1
|
Li Y, Li W, Zheng Y, Wang T, Pu R, Zhang Z. Desalting strategies for native mass spectrometry. Talanta 2025; 281:126824. [PMID: 39250868 DOI: 10.1016/j.talanta.2024.126824] [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: 09/02/2024] [Accepted: 09/05/2024] [Indexed: 09/11/2024]
Abstract
In native mass spectrometry (MS) salts are indispensable for preserving the native structures of biomolecules, but detrimental to mass sensitivity, resolution, and accuracy. Such a conflict makes desalting in native MS more challenging, distinctive, and sample-dependent than in peptide-centric MS. This review first briefly introduces the charged residue mechanism whereby native-like gaseous protein ions are released from electrospray droplets, revealing a higher degree of salt adduction than denatured proteins. Subsequently, this review summarizes and explores the existing strategies, underlying mechanisms and future perspectives of desalting in native MS. These strategies mainly focus on buffer exchange into volatile salts (offline and online approaches), addition of solution additives (e.g., anion, supercharging reagent, solution phase chelator and amino acid), use of submicron electrospray emitters (down to 60 nm), and other potential approaches (e.g., induced and electrophoretic nanoelectrospray ionization). The strategies of online buffer exchange and using nanoscale electrospray emitters are highlighted. This review would not only be a valuable addition to the field of sample preparation in MS, but would also serve as a beginner's guide to desalting in native MS.
Collapse
Affiliation(s)
- Yun Li
- School of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, China
| | - Weijie Li
- School of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, China
| | - Yajun Zheng
- School of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, China.
| | - Tong Wang
- School of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, China
| | - Ruijin Pu
- School of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, China
| | - Zhiping Zhang
- School of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, China.
| |
Collapse
|
2
|
Szabo R, Gyemant G, Nagy C, Andrasi M, Gaspar A. Taylor-Aris Dispersion-Assisted Mass Spectrometry for the Analysis of Native Proteins. Anal Chem 2024; 96:11309-11317. [PMID: 38946421 DOI: 10.1021/acs.analchem.4c01270] [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: 07/02/2024]
Abstract
As has recently been shown, Taylor-Aris dispersion-assisted mass spectrometry (TADA-MS) can offer direct injection MS determinations in fields where the targets of the analyses are large molecules present in a matrix that would otherwise cause serious interferences. In the present study, we demonstrated the exceptional utility of TADA-MS in native protein analysis: (i) a dramatic improvement in detection sensitivity was found due to its ability to strongly reduce matrix interferences, (ii) more "native-like" conditions can be used during analyses, (iii) the direct injection of non-MS-compatible matrices is allowed into MS, and (iv) a considerable simplification and economization of the workflow is ensured. We investigated the behavior of different types of proteins and protein complexes present under native conditions, demonstrating the unambiguous benefits and simplicity of the method.
Collapse
Affiliation(s)
- Ruben Szabo
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Debrecen 4032, Hungary
| | - Gyongyi Gyemant
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Debrecen 4032, Hungary
| | - Cynthia Nagy
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Debrecen 4032, Hungary
| | - Melinda Andrasi
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Debrecen 4032, Hungary
| | - Attila Gaspar
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Debrecen 4032, Hungary
| |
Collapse
|
3
|
Wu G, Yu C, Yin S, Du J, Zhang Y, Fu Z, Wang L, Wang J. A native SEC-MS workflow and validation for analyzing drug-to-antibody ratio and drug load distribution in cysteine-linked antibody-drug conjugates. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1241:124167. [PMID: 38823148 DOI: 10.1016/j.jchromb.2024.124167] [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: 02/06/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/03/2024]
Abstract
The development and optimization of Antibody-Drug Conjugates (ADCs) hinge on enhanced analytical and bioanalytical characterization, particularly in assessing critical quality attributes (CQAs). The ADC's potency is largely determined by the average number of drugs attached to the monoclonal antibody (mAb), known as the drug-to-antibody ratio (DAR). Furthermore, the drug load distribution (DLD) influences the therapeutic window of the ADC, defining the range of dosages effective in treating diseases without causing toxic effects. Among CQAs, DAR and DLD are vital; their control is essential for ensuring manufacturing consistency and product quality. Typically, hydrophobic interaction chromatography (HIC) or reversed-phase liquid chromatography (RPLC) with UV detector have been used to quantitate DAR and DLD in quality control (QC) environment. Recently, Native size-exclusion chromatography-mass spectrometry (nSEC-MS) proves the potential as a platformable quantitative method for characterizing DAR and DLD across various cysteine-linked ADCs in research or early preclinical development. In this work, we established and assessed a streamlined nSEC-MS workflow with a benchtop LC-MS platform, to quantitatively monitor DAR and DLD of different chemotype and drug load level cysteine-linked ADCs. Moreover, to deploy this workflow in QC environment, complete method validation was conducted in three independent laboratories, adhering to the International Council for Harmonization of Technical Requirements for Pharmaceuticals for Human Use (ICH) Q2(R1) guidelines. The results met the predefined analytical target profile (ATP) and performance criteria, encompassing specificity/selectivity, accuracy, precision, linearity, range, quantification/detection limit, and robustness. Finally, the method validation design offers a reference for other nSEC-MS methods that are potentially used to determine the DAR and DLD on cysteine-linker ADCs. To the best of our knowledge, this study is the first reported systematic validation of the nSEC-MS method for detecting DAR and DLD. The results indicated that the co-validated nSEC-MS workflow is suitable for DAR and DLD routine analysis in ADC quality control, release, and stability testing.
Collapse
Affiliation(s)
- Gang Wu
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 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, China
| | - Sicheng Yin
- Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai, 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, China
| | - Yifan Zhang
- Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai, 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, 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, China
| | - Junzhi Wang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China.
| |
Collapse
|
4
|
Zhai Z, Schoenmakers PJ, Gargano AFG. Identification of heavily glycated proteoforms by hydrophilic-interaction liquid chromatography and native size-exclusion chromatography - High-resolution mass spectrometry. Anal Chim Acta 2024; 1304:342543. [PMID: 38637052 DOI: 10.1016/j.aca.2024.342543] [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: 12/25/2023] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND The non-enzymatic glycation of proteins and their advanced glycation end products (AGEs) are associated with protein transformations such as in the development of diseases and biopharmaceutical storage. The characterization of heavily glycated proteins at the intact level is of high interest as it allows to describe co-occurring protein modifications. However, the high heterogeneity of glycated protein makes this process challenging, and novel methods are required to accomplish this. RESULTS In this study, we investigated two novel LC-HRMS methods to study glycated reference proteins at the intact protein level: low-flow hydrophilic-interaction liquid chromatography (HILIC) and native size-exclusion chromatography (SEC). Model proteins were exposed to conditions that favored extensive glycation and the formation of AGEs. After glycation, complicated MS spectra were observed, along with a sharply reduced signal response, possibly due to protein denaturation and the formation of aggregates. When using HILIC-MS, the glycated forms of the proteins could be resolved based on the number of reducing monosaccharides. Moreover, some positional glycated isomers were separated. The SEC-MS method under non-denaturing conditions provided insights into glycated aggregates but offered only a limited separation of glycated species based on molar mass. Overall, more than 25 different types of species were observed in both methods, differing in molar mass by 14-162 Da. 19 of these species have not been previously reported. SIGNIFICANCE The proposed strategies show great potential to characterize highly glycated intact proteins from native and denaturing perspectives and provide new opportunities for fast clinical diagnoses and investigating glycation-related diseases.
Collapse
Affiliation(s)
- Ziran Zhai
- Van't Hoff Institute for Molecular Sciences, University of Amsterdam, Amsterdam, the Netherlands; Centre for Analytical Sciences Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands.
| | - Peter J Schoenmakers
- Van't Hoff Institute for Molecular Sciences, University of Amsterdam, Amsterdam, the Netherlands; Centre for Analytical Sciences Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands
| | - Andrea F G Gargano
- Van't Hoff Institute for Molecular Sciences, University of Amsterdam, Amsterdam, the Netherlands; Centre for Analytical Sciences Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands.
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Makey DM, Ruotolo BT. Liquid-phase separations coupled with ion mobility-mass spectrometry for next-generation biopharmaceutical analysis. Expert Rev Proteomics 2024; 21:259-270. [PMID: 38934922 PMCID: PMC11299228 DOI: 10.1080/14789450.2024.2373707] [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: 03/28/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024]
Abstract
INTRODUCTION The pharmaceutical industry continues to expand its search for innovative biotherapeutics. The comprehensive characterization of such therapeutics requires many analytical techniques to fully evaluate critical quality attributes, making analysis a bottleneck in discovery and development timelines. While thorough characterization is crucial for ensuring the safety and efficacy of biotherapeutics, there is a need to further streamline analytical characterization and expedite the overall timeline from discovery to market. AREAS COVERED This review focuses on recent developments in liquid-phase separations coupled with ion mobility-mass spectrometry (IM-MS) for the development and characterization of biotherapeutics. We cover uses of IM-MS to improve the characterization of monoclonal antibodies, antibody-drug conjugates, host cell proteins, glycans, and nucleic acids. This discussion is based on an extensive literature search using Web of Science, Google Scholar, and SciFinder. EXPERT OPINION IM-MS has the potential to enhance the depth and efficiency of biotherapeutic characterization by providing additional insights into conformational changes, post-translational modifications, and impurity profiles. The rapid timescale of IM-MS positions it well to enhance the information content of existing assays through its facile integration with standard liquid-phase separation techniques that are commonly used for biopharmaceutical analysis.
Collapse
Affiliation(s)
- Devin M Makey
- Department of Chemistry, University of Michigan, Ann Arbor, MI, USA
| | | |
Collapse
|
8
|
Khalikova M, Jireš J, Horáček O, Douša M, Kučera R, Nováková L. What is the role of current mass spectrometry in pharmaceutical analysis? MASS SPECTROMETRY REVIEWS 2024; 43:560-609. [PMID: 37503656 DOI: 10.1002/mas.21858] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 06/02/2023] [Accepted: 06/25/2023] [Indexed: 07/29/2023]
Abstract
The role of mass spectrometry (MS) has become more important in most application domains in recent years. Pharmaceutical analysis is specific due to its stringent regulation procedures, the need for good laboratory/manufacturing practices, and a large number of routine quality control analyses to be carried out. The role of MS is, therefore, very different throughout the whole drug development cycle. While it dominates within the drug discovery and development phase, in routine quality control, the role of MS is minor and indispensable only for selected applications. Moreover, its role is very different in the case of analysis of small molecule pharmaceuticals and biopharmaceuticals. Our review explains the role of current MS in the analysis of both small-molecule chemical drugs and biopharmaceuticals. Important features of MS-based technologies being implemented, method requirements, and related challenges are discussed. The differences in analytical procedures for small molecule pharmaceuticals and biopharmaceuticals are pointed out. While a single method or a small set of methods is usually sufficient for quality control in the case of small molecule pharmaceuticals and MS is often not indispensable, a large panel of methods including extensive use of MS must be used for quality control of biopharmaceuticals. Finally, expected development and future trends are outlined.
