throbber
Journal of Pharmaceutical Sciences 107 (2018) 2653-2662
`
`Contents lists available at ScienceDirect
`
`Journal of Pharmaceutical Sciences
`
`j o u r n a l h o m e p a g e : w w w . j p h a r m s c i . o r g
`
`Pharmaceutics, Drug Delivery and Pharmaceutical Technology
`A Tutorial for Developing a Topical Cream Formulation Based on
`the Quality by Design Approach
`Ana Sim~oes 1, 2, Francisco Veiga 1, 2, Carla Vitorino 1, 2, 3, Ana Figueiras 1, 2, *
`1 Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
`2 LAQV. REQUIMTE, Group of Pharmaceutical Technology, Coimbra, Portugal
`3 Center for Neurosciences and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
`
`a r t i c l e i n f o
`
`a b s t r a c t
`
`Article history:
`Received 9 April 2018
`Revised 7 June 2018
`Accepted 12 June 2018
`Available online 20 June 2018
`
`Keywords:
`quality by design
`quality target product profile
`critical quality attributes
`critical material attributes
`critical process parameters
`
`The pharmaceutical industry has entered in a new era, as there is a growing interest in increasing the
`quality standards of dosage forms, through the implementation of more structured development and
`manufacturing approaches. For many decades, the manufacturing of drug products was controlled by a
`regulatory framework to guarantee the quality of the final product through a fixed process and
`exhaustive testing. Limitations related to the Quality by Test system have been widely acknowledged. The
`emergence of Quality by Design (QbD) as a systematic and risk-based approach introduced a new quality
`concept based on a good understanding of how raw materials and process parameters influence the final
`quality profile. Although the QbD system has been recognized as a revolutionary approach to product
`development and manufacturing, its full implementation in the pharmaceutical field is still limited. This
`is particularly evident in the case of semisolid complex formulation development. The present review
`aims at establishing a practical QbD framework to describe all stages comprised in the pharmaceutical
`development of a conventional cream in a comprehensible manner.
`© 2018 American Pharmacists Association®. Published by Elsevier Inc. All rights reserved.
`
`Introduction
`
`Over the last decades, our understanding about physicochemical
`properties of topical formulations and their excipients, result in the
`ability to develop physical, chemical and biologically stable prod-
`ucts. To design and develop a successful pharmaceutical dosage form
`for skin delivery, preformulation and formulation studies require
`particular considerations. Moreover, the knowledge of skin barrier
`structure and drug permeation properties is essential for a rational
`progress in the development of topical formulations.
`Dosage forms for topical application are intended to produce the
`required therapeutic action at specific targets in the skin with the
`least probable adverse effects.1,2 Topical formulations can be easily
`administered and transported, and are used for the treatment of
`several disorders. For any topical formulation, the onset, rate, and
`extent of
`therapeutic response depend on the efficiency of
`sequential processes: release of the active substance from the
`dosage form, penetration/diffusion of the drug through the stratum
`corneum (SC), and other skin layers, before producing the
`
`* Correspondence to: Ana Figueiras (Telephone: þ351-239-488-431).
`E-mail address: rfigueiras@ff.uc.pt (A. Figueiras).
`
`pharmacological effect. These different processes are variables
`which result in formulation safety and efficacy differences.3 There
`are several topical formulations available in the market; however,
`semisolids (e.g., ointments, creams and gels) are the most
`commonly used for this purpose.4
`Included in the latter category, conventional creams/emulsions
`represent a promising pharmaceutical vehicle for skin drug
`delivery despite their thermodynamic instability and complex
`formulation remaining a challenge for pharmaceutical technology.5
`Depending on the physicochemical properties, desired site of
`action, and drug delivery strategies, drugs incorporated into
`semisolid products can be applied for different purposes. A cream is
`a semisolid emulsion containing one or more active substances,
`dissolved or dispersed, and may be defined as a biphasic system in
`which the dispersed or internal phase is finely and uniformly
`dispersed in the continuous or external phase. According to the
`dispersed phase nature, it is possible to acquire an oil-in-water
`cream (o/w) or a water-in-oil cream (w/o).4,6
`Besides the several aspects taken into account in cream product
`design, during the whole process, it is imperative to preserve a high
`quality level. Thereby, in cream research and development, the
`application of a systematic approach is demanded to avoid product
`rejections during manufacturing and to achieve regulatory approval.
`
`https://doi.org/10.1016/j.xphs.2018.06.010
`0022-3549/© 2018 American Pharmacists Association®. Published by Elsevier Inc. All rights reserved.
`
`FlatWing Ex. 1049, p. 1
`
`

