throbber
mm A
`
`fin
`
`as
`
`MYLAN INST. EXHIBIT 1026 PAGE 1
`
`

`

`
`
`
`
`WMYLANIN T. EXHI‘ L I "
`
`
`
`
`
`
`
`
`
`MYLAN INST. EXHIBIT 1026 PAGE 2
`
`

`

`4 P
`
`reformulation Development of
`Parenteral Biopharmaceuticals
`
`JOHN A. BONTEMPO
`
`Biopharmacautieal Product Development. East Brunswick. New Jersey
`
`INTRODUCFION
`A. Considerations of Domestic and lntemational
`Distribution of the Product
`8. Points to Consider for Constituted Versus
`
`Lyophilized Formulations
`C. Unit Dose or Multidose
`
`D. Physicoehemieal Factors to Be Considered for
`Protein Drug Formulations
`
`11.
`
`INITIAL PREFORMULA’I‘ION STUDIES:
`PARAMETERS AND VARIABLES TO BE TESTED
`A.
`Initial Variables to Be Tested
`
`B. Preliminary Analytical Development
`C. Experimental Conditions for the Initial
`Preformulation Studies
`
`III.
`
`MECHANICAL AND PHYSICAL STRESSES
`
`A. Shaking Effect on Protein Solution at the
`Preformulation Level
`
`B. Freeze—Thaw Experiments
`
`92
`
`93
`
`93
`94
`
`94
`
`95
`95
`96
`
`97
`
`99
`
`100
`
`91
`MYLAN INST. EXHIBIT 1026 PAGE 3
`
`MYLAN INST. EXHIBIT 1026 PAGE 3
`
`

`

`92
`
`Bantampo
`
`C. FillingSystems
`D. Stability Evaluation
`
`W. DEGRADATION MECHANISMS
`A. Oxidation
`B. Deamidation
`
`C. Hydrolysis
`D. Racemiaation
`E.
`Isomerization
`
`F. Disulfide Exchange
`G. Beta-Elimination
`
`V.
`
`PHYSICALDEGRADATIONS
`
`A. Covalent Aggregation
`B. Noncovaient
`
`C. Aggregation
`D. Denaturation
`
`E. Precipitation
`F. Adsorption
`
`VI.
`
`SUMMARY
`
`REFERENCES
`
`1.
`
`INTRODUCTION
`
`100
`10]
`
`102
`102
`103
`
`103
`103
`103
`
`103
`104
`
`104
`
`104
`104
`
`104
`105
`
`105
`106
`
`106
`
`107
`
`Preformulation research studies of 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‘
`tant to obtain significant, measurable progress with preformuiations studies
`in a timely manners 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 studies are initiated.
`From an industrial point of view, the preforrnulation 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).
`
`MYLAN INST. EXHIBIT 1026 PAGE 4
`
`MYLAN INST. EXHIBIT 1026 PAGE 4
`
`

`

`Pretormulation Davampmont
`
`93
`
`As previously stated. there must be a strong interdisciplinary collabo-
`ration team to review. identify. and maximize the most productive leads
`toward formulation development. Preformulation studies are short in do—
`ration, two to three months. and some of these are performed undervarying
`stress conditions which will be described later in this chapter. It is important
`to remember that no two proteins are alike and studies designs will vaxy
`case by case.
`Prior to the onset of preforrnulations. the pharmaceutical team must
`review some very important factors which will have an impact on the pre-
`formulation and formulation development.
`
`A. Considerations of Domestic and International
`Distribution ot‘ the Product
`
`Many global joint ventures and partnerships today in the biopharrnaceuti-
`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 andlor
`Iyophilized dosage forms must also be considered. The development of for-
`mulation considerations should be on a worldwide acceptance basis.
`
`B. Points to Consider for Constituted Versus Lyophilized
`Formulations
`
`Some of the key points to be considered for a constituted formulation are:
`
`- A constituted formulation may be 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
`
`Some of the key points to be considered for a lyophilized formulation
`
`are:
`
`- Better stability than a constituted product
`‘- Determination of an optimal iyophilization qrclc
`- 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.
`
`MYLAN INST. EXHIBIT 1026 PAGE 5
`
`
`
`MYLAN INST. EXHIBIT 1026 PAGE 5
`
`