Collapse
Affiliation(s)
- Maria Khalikova
- Department of Analytical Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czech Republic
| | - Jakub Jireš
- Department of Analytical Chemistry, Faculty of Chemical Engineering, UCT Prague, Prague, Czech Republic
- Department of Development, Zentiva, k. s., Praha, Praha, Czech Republic
| | - Ondřej Horáček
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis, Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Michal Douša
- Department of Development, Zentiva, k. s., Praha, Praha, Czech Republic
| | - Radim Kučera
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis, Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Lucie Nováková
- Department of Analytical Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| |
Collapse
|
9
|
Poskute R, Sankaran PK, Sewell L, Lepore G, Shrubsall R, Dewis L, Watanabe Y, Wong V, Pascual Fernandez L, Mishra R, Holt A, Sou S, Harris C, Moreno Rodriguez C, Cankorur-Cetinkaya A, Smith J, Lonska N, Powell A, Cui T, Cheeks M, Lindo V. Identification and quantification of chain-pairing variants or mispaired species of asymmetric monovalent bispecific IgG1 monoclonal antibody format using reverse-phase polyphenyl chromatography coupled electrospray ionization mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1237:124085. [PMID: 38513430 DOI: 10.1016/j.jchromb.2024.124085] [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: 01/10/2024] [Revised: 03/08/2024] [Accepted: 03/09/2024] [Indexed: 03/23/2024]
Abstract
Developing a knob-into-hole asymmetric bispecific IgG1 monoclonal antibody (mAb) poses manufacturing challenges due to the expression of chain pairing variants, also called mispaired species, in the desired product. The incorrect pairing of light and heavy chains could result in heterogeneous mispaired species of homodimers, heterodimers, light chain swapping, and low molecular weight species (LMWS). Standard chromatography, capillary electrophoretic, or spectroscopic methods poorly resolve these from the main variants. Here, we report a highly sensitive reverse-phase polyphenyl ultra-high-performance liquid chromatography (RP-UHPLC) method to accurately measure mispaired species of Duet mAb format, an asymmetric IgG1 bispecific mAb, for both process development and quality control analytical tests. Coupled with electrospray ionization mass spectrometry (ESI-MS), it enabled direct online characterization of mispaired species. This single direct assay detected diverse mispaired IgG-like species and LMWS. The method resolved eight disulfide bonds dissociated LMWS and three mispaired LMWS. It also resolved three different types of IgG-like mispaired species, including two homodimers and one heterodimer. The characterization and quantification simultaneously enabled the cell line selection that produces a lesser heterogeneity and lower levels of mispaired species with the desired correctly paired product. The biological activity assessment of samples with increased levels of these species quantified by the method exhibited a linear decline in potency with increasing levels of mispaired species in the desired product. We also demonstrated the utility of the technique for testing in-process intermediate materials to determine and assess downstream purification process capability in removing diverse mispaired IgG-like species and LMWS to a certain level during the downstream purification process. Our investigation demonstrates that adopting this method was vital in developing asymmetric bispecific mAb from the initial stage of cell line development to manufacturing process development. Therefore, this tool could be used in the control strategy to monitor and control mispaired species during manufacturing, thus improving the quality control of the final product.
Collapse
Affiliation(s)
- Ryte Poskute
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | | | - Laura Sewell
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Giordana Lepore
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Rebecca Shrubsall
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Lydia Dewis
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Yasunori Watanabe
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Vanessa Wong
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | | | - Rahul Mishra
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Alexander Holt
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Susie Sou
- Cell Culture & Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Claire Harris
- Cell Culture & Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Cristina Moreno Rodriguez
- Cell Culture & Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Ayca Cankorur-Cetinkaya
- Cell Culture & Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Jennifer Smith
- Cell Culture & Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Nikola Lonska
- Purification Process Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Adam Powell
- Purification Process Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Tingting Cui
- Purification Process Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Matthew Cheeks
- Cell Culture & Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Viv Lindo
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK.
| |
Collapse
|
10
|
Dhenin J, Lafont V, Dupré M, Krick A, Mauriac C, Chamot-Rooke J. Monitoring mAb proteoforms in mouse plasma using an automated immunocapture combined with top-down and middle-down mass spectrometry. Proteomics 2024; 24:e2300069. [PMID: 37480175 DOI: 10.1002/pmic.202300069] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/27/2023] [Accepted: 07/10/2023] [Indexed: 07/23/2023]
Abstract
Monoclonal antibodies (mAbs) have established themselves as the leading biopharmaceutical therapeutic modality. Once the developability of a mAb drug candidate has been assessed, an important step is to check its in vivo stability through pharmacokinetics (PK) studies. The gold standard is ligand-binding assay (LBA) and liquid chromatography-mass spectrometry (LC-MS) performed at the peptide level (bottom-up approach). However, these analytical techniques do not allow to address the different mAb proteoforms that can arise from biotransformation. In recent years, top-down and middle-down mass spectrometry approaches have gained popularity to characterize proteins at the proteoform level but are not yet widely used for PK studies. We propose here a workflow based on an automated immunocapture followed by top-down and middle-down liquid chromatography-tandem mass spectrometry (LC-MS/MS) approaches to characterize mAb proteoforms spiked in mouse plasma. We demonstrate the applicability of our workflow on a large concentration range using pembrolizumab as a model. We also compare the performance of two state-of-the-art Orbitrap platforms (Tribrid Eclipse and Exploris 480) for these studies. The added value of our workflow for an accurate and sensitive characterization of mAb proteoforms in mouse plasma is highlighted.
Collapse
Affiliation(s)
- Jonathan Dhenin
- Institut Pasteur, Université Paris Cité, CNRS UAR2024, Mass Spectrometry for Biology, Paris, France
- Université Paris Cité, Sorbonne Paris Cité, Paris, France
- DMPK, Sanofi R&D, Chilly-Mazarin, France
| | | | | | | | | | - Julia Chamot-Rooke
- Institut Pasteur, Université Paris Cité, CNRS UAR2024, Mass Spectrometry for Biology, Paris, France
| |
Collapse
|
11
|
Coliat P, Erb S, Diemer H, Karouby D, Martin T, Banerjee M, Zhu C, Demarchi M, Cianférani S, Detappe A, Pivot X. Influence of pneumatic transportation on the stability of monoclonal antibodies. Sci Rep 2023; 13:21875. [PMID: 38072852 PMCID: PMC10710995 DOI: 10.1038/s41598-023-49235-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 12/06/2023] [Indexed: 12/18/2023] Open
Abstract
Pneumatic transportation systems (PTS) were recently proposed as a method to carry ready-for-injection diluted monoclonal antibodies (mAbs) from the pharmacy to the bedside of patients. This method reduces transportation time and improves the efficiency of drug distribution process. However, mAbs are highly sensitive molecules for which subtle alterations may lead to deleterious clinical effects. These alterations can be caused by various external factors such as temperature, pH, pressure, and mechanical forces that may occur during transportation. Hence, it is essential to ensure that the mAbs transported by PTS remain stable and active throughout the transportation process. This study aims to determine the safety profile of PTS to transport 11 routinely used mAbs in a clinical setting through assessment of critical quality attributes (CQA) and orthogonal analysis. Hence, we performed aggregation/degradation profiling, post-translational modifications identification using complementary mass spectrometry-based methods, along with visible and subvisible particle formation determination by light absorbance and light obscuration analysis. Altogether, these results highlight that PTS can be safely used for this purpose when air is removed from the bags during preparation.
Collapse
Affiliation(s)
- Pierre Coliat
- Institut de Cancérologie Strasbourg Europe, ICANS, 17 Rue Albert Calmette, Strasbourg, France.
| | - Stéphane Erb
- Institut Pluridisciplinaire Hubert Curien, CNRS UMR7178, Université de Strasbourg, Strasbourg, France
- Institut du Médicament Strasbourg, Strasbourg, France
- Infrastructure Nationale de Protéomique ProFI-FR2048, Strasbourg, France
| | - Hélène Diemer
- Institut Pluridisciplinaire Hubert Curien, CNRS UMR7178, Université de Strasbourg, Strasbourg, France
- Institut du Médicament Strasbourg, Strasbourg, France
- Infrastructure Nationale de Protéomique ProFI-FR2048, Strasbourg, France
| | - Dan Karouby
- Institut de Cancérologie Strasbourg Europe, ICANS, 17 Rue Albert Calmette, Strasbourg, France
| | - Tristan Martin
- Institut de Cancérologie Strasbourg Europe, ICANS, 17 Rue Albert Calmette, Strasbourg, France
| | - Mainak Banerjee
- Institut de Cancérologie Strasbourg Europe, ICANS, 17 Rue Albert Calmette, Strasbourg, France
| | - Chen Zhu
- Institut de Cancérologie Strasbourg Europe, ICANS, 17 Rue Albert Calmette, Strasbourg, France
| | - Martin Demarchi
- Institut de Cancérologie Strasbourg Europe, ICANS, 17 Rue Albert Calmette, Strasbourg, France
| | - Sarah Cianférani
- Institut Pluridisciplinaire Hubert Curien, CNRS UMR7178, Université de Strasbourg, Strasbourg, France
- Institut du Médicament Strasbourg, Strasbourg, France
- Infrastructure Nationale de Protéomique ProFI-FR2048, Strasbourg, France
| | - Alexandre Detappe
- Institut de Cancérologie Strasbourg Europe, ICANS, 17 Rue Albert Calmette, Strasbourg, France
- Institut Pluridisciplinaire Hubert Curien, CNRS UMR7178, Université de Strasbourg, Strasbourg, France
- Institut du Médicament Strasbourg, Strasbourg, France
| | - Xavier Pivot
- Institut de Cancérologie Strasbourg Europe, ICANS, 17 Rue Albert Calmette, Strasbourg, France
| |
Collapse
|
12
|
Castel J, Delaux S, Hernandez-Alba O, Cianférani S. Recent advances in structural mass spectrometry methods in the context of biosimilarity assessment: from sequence heterogeneities to higher order structures. J Pharm Biomed Anal 2023; 236:115696. [PMID: 37713983 DOI: 10.1016/j.jpba.2023.115696] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/17/2023]
Abstract
Biotherapeutics and their biosimilar versions have been flourishing in the biopharmaceutical market for several years. Structural and functional characterization is needed to achieve analytical biosimilarity through the assessment of critical quality attributes as required by regulatory authorities. The role of analytical strategies, particularly mass spectrometry-based methods, is pivotal to gathering valuable information for the in-depth characterization of biotherapeutics and biosimilarity assessment. Structural mass spectrometry methods (native MS, HDX-MS, top-down MS, etc.) provide information ranging from primary sequence assessment to higher order structure evaluation. This review focuses on recent developments and applications in structural mass spectrometry for biotherapeutic and biosimilar characterization.
