Effects of Surfaces and Leachables On The Stability of Biopharmas

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

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
1
Formulation Sciences Department, MedImmune, Gaithersburg, Maryland 20878
2
Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado 80309
3
Department 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 formation, with the resulting degradation products


normally making up a very small mass fraction of the
Therapeutic proteins are used to treat a wide range
therapeutic protein product. Not all molecular vari-
of serious medical conditions, providing substantial
ants or degradation products necessarily result in a
benefits to patients. Proteins are complex molecules,
loss of efficacy or a decrease in safety. Some types of
subject to both intrinsic variation (e.g., glycosylation
protein aggregates may elicit immune responses in
pattern and charge isoforms) and a variety of chemi-
patients.1,2 However, the mechanisms for immuno-
cal (e.g., deamidation and oxidation) and physical (for-
genicity of therapeutic proteins in patients are still
mation of soluble aggregates, particle formation, and
not well understood and a link between immunogenic-
reversible association) degradation pathways. Most
ity and aggregates or particles in products remains
common intrinsic degradation pathways for protein
unclear in many cases.3,4
therapeutics include aggregation and often particle
Using state-of-the-art technology, biotechnology
companies use formulation and process control strate-
Abbreviations used: mAb, monoclonal antibody. gies to obtain high purity and stability in order to
Correspondence to: Jared S. Bee (Telephone: +301–398-5912; meet a typical goal of a 2-year shelf life.5 For the
Fax: +303-492-4341; E-mail: BeeJ@medimmune.com, jaredsbee
@gmail.com) general case of bulk protein aggregation as described
Journal of Pharmaceutical Sciences, Vol. 100, 4158–4170 (2011) by Chi et al.,6 either partial unfolding or aggre-
© 2011 Wiley-Liss, Inc. and the American Pharmacists Association gate assembly can be the rate-determining step for

4158 JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 10, OCTOBER 2011
SURFACES AND LEACHABLES EFFECTS ON BIOPHARMACEUTICALS 4159

aggregation of proteins. The conformational and col- surface adsorption can be driven by a combination
loidal stability of the protein can be optimized through of electrostatic forces, hydrophobic binding interac-
the appropriate use of formulation buffer type, pH, tions, and entropy changes due to contributions from
and excipients.5–10 Similarly, formulation conditions both water and protein.14,15 These surface adsorption
can also be used to maximize chemical stability of processes may be reversible or irreversible and may
proteins.5–10 High-concentration monoclonal antibody lead to either unfolding or partial unfolding of the ad-
(mAb) products present their own unique challenges sorbed protein or only minimal perturbations to the
such as self-association, viscosity, opalescence, and protein structure. Depending on these factors, the ad-
protein particle formation.11,12 sorption of protein may be minimal and not cause any
Protein stability in bulk solution is only one of additional aggregation or particle formation. Simple
the key issues. During manufacturing, final fill–fin- adsorption can result in a reduction in the bulk pro-
ish, storage, and delivery, proteins may adsorb to sur- tein concentration that can be more of a concern for
faces or react/bind with leachables. In some cases, low concentration formulations. In other cases, pro-
this has resulted in aggregation, particle formation, tein adsorption could nucleate further aggregation
or adsorption losses.5,10 Figure 1 depicts some of the and particle formation. If adsorption is reversible,
processes of how solid and liquid contact surfaces and it is possible that the desorbed proteins may be re-
leachables have caused instabilities in protein prod- leased in a structurally perturbed form that could
ucts. Adsorption of proteins to surfaces is a complex lead to further aggregation or particle formation in
process that is important in many fields.13,14 Protein the bulk.16 However, the detailed mechanism(s) has

N P
A –

P I
N
V

P S
P

M
L

H
C

A 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
4160 BEE ET AL.

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

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 10, OCTOBER 2011 DOI 10.1002/jps
SURFACES AND LEACHABLES EFFECTS ON BIOPHARMACEUTICALS 4161

