1
|
Schairer J, Römer J, Neusüß C. CE-MS and CE-MS/MS for the multiattribute analysis of monoclonal antibody variants at the subunit level. J Pharm Biomed Anal 2025; 252:116495. [PMID: 39368136 DOI: 10.1016/j.jpba.2024.116495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/21/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
The analysis of product-related substances and impurities is a critical step in the biopharmaceutical quality control of multiattribute monoclonal antibodies (mAbs), as posttranslational modifications or other variants can influence the product's biological activity. Many approaches are available for variant analysis; however, they are either variant-specific, mAb-specific, time-consuming, or require expensive equipment. Here, we present a generic capillary electrophoretic method based on a neutral-coated capillary which was coupled to mass spectrometry (MS) via the nanoCEasy interface for mAb variant analysis at the subunit level (enzymatically digested and reduced mAb). The method enabled the separation of several (i) size variants (e.g. glycosylation variants) and (ii) charge variants (e.g. c-terminal lysin clipping) as well as (iii) multiple other proteoforms (e.g. additional glycation) and (iv) incompletely reduced subunits. Separated variants were confirmed by MS/MS fragmentation even for small mass deviations like deamidation or open disulfide bridges. The system, initially developed for one mAb, was tested with nine other IgG1s to show the general applicability of the system. The presented multiattribute method enables fast and detailed characterization of mAb variants with little sample preparation and relatively simple separation equipment enabling the separation of a large set of mAb variants.
Collapse
Affiliation(s)
- Jasmin Schairer
- Faculty of Chemistry, Aalen University, Beethovenstraße 1, Aalen 73430, Germany; Faculty of Science, University of Tübingen, Auf der Morgenstelle 8, Tübingen 72076, Germany
| | - Jennifer Römer
- Rentschler Biopharma SE, Erwin-Rentschler-Straße 21, Laupheim 88471, Germany
| | - Christian Neusüß
- Faculty of Chemistry, Aalen University, Beethovenstraße 1, Aalen 73430, Germany.
| |
Collapse
|
2
|
Dykstra AB, Lubinsky TG, Vitrac H, Campuzano IDG, Bondarenko PV, Simone AR. Utilization of Liquid Chromatography-Mass Spectrometry and High-Resolution Ion Mobility-Mass Spectrometry to Characterize Therapeutically Relevant Peptides with Asparagine Deamidation and Isoaspartate. Anal Chem 2024. [PMID: 39714115 DOI: 10.1021/acs.analchem.4c05246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Rapid identification of asparagine (Asn) deamidation and isoaspartate (isoAsp) in proteins remains a challenging analytical task during the development of biological therapeutics. For this study, 46 therapeutically relevant peptides corresponding to 13 peptide families (13 unmodified peptides and 33 modified peptides) were obtained; modified peptides included Asn deamidation and isoAsp. The peptide families were characterized by three methods: reversed-phase ultrahigh performance liquid chromatography-mass spectrometry (RP-UHPLC-MS); flow injection analysis high-resolution ion mobility-mass spectrometry (FIA-HRIM-MS); and shortened gradient RP-UHPLC-HRIM-MS. UHPLC-MS data acquisition was 2 h per injection, in contrast to high-throughput 1 min data acquisition of the FIA-HRIM-MS technique. A rapid 2D peptide map has been demonstrated by combining shortened gradient RP-UHPLC with HRIM, to optimize the resolution of the Asn-, Asp-, and isoAsp-containing peptides, increasing the likelihood of detecting peptides containing these quality attributes with expedited data acquisition. Additionally, this paper provides an ion mobility calibration data set for therapeutically relevant peptides (unmodified and modified) over an ion-neutral collisional cross-section range of 300-800 Å2.
Collapse
Affiliation(s)
- Andrew B Dykstra
- Pre-Pivotal Attribute Sciences, Amgen, Inc, Thousand Oaks, California 91320, United States
| | | | - Heidi Vitrac
- MOBILion Systems, Chadds Ford, Pennsylvania 19317, United States
| | - Iain D G Campuzano
- Molecular Analytics, Amgen, Inc, Thousand Oaks, California 91320, United States
| | - Pavel V Bondarenko
- Pre-Pivotal Attribute Sciences, Amgen, Inc, Thousand Oaks, California 91320, United States
| | - Ashli R Simone
- MOBILion Systems, Chadds Ford, Pennsylvania 19317, United States
| |
Collapse
|
3
|
van der Zon AAM, Höchsmann A, Bos TS, Neusüß C, Somsen GW, Jooß K, Haselberg R, Gargano AFG. Characterization of monoclonal antibody charge variants under near-native separation conditions using nanoflow sheath liquid capillary electrophoresis-mass spectrometry. Anal Chim Acta 2024; 1331:343287. [PMID: 39532401 DOI: 10.1016/j.aca.2024.343287] [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: 07/19/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Monoclonal antibodies (mAbs) undergo multiple post-translational modifications (PTMs) during production and storage, resulting for instance in charge and oxidized variants. PTMs need to be assessed as critical quality attributes to assure protein quality and safety. Capillary zone electrophoresis (CZE) enables efficient charge-based separation. The CZE method developed by He et al. (2011) is currently applied routinely in the pharmaceutical industry for profiling charge heterogeneity of mAbs. However, as the method relies on a non-volatile background electrolyte (BGE), it cannot be directly hyphenated with mass spectrometry (MS), hampering the identification of separated charge variants. RESULTS This study presents a CZE-UV/MS method using a neutral static capillary coating of hydroxypropyl methylcellulose combined with a volatile BGE at pH 5.0 to allow for MS-compatible mAb charge variant separations. The effect of several parameters, including pH and concentration of the BGE, applied voltage, and injected mAb concentrations on separation performance was investigated using a panel of commercially available mAbs. The optimized method was evaluated with IgG1 and IgG4 mAbs of varying pI (7.4-9.2) and degrees of heterogeneity. Basic and acidic variants were separated from the parent mAb using a BGE of 50 mM acetic acid adjusted to pH 5.0 with ammonium hydroxide. The relative abundances of charge variants determined with the new method showed a good correlation with the corresponding relative levels obtained with the method of He et al. CZE-MS coupling was accomplished using the nanoCEasy, a low-flow sheath liquid interface, which enabled the identification and quantitation of basic, acidic, and incomplete pyroglutamate variants, and glycoforms of the tested mAbs. SIGNIFICANCE This manuscript describes a new CZE-MS method that permits heterogeneity assessment of mAbs under MS-compatible conditions, providing charge variant separation.
Collapse
Affiliation(s)
- Annika A M van der Zon
- University of Amsterdam, van 't Hoff Institute for Molecular Sciences, Analytical Chemistry Group, Science Park 904, 1098 XH, Amsterdam, the Netherlands; Centre of Analytical Sciences Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands.
| | - Alisa Höchsmann
- Aalen University, Department of Chemistry, Beethovenstraße 1, 73430, Aalen, Germany; Eberhard Karls University of Tübingen, Faculty of Science, 72074, Tübingen, Germany
| | - Tijmen S Bos
- University of Amsterdam, van 't Hoff Institute for Molecular Sciences, Analytical Chemistry Group, Science Park 904, 1098 XH, Amsterdam, the Netherlands; Centre of Analytical Sciences Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands
| | - Christian Neusüß
- Aalen University, Department of Chemistry, Beethovenstraße 1, 73430, Aalen, Germany
| | - Govert W Somsen
- Centre of Analytical Sciences Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands; Vrije Universiteit Amsterdam, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, De Boelelaan 1085, 1081 HV, Amsterdam, the Netherlands
| | - Kevin Jooß
- Centre of Analytical Sciences Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands; Vrije Universiteit Amsterdam, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, De Boelelaan 1085, 1081 HV, Amsterdam, the Netherlands.
| | - Rob Haselberg
- Centre of Analytical Sciences Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands; Vrije Universiteit Amsterdam, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, De Boelelaan 1085, 1081 HV, Amsterdam, the Netherlands.
| | - Andrea F G Gargano
- University of Amsterdam, van 't Hoff Institute for Molecular Sciences, Analytical Chemistry Group, Science Park 904, 1098 XH, Amsterdam, the Netherlands; Centre of Analytical Sciences Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands.
| |
Collapse
|
4
|
Yang Y, Dalvie NC, Brady JR, Naranjo CA, Lorgeree T, Rodriguez-Aponte SA, Johnston RS, Tracey MK, Elenberger CM, Lee E, Tié M, Love KR, Love JC. Adaptation of Aglycosylated Monoclonal Antibodies for Improved Production in Komagataella phaffii. Biotechnol Bioeng 2024. [PMID: 39543843 DOI: 10.1002/bit.28878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/17/2024] [Accepted: 10/26/2024] [Indexed: 11/17/2024]
Abstract
Monoclonal antibodies (mAbs) are a major class of biopharmaceuticals manufactured by well-established processes using Chinese Hamster Ovary (CHO) cells. Next-generation biomanufacturing using alternative hosts like Komagataella phaffii could improve the accessibility of these medicines, address broad societal goals for sustainability, and offer financial advantages for accelerated development of new products. Antibodies produced by K. phaffii, however, may manifest unique molecular quality attributes, like host-dependent, product-related variants, that could raise potential concerns for clinical use. We demonstrate here conservative modifications to the amino acid sequence of aglycosylated antibodies based on the human IgG1 isotype that minimize product-related variations when secreted by K. phaffii. A combination of 2-3 changes of amino acids reduced variations across six different aglycosylated versions of commercial mAbs. Expression of a modified sequence of NIST mAb in both K. phaffii and CHO cells showed comparable biophysical properties and molecular variations. These results suggest a path toward the production of high-quality mAbs that could be expressed interchangeably by either yeast or mammalian cells. Improving molecular designs of proteins to enable a range of manufacturing strategies for well-characterized biopharmaceuticals could accelerate global accessibility and innovations.
Collapse
Affiliation(s)
- Yuchen Yang
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Neil C Dalvie
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Joseph R Brady
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Christopher A Naranjo
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Timothy Lorgeree
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Sergio A Rodriguez-Aponte
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Ryan S Johnston
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Mary K Tracey
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Carmen M Elenberger
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Eric Lee
- Biogen, Cambridge, Massachusetts, USA
| | - Mark Tié
- Biogen, Cambridge, Massachusetts, USA
| | - Kerry R Love
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - J Christopher Love
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
5
|
Verscheure L, Vandenheede I, De Rore E, Meersseman M, Hanssens V, Meerschaert K, Stals H, Sandra P, Lynen F, Borgions F, Sandra K. 2D-CEX-FcRn-MS to Study Structure/Function Relation of mAb Charge Variants. Anal Chem 2024; 96:18122-18131. [PMID: 39470991 DOI: 10.1021/acs.analchem.4c04158] [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: 11/01/2024]
Abstract
The automated elucidation of the interplay between monoclonal antibody (mAb) structure and function using two-dimensional liquid chromatography-mass spectrometry (2D-LC-MS) is reported. Charge variants, induced through forced degradation, are resolved by first-dimension (1D) cation-exchange chromatography (CEX) and subsequently collected in loops installed on a multiple heart-cutting valve prior to transfer to second-dimension (2D) neonatal crystallizable fragment receptor (FcRn) affinity chromatography coupled with MS. As such, binding affinity of the latter mAb variants can elegantly be assessed and a first glimpse of identity provided. To maximize MS sensitivity, charge variants are unfolded upon eluting from the 2D affinity column by postcolumn addition of a denaturing solution. Further structural details, i.e., modification sites and chain distribution, are unraveled by a multidimensional LC-MS (mD-LC-MS) setup incorporating 1D CEX and parallel online middle-up and bottom-up LC-MS analysis in the subsequent dimensions. Identified charge variants could be ranked according to their affinity for FcRn. Binding is predominantly impacted by heavy chain (HC) M253 oxidation and to a lesser extend, M429 oxidation. Oxidation of both HCs more drastically affects FcRn interaction compared to single-chain oxidation, and the more oxidation, the less binding. Other modifications, such as HC glycosylation, HC N385/390, and N326 deamidation or HC C-terminal processing, are not shown to affect binding. The streamlined platform is challenged against the established workflow involving offline collection of charge variants and structural and functional assessment by, respectively, LC-MS and enzyme-linked immunosorbent assay (ELISA). A decent correlation is demonstrated between the binding affinity measured with ELISA and 2D FcRn affinity chromatography. In addition, throughput is improved (7-fold), material requirements are substantially reduced (2 orders of magnitude), and sample preparation artifacts and loss are minimized. With the simultaneous determination of mAb structure and function, the current study takes the concept of multiattribute analysis to the next level, thereby contributing to the future development of safer and more effective antibody therapeutics.
