throbber
-_°=aoSNo=xudate)N=°°oNoN4a@#<x2a4OozOo2Ooz>0z4udNLea
`
`BONITO
`
`John A.
`
`TTe]
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 1 of 54
`
`

`

`onacid-free
`
`Ma |attheaddressbelow.
`
`This bookis printed
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 2 of 54
`
`

`

`r
`
`4
`
`Preformulation
`Development of
`
`Parenteral Biopharmaceuticals
`
`JOHN A. BONTEMPO
`
`
`
`
`Biopharmaceutioal Product Development, East Brunswick, New Jersey
`
`I.INTRODUCTION
`92
`A.Considerations of Domestic and International
`Distribution of the Product
`93
`
`Points to Consider for Constituted Versus
`B.
`
`Lyophilized Fonnulations
`93
`c.Unit Dose or Muleidose
`94
`
`D.Physicochemical Factors to Be Considered for
`Protein Drug Formulations
`
`94
`
`II, INITIAL PREFORMULATION STUDIES:
`
`PARAMETERS AND VARIABLES TO BE TESTED
`95
`95
`A.Initial Variables to Be Tested
`B.Preliminary Analytical Development
`96
`
`C.Experimental Conditions for the Initial
`
`
`Preformulation Studies
`
`97
`
`Ill. MECHANICAL AND PHYSICAL STRESSES
`
`99
`A.Shaking Effect on Protein Solution at the
`
`Preformulation Level
`99
`
`Freeze-Thaw Experiments
`B.
`
`100
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 3 of 54
`
`

`

`C. Aggregation
`D, Denaturation
`E. Precipitation
`F. Adsorption
`
`VI.
`
`SUMMARY
`
`REFERENCES
`
`1.
`
`INTRODUCTION
`
`104
`105
`105
`106
`
`106
`
`107
`
`Preformulation research studiesof protein therapeutics encompass biophar-
`maceutical, physicochemical, and analytical investigations in support of
`subsequent stable formulations for preclinical, clinical, and market usage.
`In this highly competitive protein therapeutics field, it is very impor-
`(ant to obtain significant, measurable progress with preformulationsstudies
`in a timely manner. How extensive these studies are will depend on the
`availability of the crude, active drug substance and the intended route
`of administration. Most often, these studies begin with extremely small
`amounts of crude bulk active substance and, as more material becomes
`available with greater purity, more studiesareinitiated,
`From an industrial point of view, the preformulation studies are de-
`signed to cover a wide range of properties in a short time to learn as much
`as possible, but not in great depth. The pharmaceutical formulation scien-
`tist is very much interested in identifying potential problems early enough
`to evaluate potential alternatives to stabilize future formulation(s).
`
`
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 4 of 54
`
`

`

`Manyglobaljoint ventures and partnerships today in the biopharmaceuti-
`cal industry dictate various pharmaceutical, clinical, and marketing strate-
`gies, The regulatory requirements and acceptance of formulation excipi-
`ents, packaging components, unit dose versus multidose product, and
`stability conditions vary from continent to continent. Constituted and/or
`lyophilized dosage forms must also be considered. The developmentoffor-
`mulation considerations should be on a worldwide acceptancebasis,
`
`B, Points to Consider for Constituted Versus Lyophilized
`Formulations
`
`Someofthe key points to be considered for a constituted formulation are:
`* Aconstituted formulation maybe less stable than a lyophilized one
`* Effect of agitation during manufacturing and shipping
`*
`Interaction of the liquid with the inner wall of the glass vial and
`with the elastomeric closure
`* Aggregation problems
`* Head space within the vial
`* Preservative effectiveness
`
`Someof the key points to be considered for a lyophilized formulation
`
`are:
`
`* Better stability than a constituted product
`* Determination of an optimal lyophilization cycle
`* Effects of residual moisture on the activity and stability of the product
`* Ease of reconstitutability. Clinicians, nurses and trained home us-
`ers, prefer reconstitutability of the product within two minutes.
`
`
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 5 of 54
`
`

