Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

REVIEWS

Forced Degradation of Therapeutic Proteins


ANDREA HAWE,1,2 MICHAEL WIGGENHORN,2 MARCO VAN DE WEERT,3 JOERG H. O. GARBE,4
HANNS-CHRISTIAN MAHLER,5 WIM JISKOOT1
1
Division of Drug Delivery Technology, Leiden/Amsterdam Center for Drug Research, 2300 RA Leiden, The Netherlands
2
Coriolis Pharma, Am Klopferspitz 19, 82152 Martinsried-Munich, Germany
3
Department of Pharmaceutics and Analytical Chemistry, Faculty of Pharmaceutical Sciences, University of Copenhagen,
Universitetsparken 2, 2100 Copenhagen, Denmark
4
Analytical Development & Quality Control, Pharma Technical Development Biologics Europe, F. Hoffmann- La Roche Ltd.,
Basel, Switzerland
5
Pharmaceutical & Device Development, Pharma Technical Development Biologics Europe, F. Hoffmann- La Roche Ltd.,
Basel, Switzerland

Received 4 January 2011; revised 17 October 2011; accepted 18 October 2011


Published online 14 November 2011 in Wiley Online Library (wileyonlinelibrary.com). DOI 10.1002/jps.22812

ABSTRACT: The scope of this paper is to review approaches used for forced degradation (syn-
onym, stress testing) of therapeutic proteins. Forced degradation studies play a central role in
the development of therapeutic proteins, for example, for candidate selection, molecule char-
acterization, formulation development, assay development, and comparability studies. Typical
stress methods are addressed within this review, such as exposure to elevated temperatures,
freeze–thawing, mechanical stress, oxidation, light, as well as various materials and devices
used in the clinics during final administration. Stability testing is briefly described as far as
relevant to the discussion of forced degradation studies. Whereas stability-testing requirements
are defined in regulatory guidelines, standard procedures for forced degradation of therapeutic
proteins are largely unavailable, except for photostability. Possible selection criteria to identify
appropriate stress conditions and recommendations for setting up forced degradation studies
for the different phases of development of therapeutic proteins are presented. © 2011 Wiley
Periodicals, Inc. and the American Pharmacists Association J Pharm Sci 101:895–913, 2012
Keywords: Comparability studies; Forced degradation (stress testing); Analysis; Deamida-
tion; Oxidation; Protein aggregation; Protein formulation; Protein structure; Proteins Stability

INTRODUCTION an acceptable level of impurities, stable and robust


formulations can be reasonably designed to preserve
Therapeutic proteins have acquired a key role in
this state until the final administration to the patient.
the treatment of various diseases such as diabetes,
The robustness of the formulations against external
several types of cancer, and inflammatory diseases.
stress factors is especially important, because thera-
Side effects, immunogenicity, and allergic reactions
peutic proteins are exposed to various types of stress
have been caused when administering rather impure
during production, fill and finish, shipment, stor-
preparations of plasma-derived immunoglobulins,1,2
age, and final administration.6,7 Among these stress
factor VIII,3 and growth hormone from pituitary
factors are temperature changes, elevated tempera-
glands of deceased individuals.4 To prevent such in-
ture, freezing, thawing, mechanical stress, light expo-
cidents, a strong awareness to strive for highly pure
sure, or interaction with specific components derived
and stable protein products has emerged. High purity
from production equipment, primary packaging ma-
begins with production and downstream processing of
terial, or administration devices used, all of which can
therapeutic proteins.5 Only if the bulk material has
potentially impede stability (Table 1).
The scope of our review is to provide an overview
Correspondence to: Wim Jiskoot (Telephone: +31(0)715274314;
Fax: +31(0)715274565; E-mail: w.jiskoot@lacdr.leidenuniv.nl) of forced degradation (stress-testing) methods that
Journal of Pharmaceutical Sciences, Vol. 101, 895–913 (2012) are commonly used during the development of thera-
© 2011 Wiley Periodicals, Inc. and the American Pharmacists Association peutic proteins. Stability testing is briefly addressed,

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 3, MARCH 2012 895


896 HAWE ET AL.

Table 1. Stress Factors a Protein Can Encounter During Real-Life Conditions

Stress Factor Encountered (During Real-Life Conditions)


Elevated temperature Production; improper shipment, storage or handling or deviations; specific production processes (e.g.,
spray-drying)
Freezing, freeze–thawing Accidental freezing during storage or shipment; storage of frozen (bulk) material; lyophilization
Mechanical stress Production (pumping, filtration, stirring, and so on); shipment; handling (e.g., shaking)
Light Exposure to daylight or artificial light during production, shipment, storage or handling; UV detection
during downstream processing
Oxidative stress Contact with oxygen (air, dissolved O2 ); excipients, for example, peroxide impurities in polysorbate;
metal ion traces from production equipment or excipients; light; vaporized sanitation agents, for
example, vaporized hydrogen peroxide; cavitation
pH changes Production (downstream processing); freezing; formulation; dilution in infusion liquids
Interfaces Air–water interface; filters; primary packaging material, for example, vials, stoppers, syringes; infusion
bags and administration lines; silicone oil; extraneous particles
X-ray Air freight transportation

because it is closely related to stress testing at ele- phase of development, with a brief overview given in
vated temperatures. Within the review, we use the Table 2.
following definitions: According to ICH Q5C,9 forced degradation studies
in general can help to (1) assess whether acciden-
r Forced degradation (or stress testing): an um- tal (or intended) exposure to conditions other than
brella term covering all forms of applying stress those proposed, for example, during transportation or
to drug substance or drug product exceeding the storage, is deleterious to the product and (2) evaluate
conditions used for stability testing; test parameters (analytical methods) as indicators of
r Stability testing: refers to studies performed to product stability.
assess the stability of a formulation according to For accelerated testing, ICH Q5C9 states that this
the international requirements, in particular In- may (1) provide useful data for establishing shelf life,
ternational Conference on Harmonisation (ICH) (2) provide product stability information for future
guidelines ICH Q1A8 (for drugs in general) and product development, for example, preliminary as-
ICH Q5C9 (specific for biotech products); and sessment of proposed manufacturing changes, such as
r Accelerated testing: stability testing at elevated changes in formulation or scale-up, (3) assist in setup
temperatures under quiescent conditions, ac- and validation of analytical methods for the stabil-
cording to ICH Q1A8 and ICH Q5C.9 ity program, and (4) generate information that may
help elucidating the degradation profile of the drug
Within our review, we discuss the relevance of substance or drug product.
forced degradation data for product development, the During formulation development, forced degrada-
importance of suitable analytical methodology, and tion studies are essential for the identification of sta-
factors that could influence the results of these stud- ble and robust formulations within a reasonably short
ies. Test conditions for studying the effect of stress fac- time frame. This requires the assumption that the
tors such as temperature, freeze–thawing, mechan- degradation pathways under the applied stress con-
ical stress, oxidative stress, as well as exposure to ditions, for example, at elevated temperature, corre-
light and various materials on protein stability are late back to the intended storage conditions, which is,
outlined. Because of its importance for successful however, not always the case. During formulation de-
product development, we also address compatibility velopment, the stability of various formulations dur-
testing of therapeutic proteins at simulated final ad- ing forced degradation conditions is compared, more
ministration conditions. Finally, we attempt to pro- recently also in high throughput formats.10–12 Using
vide recommendations and potential selection criteria a combination of stress conditions is of great impor-
for setting up forced degradation studies for therapeu- tance to identify overall robust formulations, as a
tic proteins. certain excipient might be either stabilizing or desta-
bilizing, depending on the stress factor. One exam-
ple is polysorbate, which can increase the stability of
RELEVANCE OF FORCED DEGRADATION
proteins under agitation conditions, but may lead to
STUDIES FOR THE DEVELOPMENT
the formation of soluble aggregates and protein ox-
OF PROTEIN THERAPEUTICS
idation under quiescent storage at elevated or even
Forced degradation studies are an integral part of the at the intended storage temperature, especially when
development of therapeutic proteins. Their purpose used at low and suboptimal concentrations.13,14 To
may vary depending on the application area and the ensure protein stability during clinical development

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 3, MARCH 2012 DOI 10.1002/jps
FORCED DEGRADATION OF THERAPEUTIC PROTEINS 897

Table 2. Relevance of Forced Degradation Studies for the Different Phases of Development, Including Possible Recommendations for
Selecting Suitable Conditions

Application Area Purpose of Study Recommendations


Candidate selection • Select most suitable molecule from a set of • Combine different types of stress factors to gain a
potential candidates broad insight into several aspects of stability
Molecule characterization • Elucidate degradation products, pathways, • Combine different types of stress factors to gain a
and mechanisms broad insight into several aspects of stability
• Obtain information about stability of a protein
against various stress factors
Formulation development • Identify optimum formulation conditions • Include a combination of relevant
• Test the robustness of a formulation (product-dependent) stress factors (e.g.,
temperature, mechanical stress, light stress,
freeze–thawing stress)

Assay development and • Prepare degradation products standards to • Choose the stress condition(s) that create(s) the
validation develop and validate analytical methods degradation product of choice for the particular
• Assess stability-indicating properties of method
analytical methods
Shelf life setting • Establish accelerated degradation profile in • This will mainly be testing at elevated
comparison to intended storage temperature; the temperature needs to be
• Predict shelf life selected based on possible reaction kinetics that
could be applied

Exposure to conditions • Support evaluation of accidental (or intended) • Select conditions the protein is expected to be in
other than those proposed exposure, for example, by temperature contact with during production, storage, and
excursion studies, freezing studies, and administration
interaction studies (diluents, container
materials)
Future product • Studies to compare stability behavior of • See formulation development
development/line product, for example, after changes in the
extensions production process (or formulation)
• Collect product stability information

and for commercialization, shipment studies, in-use ucts that are detected by the particular analytical
studies, and temperature excursion evaluations are method(s) of interest. For instance, chemically de-
important. Results from accelerated testing can be graded products such as oxidized and deamidated
used, with certain limitations, for shelf life prediction species may be needed for reversed-phase HPLC
of a formulation at 2–8◦ C, as discussed later. analysis intended for the analysis of these impuri-
Forced degradation studies go hand in hand with ties; for size-exclusion HPLC, the level of aggregates
analytical characterization, because the outcome of and/or fragments should be increased. In any case,
these studies depends on not only the conditions ap- these degradation products should be generated in
plied but also the analytical methods included to as- a controlled manner by exposing the protein to var-
sess stability. Appropriate analytical techniques that ious stress conditions. Information on the stability
are capable of detecting and quantifying the different of purified degradation products is crucially impor-
degradation products formed are essential. As differ- tant if these are routinely used as controls,for ex-
ent types of degradation products can be formed upon ample, for system suitability testing over a longer
applying stress to a certain protein15,16 and because time period. Moreover, the potential effect of sample
of the complexity of proteins, a combination of comple- preparation, for example, dilution, and the analyti-
mentary analytical methods is required to cover the cal methods themselves, for example, protein–column
formed degradation products as comprehensively as interactions in chromatographic separations, on the
possible.17–20 An overview of selected analytical meth- (measured) level of degradation, should be carefully
ods that can be used to characterize different types of evaluated.20–22
degradation products is given in Table 3 and is fur- Upon changes in the manufacturing process of the
ther discussed in the “Recommendations and Criteria marketed product or during development, such as
for Setting Up Forced Degradation Studies” section. scale-up, process or cell line changes, the potential
Vice versa, stressing proteins is essential for set- impact of the change on product quality needs to be
ting up analytical methods and assessing their sta- assessed. Typically, such comparability exercises in-
bility—indicating properties (see Table 2). For this clude the analytical characterization of the product
purpose, it is important to create degradation prod- including the analysis of process- and product-related

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 3, MARCH 2012
898 HAWE ET AL.