Collapse
Affiliation(s)
- Liesa Verscheure
- RIC group, President Kennedypark 26, Kortrijk B-8500, Belgium
- Separation Science Group, Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281-S4, Ghent B-9000, Belgium
| | | | - Eline De Rore
- RIC group, President Kennedypark 26, Kortrijk B-8500, Belgium
| | | | | | | | - Hilde Stals
- Argenx, Industriepark Zwijnaarde 7, Ghent B-9052, Belgium
| | - Pat Sandra
- RIC group, President Kennedypark 26, Kortrijk B-8500, Belgium
- Separation Science Group, Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281-S4, Ghent B-9000, Belgium
| | - Frederic Lynen
- Separation Science Group, Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281-S4, Ghent B-9000, Belgium
| | - Filip Borgions
- Argenx, Industriepark Zwijnaarde 7, Ghent B-9052, Belgium
| | - Koen Sandra
- RIC group, President Kennedypark 26, Kortrijk B-8500, Belgium
- Separation Science Group, Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281-S4, Ghent B-9000, Belgium
| |
Collapse
|
6
|
Webb J, Niu C, Ritter B, Albarghouthi M, Chen X, Wang C. Developing analytical ion exchange chromatography methods for antibody drug conjugates containing the hydrolysis-prone succinimide-thioether conjugation chemistry. J Pharm Sci 2024; 113:3279-3285. [PMID: 39182845 DOI: 10.1016/j.xphs.2024.08.021] [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: 03/09/2024] [Revised: 08/15/2024] [Accepted: 08/15/2024] [Indexed: 08/27/2024]
Abstract
Charge variants are one of the most important quality attributes for protein therapeutics, including antibody drug conjugates (ADCs). ADCs are conjugation products between monoclonal antibodies (mAbs) and highly potent payloads. After attaching a payload, the charge profile of a mAb can be modified due to the change in net charge or surface charge. In this study, we present a unique challenge of charge assay development that arises from a desirable engineering of ADCs that incorporates the hydrolysis-prone succinimide-thioether conjugation chemistry. This engineered hydrolysis at conjugation sites is usually not complete during conjugation process and continuously progressing during mild stress. This hydrolysis also creates a carboxylic functional group, which manifests as acidic peaks in the ADC charge profiles. As a result, ion exchange chromatograms become sensitive measurements of this hydrolysis, which often masks the charge profile change due to other important post-translational modifications. In this study, two approaches were explored to address this unique challenge: to remove the hydrolysis heterogeneity by incubating ADCs under high pH conditions to drive complete hydrolysis; and to analyze charge variants at the subunit level after IdeS digestion. Acceptable charge profiles and quantitative integration results were successfully obtained by both approaches.
Collapse
Affiliation(s)
- Jessica Webb
- Department of Analytical Sciences, AstraZeneca, One Medimmune Way, Gaithersburg, MD, 20878, USA
| | - Chendi Niu
- Department of Analytical Sciences, AstraZeneca, One Medimmune Way, Gaithersburg, MD, 20878, USA
| | - Benjamin Ritter
- Department of Analytical Sciences, AstraZeneca, One Medimmune Way, Gaithersburg, MD, 20878, USA
| | - Methal Albarghouthi
- Department of Analytical Sciences, AstraZeneca, One Medimmune Way, Gaithersburg, MD, 20878, USA
| | - Xiaoyu Chen
- Department of Analytical Sciences, AstraZeneca, One Medimmune Way, Gaithersburg, MD, 20878, USA
| | - Chunlei Wang
- Department of Analytical Sciences, AstraZeneca, One Medimmune Way, Gaithersburg, MD, 20878, USA.
| |
Collapse
|
7
|
Zheng X, Fang M, Zou Y, Wang S, Zhou W, Zhou H. A comparison of different intensified upstream processes highlighting the advantage of WuXi Biologics' Ultra-high Productivity platform (WuXiUP TM) in improved product quality and purification yield. Biotechnol Prog 2024; 40:e3487. [PMID: 38980213 DOI: 10.1002/btpr.3487] [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: 12/28/2023] [Revised: 05/17/2024] [Accepted: 05/29/2024] [Indexed: 07/10/2024]
Abstract
WuXiUPTM, WuXi Biologics' Ultra-high Productivity platform, is an intensified and integrated continuous bioprocess platform developed for production of various biologics including monoclonal antibodies, fusion proteins, and bispecific antibodies. This process technology platform has manifested its remarkable capability in boosting the volumetric productivity of various biologics and has been implemented for large-scale clinical material productions. In this paper, case studies of the production of different pharmaceutical proteins using two high-producing and intensified culture modes of WuXiUPTM and the concentrated fed-batch (CFB), as well as the traditional fed-batch (TFB) are discussed from the perspectives of cell growth, productivity, and protein quality. Both WuXiUPTM and CFB outperformed TFB regarding volumetric productivity. Additionally, distinctive advantages in product quality profiles in the WuXiUPTM process, such as reduced acidic charge variants and fragmentation, are revealed. Therefore, a simplified downstream purification process with only two chromatographic steps can be developed to deliver the target product at a satisfactory purity and an extremely-high yield.
Collapse
Affiliation(s)
- Xiang Zheng
- Cell Culture Process Development, WuXi Biologics, Shanghai, China
| | - Mingyue Fang
- Non-GMP Pilot Plant, WuXi Biologics, Shanghai, China
| | - Yanling Zou
- Manufacturing Facility Group 17, WuXi Biologics, Shanghai, China
| | - Shuo Wang
- Downstream Process Development, WuXi Biologics, Shanghai, China
| | - Weichang Zhou
- Cell Culture Process Development, WuXi Biologics, Shanghai, China
- Non-GMP Pilot Plant, WuXi Biologics, Shanghai, China
- Manufacturing Facility Group 17, WuXi Biologics, Shanghai, China
- Downstream Process Development, WuXi Biologics, Shanghai, China
| | - Hang Zhou
- Cell Culture Process Development, WuXi Biologics, Shanghai, China
- Non-GMP Pilot Plant, WuXi Biologics, Shanghai, China
- Downstream Process Development, WuXi Biologics, Shanghai, China
| |
Collapse
|
8
|
Liang G, Sha S, Wang Z, Liu H, Yoon S. Soft-sensor model development for CHO growth/production, intracellular metabolite, and glycan predictions. Front Mol Biosci 2024; 11:1441885. [PMID: 39502716 PMCID: PMC11535473 DOI: 10.3389/fmolb.2024.1441885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/30/2024] [Indexed: 11/08/2024] Open
Abstract
Efficaciously assessing product quality remains time- and resource-intensive. Online Process Analytical Technologies (PATs), encompassing real-time monitoring tools and soft-sensor models, are indispensable for understanding process effects and real-time product quality. This research study evaluated three modeling approaches for predicting CHO cell growth and production, metabolites (extracellular, nucleotide sugar donors (NSD) and glycan profiles): Mechanistic based on first principle Michaelis-Menten kinetics (MMK), data-driven orthogonal partial least square (OPLS) and neural network machine learning (NN). Our experimental design involved galactose-fed batch cultures. MMK excelled in predicting growth and production, demonstrating its reliability in these aspects and reducing the data burden by requiring fewer inputs. However, it was less precise in simulating glycan profiles and intracellular metabolite trends. In contrast, NN and OPLS performed better for predicting precise glycan compositions but displayed shortcomings in accurately predicting growth and production. We utilized time in the training set to address NN and OPLS extrapolation challenges. OPLS and NN models demanded more extensive inputs with similar intracellular metabolite trend prediction. However, there was a significant reduction in time required to develop these two models. The guidance presented here can provide valuable insight into rapid development and application of soft-sensor models with PATs for ipurposes. Therefore, we examined three model typesmproving real-time product CHO therapeutic product quality. Coupled with emerging -omics technologies, NN and OPLS will benefit from massive data availability, and we foresee more robust prediction models that can be advantageous to kinetic or partial-kinetic (hybrid) models.
Collapse
Affiliation(s)
| | | | | | | | - Seongkyu Yoon
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, MA, United States
| |
Collapse
|
9
|
Oates RN, Lieu LB, Srzentić K, Damoc E, Fornelli L. Characterization of a Monoclonal Antibody by Native and Denaturing Top-Down Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:2197-2208. [PMID: 39105725 DOI: 10.1021/jasms.4c00224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
Established in recent years as an important approach to unraveling the heterogeneity of intact monoclonal antibodies, native mass spectrometry has been rarely utilized for sequencing these complex biomolecules via tandem mass spectrometry. Typically, top-down mass spectrometry has been performed starting from highly charged precursor ions obtained via electrospray ionization under denaturing conditions (i.e., in the presence of organic solvents and acidic pH). Here we systematically benchmark four distinct ion dissociation methods─namely, higher-energy collisional dissociation, electron transfer dissociation, electron transfer dissociation/higher-energy collisional dissociation, and 213 nm ultraviolet photodissociation─in their capability to characterize a therapeutic monoclonal antibody, trastuzumab, starting from denatured and native-like precursor ions. Interestingly, native top-down mass spectrometry results in higher sequence coverage than the experiments carried out under denaturing conditions, with the exception of ultraviolet photodissociation. Globally, electron transfer dissociation followed by collision-based activation of product ions generates the largest number of backbone cleavages in disulfide protected regions, including the complementarity determining regions, regardless of electrospray ionization conditions. Overall, these findings suggest that native mass spectrometry can certainly be used for the gas-phase sequencing of whole monoclonal antibodies, although the dissociation of denatured precursor ions still returns a few backbone cleavages not identified in native experiments. Finally, a comparison of the fragmentation maps obtained under denaturing and native conditions strongly points toward disulfide bonds as the primary reason behind the largely overlapping dissociation patterns.
Collapse
Affiliation(s)
- Ryan N Oates
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Linda B Lieu
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | | | - Eugen Damoc
- Thermo Fisher Scientific, Bremen, DE-HB 28199, Germany
| | - Luca Fornelli
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
- School of Biological Sciences, University of Oklahoma, Norman, Oklahoma 73019, United States
| |
Collapse
|
10
|
Capito F, Wong TH, Faust C, Brand K, Dittrich W, Sommerfeld M, Tiwari G, Langer T. Improving Downstream Process Related Manufacturability Based on Protein Engineering-A Feasibility Study. Eng Life Sci 2024; 24:e202400019. [PMID: 39233725 PMCID: PMC11369322 DOI: 10.1002/elsc.202400019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/02/2024] [Accepted: 06/04/2024] [Indexed: 09/06/2024] Open
Abstract
While bioactivity and a favorable safety profile for biotherapeutics is of utmost importance, manufacturability is also worth of consideration to ease the manufacturing process. Manufacturability in the scientific literature is mostly related to stability of formulated drug substances, with limited focus on downstream process-related manufacturability, that is, how easily can a protein be purified. Process-related impurities or biological impurities like viruses and host cell proteins (HCP) are present in the harvest which have mostly acid isoelectric points and need to be removed to ensure patient safety. Therefore, during molecule design, the surface charge of the target molecule should preferably differ sufficiently from the surface charge of the impurities to enable an efficient purification strategy. In this feasibility study, we evaluated the possibility of improving manufacturability by adapting the surface charge of the target protein. We generated several variants of a GLP1-receptor-agonist-Fc-domain-FGF21-fusion protein and demonstrated proof of concept exemplarily for an anion exchange chromatography step which then can be operated at high pH values with maximal product recovery allowing removal of HCP and viruses. Altering the surface charge distribution of biotherapeutic proteins can thus be useful allowing for an efficient manufacturing process for removing HCP and viruses, thereby reducing manufacturing costs.
Collapse
Affiliation(s)
- Florian Capito
- Sanofi‐Aventis Deutschland GmbH, CMC Microbial Platform Downstream Process DevelopmentIndustriepark HöchstFrankfurt am MainGermany
| | - Ting Hin Wong
- Sanofi‐Aventis Deutschland GmbH, CMC Microbial Platform Downstream Process DevelopmentIndustriepark HöchstFrankfurt am MainGermany
- Technische Hochschule MittelhessenGießenGermany
| | - Christine Faust
- Sanofi‐Aventis Deutschland GmbH, Large Molecule ResearchIndustriepark HöchstFrankfurt am MainGermany
| | - Kilian Brand
- Sanofi‐Aventis Deutschland GmbH, CMC Microbial Platform Downstream Process DevelopmentIndustriepark HöchstFrankfurt am MainGermany
- Sanofi‐Aventis Deutschland GmbH, ICF API MIB FISIndustriepark HöchstFrankfurt am MainGermany
| | - Werner Dittrich
- Sanofi‐Aventis Deutschland GmbH, Large Molecule ResearchIndustriepark HöchstFrankfurt am MainGermany
| | - Mark Sommerfeld
- Sanofi‐Aventis Deutschland GmbH, Large Molecule ResearchIndustriepark HöchstFrankfurt am MainGermany
| | - Garima Tiwari
- Sanofi‐Aventis Deutschland GmbH, Large Molecule ResearchIndustriepark HöchstFrankfurt am MainGermany
| | - Thomas Langer
- Sanofi‐Aventis Deutschland GmbH, Large Molecule ResearchIndustriepark HöchstFrankfurt am MainGermany
| |
Collapse
|
11
|
Sutton H, Hong F, Han X, Rauniyar N. Analysis of therapeutic monoclonal antibodies by imaged capillary isoelectric focusing (icIEF). ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:5450-5458. [PMID: 39042476 DOI: 10.1039/d4ay00836g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
Imaged capillary isoelectric focusing (icIEF) is a preferred analytical method for determining isoelectric points (pIs) and charge heterogeneity profiles in biotherapeutic proteins. In this study, we optimized the icIEF method for an in-house IgG1κ monoclonal antibody (mAb-1) and assessed its reproducibility, robustness, and autosampler stability. The optimized method was used to evaluate batch-to-batch consistency in pIs for multiple lots of mAb-1 and determine the relative percentages of charge variants. We also tested the method's performance using multiple lots of another IgG1 mAb, commercially available as Herceptin (trastuzumab). Additionally, we designed and assessed native and denaturing platform icIEF methods for 11 other marketed mAbs, with pIs ranging from 6.0 (eculizumab) to 9.22 (tocilizumab).
