101
|
Zhang X, Jin X, Liu L, Zhang Z, Koza S, Yu YQ, Chen W. Optimized reversed phase LC/MS methods for intact protein analysis and peptide mapping of adeno-associated virus (AAV) proteins. Hum Gene Ther 2021; 32:1501-1511. [PMID: 34278837 PMCID: PMC8742267 DOI: 10.1089/hum.2021.046] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Recombinant adeno-associated viruses (AAVs) have emerged as the leading gene delivery platform owing to their nonpathogenic nature and long-term gene expression capability. The AAV capsid, in addition to protecting the viral genome, plays an important role in viral infectivity and gene transduction, indicating the value of the constituent viral proteins (VPs) being well-characterized as part of gene therapy development. However, the limited sample availability and sequence homology shared by the VPs pose challenges to adapt existing analytical methods developed for conventional biologics. In this study, we report the development of reversed-phase liquid chromatography/mass spectrometry-based methods for characterization of AAV capsid proteins at intact protein and peptide level with reduced sample consumptions. The developed methods allowed the measurement of VP expression with fluorescence detection and intact mass/post-translational modifications (PTMs) analysis through a benchtop time-of-flight mass spectrometer. The general applicability and validity of the methods for gene therapy product development were demonstrated by applying the optimized methods to multiple common AAV serotypes. A 1-h enzymatic digestion method was also developed using 1.25 μg of AAV VPs, providing >98% protein sequence coverage and reproducible relative quantification of various PTMs of the VPs. The efficient and sensitive analyses of AAV capsid proteins enabled by the reported methods provide further understanding and offer guidance in the development and manufacturing of AAV-related therapeutics.
Collapse
Affiliation(s)
- Ximo Zhang
- Waters Corp, 36565, Scientific Operation, 34 Maple Street, Milford, Milford, Massachusetts, United States, 01757-3604;
| | - Xiaoying Jin
- Sanofi-Aventis US LLC, 5269, Biopharmaceutics Development, One Mountain Road, Framingham, Massachusetts, United States, 01701;
| | - Lin Liu
- Sanofi Genzyme, 2194, 1 Mountain Rd, Framingham, Massachusetts, United States, 01701;
| | - Zichuang Zhang
- Sanofi-Aventis US LLC, 5269, Biopharmaceutics Development, Framingham, Massachusetts, United States;
| | - Stephan Koza
- Waters Corp, 36565, Scientific Operation, Milford, Massachusetts, United States;
| | - Ying Qing Yu
- Waters Corp, 36565, Scientific Operation, Milford, Massachusetts, United States;
| | - Weibin Chen
- Waters Corp, 36565, Scientific Operation, Milford, Massachusetts, United States;
| |
Collapse
|
102
|
Oyama H, Ishii K, Maruno T, Torisu T, Uchiyama S. Characterization of Adeno-Associated Virus Capsid Proteins with Two Types of VP3-Related Components by Capillary Gel Electrophoresis and Mass Spectrometry. Hum Gene Ther 2021; 32:1403-1416. [PMID: 34082578 PMCID: PMC10112878 DOI: 10.1089/hum.2021.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Recombinant adeno-associated virus is a leading platform in human gene therapy. The adeno-associated virus (AAV) capsid is composed of three viral proteins (VPs): VP1, VP2, and VP3. To ensure the safety of AAV-based gene therapy products, the stoichiometry of VPs of AAV vector should be carefully monitored. In this study, sodium dodecyl sulfate-polyacrylamide gel electrophoresis, capillary gel electrophoresis (CGE), and liquid chromatography-UV-mass spectrometry (LC-UV-MS) were performed to evaluate the VP components of AAV1, AAV2, and AAV6. Two types of VP3-related components, VP3 variant and VP3 fragment, were identified. The VP3 variant was the N-terminal shorter VP3, of which the translation started at M211, not at the conventional initiation codon, M203. The VP3 variant could be generated by leaky scanning of the first initiation codon of VP3. We also showed that the VP3 variant was identified in a minor peak before VP3 in CGE measurement. Meanwhile, the VP3 fragment was the C-terminal cleaved VP3, of which the sequence of VP3 ended at D590 or D626, indicating that cleavage occurred between D590 and P591, or D626 and G627. The cause of the cleavage of the DP or DG sequence was hydrolysis due to low pH of the mobile phase and high temperature of the column oven in the LC system, which was necessary to clearly separate the peak of VPs. VP3 fragments, detected only in LC-UV-MS in small amount account with less than 3% of total peak area, should be included in the quantification of VP3. Finally, the relationship of VP stoichiometry determined by the above three methods was discussed. From this study, we proposed that the VP components of AAV should be complementarily evaluated by CGE and LC-UV-MS.
Collapse
Affiliation(s)
- Hiroaki Oyama
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Suita, Japan
| | - Kentaro Ishii
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Suita, Japan
| | - Takahiro Maruno
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Suita, Japan
| | - Tetsuo Torisu
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Suita, Japan
| | - Susumu Uchiyama
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Suita, Japan.,Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Myodaiji-cho, Japan
| |
Collapse
|
103
|
Li M, Yu C, Wang W, Wu G, Wang L. Interlaboratory method validation of capillary electrophoresis sodium dodecyl sulfate (CE-SDS) methodology for analysis of mAbs. Electrophoresis 2021; 42:1900-1913. [PMID: 34240427 DOI: 10.1002/elps.202000396] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 11/09/2022]
Abstract
Capillary electrophoresis sodium dodecyl sulfate (CE-SDS) is an analytical method to assess the purity of proteins, commonly applied to monoclonal antibodies (mAbs) in the biopharmaceutical industry. To address the need to standardize the CE-SDS method in the pharmaceutical industry and to enhance the confidence in method transfer between laboratories operating different commercial capillary electrophoresis (CE) instrument platforms, an interlaboratory CE-SDS method validation was organized involving 13 laboratories in 13 companies on four different types of commercial capillary electrophoresis instruments. In the validation, a commercial mAb therapeutic was used as the sample. The validation process followed the analytical guidelines set by the ICH guidelines (International Conference for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use). The method's precision, accuracy, linearity and range, and limit of quantitation (LOQ) were validated in the study. Variations of all the parameters validated in the study passed the pre-set criteria defined at the beginning of the study. The definition was based on previously published works and the intended application purpose of the CE-SDS method for mAbs. The study proved that the CE-SDS method fits its intended application purpose as a size impurity assay and size heterogeneity characterization assay for mAb therapeutic products. This study is the first time a CE-SDS method is validated by multiple laboratories using different commercial CE instrument platforms and on a commercial mAb therapeutic. Its results will enhance the confidence of the biopharmaceutical industry to develop CE-SDS methods and transfer CE-SDS methods between different laboratories.
Collapse
Affiliation(s)
- Meng Li
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, Beijing, P. R. China
| | - Chuanfei Yu
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, Beijing, P. R. China
| | - Wenbo Wang
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, Beijing, P. R. China
| | - Gang Wu
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, Beijing, P. R. China
| | - Lan Wang
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, Beijing, P. R. China
| |
Collapse
|
104
|
Rouby G, Tran NT, Leblanc Y, Taverna M, Bihoreau N. Investigation of monoclonal antibody dimers in a final formulated drug by separation techniques coupled to native mass spectrometry. MAbs 2021; 12:e1781743. [PMID: 32633190 PMCID: PMC7531515 DOI: 10.1080/19420862.2020.1781743] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Therapeutic monoclonal antibodies (mAbs) are highly complex proteins that must be exhaustively characterized according to the regulatory authorities' recommendations. MAbs display micro-heterogeneity mainly due to their post-translational modifications, but also to their susceptibility to chemical and physical degradations. Among these degradations, aggregation is quite frequent, initiated by protein denaturation and then dimer formation. Here, we investigated the nature and structure of the high molecular weight species (HMW) present at less than 1% in an unstressed formulated roledumab biopharmaceutical, as a model of high purity mAb. HMW species were first purified through preparative size-exclusion chromatography (SEC) and then analyzed by a combination of chromatographic methods (ion-exchange chromatography (IEX), SEC) coupled to native mass spectrometry (MS), as well as sodium dodecyl sulfate–polyacrylamide gel electrophoresis and capillary gel electrophoresis under non-reducing conditions. Both covalently and non-covalently bound dimers were identified at a proportion of 50/50. In-depth characterization of the HMW fraction by SEC and IEX hyphenated to native MS revealed the presence of three mAb dimer forms having the same mass, but differing by their charge and size. They were attributed to different compact and elongated dimers. Finally, high-resolution middle-up approaches using different enzymes (IdeS and IgdE) were performed to determine the mAb domains implicated in the dimerization. Our results revealed that the roledumab dimers were associated mainly by a single Fab-to-Fab arm-bound association.
Collapse
Affiliation(s)
- G Rouby
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay , 92296, Châtenay-Malabry, France.,Analytical Department, LFB , Courtaboeuf (Les Ulis), France
| | - N T Tran
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay , 92296, Châtenay-Malabry, France
| | - Y Leblanc
- Analytical Department, LFB , Courtaboeuf (Les Ulis), France
| | - M Taverna
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay , 92296, Châtenay-Malabry, France.,Institut Universitaire de France , Paris, France
| | - N Bihoreau
- Analytical Department, LFB , Courtaboeuf (Les Ulis), France
| |
Collapse
|
105
|
Rezaei S, Sefidbakht Y, Uskoković V. Tracking the pipeline: immunoinformatics and the COVID-19 vaccine design. Brief Bioinform 2021; 22:6313266. [PMID: 34219142 DOI: 10.1093/bib/bbab241] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/23/2021] [Accepted: 06/04/2021] [Indexed: 12/23/2022] Open
Abstract
With the onset of the COVID-19 pandemic, the amount of data on genomic and proteomic sequences of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) stored in various databases has exponentially grown. A large volume of these data has led to the production of equally immense sets of immunological data, which require rigorous computational approaches to sort through and make sense of. Immunoinformatics has emerged in the recent decades as a field capable of offering this approach by bridging experimental and theoretical immunology with state-of-the-art computational tools. Here, we discuss how immunoinformatics can assist in the development of high-performance vaccines and drug discovery needed to curb the spread of SARS-CoV-2. Immunoinformatics can provide a set of computational tools to extract meaningful connections from the large sets of COVID-19 patient data, which can be implemented in the design of effective vaccines. With this in mind, we represent a pipeline to identify the role of immunoinformatics in COVID-19 treatment and vaccine development. In this process, a number of free databases of protein sequences, structures and mutations are introduced, along with docking web servers for assessing the interaction between antibodies and the SARS-CoV-2 spike protein segments as most commonly considered antigens in vaccine design.
Collapse
Affiliation(s)
- Shokouh Rezaei
- Protein Research Center at Shahid Beheshti University, Tehran, Iran
| | - Yahya Sefidbakht
- Protein Research Center at Shahid Beheshti University, Tehran, Iran
| | - Vuk Uskoković
- Founder of the biotech startup, TardigradeNano, and formerly a Professor at University of Illinois in Chicago, Chapman University, and University of California in Irvine
| |
Collapse
|
106
|
Chae H, Cho S, Jeong M, Kwon K, Choi D, Lee J, Nam W, Hong J, Lee J, Yoon S, Hong H. Improvement of Biophysical Properties and Affinity of a Human Anti-L1CAM Therapeutic Antibody through Antibody Engineering Based on Computational Methods. Int J Mol Sci 2021; 22:ijms22136696. [PMID: 34206616 PMCID: PMC8268072 DOI: 10.3390/ijms22136696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/18/2021] [Accepted: 06/19/2021] [Indexed: 01/14/2023] Open
Abstract
The biophysical properties of therapeutic antibodies influence their manufacturability, efficacy, and safety. To develop an anti-cancer antibody, we previously generated a human monoclonal antibody (Ab417) that specifically binds to L1 cell adhesion molecule with a high affinity, and we validated its anti-tumor activity and mechanism of action in human cholangiocarcinoma xenograft models. In the present study, we aimed to improve the biophysical properties of Ab417. We designed 20 variants of Ab417 with reduced aggregation propensity, less potential post-translational modification (PTM) motifs, and the lowest predicted immunogenicity using computational methods. Next, we constructed these variants to analyze their expression levels and antigen-binding activities. One variant (Ab612)—which contains six substitutions for reduced surface hydrophobicity, removal of PTM, and change to the germline residue—exhibited an increased expression level and antigen-binding activity compared to Ab417. In further studies, compared to Ab417, Ab612 showed improved biophysical properties, including reduced aggregation propensity, increased stability, higher purification yield, lower pI, higher affinity, and greater in vivo anti-tumor efficacy. Additionally, we generated a highly productive and stable research cell bank (RCB) and scaled up the production process to 50 L, yielding 6.6 g/L of Ab612. The RCB will be used for preclinical development of Ab612.
