A Statistician - PowerPoint PPT Presentation

1 / 21
About This Presentation
Title:

A Statistician

Description:

A Statistician s Perspective on Biomarkers in Drug Development PSI Biomarkers Special Interest Group Martin Jenkins and Chris Harbron, AstraZeneca – PowerPoint PPT presentation

Number of Views:34
Avg rating:3.0/5.0
Slides: 22
Provided by: Ratnayake
Learn more at: https://www.psiweb.org
Category:

less

Transcript and Presenter's Notes

Title: A Statistician


1
A Statisticians Perspective on Biomarkers in
Drug Development
  • PSI Biomarkers Special Interest Group
  • Martin Jenkins and Chris Harbron, AstraZeneca
  • Co-authors Aiden Flynn, Trevor Smart, Chris
    Harbron, Tony Sabin, Jayantha Ratnayake, Paul
    Delmar, Athula Herath, Philip Jarvis and James
    Matcham
  • Volume 10, Issue 6, Pages 494-507,
    November/December 2011

2
What is a biomarker?
Biological marker (biomarker) A characteristic
that is objectively measured and evaluated as an
indicator of normal biological processes,
pathogenic processes, or pharmacologic responses
to a therapeutic intervention.
NIH biomarker definitions working group, 2001
  • Many endpoints could be considered as biomarkers
  • Commonly thought of in terms of biological /
    tissue samples (eg blood), imaging techniques or
    even examinations
  • Many technology platforms (proteomics,
    histopathology etc)
  • Does not define one purpose and so a clear
    objective and terminology can be helpful
  • Appear at many stages throughout the development
    program

3
Types of biomarker
4
Biomarker Examples
Biomarker Current Use Classification Qualification
HER2, EGFR, K-RAS mutations Directing treatment in oncology Predictive biomarker Defines indication in label, diagnostic development
UGT1A1, TMPT polymorphisms Predisposition to certain toxicities Predictive biomarker Appear in label as risk factor suggesting dose adjustment
AB1-42 Diagnose prodromal Alzheimers Disease Prognostic biomarker Enrich trial populations. Example of qualification
Gene signature chips Prognosis prediction in oncology Prognostic biomarker Diagnostic qualification process applies
CRP, IL-6, TNFa in blood samples Proof of principle in inflammatory diseases Pharmacodynamic biomarker Fit for purpose assay validation
FDG-PET imaging Proof of concept (eg Tumour metabolism) Pharmacodynamic biomarker Collaborative opportunities for qualification
HbA1c Represents glycemic control in diabetics Surrogate Endpoint Primary evidence for label. Qualification burden
5
What characteristics are we interested in?
  • It is generally advisable to learn as much as
    possible about a biomarker prior to its
    application for decision-making
  • The science behind the choice of biomarker should
    be supported prior to initiating studies
  • The practical feasibility of using the marker
    should be examined
  • The statistical properties of the biomarker are
    of interest
  • Statistical properties of interest include
  • Variability estimates (and components of
    variability, within and between subjects)
  • Effect sizes (using a positive control, challenge
    model or other data sources) and dynamic range
  • Distributions and scaling approaches required
  • Appropriate analysis methods considered

6
Methodology studies
  • Feasibility / Methodology studies can be used to
    learn the characteristics of an endpoint, but
    also the practicalities
  • Feasibility of multiple assessments
  • Acceptability to the patient, degree of dropout
  • The same design and analysis methodology should
    be used as is envisaged for the subsequent
    clinical trial
  • Information learned can aid in designing or
    sizing this trial
  • Work carried out should be fit-for-purpose enough
    to ensure that there is sufficient confidence
    that decisions can be made based upon the
    biomarker, given its intended use. Further
    studies, reviews or meta-analyses can aid in this
    if required.

