1
|
Low KN, Kok YJ, Tate S, Bi X. Multilevel─Intact, Subunits, and Peptides─Characterization of Antibody-Based Therapeutics by a Single-Column LC-MS Setup. Anal Chem 2025; 97:5118-5125. [PMID: 39999080 DOI: 10.1021/acs.analchem.4c06350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
A comprehensive characterization of biotherapeutics, mandated by regulatory authorities, requires analyses of a protein drug at multiple structure levels. Such multilevel characterization can be performed by mass spectrometry (MS), with established conventional MS-based assays of product quality attributes (PQAs) comprising intact protein and subunit middle-up MS with analytes resolved on a C4 column, and bottom-up peptide mapping with analytes resolved on a C18 column. Recent advances in MS have facilitated the increasing use of middle-down analysis, expanding the qualitative analytical capability of MS for protein characterization. Recent studies using less-retentive reversed-phase LC in bottom-up MS also offer an opportunity for streamlining equipment configuration to a single-column LC-MS setup for multilevel characterization of therapeutic proteins. In this study, we developed a robust middle-down LC-MS method on a ZenoTOF 7600 and evaluated a C4 LC-MS setup for the characterization of NISTmAb, RG7221 bispecific antibody (bsAb), and Fc-fusion etanercept by intact protein, subunit middle-up/down, and bottom-up analyses. Successful multilevel characterization of the analytes using C4 LC-MS was demonstrated; notably, high sequence coverage and comprehensive post-translational modification profiling, including the mapping of all 13 O- and 3 N-glycosylation sites on etanercept in a single run, were obtained by bottom-up C4 LC-MS. This is also the first report on middle-down analysis of the major etanercept TNFR and Fc subunit glycoforms. A streamlined single-column LC-MS setup will enable more robust and efficient MS workflows for PQA assessment and simplify the integration of an LC-MS analyzer as a process analytical technology instrument for biopharma applications.
Collapse
Affiliation(s)
- Kia-Ngee Low
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Singapore
| | - Yee-Jiun Kok
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Singapore
| | | | - Xuezhi Bi
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
- Food, Chemical and Biotechnology Cluster, Singapore Institute of Technology, Singapore 138683, Singapore
| |
Collapse
|
2
|
Pezeshkpoor B, Sereda N, Becker-Gotot J, Berkemeier AC, Matuschek I, Müller J, Ramaraje Urs SU, Singh S, Klein C, Marquardt N, Oldenburg J. Comprehensive evaluation of anti-emicizumab antibodies in acquired hemophilia A: a detailed case study and methodological evaluation. J Thromb Haemost 2025; 23:85-96. [PMID: 39401737 DOI: 10.1016/j.jtha.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/19/2024] [Accepted: 10/03/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND Acquired hemophilia A (AHA) is a rare and severe bleeding disorder characterized by autoantibodies inhibiting coagulation factor (F)VIII. Current treatment of AHA involves bypassing agents or FVIII replacement therapy, yet their efficacy is limited in cases of high inhibitor titers. Emicizumab, a humanized bispecific monoclonal antibody, has shown promising hemostatic effectiveness in persons with congenital hemophilia A (HA) and AHA, but a minority of patients developed anti-drug antibodies (ADAs), compromising its efficacy. OBJECTIVES This study aims to characterize the development and impact of anti-emicizumab antibodies in a patient with acquired hemophilia A (AHA) who experienced reduced efficacy of emicizumab treatment. METHODS We present a comprehensive characterization of anti-emicizumab antibodies in a patient with AHA experiencing diminished emicizumab efficacy from week 10 of treatment. We developed a method for analysis of anti-emicizumab antibodies, distinguishing immunoglobulin (Ig) subclasses and IgG subtype profiling. ADAs' neutralizing activity was evaluated using a modified clotting assay. RESULTS The Luminex-based assay demonstrated the presence of anti-emicizumab antibodies, confirmed through competitive analysis. We detected anti-emicizumab IgG antibodies in the patient's plasma between week 16 and 29. IgG1 and IgG2 subclasses were identified. Longitudinal analysis showed IgG isotype antibodies against both emicizumab and FVIII. Anti-emicizumab antibodies exhibited noninhibitory activity, confirmed by a modified Bethesda assay. Moreover, no accelerated clearance of emicizumab was observed in plasma samples from the patient. CONCLUSION This study presents the first documented case of anti-emicizumab antibodies in AHA. Our study provides insights into ADA development against emicizumab, emphasizing the need for monitoring tools. The developed method enables comprehensive evaluation of ADAs, aiding in personalized treatment strategies. These findings contribute to understanding emicizumab's immunogenicity profile in AHA, facilitating its optimized clinical use.
Collapse
MESH Headings
- Humans
- Hemophilia A/drug therapy
- Hemophilia A/immunology
- Hemophilia A/blood
- Hemophilia A/diagnosis
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/therapeutic use
- Antibodies, Bispecific/adverse effects
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Factor VIII/immunology
- Factor VIII/antagonists & inhibitors
- Treatment Outcome
- Male
- Antibodies, Neutralizing/blood
- Blood Coagulation/drug effects
- Immunoglobulin G/blood
- Middle Aged
- Autoantibodies/blood
- Drug Monitoring/methods
Collapse
Affiliation(s)
- Behnaz Pezeshkpoor
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Medical Faculty, University of Bonn, Germany.
| | - Nadja Sereda
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Medical Faculty, University of Bonn, Germany
| | - Janine Becker-Gotot
- Institute of Molecular Medicine and Experimental Immunology (IMMEI), University of Bonn, Bonn, Germany
| | - Ann-Cristin Berkemeier
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Medical Faculty, University of Bonn, Germany
| | - Isabell Matuschek
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Medical Faculty, University of Bonn, Germany
| | - Jens Müller
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Medical Faculty, University of Bonn, Germany
| | - Samhitha Urs Ramaraje Urs
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Medical Faculty, University of Bonn, Germany
| | - Sneha Singh
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Medical Faculty, University of Bonn, Germany
| | - Claudia Klein
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Medical Faculty, University of Bonn, Germany; Center for Rare Diseases Bonn (Zentrum für seltene Erkrankungen der Universität Bonn), University Clinic Bonn, Bonn, Germany
| | - Natascha Marquardt
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Medical Faculty, University of Bonn, Germany; Center for Rare Diseases Bonn (Zentrum für seltene Erkrankungen der Universität Bonn), University Clinic Bonn, Bonn, Germany
| | - Johannes Oldenburg
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Medical Faculty, University of Bonn, Germany; Center for Rare Diseases Bonn (Zentrum für seltene Erkrankungen der Universität Bonn), University Clinic Bonn, Bonn, Germany.