Collapse
Affiliation(s)
- Jérôme Castel
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC UMR 7178, Université de Strasbourg, CNRS, Strasbourg 67087, France; Infrastructure Nationale de Protéomique ProFI, FR2048 CNRS CEA, Strasbourg 67087, France
| | - Sarah Delaux
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC UMR 7178, Université de Strasbourg, CNRS, Strasbourg 67087, France; Infrastructure Nationale de Protéomique ProFI, FR2048 CNRS CEA, Strasbourg 67087, France
| | - Oscar Hernandez-Alba
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC UMR 7178, Université de Strasbourg, CNRS, Strasbourg 67087, France; Infrastructure Nationale de Protéomique ProFI, FR2048 CNRS CEA, Strasbourg 67087, France
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC UMR 7178, Université de Strasbourg, CNRS, Strasbourg 67087, France; Infrastructure Nationale de Protéomique ProFI, FR2048 CNRS CEA, Strasbourg 67087, France.
| |
Collapse
|
13
|
Ventouri IK, Veelders S, Passamonti M, Endres P, Roemling R, Schoenmakers PJ, Somsen GW, Haselberg R, Gargano AFG. Micro-flow size-exclusion chromatography for enhanced native mass spectrometry of proteins and protein complexes. Anal Chim Acta 2023; 1266:341324. [PMID: 37244657 DOI: 10.1016/j.aca.2023.341324] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/29/2023] [Accepted: 05/03/2023] [Indexed: 05/29/2023]
Abstract
Size-exclusion chromatography (SEC) employing aqueous mobile phases with volatile salts at neutral pH combined with native mass spectrometry (nMS) is a valuable tool to characterize proteins and protein aggregates in their native state. However, the liquid-phase conditions (high salt concentrations) frequently used in SEC-nMS hinder the analysis of labile protein complexes in the gas phase, necessitating higher desolvation-gas flow and source temperature, leading to protein fragmentation/dissociation. To overcome this issue, we investigated narrow SEC columns (1.0 mm internal diameter, I.D.) operated at 15-μL/min flow rates and their coupling to nMS for the characterization of proteins, protein complexes and higher-order structures (HOS). The reduced flow rate resulted in a significant increase in the protein-ionization efficiency, facilitating the detection of low-abundant impurities and HOS up to 230 kDa (i.e., the upper limit of the Orbitrap-MS instrument used). More-efficient solvent evaporation and lower desolvation energies allowed for softer ionization conditions (e.g., lower gas temperatures), ensuring little or no structural alterations of proteins and their HOS during transfer into the gas phase. Furthermore, ionization suppression by eluent salts was decreased, permitting the use of volatile-salt concentrations up to 400 mM. Band broadening and loss of resolution resulting from the introduction of injection volumes exceeding 3% of the column volume could be circumvented by incorporating an online trap-column containing a mixed-bed ion-exchange (IEX) material. The online IEX-based solid-phase extraction (SPE) or "trap-and-elute" set-up provided on-column focusing (sample preconcentration). This allowed the injection of large sample volumes on the 1-mm I.D. SEC column without compromising the separation. The enhanced sensitivity attained by the micro-flow SEC-MS, along with the on-column focusing achieved by the IEX precolumn, provided picogram detection limits for proteins.
Collapse
Affiliation(s)
- Iro K Ventouri
- Analytical Chemistry group, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands; Centre for Analytical Sciences Amsterdam, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands.
| | - Sharene Veelders
- Analytical Chemistry group, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands; Centre for Analytical Sciences Amsterdam, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands
| | - Marta Passamonti
- Analytical Chemistry group, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands; Centre for Analytical Sciences Amsterdam, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands
| | - Patrick Endres
- Tosoh Bioscience GmbH, Im Leuschnerpark 4, 64347, Griesheim, Germany
| | - Regina Roemling
- Tosoh Bioscience GmbH, Im Leuschnerpark 4, 64347, Griesheim, Germany
| | - Peter J Schoenmakers
- Analytical Chemistry group, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands; Centre for Analytical Sciences Amsterdam, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands
| | - Govert W Somsen
- Analytical Chemistry group, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands; Division of Bioanalytical Chemistry, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081, HV Amsterdam, the Netherlands
| | - Rob Haselberg
- Analytical Chemistry group, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands; Division of Bioanalytical Chemistry, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081, HV Amsterdam, the Netherlands
| | - Andrea F G Gargano
- Analytical Chemistry group, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands; Centre for Analytical Sciences Amsterdam, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands.
| |
Collapse
|
14
|
Cotham VC, Liu AP, Wang S, Li N. A generic platform to couple affinity chromatography with native mass spectrometry for the analysis of therapeutic monoclonal antibodies. J Pharm Biomed Anal 2023; 228:115337. [PMID: 36933319 DOI: 10.1016/j.jpba.2023.115337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/06/2023] [Accepted: 03/10/2023] [Indexed: 03/16/2023]
Abstract
Affinity chromatography coupled with native mass spectrometry has emerged as a powerful tool for the analysis of therapeutic monoclonal antibodies (mAbs). Exploiting the specific interactions between mAbs and their ligands, these methods not only provide orthogonal means to study the highly complex mAb attributes, but also offer insights on their biological relevance. Despite the great promise, application of affinity chromatography - native mass spectrometry in routine mAb characterization has been limited, largely due to the complicated experimental set up. In this study, we introduced a generic platform to facilitate the online coupling of different affinity separation modes with native mass spectrometry. Built upon a recently introduced native LC-MS platform, this new strategy can accommodate a wide range of chromatographic conditions, and therefore, allow greatly simplified experimental set up and facile swapping of affinity separation modes. The utility of this platform was demonstrated by successful online coupling of three affinity chromatography methods (protein A, FcγRIIIa, and FcRn) with native mass spectrometry. The developed protein A-MS method was tested both in a "bind-and-elute" mode for rapid mAb screening and in a high-resolution resolving mode to study mAb species with altered protein A affinity. The FcγRIIIa-MS method was applied to achieve glycoform-resolved analyses of both IgG1 and IgG4 subclass molecules. The FcRn-MS method was demonstrated in two case studies, where specific post-translational modifications and Fc mutations were known to alter FcRn affinities.
Collapse
Affiliation(s)
- Victoria C Cotham
- Analytical Chemistry Group, Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | - Anita P Liu
- Analytical Chemistry Group, Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | - Shunhai Wang
- Analytical Chemistry Group, Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA.
| | - Ning Li
- Analytical Chemistry Group, Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| |
Collapse
|
15
|
Nickerson JL, Baghalabadi V, Rajendran SRCK, Jakubec PJ, Said H, McMillen TS, Dang Z, Doucette AA. Recent advances in top-down proteome sample processing ahead of MS analysis. MASS SPECTROMETRY REVIEWS 2023; 42:457-495. [PMID: 34047392 DOI: 10.1002/mas.21706] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/21/2021] [Accepted: 05/06/2021] [Indexed: 06/12/2023]
Abstract
Top-down proteomics is emerging as a preferred approach to investigate biological systems, with objectives ranging from the detailed assessment of a single protein therapeutic, to the complete characterization of every possible protein including their modifications, which define the human proteoform. Given the controlling influence of protein modifications on their biological function, understanding how gene products manifest or respond to disease is most precisely achieved by characterization at the intact protein level. Top-down mass spectrometry (MS) analysis of proteins entails unique challenges associated with processing whole proteins while maintaining their integrity throughout the processes of extraction, enrichment, purification, and fractionation. Recent advances in each of these critical front-end preparation processes, including minimalistic workflows, have greatly expanded the capacity of MS for top-down proteome analysis. Acknowledging the many contributions in MS technology and sample processing, the present review aims to highlight the diverse strategies that have forged a pathway for top-down proteomics. We comprehensively discuss the evolution of front-end workflows that today facilitate optimal characterization of proteoform-driven biology, including a brief description of the clinical applications that have motivated these impactful contributions.
Collapse
Affiliation(s)
| | - Venus Baghalabadi
- Department of Chemistry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Subin R C K Rajendran
- Department of Chemistry, Dalhousie University, Halifax, Nova Scotia, Canada
- Verschuren Centre for Sustainability in Energy and the Environment, Sydney, Nova Scotia, Canada
| | - Philip J Jakubec
- Department of Chemistry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Hammam Said
- Department of Chemistry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Teresa S McMillen
- Department of Chemistry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Ziheng Dang
- Department of Chemistry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Alan A Doucette
- Department of Chemistry, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
16
|
Deslignière E, Ollivier S, Beck A, Ropartz D, Rogniaux H, Cianférani S. Benefits and Limitations of High-Resolution Cyclic IM-MS for Conformational Characterization of Native Therapeutic Monoclonal Antibodies. Anal Chem 2023; 95:4162-4171. [PMID: 36780376 DOI: 10.1021/acs.analchem.2c05265] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Monoclonal antibodies (mAbs) currently represent the main class of therapeutic proteins. mAbs approved by regulatory agencies are selected from IgG1, IgG2, and IgG4 subclasses, which possess different interchain disulfide connectivities. Ion mobility coupled to native mass spectrometry (IM-MS) has emerged as a valuable approach to tackle the challenging characterization of mAbs' higher order structures. However, due to the limited resolution of first-generation IM-MS instruments, subtle conformational differences on large proteins have long been hard to capture. Recent technological developments have aimed at increasing available IM resolving powers and acquisition mode capabilities, namely, through the release of high-resolution IM-MS (HR-IM-MS) instruments, like cyclic IM-MS (cIM-MS). Here, we outline the advantages and drawbacks of cIM-MS for better conformational characterization of intact mAbs (∼150 kDa) in native conditions compared to first-generation instruments. We first assessed the extent to which multipass cIM-MS experiments could improve the separation of mAbs' conformers. These initial results evidenced some limitations of HR-IM-MS for large native biomolecules which possess rich conformational landscapes that remain challenging to decipher even with higher IM resolving powers. Conversely, for collision-induced unfolding (CIU) approaches, higher resolution proved to be particularly useful (i) to reveal new unfolding states and (ii) to enhance the separation of coexisting activated states, thus allowing one to apprehend gas-phase CIU behaviors of mAbs directly at the intact level. Altogether, this study offers a first panoramic overview of the capabilities of cIM-MS for therapeutic mAbs, paving the way for more widespread HR-IM-MS/CIU characterization of mAb-derived formats.
