throbber
B i o P r o c e s s Technical
`
`Stability Considerations for
`Biopharmaceuticals, Part 1
`Overview of Protein and Peptide Degradation Pathways
`
`Jalpa Patel, Ruchi Kothari, Rashbehari Tunga, Nadine M. Ritter, and Binita S. Tunga
`
`T o ensure product safety and
`
`efficacy, protein therapeutics
`must meet defined quality
`characteristics immediately after
`manufacture as well at the end of their
`designated shelf lives. Many physical
`and chemical factors can affect the
`quality and stability of
`biopharmaceutical products,
`particularly after long-term storage in a
`container–closure system likely to be
`subject to variations in temperature,
`light, and agitation with shipping and
`handling. Compared with traditional
`chemical pharmaceuticals, proteins are
`considerably larger molecular entities
`with inherent physiochemical
`complexities, from their primary amino
`acid sequences through higher-order
`secondary and tertiary structures —
`and in some cases, quaternary elements
`such as subunit associations (1).
`Many proteins are glycosylated, and
`some have other posttranslational
`modifications such as
`phosphorylation, which also affects
`
`Product Focus: PePtides and Proteins
`
`Process Focus: Formulation/stability
`
`Who should read: analytical and
`Formulation develoPment, Process
`develoPment, Qa/Qc and regulatory
`aFFairs
`
`KeyWords: Forced degradation,
`Protein degradation, oxidation,
`disulFide scrambling, deamidation,
`aggregation, hydrolysis
`
`level: intermediate
`
`20 BioProcess International
`
`January 2011
`
`their potential degradation pathways
`as well as the kinetics of their
`degradation. Proteins are typically
`sensitive to slight changes in solution
`chemistry. They remain
`compositionally and conformationally
`stable only within a relatively narrow
`range of pH and osmolarity, and many
`require additionally supportive
`formulation components to remain in
`solution, particularly over time (2).
`Even lyophilized protein products
`experience degradation (3, 4).
`Advances in analytical chemistry
`have identified many degradation
`pathways that can occur in
`recombinant protein therapeutics over
`time. These pathways generate either
`chemical or physical instability.
`Chemical instability refers to the
`formation or destruction of covalent
`bonds within a polypeptide or protein
`structures. Chemical modifications of
`protein include oxidation, deamidation,
`reduction, and hydrolysis (5).
`Unfolding, dissociation, denaturation,
`aggregation, and precipitation are
`known as conformational or physical
`instabilities (5). In some cases, protein
`degradation pathways are synergistic:
`A chemical event may trigger a
`physical event, such as when oxidation
`is followed by aggregation.
`Here, we present several protein
`degradation events: oxidation,
`photodegradation, disulfide
`scrambling, deamidation, aggregation,
`precipitation, dissociation, and
`fragmentation. We illustrate the
`biochemistry of each, showing
`potential means of induction and
`
`www.photos.com
`
`suggesting formulation considerations
`for prevention. In an upcoming issue,
`Part 2 will conclude with methods of
`detection and strategies for validation
`of stability-indicating methods. Our
`objective is to provide an introduction
`(or refresher) to the major degradation
`pathways of protein products, with
`references for each. Readers are
`encouraged to consult those references
`for expanded details on the basic
`biochemistry of each pathway, case
`studies describing experiments with
`specific proteins, and further
`information on formulation
`development strategies.
`oxidation, Photodegradation,
`and disulFide scramBling
`Proteins and peptides are susceptible
`to oxidative damage through reaction
`of certain amino acids with oxygen
`radicals present in their environment.