Collapse
Affiliation(s)
- Heesu Chae
- Department of Systems Immunology, Kangwon National University, Chuncheon 24341, Korea; (H.C.); (M.J.); (K.K.); (J.H.); (J.L.)
- APIT BIO Inc., B910, Munjeongdong Tera Tower, 167 Songpa-daero, Songpa-gu, Seoul 05855, Korea; (J.L.); (W.N.)
| | - Seulki Cho
- Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon 24341, Korea;
| | - Munsik Jeong
- Department of Systems Immunology, Kangwon National University, Chuncheon 24341, Korea; (H.C.); (M.J.); (K.K.); (J.H.); (J.L.)
| | - Kiyoung Kwon
- Department of Systems Immunology, Kangwon National University, Chuncheon 24341, Korea; (H.C.); (M.J.); (K.K.); (J.H.); (J.L.)
| | - Dongwook Choi
- Division of Drug Process Development, New Drug Development Center, Osong Medical Innovation Foundation, Chungcheongbuk-do, Cheongju-si 28160, Korea;
| | - Jaeyoung Lee
- APIT BIO Inc., B910, Munjeongdong Tera Tower, 167 Songpa-daero, Songpa-gu, Seoul 05855, Korea; (J.L.); (W.N.)
| | - Woosuk Nam
- APIT BIO Inc., B910, Munjeongdong Tera Tower, 167 Songpa-daero, Songpa-gu, Seoul 05855, Korea; (J.L.); (W.N.)
| | - Jisu Hong
- Department of Systems Immunology, Kangwon National University, Chuncheon 24341, Korea; (H.C.); (M.J.); (K.K.); (J.H.); (J.L.)
| | - Jiwoo Lee
- Department of Systems Immunology, Kangwon National University, Chuncheon 24341, Korea; (H.C.); (M.J.); (K.K.); (J.H.); (J.L.)
| | - Seonjoo Yoon
- APIT BIO Inc., B910, Munjeongdong Tera Tower, 167 Songpa-daero, Songpa-gu, Seoul 05855, Korea; (J.L.); (W.N.)
- Correspondence: (S.Y.); (H.H.); Tel.: +82-10-2305-9704 (S.Y.); +82-10-5430-0480 (H.H.)
| | - Hyojeong Hong
- Department of Systems Immunology, Kangwon National University, Chuncheon 24341, Korea; (H.C.); (M.J.); (K.K.); (J.H.); (J.L.)
- Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon 24341, Korea;
- Correspondence: (S.Y.); (H.H.); Tel.: +82-10-2305-9704 (S.Y.); +82-10-5430-0480 (H.H.)
| |
Collapse
|
107
|
Identification and Characterization of a Monoclonal Antibody Variant Species with a Clipping in the Complementarity Determining Region Isolated by Size Exclusion Chromatography Under Native Conditions. J Pharm Sci 2021; 110:3367-3374. [PMID: 34089708 DOI: 10.1016/j.xphs.2021.05.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/30/2021] [Accepted: 05/31/2021] [Indexed: 01/09/2023]
Abstract
The content of monoclonal antibody (mAb) fragments in pharmaceutical mAb products is a critical quality attribute and should be controlled for safety. Peptide bonds in the hinge region of mAbs are susceptible to hydrolysis, generating Fc-Fab fragments, which are associated with lower efficacy than the intact antibody. Fc-Fab fragments can be separated from intact antibody molecules under native conditions by size exclusion chromatography (SEC). Although several fragments generated by a clip in the complementarity determining region (CDR) have been reported, their efficacies have not been analyzed. This is because these fragments could not be separated from intact antibodies under native conditions owing to their similar molecular sizes. Here, we report that bevacizumab variant with clipping in the CDR, with the resulting fragments remaining intact in the variant, can be isolated under native conditions by selecting an adequate SEC column.
Collapse
|
108
|
Boschanski M, Krüger T, Karsten L, Falck G, Alam S, Gerlach M, Müller B, Müller KM, Sewald N, Dierks T. Site-Specific Conjugation Strategy for Dual Antibody-Drug Conjugates Using Aerobic Formylglycine-Generating Enzymes. Bioconjug Chem 2021; 32:1167-1174. [PMID: 34060308 DOI: 10.1021/acs.bioconjchem.1c00246] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Multiple, site-specific protein conjugation is increasingly attractive for the generation of antibody-drug conjugates (ADCs). As it is important to control the number and position of cargoes in an ADC, position-selective generation of reactive sites in the protein of interest is required. Formylglycine (FGly) residues are generated by enzymatic conversion of cysteine residues embedded in a certain amino acid sequence motif with a formylglycine-generating enzyme (FGE). The addition of copper ions increases FGE activity leading to the conversion of cysteines within less readily accepted sequences. With this tuned enzyme activity, it is possible to address two different recognition sequences using two aerobic formylglycine-generating enzymes. We demonstrate an improved and facile strategy for the functionalization of a DARPin (designed ankyrin repeat protein) and the single-chain antibody scFv425-Fc, both directed against the epidermal growth factor receptor (EGFR). The single-chain antibody was conjugated with monomethyl auristatin E (MMAE) and carboxyfluorescein (CF) and successfully tested for receptor binding, internalization, and cytotoxicity in cell culture, respectively.
Collapse
Affiliation(s)
- Mareile Boschanski
- Biochemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| | - Tobias Krüger
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| | - Lennard Karsten
- Cellular and Molecular Biotechnology, Faculty of Technology, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| | - Georg Falck
- Cellular and Molecular Biotechnology, Faculty of Technology, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| | - Sarfaraz Alam
- Biochemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| | - Marcus Gerlach
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| | | | - Kristian M Müller
- Cellular and Molecular Biotechnology, Faculty of Technology, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| | - Norbert Sewald
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| | - Thomas Dierks
- Biochemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| |
Collapse
|
109
|
Deslignière E, Ehkirch A, Duivelshof BL, Toftevall H, Sjögren J, Guillarme D, D’Atri V, Beck A, Hernandez-Alba O, Cianférani S. State-of-the-Art Native Mass Spectrometry and Ion Mobility Methods to Monitor Homogeneous Site-Specific Antibody-Drug Conjugates Synthesis. Pharmaceuticals (Basel) 2021; 14:ph14060498. [PMID: 34073805 PMCID: PMC8225019 DOI: 10.3390/ph14060498] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 02/06/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are biotherapeutics consisting of a tumor-targeting monoclonal antibody (mAb) linked covalently to a cytotoxic drug. Early generation ADCs were predominantly obtained through non-selective conjugation methods based on lysine and cysteine residues, resulting in heterogeneous populations with varying drug-to-antibody ratios (DAR). Site-specific conjugation is one of the current challenges in ADC development, allowing for controlled conjugation and production of homogeneous ADCs. We report here the characterization of a site-specific DAR2 ADC generated with the GlyCLICK three-step process, which involves glycan-based enzymatic remodeling and click chemistry, using state-of-the-art native mass spectrometry (nMS) methods. The conjugation process was monitored with size exclusion chromatography coupled to nMS (SEC-nMS), which offered a straightforward identification and quantification of all reaction products, providing a direct snapshot of the ADC homogeneity. Benefits of SEC-nMS were further demonstrated for forced degradation studies, for which fragments generated upon thermal stress were clearly identified, with no deconjugation of the drug linker observed for the T-GlyGLICK-DM1 ADC. Lastly, innovative ion mobility-based collision-induced unfolding (CIU) approaches were used to assess the gas-phase behavior of compounds along the conjugation process, highlighting an increased resistance of the mAb against gas-phase unfolding upon drug conjugation. Altogether, these state-of-the-art nMS methods represent innovative approaches to investigate drug loading and distribution of last generation ADCs, their evolution during the bioconjugation process and their impact on gas-phase stabilities. We envision nMS and CIU methods to improve the conformational characterization of next generation-empowered mAb-derived products such as engineered nanobodies, bispecific ADCs or immunocytokines.
Collapse
Affiliation(s)
- Evolène Deslignière
- Laboratoire de Spectrométrie de Masse BioOrganique, IPHC UMR 7178, Université de Strasbourg, CNRS, 67087 Strasbourg, France; (E.D.); (A.E.); (O.H.-A.)
- Infrastructure Nationale de Protéomique ProFI—FR2048, 67087 Strasbourg, France
| | - Anthony Ehkirch
- Laboratoire de Spectrométrie de Masse BioOrganique, IPHC UMR 7178, Université de Strasbourg, CNRS, 67087 Strasbourg, France; (E.D.); (A.E.); (O.H.-A.)
- Infrastructure Nationale de Protéomique ProFI—FR2048, 67087 Strasbourg, France
| | - Bastiaan L. Duivelshof
- School of Pharmaceutical Sciences, University of Geneva, CMU—Rue Michel-Servet 1, 1211 Geneva, Switzerland; (B.L.D.); (D.G.); (V.D.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU—Rue Michel-Servet 1, 1211 Geneva, Switzerland
| | | | | | - Davy Guillarme
- School of Pharmaceutical Sciences, University of Geneva, CMU—Rue Michel-Servet 1, 1211 Geneva, Switzerland; (B.L.D.); (D.G.); (V.D.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU—Rue Michel-Servet 1, 1211 Geneva, Switzerland
| | - Valentina D’Atri
- School of Pharmaceutical Sciences, University of Geneva, CMU—Rue Michel-Servet 1, 1211 Geneva, Switzerland; (B.L.D.); (D.G.); (V.D.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU—Rue Michel-Servet 1, 1211 Geneva, Switzerland
| | - Alain Beck
- IRPF—Centre d’Immunologie Pierre-Fabre (CIPF), 74160 Saint-Julien-en-Genevois, France;
| | - Oscar Hernandez-Alba
- Laboratoire de Spectrométrie de Masse BioOrganique, IPHC UMR 7178, Université de Strasbourg, CNRS, 67087 Strasbourg, France; (E.D.); (A.E.); (O.H.-A.)
- Infrastructure Nationale de Protéomique ProFI—FR2048, 67087 Strasbourg, France
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique, IPHC UMR 7178, Université de Strasbourg, CNRS, 67087 Strasbourg, France; (E.D.); (A.E.); (O.H.-A.)
- Infrastructure Nationale de Protéomique ProFI—FR2048, 67087 Strasbourg, France
- Correspondence:
| |
Collapse
|
110
|
Ma F, Raoufi F, Bailly MA, Fayadat-Dilman L, Tomazela D. Hyphenation of strong cation exchange chromatography to native mass spectrometry for high throughput online characterization of charge heterogeneity of therapeutic monoclonal antibodies. MAbs 2021; 12:1763762. [PMID: 32370592 PMCID: PMC7299211 DOI: 10.1080/19420862.2020.1763762] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Characterization of charge heterogeneity in monoclonal antibodies (mAbs) is needed during developability assessment and downstream development of drug candidates. Charge heterogeneity can come from post-translational modifications like deamidation, isomerization, and sialylation. Elucidation of charge variants with mass spectrometry (MS) has historically been challenging. Due to the nonvolatility and high ionic strength of conventional buffer systems, labor-intensive offline fractionation followed by MS analysis is routinely used. Here, we describe an alternative strategy that directly couples strong cation exchange (SCX) chromatography to high-resolution Orbitrap MS for online native MS analysis (SCX-MS). A combined pH and salt gradient was used for universal separation of mAbs from a wide range of pI values (6.38 ~ 9.2), including infliximab (Remicade®, chimeric IgG1/kappa), NISTmab (humanized IgG1/kappa) and trastuzumab (Herceptin®, humanized IgG1/kappa), without tailoring of chromatographic profiles. Liquid chromatography and MS parameters were optimized to achieve high-quality spectra and enhanced detection of low abundant species under high flow rate conditions. Genedata Expressionist, a vendor agnostic software, was used for data processing. This integrated strategy allows unbiased characterization of numerous charge variant species and low molecular weight fragments (<0.05%) without post-column flow splitting. The application was further expanded with middle-up approaches for subdomain analysis, which demonstrated the versatility of the strategy for analysis of various construct types. With our analysis of mAbs during developability assessment and forced degradation studies, which aimed at assessing potential critical quality attributes in antibody drug molecules, we provide, for the first time, direct visualization of molecular alterations of mAbs at intact level. Furthermore, strong correlation was observed between this novel MS approach and analysis by capillary isoelectric focusing.
