Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
J Pharm Sci ; 109(1): 566-575, 2020 01.
Article in English | MEDLINE | ID: mdl-31669653

ABSTRACT

During the manufacturing of protein biologics, product variability during cell culture production and harvest needs to be actively controlled and monitored to maintain acceptable product quality. To a large degree, variants that have previously been described are covalent in nature and are easily analyzed by a variety of techniques. Here, we describe a noncovalent post translational modification of recombinantly expressed antibodies, containing variable domain tryptophans, that are exposed to culture media components and ambient laboratory light. The modified species, designated as conformer, can be monitored by hydrophobic interaction chromatography and often exhibits reduced potency. We studied conformer formation and identified key elements driving its accelerated growth using an IgG2 monoclonal antibody. Conformer is a result of a noncovalent interaction of the antibody with riboflavin, an essential vitamin added to many production cell culture formulations. Chemical and physical factors that influence the impact of riboflavin are identified, and methods for process control of this product quality attribute are addressed in order to prevent loss of antibody potency and potential safety issues. Identifying therapeutic antibody drug candidates with the potential to form conformers can be performed early in development to avoid this undesirable product quality propensity.


Subject(s)
Antibodies, Monoclonal/metabolism , Culture Media/metabolism , Drug Contamination , Immunoglobulin G/metabolism , Protein Processing, Post-Translational , Riboflavin/metabolism , Tryptophan/chemistry , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/pharmacology , Antibody Affinity , Binding, Competitive , CHO Cells , Cell Culture Techniques , Cricetulus , Hydrophobic and Hydrophilic Interactions , Immunoglobulin G/biosynthesis , Immunoglobulin G/pharmacology , Light , Protein Binding , Recombinant Proteins/metabolism
2.
MAbs ; 7(5): 881-90, 2015.
Article in English | MEDLINE | ID: mdl-26186204

ABSTRACT

Regulatory agencies have recently recommended a Quality by Design (QbD) approach for the manufacturing of therapeutic molecules. A QbD strategy requires deep understanding at the molecular level of the attributes that are crucial for safety and efficacy and for insuring that the desired quality of the purified protein drug product is met at the end of the manufacturing process. A mass spectrometry (MS)-based approach to simultaneously monitor the extensive array of product quality attributes (PQAs) present on therapeutic molecules has been developed. This multi-attribute method (MAM) uses a combination of high mass accuracy / high resolution MS data generated by Orbitrap technology and automated identification and relative quantification of PQAs with dedicated software (Pinpoint). The MAM has the potential to replace several conventional electrophoretic and chromatographic methods currently used in Quality Control to release therapeutic molecules. The MAM represents an optimized analytical solution to focus on the attributes of the therapeutic molecule essential for function and implement QbD principles across process development, manufacturing and drug disposition.


Subject(s)
Antibodies, Monoclonal/chemistry , Biological Products/standards , Peptide Mapping/methods , Quality Control , Technology, Pharmaceutical/methods , Amino Acid Sequence , Chromatography, Affinity , Chromatography, Liquid , Mass Spectrometry , Molecular Sequence Data , Protein Processing, Post-Translational , Software
3.
Article in English | MEDLINE | ID: mdl-26079266

ABSTRACT

In this communication, a high-throughput method for automated data analysis of cysteine-related product quality attributes (PQAs) in IgG2 antibodies is reported. This method leverages recent advances in the relative quantification of PQAs to facilitate the characterization of disulfide variants and free sulfhydryls (SHs) in IgG2 antibodies. The method uses samples labeled with a mass tag (N-ethyl maleimide [NEM]) followed by enzymatic digestion under non-reducing conditions to maintain the cysteine connectivity. The digested IgG2 samples are separated and detected by mass spectrometry (MS) and the resulting peptide map is analyzed in an automated fashion using Pinpoint software (Thermo Scientific). Previous knowledge of IgG2 disulfide structures can be fed into the Pinpoint software to create workbooks for various disulfide linkages and hinge disulfide variants. In addition, the NEM mass tag can be added to the workbooks for targeted analysis of labeled cysteine-containing peptides. The established Pinpoint workbooks are a high-throughput approach to quantify relative abundances of unpaired cysteines and disulfide linkages, including complicated hinge disulfide variants. This approach is especially efficient for comparing large sets of similar samples such as those created in comparability and stability studies or chromatographic fractions. Here, the high throughput method is applied to quantify the relative abundance of hinge disulfide variants and unpaired cysteines in the IgG2 fractions from non-reduced reversed-phase high-performance liquid chromatography (nrRP-HPLC). The LC-MS data analyzed by the Pinpoint workbook suggests that the nrRP-HPLC separated peaks contain hinge disulfide isoforms and free cysteine pairs for each major disulfide isoform structure.


