Abstract
When isolator technology is applied to biotechnology drug product fill-finish process, hydrogen peroxide (H2O2) spiking studies for the determination of the sensitivity of protein to residual peroxide in the isolator can be useful for assessing a maximum vapor phase hydrogen peroxide (VPHP) level. When monoclonal antibody (mAb) drug products were spiked with H2O2, an increase in methionine (Met 252 and Met 428) oxidation in the Fc region of the mAbs with a decrease in H2O2 concentration was observed for various levels of spiked-in peroxide. The reaction between Fc-Met and H2O2 was stoichiometric (i.e., 1:1 molar ratio), and the reaction rate was dependent on the concentrations of mAb and H2O2. The consumption of H2O2 by Fc-Met oxidation in the mAb followed pseudo first-order kinetics, and the rate was proportional to mAb concentration. The extent of Met 428 oxidation was half of that of Met 252, supporting that Met 252 is twice as reactive as Met 428. Similar results were observed for free L-methionine when spiked with H2O2. However, mAb formulation excipients may affect the rate of H2O2 consumption. mAb formulations containing trehalose or sucrose had faster H2O2 consumption rates than formulations without the sugars, which could be the result of impurities (e.g., metal ions) present in the excipients that may act as catalysts. Based on the H2O2 spiking study results, we can predict the amount Fc-Met oxidation for a given protein concentration and H2O2 level. Our kinetic modeling of the reaction between Fc-Met oxidation and H2O2 provides an outline to design a H2O2 spiking study to support the use of VPHP isolator for antibody drug product manufacture.
LAY ABSTRACT: Isolator technology is increasing used in drug product manufacturing of biotherapeutics. In order to understand the impact of residual vapor phase hydrogen peroxide (VPHP) levels on protein product quality, hydrogen peroxide (H2O2) spiking studies may be performed to determine the sensitivity of monoclonal antibody (mAb) drug products to residual peroxide in the isolator. In this study, mAbs were spiked with H2O2; an increase in methionine (Met) oxidation of the mAbs with a decrease in H2O2 concentration was observed for various levels of spiked-in peroxide. The reaction between Met and H2O2 was 1:1, and its rate was dependent on mAb and H2O2 concentrations. Consumption of H2O2 by Met followed pseudo first-order kinetics; the rate was proportional to mAb concentration. Formulations containing trehalose or sucrose had faster consumption rates than formulations without the sugars, which could be due to excipient impurities. Based on H2O2 spiking study results, we can predict the amount of Met oxidation for a given mAb concentration and H2O2 level. Our modeling of the reaction between Fc-Met oxidation and H2O2 provides an outline to design a H2O2 spiking study that supports using VPHP isolators during manufacture of mAb products.
Introduction
Among all the protein pharmaceuticals that have been approved for commercial use, monoclonal antibodies (mAbs) have emerged as an important class of drugs in the biopharmaceutical industry. They can treat many diseases, with a majority of treatments being in oncology and immunology. Although mAb drug products often have a shelf life at the intended storage temperature for at least 18 months, and preferably 36 months in their formulations, they can be susceptible to physical and chemical degradation caused by processing stresses (e.g., thawing, mixing, and inspection) during product manufacturing. Chemical degradation pathways that may affect the stability of mAbs include oxidation, deamidation, isomerization, and others. The oxidation of methionine (Met) residues in mAbs is one of the chemical degradation pathways of which a common reaction product is Met sulfoxide. Met oxidation may result in the decrease of mAbs' binding affinities and loss of their biological activities (1⇓⇓⇓⇓⇓–7), and it can significantly limit the shelf life of the drug product. mAbs usually contain several Met residues that have different degrees of susceptibility to oxidation. Susceptible Met residues are typically located on the surface of the protein, and their susceptibility is also highly dependent on their solvent exposure (2⇓–4). Based on thermal stability studies, it has been reported that Met 252 and Met 428, (numbering according to Kabat et al. (8), are the two Met residues in the fragment crystallizable (Fc) region of each heavy chain in mAbs that are most vulnerable to oxidation due to surface exposure (2). These two Met residues are found in all immunoglobin G (IgG) classes and are located at the CH2–CH3 interface of the Fc region (9), which is also an important structural region for binding to Protein A and neonatal Fc receptor (FcRn). Because Met 252 and Met 428 are located in this region, oxidation of these Met residues has shown to weaken the antibody binding affinities to Protein A and FcRn (1, 4, 5).
