throbber
The AAPS Journal, Vol. 16, No. 4, July 2014 ( # 2014)
`DOI: 10.1208/s12248-014-9598-3
`
`Review Article
`
`Understanding Pharmaceutical Quality by Design
`
`Lawrence X. Yu,1,6 Gregory Amidon,2 Mansoor A. Khan,1 Stephen W. Hoag,3 James Polli,3
`G. K. Raju,4,5 and Janet Woodcock1
`
`Received 17 November 2013; accepted 24 March 2014; published online 23 May 2014
`
`Abstract. This review further clarifies the concept of pharmaceutical quality by design (QbD) and describes
`its objectives. QbD elements include the following: (1) a quality target product profile (QTPP) that identifies
`the critical quality attributes (CQAs) of the drug product; (2) product design and understanding including
`identification of critical material attributes (CMAs); (3) process design and understanding including
`identification of critical process parameters (CPPs), linking CMAs and CPPs to CQAs; (4) a control strategy
`that includes specifications for the drug substance(s), excipient(s), and drug product as well as controls for
`each step of the manufacturing process; and (5) process capability and continual improvement. QbD tools and
`studies include prior knowledge, risk assessment, mechanistic models, design of experiments (DoE) and data
`analysis, and process analytical technology (PAT). As the pharmaceutical industry moves toward the
`implementation of pharmaceutical QbD, a common terminology, understanding of concepts and expectations
`are necessary. This understanding will facilitate better communication between those involved in risk-based
`drug development and drug application review.
`
`KEY WORDS: control strategy; critical quality attributes; pharmaceutical quality by design; process
`understanding; product understanding.
`
`INTRODUCTION
`
`Quality by design (QbD) is a concept first developed by the
`quality pioneer Dr. Joseph M. Juran (1). Dr. Juran believed that
`quality should be designed into a product, and that most quality
`crises and problems relate to the way in which a product was
`designed in the first place. Woodcock (2) defined a high-quality
`drug product as a product free of contamination and reliably
`delivering the therapeutic benefit promised in the label to the
`consumer. The US Food and Drug Administration (FDA)
`encourages risk-based approaches and the adoption of QbD
`principles in drug product development, manufacturing, and
`regulation. FDA’s emphasis on QbD began with the recognition
`that increased testing does not necessarily improve product
`quality. Quality must be built into the product.
`Over the years, pharmaceutical QbD has evolved with the
`issuance of ICH Q8 (R2) (Pharmaceutical Development), ICH
`Q9 (Quality Risk Management), and ICH Q10 (Pharmaceutical
`Quality System) (3–5). In addition, the ICH Q1WG on Q8, Q9,
`and Q10 Questions and Answers; the ICH Q8/Q9/Q10 Points to
`
`1 Center for Drug Evaluation and Research, Food and Drug
`Administration, Silver Spring, Maryland 20993, USA.
`2 University of Michigan, Ann Arbor, Michigan 48109, USA.
`3 University of Maryland, Baltimore, Maryland 21201, USA.
`4 Massachusetts Institute of Technology, Cambridge, Massachusetts
`02139, USA.
`5 Light Pharm Inc., Cambridge, Massachusetts 02142, USA.
`6 To whom correspondence should be addressed. (e-mail:
`lawrence.yu@fda.hhs.gov)
`
`Consider document; and ICH Q11 (Development and Manu-
`facture of Drug Substance) have been issued, as have the
`conclusions of FDA-EMA’s parallel assessment of Quality-By-
`Design elements of marketing applications (6–9). These docu-
`ments provide high level directions with respect to the scope and
`definition of QbD as it applies to the pharmaceutical industry.
`Nonetheless, many implementation details are not
`discussed in these guidances or documents. There is confusion
`among industry scientists, academicians, and regulators despite
`recent publications (10–13). This paper is intended to describe
`the objectives of pharmaceutical QbD, detail its concept and
`elements, and explain implementation tools and studies.
`
`PHARMACEUTICAL QUALITY BY DESIGN
`OBJECTIVES
`
`Pharmaceutical QbD is a systematic approach to devel-
`opment that begins with predefined objectives and empha-
`sizes product and process understanding and control based on
`sound science and quality risk management (3). The goals of
`pharmaceutical QbD may include the following:
`
`1. To achieve meaningful product quality specifications
`that are based on clinical performance
`2. To increase process capability and reduce product
`variability and defects by enhancing product and
`process design, understanding, and control
`3. To increase product development and manufacturing
`efficiencies
`4. To enhance root cause analysis and postapproval
`change management
`
`771
`
`1550-7416/14/0400-0771/0 # 2014 American Association of Pharmaceutical Scientists
`
`Eton Ex. 1037
`1 of 13
`
`

`

`772
`
`Under QbD, these goals can often be achieved by
`linking product quality to the desired clinical performance
`and then designing a robust formulation and manufactur-
`ing process to consistently deliver the desired product
`quality.