`

`Cc. Unit Dose or Multidose
`
`The decision to select unit dose versus multidase should be based upon
`inputfrom clinical investigators, focus groups, marketing surveys, and com-
`petitors’ products. A multidose formulation will require significantly more
`time for development,
`The multidose will require the screening and incorporation of com-
`patible preservative(s) with the protein formulation. This formulation will
`be tested to determine ifit is efficacious enough to meet the United States
`Pharmacopeia (USP) requirements. Meeting these requirements, it can
`qualify as a “multidose” for the U.S. market. However,if the formulation
`is also designated for international market, there are three additional fac-
`tors that must be taken into account. Thefirst is that for the “antimicrobial
`effectiveness test,” a particular country may or may not accept the preserv-
`ative selected. Secondly, the concentrations of the preservative present in
`the formulation maybe different from the USP requirements. Thirdly, the
`time periods required for the inhibition of the bacteria and fungi strains
`tested may also differ, Consequently, I strongly suggest that the interna-
`tional regulatory requirements for compliance should be well researched
`and understood by the scientific and management staff. Other excipients
`should also be thoroughly reviewed for international acceptance.
`
`D. Physicochemical Factors to Be Considered for
`Protein Drug Formulations
`
`Someof the most important physicochemical properties of protein drugs
`required for the development of parenteral preformulations and formula-
`tions are found in Table 1.
`
`
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 6 of 54
`
`

`

`
`
`pH Surface denaturation
`
`Since this may be an early stage of process development, some of the
`properties listed in Table 1 may not be available initially, simply because
`there was not enough time or personnel to perform the work.
`
`il.
`
`INITIAL PREFORMULATION STUDIES: PARAMETERS
`AND VARIABLES TO BE TESTED
`
`The pharmaceuticalformulationscientist will consider several factors in the
`preformulation designs. The data received from the Process/Purification
`section are reviewedfor structure, pH and purity of the substance, prelimi-
`nary bioassay, and an immune assay used in terms of semiquantitative
`measurements,
`Other important information that may or may not be available are
`product solubility, preliminary stability, potential degradation routes. From
`personal experience, there is only minimal crude bulk active substance at
`this early stage.
`
`A.
`
`Initial Variables to Be Tested
`
`Perhaps 10 or more initial preformulation combinations should be consid-
`ered. Theinitial variables to be tested with various protein concentrations
`are the effects of buffer species, ionic strength, pH range, temperature,
`
`
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 7 of 54
`
`

`

`two different methods that are “stability indicators,” most often fully vali-
`dated, Dr. Sharma, in Chapter6, will cover the bioanalytical development.
`
`TABLE 2 Bioanalytical Methodsto EvaluateInitial Preformulation Development
`Method
`Function
`
`Bioassay
`
`Immunoassay
`
`pH
`SDS-PAGE (Reduced &
`nonreduced)
`RP-HPLC
`
`|EF
`
`SE-HPLC
`
`N-terminal sequencing
`UV
`
`CD (circular dichroism)
`in the UV region
`
`Measure of activity throughout shelf life of a
`formulation
`
`Purity assessment and measures concentra-
`tion of a particular molecular species
`Chemical stability
`Separation by molecular weight, characteriza-
`tion of proteins and purity
`Estimation of purity, identity, and stability of
`proteins. Separation and analysis af pro-
`tein digests,
`Determinesthe isoelectric point af the protein
`and detects modifications of the protein
`Method of separating molecules according
`to their molecular size and purity
`determination
`Elucidation of the C-terminus,identity
`Detection of individual component, concentra-
`tion, and aggregation
`Detects secondary and tertiary conformation
`and quantitates various structures
`
`
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 8 of 54
`
`