Collapse
Affiliation(s)
- Fengfei Ma
- Protein Sciences, Discovery Biologics, Merck & Co., Inc., South San Francisco, CA, USA
| | - Fahimeh Raoufi
- Protein Sciences, Discovery Biologics, Merck & Co., Inc., South San Francisco, CA, USA
| | - Marc Andre Bailly
- Protein Sciences, Discovery Biologics, Merck & Co., Inc., South San Francisco, CA, USA
| | | | - Daniela Tomazela
- Protein Sciences, Discovery Biologics, Merck & Co., Inc., South San Francisco, CA, USA
| |
Collapse
|
111
|
Schuster J, Mahler HC, Joerg S, Huwyler J, Mathaes R. Analytical Challenges Assessing Protein Aggregation and Fragmentation Under Physiologic Conditions. J Pharm Sci 2021; 110:3103-3110. [PMID: 33933436 DOI: 10.1016/j.xphs.2021.04.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 03/01/2021] [Accepted: 04/09/2021] [Indexed: 12/17/2022]
Abstract
Therapeutic proteins are administered by injection or infusion. After administration, the physiologic environment in the desired body compartment - fluid or tissue - can impact protein stability and lead to changes in the safety and/or efficacy profile. For example, protein aggregation and fragmentation are critical quality attributes of the drug product and can occur after administration to patients. In this context, the in vivo stability of therapeutic proteins has gained increasing attention. However, in vivo protein aggregation and fragmentation are difficult to assess and have been rarely investigated. This mini-review summarizes analytical approaches to assess the stability of therapeutic proteins using simulated physiologic conditions. Furthermore, we discuss factors potentially causing in vivo protein aggregation, precipitation, and fragmentation in complex biological fluids. Different analytical approaches are evaluated with respect to their applicability and possible shortcomings when it comes to these degradation events in biological fluids. Tracking protein stability in biological fluids typically requires purifying or labeling the protein of interest to circumvent matrix interference of biological fluids. Improved analytical methods are strongly needed to gain knowledge on in vivo protein aggregation and fragmentation. In vitro models can support the selection of lead candidates and accelerate the pre-clinical development of therapeutic proteins.
Collapse
Affiliation(s)
- Joachim Schuster
- Lonza Pharma and Biotech, Drug Product Services, Basel, Switzerland; University of Basel, Pharmacenter, Division of Pharmaceutical Technology, Basel, Switzerland
| | | | - Susanne Joerg
- Lonza Pharma and Biotech, Drug Product Services, Basel, Switzerland
| | - Joerg Huwyler
- University of Basel, Pharmacenter, Division of Pharmaceutical Technology, Basel, Switzerland
| | - Roman Mathaes
- Lonza Pharma and Biotech, Drug Product Services, Basel, Switzerland.
| |
Collapse
|
112
|
Römer J, Stolz A, Kiessig S, Moritz B, Neusüß C. Online top-down mass spectrometric identification of CE(SDS)-separated antibody fragments by two-dimensional capillary electrophoresis. J Pharm Biomed Anal 2021; 201:114089. [PMID: 33940498 DOI: 10.1016/j.jpba.2021.114089] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/01/2021] [Accepted: 04/19/2021] [Indexed: 12/26/2022]
Abstract
Size heterogeneity analysis by capillary sieving electrophoresis utilizing sodium dodecyl sulfate (CE(SDS)) with optical detection is a major method applied for release and stability testing of monoclonal antibodies (mAbs) in biopharmaceutical applications. Identification of mAb-fragments and impurities observed with CE(SDS) is of outstanding importance for the assessment of critical quality attributes and development of the analytical control system. Mass spectrometric (MS) detection is a powerful tool for protein identification and characterization. Unfortunately, CE(SDS) is incompatible with online MS-hyphenation due to strong ionization suppression of SDS and other separation buffer components. Here, we present a comprehensive platform for full characterization of individual CE(SDS)-separated peaks by CE(SDS)-capillary zone electrophoresis-top-down-MS. The peak of interest is transferred from the first to the second dimension via an 8-port valve to remove MS-incompatible components. Full characterization of mAb byproducts is performed by intact mass determination and fragmentation by electron transfer dissociation, higher-energy collisional dissociation, and ultraviolet photodissociation. This enables online determination of intact mass as well as sequence verification of individual CE(SDS)-separated peaks simultaneously. A more substantiated characterization of unknown CE(SDS) peaks by exact localization of modifications without prior digestion is facilitated. High sensitivity is demonstrated by successful mass and sequence verification of low abundant, unknown CE(SDS) peaks from two stressed mAb samples. Good fragmentation coverages are obtained by MS2, enabling unequivocal identification of these mAb-fragments. Also, the differentiation of reduced/non-reduced intra-protein disulfide bonds is demonstrated. In summary, a reliable and unambiguous online MS2 identification of unknown compounds of low-abundant individual CE(SDS) peaks is enabled.
Collapse
Affiliation(s)
- Jennifer Römer
- Faculty of Chemistry, Aalen University, Beethovenstraße 1, 73430, Aalen, Germany; Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Regensburg, Germany
| | - Alexander Stolz
- Faculty of Chemistry, Aalen University, Beethovenstraße 1, 73430, Aalen, Germany; Department of Pharmaceutical/Medicinal Chemistry, Friedrich Schiller University, Jena, Germany
| | - Steffen Kiessig
- F. Hoffmann-La Roche Ltd, Grenzacherstraße 124, 4070, Basel, Switzerland
| | - Bernd Moritz
- F. Hoffmann-La Roche Ltd, Grenzacherstraße 124, 4070, Basel, Switzerland
| | - Christian Neusüß
- Faculty of Chemistry, Aalen University, Beethovenstraße 1, 73430, Aalen, Germany.
| |
Collapse
|
113
|
Bigelow E, Song Y, Chen J, Holstein M, Huang Y, Duhamel L, Stone K, Furman R, Li ZJ, Ghose S. Using continuous chromatography methodology to achieve high-productivity and high-purity enrichment of charge variants for analytical characterization. J Chromatogr A 2021; 1643:462008. [PMID: 33780880 DOI: 10.1016/j.chroma.2021.462008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/27/2021] [Accepted: 02/14/2021] [Indexed: 11/28/2022]
Abstract
Charge variants of biological products, such as monoclonal antibodies (mAbs), often play an important role in stability and biological activity. Characterization of these charge variants is challenging, however, primarily due to the lack of both efficient and effective isolation methods. In this work, we present a novel use of an established, high productivity continuous chromatography method, known as multi-column counter-current solvent gradient purification (MCSGP), to create an enriched product that can be better utilized for analytical characterization. We demonstrate the principle of this separation method and compare it to traditional batch HPLC (high performance liquid chromatography) or FPLC (fast protein liquid chromatography) methods, using the isolation of charge variants of different mAbs as a case study. In a majority of cases, we are able to show that the MCSGP method is able to provide enhanced purity and quantity of samples when compared to traditional fractionation methods, using the same separation conditions. In one such case, a sample prepared by MCSGP methodology achieved 95% purity in 10 hours of processing time, while those prepared by FPLC and HPLC achieved purities of 78% and 87% in 48 and 300 hours of processing time, respectively. We further evaluate charge variant enrichment strategies using both salt and pH gradients on cation exchange chromatography (CEX) and anion exchange chromatography (AEX) resins, to provide more effective separation and less sample processing following enrichment. As a result, we find that we are able to utilize different gradients to change the enrichment capabilities of certain charged species. Lastly, we summarize the identified mAb charge variants used in this work, and highlight benefits to analytical characterization of charge variants enriched with the continuous chromatography method. The method adds a new option for charge variant enrichment and facilitates analytical characterization of charge variants.
Collapse
Affiliation(s)
- Elizabeth Bigelow
- Biologics Development, Bristol Myers Squibb, 38 Jackson Road, Devens, MA 01434.
| | - Yuanli Song
- Biologics Development, Bristol Myers Squibb, 38 Jackson Road, Devens, MA 01434
| | - Jie Chen
- Biologics Development, Bristol Myers Squibb, 38 Jackson Road, Devens, MA 01434
| | - Melissa Holstein
- Biologics Development, Bristol Myers Squibb, 38 Jackson Road, Devens, MA 01434
| | - Yunping Huang
- Biologics Development, Bristol Myers Squibb, 1 Squibb Drive, New Brunswick, NJ 08901
| | - Lauren Duhamel
- Biologics Development, Bristol Myers Squibb, 38 Jackson Road, Devens, MA 01434
| | - Kelly Stone
- Biologics Development, Bristol Myers Squibb, 38 Jackson Road, Devens, MA 01434
| | - Ran Furman
- Biologics Development, Bristol Myers Squibb, 1 Squibb Drive, New Brunswick, NJ 08901
| | - Zheng Jian Li
- Biologics Development, Bristol Myers Squibb, 38 Jackson Road, Devens, MA 01434
| | - Sanchayita Ghose
- Biologics Development, Bristol Myers Squibb, 38 Jackson Road, Devens, MA 01434
| |
Collapse
|
114
|
Schuster J, Mahler HC, Joerg S, Kamuju V, Huwyler J, Mathaes R. Stability of monoclonal antibodies after simulated subcutaneous administration. J Pharm Sci 2021; 110:2386-2394. [PMID: 33722546 DOI: 10.1016/j.xphs.2021.03.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Changes in the environment from the drug product to the human physiology might lead to physical and/or chemical modifications of the protein drug, such as in vivo aggregation and fragmentation. Although subcutaneous (SC) injection is a common route of administration for therapeutic proteins, knowledge on in vivo stability in the SC tissue is limited. In this study, we developed a physiologic in vitro model simulating the SC environment in patients. We assessed the stability of two monoclonal antibodies (mAbs) in four different protein-free fluids under physiologic conditions. We monitored protein stability over two weeks using a range of analytical methods, in analogy to testing purposes of a drug product. Both mAbs showed an increase of protein aggregates, fragments, and acidic species. mAb1 was consistently more stable in this in vitro model than mAb2, highlighting the importance of comparing the stability of different mAbs under physiologic conditions. Throughout the study, both mAbs were substantially less stable in bicarbonate buffers as compared to phosphate-buffered saline. In summary, our developed model was able to differentiate stability between molecules. Bicarbonate buffers were more suitable compared to phosphate-buffered saline in regards to simulating the in vivo conditions and evaluating protein liabilities.
Collapse
Affiliation(s)
- Joachim Schuster
- Lonza Pharma and Biotech, Drug Product Services, Hochbergerstr. 60A, 4057 Basel, Switzerland; Division of Pharmaceutical Technology, University of Basel, Pharmacenter, Basel, Switzerland
| | - Hanns-Christian Mahler
- Lonza Pharma and Biotech, Drug Product Services, Hochbergerstr. 60A, 4057 Basel, Switzerland
| | - Susanne Joerg
- Lonza Pharma and Biotech, Drug Product Services, Hochbergerstr. 60A, 4057 Basel, Switzerland
| | - Vinay Kamuju
- Lonza Pharma and Biotech, Drug Product Services, Hochbergerstr. 60A, 4057 Basel, Switzerland
| | - Joerg Huwyler
- Division of Pharmaceutical Technology, University of Basel, Pharmacenter, Basel, Switzerland
| | - Roman Mathaes
- Lonza Pharma and Biotech, Drug Product Services, Hochbergerstr. 60A, 4057 Basel, Switzerland.
| |
Collapse
|
115
|
Ulitzka M, Carrara S, Grzeschik J, Kornmann H, Hock B, Kolmar H. Engineering therapeutic antibodies for patient safety: tackling the immunogenicity problem. Protein Eng Des Sel 2021; 33:5944198. [PMID: 33128053 DOI: 10.1093/protein/gzaa025] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/21/2022] Open
Abstract
Established monoclonal antibodies (mAbs) allow treatment of cancers, autoimmune diseases and other severe illnesses. Side effects either arise due to interaction with the target protein and its biology or result from of the patient's immune system reacting to the foreign protein. This immunogenic reaction against therapeutic antibodies is dependent on various factors. The presence of non-human sequences can trigger immune responses as well as chemical and post-translational modifications of the antibody. However, even fully human antibodies can induce immune response through T cell epitopes or aggregates. In this review, we briefly describe, how therapeutic antibodies can interact with the patient's immune system and summarize recent advancements in protein engineering and in silico methods to reduce immunogenicity of therapeutic monoclonal antibodies.
