throbber
REVIEWS
`
`Effects of Surfaces and Leachables on the Stability
`of Biopharmaceuticals
`
`JARED S. BEE,1 THEODORE W. RANDOLPH,2 JOHN F. CARPENTER,3 STEVEN M. BISHOP,1 MARIANA N. DIMITROVA1
`
`1Formulation Sciences Department, MedImmune, Gaithersburg, Maryland 20878
`
`2Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado 80309
`
`3Department of Pharmaceutical Sciences, University of Colorado Health Sciences Center, Aurora, Colorado 80045
`
`Received 19 October 2010; revised 11 January 2011; accepted 12 April 2011
`
`Published online 26 April 2011 in Wiley Online Library (wileyonlinelibrary.com). DOI 10.1002/jps.22597
`
`ABSTRACT: Therapeutic proteins are exposed to various potential contact surfaces, particles,
`and leachables during manufacturing, shipping, storage, and delivery. In this review, we present
`published examples of interfacial- or leachable-induced aggregation or particle formation, and
`discuss the mitigation strategies that were successfully utilized. Adsorption to interfaces or
`interactions with leachables and/or particles in some cases has been reported to cause protein
`aggregation or particle formation. Identification of the cause(s) of particle formation involving
`minute amounts of protein over extended periods of time can be challenging. Various formulation
`strategies such as addition of a nonionic surfactant (e.g., polysorbate) have been demonstrated
`to effectively mitigate adsorption-induced protein aggregation. However, not all stability prob-
`lems associated with interfaces or leachables are best resolved by formulation optimization.
`Detectable leachables do not necessarily have any adverse impact on the protein but control
`of the leachable source is preferred when there is a concern. In other cases, preventing pro-
`tein aggregation and particle formation may require manufacturing process and/or equipment
`changes, use of compatible materials at contact interfaces, and so on. This review summarizes
`approaches that have been used to minimize protein aggregation and particle formation dur-
`ing manufacturing and fill–finish operations, product storage and transportation, and delivery
`of protein therapeutics. © 2011 Wiley-Liss, Inc. and the American Pharmacists Association J
`Pharm Sci 100:4158–4170, 2011
`Keywords: protein aggregation; formulation; stability; agitation; air–water interface; adsorp-
`tion; particles; leachables; surface; biopharmaceuticals characterization
`
`INTRODUCTION
`Therapeutic proteins are used to treat a wide range
`of serious medical conditions, providing substantial
`benefits to patients. Proteins are complex molecules,
`subject to both intrinsic variation (e.g., glycosylation
`pattern and charge isoforms) and a variety of chemi-
`cal (e.g., deamidation and oxidation) and physical (for-
`mation of soluble aggregates, particle formation, and
`reversible association) degradation pathways. Most
`common intrinsic degradation pathways for protein
`therapeutics include aggregation and often particle
`
`Abbreviations used: mAb, monoclonal antibody.
`Correspondence to: Jared S. Bee (Telephone: +301–398-5912;
`Fax: +303-492-4341; E-mail: BeeJ@medimmune.com, jaredsbee
`@gmail.com)
`Journal of Pharmaceutical Sciences, Vol. 100, 4158–4170 (2011)
`© 2011 Wiley-Liss, Inc. and the American Pharmacists Association
`
`formation, with the resulting degradation products
`normally making up a very small mass fraction of the
`therapeutic protein product. Not all molecular vari-
`ants or degradation products necessarily result in a
`loss of efficacy or a decrease in safety. Some types of
`protein aggregates may elicit immune responses in
`patients.1,2 However, the mechanisms for immuno-
`genicity of therapeutic proteins in patients are still
`not well understood and a link between immunogenic-
`ity and aggregates or particles in products remains
`unclear in many cases.3,4
`Using state-of-the-art technology, biotechnology
`companies use formulation and process control strate-
`gies to obtain high purity and stability in order to
`meet a typical goal of a 2-year shelf life.5 For the
`general case of bulk protein aggregation as described
`by Chi et al.,6 either partial unfolding or aggre-
`gate assembly can be the rate-determining step for
`
`4158
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 10, OCTOBER 2011
`
`Novartis Exhibit 2040.001
`Regeneron v. Novartis, IPR2020-01317
`
`