Previous studies have demonstrated that several agents can oxidize Met residues, including tert-butylhydroperoxide, hydrogen peroxide (H2O2), 2,2′-azobis(2-amidinopropane)dihydrochloride (also known as AAPH) and chloramine T (10⇓⇓⇓–14). Among these oxidizing agents, H2O2 is commonly used as a sterilizing agent for surface decontamination in the biopharmaceutical industry. For example, vapor phase hydrogen peroxide (VPHP) is typically used for sanitizing isolators for drug product manufacture because it is environmentally safer than other agents, such as peracetic acid (15), and it is effective in killing a variety of microorganisms at low concentrations (16). Currently, biopharmaceutical manufacturers are moving protein drug product filling operations out of traditional clean rooms or restricted-access barriers and into VPHP isolator systems for the reason of improved sterility assurance and operator comfort. However, a disadvantage of using VPHP isolators is that H2O2 may still be present in residual amounts in the isolator. Additionally, the vapor absorbed by the non-metallic materials (tubing, glove ports, fill nozzles) for product filling and the surface of the isolator during the decontamination cycle (for sterilization) can be released back into the isolator's atmosphere once production begins (due to movement and airflow). Depending on the residual peroxide level in the isolator during production, the drug product may be exposed to different levels of H2O2 as a result of this process. Therefore, the amount of acceptable peroxide for the protein drug products to be manufactured using VPHP isolators should be assessed via spiking studies. During such studies, specified amounts of H2O2 solution can be spiked into protein formulations, which are then placed on stability assessment. Throughout the stability assessment, protein oxidation and peroxide residuals in the formulations can be monitored to determine a maximum tolerated level of peroxide that does not trigger unacceptable changes in drug product quality, including oxidation. The results from H2O2 spiking studies can be used as guidance for the development of a VPHP decontamination cycle for the isolator.
Additionally, uptake studies (where vials filled with protein solution are exposed to VPHP inside the isolator) are valuable in determining the amount of H2O2 that can enter and dissolve into the drug product. Many factors can affect the H2O2 uptake rate and the dissolved amount (e.g., vial size, vial opening diameter, fill volume, time between liquid fill and stopper closing, interruptions, residual VPHP level, airflow, location of the vial, etc.). A thorough assessment combining the drug product formulation sensitivity data (spiking studies) with the dissolved H2O2 amounts into a specific primary packaging configuration (uptake studies) can assure a fill and finish process that does not lead to unacceptable oxidation triggered by residual peroxide.
In this study, we investigated the factors that can influence H2O2-induced mAb oxidation. Because the amount of Met oxidation is expected to correlate with the spiked-in H2O2 that is consumed by the Met residues, determining the correlation will be a useful tool in predicting an acceptable H2O2 level based on mAb concentration. While conducting such investigation, two additional factors were evaluated: (1) stability of H2O2 at low concentrations (stability of high concentrations have been reported in the literature), and (2) types of H2O2 solutions (stabilized and unstabilized; details can be found in Materials) commonly used for isolator disinfection. The results from such studies can enable a recommendation and optimal guidance to support the use of isolators sanitized by VPHP.
Materials
Three fully recombinant humanized monoclonal IgG1 antibodies (mAb1, mAb2, and mAb3) provided by Genentech Inc. (South San Francisco, CA) were used for the H2O2 spiking studies. The mAb formulations are listed in Table I. All chemicals and reagents used were reagent grade. Stabilized H2O2 solution (30–35%) was purchased from J. T. Baker (Center Valley, PA) or EMD Millipore (Billerica, MA). Unstabilized H2O2 solution (35%) was purchased from Solvay Chemicals (La Porte, TX).
Formulation Compositions of mAb1, mAb2, and mAb3
H2O2 is commercially supplied as a stabilized or unstabilized solution for use in sterilization of isolators in the pharmaceutical industry. Stabilized H2O2 contains small amounts (total <0.04%) of sodium pyrophosphate, phosphoric acid, ammonium nitrate, and/or stannate as preservatives (per the supplier's certificate of analysis) and has a minimum shelf life of 2 years at room temperature. However, the unstabilized form does not contain such stabilizers and has a limited shelf life of 6–12 months at the same storage temperature.
Methods
Hydrogen Peroxide Spiking Studies
The correlation between H2O2 consumption and H2O2-induced oxidation in mAbs was determined by H2O2 spiking studies. For each mAb studied, various amounts of stabilized H2O2 stock solution were spiked into the protein formulation to obtain final H2O2 concentrations ranging from 10 to 7000 ng/mL. mAb formulations without H2O2 spiking served as controls for the studies. Each of the H2O2-spiked samples and unspiked controls was filtered through a 0.22 μm filter before being aseptically filled into glass vials for real-time (5 °C) stability testing for up to 24 months and accelerated stability (30 °C) testing for up to 3 months. At each time point the samples were analyzed for protein oxidation using peptide mapping and for residual peroxide using a horseradish peroxidase (HRP) assay.