Collapse
Affiliation(s)
- Evolène Deslignière
- Laboratoire de Spectrométrie de Masse BioOrganique, IPHC UMR 7178, Université de Strasbourg, CNRS, Strasbourg 67000, France.,Infrastructure Nationale de Protéomique ProFI - FR2048, Strasbourg 67087, France
| | - Simon Ollivier
- UR BIA, INRAE, Nantes F-44316, France.,PROBE Research Infrastructure, BIBS Facility, INRAE, Nantes F-44316, France
| | - Alain Beck
- IRPF Centre d'Immunologie Pierre-Fabre (CIPF), Saint-Julien-en-Genevois 74160, France
| | - David Ropartz
- UR BIA, INRAE, Nantes F-44316, France.,PROBE Research Infrastructure, BIBS Facility, INRAE, Nantes F-44316, France
| | - Hélène Rogniaux
- UR BIA, INRAE, Nantes F-44316, France.,PROBE Research Infrastructure, BIBS Facility, INRAE, Nantes F-44316, France
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique, IPHC UMR 7178, Université de Strasbourg, CNRS, Strasbourg 67000, France.,Infrastructure Nationale de Protéomique ProFI - FR2048, Strasbourg 67087, France
| |
Collapse
|
17
|
A New Approach for Increasing Speed, Loading Capacity, Resolution, and Scalability of Preparative Size-Exclusion Chromatography of Proteins. Processes (Basel) 2022. [DOI: 10.3390/pr10122566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Low speed, low capacity, and poor scalability make size-exclusion chromatography (SEC) unattractive for use in the preparative separation of proteins. We discuss a novel z2 cuboid SEC device that addresses these challenges. A z2 cuboid SEC device (~24 mL volume) was systematically compared with a conventional SEC column having the same volume and packed with the same resin. The primary objective of this study was to use the same volume of SEC medium in a much more efficient way by using the novel device. At any given flow rate, the pressure drop across the z2 cuboid SEC device was lower by a factor of 6 to 8 due to its shorter bed height and greater cross-sectional area. Under overloaded conditions, the peaks obtained during protein separation with the conventional column were poorly resolved and showed significant fronting, while those obtained with the z2 cuboid SEC device were much better resolved and showed no fronting. At any given flow rate, better resolution was obtained with the z2 cuboid SEC device, while for obtaining a comparable resolution, the flow rate that could be used with the z2 cuboid SEC device was higher by a factor of 2 to 3. Hence, productivity in SEC could easily be increased by 200 to 300% using the z2 cuboid SEC device. The scalability of the z2 cuboid SEC device was also demonstrated based on a device with a 200 mL bed volume.
Collapse
|
18
|
Deslignière E, Ollivier S, Ehkirch A, Martelet A, Ropartz D, Lechat N, Hernandez-Alba O, Menet JM, Clavier S, Rogniaux H, Genet B, Cianférani S. Combination of IM-Based Approaches to Unravel the Coexistence of Two Conformers on a Therapeutic Multispecific mAb. Anal Chem 2022; 94:7981-7989. [PMID: 35604400 PMCID: PMC9178554 DOI: 10.1021/acs.analchem.2c00928] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
![]()
Multispecific antibodies,
which target multiple antigens at once,
are emerging as promising therapeutic entities to offer more effective
treatment than conventional monoclonal antibodies (mAbs). However,
these highly complex mAb formats pose significant analytical challenges.
We report here on the characterization of a trispecific antibody (tsAb),
which presents two isomeric forms clearly separated and identified
with size exclusion chromatography coupled to native mass spectrometry
(SEC-nMS). Previous studies showed that these isomers might originate
from a proline cis/trans isomerization
in one Fab subunit of the tsAb. We combined several innovative ion
mobility (IM)-based approaches to confirm the isomeric nature of the
two species and to gain new insights into the conformational landscape
of both isomers. Preliminary SEC-nIM-MS measurements performed on
a low IM resolution instrument provided the first hints of the coexistence
of different conformers, while complementary collision-induced unfolding
(CIU) experiments evidenced distinct gas-phase unfolding behaviors
upon activation for the two isomers. As subtle conformational differences
remained poorly resolved on our early generation IM platform, we performed
high-resolution cyclic IM (cIM-MS) to unambiguously conclude on the
coexistence of two conformers. The cis/trans equilibrium was further tackled by exploiting the IMn slicing capabilities of the cIM-MS instrument. Altogether, our results
clearly illustrate the benefits of combining state-of-the-art nMS
and IM-MS approaches to address challenging issues encountered in
biopharma. As engineered antibody constructs become increasingly sophisticated,
CIU and cIM-MS methodologies undoubtedly have the potential to integrate
the drug development analytical toolbox to achieve in-depth conformational
characterization of these products.
Collapse
Affiliation(s)
- Evolène Deslignière
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67087 Strasbourg, France.,Infrastructure Nationale de Protéomique ProFI - FR2048, 67087 Strasbourg, France
| | - Simon Ollivier
- INRAE, UR BIA, F-44316 Nantes, France.,INRAE, BIBS Facility, F-44316 Nantes, France
| | - Anthony Ehkirch
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67087 Strasbourg, France.,Infrastructure Nationale de Protéomique ProFI - FR2048, 67087 Strasbourg, France
| | - Armelle Martelet
- CMC Development, BioAnalytics department France, SANOFI R&D, 94400 Vitry-sur-Seine, France
| | - David Ropartz
- INRAE, UR BIA, F-44316 Nantes, France.,INRAE, BIBS Facility, F-44316 Nantes, France
| | - Nelly Lechat
- CMC Development, BioAnalytics department France, SANOFI R&D, 94400 Vitry-sur-Seine, France
| | - Oscar Hernandez-Alba
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67087 Strasbourg, France.,Infrastructure Nationale de Protéomique ProFI - FR2048, 67087 Strasbourg, France
| | - Jean-Michel Menet
- CMC Development, BioAnalytics department France, SANOFI R&D, 94400 Vitry-sur-Seine, France
| | - Séverine Clavier
- CMC Development, BioAnalytics department France, SANOFI R&D, 94400 Vitry-sur-Seine, France
| | - Hélène Rogniaux
- INRAE, UR BIA, F-44316 Nantes, France.,INRAE, BIBS Facility, F-44316 Nantes, France
| | - Bruno Genet
- CMC Development, BioAnalytics department France, SANOFI R&D, 94400 Vitry-sur-Seine, France
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67087 Strasbourg, France.,Infrastructure Nationale de Protéomique ProFI - FR2048, 67087 Strasbourg, France
| |
Collapse
|
19
|
Abstract
Native mass spectrometry (nMS) has emerged as an important tool in studying the structure and function of macromolecules and their complexes in the gas phase. In this review, we cover recent advances in nMS and related techniques including sample preparation, instrumentation, activation methods, and data analysis software. These advances have enabled nMS-based techniques to address a variety of challenging questions in structural biology. The second half of this review highlights recent applications of these technologies and surveys the classes of complexes that can be studied with nMS. Complementarity of nMS to existing structural biology techniques and current challenges in nMS are also addressed.
Collapse
Affiliation(s)
- Kelly R Karch
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, USA;
- Resource for Native Mass Spectrometry Guided Structural Biology, The Ohio State University, Columbus, Ohio, USA
| | - Dalton T Snyder
- Resource for Native Mass Spectrometry Guided Structural Biology, The Ohio State University, Columbus, Ohio, USA
| | - Sophie R Harvey
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, USA;
- Resource for Native Mass Spectrometry Guided Structural Biology, The Ohio State University, Columbus, Ohio, USA
| | - Vicki H Wysocki
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, USA;
- Resource for Native Mass Spectrometry Guided Structural Biology, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
20
|
Vallejo DD, Jeon CK, Parson KF, Herderschee HR, Eschweiler JD, Filoti DI, Ruotolo BT. Ion Mobility-Mass Spectrometry Reveals the Structures and Stabilities of Biotherapeutic Antibody Aggregates. Anal Chem 2022; 94:6745-6753. [PMID: 35475624 DOI: 10.1021/acs.analchem.2c00160] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Stability is a key critical quality attribute monitored throughout the development of monoclonal antibody (mAb) therapeutics. Minor changes in their higher order structure (HOS) caused by stress or environment may alter mAb aggregation, immunogenicity, and efficacy. In addition, the structures of the resulting mAb aggregates are largely unknown, as are their dependencies on conditions under which they are created. In this report, we investigate the HOS of mAb monomers and dimers under a variety of forced degradation conditions with ion mobility-mass spectrometry (IM-MS) and collision-induced unfolding (CIU) technologies. We evaluate two model IgG1 antibodies that differ significantly only in their complementarity-determinant regions: IgG1α and IgG1β. Our data covering both heat- and pH-based forced degradation conditions, aquired on two different IM-MS platforms, show that these mAbs undergo global HOS changes at both monomer and dimer levels upon degradation, but shifts in collision cross section (CCS) differ under pH or heat degradation conditions. In addition, the level of CCS change detected is different between IgG1α and IgG1β, suggesting that differences in the CDR drive differential responses to degradation that influence the antibody HOS. Dramatically different CIU fingerprints are obtained for IgG1α and IgG1β monomers and dimers for both degradation conditions. Finally, we constructed a series of computational models of mAb dimers for comparison with experimental CCS values and found evidence for a compact, overlapped dimer structure under native and heat degradation conditions, possibly adopting an inverted or nonoverlapped quaternary structure when produced through pH degredation. We conclude by discussing the potential impact of our findings on ongoing biotherapeutic discovery and development efforts.
Collapse
Affiliation(s)
- Daniel D Vallejo
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Chae Kyung Jeon
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Kristine F Parson
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Hayley R Herderschee
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | | | - Dana I Filoti
- AbbVie, North Chicago, Illinois 60064, United States
| | - Brandon T Ruotolo
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
21
|
Abstract
Native mass spectrometry (MS) is aimed at preserving and determining the native structure, composition, and stoichiometry of biomolecules and their complexes from solution after they are transferred into the gas phase. Major improvements in native MS instrumentation and experimental methods over the past few decades have led to a concomitant increase in the complexity and heterogeneity of samples that can be analyzed, including protein-ligand complexes, protein complexes with multiple coexisting stoichiometries, and membrane protein-lipid assemblies. Heterogeneous features of these biomolecular samples can be important for understanding structure and function. However, sample heterogeneity can make assignment of ion mass, charge, composition, and structure very challenging due to the overlap of tens or even hundreds of peaks in the mass spectrum. In this review, we cover data analysis, experimental, and instrumental advances and strategies aimed at solving this problem, with an in-depth discussion of theoretical and practical aspects of the use of available deconvolution algorithms and tools. We also reflect upon current challenges and provide a view of the future of this exciting field.