Collapse
Affiliation(s)
- Michael Ulitzka
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany.,Ferring Darmstadt Labs, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| | - Stefania Carrara
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany.,Ferring Darmstadt Labs, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| | - Julius Grzeschik
- Ferring Darmstadt Labs, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| | - Henri Kornmann
- Ferring International Center S.A., Chemin de la Vergognausaz 50, CH-1162 Saint-Prex, Switzerland
| | - Björn Hock
- Ferring International Center S.A., Chemin de la Vergognausaz 50, CH-1162 Saint-Prex, Switzerland
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| |
Collapse
|
116
|
Hinterholzer A, Stanojlovic V, Regl C, Huber CG, Cabrele C, Schubert M. Detecting aspartate isomerization and backbone cleavage after aspartate in intact proteins by NMR spectroscopy. JOURNAL OF BIOMOLECULAR NMR 2021; 75:71-82. [PMID: 33475951 PMCID: PMC7897204 DOI: 10.1007/s10858-020-00356-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/23/2020] [Indexed: 05/14/2023]
Abstract
The monitoring of non-enzymatic post-translational modifications (PTMs) in therapeutic proteins is important to ensure drug safety and efficacy. Together with methionine and asparagine, aspartic acid (Asp) is very sensitive to spontaneous alterations. In particular, Asp residues can undergo isomerization and peptide-bond hydrolysis, especially when embedded in sequence motifs that are prone to succinimide formation or when followed by proline (Pro). As Asp and isoAsp have the same mass, and the Asp-Pro peptide-bond cleavage may lead to an unspecific mass difference of + 18 Da under native conditions or in the case of disulfide-bridged cleavage products, it is challenging to directly detect and characterize such modifications by mass spectrometry (MS). Here we propose a 2D NMR-based approach for the unambiguous identification of isoAsp and the products of Asp-Pro peptide-bond cleavage, namely N-terminal Pro and C-terminal Asp, and demonstrate its applicability to proteins including a therapeutic monoclonal antibody (mAb). To choose the ideal pH conditions under which the NMR signals of isoAsp and C-terminal Asp are distinct from other random coil signals, we determined the pKa values of isoAsp and C-terminal Asp in short peptides. The characteristic 1H-13C chemical shift correlations of isoAsp, N-terminal Pro and C-terminal Asp under standardized conditions were used to identify these PTMs in lysozyme and in the therapeutic mAb rituximab (MabThera) upon prolonged storage under acidic conditions (pH 4-5) and 40 °C. The results show that the application of our 2D NMR-based protocol is straightforward and allows detecting chemical changes of proteins that may be otherwise unnoticed with other analytical methods.
Collapse
Affiliation(s)
- Arthur Hinterholzer
- Christian Doppler Laboratory for Innovative Tools for Biosimilar Characterization, University of Salzburg, Hellbrunnerstrasse 34, 5020, Salzburg, Austria
- Department of Biosciences, University of Salzburg, Billrothstrasse 11, 5020, Salzburg, Austria
| | - Vesna Stanojlovic
- Department of Biosciences, University of Salzburg, Billrothstrasse 11, 5020, Salzburg, Austria
| | - Christof Regl
- Christian Doppler Laboratory for Innovative Tools for Biosimilar Characterization, University of Salzburg, Hellbrunnerstrasse 34, 5020, Salzburg, Austria
- Department of Biosciences, Division of Chemistry and Bioanalytics, University of Salzburg, Hellbrunnerstrasse 34, 5020, Salzburg, Austria
| | - Christian G Huber
- Christian Doppler Laboratory for Innovative Tools for Biosimilar Characterization, University of Salzburg, Hellbrunnerstrasse 34, 5020, Salzburg, Austria
- Department of Biosciences, Division of Chemistry and Bioanalytics, University of Salzburg, Hellbrunnerstrasse 34, 5020, Salzburg, Austria
| | - Chiara Cabrele
- Christian Doppler Laboratory for Innovative Tools for Biosimilar Characterization, University of Salzburg, Hellbrunnerstrasse 34, 5020, Salzburg, Austria
- Department of Biosciences, University of Salzburg, Billrothstrasse 11, 5020, Salzburg, Austria
| | - Mario Schubert
- Christian Doppler Laboratory for Innovative Tools for Biosimilar Characterization, University of Salzburg, Hellbrunnerstrasse 34, 5020, Salzburg, Austria.
- Department of Biosciences, University of Salzburg, Billrothstrasse 11, 5020, Salzburg, Austria.
| |
Collapse
|
117
|
|
118
|
Lai PK, Fernando A, Cloutier TK, Kingsbury JS, Gokarn Y, Halloran KT, Calero-Rubio C, Trout BL. Machine Learning Feature Selection for Predicting High Concentration Therapeutic Antibody Aggregation. J Pharm Sci 2020; 110:1583-1591. [PMID: 33346034 DOI: 10.1016/j.xphs.2020.12.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/25/2020] [Accepted: 12/11/2020] [Indexed: 02/03/2023]
Abstract
Protein aggregation can hinder the development, safety and efficacy of therapeutic antibody-based drugs. Developing a predictive model that evaluates aggregation behaviors during early stage development is therefore desirable. Machine learning is a widely used tool to train models that predict data with different attributes. However, most machine learning techniques require more data than is typically available in antibody development. In this work, we describe a rational feature selection framework to develop accurate models with a small number of features. We applied this framework to predict aggregation behaviors of 21 approved monospecific monoclonal antibodies at high concentration (150 mg/mL), yielding a correlation coefficient of 0.71 on validation tests with only two features using a linear model. The nearest neighbors and support vector regression models further improved the performance, which have correlation coefficients of 0.86 and 0.80, respectively. This framework can be extended to train other models that predict different physical properties.
Collapse
Affiliation(s)
- Pin-Kuang Lai
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Amendra Fernando
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Theresa K Cloutier
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Yatin Gokarn
- Biologics Development, Sanofi, Framingham, MA, USA
| | | | | | - Bernhardt L Trout
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
119
|
Ayalew L, D'Souza AD, Chen Y, Kim MT. Identification and quantitation of hinge cysteinylation on endogenous IgG2 antibodies from human serum. MAbs 2020; 12:1854923. [PMID: 33317401 PMCID: PMC7755172 DOI: 10.1080/19420862.2020.1854923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Cysteinylation is a post-translational modification (PTM) that occurs when a cysteine residue on a protein forms a disulfide bond with a terminal cysteine molecule. This PTM has been found in the hinge region of several recombinant therapeutic IgG2 antibodies, but the impact of cysteinylation on the safety and immunogenicity of therapeutics remains unclear. In this study, we characterized recombinant and endogenous IgG2 antibodies to quantify their levels of hinge cysteinylation, if present. To the best of our knowledge, this is the first study to identify and quantify hinge cysteinylation in endogenous IgG2 antibodies from healthy human serum. We used anti-IgG2 immunopurification of human serum to specifically enrich for endogenous IgG2 antibodies, and then subjected the resulting samples to Lys-C peptide mapping coupled with targeted mass spectrometry techniques. Using this analytical workflow, we found that all healthy human serum samples tested (N = 10) contained quantifiable levels of hinge cysteinylation (0.8 ± 0.3%) in their endogenous human IgG2s (IgG2-A isoform). These findings demonstrate that hinge cysteinylation in therapeutic IgG2s, at least up to a certain level, is well tolerated in humans and pose minimal safety or immunogenicity risks.
Collapse
Affiliation(s)
- Luladey Ayalew
- Protein Analytical Chemistry, Genentech , South San Francisco, CA, USA
| | - Alicia D D'Souza
- Protein Analytical Chemistry, Genentech , South San Francisco, CA, USA
| | - Yan Chen
- Protein Analytical Chemistry, Genentech , South San Francisco, CA, USA
| | - Michael T Kim
- Protein Analytical Chemistry, Genentech , South San Francisco, CA, USA
| |
Collapse
|
120
|
Abstract
Multiple therapeutic proteins can be combined into a single dose for synergistic targeting to multiple sites of action. Such proteins would be mixed in dose-specific ratios to provide the correct potency for each component, and yet the formulations must also preserve their activity and keep degradation to a minimum. Mixing different therapeutic proteins could adversely affect their stability, and reduce the shelf life of each individual component, making the control of such products very challenging. In this study, a therapeutic monoclonal antibody and a related Fab fragment, were combined to investigate the impact of coformulation on their degradation kinetics. Under mildly destabilizing conditions, these proteins were found to protect each other from degradation. The protective effect appeared to originate from the interaction of Fab and IgG1 in small soluble oligomers, or through the rapid coalescence of pre-existing monomeric IgG1 nuclei into a dead-end aggregate, rather than through macromolecular crowding or diffusion-limitations.
Collapse
|
121
|
A synthetic mimic of phosphodiesterase type 5 based on corona phase molecular recognition of single-walled carbon nanotubes. Proc Natl Acad Sci U S A 2020; 117:26616-26625. [PMID: 33055208 DOI: 10.1073/pnas.1920352117] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Molecular recognition binding sites that specifically identify a target molecule are essential for life science research, clinical diagnoses, and therapeutic development. Corona phase molecular recognition is a technique introduced to generate synthetic recognition at the surface of a nanoparticle corona, but it remains an important question whether such entities can achieve the specificity of natural enzymes and receptors. In this work, we generate and screen a library of 24 amphiphilic polymers, preselected for molecular recognition and based on functional monomers including methacrylic acid, acrylic acid, and styrene, iterating upon a poly(methacrylic acid-co-styrene) motif. When complexed to a single-walled carbon nanotube, some of the resulting corona phases demonstrate binding specificity remarkably similar to that of phosphodiesterase type 5 (PDE5), an enzyme that catalyzes the hydrolysis of secondary messenger. The corona phase binds selectively to a PDE5 inhibitor, Vardenafil, as well as its molecular variant, but not to other potential off-target inhibitors. Our work herein examines the specificity and sensitivity of polymer "mutations" to the corona phase, as well as direct competitions with the native binding PDE5. Using structure perturbation, corona surface characterization, and molecular dynamics simulations, we show that the molecular recognition is associated with the unique three-dimensional configuration of the corona phase formed at the nanotube surface. This work conclusively shows that corona phase molecular recognition can mimic key aspects of biological recognition sites and drug targets, opening up possibilities for pharmaceutical and biological applications.
Collapse
|
122
|
Garripelli VK, Wu Z, Gupta S. Developability assessment for monoclonal antibody drug candidates: a case study. Pharm Dev Technol 2020; 26:11-20. [PMID: 32986499 DOI: 10.1080/10837450.2020.1829641] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Various screening approaches are used by industry to evaluate development risks associated with discovery candidates. This process has become more complicated with biological therapeutics, a class dominated by monoclonal antibodies (mAb), and, increasingly, their derivative constructs. Effective early assessment for drug-like properties (DLP) can save time and costs by allowing a more complete consideration of issues that could impact the desired end result of a stable drug product. Here we report a case study of four IgG1 mAbs, with sequence variations in the variable domain region, screened as a set of possible drug candidates. Our comprehensive, tiered approach used a battery of analytical tools to assess molecular characteristics, conformational stability, colloidal stability, and short-term storage stability. While most DLP for the four candidates were developmentally acceptable and comparable, mAb-2 was associated with adverse colloidal properties. Further investigation of mAb-2 in an expanded pH range revealed a propensity for phase separation, indicating a need for the additional product development effort. Our results support that comprehensive DLP assessments in an expanded pH range are beneficial in identifying development options for promising molecules that show challenging stability trends. This adaptable approach may be especially useful in the development of increasingly complex antibody constructs.
Collapse
Affiliation(s)
- Vivek Kumar Garripelli
- Pharmaceutical Development, Oncology Early Development CMC Biologics, AbbVie, Redwood City, CA, USA
| | - Zhen Wu
- ADC Therapeutics, San Mateo, CA, USA
| | - Supriya Gupta
- Pharmaceutical Development, Oncology Early Development CMC Biologics, AbbVie, Redwood City, CA, USA
| |
Collapse
|
123
|
Park D, Kim J, Yun J, Park SJ. Evaluation of the Physico-Chemical and Biological Stability of SB8 (Aybintio), a Proposed Biosimilar to Bevacizumab, Under Ambient and In-Use Conditions. Adv Ther 2020; 37:4308-4324. [PMID: 32816233 DOI: 10.1007/s12325-020-01465-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Indexed: 11/30/2022]
Abstract
INTRODUCTION No stability data of SB8 (proposed biosimilar to bevacizumab) are available for opened vials at 2-8 °C or for unopened vials stored outside of this range. Furthermore, limited stability data are available for dilutions. Stability of unopened vials of SB8 at ambient temperature and in-use stability for opened vials as well as diluted SB8 in infusion bags were evaluated. METHODS SB8 stability was assessed in three scenarios in the absence of light: unopened 100-mg vials 36 months after manufacture stored at 30 ± 2 °C with 65 ± 5% relative humidity for 1 month, opened 100-mg and 400-mg vials stored at 5 ± 3 °C for 72 h and diluted (1.4 mg/ml or 16.5 mg/ml in 100 ml 0.9% NaCl polyolefin bags) stored for 45 days at 5 ± 3 °C and then 3 days at 30 ± 2 °C; the United Kingdom's National Health Service protocol was used for the study design after dilution. Physicochemical stability (appearance, pH, protein concentration, size-exclusion high-performance liquid chromatography, non-reducing capillary electrophoresis-sodium dodecyl sulfate, imaged capillary isoelectric focusing), biological activity [vascular endothelial growth factor (VEGF) neutralization assay, VEGF binding assay] and potential safety impact properties (subvisible particulates, submicronic aggregation by dynamic light scattering) were determined. RESULTS All stability-indicating criteria including those for biological activity were met for both unopened vials at ambient condition and for in-use conditions of opened vials as well as both dilutions. No noteworthy changes in terms of physicochemical stability, biological activity and properties with a potential safety impact occurred. CONCLUSION Under the studied aspetic extreme conditions, SB8 was stable. Our data may support advanced SB8 preparation and may help prevent SB8 wastage because of exceptional temperature excursions or unused product. Sterility assurance is the responsibility of the user and is of utmost importance when opened or diluted SB8 is not immediately used.