`

`SURFACES AND LEACHABLES EFFECTS ON BIOPHARMACEUTICALS
`
`4159
`
`aggregation of proteins. The conformational and col-
`loidal stability of the protein can be optimized through
`the appropriate use of formulation buffer type, pH,
`and excipients.5–10 Similarly, formulation conditions
`can also be used to maximize chemical stability of
`proteins.5–10 High-concentration monoclonal antibody
`(mAb) products present their own unique challenges
`such as self-association, viscosity, opalescence, and
`protein particle formation.11,12
`Protein stability in bulk solution is only one of
`the key issues. During manufacturing, final fill–fin-
`ish, storage, and delivery, proteins may adsorb to sur-
`faces or react/bind with leachables. In some cases,
`this has resulted in aggregation, particle formation,
`or adsorption losses.5,10 Figure 1 depicts some of the
`processes of how solid and liquid contact surfaces and
`leachables have caused instabilities in protein prod-
`ucts. Adsorption of proteins to surfaces is a complex
`process that is important in many fields.13,14 Protein
`
`surface adsorption can be driven by a combination
`of electrostatic forces, hydrophobic binding interac-
`tions, and entropy changes due to contributions from
`both water and protein.14,15 These surface adsorption
`processes may be reversible or irreversible and may
`lead to either unfolding or partial unfolding of the ad-
`sorbed protein or only minimal perturbations to the
`protein structure. Depending on these factors, the ad-
`sorption of protein may be minimal and not cause any
`additional aggregation or particle formation. Simple
`adsorption can result in a reduction in the bulk pro-
`tein concentration that can be more of a concern for
`low concentration formulations. In other cases, pro-
`tein adsorption could nucleate further aggregation
`and particle formation. If adsorption is reversible,
`it is possible that the desorbed proteins may be re-
`leased in a structurally perturbed form that could
`lead to further aggregation or particle formation in
`the bulk.16 However, the detailed mechanism(s) has
`
`A
`
`–
`
`P
`
`V
`
`P
`
`L
`
`N
`
`N
`
`P
`
`A
`
`C
`
`A
`
`P
`
`I
`
`S
`
`M
`
`H
`
`A
`
`Figure 1. Possible physical degradation pathways of proteins caused by interfaces, foreign
`particulates, and leachables described in this review. The processes in the figure correspond to
`specific examples that have been published and are discussed in the text. Although the figure
`shows a vial as one example, these processes may also occur in other upstream operations and
`in other containers or delivery devices. These examples are also described and reviewed in this
`work.
`
`DOI 10.1002/jps
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 10, OCTOBER 2011
`
`Novartis Exhibit 2040.002
`Regeneron v. Novartis, IPR2020-01317
`
`