The effect of mAb formulation excipients on H2O2 degradation was also evaluated by H2O2 spiking studies. Formulation buffers (without protein) at pH 6.2 and 6.5 with and without surfactant (polysorbate 20 or 80), cyroprotectant (trehalose or sucrose), sodium chloride, and disodium ethylenediaminetetraacetate (EDTA) were spiked with stabilized H2O2 at final concentrations of 200 and 2000 ng/mL. For each formulation buffer, an unspiked sample served as a control. Each of the H2O2-spiked and unspiked formulation buffers was filtered through a 0.22 μm filter before it was filled into glass vials and placed at 30 °C for up to 28 days (e.g, 1, 4, 7, 14, and/or 28 days) for residual peroxide testing by the HRP assay.
To compare the consumption of H2O2 by free Met and mAbs containing Met in the Fc region, L-methionine was added to a histidine buffer at pH 5.5 consisting of sucrose and polysorbate 20 to achieve final concentrations of 0.5, 1, 5, and 10 mM. In addition, a protein solution consisting of 40 mg/mL (0.3 mM) mAb2 in phosphate buffer at pH 6.2 was used. Unstabilized H2O2 solution was added to all methionine-containing buffers and 0.3 mM mAb2 protein solution to achieve a final concentration of 1000 ng/mL, followed by incubation at 5 °C, 25 °C, and 40 °C for up to 72 h. At each storage condition, H2O2 concentration was determined by the HRP assay.
To study the degradation between stabilized and unstabilized H2O2 and their effect on Met oxidation of mAbs, each type of H2O2 was spiked into mAb1 and mAb2 formulation buffers (Table I) and 10 mg/mL mAb2 in the phosphate buffer at pH 6.2 to achieve final H2O2 concentrations of 200 ng/mL and 500 ng/mL, respectively. Samples were filtered through a 0.22 μm filter before they were filled into glass vials. The formulation buffers and mAb2 samples were placed on stability at 30 °C for up to 7 and 21 days, respectively. At each time point, the H2O2 concentration was determined by the HRP assay. The mAb2 samples were analyzed for protein oxidation by peptide mapping.
Determination of H2O2 Concentrations in mAb Formulations and Formulation Buffers
H2O2 concentrations in mAb formulations and formulation buffers were measured using the fluorometric Amplex® UltraRed HRP Assay purchased from Life Technologies (Carlsbad, CA). In this assay, the Amplex UltraRed reagent, which is sensitive to extremely low concentrations of peroxides, is used in combination with HRP to detect peroxides in solution. In the presence of HRP, the Amplex UltraRed reagent reacts with H2O2 in a 1:1 stoichiometric ratio to produce Amplex UltroxRed, a brightly fluorescent and absorbing reaction product. All H2O2 standards curves were prepared at 1–500 ng/mL in formulation buffers with or without mAb. All samples and formulation buffers for standard curves were plated onto a 96-well plate and analyzed with a Molecular Probes ME20 plate reader (Eugene, OR) with an excitation and emission at 530–560 nm and 590 nm, respectively. For formulations containing protein, the limit of detection (LOD) for this assay ranged from 6 to 16 ng/mL H2O2, depending on the protein formulation.
Determination of mAb Oxidation by Peptide Mapping
Oxidation of mAbs was determined by either trypsin or Lys-C peptide mapping using reverse-phase liquid chromatography on an Agilent 1200 high-performance liquid chromatography (HPLC) (Agilent Technologies, Santa Clara, CA) instrument equipped with a diode-array detector and a solvent delivery system with binary pump. Samples were first reduced with 1.0 M dithiothreitol for 30 min at 37 °C followed by carboxymethylation with iodoacetic acid under the same conditions. After carboxymethylation, samples were digested with trypsin (mAb1 and mAb3) or endoproteinase Lys-C (mAb2) for 4–5 h at 37 °C. A C18 column (300 Å, 5 μm particle, 4.6 × 250 mm) was controlled at 45 °C and the column effluent was monitored at 214 nm. Elution was achieved at 1.0 mL/min with a mobile phase consisting of solvent A (0.1% trifluoroacetic acid in water) and solvent B (0.08% trifluoroacetic acid in acetonitrile). The peptides were separated with a gradient from 0% to 70% solvent B over 130 min and detected by ultraviolet (UV) absorbance. For mAb1, Met oxidation that occurred in the H2O2-spiked samples was determined by the presence of new peaks in the peptide map when compared with the unspiked control. For mAb2 and mAb3, Met oxidation was quantitated from the peak area of the oxidized species.