various different mechanisms.25 Storage temperature verse interactions with interfaces. For instance, the
and freezing rate are important parameters for frozen aggregation leading to membrane fouling during ster-
stability. One other factor that is the focus of this re- ile filtration of human growth hormone was found to
view contributing to the overall destabilization of the be caused by adsorption to hydrophobic interfaces and
protein is the choice of container material. For ex- could be mitigated by addition of surfactant.31 Differ-
ample, polytetrafluoroethylene and other commercial ences in the magnitude of protein adsorption has been
freezing containers fostered more aggregation than observed between different types and brands of ster-
polypropylene during freeze–thawing of an IgG2 .26 ilizing filters (e.g., polyvinylidene fluoride (PVDF),
Cryoprotectants, such as sucrose or trehalose, are of- polyethersulfone (PES), cellulose acetate (CA), and
ten added to protein formulations to protect against Nylon).21 Various filters were found to adsorb polysor-
freezing and thawing damage.25 bate 80, requiring appropriate setup of the preflush
The ice–solution interface itself can be destabi- step to avoid decreasing the levels of surfactant below
lizing to proteins: increased intermolecular $-sheet the intended value for the final protein formulation.21
content was measured by infrared spectroscopy for Interestingly, it has been found that cellulose could
two different proteins adsorbed to the ice surface.27 preferentially adsorb soluble aggregates of a mAb
Polysorbate addition has been shown to reduce for- from solution, although this did not have any adverse
mation of nonnative intermolecular $-sheet levels in effect on the protein stability in bulk solution.32
proteins adsorbed to ice interfaces.27 In this case, Stainless steel is ubiquitous in protein produc-
the ability of polysorbate to reduce such structural tion equipment and has been reported to be a cause
changes in ice-adsorbed protein molecules was pro- of protein aggregation or fragmentation in several
tein specific.27 Polysorbate 20 protected Factor XIII cases: submicron steel particles shed from a pump in
during freeze–thawing by competing with the par- the laboratory environment caused “agglomeration of
tially unfolded protein for interfaces.28 Additionally, protein-coated particles” (see Fig. 1) and/or nucleated
polysorbate 80 protected hemoglobin from damage formation of larger aggregates of a mAb33 ; Fe ions
at interfaces during freezing.29 The 40% or greater caused hinge-region fragmentation of a mAb34 ; expo-
loss of interleukin-11 (IL-11) activity caused by ad- sure to the stainless steel surface combined with ad-
sorption to glass lyophilization vials was prevented ditional shear stress resulted in aggregation of a mAb
by polysorbate 20, although for complete protection that exhibited a first-order dependence on protein
during lyophilization, trehalose and human albumin concentration35 ; Fe ions leached from steel caused
were also necessary.30 oxidation and aggregation36 ; Fe ions directly bound
The rate of cooling and the degree of supercooling to a protein resulting in conformational destabiliza-
affect the number and sizes of ice crystals and the tion followed by aggregation,37 and surface-induced
time the protein is exposed to the ice interface. Each soluble aggregation of a mAb had a second-order de-
of these variables could potentially influence the ex- pendence on steel surface area and a zero-order de-
tent of freeze–thawing-induced protein aggregation. pendence on bulk protein concentration that could
Because there are multiple variables in freeze–thaw be completely suppressed by polysorbate.38 Stainless
stress, experimental studies to test the sensitivity of steel surfaces typically are “passivated” or “electropol-
the specific protein formulation to realistic and worst- ished” to create a more corrosion-resistant chromium
case freeze–thaw stresses can be used to determine oxide-rich surface layer. Factors that may impact the
appropriate mitigation strategies. protein in solution include the following: the grade of
steel alloy, the frequency of passivation, and chemical
Sterile Filtration and Fill–Finish
exposures of the steel. The impact of the formulation
Sterile filtration and fill–finish operations may exert may play a particularly large role in the potential ad-
adverse effects on stability by exposing the protein to verse interactions; for instance, exposure of steel to
production equipment surfaces (e.g., those presented chloride ions at low pH has been shown to result in
by membranes, tubing, and pumps). In an engineering corrosion and release of Fe ions that subsequently cat-
approach, choice of equipment to minimize air–water alyzed the oxidation of methionine residues.36 Stain-
interface exposure and turnover, particle shedding, less steel exposure is an example of where there may
leachables, and cavitation can be employed to elimi- be multiple distinct causes of aggregation or par-
nate or minimize suspected causes of aggregation.19 ticle formation: the steel surface itself, steel parti-
This type of optimization should be performed while cles shed from equipment, and the Fe ions leached
also maintaining product homogeneity (i.e., ensuring from steel equipment. These examples would corre-
mixing is adequate) and sterility, and overall robust- spond to the scenarios of “physical or chemical in-
ness and quality. These same strategies could also stability caused by leachables” (Fe ions), “nucleation
be useful to minimize aggregation or particle forma- of aggregates on heterogenous particles or surfaces”
tion in other upstream unit operations. Formulation (steel surface), and “agglomeration of protein-coated
approaches can also be very effective at reducing ad- particles” (steel particles) shown in Figure 1. Here,

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 10, OCTOBER 2011
4162 BEE ET AL.

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

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 10, OCTOBER 2011 DOI 10.1002/jps
SURFACES AND LEACHABLES EFFECTS ON BIOPHARMACEUTICALS 4163

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

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 10, OCTOBER 2011
4164 BEE ET AL.

charged species in solution (in this case tungsten by silicone oil lubricant on syringes.62,63 In a study
polyanions), leading to particles and precipitate for- of four model proteins, spiking of silicone oil at 0.5%
mation. Results for Fc fusion and "-helical proteins (v/v) was found to cause increased turbidity in pro-
spiked with extracts from actual tungsten pins used tein solutions, which was attributed to aggregation.
in syringe-forming operations were also consistent Interestingly, silicone oil did not cause measurable
with polyanion-induced coagulation.61 Particle forma- perturbation of the structures of the proteins nor alter
tion was greatest at lower pHs and higher soluble their thermal stability.64 Similarly, there were no ma-
tungsten levels, but in all cases, particles were only jor detectable structural perturbations or decreases
formed at tungsten levels 10 times higher than those in thermal stability of a mAb spiked with silicone oil
actually measured in commercial PFS.61 These ex- emulsion droplets, presumably because only a minute
amples correspond to “coagulation with leachables” fraction of the protein population was adsorbed to the
as depicted in Figure 1. In response to the potential silicone oil–aqueous solution interface.65 The silicone
tungsten-induced protein degradation, syringe man- oil could adsorb a monolayer of mAb, but formation
ufacturers have developed proprietary manufactur- of insoluble aggregate particles only occurred when
ing processes and engineering changes that control additional high-speed agitation was applied.65
or effectively eliminate residual tungsten contamina- Formulation conditions resulting in decreased col-
tion in glass-staked needle syringes. Here we have loidal stability of the mAb accelerated aggregation,
referenced work showing different tungsten sensitiv- and conversely, addition of the surfactant polysorbate
ities of a mAb, an Fc fusion protein, and an "-helical 20 completely inhibited aggregation.65 Again, in a dif-
protein.60,61 These sensitivities varied based on mul- ferent study of four varied proteins (including a com-
tiple factors such as the protein-to-tungsten ratio, the mercial mAb and a commercial Fc fusion protein),
protein properties, protein concentration, and the for- spiking of silicone oil into formulations containing
mulation conditions.60,61 Therefore, sensitivity of dif- sodium chloride, sucrose, and polysorbate resulted in
ferent proteins and different formulations to tungsten an initial protein loss due to surface adsorption with-
leached from forming pins should be evaluated on a out any additional protein loss or aggregation over a
case-by-case basis because there may, or may not, be 2-week period.66 In this particular study, the silicone
an impact at the actual potential worse case expected oil effects on protein stability in bulk solution were
tungsten levels in PFSs.60,61 It is likely that the re- minimal (without agitation) even for the range of pro-
ductions of tungsten level in syringes, combined with tein types and formulation conditions studied.66 The
knowledge of the pH effects on potential coagulation formulation acted both to alter the adsorption of pro-
with protein, can now be used to effectively mitigate tein molecules to the silicone oil droplets and to im-
this specific issue for proteins which are sensitive to pact the stability of the silicone oil emulsion itself.66
tungsten. We note that accelerated stability studies using sili-
The US Food and Drug Administration (FDA) re- cone oil spiking may not be a complete representation
ported in one case that if the specific glue used to of the actual exposure during transportation and stor-
attach the needle to the syringe tip was not allowed age of protein in syringes. Agglomeration of silicone
to fully dry, it could leach solvents, resulting in pro- oil droplets could account for some visual observations
tein oxidation.46 They reported that in this specific of “particles” after exposure to silicone oil: see the il-
case, the issue could be resolved by allowing the glue lustration for “agglomeration of protein-coated par-
to fully dry for 6 months prior to using the syringes.46 ticles” shown in Figure 1. Polysorbate stabilized the
The type of glue and method of curing was not spec- silicone oil emulsion but also dramatically reduced
ified in the FDA report but presumably other pro- adsorption of the protein to the silicone oil droplets,
cess parameters could impact the leaching of solvents whereas salt destabilized the emulsion and increased
from needle adhesives and should be considered for protein adsorption.66 In a more recent study of two
commercial container-closure configurations. different mAbs, extensive biophysical and stability
studies showed no change in structure, activity, or
Silicone Oil degradation profiles in PFS compared with vials ex-
Silicone oil is used to lubricate the barrel of glass PFSs cept in terms of subvisible particles.67 For the PFS
for smooth plunger motion with a lower injection configuration, there was a small (yet statistically sig-
force. Silicone oil is composed of polydimethylsiloxane nificant) increase in the levels of SVPs in PFSs for
but can also contain trace levels of cyclic polymers both mAbs, most likely due to the presence of sil-
such as octamethylcyclotetrasiloxane (called D4).46 icone oil droplets.67 The authors indicated that the
Silicone oil is also used to lubricate stoppers for ease results of clinical studies for the vial and PFS pre-
of handling and smooth insertion into vials.41 In early sentations indicated no differences in safety, efficacy,
studies, adverse effects of silicone oil on protein sta- or immunogenicity for patients receiving either the
bility were reported for the clouding of insulin caused vial or the PFS presentation.67 Sensitivity to silicone,