`Since the initiation of pharmaceutical QbD, the FDA
`has made significant progress
`in achieving the first
`objective: performance-based quality specifications. Some
`examples of FDA policies include tablet scoring and bead
`sizes in capsules labeled for sprinkle (14,15). The recent
`FDA discussions on the assayed potency limits for narrow
`therapeutic index drugs and physical attributes of generic
`drug products
`reflect
`this
`trend (16). Nonetheless,
`it
`should be recognized that ICH documents (3–9) did not
`explicitly acknowledge clinical performance-based specifi-
`cations as a QbD goal, although this was recognized in a
`recent scientific paper (10).
`The second objective of pharmaceutical QbD is to
`increase process capability and reduce product variability
`that often leads to product defects, rejections, and recalls.
`Achieving this objective requires robustly designed prod-
`uct and process. In addition, an improved product and
`process understanding can facilitate the identification and
`control of
`factors influencing the drug product quality.
`After
`regulatory approval, effort
`should continue to
`improve the process to reduce product variability, defects,
`rejections, and recalls.
`QbD uses a systematic approach to product design and
`development. As such,
`it enhances development capability,
`speed, and formulation design. Furthermore,
`it transfers
`resources
`from a downstream corrective mode to an
`upstream proactive mode. It enhances the manufacturer’s
`ability to identify the root causes of manufacturing
`failures. Hence,
`increasing product development and
`manufacturing efficiencies is the third objective of phar-
`maceutical QbD.
`The final objective of QbD is to enhance root cause
`analysis and postapproval change management. Without good
`product and process understanding, the ability to efficiently
`scale-up and conduct root cause analysis is limited and
`requires the generation of additional data sets on the
`proposed larger scale. FDA’s change guidances (17,18)
`provide a framework for postapproval changes. Recently,
`the FDA issued a guidance intended to reduce the regulatory
`filing requirements
`for
`specific low-risk chemistry,
`manufacturing, and control (CMC) postapproval manufactur-
`ing changes (19).
`
`ELEMENTS OF PHARMACEUTICAL QUALITY
`BY DESIGN
`
`In a pharmaceutical QbD approach to product develop-
`ment, an applicant identifies characteristics that are critical to
`quality from the patient’s perspective, translates them into the
`drug product critical quality attributes (CQAs), and estab-
`lishes the relationship between formulation/manufacturing
`variables and CQAs to consistently deliver a drug product
`with such CQAs to the patient. QbD consists of the following
`elements:
`
`Yu et al.
`
`1. A quality target product profile (QTPP) that identifies
`the critical quality attributes (CQAs) of the drug
`product
`2. Product design and understanding including the
`identification of critical material attributes (CMAs)
`3. Process design and understanding including the iden-
`tification of critical process parameters (CPPs) and a
`thorough understanding of scale-up principles, linking
`CMAs and CPPs to CQAs
`4. A control strategy that includes specifications for the
`drug substance(s), excipient(s), and drug product as
`well as controls for each step of the manufacturing
`process
`5. Process capability and continual improvement
`
`Quality Target Product Profile that Identifies the Critical
`Quality Attributes of the Drug Product
`
`QTPP is a prospective summary of the quality charac-
`teristics of a drug product that ideally will be achieved to
`ensure the desired quality, taking into account safety and
`efficacy of the drug product. QTPP forms the basis of design
`for the development of
`the product. Considerations for
`inclusion in the QTPP could include the following (3):
`
`& Intended use in a clinical setting, route of adminis-
`tration, dosage form, and delivery system(s)
`& Dosage strength(s)
`& Container closure system
`& Therapeutic moiety release or delivery and attributes
`affecting pharmacokinetic characteristics (e.g., disso-
`lution and aerodynamic performance) appropriate to
`the drug product dosage form being developed
`& Drug product quality criteria (e.g., sterility, purity,
`stability, and drug release) appropriate for the
`intended marketed product
`
`Identification of the CQAs of the drug product is the
`next step in drug product development. A CQA is a
`physical, chemical, biological, or microbiological property
`or characteristic of an output material
`including finished
`drug product that should be within an appropriate limit,
`range, or distribution to ensure the desired product
`quality (3). The quality attributes of a drug product may
`include identity, assay, content uniformity, degradation
`products, residual solvents, drug release or dissolution,
`moisture content, microbial limits, and physical attributes
`such as color, shape, size, odor, score configuration, and
`friability. These attributes can be critical or not critical.
