1,587
Views
2
CrossRef citations to date
0
Altmetric
Report

Identification and characterization of an unexpected isomerization motif in CDRH2 that affects antibody activity

, , , , , , , , , , , , , & show all
Article: 2215364 | Received 30 Jan 2023, Accepted 15 May 2023, Published online: 25 May 2023

ABSTRACT

Aspartic acid (Asp) isomerization is a spontaneous non-enzymatic post-translation modification causing a change in the structure of the protein backbone, which is commonly observed in therapeutic antibodies during manufacturing and storage. The Asps in Asp–Gly (DG), Asp–Ser (DS), and Asp–Thr (DT) motifs in the structurally flexible regions, such as complementarity-determining regions (CDRs) in antibodies, are often found to have high rate of isomerization, and they are considered “hot spots” in antibodies. In contrast, the Asp-His (DH) motif is usually considered a silent spot with low isomerization propensity. However, in monoclonal antibody mAb-a, the isomerization rate of an Asp residue, Asp55, in the aspartic acid-histidine-lysine (DHK) motif present in CDRH2 was found to be unexpectedly high. By determining the conformation of DHK motif in the crystal structure of mAb-a, we found that the Cgamma of the Asp side chain carbonyl group and the back bone amide nitrogen of successor His were in proximal contact, which facilitates the formation of succinimide intermediate, and the +2 Lys played an important role in stabilizing such conformation. The contributing roles of the His and Lys residues in DHK motif were also verified using a series of synthetic peptides. This study identified a novel Asp isomerization hot spot, DHK, and the structural-based molecular mechanism was revealed. When 20% Asp55 isomerization in this DHK motif occurred in mAb-a, antigen binding activity reduced to 54%, but the pharmacokinetics in rat was not affected significantly. Although Asp isomerization of DHK motif in CDR does not appear to have a negative impact on PK, DHK motifs in the CDRs of antibody therapeutics should be removed, considering the high propensity of isomerization and impact on antibody activity and stability.

Introduction

Therapeutic monoclonal antibodies (mAbs), in particular IgGs, are now an important class of medicines that have applications in various therapeutic areas, including cancer, inflammatory disease, organ transplantation, cardiovascular disease, pathogenic infection, respiratory disease, and ophthalmological disease, due to several desirable features, such as high specificity, high potency, good safety and long half-life.Citation1,Citation2 In the folded state, IgG contains three relatively independent fragments, two identical antigen binding fragments (Fabs) and a crystallizable fragment (Fc), linked by a hinge region.Citation3 Each Fab has six complementarity-determining regions (CDRs), including three CDRLs from light chain and three CDRHs from heavy chain, that in the structure form the antigen binding site. The formation of antigen-antibody complex is mostly governed by electrostatic and hydrophobic interactions between amino acid residues of CDRs and target epitopes.Citation4

Therapeutic mAbs degrade via multiple pathways during expression, purification, formulation, storage, and delivery. Post-translational modifications (PTMs) or chemical modifications of CDR amino acid residues may have negative effects on antigen binding, leading to a decrease in potency, and therefore characterization of chemical modifications is an important part of evaluating the stability of therapeutic mAbs.Citation5,Citation6 Asp isomerization is one of the major chemical modifications of proteins under typical processing formulation and storage conditions.Citation7 In mildly acidic buffers, aspartic acid (Asp) residues can form a cyclic imide intermediate, succinimide (Asu), via nucleophilic attack of the carbonyl group on the Asp side chain by the amide nitrogen on the backbone.Citation8,Citation9 The Asu loop is unstable under alkaline pH conditions and can be rapidly hydrolyzed into Asp and isoaspartic acid (isoAsp) residues (). Previous studies have reported a 1:3 ratio of Asp to isoAsp from the succinate intermediate. The rate of isomerization reaction depends on various factors, including pH, temperature, primary structure, higher order structure, ionic strength, and other buffer conditions. Generally, antibody isomerization is more readily observed after incubation at elevated temperature and low pH.Citation10–12 The formation of isoAsp may lead to an immune response and the loss of biological activity.Citation13,Citation14

Figure 1. The mechanism of aspartic acid isomerization via the formation of a succinimide intermediate (Asu). Theoretical monoisotopic masses of Asp, isoAsp residues and the succinimide intermediate are indicated.

The chemical structure of aspartic acid, iso-aspartic acid and the succinimide intermediate (Asu) showing the dehydration reaction of isomerization.
Figure 1. The mechanism of aspartic acid isomerization via the formation of a succinimide intermediate (Asu). Theoretical monoisotopic masses of Asp, isoAsp residues and the succinimide intermediate are indicated.

Previous studies with small flexible peptides have shown that the reactivity of the side chains of amino acids next to Asp has the greatest impact on the rate of isomerization.Citation15–17 This residue affects isomerization through steric, electrostatic, or catalytic interactions, and Gly, Ser, and Thr next to the Asp generally increase the rate of Asp isomerization. In addition, the conformational flexibility around Asp is also an important factor affecting isomerization propensity.Citation18,Citation19 The CDRs of antibodies bind to the therapeutic target and are mostly flexible in structure and solvent exposed. The observation of Asp isomerization in the CDRs of antibodies has become increasingly common. Irudayanathan et al. reported that backbone secondary structure, side-chain rotamer conformation and solvent accessibility were found to be key molecular indicators of Asp isomerization in 131 clinical-stage therapeutic antibodies.Citation20 The observation of 131 clinical-stage therapeutic antibodies showed that Asp–Gly, Asp–Ser, and Asp–Thr in the CDRs were often found to have high rates of Asp isomerization and the obligate Asu intermediate is not likely to occur when the n + 1 residue is bulky. The observations of isomerization at the DH motifs are expectedly rare, with the DH motif usually considered a silent spot with low isomerization propensity.Citation21

In this study, we found that the Asp55 in CDRH2 of an antibody, mAb-a, underwent very fast isomerization, and that this modification affected the target binding activity of the antibody. However, isomerization of Asp55 did not affect the pharmacokinetics (PK) of mAb-a in rat. Further characterization of the isomerization of Asp55 of mAb-a revealed a novel isomerization motif, DHK, and the structural roles that His and Lys of this motif played in increasing Asp isomerization propensity.