`

`2654
`
`A. Sim~oes et al. / Journal of Pharmaceutical Sciences 107 (2018) 2653-2662
`
`Over the years, pharmaceutical industries have spent significant
`efforts to ensure product quality, to achieve regulatory compliance,
`and to yield pharmaceuticals as cost efficient as possible. Therefore,
`they perform sophisticated processes and technologies that require
`steadiness among scientific progress and operational complexity.
`Nonetheless, such processes do not present a rational under-
`standing of critical variables and control strategies, which is
`imperative to ensure the product quality.7
`In this context, the U.S. Food and Drug Administration has
`highlighted Quality
`by Design (QbD)
`as
`current Good
`Manufacturing Practices initiative for the 21st century. The emer-
`gence of this approach has added a new dimension to pharma-
`ceutical development and manufacturing.8-10
`The implementation of the QbD approach includes the defini-
`tion of the quality target product profile (QTPP) and critical quality
`attributes (CQAs) of drug product, the accomplishment of risk
`assessment (International Conference on Harmonisation [ICH] Q9)
`to identify critical material attributes (CMAs) and critical process
`parameters (CPPs), the definition of a design space through design
`of experiments (DoEs), the establishment of a control strategy, and
`the continual improvement and innovation throughout the product
`life cycle.11
`As mentioned in the Q8 guideline, the aim of the pharmaceutical
`development based on a QbD approach is to design a successful
`product and its manufacturing process according to the intended
`quality performance. During product development, a detailed
`identification, understanding, and control of critical variables, with
`their optimal operating range definition, will enable to yield a
`product with the required quality profile. The information and
`knowledge gained from pharmaceutical development studies and
`manufacturing experience provide an enhancement of scientific
`understanding. Greater product and process understanding is also
`crucial for more flexible regulatory approaches. The degree of
`regulatory flexibility relies on the level of scientific knowledge
`provided in the registration dossier.
`Although the implementation of the QbD principles is one step
`forward for conventional solid form development, the case of
`conventional semisolid forms still remain largely unexplained. This
`review focuses on QbD approach for the development of semisolid
`dosage forms, particularly on cream formulations. Employing QbD
`principles to a complex formulation such as a cream, an effective
`product development, with an optimized formulation, and a
`continuous and robust manufacturing process, may be easily ach-
`ieved. Therefore, QbD approach will provide an opportunity for
`pharmaceutical
`companies
`to
`improve
`formulation
`and
`manufacturing efficiencies and productivity, with significant
`reduction in cost production, product variability, defects, and batch
`rejection, so as to get more flexible regulatory approvals, to
`decrease postapproval changes and to produce high-quality phar-
`maceuticals under real-time release.11-15 Thus, the predicted level
`established through this system is expected to be a scientific and
`technological progress for industries and regulatory authorities.
`
`QbD ApproacheCream Development Strategy
`
`Definition of Conventional Cream QTPP and CQAs
`
`The initial step when using QbD-based development is to pre-
`define the final quality profile. QTPP comprises cream quality
`parameters that should be ideally achieved at the final stage of the
`product development and production, considering its safety and
`efficacy.
`The second step of the QbD-based development is to identify the
`critical quality parameters. Derived from QTPP, CQAs are quality
`attributes that must be studied, controlled, and ensured during
`
`cream development and manufacturing to guarantee predefined
`product quality.7,11,16-18 An example of cream QTPP and its CQAs is
`provided in Table 1.
`
`Product Design and Development
`
`Once the dosage form is selected, the drug product development
`using the QbD approach is initiated. The main purpose of the
`product design is to develop a robust cream that can achieve the
`therapeutic objectives and quality attributes, remaining stable over
`the shelf life.
`
`Drug Substance
`The physicochemical and biological properties of the drug
`substance have a significant effect on drug product performance
`and manufacturability. Thereby, these properties must be identified
`to produce the right dosage form and to select the appropriate drug
`concentration, excipients, and process parameters.
`During preformulation studies, drug properties such as solubi-
`lity, partition coefficient (log p), particle size, pKa, permeability,
`melting point, and molecular weight need to be identified because
`of their role on percutaneous permeation.8,11,19,20
`The quality attributes of drug substance will ensure that the
`drug product meets its CQAs and must be controlled within the
`defined specifications.21
`
`Excipient Selection
`Special consideration needs to be given to excipient selection
`because of their influence also on the final product performance,
`manufacturability, and stability. This selection is related to the
`intended dosage form, route of administration, safety profile,
`manufacturing process, and regulatory aspects. Excipient nature
`and concentration will determine drug release from the dosage
`form, skin barrier features and drug penetration/diffusion, affecting
`the duration and extent of the therapeutic action at the target skin
`layer.22
`In cream formulation, excipients are used to improve drug sol-
`ubility and to incorporate it at the target concentration (solvents),
`to control drug release and cream viscosity (thickeners), to improve
`drug skin permeability (chemical permeation enhancers),
`to
`enhance drug and formulation stability (antioxidants, emulsifiers
`and buffers), and to prevent microbial growth and contamination
`(preservatives).23 Acceptable pharmaceutical excipients are listed
`in international pharmacopoeias for pharmaceutical product
`development.4
`At this stage, special consideration must be given to drug solu-
`bility because it will dictate the excipient selection because of its
`impact on diffusion through each skin environment, as well as on
`release pattern from the dosage form vehicle, final cream unifor-
`mity, and stability. If a suitable solvent is selected, to comprise the
`solubilizing phase of the emulsion system, an excellent skin
`permeation rate of the drug substance will be provided. According
`to drug physicochemical properties and dosage form (aqueous or
`oily solubilizing phase nature), different solvents have to be wisely
`selected and tested. The equilibrium solubility is defined as the
`maximum quantity of a drug which can be completely dissolved, at
`a given temperature and pressure, in a specific amount of solvent.
`Therefore, for a specific drug substance in the solid form, it is
`imperative to perform a solvent screening to determine the active
`equilibrium solubility in each promising solvent and later in the
`solvent blend/solubilizing phase.24-26
`Another important parameter to be considered for drug
`release performance and percutaneous absorption rate assess-
`ment is the thermodynamic activity of the drug in the formula-
`tion. Once exceeded the solubility equilibrium, a supersaturated
`FlatWing Ex. 1049, p. 2
`
`