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 10, OCTOBER 2011 DOI 10.1002/jps
SURFACES AND LEACHABLES EFFECTS ON BIOPHARMACEUTICALS 4165

if it is observed, is likely both protein and formula- shaking of a different mAb.70 Low-shear exposure to
tion specific and may sometimes also require multiple the air–water interface caused aggregation of recom-
synergistic factors to occur (e.g., transportation binant human growth hormone but shear acted to en-
shocks plus exposure to silicone). The available pub- hance the rate of aggregation by increasing turnover
lished reports suggest that use of surfactant to min- of the interface.71 A few studies have reported that
imize adsorption to silicone while formulating for particles, rather than soluble aggregates, were in-
maximum colloidal stability of the protein should duced by agitation.72–74 These particles slowly dis-
minimize protein–silicone interactions, resulting in sociated over time and did not nucleate or propagate
decreased aggregation propensity. Presumably, in- further aggregation or particle formation in the bulk
creased conformational stability should also mini- solution, suggesting that they were formed exclu-
mize unfolding at interfaces and therefore also min- sively at the air–water interface.72–74
imize potential for aggregation caused by silicone oil Although there are numerous examples of where
surfaces. exposure to the air–water interface caused aggre-
Ongoing developments in syringe siliconization to gation and/or particle formation, the fundamental
optimize and control the level and distribution of sil- underlying kinetics and mechanism(s) are still not
icone oil to maximize functionality while minimizing completely understood. Aggregation at the air–wa-
the level (and excess) of silicone oil by syringe manu- ter interface may simply be due to increased num-
facturers may lead to syringes with minimal excess bers of collisions between protein molecules at the
droplet detachment from the syringe walls during interface, leading to the formation of aggregates and/
transportation or over time. Other treatments or sy- or particles. The potency of the air–water interface
ringe materials or coatings that are being developed in inducing aggregation may also be explained in
by syringe suppliers could also provide additional fu- part by partial unfolding of proteins caused by the
ture configuration options for “silicone oil sensitive” surface tension force at the air–water interface. The
proteins. forces at the air–water interface are comparable to
the 20–150 pN forces measured in atomic force mi-
croscopy (AFM) protein unfolding studies, suggesting
PRODUCT TRANSPORTATION AND STORAGE that the air–water interface can easily mechanically
unfold sections of proteins (e.g., a 140 pN force is ap-
Agitation and Air–Water Interface Exposures
plied across a 2 nm air–water interface with 70 mN/
Proteins may be exposed to agitation and mechanical m surface tension).19 In the limiting thermodynamic
shocks during both production and shipping. Agita- case, if the surface tension of the interface is greater
tion can expose the protein to interfacial forces at the than internal forces favoring compaction of the pro-
air–water interface. It has also been reported that tein, then the area of the protein must increase (i.e.,
agitation can be a contributing factor to silicone oil it will unfold) to achieve equilibrium when adsorbed
sensitivity.65 Shaking, stirring, or vortexing methods at the interface.75 The air–water interface is regen-
have been used in accelerated stability testing of pro- erative, allowing for cumulative protein damage over
tein formulations. In an agitation study of 13 var- time. Agitation and exposure to the air–water inter-
ied model proteins, particle formation was found to face involves multiple additional factors. In one study,
depend upon both the protein identity and the so- it was found that dynamic, dilational, surface area
lution conditions, although no attempts to link the changes at the air–water interface could cause par-
results with molecular properties were made.68 De- ticle formation.72 Compression of the interface (re-
pending upon the details of the agitation procedure, duction in the area) by changing the orientation of
vastly different qualitative and quantitative results vials during gentle rotation showed that compression
may be obtained, most likely because there are mul- of the air–water interface above a critical value of
tiple different simultaneous aggregation and particle about five sharply increased particle formation for a
formation processes occurring at the air–water inter- mAb at constant air–water interface area exposure
face during agitation. This presents a potential prob- rate.72 The rate of particle formation was essentially
lem if the formulation was optimized with respect to independent of the bulk protein concentration,72 con-
a certain accelerated stability test condition but is sistent with a surface-driven process and with other
exposed to different or additional forces and factors reports of decreased aggregation on a percentage ba-
upon real filling, transportation, and storage. In one sis as bulk protein concentration is increased during
example, stirring of a mAb in Reacti VialsTM (Thermo agitation.76
Fisher Scientific, Waltham MA) was found to produce Nonionic surfactants are commonly added to for-
an additional population of smaller aggregates when mulations to protect against agitation- or interface-
compared with horizontal shaking.69 Similarly, stir- induced aggregation and this generally appears to
ring was also found to cause more aggregation than be very effective and rational strategy for many

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 10, OCTOBER 2011
4166 BEE ET AL.