Subject(s)
Chromatography, High Pressure Liquid/methods , Chromatography, Reverse-Phase/methods , Cysteine/chemistry , Immunoglobulin G/analysis , Immunoglobulin G/chemistry , Mass Spectrometry/methods , Animals , CHO Cells , Cricetinae , Cricetulus , Disulfides , Humans , Protein Conformation , Recombinant Proteins/analysis , Recombinant Proteins/chemistry , Software
4.
Anal Bioanal Chem ; 407(13): 3851-60, 2015 May.
Article in English | MEDLINE | ID: mdl-25795027

ABSTRACT

The development of sensitive techniques to detect sequence variants (SVs), which naturally arise due to DNA mutations and errors in transcription/translation (amino acid misincorporations), has resulted in increased attention to their potential presence in protein-based biologic drugs in recent years. Often, these SVs may be below 0.1%, adding challenges for consistent and accurate detection. Furthermore, the presence of false-positive (FP) signals, a hallmark of SV analysis, requires time-consuming analyst inspection of the data to sort true from erroneous signal. Consequently, gaps in information about the prevalence, type, and impact of SVs in marketed and in-development products are significant. Here, we report the results of a simple, straightforward, and sensitive approach to sequence variant analysis. This strategy employs mixing of two samples of an antibody or protein with the same amino acid sequence in a dilution series followed by subsequent sequence variant analysis. Using automated peptide map analysis software, a quantitative assessment of the levels of SVs in each sample can be made based on the signal derived from the mass spectrometric data. We used this strategy to rapidly detect differences in sequence variants in a monoclonal antibody after a change in process scale, and in a comparison of three mAbs as part of a biosimilar program. This approach is powerful, as true signals can be readily distinguished from FP signal, even at a level well below 0.1%, by using a simple linear regression analysis across the data set with none to minimal inspection of the MS/MS data. Additionally, the data produced from these studies can also be used to make a quantitative assessment of relative levels of product quality attributes. The information provided here extends the published knowledge about SVs and provides context for the discussion around the potential impact of these SVs on product heterogeneity and immunogenicity.


Subject(s)
Algorithms , Amino Acid Substitution , Antibodies, Monoclonal/chemistry , Protein Interaction Mapping/methods , Recombinant Proteins/chemistry , Sequence Analysis, Protein/methods , Amino Acid Sequence , Antibodies, Monoclonal/genetics , Genetic Variation , Molecular Sequence Data , Protein Binding , Recombinant Proteins/genetics
5.
MAbs ; 6(6): 1453-63, 2014.
Article in English | MEDLINE | ID: mdl-25484040

ABSTRACT

During cell line development for an IgG1 antibody candidate (mAb1), a C-terminal extension was identified in 2 product candidate clones expressed in CHO-K1 cell line. The extension was initially observed as the presence of anomalous new peaks in these clones after analysis by cation exchange chromatography (CEX-HPLC) and reduced capillary electrophoresis (rCE-SDS). Reduced mass analysis of these CHO-K1 clones revealed that a larger than expected mass was present on a sub-population of the heavy chain species, which could not be explained by any known chemical or post-translational modifications. It was suspected that this additional mass on the heavy chain was due to the presence of an additional amino acid sequence. To identify the suspected additional sequence, de novo sequencing in combination with proteomic searching was performed against translated DNA vectors for the heavy chain and light chain. Peptides unique to the clones containing the extension were identified matching short sequences (corresponding to 9 and 35 amino acids, respectively) from 2 non-coding sections of the light chain vector construct. After investigation, this extension was observed to be due to the re-arrangement of the DNA construct, with the addition of amino acids derived from the light chain vector non-translated sequence to the C-terminus of the heavy chain. This observation showed the power of proteomic mass spectrometric techniques to identify an unexpected antibody sequence variant using de novo sequencing combined with database searching, and allowed for rapid identification of the root cause for new peaks in the cation exchange and rCE-SDS assays.