`

`94
`
`Bontempo
`
`' Stability of the reconstituted product
`- Preservative effectiveness (if this is a multidose product)
`- Cost effectiveness. Lyophilization technology is expensive along
`with cost of utilities
`
`At the onset of preformulations studies. it is difficult to predict with
`certainty which of the two types of formulations will have a marketable
`advantage for an extended shelf life. At this early stage of devel0pment,
`there are usually very small amounts of the bulk active drug substance avail-
`able. The formulator must make very efficient use of the active drug sub-
`stance. Nevertheless, both formulations should be considered and started
`
`at the same time. Stability results should be the deciding factor as to which
`form will be selected for further deveIOpment.
`
`G. Unit Dose or Multidose
`
`The decision to select unit dose versus multidose should be based upon
`input from 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 if it is efficacious enough to meet the United States
`Pharmacopeia (USP) requirements. Meeting these requirements. it can
`qualify as a “multidose” {or the US market. However, if the formulation
`is also designated for international market. there are three additional fac-
`tors that must be taken into account. The first 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 [emulation may be 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 escipients
`should also be thoroughly reviewed for international acceptance.
`
`0. Physlcochemlcal Factors to Be Considered for
`Proteln Drug Formulations
`
`Some of the most important physieochemical properties of protein drugs
`required for the development of parenteral preformulations and formula-
`tions are found in Table l.
`
`MYLAN INST. EXHIBIT 1026 PAGE 6
`
`MYLAN INST. EXHIBIT 1026 PAGE 6
`
`

`

`
`
`Preformulatlon Development
`
`95
`
`TABLE 1 Physicochemlcal Factors to Be Considered for Protein Drug
`Formulations
`
`Structure of the protein drug
`lsoelectrlc point
`Molecular weight
`
`Amino acid composition
`
`Disuilide bonds
`
`Spectral propenies
`Agents effecting solubility:
`Detergent
`Salts
`Metal ions
`
`pH
`
`Agents affecting stability
`pH
`Temperature
`Light
`Oxygen
`Metal ions
`Freeze—thaw
`Mechanical stress
`
`Polymorphism
`Stereoisomers
`Filtration media compatibility
`Shear
`
`Surface denaturatEOn
`
`Since this may be an early stage of process development, some of the
`properties listed in Table i may not be available initially, simply because
`there was not enough time or personnel to perform the work.
`
`II.
`
`INITIAL PREFOHMULATION STUDIES: PARAMETERS
`AND VARIABLES TO BE TESTED
`
`The pharmaceutical formulation scientist will consider several factors in the
`preformMation designs. The data received from the ProcesslPurification
`section are reviewed for 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 Tasted
`
`Perhaps 10 or more initial prefonnulation combinations should be consid-
`ered. The initial variables to be tested with various protein concentrations
`are the effects of buffer species. ionic strength, pH range, temperature,
`
`MYLAN INST. EXHIBIT 1026 PAGE 7
`
`MYLAN INST. EXHIBIT 1026 PAGE 7
`
`

`

`96
`
`Bantempo
`
`initial shear, surface denaturation, agitation. and aggregation. Since it has
`been well documented that protein solutions are unstable, some selective
`excipients from various classes of stabilizers should also be included in or-
`der to evaluate stability requirements. Stabilizers will be discussed later in
`this volume.
`
`B. Preliminary Analytical Development
`
`In order to determine the initial stability results. it is necessary to have
`developed, or to have under development, analytical methods to measure
`the potency of the specific formulation under various experimental condi-
`tions. Ultimately some of these analytical methods will be needed to moni-
`tor stability to detect physical and chemical degradation. Regulatory com—
`pliance for the beginning of Phase I Clinical Studies may require at least
`two different methods that are “stability indicators." most often fully vali-
`dated. Dr. Sharma. in Chapter I5, will cover the bioanalytical development.
`
`TABLE 2 Bioanatytlcal Methods to Evaluate initial Pretormulation Development
`
`Method
`
`Bioassay
`
`immunoassay
`
`pH
`SOS-PAGE (Reduced 8t
`nonreduced)
`RP-HPLC
`
`IEF
`
`SE-HPLC
`
`Function
`
`Measure of activity throughout shell lite ot a
`formulation
`
`Purity assessment and measures concentra-
`tion of a panicalar molecular species
`Chemical stability
`Separation by molecular weight. characterizes
`tion of proteins and purity
`Estimation of purity, identity. and stability of
`proteins. Separation and analysis oi pro
`tein digests.
`Determines the isoelectric point of the protein
`and detects modifications at the protein
`Method of separating molecules according
`to their molecular size and purity
`determination
`
`N-terminal sequencing
`UV
`
`Elucidation of the C-ten'ntnus. identity
`Detection at individual component. concentra«
`tion. and aggregation
`Detects secondary and tertiary conformation
`CD (circular dichroism)
`
`in the UV region and quantitatas various structures
`
`MYLAN INST. EXHIBIT 1026 PAGE 8
`
`MYLAN INST. EXHIBIT 1026 PAGE 8
`
`