`

`A. Sim~oes et al. / Journal of Pharmaceutical Sciences 107 (2018) 2653-2662
`
`2655
`
`Table 1
`Example of General Elements of QTPP and CQAs for a Conventional Cream Formulation
`
`QTPP
`
`Target
`
`CQAs
`
`Justification
`
`Dosage form
`Route of administration
`Dosage strength
`Dosage design
`
`Appearance
`Identification
`Assay
`
`Impurities
`
`Uniformity
`API particle size
`API crystallization
`pH
`Viscosity
`
`Cream
`Topical
`% w/w
`Oil in water emulsion cream with API dispersed in
`the cream base
`White smooth cream with dispersed API
`USP <621>/Eur. Ph. 2.2.29
`80%-90% of the saturation concentration in the
`solubilizing phase
`USP <1086>/Eur. Ph. 5.20
`
`USP <905>/Eur. Ph. 2.9.40
`USP <429>/Eur. Ph. 2.9.31
`USP <854>/Eur. Ph. 2.2.24
`USP <791>/Eur. Ph. 2.2.3
`USP <911>/Eur. Ph. 2.2.9
`
`Oil droplet size
`In vitro release profile
`
`USP <776>/Eur. Ph. 2.9.37
`In vitro Release Testing Guidance
`
`Preservatives content
`Microbial limits
`
`Consult USP <51>/Eur. Ph. 5.1.3
`USP <61>/Eur. Ph. 2.6.12
`
`Residual solvents
`Stability
`Container closure system
`Package integrity
`
`USP <467>/Eur. Ph. 5.4
`ICH QI A
`Appropriate for the dosage form
`Compatible
`
`e
`e
`e
`e
`
`e
`e
`Yes
`
`Yes
`
`Yes
`Yes
`
`Yes
`Yes
`
`Yes
`Yes
`
`e
`Yes
`
`e
`Yes
`e
`e
`
`e
`Local administration avoiding systemic side effects
`e
`e
`
`e
`e
`To set up the dose that will ensure drug availability to promote
`therapeutic effect
`Should be maintained below set limits to ensure safety and efficacy of
`formulation
`To assure consistency of the delivery system performance
`Smaller size facilitates drug permeation
`Impact on formulation uniformity and stability
`Impact on physicochemical stability
`To increase drug residence time at the site of application and
`consequently its action duration/To ensure drug release
`Impact on release/permeation behavior
`To assess formulation delivery performance for enhanced therapeutic
`efficacy
`
`e
`Must be maintained below the specified limits to ensure formulation
`safety and stability
`
`e
`Quality requirement
`To ensure target shelf-life
`e
`
`API, active pharmaceutical ingredient; EE, encapsulation efficiency; Eur. Ph., European Pharmacopeia; PDI, polydispersity index; USP, United States Pharmacopeia.
`
`solution is yielded and a higher thermodynamic activity is ach-
`ieved,
`increasing the driving force drug diffusion from the
`formulation.27-32
`Compatibility among excipients and drug(s) must be evaluated
`to anticipate any stability failures and possible incompatibilities in
`the final formulation.19 These studies are imperative in the drug
`product development process because the acquired information
`from compatibility data is applied to select the suitable excipients,
`to ensure active substance stability, to understand degradation
`products and its formation pathway, and to investigate reaction
`mechanisms, which help to prevent unexpected hurdles and
`identify stable storage conditions.33
`In the pharmaceutical technological field, there are different
`accepted methods for drug and excipients compatibility analysis.
`The accelerated stability test is the most common to evaluate
`chemical compatibility for topical formulation development.34