7
Sources of variation
  • Much biomarker work is accompanied by similar
    practical challenges around controlling sources
    of variation
  • Patient-related factors
  • Individual biological factors genetics, race,
    age, comorbidities
  • Social and habitual factors smoking, physical
    fitness, diet
  • Natural biological variation eg diurnal effects
  • External factors
  • Pre-analytical variation sample collection
    and fixation practices
  • Analytical variation between laboratories,
    readers and batches,
    technical precision of
    the assay

8
Sources of bias
  • Bias can arise, especially in open-label studies,
    for several reasons
  • Verification bias because of the choice of
    locally available methods
  • Patient selection bias, for example, patients
    with breast cancer family history, may not
    consent to BRCA DNA testing
  • Treatment allocation bias, if treatment
    assignment is based on a subjective assessment,
    as in some histo-pathological endpoints
  • Being asked the same question repeatedly, for
    example in a pain challenge model, could induce
    false differences
  • If a scoring method is subjective, then measures
    should be taken to minimize bias, for example,
    with the use of scripted questions, blinding or
    standard operating procedures.
  • Conclusions may be subject to many caveats if
    they are not based upon complete and
    representative data-sets

9
Sources of missing data
  • Biomarker studies can suffer from missing data
    issues, especially in exploratory situations
  • Inaccessibility of tissues (e.g. lung cancer)
  • Low consent rates for optional samples
  • Lack of residual tissues in complete responders
  • Poor quality of fixation in archival samples
  • Patient drop-outs (lack of response or toxicity)
  • Poor data handling procedures cf. clinical data
  • Likely sources of missing data should be
    considered in advance so as to minimise potential
    implications and make appropriate assumptions

10
Multiplicity when considering many markers
  • Range of new high-dimensional technologies
  • e.g. genetics, genomics, proteomics,
    metabonomics, NGS
  • Allow understanding in detail at a molecular
    level the processes of disease and response to
    treatment and identify biomarkers that can
    identify or predict these changes.
  • Multiplicity becomes a concern and requires new
    approaches to be adopted
  • e.g. False Discovery Rate, Permutation testing,
    Significance Analysis of Microarrays (SAM)
  • Pre-analysis filtering of variables can help
  • Want to emerge with both statistical significance
    scientifically relevant
    and meaningful effect.
  • 2D FDR

11
Composites and Multivariate Analyses
  • Visualisation key
  • Principal Components Analysis (PCA), Clustering
  • Wide variety of supervised multivariate
    predictive modeling techniques are available
  • Regression-based approaches e.g. Partial least
    Squares (PLS), Elastic Nets
  • Proximity-based methods e.g. Nearest Neighbours
  • Tree-based methods e.g. Random Forests, Gradient
    Boosting
  • Distance-based approaches e.g. Support Vector
    Machines (SVM)
  • Not just Black-Box, Interpretation important
  • Importance scores
  • Study design is critical, as is visualization,
    understanding data quality and its impact on
    subsequent analyses.
  • MAQC-II

12
Biomarkers for Personalised Healthcare
  • Personalized health care offers the potential to
    identify patients more likely to derive benefit
    from treatment and as such is of great interest
    to Pharmaceutical companies, Regulatory
    Authorities and Health care Providers
  • Predictive markers marker by treatment
    interaction
  • Gives rise to new set of challenges eg. Low
    power to robustly identify biomarkers in
    exploratory studies
  • Developing from a Biomarker to a Companion
    Diagnostic
  • Understanding and quantifying the factors that
    may impact the performance of the diagnostic
  • Identifying an optimal cut-off
  • Additional regulatory process

13
Study design options
  • All-comers design
  • Subjects enrolled into groups, and
    retrospectively measured
  • Stratification design
  • Biomarker status at screening used as a
    stratification factor in randomisation to ensure
    balance
  • Targeted design
  • Only biomarker positive patients recruited into
    study
  • Enriched design
  • Hybrid, recruiting a limited number of biomarker
    negative patients with a greater representation
    of biomarker positives
  • Adaptive design
  • Many options allowing testing and refinement of a
    biomarker

14
Safety biomarkers preclinical translation
  • Predictive safety biomarkers allow early
    detection of toxicity and assessment of human
    risk
  • Preclinical qualification including mechanistic
    understanding of the relationship between the
    biomarker and organ damage
  • Use known organ toxicants
  • Determination of organ toxicity in rat by
    qualified toxicological pathologist
  • Understanding properties of translation to man
  • Animal Model Framework (AMF) project is a
    collaborative effort combining pre-clinical and
    clinical data to determine operating
    characteristics and refine thresholds of Safety
    Pharmacology models

15
Safety biomarkers - clinical
  • Clinical qualification less straightforward
    because generally verification of organ toxicity
    not possible
  • Utilise methods that can assess performance in
    absence of gold standard including Bayesian
    approaches
  • Currently lab parameters used to indicate kidney
    and liver injury, but poor performance
  • e.g. by time serum creatinine indicates drug
    induced kidney injury, a high degree of kidney
    function loss has already occurred
  • Public-private precompetitive partnerships
    established for searching for, validating and
    qualifying safety biomarkers for predicting
    drug-induced organ injury
  • Predictive Safety Testing Consortium (C-Path),
    SAFE-T (IMI)