`

`Preformulalion Development
`
`97
`
`However. some of the following bioanalytical methods listed in Table 2 can
`be applied to begin initial evaluation of the preformulations degradation
`(if any) under test.
`
`C. Experimental Conditions for the initial
`Pretormulatlon Studies
`
`Protein Concentration
`
`Protein drugs are extremely potent: therefore, very low concentrations are
`required for their respective therapeutic levels. Dosage forms development
`need to be tested at varying ranges of activity. The respective concentra-
`tions may range from nanograms to micrograms to milligrams and the con-
`centration will vary from protein to protein.
`
`pH Flange
`
`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 the solubility and stability of the for-
`mulation. pH control in pharmaceutical dosage forms is very critical (1).
`The proper pH selection is one of the key factors in deveIOping 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 bu ffers 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 0.1 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 Exciplents to Be Considered
`
`As it was stated previously. the objective of a prefonnulation study is to
`select potentially compatible excipients in order to hasten the development
`of stable formulations. Based upon protein chemical and physical instabil-
`
`MYLAN INST. EXHIBIT 1026 PAGE 9
`
`MYLAN INST. EXHIBIT 1026 PAGE 9
`
`

`

`93
`
`Bontempo
`
`ity, it is highly probable that some excipients may be included in the prefor-
`mulation. In so doing, the designs of formulations to follow can be more
`specific in selecting the proper excipient(s) to control specific degradation
`pathways.
`
`Cholating Agents
`
`The crude bulk protein drug during fermentation and purification steps has
`passed through and contacted surfaces such as metal, plastic, and glass. If
`metal ions are present in the liquid bulk active. it is highly recommended
`to use a chelating agent such as cthyienediamine tetraacidic acid (EDTA)
`to effectively bind trace metals such as copper, iron, calcium. manganese
`and others. A recommended dose of (EDTA) would be about 0.01 to
`0.05%.
`
`Antioxidants
`
`Since oxidation is one of the major facrors 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 andior nitrogen gas can also be used to flood the head space
`of a vial or ampule during sterile filling to prevent or retard oxidation. A
`recommended antioxidant dose would be about 0.05 to 0.1%.
`Preservatives
`
`If a multidose formulation is required, an antimicrobial agent, called pre-
`servative, is required to be incorporated into the formulation. "the preserv-
`ative effectiveness must comply with the USP requirements to 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»
`coho! (1.0 to 3.0%). Additional details are provided in Chapter 5.
`
`Surfactants
`
`J udicious selection of surfactants can result in the prevention of aggrega-
`tion and stabilization of proteins (2). Polysorbate 80, poloxarner 188, and
`pluronic 68 have been used in injectable formulation. The purity of the
`surfactant may have an impact on the chemicai stabiiity of the prefonnula-
`tion. Peroxide residues in the surfacrant have been implicated in oxidations
`
`of protein.
`
`MYLAN INST. EXHIBIT 1026 PAGE 10
`
`MYLAN INST. EXHIBIT 1026 PAGE 10
`
`