`

`pH Range
`
`Initially, a range of pHs should be selected, for example, 3, 5, 7 and 9. Spe-
`cific pH units will be determined during the formulation studies. The pH
`changes may have varying impacts on thesolubility and stability of the for-
`mulation. pH control in pharmaceutical dosage formsis very critical (1).
`The proper pH selection is one of the key factors in developing a stable
`product.
`
`Buffers
`
`The buffer(s) selection should be made from the USP physiological buffers
`list and should be selected based upon their optimal pH range. Some of
`these buffers are acetate pH 3.8-5.8, succinate pH 3.2-6.6, citrate pH 2.1—
`6.2, phosphate pH 6,2-8.2, and triethanolamine pH 7.0-9.0, These pH
`ranges will differ from protein to protein.
`Buffer concentrations should be in the range of 0.01 to 0, molar con-
`centration, As buffer concentration goes up, so does the pain upon injec-
`tion. In selecting the proper buffer, phosphate should be the last in one’s
`choice. Phosphate buffer reacts with calcium from the glass vial and zinc
`from the rubber stopper to cause glass laminates and eventually haziness
`of the solution during stability periods.
`
`Other Excipients to Be Considered
`
`As it was stated previously, the objective of a preformulation study is to
`select potentially compatible excipients in orderto hasten the development
`of stable formulations. Based upon protein chemical and physical instabil-
`
`
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 9 of 54
`
`

`

`Antioxidants
`
`Since oxidationis one of the majorfactors in protein degradation,it is highly
`recommended,should the use of a specific antioxidant be required, to in-
`clude into the preformulation an antioxidant such as ascorbic acid, sodium
`disulfide, monothio-glycerol, or alpha tocopherol. The role of an antioxi-
`dant is to deplete or block a specific chain reaction. Antioxidants will be the
`preferential target and eventually be depleted, or may block a specific chain
`reaction. Argon and/or nitrogen gascan also be usedto flood the head space
`of a vial or ampule duringsterile filling to prevent or retard oxidation. A
`recommended antioxidant dose would be about 0.05 to 0.1%.
`Preservatives
`
`If a multidose formulationis required, an antimicrobial agent,called pre-
`servative,is required to be incorporatedinto the formulation, The preserv-
`ative effectiveness must comply with the USP requirementsto be qualified
`as multidose. The most often used preservatives and respective concentra-
`tions are phenol (0.3 to 0.5%), chlorobutanol (0.3 to 0.5%) and benzyl al-
`cohol(1.0 to 3.0%). Additional details are provided in Chapter5.
`Surfactants
`
`Judicious selection of surfactants can result in the prevention of aggrega-
`tion and stabilization of proteins (2). Polysorbate 80, poloxamer 188, and
`pluronic 68 have been used in injectable formulation. The purity of the
`surfactant may have an impact on the chemicalstability of the preformula-
`tion. Peroxide residues in the surfactant have been implicated in oxidations
`of protein.
`
`
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 10 of 54
`
`

`

`ceiving. Simulation of some of the functions described above need to be
`
`Membranefiltration is the most often used techniqueto sterilize protein
`solutions. The chemical nature of the filter and the pH ofthe protein solu-
`tion are the two mostimportant factors affecting the protein adsorption(3).
`However, there are other issues that require consideration, The formula-
`tion scientist must be aware ofparticles or fibers released duringthefiltra-
`tion, the potential extractables that may occur, the potential toxicity of the
`filter media and the product compatibility with the membrane. Of al] the
`filters tested (unpublished data) polyvinylidene difluoride, polycarbonate,
`polysulfone, and regenerated cellulose were found to be the most compat-
`ible with various proteins and with minimal amountsof protein binding and
`deactivation.
`
`to achieve greater compatibility, flexibility, low levels of particulates, and
`machinability. In addition, adsorption, absorption, and permeation through
`the stopper are essentially eliminated. Extensive details may be found in
`Chapter8.
`Membrane Filter Selection
`
`lil. MECHANICAL AND PHYSICAL STRESSES
`
`A. Shaking Effect on Protein Solution at the
`Preformulation Level
`
`Someof the various physical modesofvialed protein solution can undergo
`begin with the bulk active formulation,filling of formulated solution into
`vials or ampules, visual inspection, labeling, packaging, shipping, and re-
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 11 of 54
`
`