| |
Collapse
|
3
|
Schairer J, Römer J, Neusüß C. CE-MS and CE-MS/MS for the multiattribute analysis of monoclonal antibody variants at the subunit level. J Pharm Biomed Anal 2025; 252:116495. [PMID: 39368136 DOI: 10.1016/j.jpba.2024.116495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/21/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
The analysis of product-related substances and impurities is a critical step in the biopharmaceutical quality control of multiattribute monoclonal antibodies (mAbs), as posttranslational modifications or other variants can influence the product's biological activity. Many approaches are available for variant analysis; however, they are either variant-specific, mAb-specific, time-consuming, or require expensive equipment. Here, we present a generic capillary electrophoretic method based on a neutral-coated capillary which was coupled to mass spectrometry (MS) via the nanoCEasy interface for mAb variant analysis at the subunit level (enzymatically digested and reduced mAb). The method enabled the separation of several (i) size variants (e.g. glycosylation variants) and (ii) charge variants (e.g. c-terminal lysin clipping) as well as (iii) multiple other proteoforms (e.g. additional glycation) and (iv) incompletely reduced subunits. Separated variants were confirmed by MS/MS fragmentation even for small mass deviations like deamidation or open disulfide bridges. The system, initially developed for one mAb, was tested with nine other IgG1s to show the general applicability of the system. The presented multiattribute method enables fast and detailed characterization of mAb variants with little sample preparation and relatively simple separation equipment enabling the separation of a large set of mAb variants.
Collapse
Affiliation(s)
- Jasmin Schairer
- Faculty of Chemistry, Aalen University, Beethovenstraße 1, Aalen 73430, Germany; Faculty of Science, University of Tübingen, Auf der Morgenstelle 8, Tübingen 72076, Germany
| | - Jennifer Römer
- Rentschler Biopharma SE, Erwin-Rentschler-Straße 21, Laupheim 88471, Germany
| | - Christian Neusüß
- Faculty of Chemistry, Aalen University, Beethovenstraße 1, Aalen 73430, Germany.
| |
Collapse
|
4
|
Niu B, Lee B, Chen W, Alberto C, Betancourt Moreira K, Compton P, Homan K, Pinckney J, Zhu Y, Vendel M, Wetterhorn K, Walrond S, Santha E, Horowitz A, Zaubi N, Johnson J. End-To-End Automated Intact Protein Mass Spectrometry for High-Throughput Screening and Characterization of Bispecific and Multispecific Antibodies. Anal Chem 2024; 96:18287-18300. [PMID: 39479787 PMCID: PMC11562946 DOI: 10.1021/acs.analchem.4c04833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 11/13/2024]
Abstract
Bispecific antibodies (bsAbs) and multispecific antibodies (msAbs) represent a promising frontier in therapeutic antibody development, offering unique capabilities not achievable with traditional monoclonal antibodies. Despite their potential, significant challenges remain due to their increased molecular complexity. One prominent challenge is the correct assembly of light and heavy chains, as improper pairing leads to mispaired or incompletely assembled species that lack therapeutic efficacy and possess undesired properties, impairing the developability, manufacturability, and safety. There is a critical need for rapid, sensitive analytical tools to monitor and control these undesired species and ensure the quality assessment of bsAbs and msAbs. To address this need, we present a novel high-throughput, format-agnostic intact mass workflow that significantly enhances the efficiency of detecting and quantifying biotherapeutic products and related impurities. This workflow integrates automated sample preparation, novel high-resolution rapid mass detection powered by SampleStream-MS, and an advanced data analysis pipeline. It offers increased throughput and data quality while substantially reducing analysis turnover time and labor. This was demonstrated in a pilot program where ∼800 multispecific antibodies were processed in 10 working days. The article details the evaluation and validation of our method, demonstrating its repeatability and intermediate precision in terms of measurement accuracy and relative quantification of various product-related species. We underscore the transformative potential of this end-to-end high-throughput workflow in expediting bispecific and multispecific antibody discovery, optimizing production processes, and ensuring high-quality development and manufacturing for therapeutic antibodies.
Collapse
Affiliation(s)
- Ben Niu
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Benjamin Lee
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Wen Chen
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Cristian Alberto
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Karen Betancourt Moreira
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Philip Compton
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Kristoff Homan
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Jason Pinckney
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Yaxing Zhu
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Michelle Vendel
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Karl Wetterhorn
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Shana Walrond
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Esrath Santha
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Amanda Horowitz
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Nicole Zaubi
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Jeffrey Johnson
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| |
Collapse
|
5
|
Wu G, Zhang X, Wang X, Du J, Li M, Xu G, Du M, Yu C. In-depth characterization of a cysteine-linked ADC disitamab vedotin by multiple LC-MS analysis methods and cutting-edge imaged capillary isoelectric focusing coupled with native mass spectrometry. J Chromatogr A 2024; 1736:465353. [PMID: 39270568 DOI: 10.1016/j.chroma.2024.465353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024]
Abstract
The characterization of cysteine-linked antibody‒drug conjugates (ADCs) can be more challenging than that of monoclonal antibodies (mAbs) and lysine-linked ADCs because the interchain disulfide bonds are reduced for payload conjugation, and the chains are noncovalently bonded to each other. Furthermore, payload conjugation and disulfide bond reduction/scrambling may introduce additional charge heterogeneity to biomolecules. This study illustrates an innovative workflow employing multiple separation techniques and tandem high-resolution mass spectrometry for comprehensive and in-depth characterization of disitamab vedotin, a recent-generation cysteine-linked ADC, including reversed-phase liquid chromatography (RPLC), ion exchange chromatography (IEX) and image capillary isoelectric focusing (icIEF). RPLC was employed for reduced chains analysis, subunit analysis and peptide mapping. IEX and icIEF were used for charge heterogeneity analysis. The innovation of the integrated methodology emphasizes the importance of cutting-edge icIEF-MS online coupling under near-native conditions to reveal the heterogeneity of disitamab vedotin.