Collapse
Affiliation(s)
- Dongkuk Park
- Quality Evaluation Team, Samsung Bioepis Co., Ltd., 107, Cheomdan-daero, Yeonsu-gu, Incheon, 21987, Republic of Korea
| | - Jihyun Kim
- Quality Evaluation Team, Samsung Bioepis Co., Ltd., 107, Cheomdan-daero, Yeonsu-gu, Incheon, 21987, Republic of Korea
| | - Jihoon Yun
- Quality Evaluation Team, Samsung Bioepis Co., Ltd., 107, Cheomdan-daero, Yeonsu-gu, Incheon, 21987, Republic of Korea
| | - Su Jin Park
- Quality Evaluation Team, Samsung Bioepis Co., Ltd., 107, Cheomdan-daero, Yeonsu-gu, Incheon, 21987, Republic of Korea.
| |
Collapse
|
124
|
Bansal R, Dash R, Rathore AS. Impact of mAb Aggregation on Its Biological Activity: Rituximab as a Case Study. J Pharm Sci 2020; 109:2684-2698. [DOI: 10.1016/j.xphs.2020.05.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 01/03/2023]
|
125
|
Enhancement of covalent aggregate quantification of protein therapeutics by non-reducing capillary gel electrophoresis using sodium hexadecyl sulfate (CE-SHS). J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1152:122230. [DOI: 10.1016/j.jchromb.2020.122230] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 04/30/2020] [Accepted: 06/08/2020] [Indexed: 12/27/2022]
|
126
|
Becheva ZR, Atanasova MK, Ivanov YL, Godjevargova TI. Magnetic Nanoparticle-Based Fluorescence Immunoassay for Determination of Ochratoxin A in Milk. FOOD ANAL METHOD 2020. [DOI: 10.1007/s12161-020-01848-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
127
|
Burton RE, Kim S, Patel R, Hartman DS, Tracey DE, Fox BS. Structural features of bovine colostral immunoglobulin that confer proteolytic stability in a simulated intestinal fluid. J Biol Chem 2020; 295:12317-12327. [PMID: 32665404 PMCID: PMC7443484 DOI: 10.1074/jbc.ra120.014327] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/06/2020] [Indexed: 01/16/2023] Open
Abstract
Bovine colostral antibodies, purified from cow's milk produced immediately after calving, have enhanced resistance to degradation by intestinal proteases relative to antibodies from human or bovine serum, making them of particular interest as orally administered therapeutic agents. However, the basis of this resistance is not well defined. We evaluated the stability of AVX-470, a bovine colostral anti-tumor necrosis factor (TNF) polyclonal antibody used in early clinical studies for treatment of ulcerative colitis, using conditions that mimic the human small intestine. AVX-470 was degraded ∼3 times more slowly than human IgG antibodies or infliximab (a monoclonal mouse-human chimeric IgG). Bovine IgG1 antibodies, the primary component of AVX-470, were slowly cleaved to F(ab')2 fragments. In contrast, bovine IgG2 and human IgG1 antibodies were cleaved rapidly into Fab and smaller fragments, pointing to specific regions where additional stability might be gained. Infliximab was modified to incorporate the sequences from these regions, including the bovine IgG1 hinge region and a predicted disulfide bonding motif linking the upper hinge region, the CH1 domain, and the light chain. This infliximab-bovine IgG1 chimera (bovinized infliximab) retained the antigen binding and neutralization activity of the WT sequence but was degraded 9-fold more slowly than the unmodified infliximab. This remarkable increase in stability with as few as 18 amino acid substitutions suggests that this bovinization process is a means to enable oral delivery of proven therapeutic antibodies as well as novel antibodies to targets that have been previously inaccessible to therapies delivered by injection.
Collapse
Affiliation(s)
| | - Skaison Kim
- Avaxia Biologics/Circle33 LLC, Jackson, Wyoming, USA
| | - Rutvij Patel
- Avaxia Biologics/Circle33 LLC, Jackson, Wyoming, USA
| | | | | | - Barbara S Fox
- Avaxia Biologics/Circle33 LLC, Jackson, Wyoming, USA
| |
Collapse
|
128
|
McAvan BS, Bowsher LA, Powell T, O'Hara JF, Spitali M, Goodacre R, Doig AJ. Raman Spectroscopy to Monitor Post-Translational Modifications and Degradation in Monoclonal Antibody Therapeutics. Anal Chem 2020; 92:10381-10389. [PMID: 32614170 PMCID: PMC7467412 DOI: 10.1021/acs.analchem.0c00627] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
![]()
Monoclonal
antibodies (mAbs) represent a rapidly expanding market
for biotherapeutics. Structural changes in the mAb can lead to unwanted
immunogenicity, reduced efficacy, and loss of material during production.
The pharmaceutical sector requires new protein characterization tools
that are fast, applicable in situ and to the manufacturing process.
Raman has been highlighted as a technique to suit this application
as it is information-rich, minimally invasive, insensitive to water
background and requires little to no sample preparation. This study
investigates the applicability of Raman to detect Post-Translational
Modifications (PTMs) and degradation seen in mAbs. IgG4 molecules
have been incubated under a range of conditions known to result in
degradation of the therapeutic including varied pH, temperature, agitation,
photo, and chemical stresses. Aggregation was measured using size-exclusion
chromatography, and PTM levels were calculated using peptide mapping.
By combining principal component analysis (PCA) with Raman spectroscopy
and circular dichroism (CD) spectroscopy structural analysis we were
able to separate proteins based on PTMs and degradation. Furthermore,
by identifying key bands that lead to the PCA separation we could
correlate spectral peaks to specific PTMs. In particular, we have
identified a peak which exhibits a shift in samples with higher levels
of Trp oxidation. Through separation of IgG4 aggregates, by size,
we have shown a linear correlation between peak wavenumbers of specific
functional groups and the amount of aggregate present. We therefore
demonstrate the capability for Raman spectroscopy to be used as an
analytical tool to measure degradation and PTMs in-line with therapeutic
production.
Collapse
Affiliation(s)
- Bethan S McAvan
- School of Chemistry, Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | - Leo A Bowsher
- UCB Celltech, UCB Pharma, Limited, 208 Bath Road, Slough, Berkshire SL1 3WE, United Kingdom
| | - Thomas Powell
- UCB Celltech, UCB Pharma, Limited, 208 Bath Road, Slough, Berkshire SL1 3WE, United Kingdom
| | - John F O'Hara
- UCB Celltech, UCB Pharma, Limited, 208 Bath Road, Slough, Berkshire SL1 3WE, United Kingdom
| | - Mariangela Spitali
- UCB Celltech, UCB Pharma, Limited, 208 Bath Road, Slough, Berkshire SL1 3WE, United Kingdom
| | - Royston Goodacre
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Biosciences Building, Crown Street, Liverpool L69 7ZB, United Kingdom
| | - Andrew J Doig
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| |
Collapse
|
129
|
Kosuge H, Nagatoishi S, Kiyoshi M, Ishii-Watabe A, Tanaka T, Terao Y, Oe S, Ide T, Tsumoto K. Highly sensitive HPLC analysis and biophysical characterization of N-glycans of IgG-Fc domain in comparison between CHO and 293 cells using FcγRIIIa ligand. Biotechnol Prog 2020; 36:e3016. [PMID: 32390308 PMCID: PMC7757244 DOI: 10.1002/btpr.3016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 04/08/2020] [Accepted: 05/06/2020] [Indexed: 11/06/2022]
Abstract
Quality control of monoclonal antibodies is challenging due in part to the diversity of post‐translational modifications present. The regulation of the N‐glycans of IgG‐Fc domain is one of the key factors to maintain the safety and efficacy of antibody drugs. The FcγRIIIa affinity column is an attractive tool for the precise analysis of the N‐glycans in IgG‐Fc domain. We used the mutant FcγRIIIa, which is produced in Escherichia coli and is therefore not glycosylated, as an affinity reagent to analyze the N‐glycans of monoclonal antibodies expressed in Expi293 and ExpiCHO cells. The monoclonal antibodies expressed in these cells showed very different chromatograms, because of differences in terminal galactose residues on the IgG‐Fc domains. We also carried out kinetic and thermodynamic analyses to understand the interaction between monoclonal antibodies and the mutant FcγRIIIa. Expi293 cell‐derived monoclonal antibodies had higher affinity for the mutant FcγRIIIa than those derived from ExpiCHO cells, due to slower off rates and lower binding entropy loss. Collectively, our results suggest that the FcγRIIIa column can be used to analyze the glycosylation of antibodies rapidly and specifically.
Collapse
Affiliation(s)
- Hirofumi Kosuge
- School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Satoru Nagatoishi
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masato Kiyoshi
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Kanagawa, Japan
| | - Akiko Ishii-Watabe
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Kanagawa, Japan
| | | | | | - Seigo Oe
- Tosoh Corporation, Kanagawa, Japan
| | | | - Kouhei Tsumoto
- School of Engineering, The University of Tokyo, Tokyo, Japan.,The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
130
|
Schuster J, Mahler HC, Koulov A, Joerg S, Racher A, Huwyler J, Detampel P, Mathaes R. Tracking the physical stability of fluorescent-labeled mAbs under physiologic in vitro conditions in human serum and PBS. Eur J Pharm Biopharm 2020; 152:193-201. [DOI: 10.1016/j.ejpb.2020.04.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 04/18/2020] [Accepted: 04/22/2020] [Indexed: 02/08/2023]
|
131
|
Chen K, Zhou J, Shao Z, Liu J, Song J, Wang R, Li J, Tan W. Aptamers as Versatile Molecular Tools for Antibody Production Monitoring and Quality Control. J Am Chem Soc 2020; 142:12079-12086. [PMID: 32516525 DOI: 10.1021/jacs.9b13370] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Antibody drugs have been used to treat many diseases, and to date, this has been the most rapidly growing drug class. However, the lack of suitable methods for real-time and high-throughput monitoring of antibody production and quality control has been a hindrance to the further advancement of antibody drugs or biosimilars. Therefore, we herein report a versatile tool for one-step fluorescence monitoring of antibody production by using aptamer probes selected through the in vitro SELEX method. In this case, DNA aptamers were selected against the humanized IgG1 antibody drug trastuzumab with high specificity and affinity with a Kd value of aptamer CH1S-3 of 10.3 nM. More importantly, the obtained aptamers were able to distinguish native from heat-treated, whereas antibodies failed this test. On the basis of the advantages of rapid detection for aptamers, we designed aptamer molecular beacons for direct and sensitive detection of trastuzumab in complex samples. Unlike traditional antibody-based ELISA, the signal was observed directly upon interaction with the target without the need for time-consuming binding and multiple washing steps. To further highlight biomedical applications, the use of aptamers as potential tools for quality control and traceless purification of antibody drugs was also demonstrated. Thus, aptamers are shown to be promising alternatives for antibody production monitoring, quality control, and purification, providing technical support to accelerate antibody drug development.