`

`These experiments will also be describedin later chapters andwill be part
`of Chapter 5. These experiments require a fair amountof active drug sub-
`stance as well as a fair amount of work.At this point of developmentthere
`may not be enough active drug substanceavailable,
`
`C. Filling Systems
`
`Ofall the filling types employed to dispenseliquid, such as time-pressure,
`piston, and rotary pump,the rolling diaphragm metering pumpis the one
`ofchoice for filling biopharmaceuticalsolutions. The internalparts ofthe
`pump do not come in contact with one another where the liquid solution
`flows. This is the “TL Systems Rolling Diaphragm Liquid Metering Pump”
`(4). One of the most important features of this pumpis that it eliminates
`the principal cause of particulate generation which is most often induced
`by parts coming togethercreating shedding of microscopic particles.
`There are three other important parameters to control while dispens-
`ing protein solutions. (1) The speed at whichliquidis filled into the vials.
`With protein solutions the maximum speed is between 25 to 30 vials per
`minute, delivering 0.5 to 2.0 mL volume per 5- or 10-mL vial per single
`filling head. If a large numberofvials need to befilled,this filling system
`can accommodate variable numbers offilling heads, thus allowingit to fill
`a large numberofvials, Filling at a faster rate will result in protein precipi-
`tation and aggregation. (2) The inner diameterofthefilling cannula should
`not be so very small as to induce shearing and aggregation of the protein
`solution, (3) Thetip of the cannula for the filling head should be bent at
`such an angle as to deliver the fluid against the inner wall of the vial and
`not perpendicular to the bottom ofthevial, This will result in a gentle flaw
`
`
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 12 of 54
`
`

`

`Continueif stable
`
`TABLE 3 Guideline for Preformulation Stability Studies
`Temperatures Timepaint
`Frozen controls (-80 and -20°C)
`Reference control sample as needed
`Refrigerated (2-8°C)
`T = 0, 6, 12, 24 & 48 weeks
`Continue if stable
`T=0, 4, 8, 12, 18, 24 weeks
`Continue if stable
`T=0,1, 2, 4,8, 12 weeks
`
`fers refrigerated temperature storage between 2 and 8°C.
`The presentstorage conditionsset up by the USP on storage require-
`ments are as follows:
`
`* Cold storage. Any temperature between 2 and 8°C
`* Cool. Any temperature between 8 and 15°C
`* Room temperature. Temperature prevailing in a working area
`* Controlled room temperature. Temperature controlled thermo-
`statically between 15 and 30°C
`« Excessive heat. Temperature exceeding 40°C
`
`Table 3 summarizes the initial guideline time points and tempera-
`tures that preformulation solutions should be exposed to. The results from
`the preformulationswill allow the review team to determine directions to
`manipulate the excipients to obtain better stability,
`
`
`
`Intermediate (20, 30, 37°C)
`
`High temperature (40, 45, 50°C)
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 13 of 54
`
`

`

`to understand how proteins degrade, how theyare affected by the compo-
`sition of the formulation, and the effects ofstability conditions. The major
`pathways of protein degradation are chemical and physical. Under chemi-
`cal degradation, changes and modifications occur due to bond formation
`or cleavage, yielding new chemicalentities. One or moreofthe following
`can occur: oxidation, deamidation, hydrolysis, racemization, isomerization,
`beta elimination, and disulfide exchange. Physical instability can occur in
`the form of denaturation, aggregation, precipitation, and adsorption with-
`out covalent changes.
`
`A. Oxidation
`
`Oxidation of protein is perhaps one of the most common degradation
`mechanisms that can take place during various stages of the processing,
`such as fermentation, purification, filling, packaging, and storage of the
`biopharmaceuticals, Under oxidative stress and in the presence of trace
`metals, amino acids such as methionine (Met) can be oxidized to methion-
`ine sulfoxide, cysteine (Cys) to cysteine disulfide, as well as tryptophane
`(Try) and histidine (His) via other modifications.
`Oxidation can be controlled or minimized by (1) the addition of an-
`tioxidants, (2) havingstrict controls on the processing operations, (3) using
`nitrogen gas to flood head space of the container.
`Oxidized human growth hormone (hGH) retains only 25 percent the
`activity of the native molecule, recombinant interferon-beta loses consid-
`erable antiviral activity due to oxidation (5). Oxidation can be detected by
`reversed phase HPLC (RP-HPLC), high-performanceisoelectric chroma-
`tography (HP-IEC), peptide mapping, amino acids analysis, and mass spec-
`
`
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 14 of 54
`
`