Collapse
Affiliation(s)
- Amber D. Rolland
- Department of Chemistry and Biochemistry, 1253 University of Oregon, Eugene, OR, USA 97403-1253
| | - James S. Prell
- Department of Chemistry and Biochemistry, 1253 University of Oregon, Eugene, OR, USA 97403-1253
- Materials Science Institute, 1252 University of Oregon, Eugene, OR, USA 97403-1252
| |
Collapse
|
22
|
Vallejo DD, Ramírez CR, Parson KF, Han Y, Gadkari VG, Ruotolo BT. Mass Spectrometry Methods for Measuring Protein Stability. Chem Rev 2022; 122:7690-7719. [PMID: 35316030 PMCID: PMC9197173 DOI: 10.1021/acs.chemrev.1c00857] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mass spectrometry is a central technology in the life sciences, providing our most comprehensive account of the molecular inventory of the cell. In parallel with developments in mass spectrometry technologies targeting such assessments of cellular composition, mass spectrometry tools have emerged as versatile probes of biomolecular stability. In this review, we cover recent advancements in this branch of mass spectrometry that target proteins, a centrally important class of macromolecules that accounts for most biochemical functions and drug targets. Our efforts cover tools such as hydrogen-deuterium exchange, chemical cross-linking, ion mobility, collision induced unfolding, and other techniques capable of stability assessments on a proteomic scale. In addition, we focus on a range of application areas where mass spectrometry-driven protein stability measurements have made notable impacts, including studies of membrane proteins, heat shock proteins, amyloidogenic proteins, and biotherapeutics. We conclude by briefly discussing the future of this vibrant and fast-moving area of research.
Collapse
Affiliation(s)
- Daniel D. Vallejo
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Carolina Rojas Ramírez
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Kristine F. Parson
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yilin Han
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Varun G. Gadkari
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Brandon T. Ruotolo
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
23
|
N-Glycosylation of monoclonal antibody therapeutics: A comprehensive review on significance and characterization. Anal Chim Acta 2022; 1209:339828. [DOI: 10.1016/j.aca.2022.339828] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/06/2022] [Accepted: 04/09/2022] [Indexed: 01/02/2023]
|
24
|
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: 1.5] [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
|
25
|
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: 0.5] [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
|
26
|
Studying protein structure and function by native separation–mass spectrometry. Nat Rev Chem 2022; 6:215-231. [PMID: 37117432 DOI: 10.1038/s41570-021-00353-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2021] [Indexed: 12/13/2022]
Abstract
Alterations in protein structure may have profound effects on biological function. Analytical techniques that permit characterization of proteins while maintaining their conformational and functional state are crucial for studying changes in the higher order structure of proteins and for establishing structure-function relationships. Coupling of native protein separations with mass spectrometry is emerging rapidly as a powerful approach to study these aspects in a reliable, fast and straightforward way. This Review presents the available native separation modes for proteins, covers practical considerations on the hyphenation of these separations with mass spectrometry and highlights the involvement of affinity-based separations to simultaneously obtain structural and functional information of proteins. The impact of these approaches is emphasized by selected applications addressing biomedical and biopharmaceutical research questions.
Collapse
|
27
|
Duivelshof BL, Beck A, Guillarme D, D'Atri V. Bispecific antibody characterization by a combination of intact and site-specific/chain-specific LC/MS techniques. Talanta 2022; 236:122836. [PMID: 34635226 DOI: 10.1016/j.talanta.2021.122836] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/26/2021] [Accepted: 08/30/2021] [Indexed: 01/07/2023]
Abstract
Bispecific antibodies (bsAbs) are considered as an important class of biopharmaceutical drugs, with about 160 products in clinical trials. From an analytical point of view, the correct chain-association is one of the most critical challenge to monitor during bsAbs development and production. In the present study, a full analytical workflow has been developed based on the use of various chromatographic modes: size exclusion chromatography (SEC), ion exchange chromatography (IEX), reversed phase liquid chromatography (RPLC), and hydrophilic interaction chromatography (HILIC), all combined with high resolution mass spectrometry (MS). This analytical strategy was applied to Hemlibra® (emicizumab), which is certainly the most successful commercial bsAb to date. Using this strategy, it was possible to monitor the presence of mispaired bsAb species and detect and identify additional post-translational modifications (PTMs).
Collapse
Affiliation(s)
- Bastiaan L Duivelshof
- Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, CMU-Rue Michel Servet 1, 1211, Geneva 4, Switzerland; School of Pharmaceutical Sciences, University of Geneva, CMU-Rue Michel Servet 1, 1211, Geneva 4, Switzerland
| | - Alain Beck
- IRPF - Centre d'Immunologie Pierre-Fabre (CIPF), 5 Avenue Napoléon III, BP 60497, Saint-Julien-en-Genevois, France
| | - Davy Guillarme
- Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, CMU-Rue Michel Servet 1, 1211, Geneva 4, Switzerland; School of Pharmaceutical Sciences, University of Geneva, CMU-Rue Michel Servet 1, 1211, Geneva 4, Switzerland
| | - Valentina D'Atri
- Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, CMU-Rue Michel Servet 1, 1211, Geneva 4, Switzerland; School of Pharmaceutical Sciences, University of Geneva, CMU-Rue Michel Servet 1, 1211, Geneva 4, Switzerland.
| |
Collapse
|
28
|
Vallejo DD, Kang J, Coghlan J, Ramírez CR, Polasky DA, Kurulugama RT, Fjeldsted JC, Schwendeman AA, Ruotolo BT. Collision-Induced Unfolding Reveals Stability Differences in Infliximab Therapeutics under Native and Heat Stress Conditions. Anal Chem 2021; 93:16166-16174. [PMID: 34808055 DOI: 10.1021/acs.analchem.1c03946] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ion mobility-mass spectrometry (IM-MS) and collision-induced unfolding (CIU) assays of monoclonal antibody (mAb)-based biotherapeutics have proven sensitive to disulfide bridge structures, glycosylation patterns, and small molecule conjugation levels. Despite promising prior reports detailing the capabilities of IM-MS and CIU to differentiate biosimilars, generic mAb therapeutics, there remain questions surrounding the sensitivity of CIU to mAb structure changes that occur upon stress, the reproducibility of such measurements across IM-MS platforms, and the correlation between CIU and differential scanning calorimetry (DSC) datasets. In this report, we describe a comprehensive IM-MS and CIU dataset acquired for three Infliximabs: Remicade, Inflectra, and Renflexis. We subject each infliximab sample to forced degradation through heat stress and observe broadly similar yet subtly different stability patterns for these three biotherapeutics. We find that CIU is capable of tracking differences in mAb higher-order structure (HOS) imparted during forced heat stress degradation and that DSC is less sensitive to these alterations in comparison. Furthermore, we collected our comprehensive IM-MS and CIU data across two instrument platforms (Waters G2 and Agilent 6560), with both producing similar abilities to differentiate mAbs while also revealing minor differences between the results obtained on the two instruments. Finally, we demonstrate that CIU-based heatmaps and classification allow for rapid assessment of the most differentiating charge states for the analysis of infliximab, and using multiplexed classification, we conservatively estimate a 30-fold improvement in the time required to perform mAb stability and HOS measurements over standard DSC tools.
Collapse
Affiliation(s)
- Daniel D Vallejo
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jukyung Kang
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jill Coghlan
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Carolina Rojas Ramírez
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Daniel A Polasky
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | | | - John C Fjeldsted
- Agilent Technologies, Santa Clara, California 95051, United States
| | - Anna A Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Brandon T Ruotolo
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
29
|
Yan Y, Xing T, Liu AP, Zhang Z, Wang S, Li N. Post-Column Denaturation-Assisted Native Size-Exclusion Chromatography-Mass Spectrometry for Rapid and In-Depth Characterization of High Molecular Weight Variants in Therapeutic Monoclonal Antibodies. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2021; 32:2885-2894. [PMID: 34786946 DOI: 10.1021/jasms.1c00289] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The high molecular weight (HMW) size variants present in therapeutic monoclonal antibody (mAb) samples need to be closely monitored and characterized due to their impact on product safety and efficacy. Because of the complexity and often low abundances in final drug substance (DS) samples, characterization of such HMW species is challenging and traditionally requires offline enrichment of the HMW species followed by analysis using various analytical tools. Here, we report the development of a postcolumn denaturation-assisted native SEC-MS method that allows rapid and in-depth characterization of mAb HMW species directly from unfractionated DS samples. This method not only provides high-confidence identification of HMW complexes based on accurate mass measurement of both the intact assembly and the constituent subunits but also allows in-depth analysis of the interaction nature and location. In addition, using the extracted ion chromatograms, derived from high-quality, native-like mass spectra, the elution profiles of each noncovalent and/or nondissociable complex can be readily reconstructed, facilitating the comprehension of a complex HMW profile. The utility of this novel method was demonstrated in different applications, ranging from enriched HMW characterization at late stage development, comparability assessment due to process changes, and forced degradation study of coformulated mAbs. As this method does not require prior enrichment, it is thus desirable for providing both rapid and in-depth characterization of HMW species during the development of therapeutic mAbs.
Collapse
Affiliation(s)
- Yuetian Yan
- Analytical Chemistry Group, Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Tao Xing
- Analytical Chemistry Group, Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Anita P Liu
- Analytical Chemistry Group, Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Zhengqi Zhang
- Analytical Chemistry Group, Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Shunhai Wang
- Analytical Chemistry Group, Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Ning Li
- Analytical Chemistry Group, Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| |
Collapse
|
30
|
Abstract
Mass spectrometry (MS) is a powerful technique for protein identification, quantification and characterization that is widely applied in biochemical studies, and which can provide data on the quantity, structural integrity and post-translational modifications of proteins. It is therefore a versatile and widely used analytic tool for quality control of biopharmaceuticals, especially in quantifying host-cell protein impurities, identifying post-translation modifications and structural characterization of biopharmaceutical proteins. Here, we summarize recent advances in MS-based analyses of these key quality attributes of the biopharmaceutical development and manufacturing processes.
Collapse
|
31
|
Calvaresi V, Redsted A, Norais N, Rand KD. Hydrogen-Deuterium Exchange Mass Spectrometry with Integrated Size-Exclusion Chromatography for Analysis of Complex Protein Samples. Anal Chem 2021; 93:11406-11414. [PMID: 34387074 DOI: 10.1021/acs.analchem.1c01171] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The growing use of hydrogen-deuterium exchange mass spectrometry (HDX-MS) for studying membrane proteins, large protein assemblies, and highly disulfide-bonded species is often challenged by the presence in the sample of large amounts of lipids, protein ligands, and/or highly ionizable reducing agents. Here, we describe how a short size-exclusion chromatography (SEC) column can be integrated with a conventional temperature-controlled HDX-MS setup to achieve fast and online removal of unwanted species from the HDX sample prior to chromatographic separation and MS analysis. Dual-mode valves permit labeled proteins eluting after SEC to be directed to the proteolytic and chromatographic columns, while unwanted sample components are led to waste. The SEC-coupled HDX-MS method allows analyses to be completed with lower or similar back-exchange compared to conventional experiments. We demonstrate the suitability of the method for the analysis of challenging protein samples, achieving efficient online removal of lipid components from protein-lipid systems, depletion of an antibody from an antigen during epitope mapping, and elimination of MS interfering compounds such as tris(2-carboxyethyl)phosphine (TCEP) during HDX-MS analysis of a disulfide-bonded protein. The implementation of the short SEC column to the conventional HDX-MS setup is straightforward and could be of significant general utility during the HDX-MS analysis of complex protein states.