Collapse
Affiliation(s)
- Kaiming Chen
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jie Zhou
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Zhentao Shao
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jia Liu
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jia Song
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ruowen Wang
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Juan Li
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.,MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Weihong Tan
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.,Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| |
Collapse
|
132
|
Zhang W, Liu X, Tang H, Zhang X, Zhou Y, Fan L, Wang H, Tan WS, Zhao L. Investigation into the impact of tyrosine on the product formation and quality attributes of mAbs in rCHO cell cultures. Appl Microbiol Biotechnol 2020; 104:6953-6966. [PMID: 32577803 DOI: 10.1007/s00253-020-10744-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 06/04/2020] [Accepted: 06/15/2020] [Indexed: 11/26/2022]
Abstract
Tyrosine (Tyr) is crucial to the maintenance of the monoclonal antibody (mAb) titers and quality attributes in fed-batch cultures of recombinant Chinese hamster ovary (rCHO) cells. However, the relation between tyrosine and these aspects is not yet fully defined. In order to further elucidate such a relation, two groups of fed-batch experiments with high tyrosine (H-T) or low tyrosine (L-T) additions producing an IgG1 monoclonal antibody against CD20 were implemented to investigate the intracellular and extracellular effects of tyrosine on the culture performance. It was found that the scarcity of tyrosine led to the distinctive reduction in both viable cell density and antibody specific production rate, hence the sharply reduced titer, possibly related to the impaired translation efficiency caused by the substrate limitation of tyrosine. In addition, alterations to the critical quality attributes were detected in the L-T group, compared to those in the H-T condition. Notable decrease in the contents of intact antibody was found under the L-T condition because of the elevated reductive level in the supernatant. Moreover, the aggregate content in the L-T condition was also reduced, probably resulting from the accumulation of extracellular cystine. In particular, the lysine variant content noticeably increased with tyrosine limitation owing to the downregulation of two carboxypeptidases, i.e., CpB and CpH. Overall, understanding the role of tyrosine in these aspects is fundamental to the increase of product titers and control of critical quality attributes in the monoclonal antibody production of rCHO cell fed-batch cultures. KEY POINTS: • Tyrosine is essential in the maintenance of product titers and the control of product qualities in high cell density cultivations in rCHO cell. • This study revealed the bottleneck of decreased qmAbupon the deficiency of tyrosine. • The impact of tyrosine on the critical product qualities and the underlying mechanisms were also thoroughly assessed.
Collapse
Affiliation(s)
- Weijian Zhang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, P. O. Box 309#, Shanghai, 200237, China
| | - Xuping Liu
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, P. O. Box 309#, Shanghai, 200237, China
| | - Hongping Tang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, P. O. Box 309#, Shanghai, 200237, China
| | - Xinran Zhang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, P. O. Box 309#, Shanghai, 200237, China
| | - Yanan Zhou
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, P. O. Box 309#, Shanghai, 200237, China
| | - Li Fan
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, P. O. Box 309#, Shanghai, 200237, China
| | - Haibin Wang
- Zhejiang Hisun Pharmaceutical Co., Ltd., Fuyang, Hangzhou, 311404, Zhejiang, China
| | - Wen-Song Tan
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, P. O. Box 309#, Shanghai, 200237, China
| | - Liang Zhao
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, P. O. Box 309#, Shanghai, 200237, China.
| |
Collapse
|
133
|
Powell T, Knight MJ, O'Hara J, Burkitt W. Discovery of a Photoinduced Histidine-Histidine Cross-Link in an IgG4 Antibody. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2020; 31:1233-1240. [PMID: 32392057 DOI: 10.1021/jasms.0c00076] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
A novel histidine-histidine (His-His) photooxidative cross-link has been identified in an IgG4 antibody. It was formed between the side chain of a histidine residue of the antibody and histidine from the formulation buffer. The structure of the cross-link was elucidated using high performance liquid chromatography (HPLC) hyphenated to tandem mass spectrometry (MS/MS) with higher energy collisional dissociation (HCD). The cross-link was found in multiple conformations, as the location of the oxygen varied. Furthermore, the extent of cross-link formation was shown to correlate with the amount of light the antibody was exposed to as well as the solvent accessibility of each modification site.
Collapse
Affiliation(s)
- Thomas Powell
- Biomolecular Formulation and Characterization Sciences, UCB Group, Slough, SL3WE, United Kingdom
| | - Michael J Knight
- Biomolecular Formulation and Characterization Sciences, UCB Group, Slough, SL3WE, United Kingdom
| | - John O'Hara
- Biomolecular Formulation and Characterization Sciences, UCB Group, Slough, SL3WE, United Kingdom
| | - William Burkitt
- Biomolecular Formulation and Characterization Sciences, UCB Group, Slough, SL3WE, United Kingdom
| |
Collapse
|
134
|
Coffman J, Marques B, Orozco R, Aswath M, Mohammad H, Zimmermann E, Khouri J, Griesbach J, Izadi S, Williams A, Sankar K, Walters B, Lin J, Hepbildikler S, Schiel J, Welsh J, Ferreira G, Delmar J, Mody N, Afdahl C, Cui T, Khalaf R, Hanke A, Pampel L, Parimal S, Hong X, Patil U, Pollard J, Insaidoo F, Robinson J, Chandra D, Blanco M, Panchal J, Soundararajan S, Roush D, Tugcu N, Cramer S, Haynes C, Willson RC. Highland games: A benchmarking exercise in predicting biophysical and drug properties of monoclonal antibodies from amino acid sequences. Biotechnol Bioeng 2020; 117:2100-2115. [PMID: 32255523 DOI: 10.1002/bit.27349] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 03/05/2020] [Accepted: 04/04/2020] [Indexed: 01/08/2023]
Abstract
Biopharmaceutical product and process development do not yet take advantage of predictive computational modeling to nearly the degree seen in industries based on smaller molecules. To assess and advance progress in this area, spirited coopetition (mutually beneficial collaboration between competitors) was successfully used to motivate industrial scientists to develop, share, and compare data and methods which would normally have remained confidential. The first "Highland Games" competition was held in conjunction with the October 2018 Recovery of Biological Products Conference in Ashville, NC, with the goal of benchmarking and assessment of the ability to predict development-related properties of six antibodies from their amino acid sequences alone. Predictions included purification-influencing properties such as isoelectric point and protein A elution pH, and biophysical properties such as stability and viscosity at very high concentrations. Essential contributions were made by a large variety of individuals, including companies which consented to provide antibody amino acid sequences and test materials, volunteers who undertook the preparation and experimental characterization of these materials, and prediction teams who attempted to predict antibody properties from sequence alone. Best practices were identified and shared, and areas in which the community excels at making predictions were identified, as well as areas presenting opportunities for considerable improvement. Predictions of isoelectric point and protein A elution pH were especially good with all-prediction average errors of 0.2 and 1.6 pH unit, respectively, while predictions of some other properties were notably less good. This manuscript presents the events, methods, and results of the competition, and can serve as a tutorial and as a reference for in-house benchmarking by others. Organizations vary in their policies concerning disclosure of methods, but most managements were very cooperative with the Highland Games exercise, and considerable insight into common and best practices is available from the contributed methods. The accumulated data set will serve as a benchmarking tool for further development of in silico prediction tools.
Collapse
Affiliation(s)
| | - Bruno Marques
- Process Development, Century Therapeutics, Philadelphia, Pennsylvania
| | | | | | - Hasan Mohammad
- ProUnlimited supporting Boehringer Ingelheim Fremont Inc., Fremont, California
| | | | - Joelle Khouri
- ProUnlimited supporting Boehringer Ingelheim Fremont Inc., Fremont, California
| | | | - Saeed Izadi
- Genentech Inc., South San Francisco, California
| | | | | | | | - Jasper Lin
- Genentech Inc., South San Francisco, California
| | | | - John Schiel
- Institute of Bioscience and Biotechnology Research, National Institute of Standards and Technology, Rockville, Maryland
| | - John Welsh
- Pall Life Sciences, Portsmouth, UK.,Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | | | | | | | | | | | | | | | | | - Siddharth Parimal
- Downstream Process Development, GlaxoSmithKline, King of Prussia, Pennsylvania
| | - Xuan Hong
- Protein Design and Informatics, GlaxoSmithKline, Collegeville, Pennsylvania
| | - Ujwal Patil
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Jennifer Pollard
- BioProcess Development, MRL, Merck & Co., Inc., Kenilworth, New Jersey
| | - Francis Insaidoo
- BioProcess Development, MRL, Merck & Co., Inc., Kenilworth, New Jersey
| | - Julie Robinson
- BioProcess Development, MRL, Merck & Co., Inc., Kenilworth, New Jersey
| | - Divya Chandra
- BioProcess Development, MRL, Merck & Co., Inc., Kenilworth, New Jersey
| | - Marco Blanco
- BioProcess Development, MRL, Merck & Co., Inc., Kenilworth, New Jersey
| | - Jainik Panchal
- BioProcess Development, MRL, Merck & Co., Inc., Kenilworth, New Jersey
| | | | - David Roush
- BioProcess Development, MRL, Merck & Co., Inc., Kenilworth, New Jersey
| | - Nihal Tugcu
- Purification Process Development, Sanofi-aventis, Cambridge, Massachusetts
| | - Steven Cramer
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | - Charles Haynes
- Department of Chemical and Biological Engineering, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Richard C Willson
- Protein Design and Informatics, GlaxoSmithKline, Collegeville, Pennsylvania.,Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas.,Escuela de Medicina y Ciencias de la Salud ITESM, Monterrey, Mexico
| |
Collapse
|
135
|
Sternke-Hoffmann R, Boquoi A, Lopez Y Niedenhoff D, Platten F, Fenk R, Haas R, Buell AK. Biochemical and biophysical characterisation of immunoglobulin free light chains derived from an initially unbiased population of patients with light chain disease. PeerJ 2020; 8:e8771. [PMID: 32211238 PMCID: PMC7083161 DOI: 10.7717/peerj.8771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 02/19/2020] [Indexed: 11/20/2022] Open
Abstract
In light chain (LC) diseases, monoclonal immunoglobulin LCs are abundantly produced with the consequence in some cases to form deposits of a fibrillar or amorphous nature affecting various organs, such as heart and kidney. The factors that determine the solubility of any given LC in vivo are still not well understood. We hypothesize that some of the biochemical properties of the LCs that have been shown to correlate with amyloid fibril formation in patients also can be used as predictors for the degree of kidney damage in a patient group that is only biased by protein availability. We performed detailed biochemical and biophysical investigations of light chains extracted and purified from the urine of a group of 20 patients with light chain disease. For all samples that contained a sufficiently high concentration of LC, we quantified the unfolding temperature of the LCs, the monomer-dimer distribution, the digestibility by trypsin and the formation of amyloid fibrils under various conditions of pH and reducing agent. We correlated the results of our biophysical and biochemical experiments with the degree of kidney damage in the patient group and found that most of these parameters do not correlate with kidney damage as defined by clinical parameters. However, the patients with the greatest impairment of kidney function have light chains which display very poor digestibility by trypsin. Most of the LC properties reported before to be predictors of amyloid formation cannot be used to assess the degree of kidney damage. Our finding that poor trypsin digestibility correlates with kidney damage warrants further investigation in order to probe a putative mechanistic link between these factors.
Collapse
Affiliation(s)
| | - Amelie Boquoi
- Department of Hematology, Oncology and Clinical Oncology, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany
| | - David Lopez Y Niedenhoff
- Department of Hematology, Oncology and Clinical Oncology, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany
| | - Florian Platten
- Condensed Matter Physics Laboratory, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany
| | - Roland Fenk
- Department of Hematology, Oncology and Clinical Oncology, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany
| | - Rainer Haas
- Department of Hematology, Oncology and Clinical Oncology, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany
| | - Alexander K Buell
- Institute of Physical Biology, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany.,Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
136
|
A Mechanism of Gold Nanoparticle Aggregation by Immunoglobulin G Preparation. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10020475] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Conjugates of gold nanoparticles (GNPs) and antibodies are widely used in various fields of biochemistry and microbiology. However, the procedure for obtaining such conjugates remains precarious, and the properties of conjugates differ significantly for different antibody clones. One of the most common problems is the aggregation of GNPs in the course of their conjugation with antibodies. This article considers an example of the conjugation of monoclonal antibodies with non-stable aggregating product. The composition of the antibody preparation was studied using electrophoresis, asymmetrical flow field-flow fractionation, and ultracentrifugation. It was shown that the component that causes the aggregation of the GNPs is the light chains of immunoglobulins that appear due to the spontaneous decay of the antibodies. After separation of the fraction with a molecular weight of less than 30 kDa, stable conjugates of antibodies with GNPs were obtained. The high functional activity of the obtained conjugates was confirmed by immunochromatography.
Collapse
|
137
|
Schuster J, Koulov A, Mahler HC, Detampel P, Huwyler J, Singh S, Mathaes R. In Vivo Stability of Therapeutic Proteins. Pharm Res 2020; 37:23. [DOI: 10.1007/s11095-019-2689-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/16/2019] [Indexed: 01/05/2023]
|
138
|
Anees P, Gauthier MA. Homogenous Scavenging Resolves Low-Purification Yield/Selectivity Caused by Secondary Binding of Protein-A to Antigen-Binding Antibody Fragments. Biomacromolecules 2020; 21:825-829. [PMID: 31841628 DOI: 10.1021/acs.biomac.9b01516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Antigen-binding fragments of antibodies are biotechnologically useful agents for decorating drug delivery systems, for blocking cell-surface receptors in cell culture, for recognizing analytes in biosensors, and potentially as therapeutics. They are typically produced by enzymatic digestion of full antibodies and isolated from the undesirable fragment crystallizable (Fc) by affinity chromatography using Protein-A columns. However, while Protein-A has a strong "classical" interaction with Fc fragments, it can also more weakly bind to an "alternative" site on the heavy chain variable region of antigen-binding fragments. As such, purifying small amounts of antibody fragments by Protein-A chromatography can result in low yield. Moreover, loading larger amounts of antibody fragments onto a Protein-A column can result in poor separation, because of competition of Fc and antigen-binding fragments for immobilized Protein-A. This study demonstrates that Protein-A-based homogeneous scavenging resolves this issue by precisely controlling the stoichiometry of Protein-A to Fc fragments, something that is not possible for conventional flow-type systems, such as affinity chromatography.