Collapse
Affiliation(s)
- Gang Wu
- National Institutes for Food and Drug Control, State Key Laboratory of Drug Regulatory Science, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Daxing District, Beijing, 102629, China
| | - Xiaoxi Zhang
- Thermo Fisher Scientific, Shanghai,200000, China
| | - Xin Wang
- Fujian Institute for Food and Drug Quality Control, Fuzhou, 350000,China
| | - Jialiang Du
- National Institutes for Food and Drug Control, State Key Laboratory of Drug Regulatory Science, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Daxing District, Beijing, 102629, China
| | - Meng Li
- National Institutes for Food and Drug Control, State Key Laboratory of Drug Regulatory Science, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Daxing District, Beijing, 102629, China
| | - Gangling Xu
- National Institutes for Food and Drug Control, State Key Laboratory of Drug Regulatory Science, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Daxing District, Beijing, 102629, China
| | - Min Du
- Thermo Fisher Scientific, Lexington, MA, 02421, US
| | - Chuanfei Yu
- National Institutes for Food and Drug Control, State Key Laboratory of Drug Regulatory Science, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Daxing District, Beijing, 102629, China.
| |
Collapse
|
6
|
Isu S, Vinskus L, Silva D, Cunningham K, Elich T, Greenhalgh P, Sokolnicki A, Raghunath B. Leveraging bioanalytical characterization of fractionated monoclonal antibody pools to identify aggregation-prone and less filterable proteoforms during virus filtration. Biotechnol Prog 2024; 40:e3451. [PMID: 38450976 DOI: 10.1002/btpr.3451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 01/11/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024]
Abstract
Monoclonal antibodies (mAbs) are an essential class of biotherapeutics. A platform process is used for mAb development to ensure clinically safe and stable molecules. Regulatory authorities ensure that mAb production processes include sufficient viral clearance steps to achieve less than one virus particle per million doses of product. Virus filtration is used for size-based removal of enveloped and nonenveloped viruses during downstream processing of mAbs. Process development in mAb purification relies on empirical approaches and often includes adsorptive prefiltration to mitigate virus filter fouling. Opportunities for molecular-level prediction of mAb filterability are needed to plug the existing knowledge gap in downstream processing. A molecular-level approach to understanding the factors influencing mAb filterability may reduce process development time, material loss, and processing costs due to oversized virus filters. In this work, pH step gradient fractionation was applied on polished bulk mAb feed to obtain concentrated pools of fractionated mAb variants. Biophysical properties and quality attributes of fractionated pools, including oligomeric state (size), isoelectric point profile, diffusion interaction parameters, and glycoform profile, were determined using bioanalytical methods. Filterability (loading and throughput) of fractionated pools were evaluated. Statistical methods were used to obtain correlations between quality attributes of mAb fractions and filterability on the Viresolve Pro virus filter.
Collapse
Affiliation(s)
- Solomon Isu
- Process Solutions, MilliporeSigma, Burlington, Massachusetts, USA
| | - Lilia Vinskus
- Process Solutions, MilliporeSigma, Burlington, Massachusetts, USA
| | - Derek Silva
- Process Solutions, MilliporeSigma, Burlington, Massachusetts, USA
| | | | - Thomas Elich
- Process Solutions, MilliporeSigma, Burlington, Massachusetts, USA
| | | | - Adam Sokolnicki
- Process Solutions, MilliporeSigma, Burlington, Massachusetts, USA
| | - Bala Raghunath
- Process Solutions, MilliporeSigma, Burlington, Massachusetts, USA
| |
Collapse
|
7
|
Benazza R, Koutsopetras I, Vaur V, Chaubet G, Hernandez-Alba O, Cianférani S. SEC-MS in denaturing conditions (dSEC-MS) for in-depth analysis of rebridged monoclonal antibody-based formats. Talanta 2024; 272:125727. [PMID: 38364570 DOI: 10.1016/j.talanta.2024.125727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 12/04/2023] [Accepted: 01/27/2024] [Indexed: 02/18/2024]
Abstract
Disulfide rebridging methods are emerging recently as new ways to specifically modify antibody-based entities and produce future conjugates. Briefly, the solvent-accessible disulfide bonds of antibodies or antigen-binding fragments (Fab) thereof are reduced under controlled conditions and further covalently attached with a rebridging agent allowing the incorporation of one payload per disulfide bond. There are many examples of successful rebridging cases providing homogeneous conjugates due to the use of symmetrical reagents, such as dibromomaleimides. However, partial rebridging due to the use of unsymmetrical ones, containing functional groups with different reactivity, usually leads to the development of heterogeneous species that cannot be identified by a simple sodium dodecyl sulfate-polyacrylamide gel eletrophoresis (SDS-PAGE) due to its lack of sensitivity, resolution and low mass accuracy. Mass spectrometry coupled to liquid chromatography (LC-MS) approaches have already been demonstrated as highly promising alternatives for the characterization of newly developed antibody-drug-conjugate (ADC) and monoclonal antibody (mAb)-based formats. We report here the in-depth characterization of covalently rebridged antibodies and Fab fragments in-development, using size-exclusion chromatography hyphenated to mass spectrometry in denaturing conditions (denaturing SEC-MS, dSEC-MS). DSEC-MS was used to monitor closely the rebridging reaction of a conjugated trastuzumab, in addition to conjugated Fab fragments, which allowed an unambiguous identification of the covalently rebridged products along with the unbound species. This all-in-one approach allowed a straightforward analysis of the studied samples with precise mass measurement; critical quality attributes (CQAs) assessment along with rebridging efficiency determination.
Collapse
Affiliation(s)
- Rania Benazza
- Laboratoire de Spectrométrie de Masse BioOrganique, IPHC UMR 7178, Université de Strasbourg, CNRS, 67087 Strasbourg, France; Infrastructure Nationale de Protéomique ProFI-FR2048, 67087 Strasbourg, France
| | - Ilias Koutsopetras
- Bio-Functional Chemistry (UMR 7199), Institut du Médicament de Strasbourg, University of Strasbourg, 74 Route du Rhin, 67400 Illkirch-Graffenstaden, France
| | - Valentine Vaur
- Bio-Functional Chemistry (UMR 7199), Institut du Médicament de Strasbourg, University of Strasbourg, 74 Route du Rhin, 67400 Illkirch-Graffenstaden, France
| | - Guilhem Chaubet
- Bio-Functional Chemistry (UMR 7199), Institut du Médicament de Strasbourg, University of Strasbourg, 74 Route du Rhin, 67400 Illkirch-Graffenstaden, France
| | - Oscar Hernandez-Alba
- Laboratoire de Spectrométrie de Masse BioOrganique, IPHC UMR 7178, Université de Strasbourg, CNRS, 67087 Strasbourg, France; 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; Infrastructure Nationale de Protéomique ProFI-FR2048, 67087 Strasbourg, France.