proteins.23,70,77,78 In one review, 13 out of 18 approved act as nuclei for further particle growth.83 In a similar
mAb products listed either polysorbate 20 or 80 as way, nuclei for insulin fibrillation were generated by
an excipient.79 As described above, accelerated agita- exposure to the air–water interface during agitation,
tion studies are often used to empirically determine followed by further growth in bulk solution.84
an optimal level of surfactant needed to inhibit ag- Although there are a large number of examples
gregation without necessarily elucidating the funda- when surfactants have been extremely effective in
mental mechanism(s) of action. Competition for ad- stabilizing proteins against agitation-induced par-
sorption at interfaces or direct binding to hydrophobic ticle formation, they do not always provide com-
patches on the protein are two of the most commonly plete protection against all aggregation. For instance,
identified mechanisms for the protective action(s) of rather than completely eliminating all aggregation,
surfactants.75 We note that polysorbate is also an ex- addition of polysorbate 80 stabilized small aggregates
cipient used to mitigate protein adsorption and un- of a mAb and prevented their conversion to particles
folding at other interfaces and may be required at during agitation studies.69 Surfactants can have “dual
higher levels to be effective for different surfaces (e.g., effects on protein stability:”85 Wang et al.85 demon-
silicone oil). Agitation studies employing multiple fac- strated that polysorbate 80 inhibited the agitation-
tors (e.g., air–water interface and silicone oil) are of- induced aggregation of IL-2 mutein, yet it increased
ten used for this purpose. In this section, we focus on the long-term rate of aggregation and oxidation in
examples relating to air–water interface-induced pro- quiescent solutions. In another case, polysorbate ef-
tein aggregation and particle formation. For Factor fectively mitigated aggregation for agitated protein
XIII, maximum protection against agitation-induced samples, yet again increased the aggregation in qui-
aggregation occurred when surfactant was added at escent samples.76 One reason for this could be in
levels near the critical micelle concentration and part because the degradation products of polysor-
there was no evidence of direct binding of polysorbate bate itself can include species that could potentially
to the protein.28 In contrast, stabilization by direct cause protein oxidation.85–87 However, the suscepti-
binding of polysorbate 20 to hydrophobic patches on bility to oxidation and potential for adverse effects
human growth hormone required a ratio of surfactant of oxidation is also likely very protein and formula-
to protein of greater than 4 for adequate protection tion specific, yet the advantages of polysorbate protec-
against agitation, rather than simply requiring lev- tion against agitation-induced particle formation are
els above the critical micelle concentration (CMC).80 great. There may also be other unexpected additional
Similarly, polysorbate 20 and 80 were able to protect effects of polysorbate such as enhanced leaching of di-
AlbutropinTM (Human Genome Sciences, Rockville, (2-ethylhexyl)phthalate (DEHP) from polyvinyl chlo-
Maryland) against agitation-induced soluble aggre- ride (PVC) intravenous (i.v.) bags discussed in the
gation by binding to the protein at molar ratios of Delivery section below. In summary, the literature
greater than 9 even when well below the CMC of the shows that polysorbate can be significantly stabiliz-
surfactants.81 Polysorbate 20 and 80 could fully pro- ing against agitation-induced aggregation of proteins
tect darbepoetin alfa from adsorption or agitation- if added at optimized levels. Control of the quality of
induced aggregation at about their respective CMC raw materials such as the expiration dates and per-
values.82 Additionally, monomeric polysorbate 20 and oxide levels in polysorbate excipients and control of
80 also bound to darbepoetin alfa resulting in (differ- possible synergistic factors (e.g., Fe ion levels, or oxy-
ent) subtle structural changes, yet darbepoetin alfa gen in the headspace of vials, and dissolved oxygen)
did not bind to the polysorbate micelles.82 could maximize the positive effects of polysorbate as
Some proteins seem quite robust with respect to a stabilizing excipient.
agitation: only 0.005% (w/v) polysorbate 20 (again be- Surfactants are not the only formulation compo-
low the CMC) was found to protect a mAb (reported to nent that may adsorb to interfaces or associate with
be extremely stable with respect to agitation) against proteins at interfaces: different buffer ions have been
particle formation induced by agitation at both low shown to change the relative affinity of the protein ad-
and high concentrations even though the “apparent sorption to the interface and the degree of unfolding
CMC” in the presence of high concentration mAb was of proteins at the interface.88,89 Cyclodextrin excip-
up to 0.04% (w/v).77 In other cases, proteins are easily ients have been reported to have stabilizing effects
aggregated during agitation, suggesting that suscep- on proteins exposed to agitation and air–water in-
tibility to aggregation induced by agitation may be terface turnover.90–92 Human serum albumin (HSA)
highly protein and formulation specific. In one exam- has been used as an effective excipient for preven-
ple, the exposure of buried cysteines caused by un- tion of adsorption losses of low concentration proteins
folding at the air–water interface was found to result and also as a stabilizing excipient for very hydropho-
in intermolecular disulfide bonds, possibly resulting bic proteins.30,93 However, HSA does have multiple
in the formation of a small number of particles that drawbacks as a potential pharmaceutical excipient.94

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 10, OCTOBER 2011 DOI 10.1002/jps
SURFACES AND LEACHABLES EFFECTS ON BIOPHARMACEUTICALS 4167