Collapse
Affiliation(s)
- Valeria Calvaresi
- Protein Analysis Group, Department of Pharmacy, University of Copenhagen, Copenhagen 2100, Denmark.,GSK, Siena 53100, Italy
| | - Andreas Redsted
- Protein Analysis Group, Department of Pharmacy, University of Copenhagen, Copenhagen 2100, Denmark.,GSK, Siena 53100, Italy
| | | | - Kasper D Rand
- Protein Analysis Group, Department of Pharmacy, University of Copenhagen, Copenhagen 2100, Denmark
| |
Collapse
|
32
|
Rizzo D, Cerofolini L, Pérez-Ràfols A, Giuntini S, Baroni F, Ravera E, Luchinat C, Fragai M. Evaluation of the Higher Order Structure of Biotherapeutics Embedded in Hydrogels for Bioprinting and Drug Release. Anal Chem 2021; 93:11208-11214. [PMID: 34339178 PMCID: PMC8382223 DOI: 10.1021/acs.analchem.1c01850] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/20/2021] [Indexed: 01/16/2023]
Abstract
Biocompatible hydrogels for tissue regeneration/replacement and drug release with specific architectures can be obtained by three-dimensional bioprinting techniques. The preservation of the higher order structure of the proteins embedded in the hydrogels as drugs or modulators is critical for their biological activity. Solution nuclear magnetic resonance (NMR) experiments are currently used to investigate the higher order structure of biotherapeutics in comparability, similarity, and stability studies. However, the size of pores in the gel, protein-matrix interactions, and the size of the embedded proteins often prevent the use of this methodology. The recent advancements of solid-state NMR allow for the comparison of the higher order structure of the matrix-embedded and free isotopically enriched proteins, allowing for the evaluation of the functionality of the material in several steps of hydrogel development. Moreover, the structural information at atomic detail on the matrix-protein interactions paves the way for a structure-based design of these biomaterials.
Collapse
Affiliation(s)
- Domenico Rizzo
- Magnetic
Resonance Center (CERM), University of Florence,
and Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine
(CIRMMP), Via L. Sacconi 6, Sesto Fiorentino 50019, Italy
- Department
of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3, Sesto Fiorentino 50019, Italy
| | - Linda Cerofolini
- Magnetic
Resonance Center (CERM), University of Florence,
and Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine
(CIRMMP), Via L. Sacconi 6, Sesto Fiorentino 50019, Italy
| | - Anna Pérez-Ràfols
- Department
of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3, Sesto Fiorentino 50019, Italy
- Giotto
Biotech, S.R.L, Via Madonna
del piano 6, Sesto Fiorentino, Florence 50019, Italy
| | - Stefano Giuntini
- Magnetic
Resonance Center (CERM), University of Florence,
and Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine
(CIRMMP), Via L. Sacconi 6, Sesto Fiorentino 50019, Italy
- Department
of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3, Sesto Fiorentino 50019, Italy
| | - Fabio Baroni
- Analytical
Development Biotech Department, Merck Serono
S.p.a, Merck KGaA, Guidonia, Rome 00012, Italy
| | - Enrico Ravera
- Magnetic
Resonance Center (CERM), University of Florence,
and Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine
(CIRMMP), Via L. Sacconi 6, Sesto Fiorentino 50019, Italy
- Department
of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3, Sesto Fiorentino 50019, Italy
| | - Claudio Luchinat
- Magnetic
Resonance Center (CERM), University of Florence,
and Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine
(CIRMMP), Via L. Sacconi 6, Sesto Fiorentino 50019, Italy
- Department
of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3, Sesto Fiorentino 50019, Italy
| | - Marco Fragai
- Magnetic
Resonance Center (CERM), University of Florence,
and Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine
(CIRMMP), Via L. Sacconi 6, Sesto Fiorentino 50019, Italy
- Department
of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3, Sesto Fiorentino 50019, Italy
| |
Collapse
|
33
|
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: 5.0] [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
|
34
|
Füssl F, Strasser L, Cari S, Bones J. Native LC-MS for capturing quality attributes of biopharmaceuticals on the intact protein level. Curr Opin Biotechnol 2021; 71:32-40. [PMID: 34157600 DOI: 10.1016/j.copbio.2021.05.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/12/2021] [Accepted: 05/24/2021] [Indexed: 11/16/2022]
Abstract
Intact protein analysis by means of mass spectrometry has become a well-established method for the characterization of biotherapeutics. However, due to the highly complex nature of recombinant proteins, prior chromatographic separation is inevitable for a comprehensive analysis. In recent years, progress in coupling a variety of liquid chromatography-based native separation modes such as size exclusion, ion exchange and hydrophobic interaction chromatography to mass spectrometry (native LC-MS) has been reported, therefore allowing for rapid assessment of molecular mass and deep characterization of the heterogeneity of complex, recombinantly produced therapeutic proteins. Here we provide a comprehensive overview of recent advances in the development and application of native LC-MS for biopharmaceutical characterization.
Collapse
Affiliation(s)
- Florian Füssl
- NIBRT - The National Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merrion, Blackrock, Co. Dublin, A94 X099, Ireland
| | - Lisa Strasser
- NIBRT - The National Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merrion, Blackrock, Co. Dublin, A94 X099, Ireland
| | - Sara Cari
- NIBRT - The National Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merrion, Blackrock, Co. Dublin, A94 X099, Ireland
| | - Jonathan Bones
- NIBRT - The National Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merrion, Blackrock, Co. Dublin, A94 X099, Ireland; School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Dublin 4, D04 V1W8, Ireland.
| |
Collapse
|
35
|
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: 16] [Impact Index Per Article: 5.3] [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
|
36
|
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: 7.0] [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
|
37
|
A structural signature motif enlightens the origin and diversification of nuclear receptors. PLoS Genet 2021; 17:e1009492. [PMID: 33882063 PMCID: PMC8092661 DOI: 10.1371/journal.pgen.1009492] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 05/03/2021] [Accepted: 03/15/2021] [Indexed: 12/11/2022] Open
Abstract
Nuclear receptors are ligand-activated transcription factors that modulate gene regulatory networks from embryonic development to adult physiology and thus represent major targets for clinical interventions in many diseases. Most nuclear receptors function either as homodimers or as heterodimers. The dimerization is crucial for gene regulation by nuclear receptors, by extending the repertoire of binding sites in the promoters or the enhancers of target genes via combinatorial interactions. Here, we focused our attention on an unusual structural variation of the α-helix, called π-turn that is present in helix H7 of the ligand-binding domain of RXR and HNF4. By tracing back the complex evolutionary history of the π-turn, we demonstrate that it was present ancestrally and then independently lost in several nuclear receptor lineages. Importantly, the evolutionary history of the π-turn motif is parallel to the evolutionary diversification of the nuclear receptor dimerization ability from ancestral homodimers to derived heterodimers. We then carried out structural and biophysical analyses, in particular through point mutation studies of key RXR signature residues and showed that this motif plays a critical role in the network of interactions stabilizing homodimers. We further showed that the π-turn was instrumental in allowing a flexible heterodimeric interface of RXR in order to accommodate multiple interfaces with numerous partners and critical for the emergence of high affinity receptors. Altogether, our work allows to identify a functional role for the π-turn in oligomerization of nuclear receptors and reveals how this motif is linked to the emergence of a critical biological function. We conclude that the π-turn can be viewed as a structural exaptation that has contributed to enlarging the functional repertoire of nuclear receptors. The origin of novelties is a central topic in evolutionary biology. A fundamental question is how organisms constrained by natural selection can divert from existing schemes to set up novel structures or pathways. Among the most important strategies are exaptations, which represent pre-adaptation strategies. Many examples exist in biology, at both morphological and molecular levels, such as the one reported here that focuses on an unusual structural feature called the π-turn. It is found in the structure of the most ancestral nuclear receptors RXR and HNF4. The analyses trace back the complex evolutionary history of the π-turn to more than 500 million years ago, before the Cambrian explosion and show that this feature was essential for the heterodimerization capacity of RXR. Nuclear receptor lineages that emerged later in evolution lost the π-turn. We demonstrate here that this loss in nuclear receptors that heterodimerize with RXR was critical for the emergence of high affinity receptors, such as the vitamin D and the thyroid hormone receptors. On the other hand, the conserved π-turn in RXR allowed it to accommodate multiple heterodimer interfaces with numerous partners. This structural exaptation allowed for the remarkable diversification of nuclear receptors.
Collapse
|
38
|
Jones J, Pack L, Hunter JH, Valliere-Douglass JF. Native size-exclusion chromatography-mass spectrometry: suitability for antibody-drug conjugate drug-to-antibody ratio quantitation across a range of chemotypes and drug-loading levels. MAbs 2021; 12:1682895. [PMID: 31769727 PMCID: PMC6927766 DOI: 10.1080/19420862.2019.1682895] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Native size-exclusion chromatography-mass spectrometry (nSEC-MS) is an analytical methodology that is appropriate for accurately quantitating the drug-to-antibody ratio (DAR) on a wide variety of interchain cysteine-linked antibody-drug conjugates (ADCs), irrespective of chemotype. In the current preclinical environment, novel ADCs conjugated with unique drug-linkers need to progress toward the clinic as quickly as possible. Platform analytical approaches can reduce time-to-clinic because key process development and optimization activities can be decoupled from the development of bespoke, molecule-specific analytical methods. In this work, we assessed the potential of nSEC-MS as a platformable, quantitative DAR method. The nSEC-MS method was evaluated according to performance characteristics and parameters described in the ICH guideline Validation of Analytical Procedures: Text and Methodology Q2(R1). In order to comprehensively assess the accuracy and bias of nSEC-MS DAR quantitation, ADCs were generated using three different drug-linker chemotypes with DARs ranging from 2 to 8. These molecules were tested by hydrophobic interaction chromatography (HIC) and nSEC-MS, and DARs obtained from both methods were compared to assess the degree to which nSEC-MS quantitation aligned with the HIC release assay. Our results indicated that there is no bias introduced by nSEC-MS quantitation of DAR and that SEC-MS data can be bridged to HIC data without the need for a correction factor or offset. nSEC-MS was also found to be suitable for unbiased DAR quantitation in the other ADC chemotypes that were evaluated. Based on the totality of our work, we conclude that, used as intended, nSEC-MS is well suited for quantitating DAR on a variety of interchain cysteine-linked ADCs in an accurate, unbiased manner.