| |
Collapse
|
8
|
D'Atri V, Imiołek M, Quinn C, Finny A, Lauber M, Fekete S, Guillarme D. Size exclusion chromatography of biopharmaceutical products: From current practices for proteins to emerging trends for viral vectors, nucleic acids and lipid nanoparticles. J Chromatogr A 2024; 1722:464862. [PMID: 38581978 DOI: 10.1016/j.chroma.2024.464862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/29/2024] [Accepted: 03/31/2024] [Indexed: 04/08/2024]
Abstract
The 21st century has been particularly productive for the biopharmaceutical industry, with the introduction of several classes of innovative therapeutics, such as monoclonal antibodies and related compounds, gene therapy products, and RNA-based modalities. All these new molecules are susceptible to aggregation and fragmentation, which necessitates a size variant analysis for their comprehensive characterization. Size exclusion chromatography (SEC) is one of the reference techniques that can be applied. The analytical techniques for mAbs are now well established and some of them are now emerging for the newer modalities. In this context, the objective of this review article is: i) to provide a short historical background on SEC, ii) to suggest some clear guidelines on the selection of packing material and mobile phase for successful method development in modern SEC; and iii) to highlight recent advances in SEC, such as the use of narrow-bore and micro-bore columns, ultra-wide pore columns, and low-adsorption column hardware. Some important innovations, such as recycling SEC, the coupling of SEC with mass spectrometry, and the use of alternative detectors such as charge detection mass spectrometry and mass photometry are also described. In addition, this review discusses the use of SEC in multidimensional setups and shows some of the most recent advances at the preparative scale. In the third part of the article, the possibility of SEC for the characterization of new modalities is also reviewed. The final objective of this review is to provide a clear summary of opportunities and limitations of SEC for the analysis of different biopharmaceutical products.
Collapse
Affiliation(s)
- Valentina D'Atri
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU - Rue Michel Servet 1,4, 1211 Geneva, Switzerland; School of Pharmaceutical Sciences, University of Geneva, CMU - Rue Michel Servet 1,4, 1211 Geneva, Switzerland
| | | | | | - Abraham Finny
- Waters Corporation, Wyatt Technology, Santa Barbara, CA, USA
| | - Matthew Lauber
- Waters Corporation, Wyatt Technology, Santa Barbara, CA, USA
| | | | - Davy Guillarme
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU - Rue Michel Servet 1,4, 1211 Geneva, Switzerland; School of Pharmaceutical Sciences, University of Geneva, CMU - Rue Michel Servet 1,4, 1211 Geneva, Switzerland.
| |
Collapse
|
9
|
Wu G, Du J, Yu C, Fu Z, Zhang X, Wang L, Wang J. Mass spectrometry study on SARS-CoV-2 recombinant vaccine with comprehensive separation techniques to characterize complex heterogeneity. Anal Chim Acta 2024; 1297:342349. [PMID: 38438233 DOI: 10.1016/j.aca.2024.342349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/01/2024] [Accepted: 02/04/2024] [Indexed: 03/06/2024]
Abstract
SARS-CoV-2, the causative agent of COVID-19, has imposed a major public health threat, which needs effective therapeutics and vaccination strategies. Several potential candidate vaccines being rapidly developed are in clinical evaluation and recombinant vaccine has gained much attention thanks to its potential for greater response predictability, improved efficacy, rapid development and reduced side effects. Recombinant vaccines are designed and manufactured using bacterial, yeast cells or mammalian cells. A small piece of DNA is taken from the virus or bacterium against which we want to protect and inserted into the manufacturing cells. Due to the extremely complex heterogeneity of SARS-CoV-2 recombinant vaccine, single technology platform cannot achieve thorough and accurate characterization of such difficult proteins so integrating comprehensive technologies is essential. This study illustrates an innovative workflow employing multiple separation techniques tandem high-resolution mass spectrometry for comprehensive and in-depth characterization of SARS-CoV-2 recombinant vaccine, including ultra-high performance liquid chromatography (UHPLC), ion exchange chromatography (IEX) and imaged capillary isoelectric focusing (icIEF). The integrated methodology focuses on the importance of cutting-edge icIEF-MS online coupling and icIEF fractionation applied to revealing the heterogeneity secret of SARS-CoV-2 recombinant vaccine.
Collapse
Affiliation(s)
- Gang Wu
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, Liaoning, 110016, China
| | - Jialiang Du
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, Daxing District, Beijing, 102629, 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, Daxing District, Beijing, 102629, China
| | - Zhihao Fu
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, Daxing District, Beijing, 102629, China
| | - Xiaoxi Zhang
- Thermo Fisher Scientific, A Building, Henggu1976, No.1976 Middle Gaoke Road, Pudong District, 201203, Shanghai, 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, Daxing District, Beijing, 102629, China
| | - Junzhi Wang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, Liaoning, 110016, China.
| |
Collapse
|
10
|
Schairer J, Römer J, Lang D, Neusüß C. CE-MS/MS and CE-timsTOF to separate and characterize intramolecular disulfide bridges of monoclonal antibody subunits and their application for the assessment of subunit reduction protocols. Anal Bioanal Chem 2024; 416:1599-1612. [PMID: 38296860 PMCID: PMC10899284 DOI: 10.1007/s00216-024-05161-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 02/02/2024]
Abstract
Characterization at the subunit level enables detailed mass spectrometric characterization of posttranslational modifications (PTMs) of monoclonal antibodies (mAbs). The implemented reduction often leaves the intramolecular disulfide bridges intact. Here, we present a capillary electrophoretic (CE) method based on a neutral-coated capillary for the separation of immunoglobulin G-degrading enzyme of Streptococcus pyogenes (IdeS) digested and reduced mAb subunits followed by mass spectrometry (MS), MS/MS identification, and trapped ion mobility mass spectrometry (timsTOF). Our CE approach enables the separation of (i) different subunit moieties, (ii) various reduction states, and (iii) positional isomers of these partly reduced subunit moieties. The location of the remaining disulfide bridges can be determined by middle-down electron transfer higher energy collisional dissociation (EThcD) experiments. All these CE-separated variants show differences in ion mobility in the timsTOF measurements. Applying the presented CE-MS/MS method, reduction parameters such as the use of chaotropic salts were studied. For the investigated antibodies, urea improved the subunit reduction significantly, whereas guanidine hydrochloride (GuHCl) leads to multiple signals of the same subunit in the CE separation. The presented CE-MS method is a powerful tool for the disulfide-variant characterization of mAbs on the subunit level. It enables understanding disulfide bridge reduction processes in antibodies and potentially other proteins.