DELIVERY that has adverse interactions with the protein and


elimination and/or control of its source is one of the
Syringes, Bags, and Lines for Intravenous Administration
most powerful strategies for improving protein stabil-
Some proteins may be susceptible to aggregation or ity (e.g., Fe ions and tungsten). Sometimes classifica-
particle formation induced by adsorption to hydropho- tion of a species as a leachable or an impurity may
bic surfaces to which therapeutic protein formula- complicate identification of the source. Protein insta-
tions are exposed during delivery of the products to bilities caused by adsorption to equipment surfaces or
patients. For instance, exposure to silicone rubber de- due to shed particles can be alleviated by operational
livery tubing decreased IL-2 activity by 97% because or procedural changes or by changing the surface or
of adsorption to the tubing surface followed by des- equipment type itself (e.g., membranes, pumps, and
orption of structurally altered inactive molecules.95 tubing). Similarly, compatibility studies can identify
Additives to polymers have resulted in adverse in- problems with particular container closures and de-
teractions with proteins. Plasticizers on the surface livery devices. Finally, the behavior of the protein
of PVC medical tubing can alter the adsorption of can be altered using formulation approaches to mini-
blood proteins and platelets, leading to thrombus mize adsorption losses, aggregation, and particle for-
formation.96,97 Similarly, adsorption of Factor VIII mation at interfaces (e.g., by reducing propensity for
onto PVC infusion bags reduced its activity by 42%.98 adsorption).
There are different types of i.v. bag materials avail- Exposure to the air–water interface during agita-
able that can be assessed in product compatibility tion is a common cause of protein aggregation. As
studies, which are performed by biotechnology compa- this interface is ubiquitous, a formulation approach to
nies to address the International Conference on Har- minimize its potential effects on stability is often uti-
monization M4Q guideline, which states that: “The lized. Addition of nonionic surfactant such as polysor-
compatibility of the drug product with reconstitution bate 20 or 80 appears to be a general and powerful
diluent(s) or dosage devices (e.g., precipitation of drug strategy for protecting proteins against adsorption-
substance in solution, sorption on injection vessels, induced aggregation. Polysorbate can also reduce ag-
stability) should be addressed to provide appropriate gregation caused by adsorption to other hydropho-
and supportive information for the labeling.”99 Many bic surfaces. Where addition of polysorbate is not
products contain specific instructions about handling practical or possible (such as before diafiltration), ad-
and delivery to ensure product quality and stabil- justment of equipment types and/or operational pro-
ity when delivered to the patient. For example, one cedures and settings could reduce protein particle
brand of antihemophilic factor comes with its own sy- formation and aggregation due to large interfacial
ringe and tubing for administration and the product area compressions, bubble entrainment, cavitation,
label specifies that the product be delivered within or surface adsorption/desorption enhanced by shear.
3 h of reconstitution. The label also notes that the Other excipients or solution conditions can also be
product “. . .contains polysorbate-80, which is known used to modulate the interfacial behavior of proteins.
to increase the rate of di-(2-ethylhexyl)phthalate
(DEHP) extraction from polyvinyl chloride (PVC).”100
Again, in this case, rather than eliminating the ad- REFERENCES
verse effects on protein stability, addition of polysor-
1. Rosenberg AS. 2006. Effects of protein aggregates: An
bate may, counter intuitively, potentially exacerbate immunologic perspective. AAPS J 8(3):E501–E507.
extractions of leachables from PVC containing bags. 2. Schellekens H. 2002. Immunogenicity of therapeutic pro-
In this case, it appears that this potential compatibil- teins: Clinical implications and future prospects. Clin Ther
ity issue was resolved by eliminating the use of PVC. 24(11):1720–1740.
3. Singh SK, Afonina N, Awwad M, Bechtold-Peters K, Blue
Of course many of these types of problem are pro-
JT, Chou D, Cromwell M, Krause H-J, Mahler H-C, Meyer
tein and/or formulation specific and might not be nec- BK, Narhi L, Nesta DP, Spitznagel T. 2010. An indus-
essary or appropriate for other products. The use of try perspective on the monitoring of subvisible particles as
DEHP has been limited or eliminated in many phar- a quality attribute for protein therapeutics. J Pharm Sci
maceutical products.41 99(8):3302–3321.
4. Singh SK. 2010. Impact of product-related factors on im-
munogenicity of biotherapeutics. J Pharm Sci 100:354–387.
5. Randolph TW, Carpenter JF. 2007. Engineering challenges
of protein formulations. AIChE J 53(8):1902–1907.
CONCLUSIONS
6. Chi EY, Krishnan S, Randolph TW, Carpenter JF. 2003.
Robust control of the manufacturing processes and Physical stability of proteins in aqueous solution: Mechanism
and driving forces in nonnative protein aggregation. Pharm
unit operations as well as formulation approaches
Res 20(9):1325–1336.
can often be used to effectively mitigate adverse ef- 7. Weiss WF, Young TM, Roberts CJ. 2009. Principles, ap-
fects of interfaces and leachables on protein stability. proaches, and challenges for predicting protein aggregation
For leachables, identification of the molecular species rates and shelf-life. J Pharm Sci 98(4):1246–1277.

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 10, OCTOBER 2011
4168 BEE ET AL.