Table 3. Selection of Analytical Methods to Assess Different Types of Degradation Products

Type of Degradation Selected Analytical Methods


Soluble aggregates (dimers, trimers, oligomers) Size-exclusion HPLC, AF4, AUC, electrophoretic techniques (SDS-PAGE,
and fragments capillary electrophoresis)
Subvisible aggregates, nanometer size range DLS, NTA, AF4, TDA, turbidity/nephelometry, SLS, MALLS
Subvisible aggregates, micrometer size range Light obscuration, flow microscopy techniques (e.g., microflow imaging, flow cam),
Coulter counter, light microscopy, fluorescence microscopy, SLS, MALLS,
turbidity/nephelometry
Visible particles Visual inspection
Secondary structure Far-UV CD spectroscopy, infrared spectroscopy, Raman spectroscopy
Tertiary structure Near-UV CD spectroscopy, intrinsic fluorescence spectroscopy, second derivative
UV spectroscopy, nuclear magnetic resonance
Changes in hydrophobicity HIC, reversed-phase HPLC, extrinsic fluorescent dyes
Chemical changes (e.g., oxidation, deamidation) (HPLC-)mass spectrometry, peptide mapping, reversed-phase HPLC,
ion-exchange HPLC, IEF (gel system or capillary IEF)

Abbreviations: AF4, asymmetrical flow field flow fractionation; AUC, analytical ultracentrifugation; CD, circular dichroism; DLS, dynamic light scattering;
HIC, hydrophobic-interaction chromatography; IEF, isoelectric focusing; MALLS, multiangle laser light scattering; NTA, nanoparticle tracking analysis; SLS,
static light scattering; and TDA, Taylor dispersion analysis.

impurities. Furthermore, the stability of the pre- and pH range should be evaluated.8 ICH Q5C mentions
postchange product(s) is compared by storage at real- several stress factors a product may be sensitive to,
time and accelerated storage conditions and based on which are temperature changes, oxidation, light, ionic
forced degradation studies. If substantial differences strength, and shear9 ; however, detailed instructions
are found during analysis with a potential impact on on how to test the influence of these stress factors on
the efficacy or safety of the drug product, further stud- stability are not available in the guideline.
ies may be necessary such as nonclinical or clinical The American Society for Testing and Materi-
studies. The comparability assessment is typically set als (ASTM) provides guidance on testing shipping
up case–by case, depending on the product, its mode containers and products in shipping containers in
of action, and the type of change made, for exam- general.26,27 This information could be used as guid-
ple, during the manufacturing process. Accelerated ance for mechanical stress testing, as discussed later.
and stress-stability studies (ICH Q1A8 and Q5C9 ) are Overall, the lack of predefined standard conditions
often used to establish degradation profiles and pro- for forced degradation studies in the pharmaceutical
vide a further direct comparison of prechange and field leaves it to each company/laboratory to identify
postchange product (ICH Q5E23 ). suitable forced degradation setups for the protein of
interest. Possible approaches range from performing
forced degradation according to standard procedures
WHICH STANDARD CONDITIONS FOR FORCED defined within the company to setting up stress condi-
DEGRADATION CAN BE FOUND? tions case–by case for each molecule,24 also depending
In contrast to stability testing, which is well regulated on the intended purpose (Table 2).
in the guidelines ICH Q1A8 and ICH Q5C,9 hardly any
standard procedures for forced degradation of thera-
peutic proteins are available. Within the guidelines OVERVIEW OF METHODS FOR FORCED
a rough concept on forced degradation studies is out- DEGRADATION STUDIES
lined, but information on how and to which extent
Thermal Stress
these studies should be performed are not provided.24
An exception is photostability testing, where condi- Proteins can be exposed to temperatures higher than
tions are defined in the ICH guideline Q1B.25 Never- the recommended storage temperature during pro-
theless, for biotechnological products, it is proposed duction, shipment, storage, and final administration.
to consider testing conditions others than those de- Elevated temperature is the most widespread method
scribed in ICH guideline Q1B for photostability test- to stress and degrade therapeutic proteins, which
ing on a case-by-case basis and to consult the regula- are typically stored under refrigerated conditions
tory authorities (ICH Q5C).9 (2–8◦ C).
With respect to forced degradation, the general With increasing temperature, proteins may un-
ICH guideline on stability testing (Q1A) recommends dergo conformational changes such as unfolding or
studying the effect of temperature, humidity where partial unfolding. These changes may subsequently
appropriate, that is, for dried preparations, oxida- lead to other degradation reactions, including aggre-
tion, and photolysis on the drug. Furthermore, the gation. Starting from the onset temperature of un-
susceptibility of the drug to hydrolysis across a wide folding (Tonset ) toward the melting temperature (Tm )

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 3, MARCH 2012 DOI 10.1002/jps
FORCED DEGRADATION OF THERAPEUTIC PROTEINS 899

of the protein, a gradual destabilization of the native involving both Fab and Fc.34 Furthermore, the ag-
protein structure becomes apparent,28–30 which often gregation pathway and kinetics of thermally stressed
leads to (irreversible) aggregation. At the same time, IgGs can vary per IgG type46 and is formulation de-
diffusion becomes faster at higher temperature, re- pendent.
sulting in more energetic collisions with other protein In some cases, the Tm has been used to predict sta-
molecules, as well as with reactive chemicals, thereby bility of liquid formulations at the anticipated storage
favoring both aggregation and chemical degradation conditions. Some examples in the literature suggest
reactions. Moreover, chemical degradation reactions a correlation between the Tm and storage stability,
may be altered in the unfolded state, for example, if which allowed identifying more stable formulations
buried amino acids become accessible for the chemical for these cases.32,33,47–50 In general, however, Tm mea-
reaction in solution. surements cannot replace stability testing under real-
When setting up the conditions for thermal stress- time and real-temperature conditions. For example, if
ing, the parameters temperature, duration, and op- the predominant degradation process or rate-limited
tionally temperature fluctuations need to be estab- step is not related with (partial) unfolding of the pro-
lished. The protein’s Tonset and Tm as measures of tein, the Tm will not be predictive for stability.51,52
the temperature-dependent conformational stability Besides the Tm , the reversibility of the unfolding re-
can be used as rough guidance for selecting an ap- action (i.e., the relative extent of refolding upon low-
propriate stress temperature for isothermal incuba- ering the temperature below the Tm ) can be used to
tion. The Tm is clearly defined as the temperature predict the stability of some proteins at temperatures
where half of the protein molecules are unfolded dur- below the Tm , as shown for the recombinant human
ing a thermal scan, assuming a fully reversible un- Flt3 ligand.53
folding process,31 whereas the Tonset as the onset Numerous attempts have been made to predict the
temperature of unfolding is less well defined. Differ- shelf life of peptide and protein formulations based on
ential scanning calorimetry,31–33 circular dichroism extrapolation of stability data from elevated temper-
(CD),34 FTIR,35 and intrinsic36 and extrinsic37 fluo- ature to real-time storage conditions. This would help
rescence spectroscopy can be used to determine the to speed up product development and to avoid waiting
Tm and Tonset of a protein. Thermal unfolding can for real-time/real-temperature data. Obviously, this
be reversible like in the case of lysozyme, which un- approach has a major caveat: In all cases, the inves-
dergoes a simple apparent two-state transition un- tigator needs to consider whether the testing setup
der most formulation conditions.38 For the majority is really predictive for long-term storage under real-
of proteins, thermal unfolding is irreversible, because time conditions. This would be the case for degra-
it is accompanied or followed by aggregation, as in dation reactions following specific reaction kinetics
the case of interferon-"2a.39 Larger, multidomain pro- with predictable increased reaction rates under the
teins exhibit several distinct, in part overlapping un- accelerated condition, such as (pseudo) first-order re-
folding transitions corresponding to unfolding of the actions following the Arrhenius equation (Eq. 1):
different domains. For immunoglobulin G (IgG), Fab
and Fc have been shown to denature independently
kobs = Ae−Ea /RT (1)
at 63–71◦ C and 73–82◦ C, respectively, with the tran-
sition as a whole being irreversible.34,40,41
In the context of forced degradation, it should be where kobs is the degradation rate constant in day−1 ,
noted that unfolded molecules are already present A is the preexponential factor (day−1 ), Ea is the acti-
below the Tm and that the predominant degrada- vation energy in J mol−1 , R is the molar gas constant
tion reaction may depend on the stress temperature. (8.314472 J K−1 mol−1 ), and T is the temperature in
In particular for aggregation this is the case, as a kelvin.
number of thermodynamic and dynamic parameters In principle, individual chemical degradation reac-
contribute to the observed aggregation mechanism tions can largely be described by the Arrhenius equa-
and kinetics.42 This is illustrated by the example tion, allowing a calculation of activation energies and
of the temperature-dependent aggregation behavior half lives, for example, at the intended storage tem-
of IgG.43,44 At temperatures below the first thermal perature. However, if several chemical reactions are
transition (Tm1 at about 63◦ C), slow aggregation rates occurring in parallel, as is often the case in protein
are typically measured,34 as a result of comparably molecules, the applicability may be limited. Arrhe-
low levels of partially unfolded IgG molecules with nius would then only be valid to the individual reac-
aggregation mainly being driven by an association of tions as long as these are not interacting. Examples
the Fab domains.45 Incubation at or above the second of the validity of the Arrhenius equation for chemi-
thermal transition (Tm2 at about 73◦ C) creates higher cal instability are chemical degradation of peptides,
levels of unfolded IgG molecules, leading to more se- for example, deamidation of oxytocin at low pH,54 hy-
vere aggregation and a different aggregate structure drolysis (deamidation) of insulin,55 and Met oxidation

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 3, MARCH 2012
900 HAWE ET AL.

in interleukin-1 receptor antagonist and peptide frac- and shelf life from accelerated data may become pos-
tions thereof.56 sible only if aggregation kinetics under accelerated/
However, the majority of complex degradation thermal stress and real-time conditions are qualita-
and aggregation processes in solution exhibit non- tively controlled by the same physicochemical factors,
Arrhenius behavior, by definition when the reaction for example, formation of a conformationally changed
does not follow (pseudo) first-order kinetics.57–59 Re- intermediate or a chemical change as starting point
combinant factor VIII SQ (r-VIII SQ) degradation for aggregation.42 Therefore, it is important to elu-
followed pseudo-first-order kinetics, with nonlinear cidate degradation mechanisms at thermal stress
Arrhenius plots, which would lead to an overestima- conditions and compare them to those occurring at
tion of shelf life at 2–8◦ C based on the data gath- the intended storage condition, in most cases 2–8◦ C.
ered between 10 and 37◦ C, when assuming linearity.57 The assessment of degradation kinetics can also help
An example of the successful prediction of the shelf comparing degradation pathways for comparability
life for non-Arrhenius-type aggregation is that of re- exercises.
combinant bovine granulocyte colony stimulating fac-
Incubation in the Frozen State and Freeze–Thawing
tor, where extended Lumry–Eyring models enabled
shelf life prediction from stability data from tem- Isothermal incubation in the frozen state,
peratures between 30 and 47◦ C.58 Another exam- freeze–freeze experiments, and freeze–thawing
ple is that of the denaturation of $ galactosidase in experiments are useful to (i) test the stability of
solution59 or aggregation of monoclonal antibodies in frozen bulk drug substance or early (pre-)clinical
liquid formulations.60 drug products, (ii) assess the robustness of a product
Not only temperature but other factors like pH, intended for refrigerated storage (2–8◦ C) against
which can also change with temperature, excipients accidental freezing, and (iii) to identify excipients
or protein concentration, may alter degradation rates that stabilize during freezing (also as step in
and mechanisms.61 Additional factors that may com- lyophilization) or frozen storage.
plicate extrapolation approaches include (i) the in- During freezing and thawing, proteins are exposed
fluence of surface-induced processes on the aggrega- to a combination of stress factors,62 including in-
tion kinetics, (ii) aggregation processes involving a terfacial stresses (protein/ice surface),63–66 tempera-
lag time prior to a highly variable onset of severe ag- ture fluctuations,67 cryoconcentration,64 crystalliza-
gregation, (iii) acceleration of aggregation processes tion of excipients,68–70 phase separation,71,72 and pH
due to the presence of aggregate species function- shifts.65,73–78 The relative importance of each of these
ing as nuclei, (iv) temperature-dependent conforma- factors with respect to protein instability depends
tional changes leading to altered reaction kinetics, on the protein, the formulation, and the freeze–thaw
and (v) inherent heterogeneity of the pharmaceutical process, and thus needs to be evaluated on a case-
protein.42 For a detailed overview of principles and ap- by-case basis. For several proteins, it has been de-
proaches for predicting protein aggregation rates and scribed that freeze–thawing may induce the forma-
shelf life, the interested reader is referred to Weiss tion of larger aggregates and proteinaceous particles,
et al.42 often characterized by a high degree of native-like
A suitable temperature for thermal stress testing protein structure.63,65,66,78–81
needs to be selected on a case-by-case approach. For Isothermal incubation of a frozen protein formula-
a drug product intended for storage at 2–8◦ C, ac- tion at temperatures between–70◦ C, and the thawing
celerated testing is generally performed at 25◦ C, as temperature of the product can in some cases provide
laid down in ICH Q1A.8 At the same time, the influ- suitable stability information and elucidate the influ-
ence of heat is suggested being tested in increments ence of cryoconcentration and excipient crystalliza-
of 10◦ C above the accelerated testing temperature,8 tion on stability. Freeze–freeze studies (temperature
which might in some cases be too extreme for sen- excursions in the frozen state) can simulate the influ-
sitive proteins. As a rule of thumb, one should stay ence of temperature fluctuations in the frozen state on
below Tonset and at least 10–20◦ C below the Tm dur- the product relevant, for example, when a bulk prod-
ing thermal stress testing. At or above the Tm , severe uct is stored at–20◦ C and is shipped at lower tem-
aggregation often impedes useful conclusions on sta- peratures. However, the short-term stability of for-
bility. Moreover, as the protein will be largely unfolded mulations at, for example,–20◦ C will not necessarily
under these conditions, degradation products may be correlate with the stability at lower temperatures
formed that are less relevant to “real-life” conditions. during long-term storage. For instance, the effect
When setting up provisional shelf life (expiration of excipient crystallization and cryoconcentration
dating) during clinical phases, data from accelerated is rather temperature specific, rendering stabil-
testing and thermal stress testing can be used in con- ity extrapolations to other temperatures a signifi-
nection with real-time/real-temperature data. How- cant challenge.68 Nevertheless, incubation studies in
ever, the prediction of real-time aggregation rates the frozen state at temperatures other than those

