Quality by Design: A Perspective From the Office of Biotechnology Products - PowerPoint PPT Presentation

1 / 34
About This Presentation
Title:

Quality by Design: A Perspective From the Office of Biotechnology Products

Description:

ADVISORY COMMITTEE FOR PHARMACEUTICAL SCIENCE October 26, 2005 Barry Cherney, Ph.D. Deputy Director DTP/OBP/CDER Overview Introduction of Biotech Products ... – PowerPoint PPT presentation

Number of Views:120
Avg rating:3.0/5.0
Slides: 35
Provided by: fdaGovohr
Category:

less

Transcript and Presenter's Notes

Title: Quality by Design: A Perspective From the Office of Biotechnology Products


1
Quality by Design A Perspective From the Office
of Biotechnology Products
  • ADVISORY COMMITTEE FOR PHARMACEUTICAL SCIENCE
    October 26, 2005
  • Barry Cherney, Ph.D.
  • Deputy Director
  • DTP/OBP/CDER

2
Overview
  • Introduction of Biotech Products defining the
    issues
  • OBP Practice
  • Designing a Quality Product
  • Designing a Quality Process
  • Implementation

3
Office of Biotechnology Products
  • Therapeutic Proteins
  • Growth Factors
  • Enzymes
  • Cytokines
  • Chemokines
  • Angiogenic factors
  • Toxins
  • Soluble Receptors/Receptor antagonists
  • mAbs (related products)
  • These proteins are produced from recombinant or
    non recombinant cell culture expression systems
    and from transgenic and non transgenic systems
  • Products transferred from CBER to CDER in October
    2003
  • Excludes ONDCQA regulated protein products

4
Biotechnology Products
  • Biotechnology products tend to be
  • Large, complex molecules
  • Mixtures of many active ingredients
  • Subject to extensive heterogeneity in quality
    attributes of the API
  • Dependent on higher ordered structures and many
    times, flexibility (e.g. changes in conformation)
  • Are sensitive to small changes in manufacturing
    and impurity profiles, conformation stability
    limited

5
Product Variability
  • Carbamylation
  • Carboxylation
  • Formylation
  • Gamma Carboxyglutamic acid
  • O-linked Glycosylation
  • N-linked Glycosylation
  • Methylation
  • Oxidation
  • Phosphorylation
  • Sulphation
  • Amino Acid Substitution
  • Truncation
  • Mismatched S-S bonds
  • N- and C-terminal difference
  • Aggregation
  • Multimer Dissociation
  • Denaturation
  • Acetylation
  • Acylation
  • Addition of lipid
  • Amidation/Deamidation

6
Biotechnology Products
  • Generally, have poorly understood
    structure/function relationships
  • These properties of the API are hard to fully
    characterize resulting in uncertainty
  • Formulations majority liquid presentations,
  • less complexity then other formulations
    (stability a main issue, sampling size needs
    improvement )
  • Control of the API is a major source of concern
    for Biotech products.

7
Current OBP Practice
8
Paradigms
  • Quality is ensured by testing and rejecting lots
    that fail to meet its stated quality
    (insufficient)
  • A guiding principle for the Biotech industry has
    been that the process is the product (can be too
    restrictive)
  • Quality by design concept
  • Quality cannot be tested into a product it has
    to be built by design. This design incorporates
    knowledge of the product and the process to
    ensure all critical quality parameters are
    adequately controlled

9
Quality Control Strategy
  • Product Testing
  • Method Validation
  • Release Testing
  • Characterization
  • Stability Testing

10
How Much of the Iceberg (desired product) Can We
See?
  • Characterization

11
Comprehensive Quality Control Strategy
  • Process
  • Facilities and Equipment
  • Control of Raw Materials
  • In-Process Testing (PAT)
  • In-Process Controls
  • Process Validation (FED)
  • cGMPs (QC/QA)
  • Product
  • Method Validation
  • Release Testing
  • Characterization
  • Stability Testing