8. Roberts CJ. 2007. Non-native protein aggregation kinetics. 30. Page C, Dawson P, Woollacott D, Thorpe R, Mire-Sluis A.
Biotechnol Bioeng 98(5):927–938. 2000. Development of a lyophilization formulation that pre-
9. Manning MC, Patel K, Borchardt RT. 1989. Stability of pro- serves the biological activity of the platelet-inducing cytokine
tein pharmaceuticals. Pharm Res 6(11):903–918. interleukin-11 at low concentrations. J Pharm Pharmacol
10. Manning MC, Chou DK, Murphy BM, Payne RW, Katayama 52(1):19–26.
DS. 2010. Stability of protein pharmaceuticals: an update. 31. Maa YF, Hsu CC. 1998. Investigation on fouling mechanisms
Pharm Res 27(4):544–575. for recombinant human growth hormone sterile filtration.
11. Wang W, Singh S, Zeng DL, King K, Nema S. 2007. An- J Pharm Sci 87(7):808–812.
tibody structure, instability, and formulation. J Pharm Sci 32. Bee JS, Chiu D, Sawicki S, Stevenson JL, Chatterjee K,
96(1):1–26. Freund E, Carpenter JF, Randolph TW. 2009. Monoclonal
12. Shire SJ, Shahrokh Z, Liu J. 2004. Challenges in the devel- antibody interactions with micro- and nanoparticles: Adsorp-
opment of high protein concentration formulations. J Pharm tion, aggregation, and accelerated stress studies. J Pharm Sci
Sci 93(6):1390–1402. 98(9):3218–3238.
13. Nakanishi K, Sakiyama T, Imamura K. 2001. On the ad- 33. Tyagi AK, Randolph TW, Dong A, Maloney KM, Hitscherich
sorption of proteins on solid surfaces, a common but very C, Carpenter JF. 2009. IgG particle formation during filling
complicated phenomenon. J Biosci Bioeng 91(3):233–244. pump operation: A case study of heterogeneous nucleation on
14. Norde W. 2008. My voyage of discovery to proteins in flatland stainless steel nanoparticles. J Pharm Sci 98(1):94–104.
. . .and beyond. Colloids and Surf B: Biointerfaces 61(1):1–9. 34. Salinas BA, Sathish HA, Shah AU, Carpenter JF,
15. Haynes CA, Norde W. 1994. Globular proteins at solid/liquid Randolph TW. 2010. Buffer-dependent fragmentation of a
interfaces. Colloids and Surf B: Biointerfaces 2(6):517–566. humanized full-length monoclonal antibody. J Pharm Sci
16. Norde W, Favier JP. 1992. Structure of adsorbed and des- 99(7):2962–2974.
orbed proteins. Colloids Surf 64(1):87–93. 35. Biddlecombe JG, Craig AV, Zhang H, Uddin S, Mulot S,
17. van Reis R, Zydney A. 2007. Bioprocess membrane technol- Fish BC, Bracewell DG. 2007. Determining antibody stabil-
ogy. J Membr Sci 297(1-2):16–50. ity: Creation of solid-liquid interfacial effects within a high
18. Narendranathan TJ, Dunnill P. 1982. The effect of shear on shear environment. Biotechnol Prog 23(5):1218–1222.
globular proteins during ultrafiltration: Studies of alcohol 36. Lam XM, Yang JY, Cleland JL. 1997. Antioxidants for pre-
dehydrogenase. Biotechnol Bioeng 24:2103–2107. vention of methionine oxidation in recombinant monoclonal
19. Bee JS, Stevenson JL, Mehta B, Svitel J, Pollastrini J, Platz antibody HER2. J Pharm Sci 86(11):1250–1255.
R, Freund E, Carpenter JF, Randolph TW. 2009. Response 37. Zhou S, Zhang B, Sturm E, Teagarden DL, Schöneich C,
of a concentrated monoclonal antibody formulation to high Kolhe P, Lewis LM, Muralidhara BK, Singh SK. 2010.
shear. Biotechnol Bioeng 103(5):936–943. Comparative evaluation of disodium edetate and diethylene-
20. Bodalo A, Gomez JL, Gomez E, Maximo MF, Montiel MC. triaminepentaacetic acid as iron chelators to prevent metal-
2004. Study of l-aminoacylase deactivation in an ultrafiltra- catalyzed destabilization of a therapeutic monoclonal anti-
tion membrane reactor. Enzyme Microb Technol 35(2–3):261– body. J Pharm Sci 99(10):4239–4250.
266. 38. Bee JS, Davis M, Freund E, Carpenter JF, Randolph TW.
21. Mahler H-C, Huber F, Kishore RSK, Reindl J, Rückert P, 2009. Aggregation of a monoclonal antibody induced by ad-
Müller R. 2010. Adsorption behavior of a surfactant and a sorption to stainless steel. Biotechnol Bioeng 105(1):121–
monoclonal antibody to sterilizing-grade filters. J Pharm Sci 129.
99(6):2620–2627. 39. Cromwell M, Hilario E, Jacobson F. 2006. Protein aggrega-
22. Rosenberg E, Hepbildikler S, Kuhne W, Winter G. 2009. tion and bioprocessing. AAPS J 8(3):E572–E579.
Ultrafiltration concentration of monoclonal antibody solu- 40. Wakankar AA, Wang YJ, Canova-Davis E, Ma S, Schmalzing
tions: Development of an optimized method minimizing ag- D, Grieco J, Milby T, Reynolds T, Mazzarella K, Hoff E,
gregation. J Membr Sci 342(1–2):50–59. Gomez S, Martin-Moe S. 2009. On developing a process
23. Patapoff TW, Esue O. 2009. Polysorbate 20 prevents the pre- for conducting extractable-leachable assessment of compo-
cipitation of a monoclonal antibody during shear. Pharm Dev nents used for storage of biopharmaceuticals. J Pharm Sci
Technol, 14(6):659–664. 99(5):2209–2218.
24. Mahler HC, Printz M, Kopf R, Schuller R, Muller R. 2008. Be- 41. Sacha GA, Saffel-Clemmer W, Abram K, Akers MJ. 2010.
haviour of polysorbate 20 during dialysis, concentration and Practical fundamentals of glass, rubber, and plactic sterile
filtration using membrane separation techniques. J Pharm packaging systems. Pharm Dev Technol 15(1):6–34.
Sci 97(2):764–774. 42. Ennis R, Pritchard R, Nakamura C, Coulon M, Yang T, Visor
25. Bhatnagar BS, Bogner RH, Pikal MJ. 2007. Protein stability G, Lee W. 2001. Glass vials for small volume parenterals:
during freezing: Separation of stresses and mechanisms of Influence of drug and manufacturing processes on glass de-
protein stabilization. Pharm Dev Technol 12(5):505–523. lamination. Pharm Dev Technol 6(3):393–405.
26. Kueltzo LA, Wang W, Randolph TW, Carpenter JF. 2008. 43. Iacocca RG, Allgeier M. 2007. Corrosive attack of glass by a
Effects of solution conditions, processing parameters, and pharmaceutical compound. J Mater Sci 42(3):801–811.
container materials on aggregation of a monoclonal antibody 44. Schwarzenbach MS, Reimann P, Thommen V, Hegner M,
during freeze-thawing. J Pharm Sci 97(5):1801–1812. Mumenthaler M, Schwob J, Guntherodt HJ. 2004. Topolog-
27. Schwegman JJ, Carpenter JF, Nail SL. 2009. Evidence of ical structure and chemical composition of inner surfaces
partial unfolding of proteins at the ice/freeze-concentrate in- of borosilicate vials. PDA J Pharm Sci Technol 58(3):169–
terface by infrared microscopy. J Pharm Sci 98(9):3239–3246. 175.
28. Krielgaard L, Jones LS, Randolph TW, Frokjaer S, Flink 45. Amgen and Centocor Ortho Biotech Products LP. 2010. Am-
JM, Manning MC, Carpenter JF. 1998. Effect of tween 20 on gen initiates voluntary nationwide recall of certain lots of
freeze-thawing- and agitation-induced aggregation of recom- Epogen R
and Procrit R
(epoetin alfa). United States Food
binant human factor XIII. J Pharm Sci 87(12):1593–1603. and Drug Administration. Accessed at: www.fda.gov un-
29. Kerwin BA, Heller MC, Levin SH, Randolph TW. 1998. der Home>Safety> Recalls, Market Withdrawals, & Safety
Effects of tween 80 and sucrose on acute short-term sta- Alerts. Sept. 24, 2010
bility and long-term storage at −20◦ C of a recombinant 46. Markovic I. 2010. PDA/FDA Extractables and Leachables
hemoglobin. J Pharm Sci 87(9):1062–1068. Workshop. Washington District of Columbia.