Collapse
Affiliation(s)
- Jay Jones
- Analytical Sciences, Seattle Genetics Inc., Bothell, WA, USA
| | - Laura Pack
- Quality, Seattle Genetics Inc., Bothell, WA, USA
| | - Joshua H Hunter
- Conjugation Process Development, Seattle Genetics Inc., Bothell, WA, USA
| | | |
Collapse
|
39
|
Verscheure L, Oosterlynck M, Cerdobbel A, Sandra P, Lynen F, Sandra K. Middle-up characterization of monoclonal antibodies by online reduction liquid chromatography-mass spectrometry. J Chromatogr A 2020; 1637:461808. [PMID: 33385741 DOI: 10.1016/j.chroma.2020.461808] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023]
Abstract
This study describes the fully automated middle-up characterization of monoclonal antibodies (mAbs) and next-generation variants by online reduction liquid chromatography-mass spectrometry (LC-MS). Proteins were trapped on-column and subjected to online desalting, denaturation and reduction prior to reversed phase elution of the created subunits in the MS. The evaluation of more than 20 different therapeutic proteins including full length mAbs (subclasses IgG1, IgG2 and IgG4), bispecific antibodies, antibody fragments, fusion proteins and antibody-drug conjugates (ADC) revealed that the online reduction method is as powerful as the widely applied offline sample preparation with dithiothreitol (DTT) as reducing agent and guanidine hydrochloride (Gnd.HCl) as denaturant and tackles some major disadvantages associated with the latter method, i.e. corrosion of stainless steel components, adduct formation impacting spectral quality and sample stability. The value of the online reduction LC-MS method is also enforced by its ability to reveal unstable antibody variants such as succinimide intermediates of asparagine deamidation and aspartic acid isomerization which are often lost when using the offline sample preparation method. The performance of the online reduction LC-MS set-up was verified and it was revealed that the method is precise with RSD values below 0.25% and 3.0% for retention time and area, respectively. Carry-over is within acceptable limits (< 0.5%) and the reducing buffer is stable up to 24 hours.
Collapse
Affiliation(s)
- Liesa Verscheure
- Research Institute for Chromatography (RIC), President Kennedypark 26, 8500 Kortrijk, Belgium; Separation Science Group, Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281-S4, B-9000 Ghent, Belgium
| | - Marie Oosterlynck
- Chemistry Department, KU Leuven, Celestijnenlaan 200F, bus 2404, 3001 Leuven, Belgium
| | - An Cerdobbel
- Research Institute for Chromatography (RIC), President Kennedypark 26, 8500 Kortrijk, Belgium
| | - Pat Sandra
- Research Institute for Chromatography (RIC), President Kennedypark 26, 8500 Kortrijk, Belgium; Separation Science Group, Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281-S4, B-9000 Ghent, Belgium
| | - Frederic Lynen
- Separation Science Group, Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281-S4, B-9000 Ghent, Belgium
| | - Koen Sandra
- Research Institute for Chromatography (RIC), President Kennedypark 26, 8500 Kortrijk, Belgium; Separation Science Group, Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281-S4, B-9000 Ghent, Belgium.
| |
Collapse
|
40
|
Deslignière E, Ehkirch A, Botzanowski T, Beck A, Hernandez-Alba O, Cianférani S. Toward Automation of Collision-Induced Unfolding Experiments through Online Size Exclusion Chromatography Coupled to Native Mass Spectrometry. Anal Chem 2020; 92:12900-12908. [DOI: 10.1021/acs.analchem.0c01426] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Evolène Deslignière
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - Anthony Ehkirch
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - Thomas Botzanowski
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - Alain Beck
- IRPF—Centre d’Immunologie Pierre-Fabre (CIPF), 74160 Saint-Julien-en-Genevois, France
| | - Oscar Hernandez-Alba
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| |
Collapse
|
41
|
Allison TM, Barran P, Benesch JLP, Cianferani S, Degiacomi MT, Gabelica V, Grandori R, Marklund EG, Menneteau T, Migas LG, Politis A, Sharon M, Sobott F, Thalassinos K. Software Requirements for the Analysis and Interpretation of Native Ion Mobility Mass Spectrometry Data. Anal Chem 2020; 92:10881-10890. [PMID: 32649184 DOI: 10.1021/acs.analchem.9b05792] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The past few years have seen a dramatic increase in applications of native mass and ion mobility spectrometry, especially for the study of proteins and protein complexes. This increase has been catalyzed by the availability of commercial instrumentation capable of carrying out such analyses. As in most fields, however, the software to process the data generated from new instrumentation lags behind. Recently, a number of research groups have started addressing this by developing software, but further improvements are still required in order to realize the full potential of the data sets generated. In this perspective, we describe practical aspects as well as challenges in processing native mass spectrometry (MS) and ion mobility-MS data sets and provide a brief overview of currently available tools. We then set out our vision of future developments that would bring the community together and lead to the development of a common platform to expedite future computational developments, provide standardized processing approaches, and serve as a location for the deposition of data for this emerging field. This perspective has been written by members of the European Cooperation in Science and Technology Action on Native MS and Related Methods for Structural Biology (EU COST Action BM1403) as an introduction to the software tools available in this area. It is intended to serve as an overview for newcomers and to stimulate discussions in the community on further developments in this field, rather than being an in-depth review. Our complementary perspective (http://dx.doi.org/10.1021/acs.analchem.9b05791) focuses on computational approaches used in this field.
Collapse
Affiliation(s)
- Timothy M Allison
- School of Physical and Chemical Sciences, Biomolecular Interaction Centre, University of Canterbury, Christchurch 8140, New Zealand
| | - Perdita Barran
- Michael Barber Centre for Collaborative Mass Spectrometry, Manchester Institute of Biotechnology, School of Chemistry, University of Manchester, Manchester M1 7DN, United Kingdom
| | - Justin L P Benesch
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, South Parks Road, Oxford OX1 3TA, United Kingdom
| | - Sarah Cianferani
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - Matteo T Degiacomi
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, South Parks Road, Oxford OX1 3TA, United Kingdom.,Department of Physics, Durham University, South Road, Durham DH1 3LE, United Kingdom
| | - Valerie Gabelica
- University of Bordeaux, INSERM and CNRS, ARNA Laboratory, IECB site, 2 Rue Robert Escarpit, 33600 Pessac, France
| | - Rita Grandori
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
| | - Erik G Marklund
- Department of Chemistry - BMC, Uppsala University, Box 576, 75123 Uppsala, Sweden
| | - Thomas Menneteau
- Division of Biosciences, Institute of Structural and Molecular Biology, University College of London, Gower Street, London WC1E 6BT, United Kingdom
| | - Lukasz G Migas
- Michael Barber Centre for Collaborative Mass Spectrometry, Manchester Institute of Biotechnology, School of Chemistry, University of Manchester, Manchester M1 7DN, United Kingdom
| | - Argyris Politis
- Department of Chemistry, King's College London, 7 Trinity Street, London SE1 1DB, United Kingdom
| | - Michal Sharon
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Frank Sobott
- Biomolecular & Analytical Mass Spectrometry, Department of Chemistry, University of Antwerp, 2020 Antwerp, Belgium.,School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom.,Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Konstantinos Thalassinos
- Division of Biosciences, Institute of Structural and Molecular Biology, University College of London, Gower Street, London WC1E 6BT, United Kingdom.,Department of Biological Sciences, Institute of Structural and Molecular Biology, Birkbeck, Malet Street, London WC1E 7HX, United Kingdom
| |
Collapse
|
42
|
Recent advances in LC–MS based characterization of protein-based bio-therapeutics – mastering analytical challenges posed by the increasing format complexity. J Pharm Biomed Anal 2020; 186:113251. [DOI: 10.1016/j.jpba.2020.113251] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 12/25/2022]
|
43
|
Ciudad S, Puig E, Botzanowski T, Meigooni M, Arango AS, Do J, Mayzel M, Bayoumi M, Chaignepain S, Maglia G, Cianferani S, Orekhov V, Tajkhorshid E, Bardiaux B, Carulla N. Aβ(1-42) tetramer and octamer structures reveal edge conductivity pores as a mechanism for membrane damage. Nat Commun 2020; 11:3014. [PMID: 32541820 PMCID: PMC7296003 DOI: 10.1038/s41467-020-16566-1] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 05/01/2020] [Indexed: 12/20/2022] Open
Abstract
Formation of amyloid-beta (Aβ) oligomer pores in the membrane of neurons has been proposed to explain neurotoxicity in Alzheimer's disease (AD). Here, we present the three-dimensional structure of an Aβ oligomer formed in a membrane mimicking environment, namely an Aβ(1-42) tetramer, which comprises a six stranded β-sheet core. The two faces of the β-sheet core are hydrophobic and surrounded by the membrane-mimicking environment while the edges are hydrophilic and solvent-exposed. By increasing the concentration of Aβ(1-42) in the sample, Aβ(1-42) octamers are also formed, made by two Aβ(1-42) tetramers facing each other forming a β-sandwich structure. Notably, Aβ(1-42) tetramers and octamers inserted into lipid bilayers as well-defined pores. To establish oligomer structure-membrane activity relationships, molecular dynamics simulations were carried out. These studies revealed a mechanism of membrane disruption in which water permeation occurred through lipid-stabilized pores mediated by the hydrophilic residues located on the core β-sheets edges of the oligomers.
Collapse
Affiliation(s)
- Sonia Ciudad
- University of Bordeaux, CBMN (UMR 5248)-CNRS-IPB, Institut Européen de Chimie et Biologie, 2 rue Escarpit, 33600, Pessac, France
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028, Barcelona, Spain
| | - Eduard Puig
- University of Bordeaux, CBMN (UMR 5248)-CNRS-IPB, Institut Européen de Chimie et Biologie, 2 rue Escarpit, 33600, Pessac, France
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028, Barcelona, Spain
- Departament de Química Inorgànica i Orgànica, Universitat de Barcelona, Martí i Franqués 1, 08028, Barcelona, Spain
| | - Thomas Botzanowski
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS UMR7178, IPHC, Strasbourg, France
| | - Moeen Meigooni
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology, Center for Biophysics and Quantitative Biology and Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Andres S Arango
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology, Center for Biophysics and Quantitative Biology and Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jimmy Do
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology, Center for Biophysics and Quantitative Biology and Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Maxim Mayzel
- Swedish NMR Centre, University of Gothenburg, Box 465, 405 30, Gothenburg, Sweden
| | - Mariam Bayoumi
- Biochemistry, Molecular and Structural Biology Section, University of Leuven, Celestijnenlaan 200G, 3001, Leuven, Belgium
| | - Stéphane Chaignepain
- University of Bordeaux, CBMN (UMR 5248)-CNRS-IPB, Institut Européen de Chimie et Biologie, 2 rue Escarpit, 33600, Pessac, France
| | - Giovanni Maglia
- Groningen Biomolecular Sciences & Biotechnology Institute, University of Groningen, 9747 AG, Groningen, The Netherlands
| | - Sarah Cianferani
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS UMR7178, IPHC, Strasbourg, France
| | - Vladislav Orekhov
- Swedish NMR Centre, University of Gothenburg, Box 465, 405 30, Gothenburg, Sweden
- Department of Chemistry and Molecular Biology, University of Gothenburg, Box 465, 405 30, Gothenburg, Sweden
| | - Emad Tajkhorshid
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology, Center for Biophysics and Quantitative Biology and Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Benjamin Bardiaux
- Structural Bioinformatics Unit, Department of Structural Biology and Chemistry, C3BI, Institut Pasteur; CNRS UMR3528; CNRS USR3756, Paris, France
| | - Natàlia Carulla
- University of Bordeaux, CBMN (UMR 5248)-CNRS-IPB, Institut Européen de Chimie et Biologie, 2 rue Escarpit, 33600, Pessac, France.