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 3, MARCH 2012 DOI 10.1002/jps
FORCED DEGRADATION OF THERAPEUTIC PROTEINS 901

intended for storage, as well as freeze–freeze studies perature profile in the container are typically differ-
may be useful to test for and/or explain instability as ent from the externally applied cooling rate used to
a result of fluctuating temperatures, which can occur freeze the formulation. The resulting freezing rate of
during shipment or storage of frozen drug substance. the product depends on various factors, such as heat
The scale of the experiments is a critical param- transfer to the product, filling volume, container, and
eter, as freezing behavior at large scale is highly formulation composition,. Fast freezing, for example,
different from small scale.82,83 Small volumes (e.g., quench cooling in liquid nitrogen or by placing small
1–5 mL) can be homogeneously frozen within min- volume vials in a freezer at–70◦ C, leads to the for-
utes at–20 or–80◦ C. Freezing of larger volumes as mation of many small ice crystals and consequently
typically applied to storage of drug substance may a large interfacial area. Depending on the sensitiv-
require several hours72,82,83 and may result in a ity of the studied protein toward surface-induced de-
heterogeneous frozen product with respect to pro- naturation, this may be detrimental for stability. The
tein and excipient concentration, pH, and osmolar- thawing protocol with respect to thawing rate, thaw-
ity. To elucidate such cryoconcentration effects, one ing temperature, and agitation can also influence the
can collect samples from various positions in the stability of the formulations. For the model proteins,
container of the frozen formulation, or of the liquid lactate dehydrogenase, alcohol dehydrogenase, and
formulation directly after quiescent thawing, prior catalase, the highest activity recovery and stability
to homogenizing.68,72,75 In most cases, a significant has been found upon slow freezing (<1◦ C/min) in com-
increase in protein and excipient concentration to- bination with a fast thawing process (>10◦ C/min),
ward the middle and the bottom of the container has as the relative surface area was lowest under these
been found in larger containers.68,72,75 Extrapolation conditions.86
from small-scale freezing experiments to large scale In the context of formulation studies, freeze–thaw-
is therefore not necessarily possible. ing experiments aim to assist selecting the formu-
Freezing studies can be carried out to inves- lations that are stable against accidental freezing
tigate excipient crystallization at low tempera- or suitable for lyophilization. Freeze–thawing exper-
tures. Excipients such as trehalose,68,69 sorbitol,70 iments are often performed by placing the formula-
or mannitol,84,85 and buffer salts65,73–78 can crystal- tions in small containers (1–10 mL, downscale model)
lize in an uncontrolled manner during frozen stor- into a freezer (mainly–20 or–80◦ C),87 freezing the
age, potentially compromising stability. Sugar crys- formulations in liquid nitrogen,88 or on the shelf of
tallization mainly occurs when the formulations are a freeze-drier at a controlled cooling rate. Homoge-
stored above the glass transition temperature of the neous freezing behavior and limited cryoconcentra-
maximally freeze-concentrated solutions (Tg  ). For in- tion effects are expected for such small volumes, as
stance, trehalose was found to crystallize above its discussed above.
Tg  of–29◦ C during frozen storage, which resulted in An illustrative example of the complex interrela-
a slow increase in aggregation of a monoclonal anti- tion of various parameters on protein stability dur-
body stored at–20◦ C.68 ing freeze–thawing has been published by Kueltzo
During freeze–thawing studies, a large number of et al.64 In this study, the influence of pH, ionic
process- and formulation-related variables can influ- strength, protein concentration, cooling and thawing
ence protein stability. Process-related variables in- rates, container type and container material on ag-
clude the cooling rate, temperature set at start and gregation, and structure of an IgG2 was investigated.
end of the freezing process, container geometry, and At pH 4, the protein at relatively low concentration
filling volume, which will all influence the actual (0.25 mg/mL) was more prone to aggregation dur-
freezing rate of the formulation. Formulation-related ing freeze–thawing than at a higher concentration
factors can be among others protein concentration, (10 mg/mL), pointing at surface-induced denatura-
pH, and excipients. All of these parameters can have tion as a predominant mechanism. The opposite was
an effect on the freezing behavior and ice formation, found at pH 3, where perturbations of the tertiary
which is a nucleation-governed process and thus typ- structure in solution and not surface-induced denat-
ically heterogeneous and difficult to reproduce. De- uration were identified as the cause of aggregation.
gree of supercooling and ice crystallization rate are In addition, the variation in monomer recovery and
in general the most important factors controlling the aggregation levels in formulations freeze–thawed in
amount and size of the formed ice crystals and the containers differing in material (glass, polyethylene,
dimension of the interfacial area between ice and TeflonTM ) and geometry (vials, FlexboyTM bag) il-
freeze–concentrated solution.62 lustrate that the container surface can additionally
Cooling rates used to freeze the studied formula- change the outcome of the freeze–thawing studies.64
tions can be classified as slow (<1◦ C/min), interme- How to decide on suitable conditions for
diate (∼5◦ C/min), and fast (10–900◦ C/min).62 Note freeze–thawing experiments, incubation at low tem-
that the actual freezing rate and the resulting tem- perature or freeze–freeze studies? In the simplest

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 3, MARCH 2012
902 HAWE ET AL.

case, the placement of vials filled with the liquid depends on the experimental setup, the sensitivity of
formulation into a freezer might be enough to gain the protein against mechanical stress, and the formu-
insight into the general robustness of the protein lation.
against freezing, without specifying freezing or thaw- Proteins are exposed to shear during processing
ing rates. However, freezing and thawing conditions steps, such as pumping, filtration, or mixing. In most
are preferably specified and controlled, for example, cases, shear itself was not a major factor with respect
by using a cooling/heating bath or a piece of equip- to aggregation induction, even at very high shear
ment to achieve adequate process control, especially rates of 250,000 s−1 .113,115,116 For typical processing
when applying these conditions routinely during for- steps such as pumping or filtration maximum shear
mulation screening for a comparative purpose. Also rates of about 20,000 s−1 (shear force of 0.06 pN) have
in the field of lyophilization process control during been estimated.116 To unfold proteins, shear forces
freezing, for example, by using a defined cooling rate between 20 and 150 pN are required, which would
of the shelves in the freeze-drier is important. In both equal shear rates of up to 5 × 107 s−1 . In compari-
cases, the resulting freezing and thawing rates within son, upon contact with air–liquid interfaces, proteins
the formulation could, for example, be measured by are exposed to forces of about 140 pN, clearly in the
thermocouples placed within the containers. By us- required range for unfolding.116 These estimations
ing multiple freeze–thawing cycles during formula- confirm the low aggregation propensity of proteins in
tion screening, the damage caused to the protein can pure shear experiments and point toward other fac-
be exacerbated, enabling a better discrimination be- tors than shear being responsible for unfolding and
tween poor and stable formulations.87,88 aggregation. Adsorption and unfolding of protein at
The scale of the experiment is crucially important liquid–air and liquid–solid interfaces, enhanced re-
in all cases, because of the discussed differences in newal rate of the air–liquid interface, cavitation, air
freezing time, heterogeneity after freezing, as well as bubble entrainment, and contamination with parti-
crystallization effects. To study the influence of excip- cles or leachables from shearing devices have been
ient crystallization on protein stability, both incuba- proposed as causative factors for aggregation and in-
tion above and below the Tg  should be carried out. Ex- stability during mechanical stress testing.111,114,116
cipient crystallization often cannot be predicted easily The occurrence of minor conformational changes in-
as it is considered to be a statistical event more likely duced by shear stress has been reported only in
to happen in larger volume.69,70 Therefore, it is sug- few cases, for example, for lysozyme110 and rhGH.113
gested to assess nucleation of excipients in the frozen Overall, in shear stress experiments, the combina-
state, with models that use seeding of freezing nucle- tion of various stress factors associated with apply-
ation in small scale to induce crystallization.89 Next to ing shear appears to be critical. The relative contri-
methods for protein stability testing, physicochemical bution of these factors will strongly depend on the
characterization of the excipients’ morphology, for ex- experimental conditions used in mechanical stress
ample, by differential scanning calorimetry and X-ray studies, including parameters such as container
diffraction, is required. material, container geometry, and interface/volume
ratio.
Mechanical Stress
The extent of the air–liquid interfaces created dur-
As therapeutic protein formulations are exposed to ing mechanical stress testing experiments has a huge
mechanical stress during manufacturing, transport, impact on the outcome of mechanical stress studies.
and final administration, it is crucial to test their Upon contact of a protein with air–liquid interfaces,
robustness against these stress factors. Mechanical (partial) unfolding followed by irreversible aggrega-
stress testing is often performed to evaluate the sta- tion and/or particle formation can occur.6,7,18 Con-
bilizing properties of excipients, such as surfactants sequently, one can expect more severe aggregation
added to formulations with the intention of reducing during agitation of vials with air–headspace as com-
agitation- and surface-induced aggregation.14,90 Me- pared with agitation of fully filled vials.94 Also vari-
chanical stress testing can be performed as agitation ations in the dimension of the liquid–air interface,
(shaking)14,90–97 or stirring studies,94,95,98–101 but also for example, by using different filling volumes or vial
pumping,5,102–104 vortexing,105–107 sonication,108,109 sizes, can alter the extent of aggregation or particle
and special shearing devices110–114 have been used for formation.92,94 Furthermore, parameters such as the
this purpose. velocity of the agitation movements (mainly specified
During mechanical stress testing, proteins expe- in rounds per minutes, rpm), the type of movement,
rience several types of stress factors, such as shear- the placement of the vials in the agitation device, tem-
ing, exposure to liquid–air and liquid–container inter- perature, and duration of the agitation stress have an
faces, cavitation, and local warming effects,18 which influence on the outcome of the stress studies. Foam-
can result in physical and chemical instability. The ing as a consequence of protein unfolding during me-
relevance of the different stresses on protein stability chanical stress testing is another factor influencing

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 3, MARCH 2012 DOI 10.1002/jps
FORCED DEGRADATION OF THERAPEUTIC PROTEINS 903

the properties of the air–liquid interface and was re-

Shaking: 1-mL filling volume, 120 shakes per minute Duration: 70 h

with best stability discrimination: 1-mL protein solution in 2-mL

-coated stirring
Shaking: various filling volumes; 150, 200, and 250 rpm conditions

magnetic vanes Stirring (improved): 30 min at 50 rpm followed


ported to potentially induce oxidation.117

200 rpm at 9-mL filling volume without a headspace Shaking:


Shaking: 0.6-mL filing volume; 350 rpm with vials horizontally
The chosen experimental setup for mechanical

-coated stirring bars at

Stirring (conventional): 30 min at 50 rpm with bottom sitting


Shaking: 4-mL filling volume, 150 rpm horizontally shaking
stress testing depends on the phase of development

2.5-, 5.3-, and 9-mL filling volume at 200 rpm with vials
and the aim of the study. A selection of conditions
and experimental setups of mechanical stress testing

Stirring: 2-mL filling volume, triangular Teflon R

by 15 min at 150 rpm with top-entering stirrer)


found in the literature is provided in Table 4. Stirring
and pumping experiments can be relevant to evaluate
the influence of a manufacturing unit operation, for

Conditions
example, mixing or filling, on protein stability during

horizontally placed Duration: 168 h


Stirring: with 6 mm × 15 mm Teflon R
process development.5,99,100 In that case, the param-

vials at 200 rpm Duration: 120 h


eters should be selected to be a representative of the

bar, 600 rpm Duration: 48 h


manufacturing step of interest.
Agitation (shaking) studies can be performed dur-

placed Duration: 24 h
ing formulation development as a rapid screening test
to discriminate poor from good formulations with re-
spect to their stabilizing power against mechanical
stress. This is in particular relevant during surfac-
tant screening to identify the optimum surfactant
type and concentration. Stirring, which is generally
even harsher than shaking,94,95 may be used for this
purpose as well.
For the simulation of transport-related stress,

6-mL Fiolax vials (shaking)


Examples of Mechanical Stress-Testing Conditions for Therapeutic Proteins Found in the Literature