Collapse
Affiliation(s)
- Jasmin Schairer
- Faculty of Chemistry, Aalen University, Aalen, Germany
- Faculty of Science, University of Tübingen, Tübingen, Germany
| | | | | | | |
Collapse
|
11
|
Shah A, Cui W, Harrahy J, Ivanov AR. Characterization of charge variants, including post-translational modifications and proteoforms, of bispecific antigen-binding protein by cation-exchange chromatography coupled to native mass spectrometry. Talanta 2024; 266:125062. [PMID: 37566926 PMCID: PMC10528315 DOI: 10.1016/j.talanta.2023.125062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/29/2023] [Accepted: 08/06/2023] [Indexed: 08/13/2023]
Abstract
Charge variant characterization of biologics is critical to ensure that product meets the required quality and regulatory requirements to ensure safety and efficacy of the biotherapeutic. Charge variants arise from post-translation modifications (PTMs) during upstream processing and due to enzymatic and non-enzymatic chemical reactions that occur during downstream processing and storage. Some of these modifications may impact therapeutic potency, efficacy, or immunogenicity of a biotherapeutic. The traditional workflow for characterizing charge variants that involves fraction enrichment is time-consuming and labor-intensive. This approach can be especially challenging if the product is manufactured at low concentrations (e.g., ≤2 mg/mL). Recent advances in pH-based elution for ion-exchange chromatography utilizing volatile buffers have enabled rapid native mass-spectrometry-based identification of PTMs and proteoforms associated with protein therapeutics. In this study, we develop a novel workflow to rapidly and unambiguously characterize modifications associated with a new class of biotherapeutics known as bispecific antigen-binding protein (BsABP), including low-level modifications. A cation-exchange separation was optimized using volatile buffers to provide online hyphenation for native mass spectrometry to profile modifications and proteoforms present at the native level of a biotherapeutic, such as deamidation, O-glycosylation, amino acid substitution, N-linked glycosylation and oxidation. Furthermore, a limited proteolysis method was developed to specifically inform about modifications in the different domains of the bispecific antibody. Using this approach, we could efficiently identify PTMs in unstressed, thermally and photo-stressed samples, and provide information about the impact of downstream purification in clearing out modified BsABP species. Furthermore, peptide mapping was performed to identify and confirm modifications at the amino acid residue level. The developed workflow is less time-consumable and reduces sample processing- and analysis-related artifacts compared to traditional approaches.
Collapse
Affiliation(s)
- Arnik Shah
- Amgen Inc, 360 Binney Street, Cambridge, MA, 02141, United States; Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Ave., Boston, MA, 02115, United States
| | - Weidong Cui
- Amgen Inc, 360 Binney Street, Cambridge, MA, 02141, United States
| | - John Harrahy
- Sanofi, 225 Second Avenue, Waltham, MA 02451, United States
| | - Alexander R Ivanov
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Ave., Boston, MA, 02115, United States.
| |
Collapse
|
12
|
Blöchl C, Gstöttner C, Sénard T, Stork EM, Scherer HU, Toes REM, Wuhrer M, Domínguez-Vega E. A robust nanoscale RP HPLC-MS approach for sensitive Fc proteoform profiling of IgG allotypes. Anal Chim Acta 2023; 1279:341795. [PMID: 37827688 DOI: 10.1016/j.aca.2023.341795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/30/2023] [Accepted: 09/06/2023] [Indexed: 10/14/2023]
Abstract
The conserved region (Fc) of IgG antibodies dictates the interactions with designated receptors thus defining the immunological effector functions of IgG. Amino acid sequence variations in the Fc, recognized as subclasses and allotypes, as well as post-translational modifications (PTMs) modulate these interactions. Yet, the high similarity of Fc sequences hinders allotype-specific PTM analysis by state-of-the-art bottom-up methods and current subunit approaches lack sensitivity and face co-elution of near-isobaric allotypes. To circumvent these shortcomings, we present a nanoscale reversed-phase (RP) HPLC-MS workflow of intact Fc subunits for comprehensive characterization of Fc proteoforms in an allotype- and subclass-specific manner. Polyclonal IgGs were purified from individuals followed by enzymatic digestion releasing single chain Fc subunits (Fc/2) that were directly subjected to analysis. Chromatographic conditions were optimized to separate Fc/2 subunits of near-isobaric allotypes and subclasses allowing allotype and proteoform identification and quantification across all four IgG subclasses. The workflow was complemented by a semi-automated data analysis pipeline based on the open-source software Skyline followed by post-processing in R. The approach revealed pronounced differences in Fc glycosylation between donors, besides inter-subclass and inter-allotype variability within donors. Notably, partial occupancy of the N-glycosylation site in the CH3 domain of IgG3 was observed that is generally neglected by established approaches. The described method was benchmarked across several hundred runs and showed good precision and robustness. This methodology represents a first mature Fc subunit profiling approach allowing truly subclass- and allotype-specific Fc proteoform characterization beyond established approaches. The comprehensive information obtained paired with the high sensitivity provided by the miniaturization of the approach guarantees applicability to a broad range of research questions including clinically relevant (auto)antibody characterization or pharmacokinetics assessment of therapeutic IgGs.
Collapse
Affiliation(s)
- Constantin Blöchl
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Christoph Gstöttner
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Thomas Sénard
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Eva Maria Stork
- Department of Rheumatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Hans Ulrich Scherer
- Department of Rheumatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Rene E M Toes
- Department of Rheumatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Elena Domínguez-Vega
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands.
