quality by design for biotechnology products—part 1 - process development forum

Upload: gyro9

Post on 14-Apr-2018

223 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/29/2019 Quality by design for biotechnology productspart 1 - Process Development Forum

    1/13

    6/4/12 11:Quality by design for biotechnology productspart 1 - Process Development Forum

    Page 1ttp://www.processdevelopmentforum.com/articles/quality-by-designtechnology-productspart-1/?goback=%2Egde_2586893_member_84694706

    ARTICLES/APP NOTES Share This PageShare This Page

    Eli Lilly and Company

    Quality by design for biotechnology productspart 1

    BioPharm International

    November 29, 2011

    A PhRMA Working Group's advice on applying QbD to biotech.

    By Taruna Arora, Roger Greene, Jennifer Mercer, Paul Tsang, Meg Casais, Stuart Feldman, JuttaLook, Tony Lubiniecki, Joseph Mezzatesta, Stefanie Pluschkell, Mark Rosolowsky, Anurag S.

    Rathore, PhD, Mark Schenerman, Tim Schofield, Samantha Sheridan, Paul Smock, Sally Anliker,

    Lois Atkins, Bernerd McGarvey, Bruce Meiklejohn, Jim Precup, John Towns

    ABSTRACT

    The International Conference on Harmonization (ICH) Q8(R2), Q9, and Q10 guidelines

    provide the foundation for implementing Quality by Design (QbD). Applying those concepts

    to the manufacture of biotech products, however, involves some nuances and complexities.

    Therefore, this paper offers guidance and interpretation for implementing QbD for

    biopharmaceuticals, from early-phase development steps such as identifying critical quality

    attributes and setting specifications, followed by the development of the design space andestablishing the process control strategy; to later stages, including incorporating QbD into a

    regulatory filing and facilitating efficient commercial processes and manufacturing change

    flexibility post licensure.

    Quality by Design (QbD) is a concept applied to the design and development

    of biopharmaceutical molecules and manufacturing that entails building

    quality into the process and product in a systematic, science- and risk-based

    manner. The QbD concepts outlined in the International Conference on

    Harmonization (ICH) Q8(R2), Q9, and Q10 guidelines provide the foundation

    for implementing QbD during specific stages of product development.13 A

    critical component of QbD is understanding the needs of the patient and thespecific quality attributes of the product that are linked to safety and efficacy.

    Thus, to implement QbD, it is critical to have a fundamental understanding of

    the functional relationships between patient needs, product quality attributes,

    analytical capabilities, and the manufacturing process.

    Pharmaceutical product sponsors begin by identifying product requirements to meet patient needs

    and then determine the quality target product profile (QTPP) and the critical quality attributes

    (CQAs) required to meet those patient needs. Based on this information, the sponsor then designs

    ART

    Plug

    Com

    built

    a R

    PRA

    Out Som

    visco

    strea

    MU

    POS

    UPC

    P

    May

    The

    mono

    nowa

    Rem

    http://www.processdevelopmentforum.com/multimedia/removing-the-buffer-bottleneck-through-inline-conditioninghttps://www.surveymonkey.com/s/FVFGQ39http://www.processdevelopmentforum.com/articles/quality-by-design-for-biotechnology-productspart-1/?goback=%2Egde_2586893_member_84694706#http://www.processdevelopmentforum.com/http://www.processdevelopmentforum.com/articles/http://www.processdevelopmentforum.com/articles/http://www.processdevelopmentforum.com/http://www.processdevelopmentforum.com/http://www.processdevelopmentforum.com/https://event.on24.com/eventRegistration/EventLobbyServlet?target=registration.jsp&eventid=445133&sessionid=1&key=D4FE84A34920D9D0955A6B8F49C38D1A&sourcepage=registerhttp://www.processdevelopmentforum.com/multimedia/removing-the-buffer-bottleneck-through-inline-conditioninghttp://www.processdevelopmentforum.com/tips/out-of-clutter-find-simplicity-clarification-using-permeate-flow-controlhttp://www.processdevelopmentforum.com/articles/plugged-into-single-usehttps://www.surveymonkey.com/s/FVFGQ39http://www.processdevelopmentforum.com/articles/quality-by-design-for-biotechnology-productspart-1/?goback=%2Egde_2586893_member_84694706#http://www.processdevelopmentforum.com/webcasts/http://www.processdevelopmentforum.com/multimedia/http://www.processdevelopmentforum.com/posters/http://www.processdevelopmentforum.com/tips/http://www.processdevelopmentforum.com/articles/http://www.processdevelopmentforum.com/http://www.processdevelopmentforum.com/
  • 7/29/2019 Quality by design for biotechnology productspart 1 - Process Development Forum