`In the sections that follow, the choice of excipients, their con-
`centration, and characteristics that can influence the drug product
`performance are discussed as well as their corresponding functions.
`
`Choice of the Oily Phase
`External emulsions comprise oily compounds as active sub-
`stance carriers. There are different oily excipients suitable to use in
`cream formulations. Saturated and unsaturated fatty acids/fatty
`acid esters,35 hydrocarbons, and polyols can constitute the oily
`phase, also functioning as penetration enhancers, and consistency
`or viscosity modifiers. Therefore, the selected oily excipients may
`also influence cream viscosity, drug solubility, physical stability,
`drug release performance and transport into the skin. Controlling
`emulsion consistency will ensure cream spreadability, but it needs
`to be slimy enough to form a continuous film over the skin.36
`
`Thickeners and Emulsifying Agents
`The physicochemical principles underlying emulsion formula-
`tion and stabilization are extremely complex. When 2 immiscible
`
`liquids are mechanically mixable without any interfacial stabiliza-
`tion, both liquids will form droplets, which rapidly flocculate
`(aggregation of dispersed droplets), coalesce (aggregation of floc-
`culated droplets with possible oily and water phases separation),
`and form a creamy layer (dispersed phase droplets on the top of the
`continuous phase).37,38 Physical stability is determined by the
`ability to mitigate these physical instability phenomena, and it may
`be accomplished by increasing the viscosity of the continuous
`phase, reducing the droplet movement rate of the dispersed phase,
`or decreasing interfacial tension between both phases by an
`emulsifier addition.
`It has been described a direct relationship among cream
`viscosity and the viscosity of its continuous phase, but it is also
`possible to improve its apparent viscosity by increasing the con-
`centration of the dispersed phase or reducing mean droplet size
`during homogenization process. A high apparent viscosity is
`imperative to retard the movement of dispersed phase droplets,
`keeping an emulsion physically stable.36
`Thickeners are important excipients with impact on cream
`viscosity and, consequently, on skin retention of the topical dosage
`form and on drug penetration. Thereby, it is crucial to carry out
`in vitro skin permeation studies to assess formulation release
`behavior. For example, the inclusion of methylcellulose and paraffin
`reduces dispersed droplets mobility in an o/w emulsion and in a w/
`o emulsion, respectively.
`Apparent viscosity can be also controlled by the homogeniza-
`tion process. This unit operation allows the reduction of droplet
`size and thus increases their number and surface area, increasing
`cream viscosity.
`The inclusion of emulsifying agent(s) is also imperative to assist
`the emulsification process during cream manufacturing and to
`ensure emulsion physical stability during the product shelf life.
`When an emulsion formulation is developed, the type of emulsi-
`fying agent (anionic, cationic or nonionic), hydrophilic-lipophilic
`balance (HLB), log p, and concentration are fundamental aspects
`FlatWing Ex. 1049, p. 3
`
`