`Methionine, cysteine, histidine,
`tryptophan, and tyrosine are most
`susceptible to oxidation: Met and Cys
`because of their sulfur atoms and His,
`
`Eton Ex. 1031
`1 of 10
`
`

`

`Figure 1: oxidation of methionine (top) and cysteine (bottom) *
`
`COOH
`
`H3N+
`
`C
`
`H
`
`CH2
`
`CH2
`
`S
`
`Oxidant
`
`COOH
`
`H3N+
`
`C
`
`H
`
`CH2
`
`CH2
`
`S O
`
`COOH
`
`H3N+
`
`C
`
`H
`
`CH2
`
`CH2
`O S O
`
`Oxidant
`
`CH3
`Methionine
`
`CH3
`Methionine Sulfoxide
`
`CH3
`Methionine Sulfone
`
`Oxidant
`
`H
`
`COOH
`H3N+ C
`CH2
`CH2
`S O
`OH
`Sul(cid:30)nic Acid
`pKa ~ 1.9
`
`H
`
`COOH
`H3N+ C
`CH2
`CH2
`O S O
`OH
`Cysteic Acid
`pKa ~ (cid:31)5.7
`
`COOH
`H3N+ C
`CH2
`S
`
`H
`
`Cystine
`
`COOH
`H3N+ C
`CH2
`S
`
`H
`
`n t
`
`a
`
`x i d
`
`O
`
`Oxidant
`
`H3N+
`
`COOH
`
`H
`
`C
`CH2
`
`Oxidant
`
`S O
`
`H
`Sulfenic Acid
`pKa ~ 5.7
`
`COOH
`
`H3N+
`
`H
`
`C
`CH2
`SH
`Cysteine
`pKa ~ 8.5
`
`* Griffiths SW. Oxidation of the Sulfur-containing amino acids in Recombinant human α1-antitrypsin (thesis).
`Massachusetts Institute of Technology: Cambridge, MA, 2002.
`
`environment, cysteine oxidation
`involves nucleophilic attack of thiolate
`ions on disulfide bonds, generating
`new disulfide bonds and different
`thiolate ions. The new thiolate can
`then react with another disulfide bond
`to form cysteine.
`Such intermolecular disulfide links
`formed by protein degradation
`accumulate mispaired disulfide bonds
`and scrambled disulfide bridges, which
`can alter protein conformation and
`subunit associations (6). Cysteine
`residues may also undergo spontaneous
`oxidation to form molecular byproducts
`— sulfinic acid and cysteic acid — in
`the presence of metal ions or nearby
`thiol groups (11). For example, human
`fibroblast growth factor (FGF-1)
`exhibits copper-catalyzed oxidation
`that can create homodimers (12).
`Spatial orientation of thiol groups in
`proteins plays an important role in
`cysteine oxidation. The rate of
`oxidation is inversely proportional to
`the distance between those thiol groups
`(13). This can eventually lead to
`formation of large oligomers or
`nonfunctional monomers, as with basic
`fibroblast growth factor (bFGF), which
`contains three cysteines that are easily
`oxidized and form intermolecular or
`intramolecular disulfide bonds (13).
`
`This oxidation often induces
`conformational modifications of the
`protein because cysteine disulfide
`increases side-chain volume in the
`protein’s interior and leads to
`unfavorable van der Waals interactions
`that maintain the original structure (13).
`Histidine residues are highly
`sensitive to oxidation through reaction
`with their imidazole rings, which can
`subsequently generate additional
`hydroxyl species (6). Oxidized
`histidine can yield asparagine/
`aspartate and 2-oxo-histidine (2-O-
`His) as degradation products during
`light and/or metal oxidation (6, 14). It
`may be a transient moiety because it
`can trigger protein aggregation and
`precipitation, which can obscure
`isolation of 2-O-His as an individual
`degradant (15). Oxidation of tyrosine
`may result in covalent aggregation
`through formation of bityrosine (16).
`Spatial factors may also affect tyrosine
`and histidine oxidation. Adjacent
`negatively charged amino acids
`accelerate tyrosine oxidation because
`they have high affinity to metal ions,
`whereas positively charged amino-acid
`residues disfavor the reaction (17, 18). If
`an adjacent amino acid is bulky, it may
`mask oxidation of neighboring amino
`acids and prevent them from getting
`
`Trp, and Tyr because of their aromatic
`rings (6). Oxidation can alter a
`protein’s physiochemical
`characteristics (e.g., folding and
`subunit association) and lead to
`aggregation or fragmentation. It can
`also induce potential negative effects
`on potency and immunogenicity
`depending on the position of oxidized
`amino acids in a protein relative to its
`functional or epitope-like domain(s).