Collapse
Affiliation(s)
- Haley Sutton
- Tanvex Biopharma USA, Inc., 10394 Pacific Center Ct, San Diego, CA 92121, USA.
| | - Feng Hong
- Tanvex Biopharma USA, Inc., 10394 Pacific Center Ct, San Diego, CA 92121, USA.
| | - Xuemei Han
- Tanvex Biopharma USA, Inc., 10394 Pacific Center Ct, San Diego, CA 92121, USA.
| | - Navin Rauniyar
- Tanvex Biopharma USA, Inc., 10394 Pacific Center Ct, San Diego, CA 92121, USA.
| |
Collapse
|
12
|
Divase A, Pisal S, Dake MS, Dakshinamurthy PK, Reddy PS, Dhere R, Kamat C, Chahar DS, Pal J, Nawani N. Isolation and characterization of rabies monoclonal antibody charge variants. Electrophoresis 2024; 45:1339-1355. [PMID: 38700202 DOI: 10.1002/elps.202300221] [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: 10/08/2023] [Revised: 01/20/2024] [Accepted: 03/02/2024] [Indexed: 05/05/2024]
Abstract
Current postexposure prophylaxis of rabies includes vaccines, human rabies immunoglobulin (RIG), equine RIG, and recombinant monoclonal antibodies (mAb). In the manufacturing of rabies recombinant mAb, charge variants are the most common source of heterogeneity. Charge variants of rabies mAb were isolated by salt gradient cation exchange chromatography (CEX) to separate acidic and basic and main charge variants. Separated variants were further extensively characterized using orthogonal analytical techniques, which include secondary and tertiary structure determination by far and near ultraviolet circular dichroism spectroscopy. Charge and size heterogeneity were evaluated using CEX, isoelectric focusing (IEF), capillary-IEF, size exclusion chromatography, sodium dodecyl sulfate polyacrylamide gel electrophoresis, and western blotting. Antigen binding affinity was assessed by enzyme linked immuno-sorbent assay and rapid florescence foci inhibition test. Results from structural and physicochemical characterizations concluded that charge variants are formed due to posttranslational modification demonstrating that the charge heterogeneity, these charge variants did neither show any considerable physicochemical change nor affect its biological function. This study shows that charge variants are effective components of mAb and there is no need of deliberate removal, until biological functions of rabies mAb will get affected.
Collapse
Affiliation(s)
- Ambika Divase
- Serum Institute of India Pvt. Ltd. Hadapsar, Pune, Maharashtra, India
- Biotechnology Department, Dr. D.Y. Patil Biotechnology and Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Sambhaji Pisal
- Serum Institute of India Pvt. Ltd. Hadapsar, Pune, Maharashtra, India
| | - Manjusha Sudhakar Dake
- Biotechnology Department, Dr. D.Y. Patil Biotechnology and Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | | | | | - Rajeev Dhere
- Serum Institute of India Pvt. Ltd. Hadapsar, Pune, Maharashtra, India
| | | | | | - Jayanta Pal
- Biotechnology Department, Dr. D.Y. Patil Biotechnology and Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Neelu Nawani
- Biotechnology Department, Dr. D.Y. Patil Biotechnology and Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| |
Collapse
|
13
|
Isu S, Vinskus L, Silva D, Cunningham K, Elich T, Greenhalgh P, Sokolnicki A, Raghunath B. Leveraging bioanalytical characterization of fractionated monoclonal antibody pools to identify aggregation-prone and less filterable proteoforms during virus filtration. Biotechnol Prog 2024; 40:e3451. [PMID: 38450976 DOI: 10.1002/btpr.3451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 01/11/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024]
Abstract
Monoclonal antibodies (mAbs) are an essential class of biotherapeutics. A platform process is used for mAb development to ensure clinically safe and stable molecules. Regulatory authorities ensure that mAb production processes include sufficient viral clearance steps to achieve less than one virus particle per million doses of product. Virus filtration is used for size-based removal of enveloped and nonenveloped viruses during downstream processing of mAbs. Process development in mAb purification relies on empirical approaches and often includes adsorptive prefiltration to mitigate virus filter fouling. Opportunities for molecular-level prediction of mAb filterability are needed to plug the existing knowledge gap in downstream processing. A molecular-level approach to understanding the factors influencing mAb filterability may reduce process development time, material loss, and processing costs due to oversized virus filters. In this work, pH step gradient fractionation was applied on polished bulk mAb feed to obtain concentrated pools of fractionated mAb variants. Biophysical properties and quality attributes of fractionated pools, including oligomeric state (size), isoelectric point profile, diffusion interaction parameters, and glycoform profile, were determined using bioanalytical methods. Filterability (loading and throughput) of fractionated pools were evaluated. Statistical methods were used to obtain correlations between quality attributes of mAb fractions and filterability on the Viresolve Pro virus filter.
Collapse
Affiliation(s)
- Solomon Isu
- Process Solutions, MilliporeSigma, Burlington, Massachusetts, USA
| | - Lilia Vinskus
- Process Solutions, MilliporeSigma, Burlington, Massachusetts, USA
| | - Derek Silva
- Process Solutions, MilliporeSigma, Burlington, Massachusetts, USA
| | | | - Thomas Elich
- Process Solutions, MilliporeSigma, Burlington, Massachusetts, USA
| | | | - Adam Sokolnicki
- Process Solutions, MilliporeSigma, Burlington, Massachusetts, USA
| | - Bala Raghunath
- Process Solutions, MilliporeSigma, Burlington, Massachusetts, USA
| |
Collapse
|
14
|
Hsieh MC, Zhang J, Tang L, Huang CY, Shen Y, Matathia A, Qian J, Parekh BS. Characterization of the Charge Heterogeneity of a Monoclonal Antibody That Binds to Both Cation Exchange and Anion Exchange Columns under the Same Binding Conditions. Antibodies (Basel) 2024; 13:52. [PMID: 39051328 PMCID: PMC11270306 DOI: 10.3390/antib13030052] [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: 05/31/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Therapeutic antibodies play an important role in the public healthcare system to treat patients with a variety of diseases. Protein characterization using an array of analytical tools provides in-depth information for drug quality, safety, efficacy, and the further understanding of the molecule. A therapeutic antibody candidate MAB1 exhibits unique binding properties to both cation and anion exchange columns at neutral pH. This uniqueness disrupts standard purification processes and necessitates adjustments in manufacturing. This study identifies that the charge heterogeneity of MAB1 is primarily due to the N-terminal cyclization of glutamine to pyroglutamine and, to a lesser extent, succinimide intermediate, deamidation, and C-terminal lysine. Using three approaches, i.e., deferential chemical labeling, H/D exchange, and molecular modeling, the binding to anion exchange resins is attributed to negatively charged patches on the antibody's surface, involving specific carboxylic acid residues. The methodologies shown here can be extended to study protein binding orientation in column chromatography.
Collapse
Affiliation(s)
- Ming-Ching Hsieh
- Analytical Sciences, Eli Lilly and the Company, Branchburg, NJ 08876, USA
| | - Jingming Zhang
- Analytical Sciences, Eli Lilly and the Company, Branchburg, NJ 08876, USA
| | - Liangjie Tang
- Analytical Development, Eli Lilly and the Company, Indianapolis, IN 46221, USA
| | - Cheng-Yen Huang
- Analytical Sciences, Eli Lilly and the Company, Branchburg, NJ 08876, USA
| | - Yang Shen
- Antibody Technology, Eli Lilly and the Company, New York, NY 10016, USA
| | - Alice Matathia
- TS/MS Laboratories, Eli Lilly and the Company, Branchburg, NJ 08876, USA
| | - Jun Qian
- Analytical Development, Eli Lilly and the Company, Indianapolis, IN 46221, USA
| | | |
Collapse
|
15
|
Malani H, Shrivastava A, Nupur N, Rathore AS. LC-MS Characterization and Stability Assessment Elucidate Correlation Between Charge Variant Composition and Degradation of Monoclonal Antibody Therapeutics. AAPS J 2024; 26:42. [PMID: 38570351 DOI: 10.1208/s12248-024-00915-9] [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: 01/15/2024] [Accepted: 03/19/2024] [Indexed: 04/05/2024] Open
Abstract
Aggregation stability of monoclonal antibody (mAb) therapeutics is influenced by many critical quality attributes (CQA) such as charge and hydrophobic variants in addition to environmental factors. In this study, correlation between charge heterogeneity and stability of mAbs for bevacizumab and trastuzumab has been investigated under a variety of stresses including thermal stress at 40 °C, thermal stress at 55 °C, shaking (mechanical), and low pH. Size- and charge-based heterogeneities were monitored using analytical size exclusion chromatography (SEC) and cation exchange chromatography (CEX), respectively, while dynamic light scattering was used to assess changes in hydrodynamic size. CEX analysis revealed an increase in cumulative acidic content for all variants of both mAbs post-stress treatment attributed to increased deamidation. Higher charge heterogeneity was observed in variants eluting close to the main peak than the ones eluting further away (25-fold and 42-fold increase in acidic content for main and B1 of bevacizumab and 19-fold for main of trastuzumab, respectively, under thermal stress; 50-fold increase in acidic for main and B1 of bevacizumab and 10% rise in basic content of main of trastuzumab under pH stress). Conversely, variants eluting far away from main exhibit greater aggregation as compared to close-eluting ones. Aggregation kinetics of variants followed different order for the different stresses for both mAbs (2nd order for thermal and pH stresses and 0th order for shaking stress). Half-life of terminal charge variants of both mAbs was 2- to 8-fold less than main indicating increased degradation propensity.
Collapse
Affiliation(s)
- Himanshu Malani
- Department of Chemical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Anuj Shrivastava
- Department of Chemical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Neh Nupur
- Department of Chemical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Anurag S Rathore
- Department of Chemical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India.
| |
Collapse
|
16
|
Liang G, Madhavarao CN, Morris C, O'Connor T, Ashraf M, Yoon S. Effects of process intensification on homogeneity of an IgG1:κ monoclonal antibody during perfusion culture. Appl Microbiol Biotechnol 2024; 108:274. [PMID: 38530495 DOI: 10.1007/s00253-024-13110-9] [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: 11/12/2023] [Revised: 03/01/2024] [Accepted: 03/13/2024] [Indexed: 03/28/2024]
Abstract
The pharmaceutical industry employs various strategies to improve cell productivity. These strategies include process intensification, culture media improvement, clonal selection, media supplementation and genetic engineering of cells. However, improved cell productivity has inherent risk of impacting product quality attributes (PQA). PQAs may affect the products' efficacy via stability, bioavailability, or in vivo bioactivity. Variations in manufacturing process may introduce heterogeneity in the products by altering the type and extent of N-glycosylation, which is a PQA of therapeutic proteins. We investigated the effect of different cell densities representing increasing process intensification in a perfusion cell culture on the production of an IgG1-κ monoclonal antibody from a CHO-K1 cell line. This antibody is glycosylated both on light chain and heavy chain. Our results showed that the contents of glycosylation of IgG1-κ mAb increased in G0F and fucosylated type glycans as a group, whereas sialylated type glycans decreased, for the mAb whole protein. Overall, significant differences were observed in amounts of G0F, G1F, G0, G2FS1, and G2FS2 type glycans across all process intensification levels. G2FS2 and G2 type N-glycans were predominantly quantifiable from light chain rather than heavy chain. It may be concluded that there is a potential impact to product quality attributes of therapeutic proteins during process intensification via perfusion cell culture that needs to be assessed. Since during perfusion cell culture the product is collected throughout the duration of the process, lot allocation needs careful attention to process parameters, as PQAs are affected by the critical process parameters (CPPs). KEY POINTS: • Molecular integrity may suffer with increasing process intensity. • Galactosylated and sialylated N-glycans may decrease. • Perfusion culture appears to maintain protein charge structure.
Collapse
Affiliation(s)
- George Liang
- Division of Product Quality Research, OTR/OPQ, CDER/FDA, Silver Spring, MD, USA
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, MA, USA
| | | | - Caitlin Morris
- Division of Product Quality Research, OTR/OPQ, CDER/FDA, Silver Spring, MD, USA
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, MA, USA
| | - Thomas O'Connor
- Division of Product Quality Research, OTR/OPQ, CDER/FDA, Silver Spring, MD, USA
| | - Muhammad Ashraf
- Division of Product Quality Research, OTR/OPQ, CDER/FDA, Silver Spring, MD, USA
| | - Seongkyu Yoon
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, MA, USA
| |
Collapse
|
17
|
Defant P, Regl C, Huber CG, Schubert M. The NMR signature of maltose-based glycation in full-length proteins. JOURNAL OF BIOMOLECULAR NMR 2024; 78:61-72. [PMID: 38114873 PMCID: PMC10981599 DOI: 10.1007/s10858-023-00432-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/15/2023] [Indexed: 12/21/2023]
Abstract
Reducing sugars can spontaneously react with free amines in protein side chains leading to posttranslational modifications (PTMs) called glycation. In contrast to glycosylation, glycation is a non-enzymatic modification with consequences on the overall charge, solubility, aggregation susceptibility and functionality of a protein. Glycation is a critical quality attribute of therapeutic monoclonal antibodies. In addition to glucose, also disaccharides like maltose can form glycation products. We present here a detailed NMR analysis of the Amadori product formed between proteins and maltose. For better comparison, data collection was done under denaturing conditions using 7 M urea-d4 in D2O. The here presented correlation patterns serve as a signature and can be used to identify maltose-based glycation in any protein that can be denatured. In addition to the model protein BSA, which can be readily glycated, we present data of the biotherapeutic abatacept containing maltose in its formulation buffer. With this contribution, we demonstrate that NMR spectroscopy is an independent method for detecting maltose-based glycation, that is suited for cross-validation with other methods.
