142, 2002; Morgan, B. et al. J

Clin Oncol; 21:3955-3964 2003;

• S6K inhibition after everolimus

 Tanaka C et al J Clin Oncol 26:1596-1602, 2008

• PARP Inhibition after ABT-888

 Kinders RJ, et al. Clin Cancer Res. 2008 Nov 1;14(21):6877-

85

• ERK Inhibition after PLX-4032

 Puzanov, K. L. J Clin Oncol 27:15s, 2009 (suppl; abstr 9021)

16

PLX4032, a V600EBRAF kinase inhibitor: correlation of activity with PK and PD in a phase I trial.

Puzanov, K. L. J Clin Oncol 27:15s, 2009 (suppl; abstr 9021)

Patients

4

2 pERK

PRE range

50-100, median

60;

70 pERK range

10-40, median

11

5-fold

35-fold

2

KI67

PRE range

20-60%, median

45%;

KI67 range

5-25%, median

12.5%

4-fold

30 -50% 3-5%

10-fold

PK

µM*h mean

AUC0-

24h ~

126

µM*h

500 -

1000

Imaging

PD (4)

PR (1)

PET (2)

Target Pathway Tumor

17

Target

PARP

Hedgehog

SMO

EML4-ALK

BRAFV600E

Phase I Predictive Markers

Drug Test Phase I ORR (%)

Olaparib (AZD2281; KU-

0059436)

BRCA1/2 9/21 (44%) Ovary, breast, prostate

GDC-0449

PF-02341066

Mutation

(PTCH/SMO)

Translocation

18/33 (56%) Basal

Cell

20/31 (61%) Lung

PLX4032 (RG7204) Mutation 19/27 (70%)

Melanoma

Fong et al NEJM, 2009; von Hoff et al NEJM 2009; Kwak et al ECCO/ESMO

2009: Chapman et al ECCO/ESMO 2009;

18

Biomarkers in Phase 2/3

• Focus on developing predictive markers

• Difficult to demonstrate that the absence of predictive markers contributed to “failure” of drug

◦ Known prior to phase III

 HER2,

◦ Positive phase III subsequently analyzed for subset and marker was helpful

 Cetuximab, panitumumab and KRAS

 Erlotinib/gefitinib and EGFR FISH and mutations

Or not

 EGFR IHC in colon or lung carcinoma

◦ Negative phase III not further evaluated or under evaluation

19

Phase 3 (or 2) Trial: Effects of Biomarker Assay

Initial

Selection

Histology

Stage

Target

Tested

Strata Randomize Outcome

Agent

Marker +

Marker -

Control

Agent

Phase 3:

Survival

(Phase 2:

ORR, TTE)

Control

• Trial is designed to assess treatment effects in Marker+ and Markergroups

• Marker assessment

◦ Assay failure increases number of patients screened

◦ False positives will dilute effect

◦ False negatives will increase the number of patients screened

20

Phase 3 (or 2) Trial –Effect of Assay False Positive and

Negatives

Marker+ Treatment+

MarkerTreatment -

Ideal Marker x Treatment

Interaction

Marker x Treatment

Interaction with False Assay Results

M+/T+

M+/T+

M-/T+

M+/T-

M-/T-

M-/T+

Time

21

Suppose we have a new targeted therapy designed to be effective in patients with Marker A.

What types of clinical trials should we design?

22

Biomarker Clinical Trial Designs

• Target Selection or Enrichment Designs

• Unselected or All-comers designs

◦ Marker by treatment interaction designs (biomarker stratified design)

◦ Adaptive analysis designs

◦ Sequential testing strategy designs

◦ Biomarker-strategy designs

• Hybrid designs

Target Selection/Enrichment Designs

If we are sure that the therapy will not work in Markernegative patients

AND

We have an assay that can reliably assess the Marker

THEN

We might design and conduct clinical trials for Markerpositive patients or in subsets of patients with high likelihood of being Marker-positive

East Asian

Never smoker/light former smoker

Pulmonary

Adenocarcinoma

No prior treatment

IPASS-Schema

R

A

N

D

O

M

I

Z

E

Mok et al N Engl J Med 2009;361:947-57

Gefitinib

250 mg daily

Paclitaxel 200 mg/m 2

Carboplatin AUC 5-6

1 ° Endpoint PFS

2 ° EGFR Biomarker

25

IPASS-Gefitinib or Carboplatin–Paclitaxel in Pulmonary

Adenocarcinoma.