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028, Barcelona, Spain.
| |
Collapse
|
44
|
Farsang E, Guillarme D, Veuthey JL, Beck A, Lauber M, Schmudlach A, Fekete S. Coupling non-denaturing chromatography to mass spectrometry for the characterization of monoclonal antibodies and related products. J Pharm Biomed Anal 2020; 185:113207. [DOI: 10.1016/j.jpba.2020.113207] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/21/2020] [Accepted: 02/22/2020] [Indexed: 01/31/2023]
|
45
|
Conformation assessment of therapeutic monoclonal antibodies by SEC-MS: Unravelling analytical biases for application to quality control. J Pharm Biomed Anal 2020; 185:113252. [DOI: 10.1016/j.jpba.2020.113252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 11/18/2022]
|
46
|
Dovgan I, Ehkirch A, Lehot V, Kuhn I, Koniev O, Kolodych S, Hentz A, Ripoll M, Ursuegui S, Nothisen M, Cianférani S, Wagner A. On the use of DNA as a linker in antibody-drug conjugates: synthesis, stability and in vitro potency. Sci Rep 2020; 10:7691. [PMID: 32376903 PMCID: PMC7203131 DOI: 10.1038/s41598-020-64518-y] [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] [Received: 12/06/2019] [Accepted: 04/07/2020] [Indexed: 12/16/2022] Open
Abstract
Here we present the synthesis and evaluation of antibody-drug conjugates (ADCs), for which antibody and drug are non-covalently connected using complementary DNA linkers. These ADCs are composed of trastuzumab, an antibody targeting HER2 receptors overexpressed on breast cancer cells, and monomethyl auristatin E (MMAE) as a drug payload. In this new ADC format, trastuzumab conjugated to a 37-mer oligonucleotide (ON) was prepared and hybridized with its complementary ON modified at 5-end with MMAE (cON-MMAE) in order to obtain trastuzumab-DNA-MMAE. As an advantage, the cON-MMAE was completely soluble in water, which decreases overall hydrophobicity of toxic payload, an important characteristic of ADCs. The stability in the human plasma of these non-engineered ON-based linkers was investigated and showed a satisfactory half-life of 5.8 days for the trastuzumab-DNA format. Finally, we investigated the in vitro cytotoxicity profile of both the DNA-linked ADC and the ON-drug conjugates and compared them with classical covalently linked ADC. Interestingly, we found increased cytotoxicity for MMAE compared to cON-MMAE and an EC50 in the nanomolar range for trastuzumab-DNA-MMAE on HER2-positive cells. Although this proved to be less potent than classically linked ADC with picomolar range EC50, the difference in cytotoxicity between naked payload and conjugated payload was significant when an ON linker was used. We also observed an interesting increase in cytotoxicity of trastuzumab-DNA-MMAE on HER2-negative cells. This was attributed to enhanced non-specific interaction triggered by the DNA strand as it could be confirmed using ligand tracer assay.
Collapse
Affiliation(s)
- Igor Dovgan
- Bio-Functional Chemistry (UMR 7199), LabEx Medalis, University of Strasbourg, 74 Route du Rhin, 67400, Illkirch-Graffenstaden, France
| | - Anthony Ehkirch
- BioOrganic Mass Spectrometry Laboratory (LSMBO), IPHC, University of Strasbourg, 25 rue Becquerel, 67087, Strasbourg, France
| | - Victor Lehot
- Bio-Functional Chemistry (UMR 7199), LabEx Medalis, University of Strasbourg, 74 Route du Rhin, 67400, Illkirch-Graffenstaden, France
| | - Isabelle Kuhn
- Bio-Functional Chemistry (UMR 7199), LabEx Medalis, University of Strasbourg, 74 Route du Rhin, 67400, Illkirch-Graffenstaden, France
| | - Oleksandr Koniev
- Syndivia SAS, 650 Boulevard Gonthier d'Andernach, 67400, Illkirch-Graffenstaden, France
| | - Sergii Kolodych
- Syndivia SAS, 650 Boulevard Gonthier d'Andernach, 67400, Illkirch-Graffenstaden, France
| | - Alexandre Hentz
- Bio-Functional Chemistry (UMR 7199), LabEx Medalis, University of Strasbourg, 74 Route du Rhin, 67400, Illkirch-Graffenstaden, France
| | - Manon Ripoll
- Bio-Functional Chemistry (UMR 7199), LabEx Medalis, University of Strasbourg, 74 Route du Rhin, 67400, Illkirch-Graffenstaden, France
| | - Sylvain Ursuegui
- Bio-Functional Chemistry (UMR 7199), LabEx Medalis, University of Strasbourg, 74 Route du Rhin, 67400, Illkirch-Graffenstaden, France
| | - Marc Nothisen
- Bio-Functional Chemistry (UMR 7199), LabEx Medalis, University of Strasbourg, 74 Route du Rhin, 67400, Illkirch-Graffenstaden, France
| | - Sarah Cianférani
- BioOrganic Mass Spectrometry Laboratory (LSMBO), IPHC, University of Strasbourg, 25 rue Becquerel, 67087, Strasbourg, France.,IPHC, CNRS, UMR7178, University of Strasbourg, 67087, Strasbourg, France
| | - Alain Wagner
- Bio-Functional Chemistry (UMR 7199), LabEx Medalis, University of Strasbourg, 74 Route du Rhin, 67400, Illkirch-Graffenstaden, France.
| |
Collapse
|
47
|
Wagner E, Colas O, Chenu S, Goyon A, Murisier A, Cianferani S, François Y, Fekete S, Guillarme D, D’Atri V, Beck A. Determination of size variants by CE-SDS for approved therapeutic antibodies: Key implications of subclasses and light chain specificities. J Pharm Biomed Anal 2020; 184:113166. [DOI: 10.1016/j.jpba.2020.113166] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/06/2020] [Accepted: 02/09/2020] [Indexed: 12/27/2022]
|
48
|
Gargano A, Schouten O, van Schaick G, Roca L, van den Berg-Verleg J, Haselberg R, Akeroyd M, Abello N, Somsen G. Profiling of a high mannose-type N-glycosylated lipase using hydrophilic interaction chromatography-mass spectrometry. Anal Chim Acta 2020; 1109:69-77. [DOI: 10.1016/j.aca.2020.02.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/17/2020] [Accepted: 02/23/2020] [Indexed: 10/24/2022]
|
49
|
Ventouri IK, Malheiro DBA, Voeten RLC, Kok S, Honing M, Somsen GW, Haselberg R. Probing Protein Denaturation during Size-Exclusion Chromatography Using Native Mass Spectrometry. Anal Chem 2020; 92:4292-4300. [PMID: 32107919 PMCID: PMC7081181 DOI: 10.1021/acs.analchem.9b04961] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
![]()
Size-exclusion chromatography
employing aqueous mobile phases with
volatile salts at neutral pH combined with electrospray-ionization
mass spectrometry (SEC-ESI-MS) is a useful tool to study proteins
in their native state. However, whether the applied eluent conditions
actually prevent protein–stationary phase interactions, and/or
protein denaturation, often is not assessed. In this study, the effects
of volatile mobile phase additives on SEC retention and ESI of proteins
were thoroughly investigated. Myoglobin was used as the main model
protein, and eluents of varying ionic strength and pH were applied.
The degree of interaction between protein and stationary phase was
evaluated by calculating the SEC distribution coefficient. Protein-ion
charge state distributions obtained during offline and online native
ESI-MS were used to monitor alterations in protein structure. Interestingly,
most of the supposedly mild eluent compositions induced nonideal SEC
behavior and/or protein unfolding. SEC experiments revealed that the
nature, ionic strength, and pH of the eluent affected protein retention.
Protein–stationary phase interactions were effectively avoided
using ammonium acetate at ionic strengths above 0.1 M. Direct-infusion
ESI-MS showed that the tested volatile eluent salts seem to follow
the Hofmeister series: no denaturation was induced using ammonium
acetate (kosmotropic), whereas ammonium formate and bicarbonate (both
chaotropic) caused structural changes. Using a mobile phase of 0.2
M ammonium acetate (pH 6.9), several proteins (i.e., myoglobin, carbonic
anhydrase, and cytochrome c) could be analyzed by SEC-ESI-MS using
different column chemistries without compromising their native state.
Overall, with SEC-ESI-MS, the effect of nonspecific interactions between
protein and stationary phase on the protein structure can be studied,
even revealing gradual structural differences along a peak.
Collapse
Affiliation(s)
- Iro K Ventouri
- Division of Bioanalytical Chemistry, AIMMS Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands.,Centre for Analytical Sciences Amsterdam, 1098XH Amsterdam, The Netherlands.,TI-COAST, 1098 XH Amsterdam, The Netherlands.,Analytical Chemistry Group, van't Hoff Institute for Molecular Sciences, University of Amsterdam, PO Box 94720, 1090 GE Amsterdam, The Netherlands
| | - Daniel B A Malheiro
- Division of Bioanalytical Chemistry, AIMMS Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands.,TI-COAST, 1098 XH Amsterdam, The Netherlands
| | - Robert L C Voeten
- Division of Bioanalytical Chemistry, AIMMS Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands.,Centre for Analytical Sciences Amsterdam, 1098XH Amsterdam, The Netherlands.,TI-COAST, 1098 XH Amsterdam, The Netherlands
| | - Sander Kok
- DSM Materials Science Center, 6167 RD Geleen, The Netherlands
| | - Maarten Honing
- Division of Bioanalytical Chemistry, AIMMS Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands.,DSM Materials Science Center, 6167 RD Geleen, The Netherlands
| | - Govert W Somsen
- Division of Bioanalytical Chemistry, AIMMS Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands.,Centre for Analytical Sciences Amsterdam, 1098XH Amsterdam, The Netherlands
| | - Rob Haselberg
- Division of Bioanalytical Chemistry, AIMMS Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands.,Centre for Analytical Sciences Amsterdam, 1098XH Amsterdam, The Netherlands
| |
Collapse
|
50
|
Analytical challenges and advancements in bioanalysis of therapeutic proteins. Bioanalysis 2020; 12:207-209. [DOI: 10.4155/bio-2020-0006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|