`Criticality of an attribute is primarily based upon the
`severity of harm to the patient should the product fall
`outside the acceptable range for that attribute. Probability
`of occurrence, detectability, or controllability does not
`impact criticality of an attribute.
`It seems obvious that a new product should be ade-
`quately defined before any development work commences.
`However, over the years, the value of predefining the target
`characteristics of the drug product is often underestimated.
`Consequently, the lack of a well-defined QTPP has resulted in
`wasted time and valuable resources. A recent paper by Raw
`
`Eton Ex. 1037
`2 of 13
`
`

`

`Understanding Pharmaceutical Quality by Design
`
`773
`
`et al. (12) illustrates the significance of defining the correct
`QTPP before conducting any development. Also, QbD exam-
`ples exemplify the identification and use of QTPPs (20–22).
`
`Product Design and Understanding
`
`Over the years, QbD’s focus has been on the process
`design, understanding, and control, as discussed in the ICH
`Q8 (R2) guidance (3). It should be emphasized that product
`design, understanding, and control are equally important.
`Product design determines whether the product is able to
`meet patients’ needs, which is confirmed with clinical studies.
`Product design also determines whether the product is able to
`maintain its performance through its shelf life, which is
`confirmed with stability studies. This type of product under-
`standing could have prevented some historical stability
`failures.
`The key objective of product design and understanding is
`to develop a robust product that can deliver the desired
`QTPP over the product shelf life. Product design is open-
`ended and may allow for many design pathways. Key
`elements of product design and understanding include the
`following:
`
`& Physical, chemical, and biological characterization of
`the drug substance(s)
`& Identification and selection of excipient type and
`grade, and knowledge of intrinsic excipient variability
`& Interactions of drug and excipients
`& Optimization of formulation and identification of
`CMAs of both excipients and drug substance
`
`To design and develop a robust drug product that has the
`intended CQAs, a product development scientist must give
`serious consideration to the physical, chemical, and biological
`properties of the drug substance. Physical properties include
`physical description (particle size distribution and particle
`morphology), polymorphism and form transformation, aqueous
`solubility as a function of pH,
`intrinsic dissolution rate,
`hygroscopicity, and melting point(s). Pharmaceutical solid
`polymorphism, for example, has received much attention
`recently since it can impact solubility, dissolution, stability, and
`manufacturability. Chemical properties include pKa, chemical
`stability in solid state and in solution, as well as photolytic and
`oxidative stability. Biological properties include partition coef-
`ficient, membrane permeability, and bioavailability.
`Pharmaceutical excipients are components of a drug
`product other than the active pharmaceutical
`ingredient.
`Excipients can (1) aid in the processing of the dosage
`form during its manufacture; (2) protect,
`support, or
`enhance stability, bioavailability, or patient acceptability;
`(3) assist
`in product
`identification; or (4) enhance any
`other attribute of
`the overall safety, effectiveness, or
`delivery of
`the drug during storage or use (23). They
`are classified by the functions they perform in a pharma-
`ceutical dosage form. Among 42 functional excipient
`categories listed in USP/NF (24), commonly used excipi-
`ents include binders, disintegrants, fillers (diluents), lubri-
`cants, glidants (flow enhancers), compression aids, colors,
`sweeteners, preservatives,
`suspending/dispersing agents,
`pH modifiers/buffers, tonicity agents, film formers/coatings,
`flavors, and printing inks. The FDA’s inactive ingredients
`
`database (25) lists the safety limits of excipients based on
`prior use in FDA-approved drug products.
`It is well recognized that excipients can be a major
`source of variability. Despite the fact that excipients can alter
`the stability, manufacturability, and bioavailability of drug
`products, the general principles of excipient selection are not
`well-defined, and excipients are often selected ad hoc without
`systematic drug-excipient compatibility testing. To avoid
`costly material wastage and time delays, ICH Q8 (R2)
`recommends drug-excipient compatibility studies to facilitate
`the early prediction of compatibility (3). Systematic drug-
`excipient compatibility studies offer several advantages as
`follows: minimizing unexpected stability failures which usual-
`ly lead to increased development time and cost, maximizing
`the stability of a formulation and hence the shelf life of the
`drug product, and enhancing the understanding of drug-
`excipient interactions that can help with root cause analysis
`should stability problems occur.