`

`4160
`
`BEE ET AL.
`
`not been fully determined for many published reports
`of adverse protein interactions with surfaces. Figure 1
`illustrates alternative mechanisms of particle forma-
`tion. These include agglomeration of protein-coated
`particles or silicone oil droplets and coagulation of
`proteins with leachables. This might occur when a
`few subvisible particles (SVPs) that were initially col-
`loidally stable (due to a high negative surface charge
`in the case of glass and silicone oil at common formu-
`lation pH conditions) become less stable when the sur-
`face charge is reduced by adsorption of protein. These
`protein-coated particles may then simply agglomerate
`together to form larger, more easily detectable parti-
`cles. It is also possible that foreign particles could ag-
`glomerate even if there is little or no protein adsorp-
`tion to the particles. A similar process of binding of
`leachables to proteins can lead to particle formation
`through colloidal destabilization of the protein, fol-
`lowed by precipitation of particles. Other leachables
`may also cause protein damage by directly reacting
`with the protein, potentially creating an aggregation-
`competent protein species. The last process for ag-
`gregation and particle formation we discuss is expo-
`sure to the air–water interface. Air–water interface
`exposure is one of the more common causes of par-
`ticle formation and aggregation described in the lit-
`erature. As with other interfaces, the details of the
`mechanism(s) of air–water interface-induced aggre-
`gation are not well described for many proteins. In
`this review, we present examples of the published ev-
`idence for these aggregation and particle formation
`processes and discuss rational mitigation strategies.
`Many of these examples of interface- and leachable-
`induced aggregation and particle formation processes
`are specific to certain products or conditions. We also
`note that detectable leachables may have no adverse
`impact of product safety, efficacy, quality, or protein
`stability. In this review, we have included many dif-
`ferent examples (even if they are less common) so that
`the lessons learned may be used to help in the practi-
`cal resolution of other similar issues in the future.
`
`MANUFACTURING AND FILL–FINISH
`OPERATIONS
`Manufacturing of therapeutic proteins is a complex
`process, which begins with production of the protein
`in cells cultured in a bioreactor wherein the protein
`is exposed to a multitude of solution species in the
`growth medium. The protein is then separated from
`the cell culture media by filtration or centrifugation.
`Recovery from inclusion bodies and refolding are per-
`formed if necessary. In downstream protein purifica-
`tion, viral inactivation and removal steps are often
`performed (e.g., low pH incubation, nanofiltration,
`and solvent–detergent addition). Multiple chromatog-
`raphy (e.g., affinity, ion exchange, and hydrophobic
`
`interaction) and filtration steps are used to purify
`the protein further. The protein may be concentrated
`and formulated using diafiltration. The formulated
`bulk may then be frozen or held before the final
`sterile filtration and fill–finish operations. Following
`the final sterile filtration step, the product is filled
`into vials, syringes, or cartridges. Each of these steps
`may expose the protein to interfaces (i.e., solid–liq-
`uid and air–liquid) under a variety of solution condi-
`tions. In this review, we focus on downstream exam-
`ples of interfacial protein instabilities, although many
`of the aggregation and particle formation processes to
`which proteins are exposed during downstream unit
`operations could also be relevant to the cell culture
`environment.
`
`Diafiltration
`Air bubble entrainment and/or microcavitation have
`been cited as a cause of aggregation during diafil-
`tration operations.17–19 Adsorption to solid surfaces,
`contamination by particulates, and increased rate of
`aggregate assembly due to mixing could also be causes
`of aggregation.19 Simple adsorption losses and fouling
`of the protein onto the membrane can also occur. For
`instance, deactivation of aminoacylase was directly
`caused by adsorption losses to an ultrafiltration mem-
`brane surface.20 The type and brand of filtration mem-
`brane have been shown to result in different levels of
`protein adsorption.21
`Process controls may be used to minimize aggrega-
`tion during diafiltration by optimization of the oper-
`ation parameters such as the transmembrane pres-
`sure and cross-flow rate.22 It has been suggested
`that reducing turnover of the air–water interface and
`bubble entrainment would also reduce the formation
`of particles in biotherapeutics during diafiltration
`operations.18,19 It is possible that some formulation
`excipients can provide additional protection during
`diafiltration. This of course depends upon whether ex-
`cipients are added during the diafiltration operation
`or afterward by addition of a concentrated stock of
`the excipients. Although addition of a surfactant can
`suppress the formation of aggregates at the air–water
`interface when the protein is also exposed to shear,23
`this strategy may not be practical for diafiltration op-
`erations. Surfactants are normally added after the
`diafiltration operation because of the difficulties in
`controlling and predicting the final surfactant level
`in the retentate.24
`
`Freezing and Thawing
`Freezing is a common unit operation during the pro-
`duction of therapeutic protein products. Bulk inter-
`mediates are often frozen to increase their stability
`during production hold steps and freezing is the first
`step in lyophilization. Freezing and thawing can trig-
`ger aggregation and particle formation in proteins by
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 10, OCTOBER 2011
`
`DOI 10.1002/jps
`
`Novartis Exhibit 2040.003
`Regeneron v. Novartis, IPR2020-01317
`
`