`For example, parathyroid hormone
`biological activity was differentially
`affected by a single oxidation of either
`Met-8 or Met-18 and double oxidation
`(Met-8 with Met-18) when each
`specie was isolated and testing using
`in vitro bioassays (7, 8). Similarly,
`oxidation of Met-36 and Met-48 in
`human stem cell factor (huSCF)
`derived from Escherichia coli decreased
`its potency 40% and 60% (respectively)
`while increasing the dissociation rate
`constant of SCF dimer by two- to
`threefold, which suggests an effect on
`subunit binding and tertiary structure
`(9). In other cases, oxidation had no
`measurable impact on protein potency
`even when substantial structural
`changes were seen. For example,
`oxidized Met-111 in interferon α-2b
`affected the molecule’s primary,
`secondary, and tertiary structure and
`prevented site-specific epitope
`recognition by a monoclonal antibody
`(MAb) without altering in vitro
`biological activity (10).
`Mechanism and Factors Involved:
`Figure 1 shows biochemical pathways
`for oxidation of methionine and
`cysteine residues. Methionine is
`oxidized by atmospheric oxygen and
`oxygen radicals in solution to form
`methionine sulfoxide and methionine
`sulfone. Both species are larger and
`more polar than nonoxidized
`methionine, which can alter protein
`folding and structural stability (11).
`The rate of methionine oxidation in
`recombinant human parathyroid
`hormone (rHu-PTH) by hydrogen
`peroxide is enhanced at alkaline pH (8).
`Cysteine oxidation is also more
`prevalent at alkaline pH, which
`deprotonates thiol groups. Oxidation
`of cysteine induces disulfide bond
`breakage in a reducing environment
`(Figure 1, bottom). In such an
`
`22 BioProcess International
`
`January 2011
`
`Eton Ex. 1031
`2 of 10
`
`

`

`Figure 3: hydrolysis of a peptide bond
`
`N H
`
`H2O
`
`O
`
`H
`
`OH
`
`N H
`
`O
`
`presence of denaturing/unfolding
`reagents in solution can increase the
`extent of protein oxidation. Excipients
`such as polyols and sugars involved in
`stabilizing protein structure can
`decrease the rate of oxidation (6).
`Oxidative modification depends on
`intrinsic structural features such as
`buried and exposed amino acids. In the
`case of human growth hormone, Met-
`14 and Met-125 are readily oxidized by
`H2O2 because they are exposed to the
`surface of the protein, whereas Met-170
`in its buried position can be oxidized
`only when the molecule is unfolded
`(21). Also, atmospheric oxygen can
`cause protein oxidation over time.
`Headspace oxygen contributed to the
`loss of 50% potency by four months in
`multidose vials of tuberculin purified
`protein (TPP) (25).
`Oxidation can be induced during
`protein processing and storage by
`peroxide contamination resulting from
`polysorbates and polyethylene glycols
`(PEGs), both commonly used as
`pharmaceutical excipients. A
`correlation has been observed between
`the peroxide content in Tween-80 and
`the degree of oxidation in rhG-CSF,
`and peroxide-induced oxidation
`appeared more severe than that from
`atmospheric oxygen (26). Peroxide can
`also leach from plastic or elastomeric
`materials used in primary packaging
`container–closure systems, including
`prefilled syringes (27, 28).
`Preventive Measures: One
`molecular engineering strategy for
`minimizing oxidative degradation is to
`replace oxygen-labile amino acids with
`oxygen-resistant ones if a protein’s
`nature permits. In therapeutic
`Interferon beta (IFN-β), cysteine at
`position 17 was replaced by serine,
`because the former loses antiviral
`activity during storage to oxidation
`and disulfide scrambling (29).
`Substitution of methionine of
`epidermal growth factor (EGF) with a
`nonnaturally occurring norleucine also
`prevented oxidative degradation (30).
`
`Ala
`
`O C
`
`CH2
`
`Figure 2: Deamidation pathway of asparagine *
`O
`C
`
`Val Tyr Pro
`
`CH2
`CH
`
`NH2
`NH
`
`?
`
`C
`O
`L-Asn Peptide
`
`O
`C
`
`CH2
`CH
`
`O
`NH
`
`O
`C
`
`CH2
`
`C
`O
`L-Normal Peptide
`?