Results

Dramatic increase of basic peak species of mAb-a incubated under mildly acidic conditions

Samples of mAb-a stored under mildly acidic pH conditions (5.0 and 6.0) were analyzed by cation ion exchange (CIEX) chromatography, and showed charge variant heterogeneity (). The chromatogram consisted of three fractions designated M, B1, and B2, of which both B1 and B2 were the basic peaks of mAb-a, indicating charge heterogeneity of mAb-a. As the incubation time increased, the ratio of the basic peaks (B1 and B2) of mAb-a also increased. After incubation at pH 5.0 and 25C for only 5 days, the level of the base peaks increased dramatically from 27.1% to 72.9%, and the level of the main peaks decreased from 57.0% to 21.4%. Moreover, after incubation at pH 6.0 for 5 days, the basic peaks of mAb-a also increased significantly to approximately 45.8% (). Surprisingly, under neutral conditions (pH 7.0) and extremely acidic conditions (pH 3.5), the base peaks of mAb-a did not change significantly in relative intensity. In summary, the CIEX chromatograms indicated that mAb-a underwent a rapid change over a narrow range of pH of around pH 5.0, specifically under slightly acidic conditions. The main peak and basic peaks (B1 and B2) of pH 5, 5 days pre-incubated sample were individually collected and analyzed through intact mass spectrometry (MS). The results revealed that the B1 peak had a molecular mass 18 Da lower than that of the main peak, while B2 peak had a 36 Da lower mass. These findings indicate that succinimide, rather than IsoD, was the primary isomerization product following mild acidic treatment (). Moreover, the B1 and B2 peaks were attributed to species with a single heavy chain and both heavy chains containing succinimide modifications, respectively.

Figure 2. Identification of the charge variants in mAb-a pre-incubated under different pH conditions. (A) Cation-exchange HPLC (CEX) profiles of mAb-a pre-incubated at different pH values (pH 5.0, 6.0, and 7.0) and 25°C over a period of 5 days. The two basic peaks with significant intensity increases after acidic pH pre-incubation, B1 and B2, are indicated in the chromatograms. (B) Identification of B1 peak and B2 peak in pH5, 5 day treated mAb-a by intact mass analysis. The B1 peak has 18 Da lower mass while B2 peak has 36 Da lower mass compared to the main peak.

(A) A four section CEX profiles showing main peak and basic peaks B1/B2 of mAb-a with different treatment (control, and pH 5.0/6.0/7.0). In the figure, there are three separate chromatograms, one for each pH value. Two basic fractions B1 and B2 with significant intensity increases in pH 5.0 treatment. (B) Intact mass of charge variant peaks B1 and B2, the B1 peak had an 18 Da lower molecular mass while the B2 peak had a 36 Da lower molecular mass.
Figure 2. Identification of the charge variants in mAb-a pre-incubated under different pH conditions. (A) Cation-exchange HPLC (CEX) profiles of mAb-a pre-incubated at different pH values (pH 5.0, 6.0, and 7.0) and 25°C over a period of 5 days. The two basic peaks with significant intensity increases after acidic pH pre-incubation, B1 and B2, are indicated in the chromatograms. (B) Identification of B1 peak and B2 peak in pH5, 5 day treated mAb-a by intact mass analysis. The B1 peak has 18 Da lower mass while B2 peak has 36 Da lower mass compared to the main peak.

Table 1. CEX and post-translation modification (PTM) analyses of mAb-a pre-incubated under different pHs.

Isomerization occurs at Asp55 in CDRH2

To identify the major changes in mAb-a at low pH, mAb-a was incubated under different pH conditions (pH 5.0, 6.0, and 7.0) at room temperature, and tryptic peptide mapping was used to measure the chemical modification levels. We found that under mildly acidic pH conditions, a high proportion of succinimide was formed from Asp55 in CDRH2 of mAb-a. As mentioned above, succinimide is the intermediate of aspartic acid isomerization. shows the combined extracted ion chromatograms of tryptic peptide P1 containing aspartic acid, isoaspartic acid, or succinimide extracted from mAb-a incubated under the indicated pHs. P1 contains two aspartic acids (Asp52 and Asp55) that can potentially undergo isomerization. In the chromatogram of peptide P1 containing the succinimide residue, there were multiple peaks corresponding to the calculated b and y ions, confirming the presence of the succinimide residue at the position of Asp55 (). At the retention time of 30.54 min, the observed peak corresponded to m/z of 699.08 (3+), which matched the calculated molecular weight, and this peak was assigned to the peptide with the original aspartic acid residue. The peak with the retention time of 31.24 min had the same molecular weight as the main peak, and MS/MS indicated that the two peaks had the “same” amino acid sequence, indicating that the later peak of 31.24 min could correspond to the same peptide containing the isoaspartic acid residue. The calculated m/z of the peak at 29.75 min was 694.58, which was 18 Da below the theoretical molecular weight and thus corresponded to a peptide with the succinimide residue. Because Asp and isoAsp were indistinguishable in the MS/MS spectra, synthetic peptide P1 and its isoAsp isomer, iso-P1, were generated for verification of peak identification. The synthetic peptide P1 was eluted at 30.54 min, while iso-P1 had the same retention time at 31.24 min as each later eluting peak, which confirmed the later eluting peak at 31.24 min was isomerized peptide iso-P1 (data not shown).

Figure 3. Extracted ion chromatograms of tryptic peptides of mAb-a that were pre-incubated at pH 5.0, 6.0, or 7.0 over five days and contain either aspartate, iso-aspartate, or succinimide (A). The figure also includes MS/MS spectra of the tryptic peptides that contain aspartate or iso-aspartate (B) and succinimide (C) located at amino acid residue position 55. The spectra and chromatograms indicate the presence of Asp55 isomerization products in the samples.

(A) Extracted ion chromatograms of tryptic target peptides of mAb-a incubated at pH 5.0/6.0/7.0, Asu-peptide, aspartate peptide and iso-aspartate peptide were sequentially eluted. The intensity of iso-aspartate peptide significantly increased. (B) MS/MS spectrum of iso-aspartate peptide shows matched b/y ions. (C) MS/MS spectrum of Asu-peptide shows matched b/y ions.
Figure 3. Extracted ion chromatograms of tryptic peptides of mAb-a that were pre-incubated at pH 5.0, 6.0, or 7.0 over five days and contain either aspartate, iso-aspartate, or succinimide (A). The figure also includes MS/MS spectra of the tryptic peptides that contain aspartate or iso-aspartate (B) and succinimide (C) located at amino acid residue position 55. The spectra and chromatograms indicate the presence of Asp55 isomerization products in the samples.