`Formulation optimization studies are essential in developing a
`robust formulation that is not on the edge of failure. Without
`optimization studies, a formulation is more likely to be high risk
`because it is unknown whether any changes in the formulation itself
`or in the raw material properties would significantly impact the
`quality and performance of the drug product, as shown in recent
`examples (26,27). Formulation optimization studies provide impor-
`tant information on the following:
`
`& Robustness of the formulation including establishing
`functional relationships between CQAs and CMAs
`& Identification of CMAs of drug substance, excipients,
`and in-process materials
`& Development of control strategies for drug substance
`and excipients
`
`In a QbD approach, it is not the number of optimization
`studies conducted but rather the relevance of the studies and
`the utility of the knowledge gained for designing a quality
`drug product that is paramount. As such, the QbD does not
`equal design of experiments (DoE), but the latter could be an
`important component of QbD.
`Drug substance, excipients, and in-process materials may
`have many CMAs. A CMA is a physical, chemical, biological,
`or microbiological property or characteristic of an input
`material that should be within an appropriate limit, range,
`or distribution to ensure the desired quality of that drug
`substance, excipient, or in-process material. For the purpose
`of this paper, CMAs are considered different from CQAs in
`that CQAs are for output materials including product
`intermediates and finished drug product while CMAs are for
`input materials including drug substance and excipients. The
`CQA of an intermediate may become a CMA of that same
`intermediate for a downstream manufacturing step.
`Since there are many attributes of the drug substance
`and excipients that could potentially impact the CQAs of the
`intermediates and finished drug product, it is unrealistic that a
`formulation scientist investigate all the identified material
`attributes during the formulation optimization studies. There-
`fore, a risk assessment would be valuable in prioritizing which
`material attributes warrant further study. The assessment
`should leverage common scientific knowledge and the
`formulator’s expertise. A material attribute is critical when a
`realistic change in that material attribute can have a
`
`Eton Ex. 1037
`3 of 13
`
`

`

`774
`
`Yu et al.
`
`Table I. Typical Input Material Attributes, Process Parameters, and Quality Attributes of Pharmaceutical Unit Operations
`
`Pharmaceutical unit operation
`
`Input material attributes
`
`(cid:129) Particle size
`(cid:129) Particle size distribution
`(cid:129) Fines/oversize
`(cid:129) Particle shape
`(cid:129) Bulk/tapped/true density
`(cid:129) Cohesive/adhesive properties
`(cid:129) Electrostatic properties
`(cid:129) Moisture content
`
`(cid:129) Particle/granule size
`(cid:129) Particle/granule size
`distribution
`(cid:129) Fines
`(cid:129) Particle/granule shape
`(cid:129) Bulk/tapped/true density
`(cid:129) Adhesive properties
`(cid:129) Electrostatic properties
`(cid:129) Hardness/plasticity
`(cid:129) Viscoelasticity
`(cid:129) Brittleness
`(cid:129) Elasticity
`(cid:129) Solid form/polymorph
`(cid:129) Moisture content
`(cid:129) Granule porosity/density
`
`(cid:129) Particle size distribution
`(cid:129) Fines/Oversize
`(cid:129) Particle shape
`(cid:129) Bulk/tapped/true density
`(cid:129) Cohesive/adhesive properties
`(cid:129) Electrostatic properties
`(cid:129) Hardness/plasticity
`(cid:129) Viscoelasticity
`(cid:129) Brittleness
`(cid:129) Elasticity
`(cid:129) Solid form/polymorph
`(cid:129) Moisture content
`
`Process parameters
`Blending/mixing
`(cid:129) Type and geometry of mixer
`(cid:129) Mixer load level
`(cid:129) Order of addition
`(cid:129) Number of revolutions (time and speed)
`(cid:129) Agitating bar (on/off pattern)
`(cid:129) Discharge method
`(cid:129) Holding time
`(cid:129) Environment temperature and RH
`
`Size reduction/comminution
`
`Ribbon milling
`(cid:129) Ribbon dimensions
`(cid:129) Ribbon density
`(cid:129) Ribbon porosity/solid fraction
`
`Impact/cutting/screening mills
`(cid:129) Mill type
`(cid:129) Speed