12
Designing a Quality Product
  • Design a high quality product that maximizes
    efficacy while minimizing adverse affects
  • Design a robust quality process to efficiently
    deliver a consistant product with the expected Q,
    S, and E profile

13
Q by D General Requirements for Biotech Products
  • Full Characterization of the products attributes
    (establish product variability the earlier the
    better)
  • Understanding the relationship between the
    products quality attributes and safety and
    efficacy
  • Understanding the mechanism of action both in
    terms of efficacy and safety (Biological
    characterization)
  • Understand how process affects critical quality
    attributes
  • This knowledge is limited for many Biotech
    products

14
The Desired Product
  • Dosage form is usually a given, liquid (some
    vialed as lyophilized power)
  • Excipients vary from product to product but
    mostly affect product stability
  • Desired attributes of the API (Focus for
    Biotech)
  • Opportunity for protein engineering -
    understanding protein structure/function
    relationship
  • Limit variability for attributes that negatively
    impact on product quality (via process or
    product)

15
Protein Engineering (rational design)
  • Increase manufacturability
  • Improving function/new properties
  • Increase specificity/affinity
  • Increasing Bioavailability
  • Pegylation
  • Glycoslation
  • Adding protein domains with increased half life
    (Fc)
  • Adding domains that bind to endogenous long lived
    proteins

16
Protein Engineering
  • Reduce tendency for aggregation
  • Increase conformational stability
  • Reducing immunogenicity
  • Eliminate sequences that promote aggregation
  • Humanizing foreign proteins (mAb)
  • Pegylation
  • Incorporate structures that are less immunogenic
    (disulfide bond scaffolds)
  • T cell epitope engineering

17
Protein Engineering
  • OBP has encouraged development of innovative
    products (not a regulatory requirement)
  • Less enthusiastic concerning the use of products
    whose design increases uncertainly and has no
    expected value clinically (premise limit product
    variability)
  • Histidine tag proteins (Quality versus
    Manufacturability)
  • Protein domains that potentially adversely impact
    safety

18
Designing A Quality Process
19
Examples of Problematic Process Designs
  • Manufacturing capacity to clear viruses is
    limited
  • Following elimination of aggregates by SEC, the
    manufacturer performs a heat treatment step for
    viral inactivation thus reintroduces aggregates
    back into the process
  • Process performed at room temperature with
    negative impact on quality
  • Roller bottle processes (open, multiple
    fermentations difficult to control)
  • Recloning is used to establish new cell banks
    introducing variability
  • Manufacturer recognized the limitations but
    regulatory hurdles are difficult to overcome
    particularly after approval

20
Process Control
  • Current OBP expectations are that critical
    sources of variation should be identified and
    controlled (raw materials/ unit operations)
  • Controlled through in-process testing (PAT or
    other tests), monitoring operating parameters and
    process validation
  • Based on long standing paradigm that process
    consistency product consistency

21
Biotechnology Process Control
Some steps controlled by volume or time few
measure product attributes directly
Turbidity Conductivity
Harvest
Chromatography Columns
D02 pH
Fermentor
280nm ABS Conductivity
22
The Essence of PAT
  • Process decisions (in real time) are based on
    assessments of critical material attributes
  • Forward-feed of incoming material
  • Feedback by in-process monitoring
  • Product quality is monitored and controlled
    during the manufacturing process
  • End points achievement of the desired material
    attribute
  • Currently, limited use of PAT in Biotech products
    but applicability is promising

23
Process Control of Unit Operations
  • Identify intended functions of unit operations
    and the critical product attributes potentially
    affected
  • Establish desired limits of attribute (typically
    established by estimates of process capability)
  • Identify critical variables for the process step
  • Establish the range of the variables that
    provides assurance that you can meet your quality
    expectations
  • First principles ??
  • Empirical approach using multi variant analysis
    FED, but can you extrapolate to larger scales?