The intensity of peaks eluting later (31.24 min) or earlier (29.75 min) increased under acidic (pH 5.0 or 6.0) incubation conditions. As shown in , after mAb-a was incubated for 5 days at pH 5.0, the level of the isoAsp55 reached 26.5% and the level of succinimide reached 16.8%, indicating that modification at this Asp55 site was the major cause of basic peaks increase in the CIEX chromatograms. At the intact mass level, our results revealed that the primary isomerization product of mAb-a induced at acidic pH was succinimide. However, during peptide mapping analysis, the experimental conditions could disrupt the succinimide and convert it into either isoAsp or Asp. Therefore, the high proportion of isoAsp observed in the peptide mapping analysis was due to the degradation of succinimide over the analytical experiment process.

Asp55 isomerization affects the binding affinity of mAb-a

mAb-a was pre-incubated in different pH conditions (pH 5.0, 6.0, and 7.0) to determine the effect of isomerization on antibody binding potency. CHO-K1 cells that overexpressed the target antigen were used to determine the relative binding activity of mAb-a using fluorescence-activated cell sorting (FACS). As described in the methods section, we normalized the binding activity (measured by EC50) of untreated mAb-a as 100%. The relative binding activity of the treated sample was estimated by normalizing it against untreated mAb-a. The binding potency showed a clear trend of decrease over pre-treatment incubation under acidic pHs ().

Figure 4. The relative binding potency of mAb-a to target cells as measured by FACS. Panel a illustrates the varying potency of mAb-a samples, which were pre-incubated at different pHs to obtain different proportions of Asp55 isomerization. The different treatment conditions are color-coded (red for pH 5.0, blue for pH 6.0, and black for pH 7.0). The pre-incubation time are plotted on the x-axis, while the corresponding potency levels are on the y-axis. Panel B shows the correlation between the level of Asp55 modification (IsoD+asu%) and the binding potency of mAb-a.

(A) Three lines graph plotting the percentage of the potency of mAb-a with different proportion of isomerization. mAb-a treated in pH 5.0 with highest isomerization has the poorest binding affinity. (B) A line graph plotting the correlation between Asp55 isomerization level and potency of mAb-a. There is good linearity with R2 is 0.9732.
Figure 4. The relative binding potency of mAb-a to target cells as measured by FACS. Panel a illustrates the varying potency of mAb-a samples, which were pre-incubated at different pHs to obtain different proportions of Asp55 isomerization. The different treatment conditions are color-coded (red for pH 5.0, blue for pH 6.0, and black for pH 7.0). The pre-incubation time are plotted on the x-axis, while the corresponding potency levels are on the y-axis. Panel B shows the correlation between the level of Asp55 modification (IsoD+asu%) and the binding potency of mAb-a.

As previously discussed, succinimide was found to be the primary product of mAb-a isomerization under mild acidic pH conditions. However, during the liquid chromatography (LC)-MS peptide mapping analysis, a significant portion of succinimide was converted to isoAsp as well as Asp. Therefore, the sum of %isoD and %succinimide was used to assess the correlation between Asp55 isomerization and the relative binding potency of mAb-a. There was a good correlation between mAb-a potency and Asp isomerization; specifically, target cell binding activity decreased when the amount of isomeric Asp increased (). Size-exclusion chromatography (SEC) and capillary gel electrophoresis (CGE) testing results showed minimal changes in mAb-a aggregation and fragmentation levels for pH 5.0 and 6.0 incubated samples (data not shown). The results showed the isomerization of this Asp55 residue led to decreased antibody activity, and suggested Asp55 in CDRH2 was directly involved in target binding.

Isomerization of mAb-a Asp55 in vivo

To determine the rate of Asp55 isomerization in vivo, Sprague Dawley (SD) rats were injected with a dose of 10 mg/kg mAb-a pre-treated under indicated pH conditions for 5 days. mAb-a in rat plasma sample was captured and analyzed using the immunoprecipitation (IP)-LC-MS/MS method (). A portion of the captured antibody was digested by trypsin for PTM analysis using LC-MS/MS and another portion was used for antibody quantitation analysis.

Figure 5. The workflow of affinity capture LC-MS assay. Use immunoprecipitation with antigen to recover mAb-a from rat plasma. Following IdeS cleavage and tryptic digestion, peptides, including the liability peptide containing Asp55 residue, are detected using LC – MS/MS.

Antigen was conjunct with biotin and coated in magnetic beads with streptavidin. Immunoprecipitation with antigen was applied to recover mAb-a from rat plasma. Following IdeS cleavage and tryptic digestion, peptides, including the liability peptide containing Asp55 residue, are detected using LC–MS/MS.
Figure 5. The workflow of affinity capture LC-MS assay. Use immunoprecipitation with antigen to recover mAb-a from rat plasma. Following IdeS cleavage and tryptic digestion, peptides, including the liability peptide containing Asp55 residue, are detected using LC – MS/MS.

shows the relationship between isoAsp% and time after injection of different mAb-a in rats. After 7 days in vivo, the level of the isoAsp55 of mAb-a pre-treated at pH 5.0 (mAb-a-pH5), 6.0 (mAb-a-pH6), and 7.0 (mAb-a-pH5) increased by approximately 15%, 14%, and 14%, respectively. Although the plasma was considered to maintain the neutral pH (pH7.3-pH7.5) in vivo, it did not prevent the isomerization of Asp55 in mAb-a, indicating that mAb-a was unstable in vivo irrespective of the initial state of mAb-a Asp55, that is, pre-treatment of mAb-a under different pH conditions did not affect Asp55 isomerization rate in vivo.

Figure 6. Time-dependent changes in the proportion of mAb-a isomerization product (IsoD) after dosing in SD-rats. mAb-a was pre-incubated at different pH levels (pH 5.0, 6.0, and 7.0) for five days before intravenous injection in rats to achieve varying starting levels of Asp55 isomerization. At the indicated time point, mAb-a was affinity captured from rat plasma and analyzed by LC-MS peptide mapping to measure the percentage of IsoD. The lines representing the mAb-a pre-treatment conditions are color coded. (red for pH 5.0, blue for pH 6.0, and black for pH 7.0).