    2/13

    6/4/12 11:Quality by design for biotechnology productspart 1 - Process Development Forum

    Page 2ttp://www.processdevelopmentforum.com/articles/quality-by-designtechnology-productspart-1/?goback=%2Egde_2586893_member_84694706

    the molecule, the manufacturing process, and the control strategy to ensure that the desired

    product quality is met consistently.

    The QbD approach can be maintained throughout the lifecycle of the product to facilitate innovation

    and continuous improvement based on expanded knowledge and new technologies. This

    knowledge is gained during development and grows with more manufacturing experience through

    process characterization, scale-up, technology transfer, and manufacture, as well as through

    increased patient exposure to the product. This approach encourages incorporating prior process

    and product knowledge and experience-based methodologies (i.e., platform knowledge) into

    manufacturing throughout the life of the product.

    The QbD approach was introduced into the US Food and Drug Administration's chemistry,

    manufacturing, and controls (CMC) review process in 2004 with the aim of enhancing and

    modernizing the regulation of pharmaceutical manufacturing and product quality.4 The goal of QbD

    is to develop robust, well-understood processes that 1) deliver a product meeting the QTPP and 2)

    are controlled by defined steps within the manufacturing process and that allow for manufacturing

    process changes within an established design space of input variables and operating parameters

    without negatively affecting process attributes or identified CQAs. When a product is developed

    using a QbD approach, the impact of raw and starting materials and process parameters on product

    quality are well understood and the sources of process and product variability are well-known and

    controlled. In contrast, traditional pharmaceutical manufacturing relies heavily on end-product

    testing and the process typically lacks the flexibility needed to respond to variables encountered

    during manufacturing. The outcome is often "quality by conformance" and a rigid, static

    manufacturing operation that is more susceptible to multiple manufacturing changes post licensure,

    thus requiring continuous updating of the application through burdensome global regulatory

    supplements, variations, or amendments.

    This paper focuses on the factors to consider when applying the QbD concepts outlined in ICH

    Q8(R2), Q9, and Q10 to biotechnology products. The paper is intended to capture and reflect both

    the current and future state of QbD implementation. Therefore, new tools and terminology (e.g.,

    expanded change protocol and the post-approval management plan) have been included with anexpectation that the paper will be revised in the future based on new information and experiences,

    such as the mock biotech common technical document (CTD) sections being developed by the

    European Federation of Pharmaceutical Industries and Associations (EFPIA) and the CMC Biotech

    Working Group (formerly Conformia), and the US FDA's CMC Biotech Pilot Program.

    This position paper begins by addressing patient needs and product quality attributes, followed by a

    discussion of characterization development studies and the development of the design space and

    control strategies. It then addresses how to incorporate QbD into a regulatory filing and finally, how

    to facilitate efficient, cost-effective commercial processes and manufacturing change flexibility post

    licensure. Although biologic and biotechnology products often present a higher level of complexity

    than small molecules in terms of manufacturing process or product structure, the concepts of QbDare the same as those for small molecules. This paper describes the nuances and complexities

    involved in implementing QbD in the manufacture of biotech products and offers guidance and

    interpretation for doing so. The scope of this paper is limited to well-characterized protein products,

    in which the natural molecular heterogeneity, impurity profile, and potency can be defined with a

    high degree of confidence.

    ESTABLISHING CRITICAL QUALITY ATTRIBUTES, SPECIFICATIONS, DESIGN SPACE, AND

    CONTROL STRATEGY FOR A BIOTECH PRODUCT

  • 7/29/2019 Quality by design for biotechnology productspart 1 - Process Development Forum

    3/13

    6/4/12 11:Quality by design for biotechnology productspart 1 - Process Development Forum

    Page 3ttp://www.processdevelopmentforum.com/articles/quality-by-designtechnology-productspart-1/?goback=%2Egde_2586893_member_84694706

    Figure 1. Steps taken toward establishing critical quality attributes, specifications, design space, andcontrol strategy

    QbD is centered on the patient.Understanding the patient's needs for managing his or her disease

    state helps define the QTPP. Safety, efficacy, convenience, compliance, and cost effectiveness of

    the product must be considered. Next, CQAs of the product are identified, based on the

    understanding of the impact of various quality attributes on the safety and efficacy of the product.