`(cid:129) Blade configuration, type, orientation
`(cid:129) Screen size and type
`(cid:129) Feeding rate
`
`Fluid energy mill
`(cid:129) Number of grinding nozzles
`(cid:129) Feed rate
`(cid:129) Nozzle pressure
`(cid:129) Classifier
`
`Granule/ribbon milling
`(cid:129) Mill type
`(cid:129) Speed
`(cid:129) Blade configuration, type, orientation
`(cid:129) Screen size and type
`(cid:129) Feeding rate
`
`location, power
`
`Wet granulation
`High/low shear granulation
`(cid:129) Type of granulator (High/low shear, top/bottom drive)
`(cid:129) Fill level
`(cid:129) Pregranulation mix time
`(cid:129) Granulating liquid or solvent quantity
`(cid:129) Impeller speed, tip speed, configuration,
`consumption/torque
`(cid:129) Chopper speed, configuration, location, power consumption
`(cid:129) Spray nozzle type and location
`(cid:129) Method of binder excipient addition (dry/wet)
`(cid:129) Method of granulating liquid addition (spray or pump)
`(cid:129) granulating liquid temperature
`(cid:129) granulating liquid addition rate and time
`(cid:129) Wet massing time (post-granulation mix time)
`(cid:129) Bowl temperature(jacket temperature)
`(cid:129) Product temperature
`(cid:129) Post mixing time
`(cid:129) Pump Type: Peristaltic, Gear type
`(cid:129) Granulating liquid vessel (e.g., pressurized, heated)
`
`Fluid bed granulation
`(cid:129) Type of fluid bed
`(cid:129) Inlet air distribution plate
`(cid:129) Spray nozzle (tip size, type/quantity/ pattern/configuration/position)
`(cid:129) Filter type and orifice size
`
`Quality attributes
`
`(cid:129) Blend uniformity
`(cid:129) Potency
`(cid:129) Particle size
`(cid:129) Particle size distribution
`(cid:129) Bulk/tapped/true density
`(cid:129) Moisture content
`(cid:129) Flow properties
`(cid:129) Cohesive/adhesive properties
`(cid:129) Powder segregation
`(cid:129) Electrostatic properties
`
`(cid:129) Particle/granule size
`(cid:129) Particle/granule size distribution
`(cid:129) Particle/granule shape
`(cid:129) Particle/granule shape factor
`(e.g., aspect ratio)
`(cid:129) Particle/granule density/Porosity
`(cid:129) Bulk/tapped/true density
`(cid:129) Flow properties
`(cid:129) API polymorphic form
`(cid:129) API crystalline morphology
`(cid:129) Cohesive/adhesive properties
`(cid:129) Electrostatic properties
`(cid:129) Hardness/Plasticity
`(cid:129) Viscoelasticity
`(cid:129) Brittleness
`(cid:129) Elasticity
`
`torque,
`
`(cid:129) Endpoint measurement
`(e.g., power consumption,
`etc.)
`(cid:129) Blend uniformity
`(cid:129) Potency
`(cid:129) Flow
`(cid:129) Moisture content
`(cid:129) Particle size and distribution
`(cid:129) Granule size and distribution
`(cid:129) Granule strength and uniformity
`(cid:129) Bulk/tapped/true density
`(cid:129) API polymorphic form
`(cid:129) Cohesive/adhesive properties
`(cid:129) Electrostatic properties
`(cid:129) Granule brittleness
`(cid:129) Granule elasticity
`(cid:129) Solid form/polymorph
`
`Eton Ex. 1037
`4 of 13
`
`

`

`Understanding Pharmaceutical Quality by Design
`
`775
`
`Pharmaceutical unit operation
`
`Input material attributes
`
`Process parameters
`
`Quality attributes
`
`Table I. (continued)
`
`(cid:129) Fill level
`(cid:129) Bottom screen size and type
`(cid:129) Preheating temperature/time
`(cid:129) Method of binder excipient addition (dry/wet)
`(cid:129) Granulating liquid temperature
`(cid:129) Granulating liquid quantity
`(cid:129) Granulating liquid concentration/viscosity
`(cid:129) Granulating liquid holding time
`(cid:129) Granulating liquid delivery method
`(cid:129) Granulating liquid spray rate
`(cid:129)
`Inlet air, volume, temperature, dew point
`(cid:129) Atomization air pressure
`(cid:129) Product and filter pressure differentials
`(cid:129) Product temperature
`(cid:129) Exhaust air temperature, flow
`(cid:129) Filter shaking interval and duration
`
`Drying
`
`Fluidized bed
`(cid:129)
`Inlet air volume, temperature, dew point
`(cid:129) Product temperature
`(cid:129) Exhaust air temperature, flow
`(cid:129) Filter type and orifice size
`(cid:129) Shaking interval and duration
`(cid:129) Total drying time
`
`Tray
`(cid:129) Type of tray dryer
`(cid:129) Bed thickness/tray depth (depth of product per tray)
`(cid:129) Type of drying tray liner (e.g., paper, plastic,
`synthetic fiber, etc.)