`

`Preformulation Development
`
`99
`
`Glass Vial Selection
`
`Type 1 glass. as classified in the USP. should be used. The selection of a
`glass vial must also be taken into consideration when dealing with adsorp-
`tive properties of the respective protein. Adsorption of proteins will be
`treated later in this volume.
`
`Rubber Stopper Selection
`
`In studying both the liquid and reconstituted protein drugs. the selection of
`a rubber stepper is also of major concern considering the potential reactiv—
`ity of a protein solution with a rubber stopper. as well as the reactivity of
`the reconstituted lyophilized solution during storage conditions prior to
`use. For parenteral formulations, the biopharrnaceutical industry has been
`using rubber stoppers with a veryr thin film of various inert polymers in order
`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
`Chapter 8.
`
`Membrane Filter Selection
`
`Membrane filtration is the most often used technique to sterilize protein
`solutions. The chemical nature of the filter and the pH of the protein solu~
`tion are the two most important factors affecting the protein adsorption (3).
`However. there are other issues that require consideration. The female-
`tion scientist must be aware of particles or fibers released during the filtra-
`tion, the potential extractables that may about, the potential toxicity of the
`filter media and the product compatibility with the membrane. Of all 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 amoants of protein binding and
`deactivation,
`
`Ill. MECHANICAL AND PHYSICAL STRESSES
`
`A. Shaking Ettect on Protein Solution at the
`Pretormulation Level
`
`Some of the various physical modes of vialed 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-
`ceiving. Simulation of some of the functions described above need to be
`
`MYLAN INST. EXHIBIT 1026 PAGE 11
`
`MYLAN INST. EXHIBIT 1026 PAGE 11
`
`

`

`mo
`
`Bantempo
`
`performed by doing some shaking experiments to determine their affect on
`aggregation induction.
`Some of these preformulation experiments should also contain vary-
`ing concentrations of surfactant(s) with appropriate controls. These short
`and inexpensive experiments can be set up on reciprocal shakers for periods
`of time from 1 to 6 to 24 hours. shaking from 10, 30, and 60 reciprocal
`strokes per minute. Reciprocal strokes disrupt and break up the flow of the
`liquid, while rotary strokes move the liquid circularly without breakup.
`These studies are intended to determine precipitation and aggregation ef-
`fects. Detailed aggregation experiments and results will be described later
`in these chapters.
`
`B. Freeze—Thaw Experiments
`
`These experiments will also be described in later chapters and will be part
`of Chapter 5. These experiments require a fair amount of active drug sub-
`stance as well as a fair amount of work. At this point of development there
`may not be enough active drug substance available.
`
`C. Filling Systems
`
`or all the filling types employed to dispense liquid, such as time«pressure.
`piston, and rotary pump, the rolling diaphragm metering pump is the one
`of choice for filling biOphannaceutical solutions. The internal pans of the
`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 pump is that it eliminates
`the principal cause of particulate generation which is most often induced
`by parts coming together creating shedding of microscopic particles.
`There are three other important parameters to control while dispens-
`ing protein solutions. (1) The speed at which liquid is 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 Ill-ml. vial per single
`filling head. If a large number of vials need to be filled. this filling system
`can accommodate variable numbers of filling heads. thus allowing it to fill
`a large number of vials. Filling at a faster rate will result in protein precipi-
`tation and aggregation. (2) The inner diameter of the filling cannula should
`not be so very small as to induce shearing and aggregation of the protein
`solution. (3) The tip 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 of the vial. This will result in a gentle flow
`
`MYLAN INST. EXHIBIT 1026 PAGE 12
`
`MYLAN INST. EXHIBIT 1026 PAGE 12
`
`

`

`Prelormulatlon Development
`
`tat
`
`touching the inner wall of the vial when the cannula enters the vial and
`delivers the required amount of fluid. The proper bend on the tip of the
`cannula may also eliminate aggregation andlor shearing of the protein
`solution.
`
`0. Stability Evaluation
`
`The development of acceptable analytical methods while isolation, charac-
`terization, and purification of a bulk active drug substance are going an is
`very important. It can be an aid in generating semiquantitative and quanti-
`tative measurements of the active bulk drug at various stages of the process.
`Significant marketing advantages in this competitive pharmaceutical
`market wOuld be to achieve a longer shelf life of the product and storage
`temperature at room temperature. Today the lyophilized protein drug of-
`fers refrigerated temperature storage between 2 and 8°C.
`The present storage conditions set 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
`0 Excessive heat. Temperature exceeding 40°C
`
`Table 3 summarizes the initial guideline time points and tempera-
`tures that preforrnulation solutions should be exposed to. The results from
`the prefonnulations will allow the review team to determine directions to
`manipulate the excipients to obtain better stability.
`
`TABLE 3 Guideline tor Pretormulation Stability Studies
`
`Temperatures
`
`Timepolnt
`
`Frozen controls (~80 and —20'C}
`Refrigerated (24"0)
`
`Intermediate (20. 30. 37°C}
`
`High temperature {40. 45. 50°C)
`
`Reference control sample as needed
`T = O. B. 12. 24 8. 48 weeks
`Continue it stable
`
`T = 0. 4, 8, 12. 18. 24 weeks
`Continue it stable
`
`T = 0. 1. 2. 4. 8, 12 weeks
`Continue it stable
`
`
`
`MYLAN INST. EXHIBIT 1026 PAGE 13
`
`MYLAN INST. EXHIBIT 1026 PAGE 13
`
`