`

`2656
`
`A. Sim~oes et al. / Journal of Pharmaceutical Sciences 107 (2018) 2653-2662
`
`to be considered in the emulsifier selection. Although this task
`seems to be limitless, there are some guidelines that may help in
`their correct choice. First of all, the use of pharmaceutically
`approved excipients will reduce regulatory justification periods.
`Emulsifier agent nature is extremely important for emulsion
`stabilization and generalization; ionic surfactants are used in o/w
`emulsions, whereas nonionic surfactants can be used in both o/w
`and w/o formulations. Essential information is provided by HLB
`system, a useful method for calculating the relative emulsifier
`amounts necessary to produce the most physically stable emulsion.
`Each surfactant is associated to an HLB number (usually between
`0 and 20), representing the relative ratio of surfactant lipophilic and
`hydrophilic proportions. Emulsifiers with high HLB values (hydro-
`philic or polar proprieties) enable to develop o/w emulsions,
`whereas surfactants with low HLB values (lipophilic or nonpolar
`characteristics) allow to formulate w/o emulsions.36
`Physical stability of semisolid formulations can be assessed
`through different analytical methodologies. Microscopic examina-
`tion, centrifugation tests, and viscosity analysis are essential tech-
`niques performed for this purpose.36,39,40
`
`Preservatives and Antioxidants
`Oils and fats used in emulsion formulation are susceptible to
`oxidation by atmospheric oxygen or the action of microorganisms.
`The atmosphere oxidation results in degradation products which
`confer unpleasant cream characteristics. The resulting instability
`can be prevented by introduction of excipients with antioxidant
`properties. The selection of the antioxidant and its concentration
`can only be determined by testing its effectiveness on the final
`product, according to pharmacopoeial information. Their efficiency
`depends on its compatibility with other excipients and on its oil/
`water partition coefficient.
`Oxidation from microbiological source influences the physico-
`chemical properties of the emulsion, such as color and odor
`changes, hydrolysis of fats and oils, pH changes in the aqueous
`phase, and breaking of emulsion. Emulsions with aqueous contin-
`uous phase nature are more susceptible to microbial contamina-
`tion. Therefore,
`it is required to include antimicrobial agents
`(preservatives) to prevent any microorganism growth. A preser-
`vative suitable for an emulsion must present wide spectrum of
`bactericidal activity, low log p, compatibility with other excipients,
`stability, and effectiveness over a wide range of pH and
`temperatures.
`
`Buffer Agents
`To provide chemical stability and to ensure physical compati-
`bility, it is necessary to include buffer agents, but these electrolytes
`have to be carefully added to avoid undesirable effects on physical
`stability (e.g., rheological behavior).36,40
`All components are related to the requirement that formulation
`must be capable of delivering the correct amount of drug to the
`therapeutic application site, be free from microbial contamination,
`and physically unchanged since the manufacturing day.
`
`Process Design and Development
`
`Since a formulation cannot become a product without a process,
`process and product design and development cannot be separated.
`Process design is an initial phase of the process development and
`should include all factors that need to be considered in cream
`production,
`such
`as
`equipment, material
`transfer,
`and
`manufacturing variables. Therefore, process selection depends on
`the product design and material properties. Once process design
`and development has been concluded, preliminary studies are
`
`conducted in accordance to the cream QTPP, product, and process
`knowledge.
`To produce the intended quality product, a series of unit oper-
`ations are accomplished throughout the cream manufacturing
`process. A unit operation involves physical or chemical changes
`while process parameters are referred to the input operating
`parameters (e.g., speed) or variables associated with a specific
`operation (e.g., temperature).
`
`Manufacturing Process
`In cream formulation production, the first mechanical process
`carried out is the mixture of both the aqueous and oily phases by
`adding the dispersed to the continuous phase or of the continuous
`to the dispersed phase. Sometimes, o/w emulsions are prepared
`through the phase inversion technique, in which the aqueous phase
`is slowly added to the oily phase. Initially, a w/o emulsion is formed,
`but as further aqueous phase is added, the emulsion inverts to form
`an o/w emulsion. It should be evaluated the effect of the order of
`addition and the rate of addition on the drug product quality
`attributes.
`Prior to mixing, different excipients are dissolved in the phase in
`which they are soluble. The initial mixing temperature of both
`phases should be high enough to ensure intimate liquid mixing and
`avoid premature solidification of the oily phase by the colder water.
`Aqueous phase should be warmed to a temperature slightly higher
`than the oily phase.
`Some active pharmaceutical ingredients can be dissolved at high
`temperature and recrystallized during the cooling stage. In this
`case, the active substance can be carried to the cooled down cream
`base via a powder eduction system or through a slurry addition and
`simultaneously mixed into the cream base, thus preventing the
`recrystallization problem. The stage to introduce the active sub-
`stance into the semisolid mixture may be critical and should be
`identified.20,39
`The next step in cream production is the homogenization stage.
`Agitators, mechanical mixers, rotor stators, homogenizers, or
`ultrasonic devices could be employed to ensure uniform excipient
`dispersion and droplet size reduction. To remove cream air pockets,
`a deaeration via vacuum with low-speed mixing may be turned on
`to the system. Homogenization time and vacuum pressure are
`significant process variables that can affect physical stability (e.g.,
`coalescence of droplets, phase separation) and homogeneity.
`During cream process development and manufacturing, time,
`temperature, and mechanical energy are high-risk variables of the
`homogenization equipment that must be controlled to produce
`consistent quality.
`Furthermore, as cooling rate can influence the final product
`quality, different cooling rates after melting, mixing, and homog-
`enization steps should be additionally investigated as process
`variables.
`Visual inspection is a useful and simple confirmatory test to
`ensure solid dissolution or uniformity system before proceeding to
`the next step. Microscopic visualizations should be also performed
`to select homogenization speed and time, so as to enable proper
`incorporation of the active substance into the base and conform the
`microscopic appearance, including drug particle size and droplet
`size.14,41
`
`Risk Assessment
`
`Based on prior knowledge, an initial risk assessment is per-
`formed to identify and prioritize potential high-risk variables that
`may influence identified cream CQAs. The outcome of this pro-
`cedure is to determine which material attributes and process pa-
`rameters are critical and which ones need to be experimentally
`FlatWing Ex. 1049, p. 4
`
`