16
Biomarker qualification
  • Formal biomarker qualification process have been
    introduced
  • FDA (qualification process for drug development
    tools)
  • EMA (qualification of novel methodologies and
    biomarkers)
  • Examples on EMA and FDA websites
  • These work alongside usual scientific advice
    routes and are a way to seek regulatory opinion
    on acceptability of a marker for a given use
  • This is not mandatory and so is usually not
    required, but may be an advantageous step,
    particularly for a marker with wide applicability
    (For example for collaborative groups seeking to
    develop a new endpoint, particularly for
    toxicity)
  • Diagnostic biomarkers would need to follow the
    in-vitro diagnostic regulatory processes

17
Surrogate endpoints
  • Those outcomes for which treatment effects
    correlate well with those for an accepted
    clinical outcome at an individual and group level
    could potentially substitute for a recognized
    clinical endpoint
  • Should lead to the same decision being made
    as if the
    clinical outcome had been used
  • Examples exist (eg in diabetes or HIV), but
    a large
    body of evidence is required and this
    is not required for
    many purposes
  • Surrogacy is often an unrealistic target.
  • Statistical methods have been developed to
    examine the association between endpoints, but
    these can also be applied to other situations,
    for example when considering assurance when
    endpoints differ between studies

18
Future challenges
Issues to address in the future
i How can the traditional clinical development program be reengineered, so allow for greater learning around a biomarker without delaying the drug program?
Ii What study designs can be used to translate markers of drug toxicity into man given that the pre-clinical studies used for validation are not feasible to conduct in humans?
iii Many technologies suffer from a batch effect. What pre-processing techniques can be developed to ensure we are measuring true biology rather than sample quality?
iv New techniques have moved the degree of dimensionality by several orders of magnitude. How, both practically and analytically, can we cope with this deluge of data?
v Effective ways, possibly Bayesian, could be developed to build existing pathway knowledge into data analyses.
vi When identifying biomarkers for PHC, interaction analyses typically have low power, especially with many potential markers. How can such markers be reliably identified?
vii As PHC becomes the processes for gaining approval of a drug, diagnostic and biomarker qualification may become more burdensome. How can this evidence be streamlined?
19
Conclusions Recommendations
Key considerations for statisticians in biomarker development
i Define clear objectives shaped by the potential future use of the marker This will clarify what can reasonably be claimed based upon the research Validation work and study designs should be fit-for-purpose relative to these aims
ii Prospectively plan studies even if they are exploratory - Defining what represents success in advance will aid objective decision-making
iii Consult expert colleagues to understand and shape the endpoints definitions Learn about distributions and pre-processing Influence scoring methods so as to get maximum information
iv Gather prior knowledge on sources of variability and causes of missing data Plan ahead to avoid sample and data management issues rather than being reactive Adjust missing data assumptions accordingly
v Learn about a new marker using methodology studies or translational science - Estimates of variability or effect size can be valuable if reflective of situation of use
20
Conclusions Recommendations
Key considerations for statisticians in biomarker development
vi Analyse a biomarker using the appropriate methodology for the given endpoint type - As with other endpoints there is no universal approach
vii Be alert when using large numbers of candidate biomarkers - Particular attention should be paid to issues of multiplicity and model validation
viii Maintain a sceptical and questioning point of view to manage expectations - Consider results in relation to prior knowledge and study aims, avoiding extrapolation
ix Consider how to add confidence to findings using other sources of information - Integrate biological information as well as numerical results
x Remember that there is nothing mysterious about biomarkers - A consideration of key statistical principles can be instructive in most situations
21
PSI Biomarker Special Interest Group
  • Please get in touch if you would like to get
    involved!
  • Join the mailing list or linked-in group in
  • Help on the SIG committee
  • Review papers, training, discussion groups
  • Attend one of our free meetings /or our
    conference sessions
  • Go to www.psiweb.org and click on committees and
    sigs in the menu to find our webpage with more
    information

Biomarker Validation - Case Studies and
ApproachesĀ  MedImmune, Cambridge, 10 October 2012
Write a Comment
User Comments (0)
About PowerShow.com