Collapse
Affiliation(s)
- Pauline Defant
- Department of Biosciences and Medical Biology, University of Salzburg, Hellbrunnerstrasse 34, 5020, Salzburg, Austria
| | - Christof Regl
- Department of Biosciences and Medical Biology, University of Salzburg, Hellbrunnerstrasse 34, 5020, Salzburg, Austria
| | - Christian G Huber
- Department of Biosciences and Medical Biology, University of Salzburg, Hellbrunnerstrasse 34, 5020, Salzburg, Austria
| | - Mario Schubert
- Department of Biosciences and Medical Biology, University of Salzburg, Hellbrunnerstrasse 34, 5020, Salzburg, Austria.
- Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Takustr. 3, 14195, Berlin, Germany.
| |
Collapse
|
18
|
Schairer J, Römer J, Lang D, Neusüß C. CE-MS/MS and CE-timsTOF to separate and characterize intramolecular disulfide bridges of monoclonal antibody subunits and their application for the assessment of subunit reduction protocols. Anal Bioanal Chem 2024; 416:1599-1612. [PMID: 38296860 PMCID: PMC10899284 DOI: 10.1007/s00216-024-05161-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 02/02/2024]
Abstract
Characterization at the subunit level enables detailed mass spectrometric characterization of posttranslational modifications (PTMs) of monoclonal antibodies (mAbs). The implemented reduction often leaves the intramolecular disulfide bridges intact. Here, we present a capillary electrophoretic (CE) method based on a neutral-coated capillary for the separation of immunoglobulin G-degrading enzyme of Streptococcus pyogenes (IdeS) digested and reduced mAb subunits followed by mass spectrometry (MS), MS/MS identification, and trapped ion mobility mass spectrometry (timsTOF). Our CE approach enables the separation of (i) different subunit moieties, (ii) various reduction states, and (iii) positional isomers of these partly reduced subunit moieties. The location of the remaining disulfide bridges can be determined by middle-down electron transfer higher energy collisional dissociation (EThcD) experiments. All these CE-separated variants show differences in ion mobility in the timsTOF measurements. Applying the presented CE-MS/MS method, reduction parameters such as the use of chaotropic salts were studied. For the investigated antibodies, urea improved the subunit reduction significantly, whereas guanidine hydrochloride (GuHCl) leads to multiple signals of the same subunit in the CE separation. The presented CE-MS method is a powerful tool for the disulfide-variant characterization of mAbs on the subunit level. It enables understanding disulfide bridge reduction processes in antibodies and potentially other proteins.
Collapse
Affiliation(s)
- Jasmin Schairer
- Faculty of Chemistry, Aalen University, Aalen, Germany
- Faculty of Science, University of Tübingen, Tübingen, Germany
| | | | | | | |
Collapse
|
19
|
Hidayah SN, Biabani A, Gaikwad M, Nissen P, Voß H, Riedner M, Schlüter H, Siebels B. Application of sample displacement batch chromatography for fractionation of proteoforms. Proteomics 2024; 24:e2200424. [PMID: 37750450 DOI: 10.1002/pmic.202200424] [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: 03/08/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/27/2023]
Abstract
Fractionation of proteoforms is currently the most challenging topic in the field of proteoform analysis. The need for considering the existence of proteoforms in experimental approaches is not only important in Life Science research in general but especially in the manufacturing of therapeutic proteins (TPs) like recombinant therapeutic antibodies (mAbs). Some of the proteoforms of TPs have significantly decreased actions or even cause side effects. The identification and removal of proteoforms differing from the main species, having the desired action, is challenging because the difference in the composition of atoms is often very small and their concentration in comparison to the main proteoform can be low. In this study, we demonstrate that sample displacement batch chromatography (SDBC) is an easy-to-handle, economical, and efficient method for fractionating proteoforms. As a model sample a commercial ovalbumin fraction was used, containing many ovalbumin proteoforms. The most promising parameters for the SDBC were determined by a screening approach and applied for a 10-segment fractionation of ovalbumin with cation exchange chromatography resins. Mass spectrometry of intact proteoforms was used for characterizing the SDBC fractionation process. By SDBC, a significant separation of different proteoforms was obtained.
Collapse
Affiliation(s)
- Siti Nurul Hidayah
- Section Mass Spectrometric Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada, Sleman, Yogyakarta, Indonesia
| | - Ali Biabani
- Section Mass Spectrometric Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manasi Gaikwad
- Section Mass Spectrometric Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Paula Nissen
- Section Mass Spectrometric Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hannah Voß
- Section Mass Spectrometric Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maria Riedner
- Technology Platform Mass Spectrometry, University of Hamburg, Hamburg, Germany
| | - Hartmut Schlüter
- Section Mass Spectrometric Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bente Siebels
- Section Mass Spectrometric Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
20
|
Li M, Beaumont VA, Akbar S, Duncan H, Creasy A, Wang W, Sackett K, Marzilli L, Rouse JC, Kim HY. Comprehensive characterization of higher order structure changes in methionine oxidized monoclonal antibodies via NMR chemometric analysis and biophysical approaches. MAbs 2024; 16:2292688. [PMID: 38117548 PMCID: PMC10761137 DOI: 10.1080/19420862.2023.2292688] [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: 10/19/2023] [Accepted: 12/05/2023] [Indexed: 12/21/2023] Open
Abstract
The higher order structure (HOS) of monoclonal antibodies (mAbs) is an important quality attribute with strong contribution to clinically relevant biological functions and drug safety. Due to the multi-faceted nature of HOS, the synergy of multiple complementary analytical approaches can substantially improve the understanding, accuracy, and resolution of HOS characterization. In this study, we applied one- and two-dimensional (1D and 2D) nuclear magnetic resonance (NMR) spectroscopy coupled with chemometric analysis, as well as circular dichroism (CD), differential scanning calorimetry (DSC), and fluorescence spectroscopy as orthogonal methods, to characterize the impact of methionine (Met) oxidation on the HOS of an IgG1 mAb. We used a forced degradation method involving concentration-dependent oxidation by peracetic acid, in which Met oxidation is site-specifically quantified by liquid chromatography-mass spectrometry. Conventional biophysical techniques report nuanced results, in which CD detects no change to the secondary structure and little change in the tertiary structure. Yet, DSC measurements show the destabilization of Fab and Fc domains due to Met oxidation. More importantly, our study demonstrates that 1D and 2D NMR and chemometric analysis can provide semi-quantitative analysis of chemical modifications and resolve localized conformational changes with high sensitivity. Furthermore, we leveraged a novel 15N-Met labeling technique of the antibody to directly observe structural perturbations at the oxidation sites. The NMR methods described here to probe HOS changes are highly reliable and practical in biopharmaceutical characterization.
Collapse
Affiliation(s)
- Mingyue Li
- Pfizer, Inc. BioTherapeutics Pharmaceutical Sciences, Analytical Research and Development, Andover, MA, USA
| | - Victor A. Beaumont
- Pfizer, Inc. Pharmaceutical Sciences Small Molecules, Analytical Research and Development, Sandwich, United Kingdom
| | - Shahajahan Akbar
- Pfizer, Inc. BioTherapeutics Pharmaceutical Sciences, Analytical Research and Development, Andover, MA, USA
| | - Hannah Duncan
- Pfizer, Inc. BioTherapeutics Pharmaceutical Sciences, Analytical Research and Development, Andover, MA, USA
| | - Arch Creasy
- Pfizer, Inc. BioTherapeutics Pharmaceutical Sciences, Bioprocess Research and Development, Andover, MA, USA
| | - Wenge Wang
- Pfizer, Inc. BioTherapeutics Pharmaceutical Sciences, Bioprocess Research and Development, Andover, MA, USA
| | - Kelly Sackett
- Pfizer, Inc. BioTherapeutics Pharmaceutical Sciences, Analytical Research and Development, Andover, MA, USA
| | - Lisa Marzilli
- Pfizer, Inc. BioTherapeutics Pharmaceutical Sciences, Analytical Research and Development, Andover, MA, USA
| | - Jason C. Rouse
- Pfizer, Inc. BioTherapeutics Pharmaceutical Sciences, Analytical Research and Development, Andover, MA, USA
| | - Hai-Young Kim
- Pfizer, Inc. BioTherapeutics Pharmaceutical Sciences, Analytical Research and Development, Andover, MA, USA
| |
Collapse
|
21
|
Aebischer-Gumy C, Moretti P, Brunstein Laplace T, Frank J, Grand Y, Mosbaoui F, Hily E, Galea A, Peltret M, Estoppey C, Ayoub D, Giovannini R, Bertschinger M. Alternative splicing for tuneable expression of protein subunits at desired ratios. MAbs 2024; 16:2342243. [PMID: 38650451 PMCID: PMC11042056 DOI: 10.1080/19420862.2024.2342243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 04/09/2024] [Indexed: 04/25/2024] Open
Abstract
The controlled expression of two or more proteins at a defined and stable ratio remains a substantial challenge, particularly in the bi- and multispecific antibody field. Achieving an optimal ratio of protein subunits can facilitate the assembly of multimeric proteins with high efficiency and minimize the production of by-products. In this study, we propose a solution based on alternative splicing, enabling the expression of a tunable and predefined ratio of two distinct polypeptide chains from the same pre-mRNA under the control of a single promoter. The pre-mRNA used in this study contains two open reading frames situated on separate exons. The first exon is flanked by two copies of the chicken troponin intron 4 (cTNT-I4) and is susceptible to excision from the pre-mRNA by means of alternative splicing. This specific design enables the modulation of the splice ratio by adjusting the strength of the splice acceptor. To illustrate this approach, we developed constructs expressing varying ratios of GFP and dsRED and extended their application to multimeric proteins such as monoclonal antibodies, achieving industrially relevant expression levels (>1 g/L) in a 14-day fed-batch process. The stability of the splice ratio was confirmed by droplet digital PCR in a stable pool cultivated over a 28-day period, while product quality was assessed via intact mass analysis, demonstrating absence of product-related impurities resulting from undesired splice events. Furthermore, we showcased the versatility of the construct by expressing two subunits of a bispecific antibody of the BEAT® type, which contains three distinct subunits in total.
Collapse
Affiliation(s)
- Christel Aebischer-Gumy
- Drug Substance Development, Ichnos Sciences SA (formerly Glenmark Pharmaceuticals SA), La Chaux-de-Fonds, Switzerland
| | - Pierre Moretti
- Drug Substance Development, Ichnos Sciences SA (formerly Glenmark Pharmaceuticals SA), La Chaux-de-Fonds, Switzerland
| | - Timothee Brunstein Laplace
- Drug Substance Development, Ichnos Sciences SA (formerly Glenmark Pharmaceuticals SA), La Chaux-de-Fonds, Switzerland
| | - Jana Frank
- Drug Substance Development, Ichnos Sciences SA (formerly Glenmark Pharmaceuticals SA), La Chaux-de-Fonds, Switzerland
| | - Ysaline Grand
- Drug Substance Development, Ichnos Sciences SA (formerly Glenmark Pharmaceuticals SA), La Chaux-de-Fonds, Switzerland
| | - Farid Mosbaoui
- Drug Substance Development, Ichnos Sciences SA (formerly Glenmark Pharmaceuticals SA), La Chaux-de-Fonds, Switzerland
| | - Emilie Hily
- Drug Substance Development, Ichnos Sciences SA (formerly Glenmark Pharmaceuticals SA), La Chaux-de-Fonds, Switzerland
| | - Anna Galea
- Drug Substance Development, Ichnos Sciences SA (formerly Glenmark Pharmaceuticals SA), La Chaux-de-Fonds, Switzerland
| | - Megane Peltret
- Drug Substance Development, Ichnos Sciences SA (formerly Glenmark Pharmaceuticals SA), La Chaux-de-Fonds, Switzerland
| | - Carole Estoppey
- Antibody Engineering, Ichnos Sciences SA (formerly Glenmark Pharmaceuticals SA), La Chaux-de-Fonds, Switzerland
| | - Daniel Ayoub
- Analytical Development, Ichnos Sciences SA (formerly Glenmark Pharmaceuticals SA), La Chaux-de-Fonds, Switzerland
| | - Roberto Giovannini
- Process Sciences, Ichnos Sciences SA (formerly Glenmark Pharmaceuticals SA), La Chaux-de-Fonds, Switzerland
| | - Martin Bertschinger
- Drug Substance Development, Ichnos Sciences SA (formerly Glenmark Pharmaceuticals SA), La Chaux-de-Fonds, Switzerland
| |
Collapse
|
22
|
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
|
23
|
Hamedani NS, Donners AAMT, van Luin M, Gasper S, Rühl H, Klein C, Albert T, El Amrani M, Pötzsch B, Oldenburg J, Müller J. Functional determination of emicizumab in presence of factor VIII activity. J Thromb Haemost 2023; 21:3490-3500. [PMID: 37741510 DOI: 10.1016/j.jtha.2023.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/25/2023]
Abstract
BACKGROUND Accurate measurement of emicizumab in the presence of factor (F) VIII is required in patients with severe hemophilia A treated with emicizumab, as well as additional need for FVIII substitution or emicizumab prophylaxis in patients with acquired or moderate to mild hemophilia A. However, the presence of FVIII potentially biases the results. OBJECTIVES To assess the impact of plasma FVIII activity on determined emicizumab levels and evaluate different strategies for correction for or preanalytical inhibition of FVIII. METHODS Evaluated strategies comprised of the following: (1) calculation of actual emicizumab plasma levels based on measured FVIII activities and FVIII-affected emicizumab values, (2) preanalytical heat treatment (56 °C for 40 minutes), and (3) neutralization of FVIII activity using FVIII inhibitors. Emicizumab levels and FVIII activities were measured using a modified FVIII one-stage clotting assay and a chromogenic FVIII assay based on bovine factors, respectively. RESULTS Spiking experiments revealed a consistent linear association between FVIII activities and determined (FVIII-affected) emicizumab results at different emicizumab input levels (∼0.12 μg/mL per IU/dL of FVIII). This principally allowed for mathematical correction of measured emicizumab levels in the presence of FVIII. While a 40% to 50% activity loss of intrinsic plasma emicizumab through heat treatment was observed in patient samples, emicizumab spiked into FVIII-deficient plasma was not or only marginally affected. Application of inhibitor-based FVIII neutralization led to good agreement of results when compared with direct quantification of emicizumab by liquid chromatography-tandem mass spectrometry. CONCLUSION Inhibitor-based FVIII neutralization appears to be a feasible strategy for accurate measurement of plasma emicizumab levels in the presence of FVIII activity.