Results and Discussion
Correlation between Met Oxidation in mAbs and Peroxide Consumption
In the H2O2-spiking studies, Met 252 and Met 428 in the Fc region (Fc-Met) of all three mAbs were monitored for oxidation because they were the most surface-exposed Met residues (8) and thus most susceptible to H2O2-induced oxidation. Fc-Met oxidation and H2O2 residual for mAb1, mAb2, and mAb3 are presented in Tables II, III, and IV, respectively. For mAb1 at 10 mg/mL protein concentration, Fc-Met oxidation was initially detected qualitatively by trypsin peptide mapping for the presence of oxidized peptide in the samples spiked with 1000 and 250 ng/mL H2O2 after storage at 5 °C for 3 and 6 months, respectively (Table II). Without spiked-in peroxide, a minimal amount of Met oxidation was observed in the control. mAb1 samples spiked with less than or equal to 100 ng/mL H2O2 also had similar oxidation results when compared with the control throughout the stability study. Peroxide content in solution decreased over time for all samples spiked with different levels of H2O2, presumably as a result of consumption by the Fc-Mets for oxidation.
Peroxide Residual and Oxidation of mAb1 Formulation (10 mg/mL) Spiked with Various Concentrations of H2O2 at 5 °C Storage
Peroxide Residual and Oxidation of mAb2 Formulation (25 mg/mL) Spiked with Various Concentrations of H2O2 at 5 °C Storage
Peroxide Residual and Oxidation of mAb3 Formulation (200 mg/mL) Spiked with Various Concentrations of H2O2 at 5 °C Storage
For mAb2 at 25 mg/mL protein concentration, Met 252 and Met 428 oxidation was measured separately and quantitatively by Lys-C peptide mapping. No difference in oxidation levels were observed in the mAb2 samples spiked with not more than 50 ng/mL when compared with the unspiked control. At 5 °C, an inverse correlation was observed between the amount of peroxide residual and the level of Met 252 oxidation in the mAb2 sample spiked with 5000 ng/mL peroxide (Figure 1). With 5000 ng/mL H2O2 spiked in, a large increase (21.2%) in Met 252 oxidation was observed throughout a period of 3 months at 5 °C that corresponded to a significant decrease of about 85% of the initial in peroxide concentration (Table III). Increased in Met oxidation and decrease in peroxide concentration were also observed at subsequent time points (e.g., 6 and 9 months). However, the degree of oxidation was much less when compared to the initial 3 month time point. Similar results were observed for mAb2 samples containing 250 and 1000 ng/mL H2O2 under the same storage conditions. Additionally, Met 428 behaved similarly to Met 252 in terms of oxidation by H2O2 in the mAb2 formulation, but the extent of oxidation was approximately one-half of that observed at Met 252. These observations suggested that H2O2 in the mAb samples might have been consumed by the Fc-Met residues in the protein molecule during the initial months, which would explain the minimal additional increase in oxidation level thereafter.
Correlation between Met 252 oxidation and loss of H2O2 in the mAb2 formulation (25 mg/mL protein concentration) spiked with 5000 ng/mL H2O2 and stored at 5 °C.
For mAb3 at 200 mg/mL protein concentration, large increases of Met 252 and Met 428 oxidation induced by H2O2 was observed only when a very high peroxide concentration (e.g., 7000 ng/mL) was spiked in. The reaction occurred within the first month at 5 °C, as no further oxidation was observed for both Met 252 and Met 428 after 1 month, and the peroxide level in the sample also decreased significantly (∼99%) at the same time (Table IV). For mAb3 samples spiked with less than or equal to 1000 ng/mL, no difference in both Met 252 and Met 428 oxidation levels were observed when compared with the unspiked control.