Subject(s)
Antibodies, Monoclonal/genetics , Codon, Terminator/genetics , DNA Mutational Analysis/methods , Immunoglobulin G/genetics , Tandem Mass Spectrometry/methods , Amino Acid Sequence , Animals , CHO Cells , Cations , Chromatography, Ion Exchange , Cricetinae , Cricetulus , DNA/genetics , DNA/metabolism , Electrophoresis, Capillary , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Light Chains/genetics , Molecular Sequence Data , Mutation , Peptide Mapping/methods , Proteomics/methods , Reproducibility of Results
6.
Protein Sci ; 19(11): 2152-63, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20836085

ABSTRACT

Novel acetone and aldimine covalent adducts were identified on the N-termini and lysine side chains of recombinant monoclonal antibodies. Photochemical degradation of citrate buffers, in the presence of trace levels of iron, is demonstrated as the source of these modifications. The link between degradation of citrate and the observed protein modifications was conclusively established by tracking the citrate decomposition products and protein adducts resulting from photochemical degradation of isotope labeled (13)C citrate by mass spectrometry. The structure of the acetone modification was determined by nuclear magnetic resonance (NMR) spectroscopy on modified-free glycine and found to correspond to acetone linked to the N-terminus of the amino acid through a methyl carbon. Results from mass spectrometric fragmentation of glycine modified with an acetone adduct derived from (13)C labeled citrate indicated that the three central carbons of citrate are incorporated onto protein amines in the presence of iron and light. While citrate is known to stoichiometrically decompose to acetone and CO(2) through various intermediates in photochemical systems, it has never been shown to be a causative agent in protein carbonylation. Our results point to a previously unknown source for the generation of reactive carbonyl species. This work also highlights the potential deleterious impact of trace metals on recombinant protein therapeutics formulated in citrate buffers.


Subject(s)
Acetone/chemistry , Antibodies, Monoclonal/chemistry , Citrates/chemistry , Recombinant Proteins/chemistry , Animals , Antibodies, Monoclonal/metabolism , CHO Cells , Carbon Isotopes/chemistry , Chromatography, High Pressure Liquid , Cricetinae , Cricetulus , Glycine/chemistry , Humans , Imines/chemistry , Immunoglobulin G , Iron/chemistry , Isotope Labeling , Mass Spectrometry , Nuclear Magnetic Resonance, Biomolecular , Photochemical Processes , Protein Carbonylation , Recombinant Proteins/metabolism
7.
Anal Chem ; 82(16): 6751-5, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20704363

ABSTRACT

Recombinant monoclonal antibodies are an important class of therapeutic agents that have found widespread use for the treatment of many human diseases. Here, we have examined the utility of ion mobility mass spectrometry (IMMS) for the rapid characterization of disulfide variants in intact IgG2 monoclonal antibodies. It is shown that IMMS reveals 2 to 3 gas-phase conformer populations for IgG2s. In contrast, a single gas-phase conformer is revealed using IMMS for both an IgG1 antibody and a Cys-232 --> Ser mutant IgG2, both of which are homogeneous with respect to disulfide bonding. This provides strong evidence that the observed IgG2 gas-phase conformers are related to disulfide bond heterogeneity. Additionally, IMMS analysis of redox enriched disulfide isoforms allows assignment of the mobility peaks to established disulfide bonding patterns. These data clearly illustrate how IMMS can be used to quickly provide information on the higher order structure of antibody therapeutics.


Subject(s)
Antibodies, Monoclonal/chemistry , Disulfides/chemistry , Immunoglobulin G/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Chromatography, High Pressure Liquid/methods , Gases/chemistry , Humans , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Mutation , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
8.
Anal Chem ; 82(9): 3510-8, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20364842