`

`102
`
`Bantampo
`
`W. DEGRADATION MECHANISMS
`
`To predict degradation pathways of new biophannaceuticals is very diff-
`cuit. Depending on the stress conditions, each protein may react differently
`than another protein. As stated previously, the objectives of preformulation
`are to evaluate stress conditions such as pH, temperatures, and buffers and
`begin evaluation of some initial breakdown products. At this particular
`stage of development, it is necessary to have some analytical method(s) with
`some reliability to detect initial degradation. it is difficult to begin evalu-
`ation of degradation products without the reliability of these preliminary
`assay methods.
`The purpose of initial preformuiation studies is to begin understand-
`ing of protein instability via chemical and physical stress conditions (5). in
`order to stabilize potential useful pharmaceutical products, it is important
`to understand how proteins degrade. how they are affected by the compo-
`sition of the formulation. and the effects of stability 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 chemical entities. One or more of the 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
`biophannaceuticals. Under oxidative stress and in the presence of trace
`metals, amino acids sach as methionine (Met) can be oxidized to methion—
`ine sulfoxide, cysteine (Qrs) to cysteine disultide. as well as tryptophane
`(Try) and histidine (His) via other modifications.
`Oxidation can be controlled or minimized by (l) the addition of an-
`tioxidants. (2) having strict 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-performance isoelectric chroma-
`tography (HP-l EC). peptide mapping. amino acids analysis. and mass spec-
`
`MYLAN INST. EXHIBIT 1026 PAGE 14
`
`MYLAN INST. EXHIBIT 1026 PAGE 14
`
`

`

`Prelormulatlon Development
`
`103
`
`trometry (MS) (6). in terms of total protein concentration. ultraviolet Spec-
`trophotometry is the method most often used (1').
`
`B. Deamldation
`
`Deamidation is another more frequent degradation mechanism affecting
`pharmaceutical protein stability. Dcarnidation is the hydrolysis of the side
`chains amide on asparagine (Asp) and glutamine (Gln) to form Asp audior
`Gln residues. Extensive reports have elucidated mechanisms of deamida-
`tion reactions (8).
`Deamidation can be detected by isoelectric focusing, ion exchange
`chromatography. tryptic mapping and HPLC (9).
`
`C. Hydrolysis
`
`Hydrolysis is another most likely cause of degradation of proteins. It in-
`volves a peptide (amide) bond in the protein backbone (5). The most influ-
`ential factor affecting the hydrolytic rate is the solution pH.
`
`D. Racemlzation
`
`Proteins may also degrade via other modifications (10) such as racemiza-
`tion. This mechanism involves the removal of the alpha proton from an
`amino acid in a peptide to yield a negatively charged planar carbonion. 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.
`
`lsomerizatloa
`
`Protein degradation is also induced by isomerization. Hydrolysis of cyclic
`amides of 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. Disuh‘ide Exchange
`
`Disulfide exchange may result from a degradation other than covalent
`modification. These reactions may include the disulfide exchange of cys-
`teine. This reaction is base, catalyzed and promoted by thin] antioxidants
`(13).
`
`MYLAN INST. EXHIBIT 1026 PAGE 15
`
`MYLAN INST. EXHIBIT 1026 PAGE 15
`
`