2-mL polypropylene vials


ASTM provides guidance on testing shipping contain-

and 3-mL Reacti vials


0.8-, 1.5-, 2.0-, and 6-mL
3-mL glass type I vials

6-mL glass type I vials


ers in general.26,27 These include performance test

Stainless steel tanks


Container

glass type I vials


schedules and conditions for testing the integrity of
filled containers in typical shipping environments,
for example, air, rail, or motor transport.26,27 These

(stirring)

(2–10 L)
guidelines are primarily intended for testing the ef-
fect of the applied stresses on the integrity of the con-
tainer itself, and not on protein stability. Nonetheless,
the ASTM guidelines on transport can provide an in- 2–8◦ C and 25◦ C
formative basis on the extent of mechanical exposure
Temperature

a product is subjected to during transport and pos-


sible conditions to simulate this. Suitable equipment
Ambient

Ambient

Ambient

Ambient
to perform studies according to the ASTM guidelines
23◦ C

is available, for example, vibration tables from vari-


ous manufactures,118 which can be applied to testing
the integrity of proteins upon exposure to transport-
Interleukin-2 mutein (5 mg/mL)

(concentration 2–5 mg/mL)

related mechanical stress. Alternatively, mock ship-


Monoclonal IgG1 (10 mg/mL)
Recombinant human growth

Monoclonal IgG (2 mg/mL)

ments can be considered and may include analysis of


Several monoclonal IgGs

product integrity.
hormone (5 mg/mL)
Protein

Exposure to Different Materials


Monoclonal IgG1
(10 mg/mL)

Proteins come into contact with various materials/


surfaces such as glass, rubber, or metal during pro-
duction (pumps, tubings, vessels, chromatography
columns, filters, containers), storage (primary pack-
aging material), and administration to the patient
Stirring and shaking93,94

(infusion bags, infusion systems, syringes). Moreover,


Stirring and shaking95

the outcome of forced degradation studies may largely


depend on the container material used to perform
these studies.
Type of Study

Exposure of protein to materials may cause aggre-


Stirring100
Shaking14

Shaking90

Shaking92
Table 4.

gation, particle formation, and other instability re-


actions such as oxidation, if a protein is sensitive
to a certain type of material or leachables thereof.

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 3, MARCH 2012
904 HAWE ET AL.

For example, it has been reported that nonproteina- Contact of a protein with other soluble impurities,
ceous particulates, for example, stainless steel parti- for example, tungsten from pins used for the manu-
cles from pumps, may cause aggregation in therapeu- facturing of certain prefilled syringes,130 iron leaching
tic protein formulations.102,119 Adsorption of protein from stainless steel surfaces,131 or impurities present
to the steel surface, as well as oxidizing effects of met- in excipients,133,134 has also been reported to result
als such as chromium oxide or leaching iron ions from in aggregation. By spiking formulations of two thera-
steel, has been found to potentially trigger aggrega- peutic proteins with different levels and types of sol-
tion and particle formation.102,119,120 Moreover, ex- uble tungsten species, it was suggested that various
tractables and leachables from filters have been found tungsten compounds (WO3 , H2 WO4 , Na2 WO4 , and an
to destabilize IgG2 formulations during agitation and extract from tungsten pins) typically present in pre-
temperatures stress studies.121 For a detailed review filled syringes can induce the formation of aggregates
on the influence of various surfaces and leachables on and particles at very low pH (e.g., pH 4) and with sen-
aggregation, the reader is referred to the review by sitive proteins, such as the "-helical protein used in
Bee et al.122 the study.130
In this section, the intentional exposure of the pro- Compatibility of proteins with solid surfaces, for
tein to different materials, for example, in form of example, of different primary packaging or container
spiking studies, at concentrations and/or surface ar- materials, can be tested during accelerated and real-
eas much higher than expected under real-life con- time stability studies of the actual product. Condi-
ditions, is considered as a stress method. Spiking tions of testing need to be carefully evaluated in a
studies can be useful to test the potential of mate- case-by-case approach. In a study by Majumdar et al.,
rial surfaces to provoke protein instability. Note that the effect of siliconized and modified syringe surfaces
elucidating the compatibility of a protein with con- on the stability of different proteins (among others
tainer materials is closely related to, but not the a protein with known sensitivity toward silicone oil)
same as, forced degradation studies in the context was assessed under quiescent storage at 2–8◦ C and
of this review. To study container compatibility, pro- 25◦ C, as well as agitation after storage.135 As another
tein adsorption and/or instability caused by different example, compatibility with stainless steel surfaces
materials such as glass,123,124 polypropylene,123,124 can be evaluated in downscaled stainless steel mini
polystyrene,125 Teflon,97 and stainless steel126 have tanks, in which the protein formulation is stored at
been studied. various temperature conditions for specified and rel-
To clarify the importance of various surface mate- evant time frames. The surface area-to-volume ratio
rials with respect to protein stability, several types in downscaled systems is usually different than the
of studies can be envisaged. Spiking of formulations ratio expected in real life, and calculations of these
with the material of choice, for example, glass, metal, ratios should be part of setting up such studies. Prior
cellulose microparticles,119 silicone oil,127–129 or solu- to this, the compatibility with stainless steel surfaces
ble metal ions (tungsten130 or iron131 ) has been de- in general can also be tested using steel chips.
scribed. In most of these spiking studies, the sur- In summary, spiking studies with different materi-
face area of particles or silicone oil droplets added als can be part of formulation screening to identify the
to the protein formulations exceeds the surface area robustness of formulations toward contact with impu-
a protein is expected to encounter in real life. For rities such as silicone oil or tungsten. The amount and
instance, microparticles with a total surface area of type of material spiked to the formulation should be
60–200 cm2 /mL have been added to a monoclonal selected case by case. With regard to testing compati-
antibody in the context of forced degradation stud- bility of a protein formulation with silicone oil, it may
ies, whereas the surface area of glass vials or sy- also be advisable to test protein stability in the speci-
ringes is only about 7 cm2 in total.119 Protein stabil- fied prefilled syringes (including silicone oil at previ-
ity has been tested upon adding silicone oil in con- ously specified ranges) over the shelf life at real time/
centrations between 0.5 and 2.0% (w/v) to protein real temperature, as well as accelerated temperature
formulations.127–129 Although exposure to silicone oil conditions.
is relevant as protein comes into contact with silicone
Oxidation Stress
in prefilled syringes and silicone may also leach into
the product over time, the amount typically present Studying oxidation of proteins is very relevant in
in prefilled syringes is far less (about 300–400 :g for formulation development, as oxidation products may
a 1-mL syringe) than in these studies. Silicone oil it- show altered bioactivity and conformation.136 Protein
self was found to cause little damage to the protein oxidation can be caused by peroxides present as im-
during quiescent storage, but did accelerate agitation- purities in excipients (e.g., polysorbates), metal ions
induced aggregation.129 However, silicone oil is not from stainless steel surfaces or present as impuri-
always detrimental to proteins, as was shown for a ties in excipients, and residues from aseptic agents
fusion protein of albumin and interferon "-2b.132 (e.g., used in isolators). Oxidation can also be caused

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 3, MARCH 2012 DOI 10.1002/jps
FORCED DEGRADATION OF THERAPEUTIC PROTEINS 905

by light, as discussed later. For a detailed descrip- pending on the temperature used during incubation
tion of oxidation mechanisms relevant to therapeu- with H2 O2 , either hydroxyl radicals or superoxide an-
tic proteins, the interested reader is referred to the ion radicals may be formed.149
literature.137–139 Oxidation stress is often used to cre- Besides H2 O2 , sometimes in combination with
ate degradation products, for example, for assay de- certain metal ions, t-butyl hydroperoxide (t-BHP)
velopment or for elucidating degradation pathways and 2,2 -azo-bis(2-amidinopropane) dihydrochloride
and kinetics. The general ICH guideline Q1A rec- (AAPH) have been used as oxidation agents. For in-
ommends assessing the susceptibility of a drug sub- stance, t-BHP can be an option to oxidize surface-
stance toward oxidation, without further specifying exposed amino acids.140,150 For metal-catalyzed oxi-
the conditions.8 dation combinations of Cu(II), Fe(II), or Fe(III) with
Amino acid residues that are particularly sensitive H2 O2 , O2 and/or ascorbic acid have been used.140 A
to oxidation include His, Met, Cys, Tyr, and Trp, but combination of Cu2+ /ascorbic acid was, for example,
also other amino acids such as Arg, Pro, and Lys can more effective than Fe3+ /ascorbic acid to oxidize Met
be oxidized under certain conditions.6 The relative residues in human relaxin.151 Metal-catalyzed oxida-
distribution of oxidation products is highly dependent tion is relevant, as metals can be introduced into the
on the oxidizer used. A good example is the work by Ji formulation from various sources, for example, during
et al. on parathyroid hormone 1–34.140 They used sev- production, from primary packaging material or from
eral conditions reported for the oxidation of Met, His, excipients. AAPH is not directly an oxidizing agent,
and Trp and found that the various procedures could but generates alkyl radicals that catalyze the forma-
lead to significantly different products and distribu- tion of reactive oxygen species.140
tion of Met, Trp, and His oxidation products even for We recommend that different oxidation protocols
this relatively simple protein. Lou et al. showed that are used, both to generate different oxidized species
metal-catalyzed oxidation of a IgG2 monoclonal anti- and to study the susceptibility of the various amino
body resulted in site-specific oxidation of Met (246), acids in the protein to oxidation. H2 O2 is an excellent
His (304), and His (427) in the Fc portion of the anti- starting point, generally mainly yielding methionine
body, whereas hydrogen peroxide treatment resulted sulfoxide. Addition of metal ions such as Fe(III) or
in oxidation of four solvent-exposed Met.15 Cu(II) alongside H2 O2 can show whether site-specific
Oxidation reactions can be nonsite specific like in oxidation may occur. The products here are likely
the case of photooxidation and oxidation involving more diverse and may include significant amounts
free radicals or reactive oxygen species.6 Site-specific of oxidized Cys, His, and Trp. If potentially radical-
oxidation also involves reactive oxygen species, but forming compounds are known to be used somewhere
mainly occurs when certain metal ions can bind to in the production process or in the formulation itself,
specific sites in the protein, for example, His oxida- it is advisable to also test these compounds (also in
tion in the presence of Cu2+ ions.141,142 higher concentrations than in the process of formula-
One common oxidation agent is H2 O2 and within tion) in combination with H2 O2 . This will then show
the European Pharmacopeia an oxidation procedure whether stability problems may be expected under
by using 0.005% (v/v) H2 O2 for 20 h at 37◦ C is de- normal conditions. To oxidize surface-exposed amino
scribed for interferon-".143 Examples on the use of acids, the use of t-BHP should be considered.
H2 O2 from the literature are numerous,140,144 includ-
Light Stress
ing Met oxidation induced by 1 mM (ca. 0.003%) H2 O2
for recombinant human interleukin-1 receptor56 or by Protein pharmaceuticals are exposed to light in vari-
0.005% (v/v) H2 O2 for recombinant human interferon ous situations. Examples include UV detection during
"-2b.145 By applying mild oxidation conditions (H2 O2 purification steps and exposure to artificial or sun-
at a molar ratio of 80:1 (H2 O2 :protein) for 4 h at light during production, shipment, storage, and ad-
20◦ C) to recombinant human growth hormone, site- ministration of the drug to the patient.118,152 Most
specific oxidation of the relatively accessible Met14 marketed therapeutic proteins are recommended to
and Met125 residues under preservation of the na- be “protected from direct (sun)light” and are sold in
tive conformation has been achieved.146,147 Not only adequate secondary packaging to avoid direct and
the H2 O2 concentration but also the solution pH can long-term light exposure.
influence the degree of oxidation, because normally Light-induced degradation reactions depend on
buried Met residues can become accessible due to pH- protein structure and conformation, formulation com-
induced conformational changes, as shown for human position, and primary packaging, as well as wave-
"-1 antitrypsin.148 The addition of low amounts of length, intensity, and duration of light exposure.
H2 O2 (e.g., 1–100 ppm) and longer incubation peri- The peptide backbone, Cys, and the aromatic amino
ods of several weeks or months can be performed to acids Trp, Tyr, and Phe are structural components
simulate contamination of a formulation with trace that are most sensitive to undergo photodegradation
amounts of H2 O2 used as a sanitization agent. De- upon absorption of light. Furthermore, light-induced

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 3, MARCH 2012
906 HAWE ET AL.