`

`Hydrolysis is another mostlikely cause of degradation ofproteins. It in-
`volves a peptide (amide) bondin the protein backbone (5). The most influ-
`ential factor affecting the hydrolytic rate is the solution pH.
`
`D. Racemization
`
`Proteins may also degrade via other modifications (10) such as racemiza-
`tion. This mechanisminvolves the removal of the alpha proton from an
`amino acid in a peptideto yield a negatively charged planar carbanion. The
`proton can then be replaced into this optically inactive intermediate, thus
`producing a mixture of D and L enantiomers (2). Racemization can yield
`enantiomers in both acidic and alkaline conditions.
`
`E.
`
`Isomerization
`
`Protein degradation is also induced by isomerization. Hydrolysis of cyclic
`amidesof asparagine, glutamine, and aspartic acid will result in isomeriza-
`tion. Low pH accelerates hydrolysis of asparagine and glutamine. However,
`high pH accelerates hydrolysis of aspartic acid and glutamic acid (2,11,12).
`
`F. Disulfide Exchange
`
`Disulfide exchange may result from a degradation other than covalent
`modification. These reactions may include the disulfide exchange ofcys-
`teine. This reaction is base, catalyzed and promoted bythiol antioxidants
`(13).
`
`
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 15 of 54
`
`

`

`A. Covalent Aggregation
`This pathway involves modification of the chemical structures resulting in
`new chemical structures and may include reactions, such as oxidation, de-
`amidation, proteolysis, disulfide interchanges, racemization, and others.
`
`B. Noncovalent
`
`This instability may be induced byagitation, shear, precipitation, and ad-
`sorption to surfaces.
`
`C. Aggregation
`Protein aggregation derived from either physical or chemicalinactivation,
`is presently a major biopharmaceutical problem (17-21). Aggregation can
`be either covalent or noncovalent, occurring during any phase of product
`development from purification to formulation, An early detection of aggre-
`gation via biochemical or spectrophotometric methods,or both, can be of
`significant guidance to formulationscientists in selecting compatible excipi-
`ents to minimize and/or prevent its formation in the experimental formu-
`lation.
`Formation of aggregation can begin by the formationofinitial parti-
`cles from protein molecules via the Brownian movement. Thisis followed
`bycollision of these molecules and aggregatesofvarying sizes can be formed.
`These aggregates can be generated by shearorcollisional forces (22).
`Detection and measurementsof aggregations can be performed by a
`numberof techniques. Visual observations,light scattering, polyacrylamide
`gel electrophoresis, UV, spectrophotometry,laserlight diffraction particu-
`
`
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 16 of 54
`
`

`

`Denaturationofproteins can be the result of several processes and reported
`by several investigators (27).
`Factors which induce denaturation are heat or cold, extreme pHs,
`organic solvents, hydrophilic surfaces, shear, agitation, mixing,filtering, shak-
`ing, freeze-thaw cycles, ionic strength, and others. Thermal inactivation
`processes will induce conformationalside reactions and destruction of amino
`acids (28). The loss of biological function may well be attributed to the
`effect of the temperature on the higher-ordered structure of the protein.
`Thermal denaturationofproteinsis of great interest to the formulation
`scientist. Thermal probesoffer tools to study protein structure andstability
`that ultimately can beofsignificant use to stabilize protein drug formula-
`tions. Modifications of protein thermaleffects have been reviewed (8).
`Theability of the protein to refold from a denatured state, a reversible
`heat denaturation, is also of considerable interest for the stability of a pro-
`tein formulation. These processes of renaturation are very complex (29),
`and each protein does have its own unique renaturation mechanisms.
`Since filtrations and volume reductions occur from the fermentation
`to process purification,thereis very likely inactivation of the protein attrib-
`utable to shearing effect.
`
`E. Precipitation
`
`Precipitation in formulations can occur by a variety of mechanismssuch as
`shaking, heating,filtration, pH, and chemical interactions, Aggregation is
`the initial onset of precipitation. The protein molecules form aggregations
`of varying sizes first, and later when the aggregates reacha critical mass,
`precipitate out of solution and are clearly visible.
`
`
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 17 of 54
`
`