Collapse
Affiliation(s)
- Nasim Shahidi Hamedani
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Anouk Anna Marie Therese Donners
- Department of Clinical Pharmacy, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Matthijs van Luin
- Department of Clinical Pharmacy, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Simone Gasper
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Heiko Rühl
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Claudia Klein
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Thilo Albert
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Mohsin El Amrani
- Department of Clinical Pharmacy, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bernd Pötzsch
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Johannes Oldenburg
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Jens Müller
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany.
| |
Collapse
|
24
|
Duivelshof BL, Bouvarel T, Pirner S, Larraillet V, Knaupp A, Koll H, D’Atri V, Guillarme D. Enhancing Selectivity of Protein Biopharmaceuticals in Ion Exchange Chromatography through Addition of Organic Modifiers. Int J Mol Sci 2023; 24:16623. [PMID: 38068945 PMCID: PMC10706461 DOI: 10.3390/ijms242316623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
Charge heterogeneity among therapeutic monoclonal antibodies (mAbs) is considered an important critical quality attribute and requires careful characterization to ensure safe and efficacious drug products. The charge heterogeneity among mAbs is the result of chemical and enzymatic post-translational modifications and leads to the formation of acidic and basic variants that can be characterized using cation exchange chromatography (CEX). Recently, the use of mass spectrometry-compatible salt-mediated pH gradients has gained increased attention to elute the proteins from the charged stationary phase material. However, with the increasing antibody product complexity, more and more selectivity is required. Therefore, in this study, we set out to improve the selectivity by using a solvent-enriched mobile phase composition for the analysis of a variety of mAbs and bispecific antibody products. It was found that the addition of the solvents to the mobile phase appeared to modify the hydrate shell surrounding the protein and alter the retention behavior of the studied proteins. Therefore, this work demonstrates that the use of solvent-enriched mobile phase composition could be an attractive additional method parameter during method development in CEX.
Collapse
Affiliation(s)
- Bastiaan Laurens Duivelshof
- School of Pharmaceutical Sciences, University of Geneva, CMU—Rue Michel Servet 1, 1211 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU—Rue Michel Servet 1, 1211 Geneva, Switzerland
| | - Thomas Bouvarel
- School of Pharmaceutical Sciences, University of Geneva, CMU—Rue Michel Servet 1, 1211 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU—Rue Michel Servet 1, 1211 Geneva, Switzerland
| | | | | | | | - Hans Koll
- Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany
| | - Valentina D’Atri
- School of Pharmaceutical Sciences, University of Geneva, CMU—Rue Michel Servet 1, 1211 Geneva, Switzerland
- 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
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU—Rue Michel Servet 1, 1211 Geneva, Switzerland
| |
Collapse
|
25
|
Vitharana S, Stillahn JM, Katayama DS, Henry CS, Manning MC. Application of Formulation Principles to Stability Issues Encountered During Processing, Manufacturing, and Storage of Drug Substance and Drug Product Protein Therapeutics. J Pharm Sci 2023; 112:2724-2751. [PMID: 37572779 DOI: 10.1016/j.xphs.2023.08.003] [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: 10/14/2022] [Revised: 07/24/2023] [Accepted: 08/07/2023] [Indexed: 08/14/2023]
Abstract
The field of formulation and stabilization of protein therapeutics has become rather extensive. However, most of the focus has been on stabilization of the final drug product. Yet, proteins experience stress and degradation through the manufacturing process, starting with fermentaition. This review describes how formulation principles can be applied to stabilize biopharmaceutical proteins during bioprocessing and manufacturing, considering each unit operation involved in prepration of the drug substance. In addition, the impact of the container on stabilty is discussed as well.
Collapse
Affiliation(s)
| | - Joshua M Stillahn
- Legacy BioDesign LLC, Johnstown, CO 80534, USA; Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA
| | | | - Charles S Henry
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA
| | - Mark Cornell Manning
- Legacy BioDesign LLC, Johnstown, CO 80534, USA; Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
26
|
Kline JT, Melani RD, Fornelli L. Mass spectrometry characterization of antibodies at the intact and subunit levels: from targeted to large-scale analysis. INTERNATIONAL JOURNAL OF MASS SPECTROMETRY 2023; 492:117117. [PMID: 38855125 PMCID: PMC11160972 DOI: 10.1016/j.ijms.2023.117117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Antibodies are one of the most formidable molecular weapons available to our immune system. Their high specificity against a target (antigen) and capability of triggering different immune responses (e.g., complement system activation and antibody-dependent cell-mediated cytotoxicity) make them ideal drugs to fight many different human diseases. Currently, both monoclonal antibodies and more complex molecules based on the antibody scaffold are used as biologics. Naturally, such highly heterogeneous molecules require dedicated analytical methodologies for their accurate characterization. Mass spectrometry (MS) can define the presence and relative abundance of multiple features of antibodies, including critical quality attributes. The combination of small and large variations within a single molecule can only be determined by analyzing intact antibodies or their large (25 to 100 kDa) subunits. Hence, top-down (TD) and middle-down (MD) MS approaches have gained popularity over the last decade. In this Young Scientist Feature we discuss the evolution of TD and MD MS analysis of antibodies, including the new frontiers that go beyond biopharma applications. We will show how this field is now moving from the "quality control" analysis of a known, single antibody to the high-throughput investigation of complex antibody repertoires isolated from clinical samples, where the ultimate goal is represented by the complete gas-phase sequencing of antibody molecules without the need of any a priori knowledge.
Collapse
Affiliation(s)
- Jake T. Kline
- Department of Biology, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Rafael D. Melani
- Thermo Fisher Scientific, San Jose, California 95134, United States
| | - Luca Fornelli
- Department of Biology, University of Oklahoma, Norman, Oklahoma 73019, United States
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| |
Collapse
|
27
|
Füssl F, Millán-Martín S, Bones J, Carillo S. Cation exchange chromatography on a monodisperse 3 µm particle enables extensive analytical similarity assessment of biosimilars. J Pharm Biomed Anal 2023; 234:115534. [PMID: 37343453 DOI: 10.1016/j.jpba.2023.115534] [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: 04/30/2023] [Revised: 06/12/2023] [Accepted: 06/15/2023] [Indexed: 06/23/2023]
Abstract
Biosimilarity assessment requires extensive characterization and comparability exercises to investigate product quality attributes of an originator product and its potential biosimilar(s) and to highlight any differences between them. Performing a thorough comparison allows a shortened approval path, which also eliminates lengthy and expensive clinical trials, ensuring comparable product quality and efficacy but at lower drug prices. The wide variety of analytical methods available for biosimilar assessment ranges from biological to analytical assays, each providing orthogonal information to fully characterize biosimilar candidates. Intact native mass spectrometry (MS) has been shown to be an excellent tool for detection and monitoring of important quality attributes such as N-glycosylation, deamidation, sequence truncation and higher order structures. When combined with efficient upfront separation methods, simplification of the proteoform heterogeneity and associated complexity prior to MS analysis can be achieved. Native mass spectrometry can provide robust and accurate results within short analysis times and requires minimal sample preparation. In this study we report the use of a monodisperse strong cation exchange chromatography phase hyphenated with Orbitrap mass spectrometry (SCX-MS) to compare the best-selling biopharmaceutical product Humira® with 7 commercially approved biosimilar products. SCX-MS analysis allowed for the identification of previously described as well as so far unreported proteoforms and their relative quantitation across all samples, revealing differences in N-glycosylation and lysine truncation, as well as unique features for some products such as sialylation and N-terminal clipping. SCX-MS analysis, powered by a highly efficient separation column, enabled deep and efficient analytical comparison of biosimilar products.
Collapse
Affiliation(s)
- Florian Füssl
- National Institute for Bioprocessing Research & Training, Fosters Avenue, Mount Merrion, Blackrock, A94 X099 Co. Dublin, Ireland
| | - Silvia Millán-Martín
- National Institute for Bioprocessing Research & Training, Fosters Avenue, Mount Merrion, Blackrock, A94 X099 Co. Dublin, Ireland
| | - Jonathan Bones
- National Institute for Bioprocessing Research & Training, Fosters Avenue, Mount Merrion, Blackrock, A94 X099 Co. Dublin, Ireland; School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Dublin 4 D04 V1W8, Ireland
| | - Sara Carillo
- National Institute for Bioprocessing Research & Training, Fosters Avenue, Mount Merrion, Blackrock, A94 X099 Co. Dublin, Ireland.
| |
Collapse
|
28
|
Millán-Martín S, Jakes C, Carillo S, Gallagher L, Scheffler K, Broster K, Bones J. Multi-Attribute Method (MAM): An Emerging Analytical Workflow for Biopharmaceutical Characterization, Batch Release and cGMP Purity Testing at the Peptide and Intact Protein Level. Crit Rev Anal Chem 2023; 54:3234-3251. [PMID: 37490277 DOI: 10.1080/10408347.2023.2238058] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
The rapid growth of biotherapeutic industry, with more and more complex molecules entering the market, forces the need for advanced analytical platforms that can quickly and accurately identify and quantify product quality attributes. Mass spectrometry has the potential to provide more detailed information about the quality attributes of complex products, and MS methods are more sensitive than UV methods for detection of impurities. The multi-attribute method (MAM), a liquid chromatography-mass spectrometry based analytical approach is an emerging platform which supports biotherapeutic characterization and cGMP testing. The main advantage lies in the ability to monitor multiple quality attributes in a single assay, both at the peptide and the intact level, facilitating streamlined biopharmaceutical production, from research and development to the QC environment. This review highlights the current landscape of the MAM approach with special attention given to increased analytical throughput, general requirements for QC in terms of instrumentation and software, regulatory requirements, and industry acceptance of the MAM platform.
Collapse
Affiliation(s)
- Silvia Millán-Martín
- National Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merrion, Co, Dublin, Ireland
| | - Craig Jakes
- National Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merrion, Co, Dublin, Ireland
| | - Sara Carillo
- National Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merrion, Co, Dublin, Ireland
| | | | | | - Kelly Broster
- Thermo Fisher Scientific, Stafford House, Hemel Hempstead, UK
| | - Jonathan Bones
- National Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merrion, Co, Dublin, Ireland
- School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Ireland
| |
Collapse
|
29
|
Shah B, Zhu Y, Wypych J, Zhang Z. Observation of Heavy-Chain C-Terminal Des-GK Truncation in Recombinant and Human Endogenous IgG4. J Pharm Sci 2023; 112:1845-1849. [PMID: 37187261 DOI: 10.1016/j.xphs.2023.05.005] [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: 03/30/2023] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 05/17/2023]
Abstract
Therapeutic IgG mAbs have shown presence of three variations of their heavy chain C-termini, including the unprocessed C-terminal lysine, the processed C-terminal lysine, and C-terminal amidation. These variants are also present in endogenous human IgGs, although the level of unprocessed C-terminal lysine is very low. Here we report a new heavy-chain C-terminal variant, i.e., the des-GK truncation, which exists in both recombinant and endogenous human IgG4. The des-GK truncation was found in negligible amount in IgG1, IgG2 and IgG3 subclasses. Observation of a significant level of heavy-chain C-terminal des-GK truncation in endogenous human IgG4 suggests that low level of this variant present in therapeutic IgG4 is unlikely to be a safety concern.