oxidation of proteins can be caused by the forma- be identifying conditions a specific product is exposed
tion of reactive oxygen species or accelerated by the to, for example, during manufacturing, fill and fin-
presence of metal ions, for example, photooxidation ish, and the clinical setup. Parameters such as wave-
of His. Photodegradation primarily leads to chemi- length of the light sources and exposure time dur-
cal changes in single amino acids, but it can also ing manufacturing and administration can be the ba-
lead to multiple cross-linking reactions due to di- sis for designing photostability studies as addition to
tyrosine or disulfide formation and to noncovalent these required by the authorities.118 Proper controls
aggregation.152 An overview of the underlying mech- are of great importance, for example, light-protected
anisms of photodegradation of proteins is given by samples to distinguish between the effect of light and
Kerwin and Remmele.152 potential warming on protein stability or placebo con-
The ICH guideline Q1B specifies the experimen- trols to distinguish between matrix effects and pro-
tal setup for photostability studies, including light tein stability.
sources and extent of light exposure.25,153 As a light
Compatibility with Final Administration Conditions
source, it is possible to use any lamp-producing light
according to emission standard D65 (outdoor day- For the clinical use of a protein drug, it is important to
light) and ID65 (indoor indirect daylight standard) as assure its compatibility with the final administration
first option, or a combination of a cool white fluores- conditions. This includes testing the stability of the
cent lamp with a near-UV lamp as a second option.25 protein after dilution in carrier solutions for infusion
The guideline distinguishes between “forced degrada- and the compatibility with delivery devices such as
tion testing studies,” that is, to estimate the overall tubing and infusion bags where losses due to adsorp-
photosensitivity or elucidate degradation pathways, tion could occur.
with conditions not further specified, and “confirma- Besides adsorption, leachables from infusion bags,
tory testing” to predict the stability. For “confirmatory such as metals, plasticizers, polymerization initiators,
testing,” the product should be exposed to at least antioxidants, and other additives, potentially impede
1.2 million lux hours in the visible range (400–800 nm) protein stability.157–159
and at least 200 W h/m2 in the UV range (320–400 nm).25 The design of these studies depends on the adminis-
Interestingly, this setup leaves two parameters to be tration conditions intended for the protein drug, such
selected by the investigator: irradiation energy per as possible dilution steps in carrier solutions, rate,
unit of time and irradiation time. In most cases, pro- and duration of the infusion, materials used for the
tein degradation under these very harsh conditions infusion, the use of in-line filters, temperature, and
is significant and thus the “confirmatory testing” is the overall contact time with the different materials.
prone to fail, resulting in severe degradation and ag- Many therapeutic proteins are administered as
gregation of the product.118 In this case, the product continuous infusion at room temperature upon dilu-
should be recommended to be protected from direct tion in a carrier solution, usually 0.9% sodium chlo-
light and adequately packaged. In official documents, ride (saline) and in some cases 5% dextrose. By do-
no testing conditions are specified on how to deter- ing so, the protein is generally exposed to a less
mine adequate levels of light that a product should be stabilizing environment, because pH, ionic strength,
able to withstand during manufacture, storage and protein concentration, and temperature differ from
use. the ideal formulation and storage conditions, and ex-
Several therapeutic proteins, for example, recom- cipients/stabilizers are typically significantly diluted.
binant human factor VIII,154 recombinant human This may lead to degradation during handling, stor-
interferon-"2a,155 and a human IgG1 monoclonal age prior to administration, or application of the ther-
antibody,156 were found to be sensitive to light ex- apeutic protein in the diluent to the patient. For in-
posure, resulting in discoloration accompanied by stance, factor VIII exhibited a greater activity loss
activity loss154,156 and the formation of various chem- in 0.9% NaCl than in 5% dextrose,154 whereas dilu-
ical and physical degradation products.155,156 Discol- tion of trastuzumab is recommended in 0.9% NaCl
oration of the solution and loss of activity was mea- and not in dextrose according to the package leaflet.
sured for a monoclonal antibody formulation (100 mg/ This illustrates the importance of determining the
mL formulated with histidine, sorbitol, and polysor- compatibility of a therapeutic protein with different
bate 80 at pH 5.5) after light exposure according to carrier solutions in a case-by-case approach. Other
the ICH conditions (1.2 million lux hours in the visible examples of the compatibility of therapeutic proteins
range and 200 W h/m2 in the range 320–400 nm).156 with diluents are found in the literature, for example,
The extent of light exposure may differ per product, for insulin160 and epoetin ",161,162 where stability is
among others depending on the manufacturing pro- typically monitored at 2–8◦ C and ambient tempera-
cess steps and the equipment used, making a stan- ture.
dardization of photostability testing difficult. A basis Another point to be considered is the loss of protein
for defining conditions for photostability testing can or excipients as a result of adsorption to the infusion

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 3, MARCH 2012 DOI 10.1002/jps
FORCED DEGRADATION OF THERAPEUTIC PROTEINS 907

bag material, in-line filters or tubings, and instability terization of the degradation taking place. In prac-
caused by surface-induced denaturation.154,163–166 In tice, this is not possible, as a result of constraints on
the case of interleukin-2, a reduction in the biological time, finances, and available methodology. The scien-
activity by about 90% within a 24-h infusion program tist must therefore initially select a set of those meth-
could be attributed to a minor degree to pure sur- ods to explore whether any significant degradation
face adsorption, but mainly to irreversible structural takes place. Ideally, at least one method for each type
changes in interleukin-2 during a transient associa- of anticipated class of degradation product should be
tion with the catheter tubings.164 used. Depending on the type of degradation products
To set up studies on testing the compatibility of identified in the first screening, a more in-depth anal-
the protein with the final administration conditions, ysis can be performed. Note that the selection of meth-
a close collaboration with clinicians and clinical phar- ods depends on the forced degradation method, the
macists is advisable. The planned administration pro- protein, and its concentration, and the formulation.
tocol, including a route of administration, conditions For more information about analytical methods, the
for infusion or injection, used materials such as tub- reader is referred to the literature.17–20,167
ing, inline filters, and infusion bags, should be the The chosen formulation conditions with respect to
basis for the study. Once this is defined, the stability pH and ionic strength most likely influence the out-
of the therapeutic protein under these conditions can come of forced degradation studies. For certain ques-
be systematically studied. To distinguish between the tions, the exposure to extreme formulation conditions,
influence of the carrier solution and the dilution itself for example, with respect to pH, ionic strength, or ex-
on stability, it is advisable to include suitable controls, cipient composition/concentration, can be considered
that is, dilution in the formulation buffer. One should as a stress method to address the robustness of a pro-
be aware of potential batch-to-batch variations with tein during processing and formulation. Formulation
respect to leachables from clinical material even if pH, ionic strength,6 and excipients133,134,168,169 can
provided by the same manufacturer, as reported for have an influence on chemical stability, as well as col-
standard polyolfin infusion bags.157 A strategy may loidal stability of a protein.7,18 As an example, the
be to first use raw materials of construction and to exposure of a protein to extreme (acidic or basic) pH
classify and evaluate suitability and compatibility of values could be used to test a protein’s susceptibility
infusion systems in general prior to testing a range of to deamidation.
infusion systems from different manufacturers. Primary packaging material and the scale of the
experiments (meaning the container volume and the
filling volume) may influence protein stability during
RECOMMENDATIONS AND CRITERIA FOR
forced degradation studies. This can be due to ad-
SETTING UP FORCED DEGRADATION STUDIES
sorption of the protein to the container, other surface-
Guidelines on how to perform forced degradation induced effects, compounds leaching from the con-
studies of proteins in the pharmaceutical field are tainer into the formulation, or the influence of fill-
widely lacking, leaving it to the pharmaceutical com- ing volume on the outcome of stress tests (e.g., as
panies or laboratories to setup the conditions for discussed in Freeze–Thawing and Mechanical Stress
stress testing. In any case, the chosen setup should Testing sections). Therefore, it is suggested to per-
be based on the particular question to be addressed form forced degradation studies in the final primary
and the phase of development. One should be very packaging material or the container of interest when
cautious in extrapolating the outcome of stress stud- testing for production-related stress. Depending on
ies to real-life conditions, as the conditions during the stage of development, this is not always feasible,
forced degradation studies generally differ from what in particular in early phases of development when
a protein is expected to encounter during real-life only limited amount of material is available or when
conditions. Thus, forced degradation studies cannot the protein concentration, volume, and final container
replace stability studies under real-time and real- have not yet been defined. In the last example, the
temperature conditions. results from forced degradation studies performed
The analytical characterization of the formed in different containers can help identifying suitable
degradation products forms an integral part of forced primary packaging materials for the formulation of
degradation studies. Here, it is highly recommended interest.
to use orthogonal analytical methods, capable of de- In some cases, it is less critical to downscale forced
tecting and quantifying the degradation products that degradation studies and use sample volumes smaller
may be formed. There are numerous analytical meth- than the final drug product formulations. When the
ods available for the characterization of proteins and aim is creating degradation product standards, the
their degradation products (see Table 3). Ideally, all scale of the experiment and the primary packag-
these methods should be used during forced degra- ing may still affect the characteristics of the degra-
dation studies to assure the most complete charac- dation products obtained, which is, however, less

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 3, MARCH 2012
908 HAWE ET AL.

critical regarding the relevance for the intended us- reasonable time frame. However, too high levels of
age. Also for certain formulation studies, when com- degradation might again impede stability discrimi-
paring the stability of a protein under different pH nation. What is sufficiently high in a reasonable time
or ionic strength conditions in quiescent storage, full- frame? In the context of forced degradation studies of
scale experiments may not necessarily be required. proteins, this should be evaluated in a case-by-case
Bridging experiments, comparing the stability of a approach. The suggestion to create not more than
protein in different primary packaging materials, or 5–15% degradation of the main compound in a reason-
different scales can give insight into the relevance of able time frame (e.g., at most 2 weeks) as described in
these parameters on the outcome of forced degrada- the context of forced degradation studies for small
tion studies. molecules24 could be a rough guidance for protein
Selection criteria and recommendations for forced therapeutics. However, because of the complexity of
degradation studies depend on the aim of the study, degradation products formed in protein formulations
with possible criteria summarized in Table 2. In some this might not be applicable to all types of instability.
cases, for example, when testing the robustness of Subvisible and visible particles formed upon forced
formulations, it is advisable to select conditions that degradation studies typically represent only a minor
are relevant to real-life stresses if a protein is poten- fraction of the overall protein present in the formu-
tially exposed during production, shipment, storage, lation, but are still an important and well-detectable
and final administration. To cover as many potential class of degradation products. The time frame should
degradation mechanisms as possible, a combination of make sense for the selected stress conditions and can
various types of stress conditions should be tested in range from minutes, for example, for vortexing or
separate experiments, for example, temperature, in- shearing studies, to several days or weeks for shaking
terfaces (ice–water, liquid–air, liquid–container) and studies or exposure to elevated temperature.
light-induced stress protocols. The conditions selected should cover potential
To gain insight into product characteristics and/or stress conditions a protein is expected to be exposed to
create degradation products for assay development, during production, storage, transportation, and han-
conditions that are less relevant to real life can still dling. Those can vary for different products, depend-
provide valuable information. Examples can be the ing on the formulation (e.g., liquid or dried), primary
exposure to extreme (both low and high) pH values, packaging (e.g., a prefilled syringe or a vial with head
or oxidation by adding chemicals such as H2 O2 . When space), and administration conditions (e.g., infusion
creating degradation products for assay development, after dilution in a carrier solution or direct injection).
it can be useful to select conditions where a certain
type of degradation product is primarily created, for
CONCLUSIONS
example, oxidized species by H2 O2 or metal-catalyzed
oxidation. Ideally, it should be possible to create differ- Forced degradation studies play a central role in
ent levels of the degradation products in a controlled the development of therapeutic proteins. In contrast
and reproducible way. This may also help assessing with the available guidelines for stability studies,
a potential further relevance of these impurities. As specific guidance for forced degradation studies is
an example, by selective oxidation of specific Met lacking. Stress conditions typically used, among oth-
residues—in the absence of other degradation pro- ers elevated temperature, freeze–thawing, mechan-
cesses—one could create specific degradation prod- ical stress, or light, can induce different types of
ucts and measure bioactivity of these species. When chemical and physical degradation reactions in ther-
degradation products cannot be “selectively gener- apeutic proteins. The prediction of real-time/real-
ated,” it can be considered to purify them for further temperature stability from accelerated testing data
testing. As an example, the isomerization products is with limitations possible, but should be performed
of Asp102 and the deamidation products of Asn30 of with care. In general, forced degradation studies do
trastuzumab were generated by thermal stress and not replace stability testing. Moreover, they are ap-
purified by ion exchange chromatography for bioac- plied to many other purposes than stability testing,
tivity testing.170 Assessing the stability of purified such as candidate selection, molecule characteriza-
degradation products is important, if those will be tion or assay development, and validation.
routinely used as standards/control solutions for an- The analytical characterization of the induced
alytical methods or stored for a longer time prior to degradation products forms an integral part of stress
activity testing. studies. It is of great importance to have suitable an-
During formulation development, the selected alytical techniques available to detect and quantify
stress conditions should most importantly allow dis- degradation products as comprehensively as possible.
crimination of stable and unstable protein formu- By doing so, a thorough understanding of underlying
lations. For this purpose, sufficiently high levels of mechanisms of protein degradation can be generated,
degradation products need to be created within a which provides a basis for finding means of preventing