Three lines graph plotting the percentage of the isomerization on mAb-a in rat plasma with pre-incubated in pH 5.0, 6.0, and 7.0. There was a significant increase in isomerization of mAb-a after injection to rats.
Figure 6. Time-dependent changes in the proportion of mAb-a isomerization product (IsoD) after dosing in SD-rats. mAb-a was pre-incubated at different pH levels (pH 5.0, 6.0, and 7.0) for five days before intravenous injection in rats to achieve varying starting levels of Asp55 isomerization. At the indicated time point, mAb-a was affinity captured from rat plasma and analyzed by LC-MS peptide mapping to measure the percentage of IsoD. The lines representing the mAb-a pre-treatment conditions are color coded. (red for pH 5.0, blue for pH 6.0, and black for pH 7.0).

The succinimide intermediate was not detected by IP-LC-MS/MS. This was expected because the IP-LC-MS/MS experiment was performed in a slightly alkaline (pH 7.5) solution for optimal binding affinity. Under this pH condition, the succinimide intermediate could be rapidly hydrolyzed to form isoaspartic acid and aspartic acid,Citation22 and thus isoaspartic acid is the major modified species detected.Citation23,Citation24

Isomerization of Asp 55 has no impact on clearance of mAb-a in rat

MAb-a was pre-incubated under different pH conditions and then intravenously (IV) administered to rats. An ELISA-based quantitative assay was used to determine the mean concentration–time curves of mAb-a, as shown in . The results showed that mAb-a samples pre-incubated under different pH conditions appeared to have similar exposure over time profiles in rat.

Figure 7. Rat pharmacokinetics of mAb-a pre-treated under indicated pH conditions. Antibody serum concentrations are plotted against time post dosing in rat. A single intravenous dose of 10 mg/kg of mAb-a was administered to SD-rats. Results are presented as mean ± SD (n = 3 rats per time point × 2 replicate measurements).

Three lines graph plotting the antibody serum concentrations with pre-incubated in pH 5.0, 6.0, and 7.0. There was no significant difference between three conditions.
Figure 7. Rat pharmacokinetics of mAb-a pre-treated under indicated pH conditions. Antibody serum concentrations are plotted against time post dosing in rat. A single intravenous dose of 10 mg/kg of mAb-a was administered to SD-rats. Results are presented as mean ± SD (n = 3 rats per time point × 2 replicate measurements).

The parametric area under the curve from days 0 to 7 (AUC0–7), was highly comparable among the three groups (). The Cl of mAb-a-pH5 was 0.650 mL/h/kg, while the Cl of mAb-a-pH6 and mAb-a-pH7 was 0.628 and 0.632 mL/h/kg, respectively, both of which met the criteria for bioequivalence. Other PK parameters such as Cmax and half-life were also highly comparable. Thus, mAb-a containing different starting levels of Asp55 isomerization exhibited similar PK profiles in rat. To verify this observation, we established a standard curve using LC-MS/MS data and quantified the concentration of mAb-a in the plasma by the IP-LC-MS/MS method. The IP-LC MS/MS results were consistent with the ELISA results and provided orthogonal confirmation of the ELISA determination of antibody level in plasma (data not shown).

Table 2. Pharmacokinetic parameters of all stressed mAb-a in SD-rats.

Overall, antibody concentration over time profiles was similar among mAb-a with different Asp55 isomerization levels after a single IV dose of 10 mg/kg in SD-rats. There were no significant differences in all PK parameters among the mAb-a isomerization variants. The results showed that the isomerization at Asp55 appeared to have no significant impact on mAb-a PK.

Identification of DHK motif as an Asp isomerization hot spot

To investigate the molecular mechanism of high isomerization propensity of this Asp55 residue, the relative contribution of the primary sequence and structural conformation to this Asp isomerization was determined. A synthetic peptide P1 with the sequence corresponding to that adjoining Asp55 of mAb-a was incubated at pH 5.0, 6.0, and 7.0, with all samples at room temperature, for 7 days. The isomerization products were injected onto the same C18 column and assayed by RP-HPLC using the same gradient as in mAb-a tryptic peptide mapping.

The results were consistent with those of previous protein-level studies, showing a typical profile of isomerization peptide and with the amount of isomerized product increasing over time (). The isomerization level rose over seven days by 17%, 12%, and 2% at pH 5.0, 6.0, and 7.0, respectively. This result demonstrates that the synthetic peptide containing the primary sequence motif exhibited a similar isomerization rate as mAb-a, suggesting that the primary sequence may be the determining factor for the observed isomerization. To evaluate the relative roles of each adjacent residues surrounding Asp in the primary sequence motif, TSDHKT, we synthesized different mutant variants of peptide P1 to obtain peptides M1, M2, M3, M4, and M5, which were subjected to different pH conditions as above. The results are shown in . When H (position +1) or K (position +2) was mutated to alanine (A), the rate of Asp isomerization was significantly reduced and increased by only 5% within 7 days at pH 5.0. In contrast, the alanine mutations at the other 3 adjacent positions (positions −2, −1, and +3) appeared to have no significant impact on the isomerization rates. These results show that the DHK motif in the synthetic peptide is sufficient to promote Asp isomerization, confirming that the primary motif, DHK, plays a crucial role in Asp reactivity.

Figure 8. Isomerization reaction results of a synthetic peptide (P1) corresponding to the tryptic peptide found in mAb-a. The peptide was incubated for seven days at 25°C under indicated pH conditions. (red line: incubated at pH 5.0; blue line: incubated at pH 6.0; and black line: incubated at pH 7.0).

Three lines graph plotting the percentage of the isomerization on target tryptic peptide of mAb-a incubated at pH 5.0 at 25°C for seven days. The isomerization level increased over seven days by 17%, 12%, and 2% at pH 5.0, 6.0, and 7.0, respectively.
Figure 8. Isomerization reaction results of a synthetic peptide (P1) corresponding to the tryptic peptide found in mAb-a. The peptide was incubated for seven days at 25°C under indicated pH conditions. (red line: incubated at pH 5.0; blue line: incubated at pH 6.0; and black line: incubated at pH 7.0).

Figure 9. Isomerization reaction results of Asp55 in the synthetic P1 and mutant peptides incubated at pH 5.0 at 25°C for seven days.

Five different mutant variants of peptide P1 were synthesized to obtain peptides M1, M2, M3, M4, and M5 (P1: xxxTSDHKTxx; M1: xxxTSDAKTxx; M2: xxTSDHATxx; M3: xxASDHKTxx; M3: xxTADHKTxx; M3: xxTSDHKAxx). Six lines graph plotting the percentage of the isomerization on mutated peptides incubated at pH 5.0 at 25°C for seven days. The proportion of isomerization on M1 and M2 is significantly lower than that of control P1.
Figure 9. Isomerization reaction results of Asp55 in the synthetic P1 and mutant peptides incubated at pH 5.0 at 25°C for seven days.