ABSTRACT

A capillary isoelectric focusing (cIEF) method has been developed as an alternative to cation exchange chromatography to determine charge heterogeneity for a therapeutic antibody. Characterization of the cIEF profile is important to understand the charged isoform distribution. A variety of preparative IEF methods have been developed over the years but have had various limitations including high levels of contaminating ampholytes and complex fractionation and isolation procedures. More recently, an off-line method that uses pI-based separation on immobilized pH gradients was developed to preparatively isolate material with convenient liquid phase recovery. This method uses the Agilent OFFGEL 3100 Fractionator and was optimized to produce fractions of antibody charge isoforms differing by as little as 0.1 pI units. The isolation of highly resolved fractions then allowed for the identification of N- and C-terminal basic charge modifications including noncyclized glutamine, signal peptide extensions, and various levels of C-terminal lysine processing and high mannose structures. These species could then be correlated to specific peaks in the cIEF profile. This work shows that a preparative IEF method using immobilized pH gradients can be optimized to generate highly resolved, pI-based fractions in solution which can be used for successful cIEF profile characterization. Access to preparative amounts of discrete charged species allows for a better understanding of the underlying covalent modifications responsible for the charge differences and facilitates evaluation of the impact of these modifications on stability and potency of therapeutic antibodies.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/analysis , Proton-Motive Force
9.
J Biol Chem ; 285(21): 16012-22, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20233717

ABSTRACT

We report the presence of oligosaccharide structures on a glutamine residue present in the V(L) domain sequence of a recombinant human IgG2 molecule. Residue Gln-106, present in the QGT sequence following the rule of an asparagine-linked consensus motif, was modified with biantennary fucosylated oligosaccharide structures. In addition to the glycosylated glutamine, analysis of a lectin-enriched antibody population showed that 4 asparagine residues: heavy chain Asn-162, Asn-360, and light chain Asn-164, both of which are present in the IgG1 and IgG2 constant domain sequences, and Asn-35, which was present in CDR(L)1, were also modified with oligosaccharide structures at low levels. The primary sequences around these modified residues do not adhere to the N-linked consensus sequon, NX(S/T). Modeling of these residues from known antibody crystal structures and sequence homology comparison indicates that non-consensus glycosylation occurs on Asn residues in the context of a reverse consensus motif (S/T)XN located on highly flexile turns within 3 residues of a conformational change. Taken together our results indicate that protein glycosylation is governed by more diversified requirements than previously appreciated.


Subject(s)
Antibodies, Monoclonal/chemistry , Asparagine/chemistry , Glutamic Acid/chemistry , Immunoglobulin G/chemistry , Oligosaccharides/chemistry , Protein Modification, Translational , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Asparagine/genetics , Asparagine/immunology , Glutamic Acid/genetics , Glutamic Acid/immunology , Glycosylation , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Models, Molecular , Oligosaccharides/genetics , Oligosaccharides/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology
10.
J Biol Chem ; 284(51): 35390-402, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-19850927

ABSTRACT

Hinge cleavage of a recombinant human IgG1 antibody, generated during production in a Chinese hamster ovary cell culture, was observed in the purified material. The cleavage products could be reproduced by incubation of the antibody with H(2)O(2) and featured complementary ladders of the C- and N-terminal residues (Asp(226)-Lys(227)-Thr(228)-His(229)-Thr(230)) in the heavy chain of the Fab domain and the upper hinge of one of the Fc domains, respectively. Two adducts of +45 and +71 Da were also observed at the N-terminal residues of some Fc fragments and were identified as isocyanate and alpha-ketoacyl derivatives generated by radical cleavage at the alpha-carbon position through the diamide and alpha-amidation pathways. We determined that the hinge cleavage was initiated by radical-induced breakage of the disulfide bond between the two hinge cysteines at position 231 (Cys(231)-Pro-Pro-Cys-Pro), followed by the formation of a thiyl radical (Cys(231)-S(*)) on one cysteine and sulfenic acid (Cys(231)-SOH) on the other. The location of the initial radical attack and the critical role of Cys(231) were demonstrated by the observation that 5,5-dimethyl-1-pyrroline N-oxide only reacted with the Cys(231) radical and completely blocked hinge cleavage, suggesting the necessity of an electron/radical transfer from the Cys(231) radical to the hinge residues where cleavage was observed. As a precursor of hydroxyl radicals, H(2)O(2) is widely produced in healthy cells and tissues and therefore could be the source for the radical-induced fragmentation of human IgG1 antibodies in vivo.