| |
Collapse
|
13
|
Bou-Jaoudeh M, Mimoun A, Delignat S, Peyron I, Capdevila L, Daventure V, Deligne C, Dimitrov JD, Christophe OD, Denis CV, Lenting PJ, Proulle V, Lacroix-Desmazes S. Imlifidase, a new option to optimize the management of patients with hemophilia A on emicizumab. J Thromb Haemost 2023; 21:2776-2783. [PMID: 37473843 DOI: 10.1016/j.jtha.2023.06.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 06/07/2023] [Accepted: 06/29/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND Emicizumab is a bispecific, chimeric, humanized immunoglobulin G (IgG)4 that mimics the procoagulant activity of factor (F) VIII (FVIII). Its long half-life and subcutaneous route of administration have been life-changing in treating patients with hemophilia A (HA) with or without FVIII inhibitors. However, emicizumab only partially mimics FVIII activity; it prevents but does not treat acute bleeds. Emergency management is particularly complicated in patients with FVIII inhibitors receiving emicizumab prophylaxis in whom exogenous FVIII is inefficient. We have shown recently that Imlifidase (IdeS), a bacterial IgG-degrading enzyme, efficiently eliminates human anti-FVIII IgG in a mouse model of severe HA with inhibitors and opens a therapeutic window for the administration of exogenous FVIII. OBJECTIVES To investigate the impact of IdeS treatment in inhibitor-positive HA mice injected with emicizumab. METHODS IdeS was injected to HA mice reconstituted with human neutralizing anti-FVIII IgG and treated with emicizumab. RESULTS IdeS hydrolyzed emicizumab in vitro and in vivo, albeit, at slower rates than another recombinant human monoclonal IgG4. While F(ab')2 fragments were rapidly cleared from the circulation, thus leading to a rapid loss of emicizumab procoagulant activity, low amounts of single-cleaved intermediate IgG persisted for several days. Moreover, the IdeS-mediated elimination of the neutralizing anti-FVIII IgG and restoration of the hemostatic efficacy of exogenous FVIII were not impaired by the presence of emicizumab and polyclonal human IgG in inhibitor-positive HA mice. CONCLUSION Our results suggest that IdeS could be administered to inhibitor-positive patients with HA receiving emicizumab prophylaxis to improve and ease the management of breakthrough bleeds or programmed major surgeries.
Collapse
Affiliation(s)
- Melissa Bou-Jaoudeh
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, CNRS, Sorbonne Université, Université Paris Cité, Paris, France
| | - Angelina Mimoun
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, CNRS, Sorbonne Université, Université Paris Cité, Paris, France
| | - Sandrine Delignat
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, CNRS, Sorbonne Université, Université Paris Cité, Paris, France
| | - Ivan Peyron
- Laboratory for Hemostasis, Inflammation & Thrombosis, Unité Mixte de Recherche 1176, Institut National de la Santé et de la Recherche Médicale, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Ladislas Capdevila
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, CNRS, Sorbonne Université, Université Paris Cité, Paris, France; Service d'Hématologie Biologique et Unité Fonctionnelle d'Hémostase, Hôpital Cochin, AP-HP Centre, Université Paris Cité, Paris, France
| | - Victoria Daventure
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, CNRS, Sorbonne Université, Université Paris Cité, Paris, France
| | - Claire Deligne
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, CNRS, Sorbonne Université, Université Paris Cité, Paris, France
| | - Jordan D Dimitrov
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, CNRS, Sorbonne Université, Université Paris Cité, Paris, France
| | - Olivier D Christophe
- Laboratory for Hemostasis, Inflammation & Thrombosis, Unité Mixte de Recherche 1176, Institut National de la Santé et de la Recherche Médicale, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Cécile V Denis
- Laboratory for Hemostasis, Inflammation & Thrombosis, Unité Mixte de Recherche 1176, Institut National de la Santé et de la Recherche Médicale, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Peter J Lenting
- Laboratory for Hemostasis, Inflammation & Thrombosis, Unité Mixte de Recherche 1176, Institut National de la Santé et de la Recherche Médicale, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Valérie Proulle
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, CNRS, Sorbonne Université, Université Paris Cité, Paris, France; Service d'Hématologie Biologique et Unité Fonctionnelle d'Hémostase, Hôpital Cochin, AP-HP Centre, Université Paris Cité, Paris, France
| | - Sébastien Lacroix-Desmazes
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, CNRS, Sorbonne Université, Université Paris Cité, Paris, France.
| |
Collapse
|
14
|
Legrand P, Dufaÿ S, Mignet N, Houzé P, Gahoual R. Modeling study of long-term stability of the monoclonal antibody infliximab and biosimilars using liquid-chromatography-tandem mass spectrometry and size-exclusion chromatography-multi-angle light scattering. Anal Bioanal Chem 2023; 415:179-192. [PMID: 36449030 PMCID: PMC9709354 DOI: 10.1007/s00216-022-04396-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 10/07/2022] [Accepted: 10/20/2022] [Indexed: 12/02/2022]
Abstract
Monoclonal antibodies (mAbs) represent a dynamic class of biopharmaceutical products, as evidenced by an increasing number of market authorizations for mAb innovator and biosimilar products. Stability studies are commonly performed during product development, for instance, to exclude unstable molecules, optimize the formulation or determine the storage limit. Such studies are time-consuming, especially for mAbs, because of their structural complexity which requires multiple analytical techniques to achieve a detailed characterization. We report the implementation of a novel methodology based on the accelerated stability assessment program (ASAP) in order to model the long-term stability of mAbs in relation to different structural aspects. Stability studies of innovator infliximab and two different biosimilars were performed using forced degradation conditions alongside in-use administration conditions in order to investigate their similarity regarding stability. Thus, characterization of post-translational modifications was achieved using liquid-chromatography-tandem mass spectrometry (LC-MS/MS) analysis, and the formation of aggregates and free chain fragments was characterized using size-exclusion chromatography-multi-angle light scattering (SEC-MALS-UV/RI) analysis. Consequently, ASAP models were investigated with regard to free chain fragmentation of mAbs concomitantly with N57 deamidation, located in the hypervariable region. Comparison of ASAP models and the long-term stability data from samples stored in intravenous bags demonstrated a relevant correlation, indicating the stability of the mAbs. The developed methodology highlighted the particularities of ASAP modeling for mAbs and demonstrated the possibility to independently consider the different types of degradation pathways in order to provide accurate and appropriate prediction of the long-term stability of this type of biomolecule.
Collapse
Affiliation(s)
- Pauline Legrand
- Université Paris Cité, Faculté de sciences pharmaceutiques et biologiques, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), CNRS UMR8258, Inserm U1267, 4, avenue de l'observatoire, 75270, Paris Cedex 06, France.