As it was unexpected based on literature knowledge that the His at position +1 and Lys at position +2 played important roles in Asp isomerization propensity, we hypothesized that this high propensity might be due to some structural conformation. The X-ray crystal structure of mAb-a Fab fragment was determined at 1.61 angstrom resolution (PDB:8JBJ). From this structure, Asp55 of mAb-a is located in a loosely ordered structure ().

Figure 10. The crystal structure of mAb-a Fab (PDB:8JBJ). (A) Overall structure of mAb-a Fab region, with the heavy chain in magenta and the light chain in yellow. The DHK motif, located in the HCDR2 is highlighted in green. (B) a detailed view of the DHK motif and its surrounding residues. All residues are depicted in stick form, and hydrogen bonds are indicated by yellow dash lines.

(A) Crystal structure of Fab region mAB-a, DHK motif which located on the CDR2 of heavy chain is highlighted in magenta. (B) Detailed interactions of DHK motif with surrounding residues.
Figure 10. The crystal structure of mAb-a Fab (PDB:8JBJ). (A) Overall structure of mAb-a Fab region, with the heavy chain in magenta and the light chain in yellow. The DHK motif, located in the HCDR2 is highlighted in green. (B) a detailed view of the DHK motif and its surrounding residues. All residues are depicted in stick form, and hydrogen bonds are indicated by yellow dash lines.

It is known that the successor residue of a reactive Asp residue had a greater effect on the reaction rates than the residues on the N-terminal.Citation25–29 The reactivity of Asp may be affected by the structure of its successor residue, including steric hindrance, residue flexibility, and ionization state.Citation30,Citation31 Studies focusing on the primary sequence have suggested that the DH motif is not a hot spot for isomerization. As shown in , the side chain of His56 forms two hydrogen bonds with its own backbone carbonyl and backbone nitrogen of Arg72. These two hydrogen bonds stabilize the conformation that exposes the backbone amide nitrogen of His56. In addition, the salt bridge between the side chain of Lys57 and Asp55 further stabilize the ring-like conformation and narrow the distance between Cgamma of Asp55 and backbone nitrogen of His56. Therefore, the special structure of His56 and Lys57 contribute to the high rate of Asp isomerization. Moreover, synthetic DHK-containing peptide showed a similar conformation to the antibody structure through structural simulations, which were consistent with the high rate of Asp isomerization. Taken together, both His56 and Lys57 made a substantial contribution to the Asp isomerization observed in both antibody and DHK-containing peptide, indicating the DHK motif leads to a high propensity of Asp isomerization.

Discussion

In this study, we found that isomerization of mAb-a Asp55, which is not in a conventional isomerization hot spot, occurred at a high rate and changed the basic peak levels of mAb-a in the CIEX analyses. In vivo experimental results showed that mAb-a Asp55 was also unstable in rat plasma at neutral pH, and the level of isomerized Asp55 increased 7 days post dose in rat. The Asp55 was located in the CDRH2 region of mAb-a, and we found that Asp55 isomerization affected the binding between the antibody and the target, thereby reducing the potency. However, Asp55 isomerization did not significantly affect the PK profile of mAb-a in rat, probably owing to the overall conformational stability of Asp55 isomerized mAb-a variant.

To determine the mechanism of the high rate of Asp55 isomerization in the CDR, we further analyzed the adjacent primary sequence and local structural features from the crystal structure of the mAb-a Fab. We found that mutation of the DHK motif, that is, replacing histidine or lysine with alanine, dramatically slowed down Asp isomerization. From detailed structural analyses, the DHK motif is in the flexible β-turn, which is completely exposed and close in space. The +2 Lys residue forms a direct salt bridge with the side chain of Asp, which further reduces the distance between carbon atom of carboxylic acid side chain of Asp and backbone amide nitrogen of + 1 His. These effects indicate how the DHK motif in such a conformation promotes the isomerization of Asp.

DHK motif is an unexpected hot spot that undergoes fast isomerization, which is a part of the critical quality attributes of the therapeutic antibody mAb-a. The identification of this DHK motif expands our understanding of the mechanism and kinetics associated with the aspartic acid isomerization reaction. It also provides atomic level insight into a favorable structure for the reaction. The newly identified DHK isomerization motif and structural insight of Asp isomerization may facilitate a better molecular design in the discovery stage of therapeutic biologics to minimize Asp isomerization risk.

Materials and methods

Materials

MAb-a and its recognized Antigen-a were produced at BeiGene. The specific peptides were synthesized at the BankPeptide Co. (China). Dynabeads™ M-280 Streptavidin and EZ-Link™ NHS-biotin (Thermo Scientific) were used for IP-LC-MS/MS. Modified trypsin was purchased from Promega and IdeS was purchased from FabRICATOR. Other chemical reagents used in experiments were from Sigma.

MAb-a was incubated in 20 mM NaAc buffer (pH5.0), 20 mM Histidine-HCl buffer (pH 6.0), and 20 mM Tris-HCl buffer (pH7.0) at room temperature for 2 h, 6 h, 24 h, 48 h and 120 h to obtain mAb-a-pH5, mAb-a-pH6, mAb-a-pH7 samples.

Cation-exchange HPLC and fraction purification

Cation-exchange chromatography was performed by salt gradient separation on a MabPac SCX-10 (Thermo Fisher Scientific) column using an Agilent 1200 HPLC system. The elution system had a gradient change from 0% to 50% B in 20 min at a flow rate of 1 ml/min, while mobile phase A was 20 mM sodium phosphate, pH 6.5, and mobile phase B was 20 mM sodium phosphate, 0.2 M NaCl, pH 6.5. The column temperature was kept at 37°C and UV absorbance was measured at 280 nm with Agilent G4212B Diode Array Detector. Data evaluation was performed via the evaluation function of chromatography software 32Karat. Below peak (main peak) or peak groups (acidic peak group, basic peak group) were identified for samples or standards.

Fluorescence-activated cell sorting by target expressing CHO-K1 cells

MAb-a samples were diluted to 1 mg/mL, and then serially diluted in 96-well V-bottom plates at 1:4 ratio to a final concentration to 96-well plate. The concentrations of 10 points-serial dilution are as follows: 30 μg/mL, 7.5 μg/mL, 1.875 μg/mL, 0.46875 μg/mL, 0.117188 μg/mL, 0.029297 μg/mL, 0.007324 μg/mL, 0.001831 μg/mL, 0.000458 μg/mL, and 0.000114 μg/mL.