`D-Normal Peptide
`
`O
`C
`
`C
`
`CH2
`CH
`
`O
`
`C
`
`N
`
`CH2
`
`O
`L-Imide Peptide
`
`D-Imide Peptide
`
`O
`C
`
`CH2
`CH
`
`O
`C
`
`NH
`
`CH2
`
`O
`
`C
`O
`L-Iso Peptide
`
`?
`D-Iso Peptide
`
`* Deamidation, Isomerization, and Racemization at Asparaginyl and Aspartyl Residues in Peptides:
`Succinimide-Linked Reactions that Contribute to Protein Degradation. J. Biolog. chem. 262(2) 1987: 785–794.
`
`oxidized. It has been observed that
`histidine present in a sequence
`markedly increases both the peptide
`oxidation rate and methionine
`sulfoxide production. The strong
`metal binding affinity of the
`imidazole ring on the histidine side
`chain brings oxidizing species close to
`the substrate methionine (6).
`Light Degradation: Photooxidation
`can change the primary, secondary,
`and tertiary structures of proteins and
`lead to differences in long-term
`stability, bioactivity, or
`immunogenicity (19). Exposure to
`light can trigger a chain of
`biochemical events that continue to
`affect a protein even after the light
`source is turned off. These effects
`depend on the amount of energy
`imparted to a protein and the presence
`of environmental oxygen.
`Photooxidation is initiated when a
`compound absorbs a certain
`wavelength of light, which provides
`energy to raise the molecule to an
`excited state. The excited molecule
`can then transfer that energy to
`molecular oxygen, converting it to
`reactive singlet oxygen atoms. This is
`how tryptophan, histidine, and
`tyrosine can be modified under light
`in the presence of O2 (6). Tyrosine
`photooxidation can produce mono-,
`di-, tri-, and tetrahydroxyl tyrosine as
`byproducts (18). Aggregation is
`observed in some proteins due to
`
`cross-linking between oxidized
`tyrosine residues (20). Photooxidation
`reaction is predominately site specific
`(21). For example, in human growth
`hormone treated with intense light,
`oxidation is carried out predominantly
`at histidine-21 (22). In addition, the
`peptide backbone is also a
`photodegradation target (23).
`Alternatively, the energized protein
`itself can react directly with another
`protein molecule in a photosensitized
`manner, typically via methionine and
`tryptophan residues at low pH (6).
`Excipients and leachables can
`synergistically affect the oxidation
`(and hence, degradation) of a protein.
`Formulation components influence the
`rate of photooxidation in some
`instances: e.g., phosphate buffer
`accelerates the rate of methionine
`degradation more than other buffer
`systems do (22). Metal-ion–catalyzed
`oxidation depends on concentration of
`metal ions in the environment. The
`presence of 0.15-ppm chloride salts of
`Fe3+, Ca2+, Cu2+, Mg2+, or Zn2+ does
`not affect the rate of oxidation for
`human insulin-like growth factor-1,
`but when the metal concentration
`increased to 1 ppm, a significant
`increase in oxidation was observed
`(23). Oxidation can be exacerbated in
`the presence of a reducing agent such
`as ascorbate. Ascorbic acid increased
`oxidation of human ciliary
`neurotrophic factor (24). Also, the
`
`24 BioProcess International
`
`January 2011
`
`Eton Ex. 1031
`3 of 10
`
`

`

`
`
`Removal of headspace oxygen by
`degassing may be effective for
`preventing oxidation in some cases.
`Filling steps are carried out under
`nitrogen pressure, and vial headspace
`oxygen is replaced with an inert gas
`such as nitrogen to prevent oxidation
`(21, 25). With some oxidation-sensitive
`proteins, processing is carried out in
`the presence of an inert gas such as
`nitrogen or argon. For multidose drug
`preparations, use of cartridges with
`negligible headspace overcomes
`oxidation and related consequences (25).
`Care must be exercised when
`container–closure changes are
`considered. Many such changes for
`protein therapeutics (from vials to
`prefilled syringes or prefilled syringes
`to pen devices, for example) are
`considered to enhance patient
`convenience and ease of use. But
`historical experience with container–
`closure systems based only on chemical
`pharmaceuticals should be reevaluated
`when the same materials are used with
`protein-based products because of
`potential for unexpected, unique
`impacts on protein degradation.