`

`SURFACES AND LEACHABLES EFFECTS ON BIOPHARMACEUTICALS
`
`4161
`
`various different mechanisms.25 Storage temperature
`and freezing rate are important parameters for frozen
`stability. One other factor that is the focus of this re-
`view contributing to the overall destabilization of the
`protein is the choice of container material. For ex-
`ample, polytetrafluoroethylene and other commercial
`freezing containers fostered more aggregation than
`polypropylene during freeze–thawing of an IgG2.26
`Cryoprotectants, such as sucrose or trehalose, are of-
`ten added to protein formulations to protect against
`freezing and thawing damage.25
`The ice–solution interface itself can be destabi-
`lizing to proteins: increased intermolecular $-sheet
`content was measured by infrared spectroscopy for
`two different proteins adsorbed to the ice surface.27
`Polysorbate addition has been shown to reduce for-
`mation of nonnative intermolecular $-sheet levels in
`proteins adsorbed to ice interfaces.27 In this case,
`the ability of polysorbate to reduce such structural
`changes in ice-adsorbed protein molecules was pro-
`tein specific.27 Polysorbate 20 protected Factor XIII
`during freeze–thawing by competing with the par-
`tially unfolded protein for interfaces.28 Additionally,
`polysorbate 80 protected hemoglobin from damage
`at interfaces during freezing.29 The 40% or greater
`loss of interleukin-11 (IL-11) activity caused by ad-
`sorption to glass lyophilization vials was prevented
`by polysorbate 20, although for complete protection
`during lyophilization, trehalose and human albumin
`were also necessary.30
`The rate of cooling and the degree of supercooling
`affect the number and sizes of ice crystals and the
`time the protein is exposed to the ice interface. Each
`of these variables could potentially influence the ex-
`tent of freeze–thawing-induced protein aggregation.
`Because there are multiple variables in freeze–thaw
`stress, experimental studies to test the sensitivity of
`the specific protein formulation to realistic and worst-
`case freeze–thaw stresses can be used to determine
`appropriate mitigation strategies.
`
`Sterile Filtration and Fill–Finish
`Sterile filtration and fill–finish operations may exert
`adverse effects on stability by exposing the protein to
`production equipment surfaces (e.g., those presented
`by membranes, tubing, and pumps). In an engineering
`approach, choice of equipment to minimize air–water
`interface exposure and turnover, particle shedding,
`leachables, and cavitation can be employed to elimi-
`nate or minimize suspected causes of aggregation.19
`This type of optimization should be performed while
`also maintaining product homogeneity (i.e., ensuring
`mixing is adequate) and sterility, and overall robust-
`ness and quality. These same strategies could also
`be useful to minimize aggregation or particle forma-
`tion in other upstream unit operations. Formulation
`approaches can also be very effective at reducing ad-
`
`verse interactions with interfaces. For instance, the
`aggregation leading to membrane fouling during ster-
`ile filtration of human growth hormone was found to
`be caused by adsorption to hydrophobic interfaces and
`could be mitigated by addition of surfactant.31 Differ-
`ences in the magnitude of protein adsorption has been
`observed between different types and brands of ster-
`ilizing filters (e.g., polyvinylidene fluoride (PVDF),
`polyethersulfone (PES), cellulose acetate (CA), and
`Nylon).21 Various filters were found to adsorb polysor-
`bate 80, requiring appropriate setup of the preflush
`step to avoid decreasing the levels of surfactant below
`the intended value for the final protein formulation.21
`Interestingly, it has been found that cellulose could
`preferentially adsorb soluble aggregates of a mAb
`from solution, although this did not have any adverse
`effect on the protein stability in bulk solution.32
`Stainless steel is ubiquitous in protein produc-
`tion equipment and has been reported to be a cause
`of protein aggregation or fragmentation in several
`cases: submicron steel particles shed from a pump in
`the laboratory environment caused “agglomeration of
`protein-coated particles” (see Fig. 1) and/or nucleated
`formation of larger aggregates of a mAb33; Fe ions
`caused hinge-region fragmentation of a mAb34; expo-
`sure to the stainless steel surface combined with ad-
`ditional shear stress resulted in aggregation of a mAb
`that exhibited a first-order dependence on protein
`concentration35; Fe ions leached from steel caused
`oxidation and aggregation36; Fe ions directly bound
`to a protein resulting in conformational destabiliza-
`tion followed by aggregation,37 and surface-induced
`soluble aggregation of a mAb had a second-order de-
`pendence on steel surface area and a zero-order de-
`pendence on bulk protein concentration that could
`be completely suppressed by polysorbate.38 Stainless
`steel surfaces typically are “passivated” or “electropol-
`ished” to create a more corrosion-resistant chromium
`oxide-rich surface layer. Factors that may impact the
`protein in solution include the following: the grade of
`steel alloy, the frequency of passivation, and chemical
`exposures of the steel. The impact of the formulation
`may play a particularly large role in the potential ad-
`verse interactions; for instance, exposure of steel to
`chloride ions at low pH has been shown to result in
`corrosion and release of Fe ions that subsequently cat-
`alyzed the oxidation of methionine residues.36 Stain-
`less steel exposure is an example of where there may
`be multiple distinct causes of aggregation or par-
`ticle formation: the steel surface itself, steel parti-
`cles shed from equipment, and the Fe ions leached
`from steel equipment. These examples would corre-
`spond to the scenarios of “physical or chemical in-
`stability caused by leachables” (Fe ions), “nucleation
`of aggregates on heterogenous particles or surfaces”
`(steel surface), and “agglomeration of protein-coated
`particles” (steel particles) shown in Figure 1. Here,
`
`DOI 10.1002/jps
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 10, OCTOBER 2011
`
`Novartis Exhibit 2040.004
`Regeneron v. Novartis, IPR2020-01317
`
`