Extent of Fc-Met Oxidation in mAb Formulation
The results from the H2O2 spiking studies for mAb1, mAb2, and mAb3 are in agreement with published reports that Met and H2O2 react in a one-to-one stoichiometric ratio (17, 18), and Met 252 is twice as reactive as Met 428 (14). Previous studies demonstrated that Fc-Met oxidation rates of mAbs are similar in solutions with a pH range of 3–8 (19); the extent of Met 252 oxidation should be comparable among mAbs in commonly used formulation buffers that have pH values of 5.5–6.5. Based on these factors, the amount of Met 252 and Met 428 oxidation, as determined by peptide mapping, can be expressed as the following:
where 34 represents the molecular weight (MW) of H2O2 in Da, and 150 represents the MW (in kDa) of mAb, and 2 represents two Met252 or 428 residues; and
where CH2O2 and CMet are the concentrations of H2O2 and Met, respectively. The factors of 2/3 in eq 3 and 1/3 in eq 4 are introduced because Met 252 is twice reactive over Met 428 (e.g., 2/3 of total H2O2 will react with Met 252, whereas 1/3 will react with Met 428). Based on these equations, the amount of oxidized Met 252 (in percent) can be calculated for a given concentration of H2O2 present in mAb solution at a given protein concentration. A plot of the theoretical percentage of Met 252 oxidation calculated versus H2O2 concentrations for various mAb concentrations is shown in Figure 2. For a mAb at 25 mg/mL (with Met 252 or Met 428) at a concentration of 0.333 × 10–3 M, the expected Met 252 and Met 428 oxidation by H2O2 at 1000 ng/mL (i.e., 0.03 × 10–3 M) should be 6% and 3%, respectively. The actual observed Met 252 and Met 428 oxidation in mAb2 (25 mg/mL) formulation spiked with 1000 ng/mL H2O2 was 4.7% and 2.4%, respectively, when the peroxide was totally consumed after 6 months at 5 °C (Table III). The theoretical and observed oxidations of Met residues in the mAb2 formulation are considered comparable.
Theoretical percent Met 252 oxidation based on protein concentration and a ratio of 2:1 for Met 252 to Met 428 oxidation.
Consumption of H2O2 by Fc-Met Oxidation in mAb Formulations Can Be Pseudo First-Order Kinetics
When the spiked-in H2O2 is consumed in a protein formulation, it is expected that the measured H2O2 in the protein solution would slowly disappear until all the H2O2 has been expended. The time course of the consumption of H2O2 by mAb1 during storage at 5 °C is shown in Figure 3. When taking the log of the H2O2 concentrations as a function of time, a linear fit of the results was observed (Figure 4A). Similar results were also observed for the mAb2 and mAb3 formulations when the same calculation was applied to the H2O2 concentrations (Figures 4B and 4C). The observed linear regressions indicate that consumption of H2O2 in the presence of the mAb follows a pseudo first-order kinetic reaction where the rate constants (k) can be obtained from the slope of the linear regression from the plots. This is assuming that the H2O2 is only consumed by Fc-Met residues. The excipients in the formulation have no effect on H2O2 consumption, and no other reactive Met residues are present in the mAb. Because the Met residues of the mAbs are most likely being oxidized by the spiked-in H2O2, the bi-molecular reaction can be written as
With the reaction rate equation:
where [Met] is the concentration of Met residues in the protein formulation, and [H2O2] is the concentration that is spiked into the samples. When the protein concentration is in excess (i.e., [Met] >>> [H2O2]), the Met concentration remains unchanged and the first-order rate equation can be reduced to
where
Or it can be written as
where [H2O2]t0 and [H2O2]t are the concentrations of H2O2 at the initial time point and at another specified time point, respectively. The assumption that the protein concentration is always in excess compared with the H2O2 concentration is reasonable (in these spiking studies) because the amount of protein is always much greater than the H2O2 that is spiked in. The concentration of 10 mg/mL mAb, which has a MW of 150 kDa, is equal to 0.066 mM with two chains each having Met 252 and Met 428 residues, which results in a total concentration of 0.26 mM (0.066 × 4) of Met residues that can be potentially oxidized by H2O2—whereas H2O2 has a MW of 34 Da, and it can range from 0.00029 mM at 10 ng/mL to 0.21 mM at 7000 ng/mL. Based on these observations, it can be concluded that the H2O2 consumption follows the pseudo first-order kinetics (eq 8), where the straight lines were observed in the semi-log plots as shown in Figures 4A, 4B, and 4C. In addition, the reaction rate constant is proportional to the Met concentration, which is proportional to the protein concentration (eq 7).
Time course of H2O2 consumption in mAb1 formulation (10 mg/mL protein concentration) spiked with 100, 250, and 1000 ng/mL H2O2 and stored at 5 °C.
Pseudo first-order kinetic reactions of H2O2 consumption by Met oxidation observed for various concentrations of H2O2 spiked in (A) mAb1 formulation (10 mg/mL protein concentration); (B) mAb2 formulation (25 mg/mL protein concentration); and (C) mAb3 formulation (200 mg/mL protein concentration).