`(cid:129) Quantity carts and trays per chamber
`(cid:129) Quantity of product per tray
`(cid:129) Drying time and temperature
`(cid:129) Air flow
`(cid:129)
`Inlet dew point
`
`Vacuum/microwave
`(cid:129)
`Jacket temperature
`(cid:129) Condenser temperature
`(cid:129)
`Impeller speed
`(cid:129) Bleed air volume
`(cid:129) Vacuum pressure
`(cid:129) Microwave power
`(cid:129) Electric field
`(cid:129) Energy supplied
`(cid:129) Product temperature
`(cid:129) Bowl and lid temperature
`(cid:129) Total drying time
`Roller compaction/chilsonation
`(cid:129) Type of roller compactor
`(cid:129) Auger (feed screw) type/design (horizontal,
`vertical or angular)
`(cid:129) Deaeration (e.g., vacuum)
`(cid:129) Auger (feed screw) speed
`(cid:129) Roll shape (cylindrical or interlocking).
`(cid:129) Roll surface design (smooth, knurled, serrated,
`or pocketed)
`(cid:129) Roll gap width (e.g., flexible or fixed)
`(cid:129) Roll speed
`(cid:129) Roll pressure
`
`(cid:129) Particle size, distribution
`(cid:129) Fines/oversize
`(cid:129) Particle shape
`(cid:129) Cohesive/adhesive properties
`(cid:129) Electrostatic properties
`(cid:129) Hardness/plasticity
`(cid:129) Viscoelasticity
`(cid:129) Brittleness
`(cid:129) Elasticity
`(cid:129) Solid form/polymorph
`(cid:129) Moisture content
`
`(cid:129) Particle size, distribution
`(cid:129) Fines/oversize
`(cid:129) Particle shape
`(cid:129) Cohesive/adhesiveproperties
`(cid:129) Electrostatic properties
`(cid:129) Hardness/plasticity
`(cid:129) Bulk/tapped/true density
`(cid:129) Viscoelasticity
`(cid:129) Brittleness
`(cid:129) Elasticity
`
`(cid:129) Granule size and distribution
`(cid:129) Granule strength, uniformity
`(cid:129) Flow
`(cid:129) Bulk/tapped/true density
`(cid:129) Moisture content
`(cid:129) Residual solvents
`(cid:129) API polymorphic form or transition
`(cid:129) Purity profile
`(cid:129) Moisture profile (e.g. product
`temperature vs. LOD)
`(cid:129) Potency
`(cid:129) Cohesive/adhesive properties
`(cid:129) Electrostatic properties
`
`(cid:129) Ribbon appearance (edge attrition,
`splitting, lamination, color, etc.)
`(cid:129) Ribbon thickness
`(cid:129) Ribbon density (e.g., envelop
`density)
`(cid:129) Ribbon porosity/solid fraction
`(cid:129) Ribbon tensile strength/breaking
`force
`(cid:129) Throughput rate
`(cid:129) API polymorphic form and transition
`
`Eton Ex. 1037
`5 of 13
`
`

`

`776
`
`Pharmaceutical unit operation
`
`Table I. (continued)
`
`Yu et al.