`

`A. Sim~oes et al. / Journal of Pharmaceutical Sciences 107 (2018) 2653-2662
`
`2657
`
`Figure 1. Ishikawa diagram showing critical parameters of a conventional cream pharmaceutical development. Log p, octanol-water partition coefficient.
`
`investigated and controlled within appropriate ranges to ensure
`cream quality.
`Using a risk approach, the starting point must be the identifi-
`cation of all material attributes and process parameters that can
`influence product CQAs and hence generate quality failure. To
`ascertain which of these parameters need to be further studied and
`controlled, an Ishikawa diagram is constructed (Fig. 1).42,43 In
`addition, a risk estimation matrix (REM) is carried out to prioritize
`material attributes and process parameters that were demon-
`strated to be a potential risk factor for cream CQAs (Fig. 2).44-46
`In pharmaceutical development, risk assessment analysis must
`be accomplished in early phases, but it is important to be updated
`at different development stages as further information becomes
`available and greater knowledge is obtained.47,48 After risk assess-
`ment data analysis, specific parameters are, then, selected for
`subsequent screening studies (DoEs).
`
`Formulation Optimization
`
`After performing screening experiments, variables that show
`criticality in the previous phase are also optimized through a DoEs.
`The optimization stage aids to specify CMAs and CPPs optimal
`settings, and, ultimately, the definition of design space. Note that
`any manufacturing process developed within the design space will
`be regulatory acceptable.
`Response surface designs, such as Central Composite Design and
`Box Behnken are the most usual models to predict the optimal
`CMAs and CPPs ranges. Currently, some software packages are
`available to simplify experimental design procedure and assist in
`results interpretation: MODDE, Design Expert Design-Ease, and
`JMP.52-56 The application of the QbD concepts to optimize formu-
`lation, single-unit operation, or to the entire manufacturing pro-
`cess, is intended to support end-product quality and real-time
`release.11,14
`
`Identification of CMAs and CPPs
`
`Formulation Performance Testing
`
`In cream development, several variables need to be considered
`and investigated to reach predefined QTPP. CMAs and CPPs that
`may specifically influence one or more cream CQAs must be iden-
`tified to develop an adequate cream formulation (Tables 2 and 3).
`Critical variable identification is the preliminary step in the
`optimization methodology, which is established through a
`screening process. A screening design is an experimental planning
`where a relatively large number of
`factors is simultaneously
`evaluated using a small number of experiments. During the
`screening phase, all factors are tested to indicate the most critical
`ones. These designs enable to detect which variables are respon-
`sible for the final product quality to eliminate all those ones which
`do not play an important role in ensuring quality constancy.
`Different experimental designs, such as, full factorial, fractional
`factorial, and Placket-Burman designs are usually used for
`screening purposes.18,49-51
`
`According to the predefined QTPP, cream performance needs to
`be assessed. There are 3 important CQAs to consider in topical
`formulation development: formulation stability, drug release pro-
`file, and skin permeation behavior.11,33,57
`
`Stability Testing
`To identify formulation stability issues, formulation stability
`studies must be performed.34 This stage aims at developing a sta-
`bility screen of prototype formulations to select the most promising
`ones for in vitro drug release and permeation testing.
`
`In Vitro Release Studies
`The release rate of an active substance from a dosage form is a
`critical feature for semisolid dermatological products because the
`active must be released from the vehicle and diffused through the
`FlatWing Ex. 1049, p. 5
`
`