H2O2 Consumption Rates are Proportional to mAb Concentration
Based on the pseudo first-order kinetics observed in the three mAb formulations (Figures 4A, 4B, and 4C), the consumption rate of H2O2 can be based on protein concentration. In the H2O2 spiking studies, the higher the protein concentration in the formulation, the faster the H2O2 was consumed. As a result, the amount of time required for the mAb to consume the spiked-in H2O2 can be calculated theoretically based on its concentration given the assumptions presented. Based on this theoretical calculation, the required duration for the H2O2 spiking stability study may be estimated. The spiking study can be carried out until the spiked-in H2O2 can no longer be detected. If residual amounts of H2O2 are still detectable in the protein formulation, the potential for the mAb to be oxidized still exists. Based on Figure 3, it took approximately 24 months at 5 °C for the H2O2 in the mAb1 (10 mg/mL) formulation spiked with 1000 ng/mL H2O2 to reach an undetectable level. Because the mAb2 formulation is 2.5 times the protein concentration (25 mg/mL) of the mAb1 formulation, the consumption rate of H2O2 in the mAb2 formulation is expected to be 2.5 times faster than that which was observed for mAb1, as both mAbs have the same MW of 150 kDa and have the same IgG1 backbone and Met residues are similarly surface-exposed. The spiked-in 1000 ng/mL H2O2 in the mAb2 (25 mg/mL) formulation should be consumed in 9.6 months of storage at 5 °C, theoretically. For the mAb3 formulation with a protein concentration of 200 mg/mL, the H2O2 is expected to reach an undetectable level within 1.2 months under the same storage condition because the protein concentration is 20 times that of mAb1. The actual H2O2 consumption rates observed for the mAb2 and mAb3 formulations in this study basically followed this assumption with some exceptions. The consumption rates observed were faster than the predicted rates, as shown in Table V. Additionally, the predicted consumption rates of mAb2 and mAb3 were not within the calculated 95% confidence intervals of the observed consumption rates from the spiking studies. The fact that all three mAb formulations were formulated using different excipients may have also contributed to the increase in consumption rates of the H2O2.
Consumption Rates (reciprocal month) of Peroxide in mAb Formulations Spiked with 1000 ng/mL H2O2
Formulation Excipients' Impact on H2O2 Degradation
The observation of faster-than-expected H2O2 consumption rates in the spiking studies for mAb2 and mAb3 led us to investigate if excipients in the mAb formulations may affect the degradation of H2O2. To determine the formulation excipient effect, H2O2 at 200 ng/mL was spiked into the mAb1 formulation buffer (citrate buffer at pH 6.5 with sodium chloride and polysorbate 80) and the mAb2 formulation buffer (phosphate buffer at pH 6.2 with trehalose and polysorbate 20). In the absence of protein, it is expected that the H2O2 should remain fairly constant in both formulation buffers because there is nothing present that can consume the spiked-in H2O2. After 28 days at 30 °C, the amount of H2O2 (200 ng/mL) in the mAb1 formulation buffer remained constant. However, the 200 ng/mL H2O2 spiked into the mAb2 formulation buffer was depleted after 14 days at 30 °C as shown in Figure 5. Similar results were also observed when a higher level of H2O2 (2000 ng/mL) was spiked into the same buffers (data not shown).
Degradation of 200 ng/mL H2O2 in mAb1 and mAb2 formulation buffers (without protein) over time at 30 °C.
As a result of the H2O2 degradation observed in the mAb2 formulation buffer, each component of the buffer (i.e., phosphate, trehalose, and polysorbate 20) was investigated to determine if any of the excipient(s) may potentially degrade or consume the peroxide. H2O2 was spiked into the following five buffers at a final concentration of 200 ng/mL and placed at 30 °C for up to 7 days: (1) mAb2 buffer as positive control, (2) mAb2 buffer without trehalose and polysorbate 20, (3) mAb2 buffer without trehalose, (4) mAb2 buffer without polysorbate 20, and (5) mAb2 buffer plus 0.05% EDTA. EDTA was added to the mAb2 formulation buffer to evaluate whether a chelating agent might help prevent the degradation of H2O2. When comparing the residual peroxide concentration across all five buffer solutions within 7 days at 30 °C, significant degradation of H2O2 was only observed in the buffer solutions that contained trehalose (Figure 6). In fact, the peroxide was completely depleted after 3–4 days at 30 °C for the trehalose-containing buffer solutions. For the buffer solutions containing no trehalose, minimal loss of the peroxide was detected for the same incubation period. The presence of EDTA as chelating agent reduced the degradation rate of the H2O2 in the mAb2 buffer, suggesting that trace impurities of metal ions may be present in the buffer (possibly from trehalose) that might have catalyzed the degradation of H2O2 and resulted in a faster degradation rate.