5 × 10Citation4 cells/well were resuspended and distributed in 200 μL of FACS buffer to new V-bottom 96-well plates, and blocked for 20 min. After blocking, the cells were added with 100 μL/well pre-diluted mAb-a samples, incubated at 4°C for 1 h, and washed by FACS buffer three times. After that, cells were added with Goat IgG F(ab’)2 anti-human IgG [F(ab’)2 specific]-488 (Jackson, 109-546-097, 1:200) as the secondary antibody, incubated at 4°C for 0.5 h, and washed by FACS buffer thrice. Following by the fixation with 1% paraformaldehyde, the cells were subjected to flow cytometry with Guava easyCyte 6HT Flow Cytometer (Millipore, USA).

PTM identification by LC-MS/MS

2.4 mg mAb-a samples were denatured with 8 M GuHCl in 200 mM Histidine-HCl solution (pH 6.0) to obtain 120 μL, 2 mg/mL samples and then slightly vortex for a short time. 1.2 µL of 1.0 M dithiothreitol solution was added followed by incubation at 56°C for 1 h for reduction. After addition of 2.4 µL of the freshly prepared 1 M iodoacetamide solution, the samples were incubated at room temperature in dark for 30 min. MAb-a were then buffer-exchanged into 120 μL of 20 mM Histidine-HCl solution by Zeba Spin desalting column (Thermo Scientific, 0.5 mL Ref# 89882) following the manufacturer’s protocol. After desalting treatment, the concentration of mAb-a was determined by using Nanodrop with the UV absorption at 280 nm to confirm the loss of desalting step. Before digestion, samples were diluted with 20 mM, pH 6.0 Histidine-HCl buffer at a final concentration of 1.0 mg/mL, and tryptic digestion was performed at 37°C for 30 min using an enzyme/protein weight ratio of 1/20. Formic acid (FA) solution is added to sample to stop the digestion (final concentration 1% FA). After centrifugation, 5 µg sample in supernatant was injected into LC/MS. Digested samples can be stored at −80°C up to 30 days until LC-MS analysis.

RP-HPLC-MS/MS analysis of peptides

The synthetic peptides were incubated in 20 mM NaAc buffer (pH5.0), 20 mM Histidine-HCl buffer (pH 6.0), and 20 mM Tris-HCl buffer (pH7.0), respectively, at room temperature for 2 h, 6 h, 24 h, 48 h, 72 h, 120 h and 168 h. The incubated products were separated using a CSH-C18, 2.1 × 100 mm, 1.7 µm column (Waters) at 65°C with a flow rate of 0.2 ml/min. The chromatographic condition was the same as that in the PTM identification method.

Rat PK study

Male SD-rats were dosed at 10 mg/kg (mAb-a-pH5, mAb-a-pH6, mAb-a-pH7) during a 5-min intravenous infusion for three replicates. Blood from rats was collected via a vascular access port at 5 min, 2, 6, 24, 48, 120, and 168 h, and processed for plasma. The plasma samples were stored at −80°C until IP-LC-MS/MS and ELISA analysis. All procedures were performed in accordance with protocols approved by the Institutional Animal Care and Use Committee (IACUC).

IP-LC-MS/MS

Antigen-a was biotinylated by mixing with biotin-NHS at w/w 10:1 ratio and incubating at room temperature for 1 h. Then extra Biotin-NHS was removed by Zeba desalting column, and biotinylated antigen-a was exchanged to HBS-EP buffer. 7 mL of 10 mg/mL M280 Streptavidin Dynabeads was mixed with 1 mg biotinylated antigen-a at room temperature for 1 h after 3 times wash with HBS – EP buffer (pH7.5). After incubation, Dynabeads were washed with 10 mL of HBS – EP buffer twice and Dynabeads binding with Antigen-a were resuspended in 7 mL HBS – EP buffer.

Samples (100 μL of rat plasma) and 100 μL of Dynabeads were mixed in a 96-well plate, capped, and placed on a shaker for overnight at 4°C. The Dynabeads were washed three times with HBS – EP buffer and resuspended with 300 uL of HBS – EP buffer containing IdeS, incubated at 37°C for 1 h with gentle shaking to remove Fc fragments. The Dynabeads were washed with HBS – EP buffer twice, followed by 500 uL of water twice and 10% acetonitrile once. Following the bead washing steps, a 100 μL aliquot of 10% acetonitrile with 1% formic acid was added to each sample and vortexed for 15 min at room temperature. The supernatant was the final product containing mAb-a Fab enriched from rat plasma. The PTM analysis of mAb-a Fab was followed by the PTM identification assay by LC-MS/MS described above.Citation32,Citation33

ELISA

The microtiter plate (Nunc, Cat#442404) was immobilized with 1 µg/mL antigen-a (prepared in house, 3 mg/mL) overnight at 2–8ºC. On the next day, the plate was washed and blocked by 3% bovine serum albumin (BSA) in PBST (PBS with 0.05% Tween-20) for 2 hours at room temperature. At the same time, the range of the mAb-a standard calibration samples were prepared from 0 ng/mL to 120 ng/mL in 3% BSA/PBST. Plasma samples were prepared at 0.1% required dilution in 3% BSA/PBST buffer. The samples were added to the plate and incubated at room temperature for 2 h. The unbound material was washed away and the captured mAb-a was detected by incubating with labeled goat anti-human polyclonal antibody at room temperature for 1 h. After thoroughly washing, streptavidin−horseradish peroxidase (Sigma, Cat#5512) was added and incubated for another 1 h. Following addition of 3,3′,5,5′-tetramethylbenzidine (Sigma, Cat#T0440) to each well, the plates were incubated at room temperature for 10 minutes and add stop solution (0.32 M H2SO4) to stop the reaction. The concentration of mAb-a in plasma samples was calculated from electrochemiluminescence intensity as measured by SpectraMax Paradigm using a 4-parameter logistic calibration curve generated from mAb-a calibrators.