`

`air during mixing (process),filtration (process), and air in the process steps,
`a significant surface area has been encounteredto yield interphases.
`During the actual final manufacturing of vials. ampules, syringes,
`catheters, pumps, and their respective storage conditions, the proteins
`could be adsorbedat the interphase and removed from the solution.
`Several researchers (22,33,34,35,36) have investigated these bio-
`chemical mechanism problems. Since proteins have surfactant charac-
`teristics, they have a high affinity to adsorptionat the air—liquid and solid-
`liquid interphase. Hydrophobic and hydrophilic interactions which are con-
`centration dependent, determine the extent and the rate of adsorption. The
`adsorption effect on the protein is the unfolding of the protein. When this
`occurs at an interphase, it can lead to (1) inactivation of the protein solu-
`tion, (2) insoluble protein aggregates being formed at the adsorbedsite, (3)
`additional conformational changes occurring, and (4) chemical degrada-
`tion of the protein continuing duringstability periods.
`
`Vi. SUMMARY
`
`Theinitial critical parameters of preformulations have been addressed in
`this chapter, The formulation team, at this point of development,will re-
`view and evaluateall the results obtained from the preformulation studies.
`The pharmaceutical formulator will design several approachesfor the next
`stage of formulation development taking into accountall the parameters
`that may achieve one or more stable marketable formulations. In the for-
`mulation studies ahead, a numberofstabilizing ingredients should be con-
`sidered to achieve acceptable industrial stability.
`
`
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 18 of 54
`
`

`

`. Fischer G, et al. Biochem Biophys Acta 1984; 87:791.
`. Bachinger HP, J Biol Chem 1987; 262:17144.
`. Kenney J, et al. Lymph Res 1986; 5:23.
`. Constantino RH,et al. Pharm Res 1994; 71:21.
`. Kenney WC,et al. Lymph Res 1986; 5:23.
`. Shihong L, et al. Pharm News 1995; 2:12.
`. Klibanov AM. Adv Appl Microbiol 1983; 29:1.
`. Manning MC,Patel K, Borchardt RT. Pharm Res 1989, 6:903.
`. Weiss M. Genetic Engineering News 1994, Jan.
`. Creighton TE.Proteins: Structures and Molecular Properties. New York: WH
`Freeman, 1984.
`. Scopes RK. Protein Purification Principles and Practices. 2d ed. Berlin: Sprin-
`ger-Verlag, 1987.
`. Glatz EC. Chemical and physical pathways of protein degradation. In: Ahern
`JT, Manning M,eds.Stability of Protein Pharmaceuticals, Part A. Plenum Press,
`1992.
`. Dingledine M,et al, Pharm Res Abstracts 1993; 10:5-82.
`. Watson E, Kenney WC. J Chromatogr 1988; 436:289.
`. Quinn R, Andrade JD. J Pharm Sci 1983; 72:1472.
`. Cantor CR, Timasheff SM. The Proteins. Vol. 5, 3d ed. New York; Academic
`Press, 1982:145,
`. Shirley AB. Chemical and physical pathways of protein degradation. In: Ahern
`JT, Manning M,eds, Stability of Protein Pharmacsuticals, Part A. Plenum Press,
`1992.
`. Volkin BD, Middaugh ER. Chemical and physical pathways of protein degra-
`dation. In: Ahern JT, Manning M, eds. Stability of Protein Pharmaceuticals,
`Part A. Plenum Press, 1992.
`. Jaenicke R. Prog Biophys Molec Biol 1987; 49:117.
`. Mizutani T. J Pharm Sci 1980; 69:279.
`
`
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 19 of 54
`
`