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 3, MARCH 2012 DOI 10.1002/jps
FORCED DEGRADATION OF THERAPEUTIC PROTEINS 909

those. Overall, forced degradation studies can provide 17. Carpenter JF, Randolph TW, Jiskoot W, Crommelin DJA,
important insights into the sensitivity of a protein to- Middaugh CR, Winter G, Fan YX, Kirshner S, Verthelyi D,
Kozlowski S, Clouse KA, Swann PG, Rosenberg A, Cherney
ward various stress conditions important for the de-
B. 2009. Overlooking subvisible particles in therapeutic pro-
velopment of stable products and user instructions in tein products: Gaps that may compromise product quality. J
package inserts. Pharm Sci 98(4):1201–1205.
18. Mahler HC, Friess W, Grauschopf U, Kiese S. 2009. Pro-
tein aggregation: Pathways, induction factors and analysis.
ACKNOWLEDGMENTS J Pharm Sci 98(9):2909–2934.
19. Philo JS. 2006. Is any measurement method optimal for all
This work was supported, in part, by the Dutch aggregate sizes and types? AAPS J 8(3):E564–E571.
Technology Foundation (Stichting Technische Weten- 20. Philo JS. 2009. A critical review of methods for size charac-
schappen, STW), Applied Science Division of Neder- terization of non-particulate protein aggregates. Curr Pharm
landse Organisatie voor Wetenschappelijk Onderzoek Biotechnol 10(4):359–372.
21. den Engelsman J, Garidel P, Smulders R, Koll H, Smith B,
(NWO), and the Technology Program of the Ministry Bassarab S, Seidl A, Hainzl O, Jiskoot W. 2010. Strategies
of Economic Affairs. The authors thank Connie van for the assessment of protein aggregates in pharmaceutical
Gent for the literature supply and the anonymous re- biotech product development. Pharm Res 28(4):920–933.
viewers for their thoughtful, constructive comments. 22. Carpenter JF, Randolph TW, Jiskoot W, Crommelin DJ,
Middaugh CR, Winter G. 2010. Potential inaccurate quan-
titation and sizing of protein aggregates by size exclusion
REFERENCES chromatography: Essential need to use orthogonal methods
to assure the quality of therapeutic protein products. J Pharm
1. Barandun S, Kistler P, Jeunet F, Isliker H. 1962. Intra- Sci 99(5):2200–2208.
venous administration of human gamma-globulin. Vox Sang 23. Q5E. 2004. Comparability of biotechnological/biological prod-
7:157–174. ucts subject to changes in their manufacturing process. Fed
2. Ellis EF, Henney CS. 1969. Adverse reactions follow- Regis 70(125):37861–37862.
ing administration of human gamma globulin. J Allergy 24. Klick S, Mujselaar PG, Waterval J, Eichinger T, Korn C,
43(1):45–54. Gerding TK, Debets AJ, Sänger-van de Griend C, van den
3. Jacquemin MG, Saint-Remy JMR. 1998. Factor VIII immuno- Beld C, Somsen GW, De Jong GJ. 2005. Toward a generic ap-
genicity. Haemophilia 4(4):552–557. proach for stress testing of drug substance and drug product.
4. Milner RD. 1985. Growth hormone 1985. Br Med J (Clin Res Pharm Technol 29:48–64.
Ed) 291(6509):1593–1594. 25. Q1B. 1997. Stability testing: Photostability testing of new
5. Cromwell ME, Hilario E, Jacobson F. 2006. Protein aggrega- drug substances and products. Fed Regis 62(95):27115–
tion and bioprocessing. AAPS J 8(3):E572–E579. 27122.
6. Manning MC, Chou DK, Murphy BM, Payne RW, Katayama 26. ASTM. 2007. D 999-08: Standard test methods for vibration
DS. 2010. Stability of protein pharmaceuticals: An update. testing of shipping containers. ASTM International.
Pharm Res 27(4):544–575. 27. ASTM. 2008. D 4169-09: Standard practice for performance
7. Wang W, Nema S, Teagarden D. 2010. Protein aggregation— testing of shipping containers and systems. ASTM Interna-
pathways and influencing factors. Int J Pharm 390(2):89– tional.
99. 28. Jaenicke R. 1991. Protein stability and molecular adaptation
8. Q1A(R2). 2003. Stability testing of new drug substances and to extreme conditions. Eur J Biochem 202(3):715–728.
products (second revision). Fed Regis 68(225):65717–65718. 29. Jaenicke R. 2000. Stability and stabilization of globular
9. Q5C. 1996. Quality of biotechnological products: Stabil- proteins in solution. J Biotechnol 79(3):193–203.
ity testing of biotechnological/biological products. Fed Regis 30. Schellman JA. 1997. Temperature, stability, and the
61:36466–36474. hydrophobic interaction. Biophys J 73(6):2960–2964.
10. Capelle MA, Gurny R, Arvinte T. 2007. High throughput 31. Schoen A, Velazquez-Campoy A. 2005. Calorimetry. In Meth-
screening of protein formulation stability: practical consid- ods for structural analysis of protein pharmaceuticals,
erations. Eur J Pharm Biopharm 65(2):131–148. Jiskoot W, Crommelin DJA, Eds. Arlington, VA: AAPS Press,
11. Capelle MA, Gurny R, Arvinte T. 2009. A high throughput pp. 573–589.
protein formulation platform: Case study of Salmon calci- 32. Burton L, Gandhi R, Duke G, Paborji M. 2007. Use of mi-
tonin. Pharm Res 26(1):118–128. crocalorimetry and its correlation with size exclusion chro-
12. Zhao H, Graf O, Milovic N, Luan XS, Bluemel M, Smolny matography for rapid screening of the physical stability
M, Forrer K. 2010. Formulation development of antibodies of large pharmaceutical proteins in solution. Pharm Dev
using robotic system and high-throughput laboratory (HTL). Technol 12(3):265–273.
J Pham Sci 99(5):2279–2294. 33. Remmele RL, Gombotz WR. 2000. Differential scanning
13. Treuheit MJ, Kosky AA, Brems DN. 2002. Inverse relation- calorimetry—A practical tool for elucidating stability of liq-
ship of protein concentration and aggregation. Pharm Res uid biopharmaceuticals. Pharm Tech Eur 12:56–s65.
19(4):511–516. 34. Vermeer AW, Norde W. 2000. The thermal stability of im-
14. Wang W, Wang YJ, Wang DQ. 2008. Dual effects of Tween 80 munoglobulin: unfolding and aggregation of a multi-domain
on protein stability. Int J Pharm 347(1–2):31–38. protein. Biophys J 78(1):394–404.
15. Luo Q, Joubert MK, Stevenson R, Ketchem RR, Narhi LO, 35. Matheus S, Friess W, Mahler HC. 2006. FTIR and nDSC as
Wypych J. 2011. Chemical modifications in therapeutic pro- analytical tools for high-concentration protein formulations.
tein aggregates generated under different stress conditions. Pharm Res 23(6):1350–1363.
J Biol Chem 286(28):25134–25144. 36. Garidel P, Hegyi M, Bassarab S, Weichel M. 2008. A rapid,
16. Joubert MK, Luo Q, Nashed-Samuel Y, Wypych J, Narhi sensitive and economical assessment of monoclonal antibody
LO. 2011. Classification and characterization of therapeutic conformational stability by intrinsic tryptophan fluorescence
antibody aggregates. J Biol Chem 286(28):25118–25133. spectroscopy. Biotechnol J 3(9–10):1201–1211.

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 3, MARCH 2012
910 HAWE ET AL.

37. He F, Hogan S, Latypov RF, Narhi LO, Razinkov VI. 2010. Oxidation of methionine residues in recombinant human
High throughput thermostability screening of monoclonal an- interleukin-1 receptor antagonist: implications of conforma-
tibody formulations. J Pharm Sci 99(4):1707–1720. tional stability on protein oxidation kinetics. Biochemistry
38. Fischer B. 1996. Folding of lysozyme. EXS 75:143–161. 46(21):6213–6224.
39. Sharma VK, Kalonia DS. 2003. Temperature- and pH- 57. Fatouros A, Osterberg T, Mikaelsson M. 1997. Recombinant
induced multiple partially unfolded states of recombinant factor VIII SQ–inactivation kinetics in aqueous solution and
human interferon-alpha2a: Possible implications in protein the influence of disaccharides and sugar alcohols. Pharm Res
stability. Pharm Res 20(11):1721–1729. 14(12):1679–1684.
40. Ionescu RM, Vlasak J, Price C, Kirchmeier M. 2008. Contri- 58. Roberts CJ, Darrington RT, Whitley MB. 2003. Irreversible
bution of variable domains to the stability of humanized IgG1 aggregation of recombinant bovine granulocyte-colony stimu-
monoclonal antibodies. J Pharm Sci 97(4):1414–1426. lating factor (bG-CSF) and implications for predicting protein
41. Vermeer AW, Norde W, van Amerongen A. 2000. The unfold- shelf life. J Pharm Sci 92(5):1095–1111.
ing/denaturation of immunogammaglobulin of isotype 2b and 59. Yoshioka S, Aso Y, Izutsu K, Kojima S. 1994. Is stability pre-
its F(ab) and F(c) fragments. Biophys J 79(4):2150–2154. diction possible for protein drugs—denaturation kinetics of
42. Weiss WFT, Young TM, Roberts CJ. 2009. Principles, ap- beta-galactosidase in solution. Pharm Res 11(12):1721–1725.
proaches, and challenges for predicting protein aggregation 60. Kayser V, Chennamsetty N, Voynov V, Helk B, Forrer K,
rates and shelf life. J Pharm Sci 98(4):1246–1277. Trout BL. 2011. Evaluation of a non-Arrhenius model for
43. McCarthy D, Goddard DH, Embling PH, Holborow EJ. therapeutic monoclonal antibody aggregation. J Pharm Sci
1981. A simple procedure for assessing the stability of 100(7):2526–2542.
heat-aggregated IgG preparations. J Immunol Methods 61. Perico N, Purtell J, Dillon TM, Ricci MS. 2009. Conforma-
41(1):75–79. tional implications of an inversed pH-dependent antibody
44. McCarthy D, Goddard DH, Pell BK, Holborow EJ. 1981. aggregation. J Pharm Sci 98(9):3031–3042.
Intrinsically stable IgG aggregates. J Immunol Methods 62. Bhatnagar BS, Bogner RH, Pikal MJ. 2007. Protein stability
41(1):63–74. during freezing: separation of stresses and mechanisms of
45. McCarthy DA, Drake AF. 1989. Spectroscopic studies on IgG protein stabilization. Pharm Dev Technol 12(5):505–523.
aggregate formation. Mol Immunol 26(9):875–881. 63. Chang BS, Kendrick BS, Carpenter JF. 1996. Surface-
46. Hartmann WK, Saptharishi N, Yang XY, Mitra G, Soman induced denaturation of proteins during freezing and its in-
G. 2004. Characterization and analysis of thermal denatu- hibition by surfactants. J Pharm Sci 85(12):1325–1330.
ration of antibodies by size exclusion high-performance liq- 64. Kueltzo LA, Wang W, Randolph TW, Carpenter JF. 2008.
uid chromatography with quadruple detection. Anal Biochem Effects of solution conditions, processing parameters, and
325(2):227–239. container materials on aggregation of a monoclonal antibody
47. Remmele RL, Jr., Nightlinger NS, Srinivasan S, Gombotz during freeze-thawing. J Pharm Sci 97(5):1801–1812.
WR. 1998. Interleukin-1 receptor (IL-1R) liquid formulation 65. Sarciaux JM, Mansour S, Hageman MJ, Nail SL. 1999. Ef-
development using differential scanning calorimetry. Pharm fects of buffer composition and processing conditions on ag-
Res 15(2):200–208. gregation of bovine IgG during freeze–drying. J Pharm Sci
48. Goldberg DS, Bishop SM, Shah AU, Sathish HA. 2010. For- 88(12):1354–1361.
mulation development of therapeutic monoclonal antibodies 66. Strambini GB, Gabellieri E. 1996. Proteins in frozen solu-
using high-throughput fluorescence and static light scatter- tions: Evidence of ice-induced partial unfolding. Biophys J
ing techniques: Role of conformational and colloidal stability. 70(2):971–976.
J Pharm Sci 100(4)1306–1315. 67. Privalov PL. 1990. Cold denaturation of proteins. Crit Rev
49. Huus K, Havelund S, Olsen HB, van de Weert M, Frokjaer Biochem Mol Biol 25(4):281–305.
S. 2006. Chemical and thermal stability of insulin: Effects of 68. Singh SK, Kolhe P, Mehta AP, Chico SC, Lary AL, Huang M.
zinc and ligand binding to the insulin zinc-hexamer. Pharm 2011. Frozen state storage instability of a monoclonal anti-
Res 23(11):2611–2620. body: Aggregation as a consequence of trehalose crystalliza-
50. Singh S, Singh J. 2003. Effect of polyols on the conformational tion and protein unfolding. Pharm Res 28(4):873–885.
stability and biological activity of a model protein lysozyme. 69. Sundaramurthi P, Suryanarayanan R. 2010. Trehalose crys-
AAPS PharmSciTech 4(3):E42. tallization during freeze–drying: implications on lyoprotec-
51. Matheus S, Mahler HC, Friess W. 2006. A critical evaluation tion. J Phys Chem Lett 1(2):510–514.
of Tm (FTIR) measurements of high-concentration IgG1 anti- 70. Piedmonte DM, Summers C, McAuley A, Karamujic L,
body formulations as a formulation development tool. Pharm Ratnaswamy G. 2007. Sorbitol crystallization can lead to pro-
Res 23(7):1617–1627. tein aggregation in frozen protein formulations. Pharm Res
52. Papadimitrou A. 2001. Liquid pharmaceutical composi- 24(1):136–146.
tion containing an erythropoietin derivative. PCT/EP2001/ 71. Izutsu K, Heller MC, Randolph TW, Carpenter JF.
005187. 1998. Effect of salts and sugars on phase separation of
53. Remmele RL, Jr., Bhat SD, Phan DH, Gombotz WR. 1999. polyvinylpyrrolidone-dextran solutions induced by freeze-
Minimization of recombinant human Flt3 ligand aggregation concentration. J Chem Soc, Faraday Trans 94(3):411–417.
at the Tm plateau: a matter of thermal reversibility. Biochem- 72. Maity H, Karkaria C, Davagnino J. 2009. Mapping of solution
istry 38(16):5241–5247. components, pH changes, protein stability and the elimina-
54. Hawe A, Poole R, Romeijn S, Kasper P, van der Heijden R, tion of protein precipitation during freeze–thawing of fibrob-
Jiskoot W. 2009. Towards heat-stable oxytocin formulations: last growth factor 20. Int J Pharm 378(1–2):122–135.
Analysis of degradation kinetics and identification of degra- 73. Anchordoquy TJ, Carpenter JF. 1996. Polymers protect
dation products. Pharm Res 26(7):1679–1688. lactate dehydrogenase during freeze–drying by inhibiting
55. Dotsikas Y, Loukas YL. 2002. Kinetic degradation study of in- dissociation in the frozen state. Arch Biochem Biophys
sulin complexed with methyl-beta cyclodextrin. Confirmation 332(2):231–238.
of complexation with electrospray mass spectrometry and 1 H 74. Eriksson JH, Hinrichs WL, de Jong GJ, Somsen GW, Frijlink
NMR. J Pharm Biomed Anal 29(3):487–494. HW. 2003. Investigations into the stabilization of drugs by
56. Thirumangalathu R, Krishnan S, Bondarenko P, Speed- sugar glasses: III. The influence of various high-pH buffers.
Ricci M, Randolph TW, Carpenter JF, Brems DN. 2007. Pharm Res 20(9):1437–1443.