Effect of formulation excipients on degradation of 200 ng/mL H2O2 over time at 30 °C in the following buffers: (#1) mAb2 formulation buffer (phosphate buffer at pH 6.2 with trehalose and polysorbate 20); (#2) mAb2 formulation buffer without trehalose and polysorbate 20; (#3) mAb2 formulation buffer without trehalose; (#4) mAb2 formulation buffer without polysorbate 20; and (#5) mAb2 formulation buffer with 0.05% EDTA.
Because many protein formulations contain either trehalose or sucrose as a cryo-protectant for drug substance storage, the effect of sucrose on H2O2 degradation was also evaluated. With the presence of 120 mM or 240 mM sucrose and 200 ng/mL H2O2 in phosphate buffer at pH 6.2, the degradation rates of H2O2 were similar to the trehalose-containing buffer solutions at 30 °C (Figure 7). However, the rate of peroxide depletion was not dependent on sugar concentration, and it was reduced by the addition of EDTA. Keeping this observation in mind, the kinetic model presented earlier in this report may be considered a worst-case assumption in regards to the amount of H2O2 and time required for the spiking studies. Therefore, the kinetic model is considered a valuable tool for planning such spiking studies to support the acceptable level of residual peroxide for manufacture of protein drug products using VPHP for sterilization in isolators.
Effect of 120 mM sucrose and 240 mM sucrose with and without 0.05% EDTA on degradation of 200 ng/mL H2O2 in phosphate buffer at pH 6.2 (PB) over 7 days at 30 °C.
Consumption of H2O2 by L-Methionine (Free) in Formulation Buffers and Activation Energy
Because the duration of a H2O2 spiking study can be calculated based on the correlation between Met oxidation in the mAb and peroxide consumption, similar calculations may also be useful if it can be demonstrated that free L-methionine behaves similarly in H2O2 spiking studies.
Because Fc-Met residues in a mAb can consume the spiked-in H2O2, free L-methionine should act similarly as Fc-Met residues in consuming the H2O2 based on the assumption that Fc-Met residues are strongly surface-exposed and react in a similar way as L-methionine (20). A study for determining the reaction rate between L-methionine and H2O2 was conducted by spiking 1000 ng/mL H2O2 into a histidine buffer consisting of sucrose and polysorbate 20 at pH 5.5 with different concentrations of L-methionine that ranged from 0 to 10 mM. The samples were placed at 5 °C, 25 °C, and 40 °C for up to 72 h. Residual H2O2 was monitored throughout the study to determine whether the L-methionine in the formulation buffer also behaves similarly to the mAb formulations in terms of H2O2 consumption. Similar to H2O2 spiking studies with mAb formulations, pseudo first-order kinetics was observed for all L-methionine concentrations (data not shown) under accelerated and stress conditions. When plotting the H2O2 consumption rates of L-methionine in an Arrhenius plot (Figure 8), the slopes of these linear fits are nearly identical for the buffer containing various amounts of L-methionine, demonstrating that the activation energy of the reaction between L-methionine and H2O2 are comparable. When comparing the linear fits of mAb2 (phosphate buffer at pH 6.2 with 0.3 mM mAb2) and L-methionine (Figure 8), the slope of mAb2 is slightly steeper than the slopes of L-methionine at all concentrations. Using the slopes that were generated from Figure 8, the activation energies were calculated. The average activation energy between L-methionine and H2O2 was approximately 38 kJ/mol, and it was 59 kJ/mol for mAb2 (Table VI). L-methionine at all concentrations was formulated in a buffer containing a cryo-protectant that might contain trace impurities of metal ions as catalysts for the H2O2-induced oxidation to occur. Thus, less activation energy might be required for the reaction when compared with mAb2 containing no sugar. This observation is consistent with the results obtained from the formulation excipient investigation, where trehalose-containing buffer had a faster rate of H2O2 degradation as compared with the buffer containing no sugar.
Arrhenius plot of H2O2 consumption by 0.5, 1, 5, and 10 mM L-methionine in histidine buffer consisting of sucrose and polysorbate 20 at pH 5.5 and 0.3 mM mAb2 in phosphate buffer at pH 6.2. The H2O2 consumption rate constants (in reciprocal day) versus reciprocals of storage temperatures are shown. The activation energies for the consumption of H2O2 by free L-methionine and mAb2 were calculated from the slope of the lines.