24
Design Space (Fermentation)
Critical process parameters
Time
Media composition
Agitation
25
Expanding the Design Space
  • Characterize a quality attribute with regard to
    relevant, clinically important parameters, i.e.
    its affect on
  • Potency
  • Bioavailability
  • Biodistribution
  • Immunogenicity
  • This information can be used to set
    specifications to ensure product quality as it
    relates to S and E and expand the design space

26
Examples from Biotech
  • For a highly glycoslyated protein various
    isoforms were isolated and monitored for relevant
    bioactivity in a animal model suitable for Pk
    measurements. Outcome widen specs for isoform
    profile
  • Monitored product isoforms from human serum
    samples over time, showed rates of decay were
    similar concluded isoforms did not impact
    bioavailability Outcome broaden acceptance
    criteria
  • Use of multiple lots of drug product in clinical
    trials to establish a link between variability of
    product attributes and clinical performance

27
Biological Activity Matrix
Purified/induced variants
Developmental lots
Clinical lot extremes
Stressed lots
Clinical lots
Multiple binding/cellular assays
Small Animal/Complex Bioassay
Clinical/Clin Pharm
Validated bioassay
28
Implementation
29
Regulatory Relief (based on process understanding)
  • Validate the process is capable of impurity
    removal to appropriate levels (non toxic
    impurities)
  • Relief Impurity is not routinely measured when
    operating under the validated state (removed from
    specifications)
  • Different approaches depending on the nature of
    the impurity
  • Validate capacity to remove those impurities that
    are added at fixed concentrations (fixed input)
  • Validate excess capacity for removal of
    impurities that variable (alternatively control
    of input levels of impurities)
  • Examples Host Cell Proteins/DNA

30
Regulatory Relief (based on product understanding)
  • Understanding of the relationship between the
    quality attribute and its impact on safety and
    efficacy can reduce regulatory requirements
  • Relief If no likely impact on S and E dont
    include as a specification (no rejection limit)
  • use as a process consistency measure, where
    exceeding a limit initiates an investigation
  • if not a consistency measure, drop the test
    entirely
  • Transitioning to this new paradigm of action
    versus rejection limits
  • Need to discuss more extensively in-house and
    provide reviewer training

31
Implementation of Q by D
  • Q by D a major fear by industry is that
    reviewers will not understand or be receptive to
    the submission paraphrased from Dr. Ken Morris,
    Q by D presentation October 17, 2005
  • OBP review is based on scientific merits of the
    proposal and not simply reliance on existing
    practice. Guidance helps frame the issue but
    science and knowledge dictates the outcome.
  • For example, we try to stay away from
    proscriptive rules i.e. rejection limits can be
    established /- 3 SD. Instead, we evaluate the
    proposal using our best scientific judgment and
    are open to other statistical analysis but links
    between the attribute and what is known regarding
    its impact on S and E are important. Lack of
    knowledge increases uncertainty and may result in
    tighten controlled.

32
Implementation of Q by D
  • Structure of OBP
  • Product reviewers a mixture of
    research/reviewers and full time reviewers
  • Research conducted in molecular and cellular
    biology and pharmaceutical science
  • Expertise in biological characterization of
    protein products is critical for meaningful risk
    assessment
  • Provides hands on experience with latest
    techniques familiarity with fermentation/purificat
    ion processes
  • Expertise in biological characterization relevant
    to other CDER products
  • Consultations across CDER

33
PAT Future Directions
Many steps controlled by measuring product
attributes (or by monitoring all DP samples)
34
Continued and Future Directions
  • Training of OBP product reviewers in PAT (4 OBP
    product reviewers will undergo extensive training
    for Biotech products), Q by D, and new
    analytical techniques (Biosensors SPR) for
    biotech products
  • Q by D discussions within and outside Agency
  • Encourage biological characterization of products
  • Encourage industry to incorporate new or under
    utilized analytical methods for control of
    in-process materials and purified proteins
Write a Comment
User Comments (0)
About PowerShow.com