`

`
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 20 of 54
`
`

`

`Biopharmaceutical Product Development, East Brunswick, New Jersey
`
`I.
`
`FORMULATION REQUIREMENTS
`A. Characterization, Homogeneity, and
`Reproducibility of the Bulk Active Drug
`B, pH Effect on a Formulation
`C. Stabilizers Used in Protein Formulations
`D. Surfactants
`E. Buffer Selection
`F. Polyols
`G. Antioxidants
`H. Antimicrobials (Preservatives)
`lL Tonicity
`
`fl. CONTAINER-CLOSURE INTERACTIONS
`A. Glass Vials
`B. Leakage Tests
`C. Plastic Vials
`D. Sorption of Preservatives by Plastic
`E.
`Siliconization of Elastomeric Closures
`F.
`Siliconization of Vials
`
`110
`
`110
`111
`11)
`111
`114
`114
`114
`116
`117
`
`117
`117
`117
`118
`119
`120
`120
`
`ang
`
`
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 21 of 54
`
`

`

`Investigational New Drug (IND) Requirements
`D.
`E. Formulation Development Scale-Up Considerations
`F, Summary
`
`REFERENCES
`
`129
`140
`14]
`
`141
`
`i. FORMULATION REQUIREMENTS
`
`Preformulation Evaluation From the preformulation studies, there should
`be some key parameters that can be ofsignificant ‘aid in the designs of
`experimental formulations. These key parameters are (1) Initial compati-
`bility testing of the active drug substance with some excipients, (2) Effect
`ofstability factors such as temperature,light, packaging components, (3)
`Initial degradation products in the preformulation, and (4) the perform-
`anceof stability assays for the preformulation,
`The following are some of the major considerations to be taken into
`the experimental formulation designs:
`
`A. Characterization, Homogeneity, and Reproducibility
`of the Bulk Active Drug
`
`Characterization, homogeneity, and lot-to-lot reproducibility of the bulk
`active drug substance is of paramount importance. At this stage of dosage
`form development, a great deal of characterization of the bulk has been
`obtained, Regulatory compliance demandsthat the process in place yields
`reproducibility of the active drug substance, as well as whatever impurities
`
`
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 22 of 54
`
`

`

`however, oxidation of methionine, cysteine, and tryptophan can occur, as
`well as other degradative mechanisms (2), The optimal pHis essential for
`better stability.
`Achangeof one pH unit will change the reaction one way oranother.
`The solution pH maybe one ofthe mosteffective ways ta stabilize a liquid
`formulation (3).
`
`C. Stabilizers Used in Proteln Formulations
`
`Degradation ofproteins can be a major biopharmaceutical problem during
`purification, characterization, preformulation, formulation development,
`and possibly during storage. Selective excipients are incorporated into the
`formulation in order to improve the physical and chemicalstability of the
`protein drug substance.
`A variety of molecules have been used as stabilizers, such as surfac-
`tants, amino acids, polyhydric alcohols, fatty acids, proteins, antioxidants,
`reducing agents, and metal ions. Someof the most often used excipients are
`stabilizers, and an explanation for their modeof action has been reported
`in the literature andlisted in Table 1 (4-19).
`
`D. Surfactants
`
`Protein surfactant interactions have also been investigated by other re-
`searchers (20-22). Most recently, the interaction of Tween 20, Tween 40,
`Tween 80, Brij 52, and Brij 92 were studied with recombinant human growth
`hormone and recombinant human interferon gamma for surfactant:protein
`binding stoichiometry.
`
`
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 23 of 54
`
`