Collapse
Affiliation(s)
- Bhavana Shah
- Process Development, Amgen Inc. One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - YaXing Zhu
- Process Development, Amgen Inc. 360 Binney Street, Cambridge, MA, 02142, USA
| | - Jette Wypych
- Process Development, Amgen Inc. One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Zhongqi Zhang
- Process Development, Amgen Inc. One Amgen Center Drive, Thousand Oaks, CA 91320, USA.
| |
Collapse
|
30
|
Arauzo-Aguilera K, Buscajoni L, Koch K, Thompson G, Robinson C, Berkemeyer M. Yields and product comparison between Escherichia coli BL21 and W3110 in industrially relevant conditions: anti-c-Met scFv as a case study. Microb Cell Fact 2023; 22:104. [PMID: 37208750 PMCID: PMC10197847 DOI: 10.1186/s12934-023-02111-4] [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: 01/17/2023] [Accepted: 05/01/2023] [Indexed: 05/21/2023] Open
Abstract
INTRODUCTION In the biopharmaceutical industry, Escherichia coli is one of the preferred expression hosts for large-scale production of therapeutic proteins. Although increasing the product yield is important, product quality is a major factor in this industry because greatest productivity does not always correspond with the highest quality of the produced protein. While some post-translational modifications, such as disulphide bonds, are required to achieve the biologically active conformation, others may have a negative impact on the product's activity, effectiveness, and/or safety. Therefore, they are classified as product associated impurities, and they represent a crucial quality parameter for regulatory authorities. RESULTS In this study, fermentation conditions of two widely employed industrial E. coli strains, BL21 and W3110 are compared for recombinant protein production of a single-chain variable fragment (scFv) in an industrial setting. We found that the BL21 strain produces more soluble scFv than the W3110 strain, even though W3110 produces more recombinant protein in total. A quality assessment on the scFv recovered from the supernatant was then performed. Unexpectedly, even when our scFv is correctly disulphide bonded and cleaved from its signal peptide in both strains, the protein shows charge heterogeneity with up to seven distinguishable variants on cation exchange chromatography. Biophysical characterization confirmed the presence of altered conformations of the two main charged variants. CONCLUSIONS The findings indicated that BL21 is more productive for this specific scFv than W3110. When assessing product quality, a distinctive profile of the protein was found which was independent of the E. coli strain. This suggests that alterations are present in the recovered product although the exact nature of them could not be determined. This similarity between the two strains' generated products also serves as a sign of their interchangeability. This study encourages the development of innovative, fast, and inexpensive techniques for the detection of heterogeneity while also provoking a debate about whether intact mass spectrometry-based analysis of the protein of interest is sufficient to detect heterogeneity in a product.
Collapse
Affiliation(s)
| | - Luisa Buscajoni
- Biopharma Austria, Process Science, Boehringer-Ingelheim RCV GmbH & Co KG, Dr. Boehringer-Gasse 5-11, 1121 Vienna, Austria
| | - Karin Koch
- Biopharma Austria, Process Science, Boehringer-Ingelheim RCV GmbH & Co KG, Dr. Boehringer-Gasse 5-11, 1121 Vienna, Austria
| | - Gary Thompson
- Wellcome Trust Biological NMR Facility, School of Biosciences, University of Kent, Canterbury, CT2 7NJ UK
| | - Colin Robinson
- School of Biosciences, University of Kent, Canterbury, CT2 7NJ UK
| | - Matthias Berkemeyer
- Biopharma Austria, Process Science, Boehringer-Ingelheim RCV GmbH & Co KG, Dr. Boehringer-Gasse 5-11, 1121 Vienna, Austria
| |
Collapse
|
31
|
Schlecht J, Moritz B, Kiessig S, Neusüß C. Characterization of therapeutic mAb charge heterogeneity by iCIEF coupled to mass spectrometry (iCIEF-MS). Electrophoresis 2023; 44:540-548. [PMID: 36148605 DOI: 10.1002/elps.202200170] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/27/2022] [Accepted: 08/31/2022] [Indexed: 11/07/2022]
Abstract
Imaged capillary isoelectric focusing (iCIEF) has emerged as an important technique for therapeutic monoclonal antibody (mAb) charge heterogeneity analysis in the biopharmaceutical context, providing imaged detection and quantitation by UV without a mobilization step. Besides quantitation, the characterization of separated charge variants ideally directly by online electrospray ionization-mass spectrometry (ESI-MS) is crucial to ensure product quality, safety, and efficacy. Straightforward direct iCIEF-MS coupling combining high separation efficiency and quantitative results of iCIEF with the characterization power of MS enables deep characterization of mAb charge variants. A short technical setup and optimized methodical parameters (30 nl/min mobilization rate, 2%-4% ampholyte concentration, 0.5-2 mg/ml sample concentration) allow successful mAb charge variant peak assignment from iCIEF to MS. Despite a loss of separation resolution during the transfer, separated intact mAb charge variants, including deamidation as well as major and minor glycoforms even from low abundant charge variants, could be characterized by online ESI-MS with high precision. The presented setup provides a large potential for mAb charge heterogeneity characterization in biopharmaceutical applications.
Collapse
Affiliation(s)
- Johannes Schlecht
- Department of Chemistry, Aalen University, Aalen, Germany.,Department of Pharmaceutical and Medicinal Chemistry, Friedrich Schiller University Jena, Jena, Germany
| | | | | | | |
Collapse
|
32
|
Kaur R, Jain R, Budholiya N, Rathore AS. Long term culturing of CHO cells: phenotypic drift and quality attributes of the expressed monoclonal antibody. Biotechnol Lett 2023; 45:357-370. [PMID: 36707452 DOI: 10.1007/s10529-023-03346-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 12/13/2022] [Accepted: 01/05/2023] [Indexed: 01/29/2023]
Abstract
OBJECTIVE Establishing cell lines with enhanced protein production requires a deep understanding of the cellular dynamics and cell line stability. The aim of the study is to investigate the impact of long term culturing (LTC) on cell morphology and altered cellular functions possibly leading to phenotypic drift, impacting product yield and quality. Study highlights the orthogonal cellular and analytical assay toolbox to define cell line stability for optimal culture performance and product quality. METHODS We investigated recombinant monoclonal antibody (mAb) expressing CHO cells for 60 passages or 180 generations and assessed the cell growth characteristics and morphology by confocal and scanning electron microscopy. Quality attributes of expressed mAb is accessed by performing charge variants, glycan, and host cell protein analysis. RESULTS We observed a 1.65-fold increase in viable cell population and 1.3-fold increase in cell specific growth rate. A 2.5-fold decrease in antibody titer and abatement of actin filament indicate cellular phenotypic drift. Mitochondrial membrane potential (∆ΨM) signified cell health and metabolic activity during LTC. Host cell protein production is reduced by 1.8-fold. Charge heterogeneity was perturbed with 12.5% and 43% reduction in abundance of acidic and basic charge variants respectively. Glycan profile indicated a decline in fucosylation with 17% increase in galactosylated species as compared with early passaged cells. CONCLUSION LTC impinges on cellular phenotype as well as the quality of the expressed antibody, suggesting a defined subculturing limit to retain stable protein expression and cell morphology to achieve consistent product quality. Study signifies the changes in cellular and metabolic markers, suggesting cellular and analytical toolbox which could play a significant role in defining cell characteristics and ensured product quality.
Collapse
Affiliation(s)
- Rajinder Kaur
- Department of Chemical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Ritu Jain
- Department of Chemical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Niharika Budholiya
- Department of Chemical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Anurag S Rathore
- Department of Chemical Engineering, Indian Institute of Technology Delhi, New Delhi, India.
| |
Collapse
|
33
|
Schlecht J, Jooß K, Moritz B, Kiessig S, Neusüß C. Two-Dimensional Capillary Zone Electrophoresis-Mass Spectrometry: Intact mAb Charge Variant Separation Followed by Peptide Level Analysis Using In-Capillary Digestion. Anal Chem 2023; 95:4059-4066. [PMID: 36800441 DOI: 10.1021/acs.analchem.2c04578] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
Characterization of charge heterogeneity is an essential pillar for pharmaceutical development and quality control of therapeutic monoclonal antibodies (mAbs). The highly selective and commonly applied capillary zone electrophoresis (CZE) method containing high amounts of ε-aminocaproic acid (EACA) provides a detailed and robust charge heterogeneity profile of intact mAb variants. Nevertheless, the exact location of protein modifications within these charge profiles remains ambiguous. Electrospray ionization mass spectrometry (ESI-MS) is a promising tool for this purpose; however, EACA is incompatible with electrospray. In this context, we present a two-dimensional CZE-CZE-MS system to combine efficient charge variant separation of intact mAbs with subsequent peptide analysis after in-capillary digestion of selected charge variants. The first dimension is based on a generic CZE(EACA) method in a fused silica capillary. In the second dimension, a neutral-coated capillary is used for in-capillary reduction and digestion with Tris(2-carboxyethyl)phosphine (TCEP) and pepsin, followed by CZE separation and MS/MS-characterization of the resulting peptides. The setup is demonstrated using stressed and nonstressed mAbs where peaks of basic, main, and acidic variants were transferred in a heart-cut fashion, digested, and characterized on the peptide level. Sequence coverages of more than 90% were obtained for heavy chain (HC) and light chain (LC) for four different mAbs, including low-abundance variants (<2% of the main peak). Frequently observed modifications (deamidation, oxidation, etc.) could be detected and localized. This study demonstrates a proof-of-concept for identification and localization of protein modifications from CZE charge heterogeneity profiles and, in this way, is expected to support the development and quality control testing of protein pharmaceuticals.
Collapse
Affiliation(s)
- Johannes Schlecht
- Department of Chemistry, Aalen University, Beethovenstrasse 1, 73430 Aalen, Germany.,Department of Pharmaceutical and Medicinal Chemistry, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Kevin Jooß
- Departments of Chemistry and Molecular Biosciences, the Chemistry of Life Processes Institute, and the Proteomics Center of Excellence, Northwestern University, Evanston, Illinois 60208, United States
| | - Bernd Moritz
- F. Hoffmann La-Roche Ltd., Grenzacherstraße 124, 4058 Basel, Switzerland
| | - Steffen Kiessig
- F. Hoffmann La-Roche Ltd., Grenzacherstraße 124, 4058 Basel, Switzerland
| | - Christian Neusüß
- Department of Chemistry, Aalen University, Beethovenstrasse 1, 73430 Aalen, Germany
| |
Collapse
|
34
|
Alhazmi HA, Albratty M. Analytical Techniques for the Characterization and Quantification of Monoclonal Antibodies. Pharmaceuticals (Basel) 2023; 16:291. [PMID: 37259434 PMCID: PMC9967501 DOI: 10.3390/ph16020291] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 08/12/2023] Open
Abstract
Monoclonal antibodies (mAbs) are a fast-growing class of biopharmaceuticals. They are widely used in the identification and detection of cell makers, serum analytes, and pathogenic agents, and are remarkably used for the cure of autoimmune diseases, infectious diseases, or malignancies. The successful application of therapeutic mAbs is based on their ability to precisely interact with their appropriate target sites. The precision of mAbs rely on the isolation techniques delivering pure, consistent, stable, and safe lots that can be used for analytical, diagnostic, or therapeutic applications. During the creation of a biologic, the key quality features of a particular mAb, such as structure, post-translational modifications, and activities at the biomolecular and cellular levels, must be characterized and profiled in great detail. This implies the requirement of powerful state of the art analytical techniques for quality control and characterization of mAbs. Until now, various analytical techniques have been developed to characterize and quantify the mAbs according to the regulatory guidelines. The present review summarizes the major techniques used for the analyses of mAbs which include chromatographic, electrophoretic, spectroscopic, and electrochemical methods in addition to the modifications in these methods for improving the quality of mAbs. This compilation of major analytical techniques will help students and researchers to have an overview of the methodologies employed by the biopharmaceutical industry for structural characterization of mAbs for eventual release of therapeutics in the drug market.
Collapse
Affiliation(s)
- Hassan A. Alhazmi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan 45142, Saudi Arabia
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| |
Collapse
|
35
|
Simple N-Glycan Profile Analysis Using Lectin Staining, Mass Spectrometry, and GlycoMaple. Methods Mol Biol 2022; 2557:691-707. [PMID: 36512245 DOI: 10.1007/978-1-0716-2639-9_41] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The Golgi apparatus is one of the major sites of protein and lipid glycosylation and processing. Protein N-glycosylation plays critical roles in protein folding, transport, stability, and activity. Various glycosyltransferases and glycoside hydrolases are localized at each cisterna in the Golgi apparatus and synthesize a large variety of N-glycan structures. The biosynthetic pathways of N-glycans are complicated, which hiders the rational design of glycan metabolic pathways. In addition, the analysis of glycan structure requires specialized instruments for analyses such as mass spectrometry, high-performance liquid chromatography, and nuclear magnetic resonance spectroscopy, which are not familiar to all laboratories. Here, we introduce relatively simple methods for N-glycan analysis, including disruption of genes encoding glycosyltransferases or glycoside hydrolases, glycan structural analysis using lectins and mass spectrometry, and visualization of glycan metabolic pathways in silico.
Collapse
|
36
|
Beck A, Nowak C, Meshulam D, Reynolds K, Chen D, Pacardo DB, Nicholls SB, Carven GJ, Gu Z, Fang J, Wang D, Katiyar A, Xiang T, Liu H. Risk-Based Control Strategies of Recombinant Monoclonal Antibody Charge Variants. Antibodies (Basel) 2022; 11:73. [PMID: 36412839 PMCID: PMC9703962 DOI: 10.3390/antib11040073] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/27/2022] [Accepted: 11/11/2022] [Indexed: 09/28/2023] Open
Abstract
Since the first approval of the anti-CD3 recombinant monoclonal antibody (mAb), muromonab-CD3, a mouse antibody for the prevention of transplant rejection, by the US Food and Drug Administration (FDA) in 1986, mAb therapeutics have become increasingly important to medical care. A wealth of information about mAbs regarding their structure, stability, post-translation modifications, and the relationship between modification and function has been reported. Yet, substantial resources are still required throughout development and commercialization to have appropriate control strategies to maintain consistent product quality, safety, and efficacy. A typical feature of mAbs is charge heterogeneity, which stems from a variety of modifications, including modifications that are common to many mAbs or unique to a specific molecule or process. Charge heterogeneity is highly sensitive to process changes and thus a good indicator of a robust process. It is a high-risk quality attribute that could potentially fail the specification and comparability required for batch disposition. Failure to meet product specifications or comparability can substantially affect clinical development timelines. To mitigate these risks, the general rule is to maintain a comparable charge profile when process changes are inevitably introduced during development and even after commercialization. Otherwise, new peaks or varied levels of acidic and basic species must be justified based on scientific knowledge and clinical experience for a specific molecule. Here, we summarize the current understanding of mAb charge variants and outline risk-based control strategies to support process development and ultimately commercialization.