`

`4162
`
`BEE ET AL.
`
`correct identification of the cause of the aggregation,
`fragmentation, or particle formation is crucial for cre-
`ating an effective mitigation strategy. Addition of a
`surfactant would be expected to reduce aggregation
`induced by surface adsorption, yet may not be effec-
`tive in eliminating oxidation, fragmentation, or con-
`formational destabilization caused by Fe ions. Rather,
`direct reduction of Fe ion levels by frequent passiva-
`tion of equipment and avoiding exposures of steel to
`extreme low pH in the presence of chloride or other
`corrosive ions might be a better strategy to eliminate
`negative effects of Fe ions on protein stability.36 Ad-
`dition of metal chelators has also been shown to be
`effective in eliminating the multiple adverse effects
`of Fe ions on protein stability, although care must be
`taken in the choice and level of the chelator.37 Nu-
`cleation of larger visible particles from smaller steel
`particles shed from pumps may not be completely sup-
`pressed by surfactant.26 In this scenario, a change in
`the process equipment has been shown to be effective.
`For instance, protein particle formation during fill-
`ing of an IgG was eliminated by replacement of a ra-
`dial piston pump with a rolling diaphragm pump.39 In
`some cases, there may be synergistic or compounded
`effects that may make identification of the problem
`and correct mitigation more difficult. A good example
`is where buffer-dependent conformational changes in
`a mAb increased the exposure of a site sensitive to
`Fe-catalyzed fragmentation.34
`Stainless steel is not the only important in-process
`surface to consider. In recent years, use of dis-
`posable containers has become a common practice
`in various steps of the manufacturing of protein
`therapeutics. Disposable containers pose potential
`challenges associated with leachables and possible
`shedding of particles, and are usually subjected to ex-
`tensive evaluation by biopharmaceutical companies
`before implementation.40
`
`CONTAINER CLOSURE
`Glass vials with rubber stoppers made of various poly-
`mers and coatings are commonly used primary con-
`tainers for protein therapeutics. Most recently, vials
`or syringes made of cyclic polyolefin (clear plastic) are
`being evaluated as options for container-closure ma-
`terials for some biopharmaceuticals.41 Container clo-
`sures can be exposed to various solvents to extract and
`identify compounds that are then monitored as leach-
`ables under realistic product contact conditions.40
`This can result in an identification of a large number
`of extractables that are often not actually detectable
`in the formulation upon extended product contact. Di-
`rect health-based risk assessments can then be per-
`formed based on the extractables–leachables data for
`a given product configuration.40 Indirect effects of
`leachables could potentially include aggregation or
`
`particle formation.40 Detectable leachables may not
`necessarily have any adverse effects on product safety,
`efficacy, or quality.
`Rapid growth in the applications of targeted
`biotechnology products is driving the development of
`alternative delivery systems including prefilled sy-
`ringes (PFSs), autoinjectors (AIs), and infusion de-
`vices. Multiple commercial products are currently of-
`fered as PFSs and AI devices, and the number is
`expected to rapidly grow. The development of PFSs,
`AI, and infusion devices are associated with potential
`for component compatibility challenges. These poten-
`tial challenges include sensitivity of proteins to the
`silicone oil often used to enhance the gliding perfor-
`mance of the syringe/device, sensitivity to trace lev-
`els of metals such as tungsten, which may be used in
`the manufacturing of glass syringes with staked nee-
`dles, and potential leachables from the glass, silicone,
`rubber, and adhesive contact surfaces. These possible
`adverse interactions are addressed during compati-
`bility and stability studies during development. In
`addition, various types of syringes are being devel-
`oped currently by multiple vendors including silicone
`oil- and tungsten-free syringes, enabling a greater se-
`lection of container-closure systems to be potentially
`chosen from and/or evaluated during development.
`
`Glass
`Borosilicate glass is the most commonly used primary
`container material for biopharmaceuticals.41 During
`development, each product formulation is generally
`assessed and optimized for stability in glass vials
`(with stopper). Glass vials surface properties can vary
`between manufacturers and may change due to in-
`teractions with the solution or sterilization proce-
`dures, which could potentially result in pitting or
`delamination.41–44 Glass has been successfully used
`for many commercial protein products without caus-
`ing aggregation or particle formation. Although re-
`ports of glass delamination are extremely rare for
`biotechnology products, the recent voluntary recall of
`a commercial protein therapeutic because some lots
`“. . .may contain extremely thin glass flakes (lamellae)
`that are barely visible in most cases” shows that de-
`lamination is still an important quality factor to be
`considered.45 We note that the voluntary recall also
`states that “To date, there have been no complaints
`or adverse events reported which can be directly at-
`tributed to the presence of glass lamellae.”45
`Excipients can also potentially interact with leach-
`ables from glass. Depending upon the exact supplier,
`glass can potentially leach ions such as barium or
`aluminum forming insoluble visible particles of bar-
`ium sulfate or aluminum phosphate when exposed
`to formulation excipients (sulfate and phosphate).46
`Proteins can adsorb to glass surfaces. In one case,
`the adsorption of protein to glass was minimized by
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 10, OCTOBER 2011
`
`DOI 10.1002/jps
`
`Novartis Exhibit 2040.005
`Regeneron v. Novartis, IPR2020-01317
`
`