Collapse
Affiliation(s)
- Palapuravan Anees
- Institut National de la Recherche Scientifique (INRS), EMT Research Center , 1650 boul. Lionel-Boulet , Varennes , J3X 1S2 , Canada
| | - Marc A Gauthier
- Institut National de la Recherche Scientifique (INRS), EMT Research Center , 1650 boul. Lionel-Boulet , Varennes , J3X 1S2 , Canada
| |
Collapse
|
139
|
Le Basle Y, Chennell P, Tokhadze N, Astier A, Sautou V. Physicochemical Stability of Monoclonal Antibodies: A Review. J Pharm Sci 2020; 109:169-190. [DOI: 10.1016/j.xphs.2019.08.009] [Citation(s) in RCA: 258] [Impact Index Per Article: 51.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/19/2019] [Accepted: 08/19/2019] [Indexed: 01/10/2023]
|
140
|
Vogg S, Pfeifer F, Ulmer N, Morbidelli M. Process intensification by frontal chromatography: Performance comparison of resin and membrane adsorber for monovalent antibody aggregate removal. Biotechnol Bioeng 2019; 117:662-672. [PMID: 31788778 DOI: 10.1002/bit.27235] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 10/23/2019] [Accepted: 11/22/2019] [Indexed: 01/22/2023]
Abstract
Aggregates are amongst the most important product-related impurities to be removed during the downstream processing of antibodies due to their potential immunogenicity. Traditional operations use cation-exchange resins in bind-elute mode for their separation. However, frontal analysis is emerging as an alternative. In this study, a three-step process development for a membrane adsorber and a resin material is carried out, allowing the comparison between the stationary phases. Based on a screening study, optimal loading conditions are determined, which show that weak binding is favored on the membrane and strong binding on the resin. Transfer of these findings to breakthrough experiments shows that at 99% pool purity the yield is higher for the membrane, while the resin can be loaded twice as high, exceeding yields of 85%. For the investigated antibody and based on a given regeneration protocol, the productivity of the two phases is similar, ranging around 200 g/(L·h). Due to the higher loading, the resin requires about one-third less buffer than the membrane. Furthermore, the implementation of a wash step after loading allows to further increase yield by about 5%. In comparison to a generic bind-elute process, productivity and buffer consumption are improved by an order of magnitude.
Collapse
Affiliation(s)
- Sebastian Vogg
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, ETH Zurich, Zurich, Switzerland
| | - Felix Pfeifer
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, ETH Zurich, Zurich, Switzerland
| | - Nicole Ulmer
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, ETH Zurich, Zurich, Switzerland
| | - Massimo Morbidelli
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
141
|
Schneck NA, Ivleva VB, Rosales-Zavala E, Wang X, Gollapudi D, Cooper JW, Lei QP. Using LC-MS to Identify Clipping in Self-Assembled Nanoparticles During Vaccine Development. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2019; 30:2576-2579. [PMID: 31595432 PMCID: PMC7780558 DOI: 10.1007/s13361-019-02318-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/21/2019] [Accepted: 08/10/2019] [Indexed: 06/10/2023]
Abstract
A hemagglutinin stabilized stem nanoparticle (HA-SS-np) that is designed to provide broad protection against influenza is being developed as a potential vaccine. During an early formulation screening study, reducing gel (rCGE) analysis indicated product degradation in a few candidate buffers at the first-week accelerated stability point, whereas no change was shown in the size exclusion chromatography (SEC) measurement. A LC-MS workflow was therefore applied to investigate the integrity of this large HA-SS-np vaccine molecule (≈ 1 MDa). Application of LC-MS was critical to rationalize the conflicting results from the rCGE and SEC assays and led to the discovery that (1) an unexpected sequence clipping in the HA-SS-np subunits occurred, explaining the atypical reducing gel profile, and (2) an undisrupted disulfide bond held the two fragments together, explaining the unchanged SEC profile. This analytical case study led to a formulation buffer redesign, which mitigated the issue.
Collapse
Affiliation(s)
- Nicole A Schneck
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd., Gaithersburg, MD, 20878, USA
| | - Vera B Ivleva
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd., Gaithersburg, MD, 20878, USA
| | - Erwin Rosales-Zavala
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd., Gaithersburg, MD, 20878, USA
| | - Xiangchun Wang
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd., Gaithersburg, MD, 20878, USA
| | - Deepika Gollapudi
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd., Gaithersburg, MD, 20878, USA
| | - Jonathan W Cooper
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd., Gaithersburg, MD, 20878, USA
| | - Q Paula Lei
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9 West Watkins Mill Rd., Gaithersburg, MD, 20878, USA.
| |
Collapse
|
142
|
Brown CJ, Woodall DW, El-Baba TJ, Clemmer DE. Characterizing Thermal Transitions of IgG with Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2019; 30:2438-2445. [PMID: 31363989 PMCID: PMC6866664 DOI: 10.1007/s13361-019-02292-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/15/2019] [Accepted: 07/16/2019] [Indexed: 06/02/2023]
Abstract
Variable temperature electrospray ionization (ESI) is coupled with mass spectrometry techniques in order to investigate structural transitions of monoclonal antibody immunoglobulin G (IgG) in a 100-mM ammonium acetate (pH 7.0) solution from 26 to 70 °C. At 26 °C, the mass spectrum for intact IgG shows six charge states + 22 to + 26. Upon increasing the solution temperature, the fraction of low-charge states decreases and new, higher-charge state ions are observed. Upon analysis, it appears that heating the solution aids in desolvation of the intact IgG precursor. Above ~ 50 °C, a cleavage event between the light and heavy chains is observed. An analysis of the kinetics for these processes at different temperatures yields transition state thermochemistry of ΔH‡ = 95 ± 10 kJ mol-1, ΔS‡ = 8 ± 1 J mol-1 K-1, and ΔG‡ = 92 ± 11 kJ mol-1. The mechanism for light chain dissociation appears to involve disulfide bond scrambling that ultimately results in a non-native Cys199-Cys217 disulfide bond in the light chain product. Above ~ 70 °C, we are unable to produce a stable ESI signal. The loss of signal is ascribed to aggregation that is primarily associated with the remaining portion of the antibody after having lost the light chain. Graphical Abstract.
Collapse
Affiliation(s)
- Christopher J Brown
- Department of Chemistry, Indiana University, 800 Kirkwood Avenue, Bloomington, IN, 47401, USA
| | - Daniel W Woodall
- Department of Chemistry, Indiana University, 800 Kirkwood Avenue, Bloomington, IN, 47401, USA
| | - Tarick J El-Baba
- Department of Chemistry, Indiana University, 800 Kirkwood Avenue, Bloomington, IN, 47401, USA
| | - David E Clemmer
- Department of Chemistry, Indiana University, 800 Kirkwood Avenue, Bloomington, IN, 47401, USA.
| |
Collapse
|
143
|
Römer J, Montealegre C, Schlecht J, Kiessig S, Moritz B, Neusüß C. Online mass spectrometry of CE (SDS)-separated proteins by two-dimensional capillary electrophoresis. Anal Bioanal Chem 2019; 411:7197-7206. [PMID: 31616969 DOI: 10.1007/s00216-019-02102-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/01/2019] [Accepted: 08/23/2019] [Indexed: 10/25/2022]
Abstract
Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) is the fundamental technique for protein separation by size. Applying this technology in capillary format, gaining high separation efficiency in a more automated way, is a key technology for size separation of proteins in the biopharmaceutical industry. However, unequivocal identification by online mass spectrometry (MS) is impossible so far, due to strong interference in the electrospray process by SDS and other components of the SDS-MW separation gel buffer. Here, a heart-cut two-dimensional electrophoretic separation system applying an electrically isolated valve with an internal loop of 20 nL is presented. The peak of interest in the CE (SDS) separation is transferred to the CZE-MS, where electrospray-interfering substances of the SDS-MW gel are separated prior to online electrospray ionization mass spectrometry. An online SDS removal strategy for decomplexing the protein-SDS complex is implemented in the second dimension, consisting of the co-injection of organic solvent and cationic surfactant. This online CE (SDS)-CZE-MS system allows MS characterization of proteoforms separated in generic CE (SDS), gaining additional separation in the CZE and detailed MS information. In general, the system can be applied to all kinds of proteins separated by CE (SDS). Here, we present results of the CE (SDS)-CZE-MS system on the analysis of several biopharmaceutically relevant antibody impurities and fragments. Additionally, the versatile application spectrum of the system is demonstrated by the analysis of extracted proteins from soybean flour. The online hyphenation of CE (SDS) resolving power and MS identification capabilities will be a powerful tool for protein and mAb characterization. Graphical abstract Two-dimensional capillary electrophoresis system hyphenated with mass spectrometry for the characterization of CE (SDS)-separated proteins. As first dimension, a generic and high MS-interfering CE (SDS) separation is performed for size separation. After heart-cut transfer of the unknown CE (SDS) protein peak, via a four-port nanoliter valve to a volatile electrolyte system as second dimension, interference-free mass spectrometric data of separated mAb fragments and soybean proteins are obtained.
Collapse
Affiliation(s)
- Jennifer Römer
- Faculty of Chemistry, Aalen University, Beethovenstraße 1, 73430, Aalen, Germany
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany
| | - Cristina Montealegre
- Faculty of Chemistry, Aalen University, Beethovenstraße 1, 73430, Aalen, Germany
| | - Johannes Schlecht
- Faculty of Chemistry, Aalen University, Beethovenstraße 1, 73430, Aalen, Germany
- F. Hoffmann-La Roche Ltd, Grenzacherstraße 124, 4070, Basel, Switzerland
| | - Steffen Kiessig
- F. Hoffmann-La Roche Ltd, Grenzacherstraße 124, 4070, Basel, Switzerland
| | - Bernd Moritz
- F. Hoffmann-La Roche Ltd, Grenzacherstraße 124, 4070, Basel, Switzerland
| | - Christian Neusüß
- Faculty of Chemistry, Aalen University, Beethovenstraße 1, 73430, Aalen, Germany.
| |
Collapse
|
144
|
Sterjova M, Džodić P, Ruskovska T, Apostolova P, Risteski M, Janevik-Ivanovska E. PREPARATION AND INTEGRITY EXAMINATION OF FREEZE DRIED KIT OF TRASTUZUMAB-IMMUNOCONJUGATES AND COLD LABELED IMMUNOCONJUGATES BY APPLYING SDS-PAGE ELECTROPHORESIS. ACTA MEDICA MEDIANAE 2019. [DOI: 10.5633/amm.2019.0303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
145
|
Wang WH, Cheung-Lau J, Chen Y, Lewis M, Tang QM. Specific and high-resolution identification of monoclonal antibody fragments detected by capillary electrophoresis-sodium dodecyl sulfate using reversed-phase HPLC with top-down mass spectrometry analysis. MAbs 2019; 11:1233-1244. [PMID: 31348730 DOI: 10.1080/19420862.2019.1646554] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
In recent years, capillary electrophoresis-sodium dodecyl sulfate (cSDS) has been widely used for high resolution separation and quantification of the fragments and aggregates of monoclonal antibodies (mAbs) to ensure the quality of mAb therapeutics. However, identification of the low-molecular-weight (LMW) and high-molecular-weight (HMW) species detected in cSDS electropherograms has been based primarily on the approximate MWs calculated from standard curves using known MW standards and correlations with fragments and aggregates identified by other methods. It is not easy to collect sufficient amounts of H/LMW species from cSDS for analysis by orthogonal methods and the direct coupling of cSDS with mass spectrometry (MS) is very difficult due to interference from SDS. In this study, we describe the precise identification of H/LMW species detected by cSDS using reversed-phase high performance liquid chromatography (RP-HPLC) coupled with top-down tandem MS analysis. The H/LMW species were first identified by on-line RP-HPLC MS analysis and the RP-HPLC fractions were then analyzed by cSDS to connect the identified H/LMW species with the peaks in the cSDS electropherogram. With this method, 58 unique H/LMW species were identified from an immunoglobulin G1 (IgG1) mAb. The identified fragments ranged from 10 kDa single chain fragments to 130 kDa triple chain fragments, including some with post-translational modifications. This is the first study to clearly identify the antibody fragments, including the exact clipping sites, observed in cSDS electropherograms. The methodology and results presented here should be applicable to most other IgG1 mAbs.