Collapse
Affiliation(s)
- Alain Beck
- Centre d’Immunologie Pierre-Fabre (CIPF), 5 Avenue Napoléon III, 74160 Saint-Julien-en-Genevois, France
| | - Christine Nowak
- Protein Characterization, Alexion AstraZeneca Rare Disease, 100 College St., New Haven, CT 06510, USA
| | - Deborah Meshulam
- Technical Operations/CMC, Scholar Rock, 301 Binney Street, 3rd Floor, Cambridge, MA 02142, USA
| | - Kristina Reynolds
- Technical Operations/CMC, Scholar Rock, 301 Binney Street, 3rd Floor, Cambridge, MA 02142, USA
| | - David Chen
- Technical Operations/CMC, Scholar Rock, 301 Binney Street, 3rd Floor, Cambridge, MA 02142, USA
| | - Dennis B. Pacardo
- Technical Operations/CMC, Scholar Rock, 301 Binney Street, 3rd Floor, Cambridge, MA 02142, USA
| | - Samantha B. Nicholls
- Protein Sciences, Scholar Rock, 301 Binney Street, 3rd Floor, Cambridge, MA 02142, USA
| | - Gregory J. Carven
- Research, Scholar Rock, 301 Binney Street, 3rd Floor, Cambridge, MA 02142, USA
| | - Zhenyu Gu
- Jasper Therapeutics, Inc., 2200 Bridge Pkwy Suite 102, Redwood City, CA 94065, USA
| | - Jing Fang
- Biological Drug Discovery, Biogen, 225 Binney St., Cambridge, MA 02142, USA
| | - Dongdong Wang
- Global Biologics, Takeda Pharmaceuticals, 300 Shire Way, Lexington, MA 02421, USA
| | - Amit Katiyar
- CMC Technical Operations, Magenta Therapeutics, 100 Technology Square, Cambridge, MA 02139, USA
| | - Tao Xiang
- Downstream Process and Analytical Development, Boston Institute of Biotechnology, 225 Turnpike Rd., Southborough, MA 01772, USA
| | - Hongcheng Liu
- Technical Operations/CMC, Scholar Rock, 301 Binney Street, 3rd Floor, Cambridge, MA 02142, USA
| |
Collapse
|
37
|
Schwahn AB, Baek J, Lin S, Pohl CA, Cook K. A Universal Eluent System for Method Scouting and Separation of Biotherapeutic Proteins by Ion-Exchange, Size-Exclusion, and Hydrophobic Interaction Chromatography. Anal Chem 2022; 94:16369-16375. [DOI: 10.1021/acs.analchem.2c03531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
| | - Julia Baek
- Thermo Fisher Scientific, Sunnyvale, California94085, United States
| | - Shanhua Lin
- Thermo Fisher Scientific, Sunnyvale, California94085, United States
| | | | - Ken Cook
- Thermo Fisher Scientific, Hemel Hempstead, HertfordshireHP2 7GE, U.K
| |
Collapse
|
38
|
Zhang W, Wang H, Feng N, Li Y, Gu J, Wang Z. Developability assessment at early-stage discovery to enable development of antibody-derived therapeutics. Antib Ther 2022; 6:13-29. [PMID: 36683767 PMCID: PMC9847343 DOI: 10.1093/abt/tbac029] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/13/2022] Open
Abstract
Developability refers to the likelihood that an antibody candidate will become a manufacturable, safe and efficacious drug. Although the safety and efficacy of a drug candidate will be well considered by sponsors and regulatory agencies, developability in the narrow sense can be defined as the likelihood that an antibody candidate will go smoothly through the chemistry, manufacturing and control (CMC) process at a reasonable cost and within a reasonable timeline. Developability in this sense is the focus of this review. To lower the risk that an antibody candidate with poor developability will move to the CMC stage, the candidate's developability-related properties should be screened, assessed and optimized as early as possible. Assessment of developability at the early discovery stage should be performed in a rapid and high-throughput manner while consuming small amounts of testing materials. In addition to monoclonal antibodies, bispecific antibodies, multispecific antibodies and antibody-drug conjugates, as the derivatives of monoclonal antibodies, should also be assessed for developability. Moreover, we propose that the criterion of developability is relative: expected clinical indication, and the dosage and administration route of the antibody could affect this criterion. We also recommend a general screening process during the early discovery stage of antibody-derived therapeutics. With the advance of artificial intelligence-aided prediction of protein structures and features, computational tools can be used to predict, screen and optimize the developability of antibody candidates and greatly reduce the risk of moving a suboptimal candidate to the development stage.
Collapse
Affiliation(s)
- Weijie Zhang
- Biologicals Innovation and Discovery, WuXi Biologicals, 1951 Huifeng West Road, Fengxian District, Shanghai 201400, China
| | - Hao Wang
- Biologicals Innovation and Discovery, WuXi Biologicals, 1951 Huifeng West Road, Fengxian District, Shanghai 201400, China
| | - Nan Feng
- Biologicals Innovation and Discovery, WuXi Biologicals, 1951 Huifeng West Road, Fengxian District, Shanghai 201400, China
| | - Yifeng Li
- Technology and Process Development, WuXi Biologicals, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Jijie Gu
- Biologicals Innovation and Discovery, WuXi Biologicals, 1951 Huifeng West Road, Fengxian District, Shanghai 201400, China
| | - Zhuozhi Wang
- To whom correspondence should be addressed. Biologics Innovation and Discovery, WuXi Biologicals, 1951 Huifeng West Road, Fengxian District, Shanghai 201400, China, Phone number: +86-21-50518899
| |
Collapse
|
39
|
Krištić J, Lauc G, Pezer M. Immunoglobulin G glycans - Biomarkers and molecular effectors of aging. Clin Chim Acta 2022; 535:30-45. [PMID: 35970404 DOI: 10.1016/j.cca.2022.08.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/04/2022] [Accepted: 08/04/2022] [Indexed: 11/28/2022]
Abstract
Immunoglobulin G (IgG) antibodies are post-translationally modified by the addition of complex carbohydrate molecules - glycans, which have profound effects on the IgG function, most significantly as modulators of its inflammatory capacity. Therefore, it is not surprising that the changes in IgG glycosylation pattern are associated with various physiological states and diseases, including aging and age-related diseases. Importantly, within the inflammaging concept, IgG glycans are considered not only biomarkers but one of the molecular effectors of the aging process. The exact mechanism by which they exert their function, however, remains unknown. In this review, we list and comment on, to our knowledge, all studies that examined changes in IgG glycosylation during aging in humans. We focus on the information obtained from studies on general population, but we also cover the insights obtained from studies of long-lived individuals and people with age-related diseases. We summarize the current knowledge on how levels of different IgG glycans change with age (i.e., the extent and direction of the change with age) and discuss the potential mechanisms and possible functional roles of changes in IgG glycopattern that accompany aging.
Collapse
Affiliation(s)
| | - Gordan Lauc
- Genos Glycoscience Research Laboratory, Zagreb, Croatia; Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - Marija Pezer
- Genos Glycoscience Research Laboratory, Zagreb, Croatia.
| |
Collapse
|
40
|
Gurel B, Berksoz M, Capkin E, Parlar A, Pala MC, Ozkan A, Capan Y, Daglikoca DE, Yuce M. Structural and Functional Analysis of CEX Fractions Collected from a Novel Avastin® Biosimilar Candidate and Its Innovator: A Comparative Study. Pharmaceutics 2022; 14:pharmaceutics14081571. [PMID: 36015197 PMCID: PMC9415858 DOI: 10.3390/pharmaceutics14081571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 02/01/2023] Open
Abstract
Avastin® is a humanized recombinant monoclonal antibody used to treat cancer by targeting VEGF-A to inhibit angiogenesis. SIMAB054, an Avastin® biosimilar candidate developed in this study, showed a different charge variant profile than its innovator. Thus, it is fractionated into acidic, main, and basic isoforms and collected physically by Cation Exchange Chromatography (CEX) for a comprehensive structural and functional analysis. The innovator product, fractionated into the same species and collected by the same method, is used as a reference for comparative analysis. Ultra-Performance Liquid Chromatography (UPLC) ESI-QToF was used to analyze the modifications leading to charge heterogeneities at intact protein and peptide levels. The C-terminal lysine clipping and glycosylation profiles of the samples were monitored by intact mAb analysis. The post-translational modifications, including oxidation, deamidation, and N-terminal pyroglutamic acid formation, were determined by peptide mapping analysis in the selected signal peptides. The relative binding affinities of the fractionated charge isoforms against the antigen, VEGF-A, and the neonatal receptor, FcRn, were revealed by Surface Plasmon Resonance (SPR) studies. The results show that all CEX fractions from the innovator product and the SIMAB054 shared the same structural variants, albeit in different ratios. Common glycoforms and post-translational modifications were the same, but at different percentages for some samples. The dissimilarities were mostly originating from the presence of extra C-term Lysin residues, which are prone to enzymatic degradation in the body, and thus they were previously assessed as clinically irrelevant. Another critical finding was the presence of different glyco proteoforms in different charge species, such as increased galactosylation in the acidic and afucosylation in the basic species. SPR characterization of the isolated charge variants further confirmed that basic species found in the CEX analyses of the biosimilar candidate were also present in the innovator product, although at lower amounts. The charge variants’ in vitro antigen- and neonatal receptor-binding activities varied amongst the samples, which could be further investigated in vivo with a larger sample set to reveal the impact on the pharmacokinetics of drug candidates. Minor structural differences may explain antigen-binding differences in the isolated charge variants, which is a key parameter in a comparability exercise. Consequently, such a biosimilar candidate may not comply with high regulatory standards unless the binding differences observed are justified and demonstrated not to have any clinical impact.
Collapse
Affiliation(s)
- Busra Gurel
- SUNUM Nanotechnology Research and Application Center, Sabanci University, Istanbul 34956, Turkey;
| | - Melike Berksoz
- ILKO ARGEM Biotechnology R&D Center, Istanbul 34906, Turkey; (M.B.); (E.C.); (M.C.P.); (A.O.); (Y.C.)
| | - Eda Capkin
- ILKO ARGEM Biotechnology R&D Center, Istanbul 34906, Turkey; (M.B.); (E.C.); (M.C.P.); (A.O.); (Y.C.)
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey;
| | - Ayhan Parlar
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey;
| | - Meltem Corbacioglu Pala
- ILKO ARGEM Biotechnology R&D Center, Istanbul 34906, Turkey; (M.B.); (E.C.); (M.C.P.); (A.O.); (Y.C.)
| | - Aylin Ozkan
- ILKO ARGEM Biotechnology R&D Center, Istanbul 34906, Turkey; (M.B.); (E.C.); (M.C.P.); (A.O.); (Y.C.)
| | - Yılmaz Capan
- ILKO ARGEM Biotechnology R&D Center, Istanbul 34906, Turkey; (M.B.); (E.C.); (M.C.P.); (A.O.); (Y.C.)
| | - Duygu Emine Daglikoca
- ILKO ARGEM Biotechnology R&D Center, Istanbul 34906, Turkey; (M.B.); (E.C.); (M.C.P.); (A.O.); (Y.C.)
- Correspondence: (D.E.D.); (M.Y.)
| | - Meral Yuce
- SUNUM Nanotechnology Research and Application Center, Sabanci University, Istanbul 34956, Turkey;
- Correspondence: (D.E.D.); (M.Y.)
| |
Collapse
|
41
|
Shah B, Li M, Wypych J, Joubert MK, Zhang Z. Observation of Heavy-Chain C-Terminal Amidation in Human Endogenous IgG. J Pharm Sci 2022; 111:2445-2450. [PMID: 35718127 DOI: 10.1016/j.xphs.2022.06.012] [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: 04/15/2022] [Revised: 06/10/2022] [Accepted: 06/10/2022] [Indexed: 10/18/2022]
Abstract
Therapeutic IgG mAbs expressed from Chinese hamster ovary (CHO) cells are known to contain three C-terminal variants in their heavy chains, namely, the unprocessed C-terminal lysine, the processed C-terminal lysine, and C-terminal amidation. Although the presence of C-terminal amidation in CHO-expressed IgGs is well studied, the biological impact of the variant on the safety and efficacy of biotherapeutics has not been well understood. To further our biological understanding of C-terminal amidation, we analyzed a series of IgG samples, including both endogenous human IgGs as well as recombinant IgGs of different subclasses expressed from both CHO and murine cell lines, for their heavy-chain C-terminal variants by LC-MS/MS based peptide mapping. The results demonstrate that heavy-chain C-terminal amidation is a common variant occurring in IgG of all four subclasses (IgG1, IgG2, IgG3 and IgG4). The variant is generally present in recombinant IgG mAbs expressed from CHO cell lines but not in IgG mAbs expressed from murine cell lines, whereas the IgGs expressed from murine cell lines contain a much larger amount of unprocessed C-terminal lysine. Additionally, a significant amount of heavy-chain C-terminal amidation is observed in endogenous human IgGs, indicating that small amount of the variant present in therapeutic IgGs does not pose a safety concern.