`

`2658
`
`A. Sim~oes et al. / Journal of Pharmaceutical Sciences 107 (2018) 2653-2662
`
`Figure 2. Risk estimation matrix presenting initial risk assessment levels of individual formulation and manufacturing parameters: Low, low risk parameter; Medium, medium risk
`parameter; High, high-risk parameter; Log p, Octanol-water partition coefficient.
`
`skin. When a topical formulation is applied to the skin, drug ther-
`modynamic activity must be suitable to ensure an adequate release
`rate from the final topical dosage form. To characterize and
`
`optimize formulation release performance, in vitro release tests are
`performed. In QbD context, it is imperative to select a suitable
`release study to discriminate differences on cream release rate
`FlatWing Ex. 1049, p. 6
`
`

`

`A. Sim~oes et al. / Journal of Pharmaceutical Sciences 107 (2018) 2653-2662
`
`2659
`
`Table 2
`CMAs of a Cream Formulation
`
`Formulation
`Components
`
`Parameter
`Type
`
`CQAs
`
`Table 3
`CPPs of a Cream Formulation
`
`Risk Assessment
`
`Parameter
`Type
`
`CQAs
`
`Drug substance
`Physical attributes
`Log p
`Molecular weight
`Melting point
`
`Equilibrium
`Solubility and
`Concentration
`Assay
`Related substances
`Excipients
`Compatibility
`Viscosity
`Concentration
`Melting point
`Oil/water ratio
`Emulsifying agent
`Type
`Log p
`HLB
`Concentration
`Melting point
`Preservatives
`Log p
`Concentration
`Melting point
`Antioxidants
`Log p
`Concentration
`Melting point
`
`e
`CMA
`CMA
`CMA
`
`CMA
`
`CMA
`CMA
`
`CMA
`CMA
`CMA
`CMA
`CMA
`
`CMA
`CMA
`CMA
`CMA
`CMA
`
`CMA
`CMA
`CMA
`
`CMA
`CMA
`CMA
`
`e
`Drug release/permeation, stability
`Permeation
`Drug release/permeation, viscosity,
`stability
`Drug release/permeation, uniformity,
`viscosity, stability
`
`Content uniformity, drug release
`Degradation products
`
`Stability
`Viscosity, stability
`Uniformity, viscosity, stability
`Viscosity, stability
`Stability
`
`Droplet size, uniformity, stability
`Stability
`Droplet size, uniformity, stability
`Droplet size, uniformity, stability
`Viscosity, stability
`
`Stability
`Stability
`Viscosity, stability
`
`Stability
`Stability
`Viscosity, stability
`
`Log p, octanol-water partition coefficient.
`
`when variations in formulation and manufacturing parameters of
`the drug product are applied and assessed.3,58-62
`commonly
`Topical
`formulations
`release performance is
`performed by Franz diffusion cells, where synthetic membranes
`(silicone, polycarbonate, or cellulose) have received distinct
`attention because of their ability to mimic physiological and
`anatomical skin conditions. This is a well-established method
`where a specific dose of formulation is applied on a membrane
`surface in the open donor chamber of the cell. The diffusion of
`drug from the topical product across the membrane is monitored
`by assay of sequentially collected samples from the receptor
`chamber. The receptor compartment of the Franz cells is filled
`with a suitable receiver fluid to maintain sink conditions
`embedded in a water bath at 37C, so as to ensure a temperature of
`32C at the surface. The receptor chamber content is agitated by

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