Subject(s)
Hydrogen Peroxide/chemistry , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin G/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin G/genetics , Oxidation-Reduction , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
11.
J Biol Chem ; 284(47): 32493-506, 2009 Nov 20.
Article in English | MEDLINE | ID: mdl-19767389

ABSTRACT

We report that N-linked oligosaccharide structures can be present on an asparagine residue not adhering to the consensus site motif NX(S/T), where X is not proline, described in the literature. We have observed oligosaccharides on a non-consensus asparaginyl residue in the C(H)1 constant domain of IgG1 and IgG2 antibodies. The initial findings were obtained from characterization of charge variant populations evident in a recombinant human antibody of the IgG2 subclass. HPLC-MS results indicated that cation-exchange chromatography acidic variant populations were enriched in antibody with a second glycosylation site, in addition to the well documented canonical glycosylation site located in the C(H)2 domain. Subsequent tryptic and chymotryptic peptide map data indicated that the second glycosylation site was associated with the amino acid sequence TVSWN(162)SGAL in the C(H)1 domain of the antibody. This highly atypical modification is present at levels of 0.5-2.0% on most of the recombinant antibodies that have been tested and has also been observed in IgG1 antibodies derived from human donors. Site-directed mutagenesis of the C(H)1 domain sequence in a recombinant-human IgG1 antibody resulted in an increase in non-consensus glycosylation to 3.15%, a greater than 4-fold increase over the level observed in the wild type, by changing the -1 and +1 amino acids relative to the asparagine residue at position 162. We believe that further understanding of the phenomenon of non-consensus glycosylation can be used to gain fundamental insights into the fidelity of the cellular glycosylation machinery.


Subject(s)
Antibodies/chemistry , Asparagine/chemistry , Immunoglobulin G/chemistry , Oligosaccharides/chemistry , Antibodies/metabolism , Chromatography, High Pressure Liquid , Chymotrypsin/pharmacology , Glycosylation , Humans , Hydrogen Bonding , Mass Spectrometry/methods , Mutagenesis, Site-Directed , Peptides/chemistry , Protein Processing, Post-Translational , Protein Structure, Tertiary , Trypsin/pharmacology
12.
J Pharm Sci ; 98(10): 3509-21, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19475547

ABSTRACT

We investigated the formation and stability of succinimide, an intermediate of deamidation events, in recombinant monoclonal antibodies (mAbs). During the course of an analytical development study of an IgG1 mAbs, we observed that a specific antibody population could be separated from the main product by cation-exchange (CEX) chromatography. The cell-based bioassay measured a approximately 70% drop in potency for this fraction. Liquid chromatography time-of-flight mass spectrometry (LC-TOF/MS) and tandem mass spectrometry (LC-MS/MS) analyses showed that the modified CEX fraction resulted from the formation of a succinimide intermediate at Asn 55 in the complementarity determining region (CDR) of the heavy chain. Biacore assay revealed a approximately 50% decrease in ligand binding activity for the succinimide-containing Fab with respect to the native Fab. It was found that the succinimide form existed as a stable intermediate with a half-life of approximately 3 h at 37 degrees C and pH 7.6. Stress studies indicated that mildly acidic pH conditions (pH 5) favored succinimide accumulation, causing a gradual loss in potency. Hydrolysis of the succinimide resulted in a further drop in potency. The implications of the succinimide formation at Asn 55, a highly conserved residue among IgG1 (mAbs), are discussed.


Subject(s)
Antibodies, Monoclonal/chemistry , Asparagine/chemistry , Complementarity Determining Regions/chemistry , Immunoglobulin G/chemistry , Immunoglobulin Heavy Chains/chemistry , Succinimides/chemical synthesis , Chromatography, High Pressure Liquid , Chromatography, Ion Exchange , Half-Life , Humans , Hydrogen-Ion Concentration , Hydrolysis , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fc Fragments/chemistry , Ligands , Mass Spectrometry , Papain/chemistry , Peptide Mapping , Trypsin
13.
Biochemistry ; 48(17): 3755-66, 2009 May 05.
Article in English | MEDLINE | ID: mdl-19254029