`

`SURFACES AND LEACHABLES EFFECTS ON BIOPHARMACEUTICALS
`
`4163
`
`use of a siloxane coating and addition of surfactant.47
`Although they do not have identical surface chem-
`istry, silica nanoparticles have been used as a surro-
`gate surface for glass and were found to nucleate the
`growth of aggregates of recombinant human platelet-
`activating factor acetylhydrolase.48 Adsorption of the
`protein to glass and resulting aggregation could be
`reduced by choosing a solution pH at which both the
`protein and the glass had a net negative charge.48
`Also, in a more recent study with a mAb, it was found
`that there were differences in adsorption affinity as
`a function of pH and a surprising tendency for pref-
`erential adsorption of aggregates when the mAb was
`formulated with 150 mM NaCl in 10 mM sodium ac-
`etate at pH 5.0 and 5.5, and 150 mM NaCl in sodium
`phosphate at pH 6.0 and 6.5.49 It has been shown that
`for several mAbs, the structural changes on adsorp-
`tion to spiked glass particles were minimal, however,
`the protein-coated glass particles were less colloidally
`stabile, resulting in formation of larger agglomerated
`clusters of the spiked glass particles.32,50 This spe-
`cific example corresponds to the general phenomenon
`of “agglomeration of protein-coated particles” illus-
`trated in Figure 1. The interactions between glass
`and protein are clearly both protein specific and for-
`mulation dependent. Simple adsorption losses at low
`concentrations may be reduced by use of coatings or
`surfactants.
`
`Stoppers
`Vial stoppers are often made from butyl and halobutyl
`rubber that may also contain other additives or poten-
`tial leachables depending upon the proprietary com-
`pound formulation.41 Not all detectable leachables
`will necessarily have any negative impact on product
`quality. In one case, vial stoppers released metal ions
`that in turn activated a metalloprotease, which then
`resulted in protein product damage.1 Polytetrafluo-
`rethylene (PTFE) or other proprietary polymers have
`been used to coat vial stoppers to reduce the leaching
`of rubber components into the bulk formulation. The
`pure red cell aplasia (PRCA) experienced by Eprex R(cid:1)
`(Johnson & Johnson, New Brunswick, NJ) patients
`has been attributed to leachables released from un-
`coated rubber stoppers in the presence of polysorbate
`used to replace human albumin as a stabilizer.51–53 In
`other work, extractables from stoppers were not able
`to elicit “danger signal” responses in dendritic cells
`that would indicate their immunogenic potential.54
`Association of protein with polysorbate micelles has
`also been suggested as a possible cause of the im-
`mune responses,55 although this particular hypothe-
`sis has been questioned after additional studies by
`other workers suggested that association with mi-
`celles does not occur.53,56 The fundamental details and
`cause(s) of the observed PRCA cases remain the sub-
`
`ject of ongoing research and debate. Ultimately, the
`incidence of PRCA decreased when a FluroTec R(cid:1) (West
`Pharmaceutical Services, Inc., Lionville, PA) stopper
`configuration replaced the uncoated stopper and the
`route of administration was changed from subcuta-
`neous to intravenous.40,52
`Container-closure changes are considered high
`risk by regulatory agencies for parenteral biotech
`products.1 Optimization of
`the formulation and
`container-closure configuration is performed together.
`Although use of a PTFE stopper apparently improved
`the stability of Eprex R(cid:1), PTFE was found to cause the
`aggregation of insulin in accelerated studies.57 For-
`mulation is important: Factor VIII adsorption to hy-
`drophobic surfaces, with concomitant unfolding and
`loss of activity of the adsorbed protein, could be re-
`duced by polysorbate 80.58 Interestingly, adsorption
`of Factor VIII to hydrophilic surfaces was not reduced
`by polysorbate, although no associated unfolding or
`loss of activity of the adsorbed protein was observed
`as was seen for hydrophobic surfaces.58 Stoppers are
`also often coated with silicone oil, and the amount of
`silicone coated is one variable that can be controlled.
`Vials are normally shipped and stored upright so the
`exposure to the stopper might be minimal for an ac-
`tual product. But for accelerated degradation studies,
`incubation in vials that are inverted or laid side-on
`can also be used to assess the impact of the stopper
`on the stability of the product.
`
`Tungsten and Needle Glue
`Contamination of glass syringes with tungsten com-
`pounds released from the tungsten pin used to form
`the needle-mounting hole was identified as a cause of
`protein particle formation,59 and has been reported
`to have resulted in the formation of visible parti-
`cles in at least two therapeutic protein products.1
`In a recent study, it was demonstrated that tung-
`sten metal particles dissolved into negatively charged
`polyanions that electrostatically aggregated with a
`mAb, resulting in rapid visible particle formation.60
`Aggregation of the mAb by tungsten species was
`only observed below about pH 6.0 and above tung-
`sten levels of about 3–9 ppm, consistent with the
`fact that tungsten polyanions are the major soluble
`species formed under these conditions.60 The small
`number of tungsten metal particles that need to dis-
`solve to achieve this level of soluble polyanions high-
`lights their potential potency in causing aggrega-
`tion at lower pHs.60 Tungsten polyanion aggregation
`with proteins was largely reversible in phosphate-
`buffered saline.60 The mechanism of this aggrega-
`tion of protein with tungsten polyanions can be
`described as “coagulation:” coagulation is a colloidal
`science term for when a colloid (in this case the pro-
`tein) is electrostatically bridged and cross-linked by
`
`DOI 10.1002/jps
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 10, OCTOBER 2011
`
`Novartis Exhibit 2040.006
`Regeneron v. Novartis, IPR2020-01317
`
`