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 3, MARCH 2012 DOI 10.1002/jps
FORCED DEGRADATION OF THERAPEUTIC PROTEINS 911

75. Kolhe P, Badkar A. 2011. Protein and solute distribution in 93. Kiese S, Pappenberger A, Friess W, Mahler HC. 2010. Equi-
drug substance containers during frozen storage and post- librium studies of protein aggregates and homogeneous nu-
thawing: A tool to understand and define freezing–thawing cleation in protein formulation. J Pharm Sci 99(2):632–644.
parameters in biotechnology process development. Biotech- 94. Kiese S, Pappenberger A, Friess W, Mahler HC. 2008.
nol Prog 27(2):494–504. Shaken, not stirred: Mechanical stress testing of an IgG1
76. Pikal-Cleland KA, Cleland JL, Anchordoquy TJ, Carpenter antibody. J Pharm Sci 97(10):4347–4366.
JF. 2002. Effect of glycine on pH changes and protein sta- 95. Mahler HC, Muller R, Friess W, Delille A, Matheus S.
bility during freeze–thawing in phosphate buffer systems. 2005. Induction and analysis of aggregates in a liquid IgG1-
J Pharm Sci 91(9):1969–1979. antibody formulation. Eur J Pharm Biopharm 59(3):407–417.
77. Gomez G, Pikal MJ, Rodriguez-Hornedo N. 2001. Effect of 96. Mahler HC, Senner F, Maeder K, Mueller R. 2009. Sur-
initial buffer composition on pH changes during far-from- face activity of a monoclonal antibody. J Pharm Sci
equilibrium freezing of sodium phosphate buffer solutions. 98(12):4525–4533.
Pharm Res 18(1):90–97. 97. Sluzky V, Klibanov AM, Langer R. 1992. Mechanism of
78. Pikal-Cleland KA, Rodriguez-Hornedo N, Amidon GL, insulin aggregation and stabilization in agitated aqueous
Carpenter JF. 2000. Protein denaturation during freez- solutions. Biotechnol Bioeng 40(8):895–903.
ing and thawing in phosphate buffer systems: Monomeric 98. Maruyama T, Katoh S, Nakajima M, Nabetani H. 2001.
and tetrameric beta-galactosidase. Arch Biochem Biophys Mechanism of bovine serum albumin aggregation during
384(2):398–406. ultrafiltration. Biotechnol Bioeng 75(2):233–238.
79. Eckhardt BM, Oeswein JQ, Bewley TA. 1991. Effect of freez- 99. Colombie S, Gaunand A, Lindet B. 2001. Lysozyme inactiva-
ing on aggregation of human growth hormone. Pharm Res tion under mechanical stirring: effect of physical and molec-
8(11):1360–1364. ular interfaces. Enzyme Microb Technol 28(9–10):820–826.
80. Hawe A, Kasper JC, Friess W, Jiskoot W. 2009. Struc- 100. Ishikawa T, Kobayashi N, Osawa C, Sawa E, Wakamatsu
tural properties of monoclonal antibody aggregates induced K. 2010. Prevention of stirring-induced microparticle for-
by freeze–thawing and thermal stress. Eur J Pharm Sci mation in monoclonal antibody solutions. Biol Pharm Bull
38(2):79–87. 33(6):1043–1046.
81. Natalello A, Santarella R, Doglia SM, de Marco A. 2008. Phys- 101. Toth SI, Smith LA, Ahmed SA. 2009. Extreme sensitivity
ical and chemical perturbations induce the formation of pro- of botulinum neurotoxin domains towards mild agitation.
tein aggregates with different structural features. Protein J Pharm Sci 98(9):3302–3311.
Expr Purif 58(2):356–361. 102. Tyagi AK, Randolph TW, Dong A, Maloney KM, Hitscherich
82. Singh KS, Kolhe P, Wang W, Nema S. 2009. Freezing of C, Jr., Carpenter JF. 2009. IgG particle formation during fill-
biologics—A practitioner’s review. Part II: Practical advice. ing pump operation: A case study of heterogeneous nucleation
Bioprocess Intl 7(10):34–42. on stainless steel nanoparticles. J Pharm Sci 98(1):94–104.
83. Singh SK, Kolhe P, Wang W, Nema S. 2009. Freezing of bio- 103. Meireles M, Aimar P, Sanchez V. 1991. Albumin denatu-
logics—A practitioner’s review. Part I: Fundamental aspects. ration during ultrafiltration: Effects of operating conditions
Bioprocess Intl 7(9):32–44. and consequences on membrane fouling. Biotechnol Bioeng
84. Jiang G, Akers M, Jain M, Guo J, Distler A, Swift R, 38(5):528–534.
Wadhwa MV, Jameel F, Patro S, Freund E. 2007. Mecha- 104. Morar-Mitrica S, Brisbane C, Nesta DP, Ketkar A. 2009. Bio-
nistic studies of glass vial breakage for frozen formulations. physical approaches to the detection and characterization of
I. Vial breakage caused by crystallizable excipient mannitol. particles in an antibody solution. Am Pharm Rev 12(7):34–38.
PDA J Pharm Sci Technol 61(6):441–451. 105. Loksztejn A, Dzwolak W. 2010. Vortex-induced formation of
85. Cavatur RK, Vemuri NM, Pyne A, Chrzan Z, Toledo- insulin amyloid superstructures probed by time-lapse atomic
Velasquez D, Suryanarayanan R. 2002. Crystallization be- force microscopy and circular dichroism spectroscopy. J Mol
havior of mannitol in frozen aqueous solutions. Pharm Res Biol 395(3):643–655.
19(6):894–900. 106. Charman SA, Mason KL, Charman WN. 1993. Techniques
86. Cao E, Chen Y, Cui Z, Foster PR. 2003. Effect of freezing for assessing the effects of pharmaceutical excipients on
and thawing rates on denaturation of proteins in aqueous the aggregation of porcine growth hormone. Pharm Res
solutions. Biotechnol Bioeng 82(6):684–690. 10(7):954–962.
87. Kerwin BA, Heller MC, Levin SH, Randolph TW. 1998. Ef- 107. Katakam M, Banga AK. 1997. Use of poloxamer polymers to
fects of Tween 80 and sucrose on acute short-term stabil- stabilize recombinant human growth hormone against vari-
ity and long-term storage at -20 degrees C of a recombinant ous processing stresses. Pharm Dev Technol 2(2):143–149.
hemoglobin. J Pharm Sci 87(9):1062–1068. 108. Gulseren I, Guzey D, Bruce BD, Weiss J. 2007. Structural and
88. Kreilgaard L, Jones LS, Randolph TW, Frokjaer S, Flink JM, functional changes in ultrasonicated bovine serum albumin
Manning MC, Carpenter JF. 1998. Effect of Tween 20 on solutions. Ultrason Sonochem 14(2):173–183.
freeze-thawing- and agitation-induced aggregation of recom- 109. Stathopulos PB, Scholz GA, Hwang YM, Rumfeldt JA,
binant human factor XIII. J Pharm Sci 87(12):1597–1603. Lepock JR, Meiering EM. 2004. Sonication of proteins causes
89. Patapoff TW. 2010. Presentationon freeze-thawing of biolog- formation of aggregates that resemble amyloid. Protein Sci
ics at AAPS National Biotech Conference, San Francisco, CA. 13(11):3017–3027.
90. Bam NB, Cleland JL, Yang J, Manning MC, Carpenter JF, 110. Ashton L, Dusting J, Imomoh E, Balabani S, Blanch EW.
Kelley RF, Randolph TW. 1998. Tween protects recombinant 2009. Shear-induced unfolding of lysozyme monitored in situ.
human growth hormone against agitation-induced damage Biophys J 96(10):4231–4236.
via hydrophobic interactions. J Pharm Sci 87(12):1554–1559. 111. Biddlecombe JG, Craig AV, Zhang H, Uddin S, Mulot S,
91. Brych SR, Gokarn YR, Hultgen H, Stevenson RJ, Rajan R, Fish BC, Bracewell DG. 2007. Determining antibody stabil-
Matsumura M. 2010. Characterization of antibody aggrega- ity: Creation of solid–liquid interfacial effects within a high
tion: Role of buried, unpaired cysteines in particle formation. shear environment. Biotechnol Prog 23(5):1218–1222.
J Pharm Sci 99(2):764–781. 112. Biddlecombe JG, Smith G, Uddin S, Mulot S, Spencer D, Gee
92. Eppler A, Weigandt M, Hanefeld A, Bunjes H. 2010. Rele- C, Fish BC, Bracewell DG. 2009. Factors influencing antibody
vant shaking stress conditions for antibody preformulation stability at solid–liquid interfaces in a high shear environ-
development. Eur J Pharm Biopharm 74(2):139–147. ment. Biotechnol Prog 25(5):1499–1507.

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 3, MARCH 2012
912 HAWE ET AL.