    The initial assessment is based on molecular design and known attributes of the molecule. These

    include not only a comprehensive understanding of the molecular structure but also data collected

    from clinical and nonclinical studies that have been performed with the molecule and with other

    relevant molecules, as well as other applicable published knowledge. This process of assessing

    quality attributes and determining their criticality is ongoing, and is revisited continuously throughout

    the product lifecycle as more data become available. These data are also used to set appropriate

    specifications for the drug substance and the drug product, and to develop the overall control

    strategy.

    The CQAs are also used to design the molecule and the manufacturing process to meet patient

    needs and endpoints for safety and efficacy, as well as to control process and product quality.

    Further, the process is optimized and characterized, and the cumulative data set is used to define

    the critical process parameters, process design space, and the process control strategy, which

    together ensure that appropriate quality is maintained and variability is managed during the

    manufacturing, storage, and distribution of the product. This approach is schematically illustrated in

    Figure 1. A more detailed discussion of these steps is presented below.

  • 7/29/2019 Quality by design for biotechnology productspart 1 - Process Development Forum

    4/13

    6/4/12 11:Quality by design for biotechnology productspart 1 - Process Development Forum

    Page 4ttp://www.processdevelopmentforum.com/articles/quality-by-designtechnology-productspart-1/?goback=%2Egde_2586893_member_84694706

    Applying QbD Early in Development

    Products that are early in the development process are in a unique position to participate fully in

    prospective QbD during development. Consequently, companies may be able to develop an

    efficient, cost-effective commercial process for such products, and benefit from a more flexible

    reporting mechanism for changes to the application after approval. This situation is expected to

    provide an opportunity for manufacturing to incorporate new technology more quickly and easily

    and to make changes within the design space without regulatory agency review and approval.

    Products in early stages of development usually have a limited amount of manufacturing history

    and limited commercial-scale experience. As commercial-scale manufacturing data are gathered

    post-approval, new knowledge can be continuously integrated into the pre-approval understanding.

    The resulting comprehensive knowledge can then be used to support process changes and the

    assessment of their potential impact on critical quality attributes based on all available lifecycle

    data.

    Molecular Design and Engineering

    For biotechnology products, a key element in implementing QbD is engineering the molecule itself.

    A number of strategies are currently used by investigators to alter the properties of the molecule to

    achieve the desired balance among efficacy, stability, safety (such as immunogenicity), and

    manufacturability. These strategies include engineering the primary sequence to incorporate

    chemical and post-translational modifications for improved stability; recombinant and chemical

    fusion approaches for improved half-life and potency; and affinity maturation by phage display or

    transgenic mouse (e.g., xenomouse, Humax, and velocity mouse) technologies. Understanding the

    structure and functional attributes of therapeutic proteins, including monoclonal antibodies (MAbs),

    is key to developing the design space, because that understanding facilitates the selection of

    desirable quality attributes during molecular design while ensuring that bioactivity of the protein

    therapeutic is maintained.

    Technologies based on combinatorial libraries and transgenic mice can generate a diverse panel of

    antibodies and protein-based candidates selected for binding and specificity for a target. Further

    selection and screening may involve recombinant cloning from hybridomas, humanization, andfusions (e.g., Fc or human serum albumin, PEGylation) and the use of stringent parameters like in

    vitro and in vivo assessment of biological activity. Other key factors in the selection of an efficacious

    molecule include affinity and the rate of dissociation from the target. Protein engineering also can

    be applied to increase tissue penetration and distribution.

    Switching the isotype of an antibody product to gain desired effector functions is one approach that

    has been practiced. In some cases, an IgG1 isotype is ideal for cell killing, whereas IgG2 is a

    favored isotype for eliminating the effector function of an antibody on target binding. For further

    enhancement of cell killing by an antibody, technologies focused on antibody Fc modifications by

    mutations or glycoengineering as well as fusions to toxin or drug conjugates have emerged and

    been taken to clinical trials for some targets.