Activation Energies of H2O2 Consumption by Free L-methionine and mAb2 Calculated from the Slopes and y-Intercepts of the Arrhenius Plots
Types of H2O2 and Their Effect on Fc-Met Oxidation
To determine if there is any difference between the stabilized and unstabilized forms of H2O2 in terms of degradation, both types of peroxide were spiked into the mAb1 formulation buffer (citrate buffer at pH 6.5 with sodium chloride and polysorbate 80) and the mAb2 formulation buffer (phosphate buffer at pH 6.2 with trehalose and polysorbate 20) at a final concentration of 200 ng/mL, and the degradation of the peroxides was monitored at 30 °C. No differences were observed between the stabilized and unstabilized H2O2 in terms of their degradation behavior over time in these two buffers (Figure 9). Loss of H2O2 for both types was not detected in the mAb1 formulation buffer containing no trehalose after 3 months at 30 °C. In contrast, both types of H2O2 in the mAb2 formulation buffer that contained trehalose were completely depleted within 7 days at 30 °C. Because the H2O2 concentration was quite low (diluted from 35% or 350 mg/mL to 200 ng/mL) in the formulation buffers, the amount of stabilizers originally present in the stock solution may have been too dilute to have an impact on peroxide degradation.
Degradation of 200 ng/mL H2O2 (stabilized versus unstabilized) in mAb1 formulation buffer (no trehalose) and mAb2 formulation buffer (with trehalose).
Because there were no differences in degradation between the stabilized and unstabilized H2O2 in the formulation buffers, it is expected that there should also be no difference in the kinetics of H2O2-induced Met oxidation in mAbs. However, there was a concern that the stabilizers present in the stabilized H2O2 may have an effect on Met oxidation. For this reason, both types of H2O2 (final concentration of 500 ng/mL each) were spiked into the mAb2 formulated at 10 mg/mL in phosphate buffer at pH 6.2 to assess the difference between stabilized and unstabilized H2O2 in terms of H2O2 consumption and Met oxidation at 30 °C. Trehalose and polysorbate were excluded in the buffer in order to eliminate the effect of impurities such as metal ions on H2O2 degradation. After 21 days at 30 °C, the amount of Met 428 and Met 252 oxidation and the total H2O2 consumed in the stabilized and unstabilized peroxide-spiked samples were comparable (Table VII). A slight increase in both Met 428 and Met 252 oxidation of the mAb2 control was also observed when compared with the initial time point, which is expected at an elevated incubation temperature of 30 °C. Overall, the amount of Met 252 oxidation observed was approximately twice the amount of Met 428 oxidation for both stabilized and unstabilized peroxide-spiked samples, which had a ratio of about 2:1. This result is consistent with other studies that have also shown that when mAbs are exposed to an oxidizing agent (e.g., H2O2), the amount of Met 252 oxidation observed is about twice the amount of Met 428 oxidation (6, 21). With the results from our studies (with and without protein), it can be concluded that both stabilized and unstabilized H2O2 was comparable.
Peroxide Residual and Oxidation of 10 mg/mL mAb2 in Phosphate Buffer at pH 6.2 Spiked with 500 ng/mL H2O2 at 30 °C Storage
Conclusion
H2O2 spiking studies are valuable for determining the sensitivity of a protein drug product that is exposed to residual amounts of H2O2 during fill and finish processes. A thorough understanding of the kinetics between Met oxidation and H2O2 consumption is a prerequisite to determine the need and extent of H2O2 spiking studies. Our kinetic modeling of the reaction between surface-exposed Met and H2O2 provides an outline to design a spiking study experiment for mAb formulations and a rationale to reduce effort and resources if the formulation has a high protein concentration. The kinetic modeling may also be considered a worst-case scenario in regards to H2O2 consumption, as other factors (e.g., excipient impact and/or additional surface-exposed Met) in the formulation can also contribute to the rate of H2O2 consumption.
The results from our studies are widely applicable to mAbs and different mAb formats, such as IgG1, IgG2, and IgG4 molecules as well as antibody drug conjugates, bispecific mAbs, and one-arm mAbs, because they all have similar Met oxidation sites in the Fc portion of the heavy chain.
Acknowledgments
The authors appreciate the support of this work from Drs. Jamie Moore, Dana Andersen, Michael Adler, and Hanns-Christian Mahler. Christopher Kuehl acknowledges receiving financial support at school provided by Dynamic Aspects of Chemical Biology Training Grant (T32 GM08545).
- © PDA, Inc. 2015