Mab-a expression, purification, and crystallization

DNA sequences coding for the Fab of mAb-a were synthesized with codon-optimization in mammalian cells. The sequences of heavy and light chain of Fab were cloned into pMAX vector, respectively, with a C-terminal 6×HIS tag in heavy chain. The plasmids harboring heavy chain and light chain of mAb-a Fab were transiently co-transfected into HEK293 cells for protein expression. The supernatant containing secreted Fab was purified by TALON affinity resin (Clontech Laboratories), followed by further purification using a HiLoad 16/600 Superdex 75pg column (GE Healthcare Life Sciences). The purified protein of mAb-a Fab was concentrated to around 10 mg/ml in 20 mM Tris pH 8.0, 100 mM NaCl for initial crystallization screening. Crystals of mAb-a Fab were grown in 21% PEG3350, 0.4 M potassium formate. The crystals of mAb-a Fab cryoprotected with 21% PEG3350, 0.4 M potassium formate, 5% glycerol were flash frozen in liquid nitrogen. The X-ray diffraction data was collected at beamline BL45×U at Spring-8 synchrotron radiation facility (Hyogo, Japan).

Abbreviation

Fab=

antigen-binding fragment

Asp=

aspartic acid

CIEX=

cation ion exchange

CDRs=

complementarity determining regions

FACS=

fluorescence-activated cell sorting

mAbs=

monoclonal antibodies

isoAsp/isoD=

isoaspartic acid

PK=

pharmacokinetics

PTMs=

Post-translational modifications

Asu=

succinimide

Author contributions

The manuscript was written by Meiqi Yi. All authors have given approval to the final version of the manuscript.

Acknowledgments

We thank Liwen Bianji (Edanz) (www.liwenbianji.cn) for editing the language of a draft of this manuscript.

Disclosure statement

No potential conflict of interest was reported by the authors.

Additional information

Funding

The author(s) reported there is no funding associated with the work featured in this article.