`Controlling or enhancing factors
`such as pH, temperature, light
`exposure, and buffer composition can
`also mitigate the effects of oxidation
`by affecting a protein’s environment.
`Cysteine oxidation often can be
`controlled by maintaining the correct
`redox potential of a protein
`formulation, such as with addition of
`thioredoxin and glutathione.
`Antioxidants and metal chelating
`agents also can be used to prevent
`oxidation in protein formulations.
`Antioxidants are chemical “sacrificial
`targets” with a strong tendency to
`oxidize, consuming chemical species
`that promote oxidation. Scavengers
`such as l-methionine and ascorbic
`acid are used for this purpose in
`biotherapeutic formulations (31). In
`the absence of metal ions, cysteine as a
`free amino acid may act as an effective
`antioxidant. As chelating agents,
`EDTA and citrate might form
`complexes with transition metal ions
`and inhibit metal-catalyzed, site-
`specific oxidation (6). Addition of
`sugars and polyols may also prevent
`metal-catalyzed oxidation because of
`
`their complexion with the metal ions.
`Protective effects of glucose, mannitol,
`glycerol, and ethylene glycol against
`metal-catalyzed oxidation has been
`observed with human relaxin (32).
`Physical protection from UV/white
`light exposure with either a primary or
`secondary packaging system may be
`necessary to protect light-labile
`proteins from photooxidation.
`deamidation
`With many recombinant proteins,
`changes in peptide and protein
`structure are observed through the
`nonenzymatic deamidation of
`glutamine and asparagine residues.
`This can have varying effects on their
`physiochemical and functional stability
`(33, 34). It has been observed that
`deamidation of hGH alters proteolytic
`cleavage of the human growth
`hormone (33). And it was reported that
`deamidation of IFN-beta increased its
`biological activity (35). It has been
`determined that deamidation of
`peptide growth-hormone–releasing
`factor leading to aspartyl and iso-
`aspartyl forms reduces the bioactivity
`by 25- and 500-fold, respectively, as
`compared with the native peptide (36).
`Deamidation at an Asn-Gly site in
`hemoglobin changes its affinity to
`oxygen (37). Asparagine deamidation
`perturbs antigen presentation on Class
`II major histocompatibility complex
`molecules (38). It was reported that
`isomerization of Asp 11 in human
`epidermal growth factor led to a
`fivefold reduction in its mitogenic
`activity (39). And deamidation at two
`Asn-Gly sequences in triose-phosphate
`isomerase resulted in subunit
`dissociation (40).
`Mechanism and Factors Involved:
`Deamidation is a chemical reaction in
`which an amide functional group is
`removed from an amino acid.
`Consequences include isomerization,
`racemization, and truncation of
`proteins. Figure 2 shows the
`mechanism of asparagine degradation
`by deamidation.
`Isomerization: Isomerization of
`aspartate to isoaspartate residues in a
`protein solution is the most commonly
`observed outcome of nonenzymatic
`deamidation (41, 42).
`
`Say hello to
`self-filling
`systems
`
`Say goodbye to unreliable
`manual intervention
`
`Allegro™ 3D biocontainers require no
`manual intervention during filling and
`draining, providing safer, more efficient
`processing of production scale volumes,
`combined with superior product recovery
`and performance.
`
`w: www.pall.com/allegro
`e: allegro@pall.com
`
`Providing Flexible Solutions
`
`© 2010 Pall Corporation. Pall, , and Allegro are trademarks of Pall
`Corporation. The ® indicates a trademark registered in the USA. GN10.3623
`
`BPI0101-QP-Pall.indd 1
`
`12/13/10 12:53:03 PM
`
`Eton Ex. 1031
`4 of 10
`
`

`

`Racemization: Succinimide
`intermediates formed during
`asparagine deamidation are highly
`prone to racemization and convert to
`d-asparagine residues (41, 42).
`Racemization of other amino acids,
`except glycine, is observed at alkaline
`pH (42).
`Truncation: At low pH levels,
`peptides and proteins experience
`deprotonation of the amide group on
`their asparagine side chain, followed
`by nucleophilic attack by the nitrogen
`atom of the amide anion on the
`peptide carbonyl carbon of the
`asparagine residue (43). This generates
`peptide chain cleavage by forming a
`succinimide peptide fragment.