ABSTRACT

Human IgG2 exists as a mixture of disulfide-linked structural isoforms that can show different activities. To probe the contribution of specific cysteine residues to the formation of structural isoforms, we characterized a series of Cys-->Ser mutant IgG2 recombinant monoclonal antibodies, focused on the first C(H)1 cysteine and the first two hinge cysteines. These residues participate in the formation of structural isoforms that have been noted by nonreduced capillary sodium dodecyl sulfate polyacrylamide gel electrophoresis, reversed-phase high-performance liquid chromatography, and cation exchange chromatography. We show that single Cys-->Ser mutants can greatly reduce heterogeneous disulfide bonding in human IgG2 and maintain in vitro activity. The data demonstrate the feasibility of applying site-directed mutagenesis to reduce disulfide bond heterogeneity in human IgG2 while preserving the activity of this therapeutically important class of human antibodies.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Disulfides/chemistry , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Mutagenesis, Site-Directed , Amino Acid Substitution/genetics , Antibodies, Monoclonal/metabolism , Cysteine/genetics , Cysteine/metabolism , Disulfides/metabolism , Electrophoresis, Polyacrylamide Gel , Humans , Immunoglobulin G/metabolism , Immunoglobulin kappa-Chains/chemistry , Immunoglobulin kappa-Chains/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Mapping , Recombinant Proteins/chemistry , Recombinant Proteins/classification , Recombinant Proteins/metabolism , Serine/genetics , Serine/metabolism , Spectrometry, Mass, Electrospray Ionization
14.
Glycobiology ; 19(2): 144-52, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18952827

ABSTRACT

We describe the characterization of an O-fucosyl modification to a serine residue on the light chain of a recombinant, human IgG1 molecule expressed in Chinese hamster ovary (CHO) cells. Cation exchange chromatography (CEX) and hydrophobic interaction chromatography (HIC) were used to isolate a Fab population which was 146 Da heavier than the expected mass. Isolated Fab fragments were treated with a reducing agent to facilitate mass spectrometric analysis of the reduced light chain (LC) and fragment difficult (Fd). An antibody light chain with a net addition of 146 Da was detected by mass spectrometric analysis of the modified Fab. A light chain tryptic peptide in complementarity determining region-1 (CDR-1) was subsequently identified with a net addition of 146 Da by a peptide map. Results from a nanospray infusion of the modified peptide into a linear ion trap mass spectrometer with electron transfer dissociation (ETD) functionality indicated that the modified residue was a serine at position 30 in the light chain. Acid hydrolysis of the modified tryptic peptide followed by fluorescent labeling with 2-aminoanthranilic acid (2AA) and HPLC comparison with monosaccharide standards confirmed the presence of fucose on the light chain peptide. The presence of O-fucose on an antibody has not been previously reported. Currently, O-fucose has been described as occurring on mammalian proteins with amino acid sequence motifs associated with epidermal growth factor (EGF)-like repeats or thrombospondin type 1 repeats (TSRs). The amino acid sequence around the modified Ser in the IgG1 molecule does not conform to any known O-fucosylation sequence motif and thus is the first description of this type of modification on a nonconsensus sequence in a mammalian protein.


Subject(s)
Fucose/metabolism , Immunoglobulin G/chemistry , Immunoglobulin Light Chains/metabolism , Animals , Antibodies/chemistry , Antibodies/metabolism , CHO Cells , Cricetinae , Cricetulus , Humans , Immunoglobulin G/metabolism , Immunoglobulin Light Chains/chemistry , Mass Spectrometry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
15.
J Chromatogr A ; 1214(1-2): 81-9, 2008 Dec 19.
Article in English | MEDLINE | ID: mdl-19012891

ABSTRACT

The following report describes the use of hydrophobic-interaction chromatography (HIC) to separate and characterize populations of monoclonal antibodies resulting from variable N- and C-terminal processing, stressed-induced covalent modifications and conformationally altered populations present in the drug product. We investigated the use of HIC to characterize heterogeneity in the intact molecule and the Fab and Fc sub-domains resulting from papain cleavage. We found that certain classes of covalent modifications to antibodies are highly amenable to HIC separation. Specific covalent modifications occurring on antibodies could be separated into pure fractions which contained unmodified, singly modified (on 1 heavy or light chain) and doubly modified (on both heavy or light chains) molecules. This report demonstrates the utility of HIC for assessing the heterogeneity, stability and, in some cases, potency of monoclonal antibodies.