    Engineering the protein sequence is also used to reduce the risk of an immune response. Murine

    and chimeric antibodies used as human therapeutics frequently produce an immune response that

    can result in a reduction in or loss of activity and an unfavorable pharmokinetic (PK) profile

    requiring frequent administration of a drug. Humanization, transgenic mice, and phage display

    approaches make it possible to select antibody sequences bearing high homology to human

    germline sequences with the goal of reducing the risk of immunogenicity. Nevertheless, the risk of

    immune response to a fully human antibody and other self proteins necessitates screening

  • 7/29/2019 Quality by design for biotechnology productspart 1 - Process Development Forum

    5/13

    6/4/12 11:Quality by design for biotechnology productspart 1 - Process Development Forum

    Page 5ttp://www.processdevelopmentforum.com/articles/quality-by-designtechnology-productspart-1/?goback=%2Egde_2586893_member_84694706

    therapeutic protein candidates for immunogenicity using in silico methods, and in vitro assays

    measuring T-cell responses. Sequence engineering also can be used to eliminate T-cell epitopes in

    the product.

    Besides rational design for efficacy and safety, science-based hypothesis-driven approaches are

    used for selection and early engineering to achieve balance in the optimal physical and chemical

    properties of proteins with respect to product stability during and after manufacturing. A common

    observation during the development and commercialization of protein products is the susceptibility

    of certain amino acids (e.g., asparagine and methionine) to chemical modifications. Deamidation of

    asparagines and oxidation of methionines in regions important for activity can affect efficacy in

    some products. Sequence engineering also may be applied to minimize risks of chemical instability.

    In summary, protein design and engineering are powerful tools for enhancing efficacy, modulating

    immunogenicity, altering proteolytic stability, introducing chemical modification sites, and improving

    expression. Rational design approaches make it possible to identify and engineer better protein

    therapeutic candidates right from the start, which combined with process optimization can pave the

    way for less frequent dosing, increased efficacy, decreased manufacturing costs, and an improved

    safety profile.510

    Using Laboratory, Nonclinical, and Clinical Studies to Identify and Assess CQAsAn important step in the bioproduct development process is the identification of the CQAs of the

    product that drive the assessment of the ability of the bioprocess and bioproduct to deliver the

    intended performance of the product.11 The initial step toward identifying the CQAs is achieving a

    full understanding of the molecular structure and product variants. Next, all the molecular quality

    attributes and potential quality attributes are ranked using risk-based tools. The objective of this

    exercise is to determine which quality attributes may have the potential to affect the performance

    (safety and efficacy) of the product. A typical monoclonal antibody product may have dozens of

    such quality attributes and all are considered and assessed. They include but are not limited to

    product-related variants, process-related impurities, formulation parameters, and essential

    attributes such as appearance. Examples of process-related impurities include host cell DNA, host

    cell protein, adventitious agents, residual medium supplements such as protein hydrolysates, and

    components such as nucleases or residual Protein A from chromatography resins used in the

    purification process.

    There are multiple approaches to assessing the criticality of quality attributes. One such approach

    is to focus on the severity and uncertainty of impact (to safety and efficacy), with the goal of

    process and product design and the control strategy to minimize the probability of occurrence to

    reduce overall risk. The ability to detect a quality attribute during product development is required to

    assess its occurrence, thus necessitating the development of methods suitable for this detection

    and quantitation. The need to detect a specific attribute can decrease as its severity and

    occurrence are eliminated or minimized to an acceptable level with product knowledge and process

    control. Other approaches do not incorporate an assessment of occurrence, because CQAs are nottied to the ability of the process to control an attribute.

    Prior product knowledge plays a key role in the risk assessment and consists of the accumulated

    laboratory, nonclinical, and clinical experience for any specific product quality attribute. It also can

    include relevant data from similar molecules and data from literature references. With the

    increasing use of platforms in the development of biotech products and process, such as

    monoclonal antibody products, it is expected that investments in creating this knowledge can be

  • 7/29/2019 Quality by design for biotechnology productspart 1 - Process Development Forum

    6/13

    6/4/12 11:Quality by design for biotechnology productspart 1 - Process Development Forum

    Page 6ttp://www.processdevelopmentforum.com/articles/quality-by-designtechnology-productspart-1/?goback=%2Egde_2586893_member_84694706

    capitalized by applying it to several products once the platform has been established.1219 This

    combined knowledge provides a rationale for relating the attribute to product safety and efficacy. As

    mentioned above, the assessment of the severity of the impact (to safety or efficacy) results in a list

    of quality attributes in order of criticality. The list may evolve as more product and process

    knowledge are accumulated.