`

`Surfactants
`Polysorbate 20 & 80
`Poloxamer 407
`
`Fatty Acids
`Phosphotidy! choline
`Ethanolamine
`Acathyliryptophanate
`Polymers
`Polyathylens glycol (PEG)
`Polyvinylpyrrolidone (PVP) 10, 24, 40
`Polyhydric alcohol
`Sorbitol
`
`Mannitol
`Glycerin
`Sucrose
`Glucose
`Propylene glycal
`Ethylene glycol
`Lactose
`Trehalose
`Antioxidants
`Ascorbic acid
`Cysteine HCI
`Thioglycerol
`Thioglycalic acid
`Thiasorbitol
`Glutathione
`
`Retard aggregation
`Prevent denaturation
`Stabilize cloudiness
`
`Stabilizer
`
`Stabilizer
`
`Pravent aggregation
`
`Pravent denaturation
`Aggregation
`Cryoprotectant
`May act as antioxidant
`
`Strengthen conformational
`Prevent aggregation
`
`Retard oxidation
`
`
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 24 of 54
`
`

`

`This stoichiometric relationship can be applied to protein formula-
`tions to determine stability. Poloxamer 407 (Pluronic F-127) was also tested
`with interleukin-2 and urease resulting in increased stability when the for-
`mulation was subjected to strong agitation (23). Recombinant urokinase
`losses were reduced by the addition of human serum albumin (HSA),
`Tween 80, and Pluronic F-68 (24).
`Interleukin-2 and ribonuclease A, when reconstituted with a variety
`of surfactants, amino acids, sugars and other substances, reduced aggrega-
`tion significantly (25).
`The formation of particulates with a monoclonal antibody wasinhib-
`ited by Tween 80 and recorded by visual and laser light diffraction particu-
`late analysis methods (26).
`Proteins will adsorb at interphases such asliquid/air or liquid/solid.
`When protein molecules are adsorbed they undergo physicochemical
`changes. [Insoluble particles begin to form,eventually resulting in aggrega-
`tion and precipitation and this, in turn, may lead to partial orfull loss of
`bioactivity.
`The addition of surfactants poloxamer 188 (Pluronic 68), or polysor-
`bate to a liquid formulation can prevent or reduce denaturation ofthe pro-
`tein at a liquid/air or liquid/solid interface of the protein in solution (27),
`The most recent literature concerning the use of nonionic surfactants,
`indicate that during bulk storage and usage, hydroperoxides may be formed
`and can degrade manyproteins (28).
`It is for this reason that, when these surfactants are purchased,a client
`must ask the vendorfora certificate of analysis specifying all the tests per-
`formed, including hydroperoxides.
`
`
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1026, p. 25 of 54
`
`

`

`alcohols and carbohydrates. These include mannitol, sorbitol, and glycerol.
`These have been found to stabilize proteins in solution in varying concen-
`tration from 1.0 to 10%. Although the modeof action ofprotein stabiliza-
`tion is not yet clear,it is suggested that the sugar exerts pressure to reduce
`the surface contact between the protein and the solvent (29,30).
`
`G. Antioxidants
`
`Oxidation is one of the major factors in protein degradation. A protein
`solution, from purificatioin to final product for an end user, goes through
`various equipment made of metal, glass, or plastic. At some points during
`the process, the protein solution comes in contact with catalyzing metals
`such as copper, iron, calcium, and manganese, thus inducing the potential
`loss of protein activity. A probablesolutionto this problem will be the in-
`corporation of a compatible antioxidant in the formulation. Some of the
`most often used antioxidants for parenteral preparations are ascorbic acid,
`sodium bisulfite, sodium metabiosulfite, monothio-glycerol, alpha toco-
`pherol, and others. The most frequently used concentrations are in the
`0.1% range and higher. The optimal concentrations are determined by the
`data the formulator obtains from experimental results on a case by case
`evaluation. Nitrogen and argon gasis also used to retard or prevent oxida-
`tive reactions and the gasis used by flooding the head spaceofa vial or
`ampule during sterile filling.
`Antioxidantsfall into one or more of the following categories (31):
`1. Chelating agents, Oxidative reactions catalyzed by metal ions.
`Chelating agents such as EDTA and citric acid de

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