`

`4164
`
`BEE ET AL.
`
`charged species in solution (in this case tungsten
`polyanions), leading to particles and precipitate for-
`mation. Results for Fc fusion and "-helical proteins
`spiked with extracts from actual tungsten pins used
`in syringe-forming operations were also consistent
`with polyanion-induced coagulation.61 Particle forma-
`tion was greatest at lower pHs and higher soluble
`tungsten levels, but in all cases, particles were only
`formed at tungsten levels 10 times higher than those
`actually measured in commercial PFS.61 These ex-
`amples correspond to “coagulation with leachables”
`as depicted in Figure 1. In response to the potential
`tungsten-induced protein degradation, syringe man-
`ufacturers have developed proprietary manufactur-
`ing processes and engineering changes that control
`or effectively eliminate residual tungsten contamina-
`tion in glass-staked needle syringes. Here we have
`referenced work showing different tungsten sensitiv-
`ities of a mAb, an Fc fusion protein, and an "-helical
`protein.60,61 These sensitivities varied based on mul-
`tiple factors such as the protein-to-tungsten ratio, the
`protein properties, protein concentration, and the for-
`mulation conditions.60,61 Therefore, sensitivity of dif-
`ferent proteins and different formulations to tungsten
`leached from forming pins should be evaluated on a
`case-by-case basis because there may, or may not, be
`an impact at the actual potential worse case expected
`tungsten levels in PFSs.60,61 It is likely that the re-
`ductions of tungsten level in syringes, combined with
`knowledge of the pH effects on potential coagulation
`with protein, can now be used to effectively mitigate
`this specific issue for proteins which are sensitive to
`tungsten.
`The US Food and Drug Administration (FDA) re-
`ported in one case that if the specific glue used to
`attach the needle to the syringe tip was not allowed
`to fully dry, it could leach solvents, resulting in pro-
`tein oxidation.46 They reported that in this specific
`case, the issue could

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