    Process-related components may be handled by considering their potential safety risk. One

    approach to assessing this safety risk is to evaluate an impurity safety factor (ISF) with an

    appropriate target or lower limit. The ISF is the ratio of the impurity LD50 to the maximum amount of

    an impurity potentially present in the product dose:

    ISF = LD50 Level in product dose

    in which LD50 is the dose of an impurity that results in lethality in 50% of the animals tested, and

    the level in product dose refers to the maximum amount of an impurity that could potentially be

    present and co-administered in a dose of product. The ISF is a normalized measure of the

    relationship between the level of an impurity resulting in a quantifiable toxic effect and the potential

    exposure of a patient to an impurity in the product. In the absence of an assay to detect an impurity,

    a conservative assumption is that all of the impurity in the process co-purifies with the product, andno clearance is achieved by the purification process. In cases where a sufficiently sensitive assay is

    available, the actual level of an impurity in the product is measured. Impurities can be eliminated

    from further consideration at any step where the safety risk is determined to be minimal, with an

    eye toward any impurity being removed or eliminated to as low a level as possible. An alternative

    strategy for process-related impurities, such as DNA or host cell proteins, is to demonstrate multi-

    log removal by validation that is conducted in a fashion similar to the validation of viral reduction or

    removal, thus obviating the need for a specification.

    The criticality of quality attributes may be explored in appropriately designed in vitro and in vivo

    studies. An example of the assessment of the affect of deamidation on the bioavailability, potency,

    and immunogenicity of a monoclonal antibody has been detailed elsewhere.15

    A combination oftools may be used to establish correlations between quality attributes and preclinical or clinical

    performance. Examples include:

    analytical tools such as native isoelectric focusing (IEF), ion-exchange chromatography

    (IEC), and online mass spectrometry to quantify the level of deamidation

    preclinical pharmacokinetics studies with the native and deamidated molecules

    in vivo potency studies with the native and deamidated molecules

    preclinical immunogenicity studies using extended dosing with the deamidated molecule

    determining the deamidation rate that would occurin vivo and assessing its relevance

    compared to the circulating half-life of the molecule.

    All or a combination of some of these studies may help qualify the criticality of an attribute such as

    deamidation.

    Setting Drug Substance and Drug Product Specifications

  • 7/29/2019 Quality by design for biotechnology productspart 1 - Process Development Forum

    7/13

    6/4/12 11:Quality by design for biotechnology productspart 1 - Process Development Forum

    Page 7ttp://www.processdevelopmentforum.com/articles/quality-by-designtechnology-productspart-1/?goback=%2Egde_2586893_member_84694706

    Figure 2. Illustration of an approach towards setting specifications for product quality attributes.

    Drug substance and drug product specifications are two components of the control strategy that

    enable delivery of the QTPP and represent the ranges within which the product is considered to

    meet the desired product quality. It should be pointed out that there is not a one-to-onecorrespondence between a list of CQAs and specifications. Although some CQAs will have

    specifications (i.e., acceptance criteria) associated with them, there may be others (such as host

    cell proteins or DNA) that will not have associated specifications if it has been demonstrated that

    the process has satisfactory clearance through multi-log removal validation carried out in small-or

    large-scale studies.

    Product specifications can be proposed using a combination of approaches, consistent with the

    guidance in ICH Q6B (Figure 2).20 Where appropriate, certain attribute specifications are based on

    existing regulatory guidance. For other attributes, relevant limits or ranges are established based on

    several sources of information that link the attributes to the safety and efficacy of the product,

    similar to the way this information is used to assess the criticality of these attributes. These sourcesinclude but are not limited to:

    clinical experience with the product

    nonclinical studies with the product or the isolated variant or impurity such as binding assays,

    in vivo assays, and cell-based assays

    clinical and nonclinical studies with similar platform products

    published literature on other similar products, variants, or impurities

    the process control strategy and the extent of variability observed in the manufactured lots.