Subject(s)
Antibodies, Monoclonal/isolation & purification , Chromatography/methods , Hydrophobic and Hydrophilic Interactions , Antibodies, Monoclonal/chemistry , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/isolation & purification , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/isolation & purification , Isomerism , Papain/metabolism , Protein Stability , Succinimides/metabolism
16.
Anal Chem ; 80(18): 7022-8, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18707131

ABSTRACT

A hydrophobic interaction chromatography method was developed to analyze recombinant soluble Interleukin 1 receptor type II (sIL-1R type II) drug substance and assess the stability of the drug under accelerated degradation studies. HIC resolved the degraded molecules into three peaks. A combination of several analytical techniques, including cyanogen bromide cleavage, reversed-phase chromatography, mass spectrometry, and N-terminal sequencing, were used to identify the origins of these peaks. We found that accelerated degradation resulted from three different events, deamidation and isomerization at asparagine 317 (Asn317), C-terminal cleavage, and aggregation. The iso-aspartate 317 (iso-Asp317)-containing species were shown to elute in HIC peak I and the Asp317-containing species in HIC peak II, respectively. Deamidation-isomerization to iso-Asp317, but not deamidation to Asp317, resulted in altered retention time on HIC companied by loss of potency, presumably by introducing a significant conformational change. CNBr C-terminal analysis showed that the inactive HIC peak I consisted of sIL-1R type II with "large" C-terminal truncations of 13 or 14 amino acids, whereas the active HIC peak II contained C-terminally full length and "small" C-terminal clips of two amino acids. Molecular modeling indicates that the short loop D317-S320, in the third domain of IL-1R type II, has a crucial impact on the stability of the molecule.


Subject(s)
Chromatography/methods , Hydrophobic and Hydrophilic Interactions , Receptors, Interleukin-1 Type I/chemistry , Receptors, Interleukin-1 Type I/metabolism , Amino Acid Sequence , Animals , Cattle , Cyanogen Bromide/metabolism , Cyanogen Bromide/pharmacology , Humans , Models, Molecular , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Solubility , Temperature
17.
Biochemistry ; 47(28): 7496-508, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18549248

ABSTRACT

In this communication we present the detailed disulfide structure of IgG2 molecules. The consensus structural model of human IgGs represents the hinge region positioned as a flexible linker connecting structurally isolated Fc and Fab domains. IgG2 molecules are organized differently from that model and exhibit multiple structural isoforms composed of (heavy chain-light chain-hinge) covalent complexes. We describe the precise connection of all the disulfide bridges and show that the IgG2 C H1 and C-terminal C L cysteine residues are either linked to each other or to the two upper hinge cysteine residues specific to the IgG2 subclass. A defined arrangement of these disulfide bridges is unique to each isoform. Mutation of a single cysteine residue in the hinge region eliminates these natural complexes. These results show that IgG2 structure is significantly different from the conventionally accepted immunoglobulin structural model and may help to explain some of the unique biological activity attributed only to this subclass.


Subject(s)
Immunoglobulin G/chemistry , Disulfides , Electrophoresis, Capillary , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin G/genetics , Immunoglobulin G/isolation & purification , Models, Molecular , Peptide Mapping , Protein Conformation , Protein Isoforms
18.
Electrophoresis ; 29(12): 2550-6, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18494039

ABSTRACT

Recombinant monoclonal antibodies of therapeutic interest were analyzed by a nonreduced CE-SDS (nrCE-SDS) method developed for the evaluation of size-based variants. We found that immunoglobulins analyzed by this technique exhibited different behavior depending on their subclasses. Under nrCE-SDS conditions, IgG1 molecules were separated in a well-resolved, single peak, whereas IgG2 molecules were consistently separated as a doublet. Investigation of these isoforms showed that they were structurally different, and that the difference was not caused by cell culture condition, glycosylation structure, or recombinant expression system. Commercially available IgG2 affinity-purified from human plasma also showed the presence of structural isoforms. The structural isoforms remained present under pH- and temperature-stressed conditions. Application of a mild cysteine/cystine redox potential converted the main peak doublet into a single peak, indicating that these isoforms were disulfide bond-related species. Bioactivity measured before and after application of a redox potential gave similar values, indicating that the structural isoforms have comparable potency. The nrCE-SDS technique described here demonstrated a unique capability to resolve IgGs, leading to the discovery of novel structural isoforms specific to the IgG2 isotype.