- Département Recherche Et Développement Pharmaceutique, Agence Générale Des Equipements Et Produits de Santé (AGEPS), Assistance Publique-Hôpitaux de Paris, (AP-HP), Paris, France.
| | - Sophie Dufaÿ
- Département Recherche Et Développement Pharmaceutique, Agence Générale Des Equipements Et Produits de Santé (AGEPS), Assistance Publique-Hôpitaux de Paris, (AP-HP), Paris, France
| | - Nathalie Mignet
- Université Paris Cité, Faculté de sciences pharmaceutiques et biologiques, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), CNRS UMR8258, Inserm U1267, 4, avenue de l'observatoire, 75270, Paris Cedex 06, France
| | - Pascal Houzé
- Université Paris Cité, Faculté de sciences pharmaceutiques et biologiques, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), CNRS UMR8258, Inserm U1267, 4, avenue de l'observatoire, 75270, Paris Cedex 06, France
- Laboratoire de Toxicologie Biologique, Hôpital Lariboisière, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France
| | - Rabah Gahoual
- Université Paris Cité, Faculté de sciences pharmaceutiques et biologiques, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), CNRS UMR8258, Inserm U1267, 4, avenue de l'observatoire, 75270, Paris Cedex 06, France
| |
Collapse
|
15
|
Shin HG, Yang HR, Yoon A, Lee S. Bispecific Antibody-Based Immune-Cell Engagers and Their Emerging Therapeutic Targets in Cancer Immunotherapy. Int J Mol Sci 2022; 23:5686. [PMID: 35628495 PMCID: PMC9146966 DOI: 10.3390/ijms23105686] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 12/16/2022] Open
Abstract
Cancer is the second leading cause of death worldwide after cardiovascular diseases. Harnessing the power of immune cells is a promising strategy to improve the antitumor effect of cancer immunotherapy. Recent progress in recombinant DNA technology and antibody engineering has ushered in a new era of bispecific antibody (bsAb)-based immune-cell engagers (ICEs), including T- and natural-killer-cell engagers. Since the first approval of blinatumomab by the United States Food and Drug Administration (US FDA), various bsAb-based ICEs have been developed for the effective treatment of patients with cancer. Simultaneously, several potential therapeutic targets of bsAb-based ICEs have been identified in various cancers. Therefore, this review focused on not only highlighting the action mechanism, design and structure, and status of bsAb-based ICEs in clinical development and their approval by the US FDA for human malignancy treatment, but also on summarizing the currently known and emerging therapeutic targets in cancer. This review provides insights into practical considerations for developing next-generation ICEs.
Collapse
Affiliation(s)
- Ha Gyeong Shin
- Department of Biopharmaceutical Chemistry, College of Science and Technology, Kookmin University, Seoul 02707, Korea; (H.G.S.); (H.R.Y.)
| | - Ha Rim Yang
- Department of Biopharmaceutical Chemistry, College of Science and Technology, Kookmin University, Seoul 02707, Korea; (H.G.S.); (H.R.Y.)
| | - Aerin Yoon
- R&D Division, GC Biopharma, Yongin 16924, Korea
| | - Sukmook Lee
- Department of Biopharmaceutical Chemistry, College of Science and Technology, Kookmin University, Seoul 02707, Korea; (H.G.S.); (H.R.Y.)
- Biopharmaceutical Chemistry Major, School of Applied Chemistry, Kookmin University, Seoul 02707, Korea
- Antibody Research Institute, Kookmin University, Seoul 02707, Korea
| |
Collapse
|
16
|
Legrand P, Dembele O, Alamil H, Lamoureux C, Mignet N, Houzé P, Gahoual R. Structural identification and absolute quantification of monoclonal antibodies in suspected counterfeits using capillary electrophoresis and liquid chromatography-tandem mass spectrometry. Anal Bioanal Chem 2022; 414:2699-2712. [PMID: 35099584 PMCID: PMC8802745 DOI: 10.1007/s00216-022-03913-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/09/2022] [Accepted: 01/18/2022] [Indexed: 11/29/2022]
Abstract
Monoclonal antibodies (mAbs) represent a major category of biopharmaceutical products which due to their success as therapeutics have recently experienced the emergence of mAbs originating from different types of trafficking. We report the development of an analytical strategy which enables the structural identification of mAbs in addition to comprehensive characterization and quantification in samples in potentially counterfeit samples. The strategy is based on the concomitant use of capillary zone electrophoresis analysis (CZE-UV), size exclusion chromatography coupled to multi-angle light scattering (SEC-MALS) and liquid chromatography hyphenated to tandem mass spectrometry (LC-MS/MS). This analytical strategy was applied to the investigation of different samples having unknown origins seized by the authorities, and potentially incorporating an IgG 4 or an IgG 1. The results achieved from the different techniques demonstrated to provide orthogonal and complementary information regarding the nature and the structure of the different mAbs. Therefore, they allowed to conclude unequivocally on the identification of the mAbs in the potentially counterfeit samples. Finally, a LC-MS/MS quantification method was developed which specificity was to incorporate a different mAbs labeled with stable isotopes as internal standard. The LC-MS/MS quantification method was validated and thus demonstrated the possibility to use common peptides with the considered IgG in order to achieve limit of quantification as low as 41.4 nM. The quantification method was used to estimate the concentration in the investigated samples using a single type of internal standard and experimental conditions, even in the case of mAbs with no stable isotope labeled homologues available.
Collapse
Affiliation(s)
- Pauline Legrand
- Faculté de Sciences Pharmaceutiques et Biologiques, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), CNRS UMR8258, Inserm U1022, Université de Paris, Paris, France.,Département Recherche Et Développement Pharmaceutique, Agence Générale Des Equipements Et Produits de Santé (AGEPS), Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France
| | - Oumar Dembele
- Faculté de Sciences Pharmaceutiques et Biologiques, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), CNRS UMR8258, Inserm U1022, Université de Paris, Paris, France
| | - Héléna Alamil
- Faculté de Sciences Pharmaceutiques et Biologiques, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), CNRS UMR8258, Inserm U1022, Université de Paris, Paris, France
| | - Catherine Lamoureux
- Service Commun de Laboratoire DGCCRF-DGCCI (SCL), Laboratoire de Paris, Massy, France
| | - Nathalie Mignet
- Faculté de Sciences Pharmaceutiques et Biologiques, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), CNRS UMR8258, Inserm U1022, Université de Paris, Paris, France
| | - Pascal Houzé
- Faculté de Sciences Pharmaceutiques et Biologiques, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), CNRS UMR8258, Inserm U1022, Université de Paris, Paris, France.,Laboratoire de Toxicologie Biologique, Hôpital Lariboisière, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France
| | - Rabah Gahoual
- Faculté de Sciences Pharmaceutiques et Biologiques, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), CNRS UMR8258, Inserm U1022, Université de Paris, Paris, France. .,Unité de Technologies Biologiques Et Chimiques Pour La Santé (UTCBS), Faculté de Pharmacie, Université Paris Descartes, 4, avenue de l'observatoire, 75270, Cedex 06, Paris, France.