`

`194
`
`Bootempo
`
`DiSulfide exchange can occur in misfolded conformers due to incor-
`rect intramolecular disulfide bonds (14).
`
`G. Beta-Elimination
`
`Another degradation residue can be the beta-elimination of ser, thr, cys, lys
`and phe residues. These reactions are accelerated by basic pH. tempera
`ture, and the presence of metal ions (16).
`
`V. PHYSICAL DEGRADATIONS
`
`Aggregation
`
`Protein aggregation can be of a covalent or noncovalem nature (11.18).
`
`A. Covalent Aggregation
`
`This pathway involves modification of the chemical structures resulting in
`newr chemical structures and may include reactions. such as oxidation. de-
`amidation, proteolysis. disulfide interchanges. racemization, and others.
`
`a. Noncova lent
`
`This instability may be induced by agitation. Shea r. precipitation. and ad-
`sorption to surfaces.
`
`C. Aggregation
`
`Protein aggregation derived from either physical or chemical inactivation,
`is presently a major biopharmaceutical problem (Ii—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 speetrophotometric methods, or both. can be of
`significant guidance to formulation scientists in selecting compatible excipi—
`ems to minimize andtor prevent its formation in the experimental formu—
`lation.
`Formation of aggregation can begin by the formation of initial parti-
`cles from protein molecules via the Brewnian movement. This is followed
`by collision of these molecules and aggregates ofvarying sizes can be formed.
`These aggregates can be generated by shear or collisional forces (22).
`Detection and measurements of aggregations can be performed by a
`number oftechniques. Visual observations, light scattering. polyacrylamide
`gel electrophoresis. UV, spectrophotometry. laser light diffraction particu—
`
`MYLAN INST. EXHIBIT 1026 PAGE 16
`
`MYLAN INST. EXHIBIT 1026 PAGE 16
`
`

`

`Pretormulation Development
`
`105
`
`late analysis. fluorescence spectra and differential scanning colorimetry
`(DSC), RP—HPLC. and SE-HPLC (7.14). Conformational changes can also
`lead to aggregations and can be measured by DSC (23).
`A formulation scientist should focus on some important observations
`that need to be made to answer some potential problems on aggregation.
`
`0 Determination of initial approximate number of aggregates
`- Determination of approximate size and distribution of aggregates
`. Do the aggregates increase in size and number over time?
`- Do the aggregates affect the efficacy of the proteins?
`- What is the effect of aggregation on the long—term storage of the
`potential marketable product?
`
`D. Denaturation
`
`Denaturation of proteins can be the result ofseveral 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 conformational side 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.
`Then-ital denaturation of proteins is of great interest to the formulation
`scientist. Thermal probes offer tools to study protein structure and stability
`that ultimately can be of significant use to stabilize protein drug formula-
`tions. Modifications of protein thermal effects have been reviewed (8).
`The ability of the protein to retold 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. there is very likely inactivation of the protein attribn
`utable IO shearing effect.
`
`E. Precipitation
`
`Precipitation in formulations can occur by a variety of mechanisms such 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 reach a critical mass.
`precipitate out of solution and are clearly visible.
`
`MYLAN INST. EXHIBIT 1026 PAGE 17
`
`MYLAN INST. EXHIBIT 1026 PAGE 17
`
`

`

`toe
`
`Boniempo
`
`From a biopharrnaceutical formulation point of view. precipitation
`can occur in membrane filters, filtration equipment. pumps. and tubing and
`loss of activity is very often recorded.
`Eventually. as the aggregation mechanisms are controlled and pre-
`vented. precipitation is essentially reduced or avoided. Details on the func-
`tions of stabilizers are discussed later in this volume.
`
`F. Adsorption
`
`Some of the most prevalent. ubiquitous factors of deactivation (30,31.
`32) that the protein biochemists and formulation scientists face. are the
`surface areas interactions from the purification. formulations. and stability
`stages.
`
`Essentially. at each point that the protein solution has encountered
`air during mixing (process). filtration (process). and air in the process steps.
`a significant surface area has been encountered to yield interphases.
`During the actual final manufacturing of vials. ampules. syringes.
`catheters. pumps. and their respective storage conditions. the proteins
`could be adsorbed at 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-
`ten’sties. they have a high affinity to adsorption at the air—liquid and solid—
`liquid interphase. Hydrophobic and hydrophilic interactions which are con-
`centration dependent. determine the extent a nd the rate ofadsorption. The
`adsorption effect on the protein is the unfol

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