`
`Input material attributes
`
`Process parameters
`
`Quality attributes
`
`(cid:129) Solid form/polymorph
`
`(cid:129) Roller temperature
`(cid:129) Fines recycled (yes or no, # of cycles)
`
`(cid:129) Particle size, distribution
`(cid:129) Fines/oversize
`(cid:129) Particle shape
`(cid:129) Cohesive/adhesiveproperties
`(cid:129) Electrostatic properties
`(cid:129) Hardness/plasticity
`(cid:129) Bulk/tapped/true density
`(cid:129) Viscoelasticity
`(cid:129) Brittleness
`(cid:129) Elasticity
`(cid:129) Solid form/polymorph
`
`(cid:129) Particle size, distribution
`(cid:129) Fines/oversize
`(cid:129) Particle shape
`(cid:129) Melting point
`(cid:129) Density
`(cid:129) Solid form/polymorph
`(cid:129) Moisture content
`
`(cid:129) Particle/granule size
`and distribution
`(cid:129) Fines/oversize
`(cid:129) Particle/granule shape
`(cid:129) Cohesive/adhesive
`properties
`(cid:129) Electrostatic properties
`(cid:129) Hardness/plasticity
`(cid:129) Bulk/tapped/true density
`(cid:129) Viscoelasticity
`(cid:129) Brittleness
`(cid:129) Elasticity
`(cid:129) Solid form/polymorph
`(cid:129) Moisture
`
`(cid:129) Particle/granule size and
`distribution
`(cid:129) Fines/oversize
`(cid:129) Particle/granule shape
`(cid:129) Cohesive/adhesive properties
`(cid:129) Electrostatic properties
`(cid:129) Hardness/plasticity
`(cid:129) Bulk/tapped/true density
`(cid:129) Viscoelasticity
`(cid:129) Brittleness
`
`Extrusion–Spheronization
`(cid:129) Type of extruder (screw or basket)
`(cid:129) Screw length, pitch, and diameter
`(cid:129) Screw channel depth
`(cid:129) Screw blade configuration
`(cid:129) Number of screws (single/dual)
`(cid:129) Die or screen configuration (e.g., radial or axial)
`(cid:129) Die length/diameter ratio
`(cid:129) Roll diameter (mm)
`(cid:129) Screen opening diameter (mm)
`(cid:129) Screw speed (rpm)
`(cid:129) Feeding rate (g/min)
`(cid:129) Type and scale of spheronizer
`(cid:129) Spheronizer load level
`(cid:129) Plate geometry and speed
`(cid:129) Plate groove design (spacing and pattern)
`(cid:129) Air flow
`(cid:129) Residence time
`
`Hot melt extrusion
`(cid:129) Screw design (twin/single)
`(cid:129) Screw speed
`(cid:129) Screw opening diameter (mm)
`(cid:129) Solid and liquid feed rates
`(cid:129) Feeder type/design
`(cid:129) Feed rate
`(cid:129) No. of zones
`(cid:129) Zone temperatures
`(cid:129) Chilling rate
`
`(cid:129) Extrudate
`(cid:129) Density
`(cid:129) Length/thickness/diameter
`(cid:129) Moisture content
`(cid:129) API polymorphic form and transition
`(cid:129) Content uniformity
`(cid:129) Throughput
`
`(cid:129) Pellets after spheronization
`(cid:129) Pellets size and distribution
`(cid:129) Pellets shape factor (e.g. aspect
`ratio)
`(cid:129) Bulk/Tapped density
`(cid:129) Flow properties
`(cid:129) Brittleness
`(cid:129) Elasticity
`(cid:129) Mechanical strength
`(cid:129) Friability
`
`(cid:129) Extrudate density
`(cid:129) Length/thickness/diameter
`(cid:129) Polymorphic form and transition
`(cid:129) Content uniformity
`(cid:129) Throughput
`
`Tabletting
`(cid:129) Type of press (model, geometry, number of stations)
`(cid:129) Hopper design, height, angle, vibration
`(cid:129) Feeder mechanism (gravity/forced feed, shape of wheels,
`direction of rotation, number of bars)
`(cid:129) Feed frame type and speed
`(cid:129) Feeder fill depth
`(cid:129) Tooling design (e.g., dimension, score configuration,
`quality of the metal)
`(cid:129) Maximum punch load
`(cid:129) Press speed/dwell time
`(cid:129) Precompression force
`(cid:129) Main compression force
`(cid:129) Punch penetration depth
`(cid:129) Ejection force
`(cid:129) Dwell Time
`
`(cid:129) Tablet appearance
`(cid:129) Tablet weight
`(cid:129) Weight uniformity
`(cid:129) Content uniformity
`(cid:129) Hardness/tablet breaking force/
`tensile strength
`(cid:129) Thickness/dimensions