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 10, OCTOBER 2011 DOI 10.1002/jps
SURFACES AND LEACHABLES EFFECTS ON BIOPHARMACEUTICALS 4169

47. Doran PM. 2006. Loss of secreted antibody from transgenic agitation-induced aggregation of a monoclonal antibody in
plant tissue cultures due to surface adsorption. J Biotechnol aqueous solution. J Pharm Sci 98(9):3167–3181.
122(1):39–54. 66. Ludwig DB, Carpenter JF, Hamel J-B, Randolph TW. 2010.
48. Chi EY, Weickmann J, Carpenter JF, Manning MC, Protein adsorption and excipient effects on kinetic stability
Randolph TW. 2005. Heterogeneous nucleation-controlled of silicone oil emulsions. J Pharm Sci 99(4):1721–1733.
particulate formation of recombinant human platelet- 67. Lubiniecki A, Volkin DB, Federici M, Bond MD, Nedved ML,
activating factor acetylhydrolase in pharmaceutical formu- Hendricks L, Mehndiratta P, Bruner M, Burman S, DalMonte
lation. J Pharm Sci 94(2):256–274. P, Kline J, Ni A, Panek ME, Pikounis B, Powers G, Vafa O,
49. Bee JS. 2009. Effect of interfaces and shear on therapeu- Siegel R. 2011. Comparability assessments of process and
tic protein stability. Ph.D Thesis. University of Colorado. product changes made during development of two different
www.proquest.com. monoclonal antibodies. Biologicals 39(1):9–22.
50. Hoehne M, Samuel F, Dong A, Wurth C, Mahler H-C, 68. Henson AF, Mitchell JR, Mussellwhite PR. 1970. The surface
Carpenter JF, Randolph TW. 2010. Adsorption of monoclonal coagulation of proteins during shaking. J Colloid Interface Sci
antibodies to glass microparticles. J Pharm Sci 100:123–132. 32(1):162–165.
51. Boven K, Stryker S, Knight J, Thomas A, van Regenmortel 69. Mahler H-C, Muller R, Friess W, Delille A, Matheus S.
M, Kemeny DM, Power D, Rossert J, Casadevall NE. 2005. 2005. Induction and analysis of aggregates in a liquid IgG1-
The increased incidence of pure red cell aplasia with an Eprex antibody formulation. Eur J Pharm Biopharm 59(3):407–417.
formulation in uncoated rubber stopper syringes. Kidney Int 70. Kiese S, Papppenberger A, Friess W, Mahler H-C. 2008.
67(6):2346–2353. Shaken, not stirred: Mechanical stress testing of an IgG1
52. Sharma B. 2007. Immunogenicity of therapeutic proteins. antibody. J Pharm Sci 97(10):4347–4366.
Part 2: Impact of container closures. Biotechnol Adv 71. Maa YF, Hsu CC. 1997. Protein denaturation by combined
25(3):318–324. effect of shear and air–liquid interface. Biotechnol Bioeng
53. Ryan MH, Heavner GA, Brigham-Burke M, McMahon F, 54(6):503–512.
Shanahan MF, Gunturi SR, Sharma B, Farrell FX. 2006. 72. Bee JS, Schwartz DK, Freund E, Carpenter JF, Randolph
An in vivo model to assess factors that may stimulate the TW. 2010. “DO NOT SHAKE”: Protein particle formation
generation of an immune reaction to erythropoietin. Int Im- caused by compression of the air-water interface. 2010 Work-
munopharmacol Use of Immune Globulin Intraven Dermatol shop on Protein Aggregation and Immunogenicity. Brecken-
6(4):647–655. ridge, CO.
54. Mueller R, Karle A, Vogt A, Kropshofer H, Ross A, Maeder 73. Ying W, Burton L, Gandhi M, Witmer M, Zhou R, Palm
K, Mahler H-C. 2009. Evaluation of the immuno-stimulatory T. 2009. Agitation-induced protein particulate formation
potential of stopper extractables and leachables by using den- and dissociation: A case study of heterogeneous nucleation
dritic cells as readout. J Pharm Sci 98(10):3548–3561. and thermodynamic stability of protein particulate matter
55. Hermeling S, Schellekens H, Crommelin DJA, Jiskoot W. (poster session). Breckenridge, Colorado: Protein Stability
2003. Micelle-associated protein in epoetin formulations: A Conference.
risk factor for immunogenicity? Pharm Res 20(12):1903– 74. Kiese S, Pappenberger A, Friess W, Mahler H-C. 2010.
1907. Equilibrium studies of protein aggregates and homogeneous
56. Villalobos AP, Gunturi SR, Heavner GA. 2005. Interaction of nucleation in protein formulation. J Pharm Sci 99(2):632–
polysorbate 80 with erythropoietin: A case study in protein– 644.
surfactant interactions. Pharm Res 22:1186–1194. 75. Randolph TW, Jones LS. 2002. Surfactant–protein interac-
57. Sluzky V, Klibanov AM, Langer R. 1992. Mechanism of tions. In Carpenter JF, Manning MC, Eds. Rational design
insulin aggregation and stabilization in agitated aqueous- of stable protein formulations. New York: Kluwer Academic/
solutions. Biotechnol Bioeng 40(8):895–903. Plenum Publishers.
58. Joshi O, McGuire J, Wang DQ. 2008. Adsorption and function 76. Treuheit MJ, Kosky AA, Brems DN. 2002. Inverse relation-
of recombinant factor VIII at solid–water interfaces in the ship of protein concentration and aggregation. Pharm Res
presence of Tween-80. J Pharm Sci 97(11):4741–4755. 19(4):511–516.
59. Liu W, Swift R, Torraca G, Nashed-Samuel Y, Wen Z-Q, Jiang 77. Mahler H-C, Senner F, Maeder K, Mueller R. 2009. Surface
Y, Vance A, Mire-Sluis A, Freund E, Davis J, Narhi L. 2010. activity of a monoclonal antibody. J Pharm Sci 98(12):4525–
Root cause analysis of tungsten-induced protein aggregation 4533.
in pre-filled syringes PDA. J Pharm Sci Technol 64(1):11–19. 78. Mahler H-C, Müller R, Friess W, Delille A, Matheus S.
60. Bee JS, Nelson SA, Freund E, Carpenter JF, Randolph TW. 2005. Induction and analysis of aggregates in a liquid IgG1-
2009. Precipitation of a monoclonal antibody by soluble tung- antibody formulation. Eur J Pharm Biopharm 59(3):407–417.
sten. J Pharm Sci 98(9):3290–3301. 79. Daugherty AL, Mrsny RJ. 2006. Formulation and delivery
61. Jiang Y, Nashed-Samuel Y, Li C, Liu W, Pollastrini J, issues for monoclonal antibody therapeutics. Adv Drug Deliv
Mallard D, Wen Z-Q, Fujimori K, Pallitto M, Donahue L, Rev 58(5-6):686–706.
Chu G, Torraca G, Vance A, Mire-Sluis T, Freund E, Davis 80. Bam NB, Cleland JL, Yang J, Manning MC, Carpenter JF,
J, Narhi L. 2009. Tungsten-induced protein aggregation: Kelley RF, Randolph TW. 1998. Tween protects recombinant
Solution behavior. J Pharm Sci 98(12):4695–4710. human growth hormone against agitation-induced damage
62. Bernstein RK. 1987. Clouding and deactivation of clear (reg- via hydrophobic interactions. J Pharm Sci 87(12):1554–1559.
ular) human insulin—Association with silicone oil from dis- 81. Chou DK, Krishnamurthy R, Randolph TW, Carpenter JF,
posable syringes. Diabetes Care 10(6):786–787. Manning MC. 2005. Effects of Tween 20 (R) and Tween 80
63. Chantelau E, Berger M, Bohlken B. 1986. Silicone oil (R) on the stability of albutropin during agitation. J Pharm
released from disposable insulin syringes. Diabetes Care Sci 94(6):1368–1381.
9(6):672–673. 82. Deechongkit S, Wen J, Narhi LO, Jiang Y, Park SS, Kim
64. Jones LS, Kaufmann A, Middaugh CR. 2005. Silicone oil J, Kerwin BA. 2009. Physical and biophysical effects of
induced aggregation of proteins. J Pharm Sci 94(4):918– polysorbate 20 and 80 on darbepoetin alfa. J Pharm Sci
927. 98(9):3200–3217.
65. Thirumangalathu R, Krishnan S, Ricci MS, Brems DN, 83. Brych SR, Gokarn YR, Hultgen H, Stevenson RJ, Rajan
Randolph TW, Carpenter JF. 2009. Silicone oil- and R, Matsumura M. 2010. Characterization of antibody