    The difference between actual experience in the clinic and product specifications depends on our

    level of understanding of the impact that the attribute under consideration can have on the safety

    and efficacy of the product. This is illustrated in Figure 3 for two attributes, purity by IEC and host

    cell proteins. In the case of purity, the proposed specifications of >95% compared to the clinical

    experience of >98% would require either nonclinical studies relating the impurity or impurities to

    safety and efficacy, or clinical or nonclinical data on these species from another similar platform

    molecule or the literature.

  • 7/29/2019 Quality by design for biotechnology productspart 1 - Process Development Forum

    8/13

    6/4/12 11:Quality by design for biotechnology productspart 1 - Process Development Forum

    Page 8ttp://www.processdevelopmentforum.com/articles/quality-by-designtechnology-productspart-1/?goback=%2Egde_2586893_member_84694706

    Figure 3. Illustration of differences between clinical experience and filed specifications for a product-related impurity (% purity by ion exchange chromatography, or IEC) and a process-related impurity

    (host cell protein, or HCP) for a typical monoclonal antibody product

    The proposed specification for host cell proteins (

  • 7/29/2019 Quality by design for biotechnology productspart 1 - Process Development Forum

    9/13

    6/4/12 11:Quality by design for biotechnology productspart 1 - Process Development Forum

    Page 9ttp://www.processdevelopmentforum.com/articles/quality-by-designtechnology-productspart-1/?goback=%2Egde_2586893_member_84694706

    Figure 4. Derivative/one branch Ishikawa diagram to identify process parameters in the bioreactorexpansion unit operation that may potentially affect the quality attributes. (Figure courtesy of S. Rose,

    Eli Lilly and Company; unpublished).

    A thorough understanding of the factors that could affect the process is required. A risk assessment

    should be performed to identify the process parameters that should be characterized using small-

    scale models and tools such as design of experiments (DoE). One approach commonly used to

    facilitate a cause-and-effect analysis is the "fishbone" or derivative/one branch Ishikawa diagram

    (Figure 4). Such a diagram provides a graphical representation of the relationship between process

    operating parameters and unit operation outcomes. For the reactor expansion unit operation, for

    example, the cell growth, viable cell density, culture viability, and glucose concentration may be

    affected by the inoculum, medium, bioreactor operation, and harvest. Scientific understanding and

    prior knowledge can be used together with quality risk management to prioritize which process

    parameters to study. One tool that is useful for documenting factor risks is failure mode effectsanalysis (FMEA), which uses the severity, probability, and detectability of each factor to rank risks

    within a process step, and thereby select factors for subsequent study.2 The accumulated

    information from similar molecules or experiences with similar technologies may be used to identify

    variables and define an initial design space for the process step of a new molecular entity.

  • 7/29/2019 Quality by design for biotechnology productspart 1 - Process Development Forum

    10/13

    6/4/12 11:Quality by design for biotechnology productspart 1 - Process Development Forum

    Page 10ttp://www.processdevelopmentforum.com/articles/quality-by-desigtechnology-productspart-1/?goback=%2Egde_2586893_member_84694706

    Definitions

    Whereas operating ranges of a process in early development are primarily based on experience

    with equipment and platform technologies, design space limits will become available during late

    development and finally established during process characterization using small-scale models andtools like multivariable DOE. (For a description of process characterization, see PDA Technical

    Report No. 42). The refinement of the design space is based on the QTPP and the knowledge

    gained throughout development. As knowledge increases, some of the potentially critical and key

    process parameters will be identified as noncritical or non-key so that the number of critical and key

    process parameters may be reduced during late development.

    PARTS 2 AND 3 OF THIS ARTICLE

    Parts 2 and 3 of this article appeared in the December 2009 and January 2010 issues ofBioPharm

    International. Part 2 covers the use of DOE to define the design space, unique considerations for

    process development for biopharmaceuticals, the establishment of a control strategy, and the

    placement of QbD information in a regultory application. Part 3 discusses continuous verification

    and post-approval changes, including topics such as verification at large scale, refinement of the

    design space, process changes and comparability, comparability protocols and expanded change

    protocols, marketed products, and a CMC post-approval management plan. It also includes the

    overall conclusions.