113. Maa YF, Hsu CC. 1996. Effect of high shear on proteins. 2b in the presence of silicone oil. J Pharm Sci Online first
Biotechnol Bioeng 51(4):458–465. DOI:10.1002/jps.22704
114. Maa YF, Hsu CC. 1997. Protein denaturation by combined 133. Kerwin BA. 2008. Polysorbates 20 and 80 used in the formu-
effect of shear and air–liquid interface. Biotechnol Bioeng lation of protein biotherapeutics: structure and degradation
54(6):503–512. pathways. J Pharm Sci 97(8):2924–2935.
115. Thomas CR, Geer D. 2010. Effect of shear on proteins on 134. Wasylaschuk WR, Harmon PA, Wagner G, Harman AB,
solution. Biotechnol Lett 33(3):443–456. Templeton AC, Xu H, Reed RA. 2007. Evaluation of hydroper-
116. Bee JS, Stevenson JL, Mehta B, Svitel J, Pollastrini J, Platz oxides in common pharmaceutical excipients. J Pharm Sci
R, Freund E, Carpenter JF, Randolph TW. 2009. Response 96(1):106–116.
of a concentrated monoclonal antibody formulation to high 135. Majumdar S, Ford BM, Mar KD, Sullivan VJ, Ulrich RG,
shear. Biotechnol Bioeng 103(5):936–943. D’Souza A J. 2011. Evaluation of the effect of syringe surfaces
117. Clarkson JR, Cui ZF, Darton RC. 1999. Protein denaturation on protein formulations. J Pharm Sci 100(7):2563–2573.
in foam. J Colloid Interface Sci 215(2):333–338. 136. Liu D, Ren D, Huang H, Dankberg J, Rosenfeld R, Cocco MJ,
118. Rathore N, Rajan RS. 2008. Current perspectives on stability Li L, Brems DN, Remmele RL. 2008. Structure and stability
of protein drug products during formulation, fill and finish changes of human IgG1 Fc as a consequence of methionine
operations. Biotechnol Prog 24(3):504–514. oxidation. Biochemistry 47(18):5088–5100.
119. Bee JS, Chiu D, Sawicki S, Stevenson JL, Chatterjee K, 137. Hawkins CL, Davies MJ. 2001. Generation and propaga-
Freund E, Carpenter JF, Randolph TW. 2009. Monoclonal tion of radical reactions on proteins. Biochim Biophys Acta
antibody interactions with micro- and nanoparticles: Adsorp- 1504(2–3):196–219.
tion, aggregation, and accelerated stress studies. J Pharm Sci 138. Stadtman ER. 1993. Oxidation of free amino acids and amino
98(9):3218–3238. acid residues in proteins by radiolysis and by metal-catalyzed
120. Lam XM, Yang JY, Cleland JL. 1997. Antioxidants for pre- reactions. Annu Rev Biochem 62:797–821.
vention of methionine oxidation in recombinant monoclonal 139. Hovorka S, Schoneich C. 2001. Oxidative degradation of phar-
antibody HER2. J Pharm Sci 86(11):1250–1255. maceuticals: theory, mechanisms and inhibition. J Pharm Sci
121. Huang M, Horwitz TS, Zweiben C, Singh SK. 2011. Impact 90(3):253–269.
of extractables/leachables from filters on stability of protein 140. Ji JA, Zhang B, Cheng W, Wang YJ. 2009. Methionine, trypto-
formulations. J Pharm Sci 100(11):4617–4630. phan, and histidine oxidation in a model protein, PTH: Mech-
122. Bee JS, Randolph TW, Carpenter JF, Bishop SM, anisms and stabilization. J Pharm Sci 98(12):4485–4500.
Dimitrova MN. 2011. Effects of surfaces and leachables on 141. Zhao F, Ghezzo-Schoneich E, Aced GI, Hong J, Milby T,
the stability of biopharmaceuticals. J Pharm Sci. Online first Schoneich C. 1997. Metal-catalyzed oxidation of histidine
DOI:10.1002/jps.22597. in human growth hormone. Mechanism, isotope effects,
123. Duncan MR, Lee JM, Warchol MP. 1995. Influence of surfac- and inhibition by a mild denaturing alcohol. J Biol Chem
tants upon protein/peptide adsorption to glass and polypropy- 272(14):9019–9029.
lene. Int J Pharm 120:179–188. 142. Jensen JL, Kolvenbach C, Roy S, Schoneich C. 2000. Metal-
124. Johnston TP. 1996. Adsorption of recombinant human granu- catalyzed oxidation of brain-derived neurotrophic factor
locyte colony stimulating factor (rhG-CSF) to polyvinyl chlo- (BDNF): analytical challenges for the identification of modi-
ride, polypropylene, and glass: Effect of solvent additives. fied sites. Pharm Res 17(2):190–196.
PDA J Pharm Sci Technol 50(4):238–245. 143. Ph. Eur. 2007. Interferon alfa-2 concentrated solution. Eur
125. Zhou B, Pellett S, Tepp WH, Zhou H, Johnson EA, Janda Pharmacopoiea 62:2150–2153.
KD. 2008. Delineating the susceptibility of botulinum neu- 144. Chu JW, Brooks BR, Trout BL. 2004. Oxidation of methionine
rotoxins to denaturation through thermal effects. FEBS Lett residues in aqueous solutions: Free methionine and methion-
582(10):1526–1531. ine in granulocyte colony-stimulating factor. J Am Chem Soc
126. Bee JS, Davis M, Freund E, Carpenter JF, Randolph TW. 126(50):16601–16607.
2010. Aggregation of a monoclonal antibody induced by ad- 145. Hermeling S, Aranha L, Damen JM, Slijper M, Schellekens
sorption to stainless steel. Biotechnol Bioeng 105(1):121–129. H, Crommelin DJ, Jiskoot W. 2005. Structural characteri-
127. Jones LS, Kaufmann A, Middaugh CR. 2005. Silicone oil in- zation and immunogenicity in wild-type and immune toler-
duced aggregation of proteins. J Pharm Sci 94(4):918–927. ant mice of degraded recombinant human interferon alpha2b.
128. Ludwig DB, Carpenter JF, Hamel JB, Randolph TW. 2010. Pharm Res 22(12):1997–2006.
Protein adsorption and excipient effects on kinetic stability 146. Mulinacci F, Capelle MA, Gurny R, Drake AF, Arvinte
of silicone oil emulsions. J Pharm Sci 99(4):1721–1733. T. 2011. Stability of human growth hormone: Influence
129. Thirumangalathu R, Krishnan S, Ricci MS, Brems DN, of methionine oxidation on thermal folding. J Pharm Sci
Randolph TW, Carpenter JF. 2009. Silicone oil- and 100(2):451–463.
agitation-induced aggregation of a monoclonal antibody in 147. Mulinacci F, Bell SE, Capelle MA, Gurny R, Arvinte T. 2011.
aqueous solution. J Pharm Sci 98(9):3167–3181. Oxidized recombinant human growth hormone that main-
130. Jiang Y, Nashed-Samuel Y, Li C, Liu W, Pollastrini J, tains conformational integrity. J Pharm Sci 100(1):110–122.
Mallard D, Wen ZQ, Fujimori K, Pallitto M, Donahue L, Chu 148. Griffiths SW, Cooney CL. 2002. Relationship between protein
G, Torraca G, Vance A, Mire-Sluis T, Freund E, Davis J, Narhi structure and methionine oxidation in recombinant human
L. 2009. Tungsten-induced protein aggregation: solution be- alpha 1-antitrypsin. Biochemistry 41(20):6245–6252.
havior. J Pharm Sci 98(12):4695–4710. 149. Kappel C, Luther K, Troe J. 2002. Shock wave study of
131. Zhou S, Zhang B, Sturm E, Teagarden DL, Schoneich C, the unimolecular dissociation of H2 O2 in its falloff range
Kolhe P, Lewis LM, Muralidhara BK, Singh SK. 2010. and of its secondary reactions. Phys Chem Chem Phys
Comparative evaluation of disodium edetate and diethylen- 4(18):4392–4398.
etriaminepentaacetic acid as iron chelators to prevent 150. Keck RG. 1996. The use of t-butyl hydroperoxide as a
metal-catalyzed destabilization of a therapeutic monoclonal probe for methionine oxidation in proteins. Anal Biochem
antibody. J Pharm Sci 99(10):4239–4250. 236(1):56–62.
132. Auge KB, Blake-Haskins AW, Devine S, Rizvi S, Li YM, 151. Li S, Nguyen TH, Schoneich C, Borchardt RT. 1995. Aggre-
Hesselberg M, Orvisky E, Affleck RP, Spitznagel TM, Perkins gation and precipitation of human relaxin induced by metal-
MD. 2011. Demonstrating the stability of albinterferon alfa- catalyzed oxidation. Biochemistry 34(17):5762–5772.

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 3, MARCH 2012 DOI 10.1002/jps
FORCED DEGRADATION OF THERAPEUTIC PROTEINS 913

152. Kerwin BA, Remmele RL, Jr. 2007. Protect from light: ter addition of a bacteriostatic diluent. Am J Hosp Pharm
Photodegradation and protein biologics. J Pharm Sci 49(6):1455–1458.
96(6):1468–1479. 162. Ohls RK, Christensen RD. 1996. Stability of human recom-
153. Baertschi SW, Alsante KM, Tonnesen HH. 2010. A critical binant epoetin alfa in commonly used neonatal intravenous
assessment of the ICH guideline on photostability testing of solutions. Ann Pharmacother 30(5):466–468.
new drug substances and products (Q1B): Recommendation 163. Ling J, Hu M., Hagerup T, Campbell RK. 1999. Lispro
for revision. J Pharm Sci 99(7):2934–2940. insulin: Adsorption and stability in selected intravenous
154. Parti R, Ardosa J, Yang L, Mankarious S. 2000. In vitro devices. Diabetes Edu 25(2):237–245
stability of recombinant human factor VIII (recombinate). 164. Tzannis ST, Hrushesky WJ, Wood PA, Przybycien TM.
Haemophilia 6(5):513–522. 1996. Irreversible inactivation of interleukin 2 in a pump-
155. Kim HH, Lee YM, Suh JK, Song NW. 2007. Photodegra- based delivery environment. Proc Natl Acad Sci USA
dation mechanism and reaction kinetics of recombinant 93(11):5460–5465.
human interferon-alpha2a. Photochem Photobiol Sci 165. Vlasveld LT, Beijnen JH, Sein JJ, Rankin EM, Melief CJ,
6(2):171–180. Hekman A. 1993. Reconstitution of recombinant interleukin-
156. Qi P, Volkin DB, Zhao H, Nedved ML, Hughes R, Bass R, 2 (rIL-2): |A comparative study of various rIL-2 muteins. Eur
Yi SC, Panek ME, Wang D, Dalmonte P, Bond MD. 2009. J Cancer 29A(14):1977–1979.
Characterization of the photodegradation of a human IgG1 166. McLeod AG, Walker IR, Zheng S, Hayward CP. 2000. Loss of
monoclonal antibody formulated as a high-concentration liq- factor VIII activity during storage in PVC containers due to
uid dosage form. J Pharm Sci 98(9):3117–3130. adsorption. Haemophilia 6(2):89–92.
157. Chang JY, Xiao NJ, Zhu M, Zhang J, Hoff E, Russell SJ, 167. Maddux NR, Joshi SB, Volkin DB, Ralston JP, Middaugh
Katta V, Shire SJ. 2010. Leachables from saline-containing CR. 2011. Multidimensional methods for the formulation of
IV bags can alter therapeutic protein properties. Pharm Res biopharmaceuticals and vaccines. J Pharm Sci Online first
27(11):2402–2413. DOI:10.1002/jps.22618.
158. Jenke DR, Zietlow D, Garber MJ, Sadain S, Reiber D, 168. Banks DD, Hambly DM, Scavezze JL, Siska CC, Stackhouse
Terbush W. 2007. Accumulation of organic compounds NL, Gadgil HS. 2009. The effect of sucrose hydrolysis on the
leached from plastic materials used in biopharmaceutical stability of protein therapeutics during accelerated formula-
process containers. PDA J Pharm Sci Technol 61(4):286–302. tion studies. J Pharm Sci 98(12):4501–4510.
159. Nuijen B, Bouma M, Manada C, Jimeno JM, Lazaro LL, Bult 169. Quan C, Alcala E, Petkovska I, Matthews D, Canova-Davis E,
A, Beijnen JH. 2001. Compatibility and stability of the inves- Taticek R, Ma S. 2008. A study in glycation of a therapeutic
tigational polypeptide marine anticancer agent kahalalide F recombinant humanized monoclonal antibody: Where it is,
in infusion devices. Invest New Drugs 19(4):273–281. how it got there, and how it affects charge-based behavior.
160. Stickelmeyer MP, Graf CJ, Frank BH, Ballard RL, Storms Anal Biochem 373(2):179–191.
SM. 2000. Stability of U-10 and U-50 dilutions of insulin 170. Harris RJ, Kabakoff B, Macchi FD, Shen FJ, Kwong M,
lispro. Diabetes Technol Ther 2(1):61–66. Andya JD, Shire SJ, Bjork N, Totpal K, Chen AB. 2001.
161. Corbo DC, Suddith RL, Sharma B, Naso RB. 1992. Stabil- Identification of multiple sources of charge heterogeneity in
ity, potency, and preservative effectiveness of epoetin alfa af- a recombinant antibody. J Chromatogr B 752(2):233–245.

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 3, MARCH 2012

You might also like