`Subsequent hydrolysis of the
`succinimide ring can yield asparaginyl
`and β-asparaginyl peptides.
`Mechanisms for the aspartate–
`isoaspartate deamidation and
`isomerization reactions are similar
`because they both proceed through an
`intramolecular cyclic imide
`intermediate (44). Deamidation rates
`for individual amide residues depends
`on their primary sequence and three-
`dimensional (3D) structure as well as
`solution properties such as pH,
`temperature, ionic strength, and buffer
`ions (45). The deamidation rate for
`glutamine residues is usually less than
`that of asparagine residues (46, 47).
`If pH is >5.0, deamidation occurs
`through very unstable cyclic imide
`intermediate formation, which
`spontaneously undergoes hydrolysis.
`Under strong acidic conditions (pH
`1–2), direct hydrolysis of the amide side
`chain becomes more favorable than
`formation of cyclic imide (48). Peptide
`bond cleavage occurs to a greater extent
`in direct amide hydrolysis. At neutral
`pH, deamidation can lead to structural
`isomerization.
`The rate of deamidation is also
`influenced by protein secondary
`structure. Increasing helical structure
`decreases the rate of deamidation in
`some proteins (50). The rate of
`deamidation in several growth-
`hormone–releasing factor analogs was
`examined as a function of methanol-
`induced α-helical structure. Addition
`of methanol increased the level of
`α-helicity and decreased the rate of
`
`26 BioProcess International
`
`January 2011
`
`deamidation (51). In its native
`structure, RNAase resists
`deamidation possibly because of the
`relatively rigid backbone in the loop
`stabilized by a disulfide between
`Cys-8 and Cys-12 and by the β-turn
`at residues 66–68, which could
`hinder the formation of the cyclic
`imide (52). But if it is reduced and
`denatured, then refolded, aspartic
`and isoaspartic forms are generated,
`demonstrating different enzymatic
`activities. Replacement of Asp-67
`with Iso-Asp-67 showed that the
`isoaspartic form refolds at half the
`rate of the fully amidated form (51).
`Storage temperatures can affect a
`protein’s deamidation rate in the
`presence of certain biological buffers.
`Because amine buffers (e.g., Tris and
`histidine) have high temperature
`coefficients, storage at temperatures
`that are different from the temperature
`of preparation could shift formulation
`pH. Deamidation and isomerization
`reactions are pH-sensitive processes, so
`those shifts in formulation pH could
`alter the rate of deamidation. Another
`indirect effect of temperature is the
`dissociation constant of water: The
`hydroxyl ion concentration of water can
`vary as a function of temperature and
`thereby affect deamidation rates (39).
`Preventive Measures: Solution pH
`can substantially affect deamidation
`(44). Formulations at pH 3–5 can
`minimize peptide deamidation (48).
`AsnA-21 and AsnB-3 of insulin forms
`isoaspartate or aspartate, depending
`on the pH of solution (52). Insulin
`deamidates rapidly at Asn A-21 in low
`pH solutions (57). Steric hindrance
`also can affect deamidation rate:
`Bulky residues following asparagine
`may inhibit the formation of
`succinimide intermediate in the
`deamidation reaction (42).
`Replacement of a glycine residue with
`more bulky leucine or proline residues
`resulted in a 30- to 50-fold decrease in
`the rate (42). In lyophilized
`formulations, the deamidation rate is
`typically reduced, probably due to
`limited availability of free water in
`which the reaction can occur.
`Formulations that incorporate
`organic cosolvents can decrease their
`deamidation rates because addition of
`
`organic solvents decreases dielectric
`constants of a solution (44). Decreasing
`solvent dielectric strength — by
`addition of cosolutes such as glycerol,
`sucrose, and ethanol to a protein
`solution — leads to significantly lower
`rates of isomerization and deamidation
`(39, 44). Lowering dielectric strength
`of the medium from 80 (water) to 35
`(PVP/glycerol/water formulations) led
`to about a sixfold decrease in peptide
`deamidation rates (54). The lower rate
`of deamidation was attributed to less
`stabilized ionic intermediates formed
`during cyclization in the asparagine
`deamidation pathway. Insulin
`prepared in neutral solutions
`containing phenol showed reduced
`deamidation probably because of its
`stabilizing effect on the tertiary
`structure (α-helix formation) around
`the deamidating residue, which
`lowered the probability for formation
`of intermediate imides (53).