Collapse
Affiliation(s)
- Bhavana Shah
- Process Development, Amgen Inc. One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Ming Li
- Quality, Amgen Inc. One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Jette Wypych
- Process Development, Amgen Inc. One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Marisa K Joubert
- Process Development, Amgen Inc. One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Zhongqi Zhang
- Process Development, Amgen Inc. One Amgen Center Drive, Thousand Oaks, CA 91320, USA.
| |
Collapse
|
42
|
Nupur N, Rathore AS. Elucidating chemical and disulfide heterogeneities in rituximab using reduced and non-reduced peptide mapping. J Sep Sci 2022; 45:2887-2900. [PMID: 35670633 DOI: 10.1002/jssc.202200290] [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: 04/12/2022] [Revised: 06/03/2022] [Accepted: 06/03/2022] [Indexed: 11/07/2022]
Abstract
Peptide mapping by liquid chromatography-mass spectrometry is the gold standard to characterize post-translational modifications and disulfide bonds. The structural integrity, heterogeneity, and quality of biotherapeutic proteins are evaluated via reduced and non-reduced peptide mapping methods. However, non-enzymatic artifacts are often induced during sample preparation when alkaline pH conditions are used. To minimize these artifacts, methods using various acidic pH conditions have been developed by multiple researchers. However, these may lead to missed and non-specific cleavages during the analysis. In this study, improved reduced and non-reduced peptide mapping method has been proposed to characterize endogenous chemical modifications and native disulfide bonds of monoclonal antibody -based products. Solubilization has been carried out at acidic pH conditions under high temperature, followed by the addition of tris (2-carboxyethyl) phosphine as a reducing agent and a low alkylating agent. It was observed that the non-enzymatic post-translational modifications and non-native disulfide scrambled peptides significantly reduced under trypsin plus Lys-C digestion conditions at acidic pH as compared to the traditional methods. The results demonstrate that the proposed peptide mapping method using trypsin plus Lys-C could identify and quantify various chemical and disulfide heterogeneities with minimal artifacts. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Neh Nupur
- Department of Chemical Engineering, IIT Delhi, Hauz Khas, New Delhi, 110016, India.,DBT Center of Excellence for Biopharmaceutical Technology, IIT Delhi, Hauz Khas, New Delhi, 110016, India
| | - Anurag S Rathore
- Department of Chemical Engineering, IIT Delhi, Hauz Khas, New Delhi, 110016, India.,DBT Center of Excellence for Biopharmaceutical Technology, IIT Delhi, Hauz Khas, New Delhi, 110016, India
| |
Collapse
|
43
|
Optimized Methods for Analytical and Functional Comparison of Biosimilar mAb Drugs: A Case Study for Avastin, Mvasi, and Zirabev. Sci Pharm 2022. [DOI: 10.3390/scipharm90020036] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Bevacizumab is a humanized therapeutic monoclonal antibody used to reduce angiogenesis, a hallmark of cancer, by binding to VEGF-A. Many pharmaceutical companies have developed biosimilars of Bevacizumab in the last decade. The official reports provided by the FDA and EMA summarize the analytical performance of biosimilars as compared to the originators without giving detailed analytical procedures. In the current study, several key methods were optimized and reported for analytical and functional comparison of bevacizumab originators (Avastin, Altuzan) and approved commercial biosimilars (Zirabev and Mvasi). This case study presents a comparative analysis of a set of biosimilars under optimized analytical conditions for the first time in the literature. The chemical structure of all products was analyzed at intact protein and peptide levels by high-resolution mass spectrometry; the major glycoforms and posttranslational modifications, including oxidation, deamidation, N-terminal PyroGlu addition, and C-terminal Lys clipping, were compared. The SPR technique was used to reveal antigen and some receptor binding kinetics of all products, and the ELISA technique was used for C1q binding affinity analysis. Finally, the inhibition performance of the samples was evaluated by an MTS-based proliferation assay in vitro. Major glycoforms were similar, with minor differences among the samples. Posttranslational modifications, except C-terminal Lys, were determined similarly, while unclipped Lys percentage was higher in Zirabev. The binding kinetics for VEGF, FcRn, FcγRIa, and C1q were similar or in the value range of originators. The anti-proliferative effect of Zirabev was slightly higher than the originators and Mvasi. The analysis of biosimilars under the same conditions could provide a new aspect to the literature in terms of the applied analytical techniques. Further studies in this field would be helpful to better understand the inter-comparability of the biosimilars.
Collapse
|
44
|
Böttinger K, Esser-Skala W, Segl M, Herwig C, Huber CG. At-line quantitative profiling of monoclonal antibody products during bioprocessing using HPLC-MS. Anal Chim Acta 2022; 1207:339813. [DOI: 10.1016/j.aca.2022.339813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 11/01/2022]
|
45
|
Jakes C, Bones J, Carillo S, Martín SM. Multi-Attribute Monitoring and the Multi-Attribute Method: A Powerful Double Act for Supporting Biopharmaceutical Manufacturing. LCGC NORTH AMERICA 2022. [DOI: 10.56530/lcgc.na.lg6280u5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
As biopharmaceutical manufacturers look towards implementing solutions for real-time process monitoring, the availability of simple, yet informative analytical methods is required. In this article, we discuss the combination of multi-attribute monitoring using Protein A affinity chromatography coupled to mass spectrometry (MS) for rapid trending of product quality attributes (PQAs) during upstream processing, with the multi-attribute method (MAM) for site-specific analysis of PQAs and new peak detection to determine adherence to specifications. Working together, these multi-attribute approaches represent a powerful combination for advanced process control.
Collapse
Affiliation(s)
- Craig Jakes
- National Institute for Bioprocessing Research and Training
| | - Jonathan Bones
- National Institute for Bioprocessing Research and Training
| | - Sara Carillo
- National Institute for Bioprocessing Research and Training, Characterization and Comparability Laboratory
| | - Silvia Millán Martín
- National Institute for Bioprocessing Research and Training, Characterization and Comparability Laboratory
| |
Collapse
|
46
|
Oxidation of specific tryptophan residues inhibits high affinity binding of cocaine and its metabolites to a humanized anti-cocaine mAb. J Biol Chem 2022; 298:101689. [PMID: 35143837 PMCID: PMC8908252 DOI: 10.1016/j.jbc.2022.101689] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/28/2022] [Accepted: 02/03/2022] [Indexed: 11/24/2022] Open
Abstract
Cocaine addiction remains a serious problem lacking an effective pharmacological treatment. Thus, we have developed a high-affinity anti-cocaine monoclonal antibody (mAb), h2E2, for the treatment of cocaine use disorders. We show that selective tryptophan (Trp) oxidation by 2,2′-azobis(2-amidinopropane) dihydrochloride (AAPH) resulted in a loss of high-affinity binding of cocaine to this mAb. The newly developed use of excess methionine (Met) to protect mAb met residues from AAPH oxidation did not substantially attenuate the effects of oxidation on cocaine binding but greatly decreased the modification of met residues in the mAb. Similar large decreases in ligand affinity (5000–10,000-fold) upon oxidation were observed using cocaine and two cocaine metabolites, cocaethylene and benzoylecgonine, which also bind with nanomolar affinity to this h2E2 mAb. The decrease in binding affinity was accompanied by a decrease of approximately 50% in Trp fluorescence, and increases in mAb 310 to 370 nm absorbance were consistent with the presence of oxidized forms of Trp. Finally, mass spectral analysis of peptides derived from control and AAPH-oxidized mAb indicated that excess free met did effectively protect mAb met residues from oxidation, and that AAPH-oxidized mAb heavy-chain Trp33 and light-chain Trp91 residues are important for cocaine binding, consistent with a recently derived h2E2 Fab fragment crystal structure containing bound benzoylecgonine. Thus, protection of the anti-cocaine h2E2 mAb from Trp oxidation prior to its clinical administration is critical for its proposed therapeutic use in the treatment of cocaine use disorders.
Collapse
|
47
|
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
|
48
|
Abstract
Monoclonal antibodies are susceptible to chemical and enzymatic modifications during manufacturing, storage, and shipping. Deamidation, isomerization, and oxidation can compromise the potency, efficacy, and safety of therapeutic antibodies. Recently, in silico tools have been used to identify liable residues and engineer antibodies with better chemical stability. Computational approaches for predicting deamidation, isomerization, oxidation, glycation, carbonylation, sulfation, and hydroxylation are reviewed here. Although liable motifs have been used to improve the chemical stability of antibodies, the accuracy of in silico predictions can be improved using machine learning and molecular dynamic simulations. In addition, there are opportunities to improve predictions for specific stress conditions, develop in silico prediction of novel modifications in antibodies, and predict the impact of modifications on physical stability and antigen-binding.
Collapse
Affiliation(s)
- Shabdita Vatsa
- Development Services, Lonza Biologics, Singapore, Singapore
| |
Collapse
|
49
|
Hao Z, Moore B, Ren C, Sadek M, Macchi F, Yang L, Harris J, Yee L, Liu E, Tran V, Ninonuevo M, Chen Y, Yu C. Multi-attribute method performance profile for quality control of monoclonal antibody therapeutics. J Pharm Biomed Anal 2021; 205:114330. [PMID: 34479173 DOI: 10.1016/j.jpba.2021.114330] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/09/2021] [Accepted: 08/14/2021] [Indexed: 11/25/2022]
Abstract
Multi-attribute method (MAM) using peptide map analysis with high resolution mass spectrometry is increasingly common in product characterization and the identification of critical quality attributes (CQAs) of biotherapeutic proteins. Capable of providing structural information specific to amino acid residues, quantifying relative abundance of product variants or degradants, and detecting profile changes between product lots, a robust MAM can replace multiple traditional methods that generate profile-based information for product release and stability testing. In an effort to provide informative and efficient analytical monitoring for monoclonal antibody (mAb) products, from early development to manufacturing quality control, we describe the desired MAM performance profile and address the major scientific challenges in MAM method validation. Furthermore, to support fast speed investigational product development, we describe a platform method validation strategy and results of an optimized MAM workflow. This strategy is applied to support the use of MAM for multiple mAb products with similar structures and physicochemical properties, requiring minimal product-specific method validation activities. Three mAb products were used to demonstrate MAM performance for common and representative product quality attributes. Method validation design and acceptance criteria were guided by the Analytical Target Profile concept, as well as relevant regulatory guidelines to ensure the method is fit-for-purpose. A comprehensive system suitability control strategy was developed, and reported here, to ensure adequate performance of the method including sample preparation, instrument operation, and data analysis. Our results demonstrated sufficient method performance for the characteristics required for quantitative measurement of product variants and degradants.
Collapse
Affiliation(s)
- Zhiqi Hao
- Analytical Development and Quality Control, 1 DNA Way, Genentech, South San Francisco, USA.
| | - Benjamin Moore
- Analytical Development and Quality Control, 1 DNA Way, Genentech, South San Francisco, USA
| | - Chengfeng Ren
- Analytical Development and Quality Control, 1 DNA Way, Genentech, South San Francisco, USA
| | - Monica Sadek
- Analytical Development and Quality Control, 1 DNA Way, Genentech, South San Francisco, USA
| | - Frank Macchi
- Analytical Development and Quality Control, 1 DNA Way, Genentech, South San Francisco, USA
| | - Lindsay Yang
- Analytical Development and Quality Control, 1 DNA Way, Genentech, South San Francisco, USA
| | - Jack Harris
- Analytical Development and Quality Control, 1 DNA Way, Genentech, South San Francisco, USA
| | - Laura Yee
- Analytical Development and Quality Control, 1 DNA Way, Genentech, South San Francisco, USA
| | - Emily Liu
- Analytical Development and Quality Control, 1 DNA Way, Genentech, South San Francisco, USA
| | - Vanessa Tran
- Analytical Development and Quality Control, 1 DNA Way, Genentech, South San Francisco, USA
| | - Milady Ninonuevo
- Analytical Development and Quality Control, 1 DNA Way, Genentech, South San Francisco, USA
| | - Yan Chen
- Analytical Development and Quality Control, 1 DNA Way, Genentech, South San Francisco, USA
| | - Christopher Yu
- Analytical Development and Quality Control, 1 DNA Way, Genentech, South San Francisco, USA.
| |
Collapse
|
50
|
Koudelka T, Winkels K, Kaleja P, Tholey A. Shedding light on both ends: An update on analytical approaches for N- and C-terminomics. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1869:119137. [PMID: 34626679 DOI: 10.1016/j.bbamcr.2021.119137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/27/2021] [Accepted: 09/06/2021] [Indexed: 02/04/2023]
Abstract
Though proteases were long regarded as nonspecific degradative enzymes, over time, it was recognized that they also hydrolyze peptide bonds very specifically with a limited substrate pool. This irreversible posttranslational modification modulates the fate and activity of many proteins, making proteolytic processing a master switch in the regulation of e.g., the immune system, apoptosis and cancer progression. N- and C-terminomics, the identification of protein termini, has become indispensable in elucidating protease substrates and therefore protease function. Further, terminomics has the potential to identify yet unknown proteoforms, e.g. formed by alternative splicing or the recently discovered alternative ORFs. Different strategies and workflows have been developed that achieve higher sensitivity, a greater depth of coverage or higher throughput. In this review, we summarize recent developments in both N- and C-terminomics and include the potential of top-down proteomics which inherently delivers information on both ends of analytes in a single analysis.
Collapse
Affiliation(s)
- Tomas Koudelka
- Systematic Proteome Research & Bioanalytics, Institute for Experimental Medicine, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Konrad Winkels
- Systematic Proteome Research & Bioanalytics, Institute for Experimental Medicine, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Patrick Kaleja
- Systematic Proteome Research & Bioanalytics, Institute for Experimental Medicine, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Andreas Tholey
- Systematic Proteome Research & Bioanalytics, Institute for Experimental Medicine, Christian-Albrechts-Universität zu Kiel, Kiel, Germany.
| |
Collapse
|