    ABOUT THE AUTHORS

    Taruna Arora is a principal scientist, protein science, Roger Greene is the director of regulatory

    affairs, Jennifer Merceris the director of regulatory affairs, CMC, and Paul Tsang is the executive

  • 7/29/2019 Quality by design for biotechnology productspart 1 - Process Development Forum

    11/13

    6/4/12 11:Quality by design for biotechnology productspart 1 - Process Development Forum

    Page 11ttp://www.processdevelopmentforum.com/articles/quality-by-desigtechnology-productspart-1/?goback=%2Egde_2586893_member_84694706

    director of quality, all at Amgen Inc.; Meg Casais is the director of global regulatory affairs, CMC,

    and Stuart Feldman is the director/CMC, global regulatory affairs, both at Schering Plough; Jutta

    Look is the senior manager, CMC regulatory affairs at Novartis Pharma AG; Tony Lubiniecki is

    the vice president of biopharmaceutical development & marketed product support at Centocor R&D,

    Inc.; Joseph Mezzatesta is the assistant director of regulatory CMC, corporate regulatory affairs, at

    Sanofi-Aventis; Stefanie Pluschkell is an associate research fellow in global CMC, biologics and

    devices, at Pfizer Inc.; Mark Rosolowsky is the executive director of global regulatory sciences

    CMC at Bristol Myers Squibb; Anurag Rathore is a biotech CMC consultant and faculty member at

    the Indian Institute of Technology, Delhi, India; Mark Schenerman is the vice president of

    analytical biochemistry at MedImmune; Tim Schofield is the director of US regulatory affairs at

    GlaxoSmithKline; Samantha Sheridan is the director of regulatory affairs at Shire Pharmaceuticals;

    Paul Smock is a director and quality product leader in Wyeth Biotech, Wyeth Pharmaceuticals;

    Sally Anlikeris the director of regulatory affairs CMC, Lois Atkins is a principal consultant, CMC

    regulatory affairs, Bernerd McGarvey is an engineering advisor in the process engineering center,

    Bruce Meiklejohn is a principal fellow, regulatory COE-biotech, Jim Precup is a research scientist

    in manufacturing, science and technology, and John Towns is the senior director of global CMC

    regulatory affairs, all at Eli Lilly and Company. Towns is also the chair of the working group,

    317.276.4079, [email protected]

    REFERENCES

    1. International Conference on Harmonization. Q8(R2), Pharmaceutical development. Geneva,

    Switzerland. 2009 Aug.

    2. US Food and Drug Administration. Guidance for industry: Q9 Quality risk management.

    Bethesda, MD; 2006.

    3. US FDA. Guidance for Industry: Q10 Quality systems approach to pharmaceutical CGMP

    regulations. Bethesda, MD; 2006.

    4. US FDA. Guidance for industry: PATA framework for innovative pharmaceutical development,

    manufacturing, and quality assurance. 2004 Sept.

    5. Lonesberg N. Human antibodies from transgenic animals. Nature Biotech. 2005;23:1:11725.

    6. Perry LC, Jones TD, Baker MP. New approaches to prediction of immune responses to

    therapeutic proteins during preclinical development. Drugs R D. 2008;99(6):38596.

    7. Beckman RA, Weiner LM, Davis HM. Antibody constructs in cancer therapy; protein engineering

    strategies to improve exposure in solid tumors. Cancer. 2007;109(2):1709.

    8. Ravetch J. Antibodies therapeutics:isotype and glycoform selection. Expert opin biol ther. 2007;

    7(9):140113.

    9. Rehder DS, Chelius D, McAuley A, Dillon TM, Xiao G, Crouse-Zeineddini J, et al. Isomerization

    of a single aspartyl residue of anti-epidermal growth factor receptor immunoglobulin gamm2

    antibody highlights the role avidity plays in antibody activity. Biochemistry. 2008;47(8):2,51830.

    10. Honegger A. Engineering antibodies for stability and efficient folding. Handb Exp Pharmacol.

    2008;181:4768.

    11. Rathore AR, Winkle H. Quality by Design for pharmaceuticals: Regulatory perspective and

    approach. Nature Biotechnol. 2009. 27(1):2634.

    12. Barrett YC, Ebling W, Pieniaszek H, Billheimer J, Seiffert D. Validation and implementation of

    drug-dependent antibody assays in clinical trials for safety monitoring of patients dosed with

    roxifiban, an orally bioavailable glycoprotein IIb/IIIa antagonist. J Pharm Biomed Anal. 2007 Aug

    15;44(4):93846.