`aggregation and PreciPitation
`Aggregated proteins are a significant
`concern for biopharmaceutical
`products because they may be
`associated with decreased bioactivity
`and increased immunogenicity.
`Macromolecular protein complexes
`can trigger a patient’s immune system
`to recognize the protein as “nonself”
`and mount an antigenic response (55).
`Large macromolecular aggregates also
`can affect fluid dynamics in organ
`systems such as eyes (56).
`Aggregation is a common problem
`encountered during manufacture and
`storage of proteins (16). The potential
`for aggregated forms is often enhanced
`by exposure of a protein to liquid–air,
`liquid–solid, and even liquid–liquid
`interfaces (57). Mechanical stresses of
`agitation (shaking, stirring, pipetting
`or pumping through tubes) can cause
`protein aggregation. Freezing and
`thawing can promote it as well.
`Solution conditions such as
`temperature, protein concentration,
`pH, and ionic strength can affect the
`rate and amount of aggregates
`observed. Formulation in sucrose can
`increase aggregation over time because
`of protein glycation when sucrose is
`hydrolyzed (58). The presence of
`certain ligands — including certain
`
`Eton Ex. 1031
`5 of 10
`
`

`

`ions — may enhance aggregation.
`Interactions with metal surfaces can
`lead to epitaxic denaturation, which
`triggers aggregate formation. Foreign
`particles from the environment,
`manufacturing process, or container–
`closure system (e.g., silicone oil) can
`also induce aggregation (21, 59, 60).
`Even handling protein products at
`compounding pharmacies can induce
`aggregation 10-fold above initially
`observed amounts (58).
`The impact of aggregation on
`product potency varies based on the
`physiochemical attributes of each
`protein relative to its functional
`domains and the nature of the activity
`being measured. Enzymes such as
`urease and catalase can lose up to 50%
`of their potency after shaking,
`fibrinogen clotting activity is
`decreased after shear stress, and
`recombinant IL-2 and recombinant
`interferon activity is substantially
`affected by aggregation from shaking
`and shearing (2). Aggregation also
`affects the mass balance of protein
`solutions, decreasing the concentration
`of the target protein. Microaggregated
`subvisible particles generated
`anywhere in a manufacturing process
`can develop into larger particles over
`time as a product is stored (62).
`Bevacizumab drug product lost 50% of
`active IgG after manipulations at the
`pharmacy triggered significant growth
`of micron-sized particles in
`repackaged solutions (58).
`Aggregates can be soluble or
`insoluble, reversible or irreversible,
`covalent or noncovalent (16). Soluble
`aggregates are usually reversible: e.g.,
`by altered solution conditions such as
`changing temperature or osmotic
`strength or by mild physical disruption
`such as swirling or filtration. Insoluble
`aggregates are typically irreversible.
`Under vigorous physical disruption
`(e.g., agitation or freezing and thawing)
`or over time in storage, they can grow
`into particles that may eventually
`precipitate. Covalent aggregates form
`when monomeric proteins become
`chemically crosslinked, e.g., though
`disulfide bonds. Although covalent
`linkages are necessary to stabilize the
`native tertiary structure of most
`polypeptide proteins, those that form
`
`by degradation can produce undesired
`crosslinks between protein moieties,
`which can lead to irreversible
`aggregation. Noncovalent aggregates
`are formed when proteins associate and
`bind based on structural regions of
`charge or polarity. Because such
`associates are weak (relative to covalent
`linkages), they are sensitive to solution
`conditions and usually reversible.
`Mechanism and Factors Involved:
`Because of the many physical and
`chemical manipulations required in
`upstream production and downstream
`processing, followed by formulation
`and filling operations, aggregation of
`protein biopharmaceuticals can be
`induced during nearly every step of the
`process including at hold points,
`shipping, and long-term storage (16, 21).