Collapse
Affiliation(s)
- Wei-Han Wang
- Bio Therapeutics Development, Janssen Research & Development, LLC , Malvern , PA , USA
| | - Jasmina Cheung-Lau
- Bio Therapeutics Development, Janssen Research & Development, LLC , Malvern , PA , USA
| | - Yan Chen
- Bio Therapeutics Development, Janssen Research & Development, LLC , Malvern , PA , USA
| | - Michael Lewis
- Bio Therapeutics Development, Janssen Research & Development, LLC , Malvern , PA , USA
| | - Qing Mike Tang
- Bio Therapeutics Development, Janssen Research & Development, LLC , Malvern , PA , USA
| |
Collapse
|
146
|
Reader PP, Olkhov RV, Reeksting S, Lubben A, Hyde CJ, Shaw AM. A rapid and quantitative technique for assessing IgG monomeric purity, calibrated with the NISTmAb reference material. Anal Bioanal Chem 2019; 411:6487-6496. [PMID: 31375854 PMCID: PMC6718376 DOI: 10.1007/s00216-019-02029-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/19/2019] [Accepted: 07/10/2019] [Indexed: 11/28/2022]
Abstract
The fraction of intact monomer in a sample (moles/moles), the monomeric purity, is measured as a quality control in therapeutic monoclonal antibodies but is often unknown in research samples and remains a major source of variation in quantitative antibody-based techniques such as immunoassay development. Here, we describe a novel multiplex technique for estimating the monomeric purity and antigen affinity of research grade antibody samples. Light scattering was used to simultaneously observe the mass of antibody binding to biosensor surfaces functionalised with antigen (revealing Fab binding kinetics) or protein A/G (PAG). Initial estimates of monomeric purity in 7 antibody samples including a therapeutic infliximab biosimilar were estimated by observing a mass deficit on the PAG surface compared to the NISTmAb standard of high monomeric purity. Monomeric purity estimates were improved in a second step by observing the mass of antigen binding to the mass of antibody on the PAG surface. The NISTmAb and infliximab biosimilar displayed tightly controlled stoichiometries for antigen binding of 1.31 ± 0.57 and 1.71 ± 0.16 (95% confidence interval)—within the theoretical limit of 1–2 antigens per antibody depending on avidity. The other antibodies in the panel displayed antigen binding stoichiometries in the range 0.06–1.15, attributed to lower monomeric purity. The monomeric purity estimates were verified by electrospray ionization mass spectrometry (ESI), the gold standard technique for structural characterization of antibodies. ESI data indicated that the NISTmAb and infliximab biosimilar samples had monomeric purity values of 93.5% and 94.7%, respectively, whilst the research grade samples were significantly lower (54–89%). Our results demonstrate rapid quality control testing for monomeric purity of antibody samples (< 15 min) which could improve the reproducibility of antibody-based experiments.
Collapse
Affiliation(s)
- Peter P Reader
- University of Exeter Medical School, Heavitree Road, Exeter, EX1 2LU, UK.,College of Life and Environmental Sciences, University of Exeter, Stocker Road, Exeter, EX4 4QD, UK
| | - Rouslan V Olkhov
- College of Life and Environmental Sciences, University of Exeter, Stocker Road, Exeter, EX4 4QD, UK
| | - Shaun Reeksting
- Chemical Characterisation and Analysis Facility, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Anneke Lubben
- Chemical Characterisation and Analysis Facility, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Christopher J Hyde
- University of Exeter Medical School, Heavitree Road, Exeter, EX1 2LU, UK
| | - Andrew M Shaw
- College of Life and Environmental Sciences, University of Exeter, Stocker Road, Exeter, EX4 4QD, UK.
| |
Collapse
|
147
|
Zhu Y, Ahluwalia D, Chen Y, Belakavadi M, Katiyar A, Das TK. Characterization of therapeutic antibody fragmentation using automated capillary western blotting as an orthogonal analytical technique. Electrophoresis 2019; 40:2888-2898. [PMID: 31271455 DOI: 10.1002/elps.201900119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 06/14/2019] [Accepted: 06/25/2019] [Indexed: 11/06/2022]
Abstract
Fragmentation in protein-based molecules continues to be a challenge during manufacturing and storage, and requires an appropriate control strategy to ensure purity and integrity of the drug product. Electrophoretic and chromatographic methods are commonly used for monitoring the fragments. However, size-exclusion chromatography often suffers from low resolution of low molecular weight fragments. Electrophoretic methods like CE-SDS are not compatible with enriching fragments for additional characterization tests such as MS. These limitations may result in inadequate control strategy for monitoring and characterizing fragments for protein-based molecules. Capillary western blotting was used in this study as an orthogonal method for characterization of fragments in an IgG1 antibody under reduced conditions. To achieve a comprehensive mapping of various fragments generated by thermal stress, capillary western profiles were generated using recognition antibodies for IgG kappa (κ) light chain, Fc, and Fab regions that enabled unambiguous fragment identification. Additionally, three different enzymatic digestion methods (IdeS, PNGase F, and IgdE) were applied coupled with capillary western blotting for clip identifications. Finally, complementary data collected using traditional chromatographic and electrophoretic methods allowed to establish a comparison of analytical profiles with an added benefit of fragment identification offered by capillary western profiling. In addition to various Fc and Fab-related low molecular weight fragments, a non-reducible thio-ether linked 75 kDa HL fragment was also identified.
Collapse
Affiliation(s)
- Yunxiao Zhu
- Methods and Analytical Development, Bristol-Myers Squibb, Pennington, NJ, USA
| | - Deepti Ahluwalia
- Methods and Analytical Development, Bristol-Myers Squibb, Pennington, NJ, USA
| | - Yingchen Chen
- Methods and Analytical Development, Bristol-Myers Squibb, Pennington, NJ, USA
| | - Madesh Belakavadi
- Methods and Analytical Development, Bristol-Myers Squibb, Pennington, NJ, USA
| | - Amit Katiyar
- Methods and Analytical Development, Bristol-Myers Squibb, Pennington, NJ, USA.,Analytical and Formulation Sciences, Patheon, Thermo Fisher Scientific, Princeton, NJ, USA
| | - Tapan K Das
- Methods and Analytical Development, Bristol-Myers Squibb, Pennington, NJ, USA
| |
Collapse
|
148
|
Manikwar P, Mulagapati SHR, Kasturirangan S, Moez K, Rainey GJ, Lobo B. Characterization of a Novel Bispecific Antibody With Improved Conformational and Chemical Stability. J Pharm Sci 2019; 109:220-232. [PMID: 31288034 DOI: 10.1016/j.xphs.2019.06.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 06/02/2019] [Accepted: 06/18/2019] [Indexed: 02/08/2023]
Abstract
Bispecific antibodies containing single-chain variable fragment (scFv) appended to immunoglobulins G offer unique development challenges. Here, we describe the stability of a novel bispecific format, BiS5, where the scFv is tethered to the CH3 domain. BiS5 showed an improved conformational and chemical stability compared with that of BiS4 in which the scFv is appended in the hinge region between the Fab and Fc. By switching the location of the scFv from hinge region to the CH3, there was an improved stabilization of CH2 and scFv domains. Interestingly, no noticeable impact was observed on the conformational stability of CH3 and Fab domains. BiS4 and BiS5 showed different aggregation and fragmentation rates under accelerated temperature stress conditions. BiS4 showed higher fragmentation rates compared with BiS5 likely owing to fragmentation in the linker region on either side of the scFv while BiS5 is more resistant toward fragmentation owing to tethering of scFv to the CH3 domain at its N and C terminus. In conclusion, the location of scFv affects both aggregation and fragmentation kinetics. These insights into the molecular structure and correlations with their physical and chemical stability will help formulation development of these novel bispecific antibodies.
Collapse
Affiliation(s)
- Prakash Manikwar
- Dosage Form Design & Development, AstraZeneca, Gaithersburg, Maryland 20878.
| | | | - Srinath Kasturirangan
- Antibody Discovery and Protein Engineering, AstraZeneca, Gaithersburg, Maryland 20878
| | - Khashayar Moez
- Dosage Form Design & Development, AstraZeneca, Gaithersburg, Maryland 20878
| | - Godfrey Jonah Rainey
- Antibody Discovery and Protein Engineering, AstraZeneca, Gaithersburg, Maryland 20878
| | - Brian Lobo
- Dosage Form Design & Development, AstraZeneca, Gaithersburg, Maryland 20878
| |
Collapse
|
149
|
Kerr RA, Keire DA, Ye H. The impact of standard accelerated stability conditions on antibody higher order structure as assessed by mass spectrometry. MAbs 2019; 11:930-941. [PMID: 30913973 PMCID: PMC6601562 DOI: 10.1080/19420862.2019.1599632] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 02/23/2019] [Accepted: 03/21/2019] [Indexed: 01/12/2023] Open
Abstract
Protein therapeutic higher order structure (HOS) is a quality attribute that can be assessed to help predict shelf life. To model product shelf-life values, possible sample-dependent pathways of degradation that may affect drug efficacy or safety need to be evaluated. As changes in drug thermal stability over time can be correlated with an increased risk of HOS perturbations, the effect of long-term storage on the product should be measured as a function of temperature. Here, complementary high-resolution mass spectrometry methods for HOS analysis were used to identify storage-dependent changes of biotherapeutics (bevacizumab (Avastin), trastuzumab (Herceptin), rituximab (Rituxan), and the NIST reference material 8671 (NISTmAb)) under accelerated or manufacturer-recommended storage conditions. Collision-induced unfolding ion mobility-mass spectrometry data showed changes in monoclonal antibody folded stability profiles that were consistent with the appearance of a characteristic unfolded population. Orthogonal hydrogen-deuterium exchange-mass spectrometry data revealed that the observed changes in unfolding occurred in parallel to changes in HOS localized to the periphery of the hinge region. Using intact reverse-phase liquid chromatography-mass spectrometry, we identified several mass species indicative of peptide backbone hydrolysis, located between the variable and constant domains of the heavy chain of bevacizumab. Taken together, our data highlighted the capability of these approaches to identify age- or temperature-dependent changes in biotherapeutic HOS.
Collapse
Affiliation(s)
- Richard A. Kerr
- Division of Pharmaceutical Analysis, Office of Testing and Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, St. Louis, USA
| | - David A. Keire
- Division of Pharmaceutical Analysis, Office of Testing and Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, St. Louis, USA
| | - Hongping Ye
- Division of Pharmaceutical Analysis, Office of Testing and Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, St. Louis, USA
| |
Collapse
|
150
|
Wang Z, Friedrich MG, Truscott RJW, Schey KL. Cleavage C-terminal to Asp leads to covalent crosslinking of long-lived human proteins. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2019; 1867:831-839. [PMID: 31226490 DOI: 10.1016/j.bbapap.2019.06.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/29/2019] [Accepted: 06/16/2019] [Indexed: 01/27/2023]
Abstract
With age, long-lived proteins in the human body deteriorate, which can have consequences both for aging and disease. The aging process is often associated with the formation of covalently crosslinked proteins. Currently our knowledge of the mechanism of formation of these crosslinks is limited. In this study, proteomics was used to characterize sites of covalent protein-protein crosslinking and identify a novel mechanism of protein-protein crosslinking in the adult human lens. In this mechanism, Lys residues are crosslinked to C-terminal Asp residues that are formed by non-enzymatic protein truncation. Ten different crosslinks were identified in major lens proteins such as αA-crystallin, αB-crystallin and AQP0. Crosslinking in AQP0 increased significantly with age and also increased significantly in cataract lenses compared with normal lenses. Using model peptides, a mechanism of formation of the Lys-Asp crosslink was elucidated. The mechanism involves spontaneous peptide cleavage on the C-terminal side of Asp residues which can take place in the pH range 5-7.4. Cleavage appears to involve attack by the side chain carboxyl group on the adjacent peptide bond, resulting in the formation of a C-terminal Asp anhydride. This anhydride intermediate can then either react with water to form Asp, or with a nucleophile, such as a free amine group to form a crosslink. If an ε-amino group of Lys or an N-terminal amine group attacks the anhydride, a covalent protein-protein crosslink will be formed. This bi-phasic mechanism represents the first report to link two spontaneous events: protein cleavage and crosslinking that are characteristic of long-lived proteins.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Biochemistry and Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Michael G Friedrich
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Roger J W Truscott
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia.
| | - Kevin L Schey
- Department of Biochemistry and Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|