| |
Collapse
|
17
|
van Schaick G, el Hajjouti N, Nicolardi S, den Hartog J, Jansen R, van der Hoeven R, Bijleveld W, Abello N, Wuhrer M, Olsthoorn MMA, Domínguez-Vega E. Native Liquid Chromatography and Mass Spectrometry to Structurally and Functionally Characterize Endo-Xylanase Proteoforms. Int J Mol Sci 2022; 23:1307. [PMID: 35163230 PMCID: PMC8835838 DOI: 10.3390/ijms23031307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 12/16/2022] Open
Abstract
Xylanases are of great value in various industries, including paper, food, and biorefinery. Due to their biotechnological production, these enzymes can contain a variety of post-translational modifications, which may have a profound effect on protein function. Understanding the structure-function relationship can guide the development of products with optimal performance. We have developed a workflow for the structural and functional characterization of an endo-1,4-β-xylanase (ENDO-I) produced by Aspergillus niger with and without applying thermal stress. This workflow relies on orthogonal native separation techniques to resolve proteoforms. Mass spectrometry and activity assays of separated proteoforms permitted the establishment of structure-function relationships. The separation conditions were focus on balancing efficient separation and protein functionality. We employed size exclusion chromatography (SEC) to separate ENDO-I from other co-expressed proteins. Charge variants were investigated with ion exchange chromatography (IEX) and revealed the presence of low abundant glycated variants in the temperature-stressed material. To obtain better insights into the effect on glycation on function, we enriched for these species using boronate affinity chromatography (BAC). The activity measurements showed lower activity of glycated species compared to the non-modified enzyme. Altogether, this workflow allowed in-depth structural and functional characterization of ENDO-I proteoforms.
Collapse
Affiliation(s)
- Guusje van Schaick
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (N.e.H.); (S.N.); (M.W.); (E.D.-V.)
| | - Nadi el Hajjouti
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (N.e.H.); (S.N.); (M.W.); (E.D.-V.)
| | - Simone Nicolardi
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (N.e.H.); (S.N.); (M.W.); (E.D.-V.)
| | - Joost den Hartog
- Center for Analytical Innovation, DSM, Alexander Fleminglaan 1, 2613 AX Delft, The Netherlands; (J.d.H.); (R.J.); (R.v.d.H.); (W.B.); (N.A.); (M.M.A.O.)
| | - Romana Jansen
- Center for Analytical Innovation, DSM, Alexander Fleminglaan 1, 2613 AX Delft, The Netherlands; (J.d.H.); (R.J.); (R.v.d.H.); (W.B.); (N.A.); (M.M.A.O.)
| | - Rob van der Hoeven
- Center for Analytical Innovation, DSM, Alexander Fleminglaan 1, 2613 AX Delft, The Netherlands; (J.d.H.); (R.J.); (R.v.d.H.); (W.B.); (N.A.); (M.M.A.O.)
| | - Wim Bijleveld
- Center for Analytical Innovation, DSM, Alexander Fleminglaan 1, 2613 AX Delft, The Netherlands; (J.d.H.); (R.J.); (R.v.d.H.); (W.B.); (N.A.); (M.M.A.O.)
| | - Nicolas Abello
- Center for Analytical Innovation, DSM, Alexander Fleminglaan 1, 2613 AX Delft, The Netherlands; (J.d.H.); (R.J.); (R.v.d.H.); (W.B.); (N.A.); (M.M.A.O.)
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (N.e.H.); (S.N.); (M.W.); (E.D.-V.)
| | - Maurien M. A. Olsthoorn
- Center for Analytical Innovation, DSM, Alexander Fleminglaan 1, 2613 AX Delft, The Netherlands; (J.d.H.); (R.J.); (R.v.d.H.); (W.B.); (N.A.); (M.M.A.O.)
| | - Elena Domínguez-Vega
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (N.e.H.); (S.N.); (M.W.); (E.D.-V.)
| |
Collapse
|
18
|
Naumann L, Schlossbauer P, Klingler F, Hesse F, Otte K, Neusüß C. High throughput glycosylation analysis of intact monoclonal antibodies by mass spectrometry coupled with capillary electrophoresis and liquid chromatography. J Sep Sci 2022; 45:2034-2044. [PMID: 35044720 DOI: 10.1002/jssc.202100865] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/05/2022] [Accepted: 01/08/2022] [Indexed: 11/07/2022]
Abstract
The analysis of monoclonal antibodies glycosylation is a crucial quality control attribute of biopharmaceutical drugs. High throughput screening approaches for antibody glycoform analysis are required in various stages of process optimization. Here, we present high throughput screening suitable mass spectrometry-based workflows for the analysis of intact antibody glycosylation out of cell supernatants. Capillary electrophoresis and liquid chromatography were coupled with quadrupole time-of-flight MS or Orbitrap MS. Both separation methods offer fast separation (10-15 min) and the capability to prevent the separated cell supernatant matrix to enter the MS by post-separation valving. Both MS instruments provide comparable results and both are sufficient to determine the glycosylation pattern of the five major glycoforms of the measured antibodies. However, the Orbitrap yields higher sensitivity of 25 μg/mL (CE-nanoCEasy-Orbitrap MS) and 5 μg/mL (LC-Orbitrap MS). Data processing was optimized for a faster processing and easier detection of low abundant glycoforms based on averaged charge-deconvoluted mass spectra. This approach combines a non-target glycoform analysis, while yielding the same glycosylation pattern as the traditional approach based on extracted ion traces. The presented methods enable the high throughput screening of the glycosylation pattern of antibodies down to low μg/mL-range out of cell supernatant without any sample preparation. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Lukas Naumann
- Department of Chemistry, Aalen University, Beethovenstraße 1, Aalen, 73430, Germany
| | - Patrick Schlossbauer
- Department of applied Biotechnology, Biberach University of Applied Sciences, Karlstraße 6-11, Biberach, 88400, Germany
| | - Florian Klingler
- Department of applied Biotechnology, Biberach University of Applied Sciences, Karlstraße 6-11, Biberach, 88400, Germany
| | - Friedemann Hesse
- Department of applied Biotechnology, Biberach University of Applied Sciences, Karlstraße 6-11, Biberach, 88400, Germany
| | - Kerstin Otte
- Department of applied Biotechnology, Biberach University of Applied Sciences, Karlstraße 6-11, Biberach, 88400, Germany
| | - Christian Neusüß
- Department of Chemistry, Aalen University, Beethovenstraße 1, Aalen, 73430, Germany
| |
Collapse
|