Subject(s)
Antibodies, Monoclonal/chemistry , Immunoglobulin G/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Electrophoresis, Capillary/methods , Enzyme-Linked Immunosorbent Assay , Humans , Hybridomas , Mass Spectrometry , Protein Isoforms/chemistry , Recombinant Proteins/chemistry , Sodium Dodecyl Sulfate
19.
J Biol Chem ; 283(23): 16194-205, 2008 Jun 06.
Article in English | MEDLINE | ID: mdl-18339624

ABSTRACT

In this work, we present studies of the covalent structure of human IgG2 molecules. Detailed analysis showed that recombinant human IgG2 monoclonal antibody could be partially resolved into structurally distinct forms caused by multiple disulfide bond structures. In addition to the presently accepted structure for the human IgG2 subclass, we also found major structures that differ from those documented in the current literature. These novel structural isoforms are defined by the light chain constant domain (C(L)) and the heavy chain C(H)1 domain covalently linked via disulfide bonds to the hinge region of the molecule. Our results demonstrate the presence of three main types of structures within the human IgG2 subclass, and we have named these structures IgG2-A, -B, and -A/B. IgG2-A is the known classic structure for the IgG2 subclass defined by structurally independent Fab domains and hinge region. IgG2-B is a structure defined by a symmetrical arrangement of a (C(H)1-C(L)-hinge)(2) complex with both Fab regions covalently linked to the hinge. IgG2-A/B represents an intermediate form, defined by an asymmetrical arrangement involving one Fab arm covalently linked to the hinge through disulfide bonds. The newly discovered structural isoforms are present in native human IgG2 antibodies isolated from myeloma plasma and from normal serum. Furthermore, the isoforms are present in native human IgG2 with either kappa or lambda light chains, although the ratios differ between the light chain classes. These findings indicate that disulfide structural heterogeneity is a naturally occurring feature of antibodies belonging to the human IgG2 subclass.


Subject(s)
Disulfides/chemistry , Immunoglobulin G/chemistry , Immunoglobulin kappa-Chains/chemistry , Immunoglobulin lambda-Chains/chemistry , Humans , Immunoglobulin G/genetics , Immunoglobulin kappa-Chains/genetics , Immunoglobulin lambda-Chains/genetics , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Structure, Quaternary/physiology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
20.
J Biol Chem ; 283(23): 16206-15, 2008 Jun 06.
Article in English | MEDLINE | ID: mdl-18339626

ABSTRACT

In the accompanying report ( Wypych, J., Li, M., Guo, A., Zhang, Z., Martinez, T., Allen, M. J., Fodor, S., Kelner, D. N., Flynn, G. C., Liu, Y. D., Bondarenko, P. V., Ricci, M. S., Dillon, T. M., and Balland, A. (2008) J. Biol. Chem. 283, 16194-16205 ), we have identified that the human IgG2 subclass exists as an ensemble of distinct isoforms, designated IgG2-A, -B, and -A/B, which differ by the disulfide connectivity at the hinge region. In this report, we studied the structural and functional properties of the IgG2 disulfide isoforms and compared them to IgG1. Human monoclonal IgG1 and IgG2 antibodies were designed with identical antigen binding regions, specific to interleukin-1 cell surface receptor type 1. In vitro biological activity measurements showed an increased activity of the IgG1 relative to the IgG2 in blocking interleukin-1beta ligand from binding to the receptor, suggesting that some of the IgG2 isoforms had lower activity. Under reduction-oxidation conditions, the IgG2 disulfide isoforms converted to IgG2-A when 1 m guanidine was used, whereas IgG2-B was enriched in the absence of guanidine. The relative potency of the antibodies in cell-based assays was: IgG1 > IgG2-A > IgG2 >> IgG2-B. This difference correlated with an increased hydrodynamic radius of IgG2-A relative to IgG2-B, as shown by biophysical characterization. The enrichment of disulfide isoforms and activity studies were extended to additional IgG2 monoclonal antibodies with various antigen targets. All IgG2 antibodies displayed the same disulfide conversion, but only a subset showed activity differences between IgG2-A and IgG2-B. Additionally, the distribution of isoforms was influenced by the light chain type, with IgG2lambda composed mostly of IgG2-A. Based on crystal structure analysis, we propose that IgG2 disulfide exchange is caused by the close proximity of several cysteine residues at the hinge and the reactivity of tandem cysteines within the hinge. Furthermore, the IgG2 isoforms were shown to interconvert in whole blood or a "blood-like" environment, thereby suggesting that the in vivo activity of human IgG2 may be dependent on the distribution of isoforms.


Subject(s)
Disulfides/chemistry , Immunoglobulin G/chemistry , Immunoglobulin lambda-Chains/chemistry , Crystallography, X-Ray , Humans , Oxidation-Reduction , Protein Isoforms/chemistry , Protein Structure, Quaternary/physiology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...