References

  • Aires da Silva F, Corte-Real S, Goncalves J. Recombinant antibodies as therapeutic agents: pathways for modeling new biodrugs. BioDrugs. 2008;22(5):301–10. PMID: 18778112. doi:10.2165/00063030-200822050-00003.
  • Reichert JM, Rosensweig CJ, Faden LB, Dewitz MC. Monoclonal antibody successes in the clinic. Nat Biotechnol. 2005 Sep;23(9):1073–78. PMID: 16151394. doi:10.1038/nbt0905-1073.
  • Chames P, Van Regenmortel M, Weiss E, Baty D. Therapeutic antibodies: successes, limitations and hopes for the future. Br J Pharmacol. 2009 May;157(2):220–33. PMID: 19459844; PMCID: PMC2697811. doi:10.1111/j.1476-5381.2009.00190.x.
  • Spanov B, Aboagye V, Olaleye O, Govorukhina N, van de Merbel NC, Bischoff R, van de Merbel NC. Effect of Trastuzumab–HER2 complex formation on Stress-Induced modifications in the CDRs of trastuzumab. Front Chem. 2022 Jan 3;9:794247. PMID: 35047480; PMCID: PMC8762049. doi:10.3389/fchem.2021.794247.
  • Cacia J, Keck R, Presta LG, Frenz J. Isomerization of an aspartic acid residue in the complementarity-determining regions of a recombinant antibody to human IgE: identification and effect on binding affinity. Biochemistry. 1996 Feb 13;35(6):1897–903. 10.1021/bi951526c. PMID: 8639672.
  • Vlasak J, Bussat MC, Wang S, Wagner-Rousset E, Schaefer M, Klinguer-Hamour C, Kirchmeier M, Corvaïa N, Ionescu R, Beck A. Identification and characterization of asparagine deamidation in the light chain CDR1 of a humanized IgG1 antibody. Anal Biochem. 2009 Sep 15;392(2):145–54. doi: 10.1016/j.ab.2009.05.043. Epub 2009 Jun 2. PMID: 19497295.
  • Zhang J, Yip H, Katta V. Identification of isomerization and racemization of aspartate in the Asp-Asp motifs of a therapeutic protein. Anal Biochem. 2011 Mar 15;410(2):234–43. doi: 10.1016/j.ab.2010.11.040. Epub 2010 Dec 2. PMID: 21130067.
  • Geiger T, Clarke S. Deamidation, isomerization, and racemization at asparaginyl and aspartyl residues in peptides. Succinimide-linked reactions that contribute to protein degradation. J Biol Chem. 1987 Jan 15;262(2):785–94. 10.1016/S0021-9258(19)75855-4. PMID: 3805008.
  • Oliyai C, Borchardt RT. Chemical pathways of peptide degradation. IV. Pathways, kinetics, and mechanism of degradation of an aspartyl residue in a model hexapeptide. Pharm Res. 1993 Jan;10(1):95–102. PMID: 8430066. doi:10.1023/a:1018981231468.
  • Harris RJ, Kabakoff B, Macchi FD, Shen FJ, Kwong M, Andya JD, Shire SJ, Bjork N, Totpal K, Chen AB. Identification of multiple sources of charge heterogeneity in a recombinant antibody. J Chromatogr B Biomed Sci Appl. 2001 Mar 10;752(2):233–45. doi: 10.1016/s0378-4347(00)00548-x. PMID: 11270864.
  • Doyle HA, Zhou J, Wolff MJ, Harvey BP, Roman RM, Gee RJ, Koski RA, Mamula MJ. Isoaspartyl post-translational modification triggers anti-tumor T and B lymphocyte immunity. J Biol Chem. 2006 Oct 27;281(43):32676–83. doi: 10.1074/jbc.M604847200. Epub 2006 Sep 1. PMID: 16950786.
  • Yang ML, Doyle HA, Gee RJ, Lowenson JD, Clarke S, Lawson BR, Aswad DW, Mamula MJ. Intracellular protein modification associated with altered T cell functions in autoimmunity. J Immunol. 2006 Oct 1;177(7):4541–49. Erratum in: J Immunol. 2006 Dec 15;177(12):8878. PMID: 16982891. doi:10.4049/jimmunol.177.7.4541.
  • Chu GC, Chelius D, Xiao G, Khor HK, Coulibaly S, Bondarenko PV. Accumulation of succinimide in a recombinant monoclonal antibody in mildly acidic buffers under elevated temperatures. Pharm Res. 2007 Jun;24(6):1145–56. Epub 2007 Mar 24. PMID: 17385019. doi:10.1007/s11095-007-9241-4.
  • Ouellette D, Chumsae C, Clabbers A, Radziejewski C, Correia I. Comparison of the in vitro and in vivo stability of a succinimide intermediate observed on a therapeutic IgG1 molecule. MAbs. 2013 May-Jun;5(3):432–44. Epub 2013 Apr 22. PMID: 23608772; PMCID: PMC4169036. doi:10.4161/mabs.24458.
  • Radkiewicz JL, Zipse H, Clarke S, Houk KN. Neighboring side chain effects on asparaginyl and aspartyl degradation: an ab initio study of the relationship between peptide conformation and backbone NH acidity. J Am Chem Soc. 2001 Apr 18;123(15):3499–506. doi: 10.1021/ja0026814. PMID: 11472122.
  • Yan B, Steen S, Hambly D, Valliere-Douglass J, Vanden Bos T, Smallwood S, Yates Z, Arroll T, Han Y, Gadgil H, et al. Succinimide formation at Asn 55 in the complementarity determining region of a recombinant monoclonal antibody IgG1 heavy chain. J Pharm Sci. 2009 Oct; 98 (10): 3509–21. 10.1002/jps.21655: PMID: 19475547
  • Yu XC, Joe K, Zhang Y, Adriano A, Wang Y, Gazzano-Santoro H, Keck RG, Deperalta G, Ling V. Accurate determination of succinimide degradation products using high fidelity trypsin digestion peptide map analysis. Anal Chem. 2011 Aug 1;83(15):5912–19. doi: 10.1021/ac200750u. Epub 2011 Jul 1. PMID: 21692515.
  • Sydow JF, Lipsmeier F, Larraillet V, Hilger M, Mautz B, Mølhøj M, Kuentzer J, Klostermann S, Schoch J, Voelger HR, et al.Kettenberger H. Structure-based prediction of asparagine and aspartate degradation sites in antibody variable regionsPLos One2014 Jun 2496e10073610.1371/journal.pone.0100736 PMID: 24959685; PMCID: PMC4069079
  • Wakankar AA, Borchardt RT, Eigenbrot C, Shia S, Wang YJ, Shire SJ, Liu JL. Aspartate isomerization in the complementarity-determining regions of two closely related monoclonal antibodies. Biochemistry. 2007 Feb 13;46(6):1534–44. 10.1021/bi061500t. Epub 2007 Jan 17. PMID: 17279618.
  • Irudayanathan FJ, Zarzar J, Lin J, Izadi S. Deciphering deamidation and isomerization in therapeutic proteins: effect of neighboring residue. MAbs. 2022 Jan-Dec;14(1):2143006. PMID: 36377085; PMCID: PMC9673968. doi:10.1080/19420862.2022.2143006.
  • Lu X, Nobrega RP, Lynaugh H, Jain T, Barlow K, Boland T, Sivasubramanian A, Vásquez M, Xu Y. Deamidation and isomerization liability analysis of 131 clinical-stage antibodies. MAbs. 2019 Jan;11(1):45–57. Epub 2018 Dec 10. PMID: 30526254; PMCID: PMC6343770. doi:10.1080/19420862.2018.1548233.
  • Clarke S. Propensity for spontaneous succinimide formation from aspartyl and asparaginyl residues in cellular proteins. Int J Pept Protein Res. 1987 Dec;30(6):808–21. PMID: 3440704. doi:10.1111/j.1399-3011.1987.tb03390.x.
  • Doyle HA, Gee RJ, Mamula MJ. Altered immunogenicity of isoaspartate containing proteins. Autoimmunity. 2007 Mar;40(2):131–37. PMID: 17453712. doi:10.1080/08916930601165180.
  • Hambly DM, Banks DD, Scavezze JL, Siska CC, Gadgil HS. Detection and quantitation of IgG 1 hinge aspartate isomerization: a rapid degradation in stressed stability studies. Anal Chem. 2009 Sep 1;81(17):7454–59. doi: 10.1021/ac901258g. PMID: 19630420.
  • Xiao G, Bondarenko PV. Identification and quantification of degradations in the Asp-Asp motifs of a recombinant monoclonal antibody. J Pharm Biomed Anal. 2008 May 12;47(1):23–30. doi: 10.1016/j.jpba.2007.11.050. Epub 2007 Dec 14. PMID: 18201853.
  • Sreedhara A, Cordoba A, Zhu Q, Kwong J, Liu J. Characterization of the isomerization products of aspartate residues at two different sites in a monoclonal antibody. Pharm Res. 2012 Jan;29(1):187–97. Epub 2011 Aug 2. PMID: 21809161. doi:10.1007/s11095-011-0534-2.
  • Xie M, Aubé J, Borchardt RT, Morton M, Topp EM, Vander Velde D, Schowen RL. Reactivity toward deamidation of asparagine residues in beta-turn structures. J Pept Res. 2000 Sep;56(3):165–71. PMID: 11007273. doi:10.1034/j.1399-3011.2000.00764.x.
  • Spiekermann K, Hiddemann W. Molekulare Zielstrukturen in der Onkologie [Molecular target structures in oncology]. Internist (Berl). 2005 Aug;46(8):856–60. German PMID: 16007411. doi:10.1007/s00108-005-1463-0.
  • Robinson NE, Robinson AB. Molecular clocks. Proc Natl Acad Sci U S A. 2001 Jan 30;98(3):944–49. doi: 10.1073/pnas.98.3.944. PMID: 11158575; PMCID: PMC14689.
  • Robinson NE. Protein deamidation. Proc Natl Acad Sci U S A. 2002 Apr 16;99(8):5283–88. doi: 10.1073/pnas.082102799. PMID: 11959979; PMCID: PMC122761.
  • Capasso S, Balboni G, Di Cerbo P. Effect of lysine residues on the deamidation reaction of asparagine side chains. Biopolymers. 2000 Feb;53(2):213–19. PMID: 10679625. 10.1002/(SICI)1097-0282(200002)53:2<213:AID-BIP11>3.0.CO;2-C.
  • Liu YD, van Enk JZ, Flynn GC, van Enk JZ. Human antibody Fc deamidation in vivo. Biologicals. 2009 Oct;37(5):313–22. Epub 2009 Jul 15. PMID: 19608432. doi:10.1016/j.biologicals.2009.06.001.
  • Mehl JT, Sleczka BG, Ciccimaro EF, Kozhich AT, Gilbertson DG, Vuppugalla R, Huang CS, Stevens B, Mo J, Deyanova EG, Wang Y, et al. Quantification of in vivo site-specific Asp isomerization and Asn deamidation of mAbs in animal serum using IP-LC–MS. Bioanalysis. 2016 Aug;8(15):1611–22. doi: 10.4155/bio-2016-0035. Epub 2016 Jul 11. PMID: 27397670.