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 10, OCTOBER 2011
4170 BEE ET AL.

aggregation: Role of buried, unpaired cysteines in particle 92. Tavornvipas S, Tajiri S, Hirayama F, Arima H, Uekama
formation. J Pharm Sci 99(2):764–781. K. 2004. Effects of hydrophilic cyclodextrins on aggrega-
84. Nielsen L, Khurana R, Coats A, Frokjaer S, Brange J, Vyas S, tion of recombinant human growth hormone. Pharm Res
Uversky VN, Fink AL. 2001. Effect of environmental factors 21(12):2369–2376.
on the kinetics of insulin fibril formation: Elucidation of the 93. Hawe A, Friess W. 2007. Stabilization of a hydrophobic re-
molecular mechanism. Biochem 40(20):6036–6046. combinant cytokine by human serum albumin. J Pharm Sci
85. Wang W, Wang YJ, Wang DQ. 2008. Dual effects of Tween 80 96(11):2987–2999.
on protein stability. Int J Pharm 347(1–2):31–38. 94. Hawe A, Friess W. 2007. Formulation development for
86. Kerwin BA. 2008. Polysorbates 20 and 80 used in the formu- hydrophobic therapeutic proteins. Pharm Dev Technol
lation of protein biotherapeutics: Structure and degradation 12(3):223–237.
pathways. J Pharm Sci 97(8):2924–2935. 95. Tzannis ST, Hrushesky WJM, Wood PA, Przybycien TM.
87. Kishore RSK, Pappenberger A, Dauphin IB, Ross A, Buergi B, 1997. Adsorption of a formulated protein on a drug delivery
Staempfli A, Mahler H-C. 2010. Degradation of polysorbates device surface. J Colloid Interface Sci 189(2):216–228.
20 and 80: Studies on thermal autoxidation and hydrolysis. 96. Kim SW, Petersen RV, Lee ES. 1976. Effect of phthalate plas-
J Pharm Sci 100:721–731. ticizer on blood compatibility of polyvinyl chloride. J Pharm
88. Fesinmeyer RM, Hogan S, Saluja A, Brych SR, Kras E, Narhi Sci 65(5):670–673.
LO, Brems DN, Gokarn YR. 2009. Effect of ions on agitation- 97. Lamba NMK, Courtney JM, Gaylor JDS, Lowe GDO. 2000.
and temperature-induced aggregation reactions of antibod- In vitro investigation of the blood response to medical grade
ies. Pharm Res 26(4):903–913. PVC and the effect of heparin on the blood response. Bioma-
89. Lakshmanan M, Dhathathreyan A, Miller R. 2008. Synergy terials 21(1):89–96.
between Hofmeister effect and coupled water in proteins: 98. Mcleod AG, Walker IR, Zheng S, Hayward CPM. 2000. Loss
Unusual dilational moduli of BSA at air/solution interface. of factor VIII activity during storage in PVC containers due
Colloids Surf A-Physicochem Eng Aspects 324(1–3):194–201. to adsorption. Haemophilia 6(2):89–92.
90. Samra HS, He F, Bhambhani A, Pipkin JD, Zimmerer R, 99. ICH. 2004. The common technical document for the registra-
Joshi SB, Middaugh CR. 2010. The effects of substituted cy- tion of pharmaceuticals for human use: quality—M4Q(R1).
clodextrins on the colloidal and conformational stability of ICH Harmonized Tripartite Guideline. International Confer-
selected proteins. J Pharm Sci 99(6):2800–2818. ence on Harmonisation of Technical Requirements for Regit-
91. Serno T, Carpenter JF, Randolph TW, Winter G. 2009. ration of Pharmaceuticals for Human Use. www.ich.org.
Inhibition of agitation-induced aggregation of an IgG- 100. Wyeth, Pharmaceuticals, Inc. 2007. ReFacto R
(antihe-
antibody by hydroxypropyl-beta-cyclodextrin. J Pharm Sci mophilic factor, recombinant). FDAProduct Label Insert.
99(3):1193–1206. Available at: www.fda.gov.

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 10, OCTOBER 2011 DOI 10.1002/jps

You might also like