    13. Klakamp SL, Lu H, Tabrizi M, Funelas C, Roskos LK, Coleman D. Application of analytical

    detection concepts to immunogenicity testing. Anal Chem. 2007 Nov 1;79(21):8,17684.

    14. Mire-Sluis AR, Barrett YC, Devanarayan V, Koren E, Liu H, Maia M, et al. Recommendations

    mailto:[email protected]
  • 7/29/2019 Quality by design for biotechnology productspart 1 - Process Development Forum

    12/13

    6/4/12 11:Quality by design for biotechnology productspart 1 - Process Development Forum

    Page 12ttp://www.processdevelopmentforum.com/articles/quality-by-desigtechnology-productspart-1/?goback=%2Egde_2586893_member_84694706

    for the design and optimization of immunoassays used in the detection of host antibodies against

    biotechnology products. J Immunol Methods. 2004 Jun;289(12):116.

    15. Schenerman MA, Axley MJ, Oliver CN, Ram K, Wasserman GF. Using a risk assessment

    process to determine criticality of product quality attributes. In: Rathore AS, Mhatre R, editors.

    Quality by design for biopharmaceuticals: principles and case studies. Hoboken: Wiley; 2009.

    16. Gupta S, Indelicato SR, Jethwa V, Kawabata T, Kelley M, Mire-Sluis AR, et al.

    Recommendations for the design, optimization, and qualification of cell-based assays used for the

    detection of neutralizing antibody responses elicited to biological therapeutics. J Immunol Methods.

    2007 Apr 10;321(12):118.

    17. Rathore A, Branning R, Cecchini D. Design space for biotech products. BioPharm Int. 2007

    April;20(4):3640.

    18. Harms J, Wang X, Kim T, Yang J, Rathore AS. Defining design space for biotech products:

    Case study of Pichia pastoris fermentation. Biotechnol Prog. 2008:24(3):65562.

    19. Godavarti R, Petrone J, Robinson J, Wright R, Kelley BD. Scale-down models for purification

    processes: approaches and applications. In Rathore AS, Sofer G, editors. Process validation in

    manufacturing of biopharmaceuticals. New York: Taylor and Francis; 2005.

    20. US FDA. Guidance for industry: Q6B specifications: Test procedures and acceptance criteria for

    biotechnological/biological products. Rockville, MD; 1999 Aug.

    21. Submission of Quality Information for Biotechnology Products in the Office of Biotechnology

    Products; Notice of Pilot Program. Federal Register. July 2 2008;73(128):379724.

    22. Parenteral Drug Association. PDA Technical Report 42. Process Validation of Protein

    Manufacturing. Bethesda, MD: PDA; Sept 2005;59(S-4):128.

    Note: This article was originally published in November 2009 in BioPharm International Volume 22,

    Issue 11, and can be found online here. See part 2 here and part 3 here.

    More Articles/App Notes

    Articles / App NotesArticles / App Notes | TipsTips | MultimediaMultimedia | WebcastsWebcasts | RSSRSS | Terms of UseTerms of Use | Privacy PolicyPrivacy Policy | AbAb

    BioPharm International | Pharmaceutical Technology | Institute of Validation Technology Journals

    http://www.ivthome.com/http://pharmtech.findpharma.com/http://biopharminternational.findpharma.com/http://www.processdevelopmentforum.com/about/http://www.processdevelopmentforum.com/about/privacy/http://www.processdevelopmentforum.com/about/terms/http://www.processdevelopmentforum.com/rss/http://www.processdevelopmentforum.com/webcasts/http://www.processdevelopmentforum.com/multimedia/http://www.processdevelopmentforum.com/tips/http://www.processdevelopmentforum.com/articles/http://www.processdevelopmentforum.com/articleshttp://www.processdevelopmentforum.com/articles/quality-by-design-for-biotechnology-productspart-3/http://www.processdevelopmentforum.com/articles/quality-by-design-for-biotechnology-productspart-2/http://biopharminternational.findpharma.com/biopharm/article/articleDetail.jsp?id=640113&pageID=1&sk=&date=
  • 7/29/2019 Quality by design for biotechnology productspart 1 - Process Development Forum

    13/13

    6/4/12 11:Quality by design for biotechnology productspart 1 - Process Development Forum

    Page 13ttp://www.processdevelopmentforum.com/articles/quality-by-desigtechnology-productspart-1/?goback=%2Egde_2586893_member_84694706