Mok et al N Engl J Med 2009;361:947-57

26

Unselected “All Comers” Trial Designs

If we are not sure that the Marker will define groups of patients that will benefit/not benefit from treatment

OR

There isn’t a validated assay that can reliably assess the status of the Marker

THEN

We might design and conduct clinical trials in unselected patients and try to identify predictive markers and robust assays.

Retrospective and Prospective Analysis Designs

Retrospective Analyses Designs

• Hypothesis generation studies

◦ Retrospective analyses based on convenience samples

• Prospective/retrospective designs

Prospective Designs

• Marker by treatment interaction designs (biomarker stratified design)

• Adaptive analysis designs

• Biomarker-strategy designs

• Sequential testing strategy designs

• Hybrid designs

Prospective/Retrospective Design

• Well-conducted randomized controlled trial

• Prospectively stated hypothesis, analysis techniques, and patient population

• Predefined and standardized assay and scoring system

• Upfront sample size and power calculation

• Samples collected during trial and available on a large majority of patients to avoid selection bias

• Biomarker status is evaluated after the analysis of clinical outcomes

• Results are confirmed by independent RCT(s)

Prospective

Retrospective

29

Suppose we want to find out if using a biomarker to select treatment is better?

30

Marker-based Strategy Design

If we think that one therapy will work in Marker-negative and another therapy will work in the Marker-positive patients

AND

We have a validated assay that can reliably assess the Marker status

THEN

We might design and conduct clinical trial to test whether using the biomarker to select treatment for patients is better than not using the marker to select treatment

Marker-based Strategy Design

Marker-Guided Randomized Design

Randomize To Use Of Marker Versus No Marker Evaluation

Control patients may receive standard or be randomized

Marker Determined

Treatment

M+

New Drug

M − Control

All Patients

New Drug

Randomize Treatment

OR

Standard Treatment

Control

Control

• Provides measure of patient willingness to follow marker-assigned therapy

• Marker guided treatment may be attractive to patients or clinicians

• Inefficient compared to completely randomized or randomized block design

Example: ERCC1: Customizing Cisplatin Based on

Quantitative Excision Repair Cross-Complementing 1 mRNA Expression

Cobo M et al. J Clin Oncol; 25:2747-2754 2007

• 444 chemotherapy-naïve patients with stage IIIB/IV NSCLC enrolled,

• 78 (17.6%) went off study before receiving chemotherapy, due insufficient tumor for

ERCC1 mRNA assessment.

• 346 patients assessable for response: Objective response was 39.3% in the control arm and 50.7% in the genotypic arm (P = .02).

33

Predictive Markers Trials: Considerations

• Is the drug/treatment active?

• Do we have a marker/markers?

• What are the treatment effects within patient subsets?

◦ Are there enough patients to assess treatment effects in

Marker+ and/or Marker- groups?

• Does the trial design distinguish predictive and prognostic effects?

• Is there a reliable assay to assess the biomarker?

• Samples requirements

34

Biomarker Translational Gaps

Laboratory

Single Centre

Trial

Multi-Centre

Trial

Clinical

Practice

Rapidity of

Science

Technology

Operations

Regulation

Standardization

Impact

• Rapid generation of new science in laboratory

• High content single institution trials can address biological questions

• Impact requires translation to multi-centre trials and ultimately clinical practice

35

Unprecedented Opportunity

• Rapid advances in understanding of cancer biology

• Rapid advances in technology

• An increasing arsenal of active agents available commercially or under clinical development

• Many opportunities for biomarker evaluation

36

8 Considerations for Biomarkers in Clinical Trials

• What is the question?

• Biomarker(s) – What we want to measure

• Assay – How we measure it

• Specimen – What we measure it in

• Study/Trial Design – Why, when, how we study it

• Study Execution – Can we get the study done

• Study Outcome – What it tells us

• Likely Impact – Whether we use it

37

Download