`Agitation (e.g., shaking, stirring, and
`shearing) of protein solutions can
`promote aggregation at the air–liquid
`interfaces, where protein molecules may
`align and unfold, exposing their
`hydrophobic regions for charge-based
`association (2). Agitation-induced
`aggregation has been seen in numerous
`protein products, including
`recombinant factor XIII, human
`growth hormone, hemoglobin, and
`insulin (2). Minimizing foaming caused
`by agitation during manufacture (as
`well as during product use) may be
`critical to preventing significant loss of
`protein activity or generation of visible
`particulate matter (62).
`Protein concentration also can
`promote aggregation, with or without
`agitation events. Results obtained
`from two PEGylated proteins and one
`Fc fusion protein demonstrated a
`direct correlation between protein
`concentration and aggregation under
`nonagitated (quiescent) conditions, but
`researchers found an inverse
`correlation between protein
`concentration and aggregation under
`conditions of shaking, vortexing, and
`simulated shipping (63).
`Antimicrobial preservatives used in
`multidose formulations also can
`induce protein aggregation. For
`example, benzyl alcohol accelerates the
`aggregation of rhGCSF because it
`favors partially unfolded
`conformations of the protein (64).
`Increasing antimicrobial preservative
`
`
`Say hello toSay hello to
`
`streamlinedstreamlined
`
`sterile connections in sterile connections in
`
`seconds, anywhereseconds, anywhere
`
`
`Say goodbye to bulky,Say goodbye to bulky,
`
`expensive tube weldersexpensive tube welders
`
`
`Available in multiple sizes, Kleenpak™ sterileAvailable in multiple sizes, Kleenpak™ sterile
`
`connectors are rigorously validated underconnectors are rigorously validated under
`
`worst-case conditions, so you can be sureworst-case conditions, so you can be sure
`
`the connection remains sterile every time...the connection remains sterile every time...
`
`even outside a controlled air environment.even outside a controlled air environment.
`
`
`w: www.pall.com/allegrow: www.pall.com/allegro
`e: allegro@pall.com e: allegro@pall.com
`
`
`
`
`Providing Flexible SolutionsProviding Flexible Solutions
`
`
`© 2010 Pall Corporation. Pall, , and Kleenpak are trademarks of Pall © 2010 Pall Corporation. Pall, , and Kleenpak are trademarks of Pall
`
`Corporation. The ® indicates a trademark registered in the USA. GN10.3621Corporation. The ® indicates a trademark registered in the USA. GN10.3621
`
`
`
`BPI0101-QP-Pall.indd 2BPI0101-QP-Pall.indd 2
`
`
`
`12/13/10 12:53:03 PM12/13/10 12:53:03 PM
`
`Eton Ex. 1031
`6 of 10
`
`

`

`levels may increase the hydrophobicity
`of a formulation and could affect a
`protein’s aqueous solubility (62).
`Phenol and m-cresol can considerably
`destabilize a protein: Phenol promotes
`formation of both soluble and
`insoluble aggregates, whereas m-cresol
`can precipitate protein (65).
`Freezing and thawing — which can
`occur multiple times throughout
`production and use of protein
`therapeutics — can dramatically affect
`protein aggregation. Generation of
`water-ice crystals at a container’s
`periphery (where heat transfer is
`greatest) can produce a “salting out”
`effect, whereby the protein and
`excipients become increasingly
`concentrated at the slower-freezing
`center of a container (21). High-salt
`and/or high-protein concentrations
`can result in precipitation and
`aggregation during freezing, which is
`not completely reversible upon
`thawing. The effect can be seen with
`thyroid-stimulating hormone: When
`stored at –80 °C, 4 °C, or 24 °C for up
`to 90 days, it remained stable, but
`when frozen to –20 °C it lost >40%
`potency in that period, which was
`attributed to subunit dissociation (66).
`Multiple freezing and thawing cycles
`can exacerbate that effect and lead to a
`cumulative impact on the generation
`and growth of subvisible and visible
`particulates. A change in pH can
`come from crystallization of buffer
`components during freezing. In one
`study, potassium phosphate buffers
`demonstrated a much smaller pH
`change on freezing than did sodium
`phosphate buffers (21).
`Compendia currently limit the
`number of particles ≥10 μm and ≥25
`μm in size that may be present in
`injectible pharmaceutical preparations
`(67, 68). However, what levels of

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