`(cid:129) Tablet porosity/density/solid fraction
`(cid:129) Friability
`(cid:129) Tablet defects
`(cid:129) Moisture content
`(cid:129) Disintegration
`(cid:129) Dissolution
`
`Encapsulation
`
`(cid:129) Machine type
`(cid:129) Machine fill speed
`(cid:129) Tamping Force
`(cid:129) No. of tamps
`(cid:129) Auger screw design/speed
`(cid:129) Powder bed height
`
`(cid:129) Capsule appearance
`(cid:129) Weight
`(cid:129) Weight uniformity
`(cid:129) Content uniformity
`(cid:129) Moisture content
`(cid:129) Slug tensile strength
`(cid:129) Disintegration
`(cid:129) Dissolution
`
`Eton Ex. 1037
`6 of 13
`
`

`

`Understanding Pharmaceutical Quality by Design
`
`777
`
`Pharmaceutical unit operation
`
`Input material attributes
`
`Process parameters
`
`Quality attributes
`
`Table I. (continued)
`
`(cid:129) Elasticity
`(cid:129) Solid form/polymorph
`(cid:129) Moisture
`
`(cid:129) Tablet dimensions
`(cid:129) Tablet defects
`(cid:129) Hardness/plasticity
`(cid:129) Density
`(cid:129) Porosity
`(cid:129) Moisture content
`
`(cid:129) Tablet dimensions
`(cid:129) Tablet defects
`(cid:129) Hardness/plasticity
`(cid:129) Density/porosity
`moisture content
`
`Pan coating
`(cid:129) Type of pan coater (conventional or side-vented)
`(cid:129) Pan (fully perforated or partial perforated)
`(cid:129) Baffle (design, number, location)
`(cid:129) Pan load level
`(cid:129) Pan rotation speed
`(cid:129) Spray nozzle (type, quantity, pattern, configuration,
`spray pattern)
`(cid:129) Nozzle to bed distance
`(cid:129) Distance between nozzles
`(cid:129) Nozzle orientation
`(cid:129) Total preheating time
`(cid:129) Inlet air flow rate, volume, temperature, dew point
`(cid:129) Product temperature
`(cid:129) Individual nozzle spray rate
`(cid:129) Total spray rate
`(cid:129) Atomization air pressure
`(cid:129) Pattern air pressure
`(cid:129) Exhaust air temperature, air flow
`(cid:129) Total coating, curing time and drying time
`
`Fluid bed coating
`(cid:129) Type of fluid bed coater
`(cid:129) Fluid bed load level
`(cid:129) Partition column diameter
`(cid:129) Partition column height
`(cid:129) Number of partition columns
`(cid:129) Air distribution plate type and size
`(cid:129) Filter type and orifice size
`(cid:129) Filter differential pressure
`(cid:129) Filter shaking interval and duration
`(cid:129) Spray nozzle (type, quantity, pattern, configuration)
`(cid:129) Nozzle port size
`(cid:129) Total preheating time
`(cid:129) Spray rate per nozzle
`(cid:129) Total spray rate
`(cid:129) Atomization air pressure
`(cid:129) Inlet air flow rate, volume, temperature, dew point
`(cid:129) Product temperature
`(cid:129) Exhaust air temperature, air flow
`(cid:129) Total coating, curing and drying time
`
`(cid:129) Size/dimensions
`(cid:129) Polymer type
`membrane thickness
`
`Laser drilling
`
`(cid:129) Conveyor type
`(cid:129) Conveyor speed
`(cid:129) Laser power
`(cid:129) Number of pulses
`(cid:129) Type(s) of lens(es)
`(cid:129) One or two sided
`(cid:129) Number of holes
`
`(cid:129) Coating efficiency
`(cid:129) Core tablet weight before and after
`preheating
`(cid:129) Moisture (gain/loss) during
`preheating
`(cid:129) Environmental equivalency factor
`(cid:129) Coated drug product (e.g., tablet or
`capsule) appearance
`(cid:129) % weight gain
`(cid:129) Film thickness
`(cid:129) Coating (polymer and /or color)
`uniformity
`(cid:129) Hardness/breaking force/Tensile
`strength
`(cid:129) Friability
`(cid:129) Moisture (gain/loss) during overall
`process
`(cid:129) Residual solvent(s)
`(cid:129) Disintegration
`(cid:129) Dissolution
`(cid:129) Tablet defects
`(cid:129) Visual attributes
`
`(cid:129) Coating efficiency
`(cid:129) Core tablet weight before and after
`preheating
`(cid:129) Moisture (gain/loss) during
`preh

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket