1
|
Marie A, Georgescauld F, Johnson KR, Ray S, Engen JR, Ivanov AR. Native Capillary Electrophoresis-Mass Spectrometry of Near 1 MDa Non-Covalent GroEL/GroES/Substrate Protein Complexes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306824. [PMID: 38191978 PMCID: PMC10953559 DOI: 10.1002/advs.202306824] [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: 09/18/2023] [Revised: 12/21/2023] [Indexed: 01/10/2024]
Abstract
Protein complexes are essential for proteins' folding and biological function. Currently, native analysis of large multimeric protein complexes remains challenging. Structural biology techniques are time-consuming and often cannot monitor the proteins' dynamics in solution. Here, a capillary electrophoresis-mass spectrometry (CE-MS) method is reported to characterize, under near-physiological conditions, the conformational rearrangements of ∽1 MDa GroEL upon complexation with binding partners involved in a protein folding cycle. The developed CE-MS method is fast (30 min per run), highly sensitive (low-amol level), and requires ∽10 000-fold fewer samples compared to biochemical/biophysical techniques. The method successfully separates GroEL14 (∽800 kDa), GroEL7 (∽400 kDa), GroES7 (∽73 kDa), and NanA4 (∽130 kDa) oligomers. The non-covalent binding of natural substrate proteins with GroEL14 can be detected and quantified. The technique allows monitoring of GroEL14 conformational changes upon complexation with (ATPγS)4-14 and GroES7 (∽876 kDa). Native CE-pseudo-MS3 analyses of wild-type (WT) GroEL and two GroEL mutants result in up to 60% sequence coverage and highlight subtle structural differences between WT and mutated GroEL. The presented results demonstrate the superior CE-MS performance for multimeric complexes' characterization versus direct infusion ESI-MS. This study shows the CE-MS potential to provide information on binding stoichiometry and kinetics for various protein complexes.
Collapse
Affiliation(s)
- Anne‐Lise Marie
- Barnett Institute of Chemical and Biological AnalysisDepartment of Chemistry and Chemical BiologyNortheastern University360 Huntington AvenueBostonMA02115USA
| | - Florian Georgescauld
- Barnett Institute of Chemical and Biological AnalysisDepartment of Chemistry and Chemical BiologyNortheastern University360 Huntington AvenueBostonMA02115USA
| | - Kendall R. Johnson
- Barnett Institute of Chemical and Biological AnalysisDepartment of Chemistry and Chemical BiologyNortheastern University360 Huntington AvenueBostonMA02115USA
| | - Somak Ray
- Barnett Institute of Chemical and Biological AnalysisDepartment of Chemistry and Chemical BiologyNortheastern University360 Huntington AvenueBostonMA02115USA
| | - John R. Engen
- Barnett Institute of Chemical and Biological AnalysisDepartment of Chemistry and Chemical BiologyNortheastern University360 Huntington AvenueBostonMA02115USA
| | - Alexander R. Ivanov
- Barnett Institute of Chemical and Biological AnalysisDepartment of Chemistry and Chemical BiologyNortheastern University360 Huntington AvenueBostonMA02115USA
| |
Collapse
|
2
|
Poltash ML, Srzentić K, Beil E, Gorre E, Damoc E, Mahan AD, Nanda H, Chowdhury P. Elucidating the Mechanism of Multispecific Antibody Aggregation through Subunit Analysis. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2023; 34:2654-2661. [PMID: 37922506 DOI: 10.1021/jasms.3c00206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2023]
Abstract
Multispecific antibody constructs are quickly becoming more common constructs in biopharmaceuticals to improve specificity and efficacy. While the advent of this technology has led to improved therapeutics, its development has challenged the analytical tools through which these therapeutics are characterized. Moreover, new critical quality attributes, such as aggregation, have challenged the approaches to characterization even further. Herein, we describe a novel native subunit analysis using IdeS and IgdE analyzed by native size exclusion chromatography coupled with mass spectrometry to interrogate the mechanism of aggregation in a multispecific antibody. Digestion by IdeS and IdgE allows for the retention and detection of noncovalent interactions thereafter. Aggregation was localized to single-chain fragment variables (scFvs) wherein a domain swapping mechanism between VH1/VL2 and VH2/VL1 occurs.
Collapse
Affiliation(s)
- Michael L Poltash
- Janssen Pharmaceuticals Research and Development, Spring House, Pennsylvania 19477, United States
| | | | - Eric Beil
- Janssen Pharmaceuticals Research and Development, Spring House, Pennsylvania 19477, United States
| | - Elsa Gorre
- Janssen Pharmaceuticals Research and Development, Spring House, Pennsylvania 19477, United States
| | - Eugen Damoc
- Thermo Fisher Scientific, 28199 Bremen, Germany
| | - Andrew D Mahan
- Janssen Pharmaceuticals Research and Development, Spring House, Pennsylvania 19477, United States
| | - Hirsh Nanda
- Janssen Pharmaceuticals Research and Development, Spring House, Pennsylvania 19477, United States
| | - Partha Chowdhury
- Janssen Pharmaceuticals Research and Development, Spring House, Pennsylvania 19477, United States
| |
Collapse
|
3
|
Ruppen I, Verscheure L, Vandenheede I, Ortiz A, de Melo IS, Liebig T, Sandra P, Beydon ME, Sandra K. Characterization of mAb size heterogeneity originating from a cysteine to tyrosine substitution using denaturing and native LC-MS. J Pharm Biomed Anal 2023; 236:115743. [PMID: 37757547 DOI: 10.1016/j.jpba.2023.115743] [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: 05/30/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023]
Abstract
Upon assessing the comparability between a biosimilar mAb and its reference product by non-reducing CE-SDS, increased levels of a heavy-heavy-light chain (HHL) variant, present as a low molecular weight (LMW) peak, were observed. RPLC-MS applied at top, middle-up and bottom-up level revealed the existence of Cys-to-Tyr substitutions, predominantly at position HC226 involved in connecting LC and HC, explaining the abundant HHL levels. Antigen binding was not impacted by the presence of this size variant suggesting a non-covalent association of Tyr substituted HHL and LC. The latter complex is not maintained in the denaturing conditions associated with CE-SDS and RPLC-MS. Its existence could, nevertheless, be confirmed by native SEC-MS which preserves non-covalent protein interactions during separation and electrospray ionization. Amino acid analysis furthermore demonstrated a depletion of Cys during the fed-batch process indicating that the observed size/sequence variant is not of genetic but rather of metabolic origin. Native SEC-MS showed that supplementing the cell culture medium with Cys halts misincorporation of Tyr and promotes the formation of the desired mAb structure. To the best of our knowledge, Cys-to-Tyr substitutions preventing interchain disulfide bridge formation have not been described earlier. This observation adds to the impressive structural heterogeneity reported to date for mAbs.
Collapse
Affiliation(s)
- Isabel Ruppen
- mAbxience Research, Manuel Pombo Angulo 28, 28050 Madrid, Spain
| | | | | | - Alexia Ortiz
- RIC group, President Kennedypark 26, 8500 Kortrijk, Belgium
| | | | - Timo Liebig
- mAbxience Research, Manuel Pombo Angulo 28, 28050 Madrid, Spain
| | - Pat Sandra
- RIC group, President Kennedypark 26, 8500 Kortrijk, Belgium
| | | | - Koen Sandra
- RIC group, President Kennedypark 26, 8500 Kortrijk, Belgium.
| |
Collapse
|
4
|
Liu GY, Zhang Z, Yan Y, Wang S, Li N. Discovery and Characterization of an Acid-Labile Serine-Lysine Cross-Link in Antibody High-Molecular-Weight Species Using a Multipronged Mass Spectrometry Approach. Anal Chem 2023; 95:13813-13821. [PMID: 37674418 PMCID: PMC10515106 DOI: 10.1021/acs.analchem.3c01602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 08/28/2023] [Indexed: 09/08/2023]
Abstract
Characterizing the cross-links responsible for the covalent high-molecular-weight (HMW) species in therapeutic monoclonal antibodies (mAbs) is of great importance as it not only provides a framework for risk assessment but also offers insights for process improvement. However, owing to the complexity and low abundance, identification of novel and unknown cross-links in mAb products can be very challenging. Here, applying a multipronged MS-based approach, we report the discovery of a novel covalent cross-link formed via an imine bond between lysine and serine residues. In particular, this Ser-Lys cross-link was found to be acid-labile and can be easily overlooked by conventional LC-MS techniques operated at low pH. It is worth noting that although imine-based cross-link has been previously reported in collagen protein cross-linking, this is the first time that a Ser-Lys cross-link has been found in a mAb product that contributes to covalent HMW species formation.
Collapse
Affiliation(s)
- Gao-Yuan Liu
- Analytical Chemistry, Regeneron
Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Zhengqi Zhang
- Analytical Chemistry, Regeneron
Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Yuetian Yan
- Analytical Chemistry, Regeneron
Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Shunhai Wang
- Analytical Chemistry, Regeneron
Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Ning Li
- Analytical Chemistry, Regeneron
Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| |
Collapse
|
5
|
Winter G, Hamp-Goldstein C, Fischer G, Kletting P, Glatting G, Solbach C, Herrmann H, Sala E, Feuring M, Döhner H, Beer AJ, Bunjes D, Prasad V. Optimization of Radiolabeling of a [ 90Y]Y-Anti-CD66-Antibody for Radioimmunotherapy before Allogeneic Hematopoietic Cell Transplantation. Cancers (Basel) 2023; 15:3660. [PMID: 37509321 PMCID: PMC10377894 DOI: 10.3390/cancers15143660] [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: 06/20/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
For patients with acute myeloid leukemia, myelodysplastic syndrome, or acute lymphoblastic leukemia, allogeneic hematopoietic cell transplantation (HCT) is a potentially curative treatment. In addition to standard conditioning regimens for HCT, high-dose radioimmunotherapy (RIT) offers the unique opportunity to selectively deliver a high dose of radiation to the bone marrow while limiting side effects. Modification of a CD66b-specific monoclonal antibody (mAb) with a DTPA-based chelating agent should improve the absorbed dose distribution during therapy. The stability and radioimmunoreactive fraction of the radiolabeled mAbs were determined. Before RIT, all patients underwent dosimetry to determine absorbed doses to bone marrow, kidneys, liver, and spleen. Scans were performed twenty-four hours after therapy for quality control. A radiochemical purity of >95% and acceptable radioimmunoreactivity was achieved. Absorbed organ doses for the liver and kidney were consequently improved compared to reported historical data. All patients tolerated RIT well with no treatment-related acute adverse events. Complete remission could be observed in 4/5 of the patients 3 months after RIT. Two patients developed delayed liver failure unrelated to the radioimmunotherapy. The improved conjugation and radiolabeling procedure resulted in excellent stability, radiochemical purity, and CD66-specific radioimmunoreactivity of 90Y-labeled anti-CD66 mAb. RIT followed by conditioning and HCT was well tolerated. Based on these promising initial data, further prospective studies of [90Y]Y-DTPA-Bn-CHX-A″-anti-CD66-mAb-assisted conditioning in HCT are warranted.
Collapse
Affiliation(s)
- Gordon Winter
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Gabriel Fischer
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Peter Kletting
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Gerhard Glatting
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Christoph Solbach
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Hendrik Herrmann
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Elisa Sala
- Department of Internal Medicine III, Ulm University Medical Center, 89081 Ulm, Germany
| | - Michaela Feuring
- Department of Internal Medicine III, Ulm University Medical Center, 89081 Ulm, Germany
| | - Hartmut Döhner
- Department of Internal Medicine III, Ulm University Medical Center, 89081 Ulm, Germany
| | - Ambros J Beer
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Donald Bunjes
- Department of Internal Medicine III, Ulm University Medical Center, 89081 Ulm, Germany
| | - Vikas Prasad
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
- Mallinckrodt Institute of Radiology, Division of Nuclear Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA
| |
Collapse
|
6
|
Gunawardena HP, Ai Y, Gao J, Zare RN, Chen H. Rapid Characterization of Antibodies via Automated Flow Injection Coupled with Online Microdroplet Reactions and Native-pH Mass Spectrometry. Anal Chem 2023; 95:3340-3348. [PMID: 36656670 PMCID: PMC10492509 DOI: 10.1021/acs.analchem.2c04535] [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] [Indexed: 01/20/2023]
Abstract
Microdroplet reactions have aroused much interest due to significant reaction acceleration (e.g., ultrafast protein digestion in microdroplets could occur in less than 1 ms). This study integrated a microdroplet protein digestion technique with automated sample flow injection and online mass spectrometry (MS) analysis, to develop a rapid and robust method for structural characterization of monoclonal antibodies (mAbs) that is essential to assess the antibody drug's safety and quality. Automated sequential aspiration and mixing of an antibody and an enzyme (IdeS or IgdE) enabled rapid analysis with high reproducibility (total analysis time: 2 min per sample; reproducibility: ∼2% coefficient of variation). Spraying the sample in ammonium acetate buffer (pH 7) using a jet stream source allowed efficient digestion of antibodies and efficient ionization of resulting antibody subunits under native-pH conditions. Importantly, it also provided a platform to directly study specific binding of an antibody and an antigen (e.g., detecting the complexes mAb/RSFV antigen and F(ab')2/RSVF in this study). Furthermore, subsequent tandem MS analysis of a resulting subunit from microdroplet digestion enabled localizing post-translational modifications on particular domains of a mAb in a rapid fashion. In combination with IdeS digestion of an antibody, additional tris(2-carboxyethyl)phosphine (TCEP) reduction and N-glycosidase F (PNGase F) deglycosylation reactions that facilitate antibody analysis could be realized in "one-pot" spraying. Interestingly, increased deglycosylation yield in microdroplets was found, simply by raising the sample temperature. We expect that our method would have a high impact for rapid characterization of monoclonal antibodies.
Collapse
Affiliation(s)
- Harsha P. Gunawardena
- Janssen Research & Development, The Janssen Pharmaceutical Companies of Johnson & Johnson, Spring House, Pennsylvania 19477, USA
| | - Yongling Ai
- Department of Chemistry & Environmental Science, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Jinshan Gao
- Department of Chemistry and Biochemistry, Montclair State University, 1 Normal Ave, Montclair, NJ 07043, USA
| | - Richard N. Zare
- Department of Chemistry, Stanford University, Stanford, California 94305-5080, USA
| | - Hao Chen
- Department of Chemistry & Environmental Science, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| |
Collapse
|
7
|
McClain A, Zhang Y, Yin Y, Wang Q, Hwang LY, Gu Y, Beckman J, Ludwig R. Using Digestion by IdeS Protease to Improve Quantification of Degradants in Monoclonal Antibodies by Non-Reducing Capillary Gel Electrophoresis. Anal Chem 2022; 94:17388-17395. [PMID: 36472948 PMCID: PMC9774260 DOI: 10.1021/acs.analchem.2c02630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022]
Abstract
Monoclonal antibodies (mAbs) have become predominant therapeutics by providing highly specific mechanisms of action enabling treatment of complex diseases. However, mAbs themselves are highly complex and require thorough testing and characterization to ensure efficacy and patient safety. In this regard, fragmentation is a degradation product of concern. The biotechnology industry uses capillary gel electrophoresis (CGE) to quantify fragmentation by electrophoretically resolving size variants, such as products resulting from partial reduction of interchain disulfides. However, standard CGE methods may not adequately separate less typical fragments, particularly when there is minimal size difference to the parent molecule. For mAb-1, a degradant only ∼11 kDa smaller than the intact mAb (∼149 kDa) was unable to be resolved under typical non-reducing conditions, preventing an accurate purity assessment and precluding tracking of product purity within stability studies. To address these deficiencies, a subunit-based non-reducing CGE method was developed to employ IdeS protease to produce F(ab')2 and Fc fragments, which resulted in baseline resolution of the clipped subunit species from its parent species. This enabled more accurate trending of purity throughout stability studies. Method characterization ensured that this subunit method monitored expected impurities observed by intact non-reducing CGE and thus could suitably replace non-reducing CGE in the release and stability testing panel. It also has the potential to replace reducing CGE based on its tracking of the deglycosylated Fc species. We believe this approach of utilizing proteases to develop subunit CGE methods for release and stability can be applied to other molecules when in need of resolving analogous fragments.
Collapse
Affiliation(s)
- Andrew McClain
- Bristol
Myers Squibb, 38 Jackson Rd, Devens, Massachusetts01434, United States
| | - Yiting Zhang
- Bristol
Myers Squibb, 38 Jackson Rd, Devens, Massachusetts01434, United States
| | - Yan Yin
- Bristol
Myers Squibb, 200 Cambridgepark Drive, Cambridge, Massachusetts02140, United States
| | - Qi Wang
- Bristol
Myers Squibb, 38 Jackson Rd, Devens, Massachusetts01434, United States
| | - Lih-Yueh Hwang
- Bristol
Myers Squibb, 556 Morris Ave, Summit, New Jersey07901, United States
| | - Yan Gu
- Bristol
Myers Squibb, 38 Jackson Rd, Devens, Massachusetts01434, United States
| | - Jeff Beckman
- Bristol
Myers Squibb, 38 Jackson Rd, Devens, Massachusetts01434, United States
| | - Richard Ludwig
- Bristol
Myers Squibb, 38 Jackson Rd, Devens, Massachusetts01434, United States
| |
Collapse
|
8
|
Mielecki M, Ziemniak M, Ozga M, Borowski R, Antosik J, Kaczyńska A, Pająk B. Structure-Activity Relationship of the Dimeric and Oligomeric Forms of a Cytotoxic Biotherapeutic Based on Diphtheria Toxin. Biomolecules 2022; 12:biom12081111. [PMID: 36009005 PMCID: PMC9406121 DOI: 10.3390/biom12081111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022] Open
Abstract
Protein aggregation is a well-recognized problem in industrial preparation, including biotherapeutics. These low-energy states constantly compete with a native-like conformation, which is more pronounced in the case of macromolecules of low stability in the solution. A better understanding of the structure and function of such aggregates is generally required for the more rational development of therapeutic proteins, including single-chain fusion cytotoxins to target specific receptors on cancer cells. Here, we identified and purified such particles as side products of the renaturation process of the single-chain fusion cytotoxin, composed of two diphtheria toxin (DT) domains and interleukin 13 (IL-13), and applied various experimental techniques to comprehensively understand their molecular architecture and function. Importantly, we distinguished soluble purified dimeric and fractionated oligomeric particles from aggregates. The oligomers are polydisperse and multimodal, with a distribution favoring lower and even stoichiometries, suggesting they are composed of dimeric building units. Importantly, all these oligomeric particles and the monomer are cystine-dependent as their innate disulfide bonds have structural and functional roles. Their reduction triggers aggregation. Presumably the dimer and lower oligomers represent the metastable state, retaining the native disulfide bond. Although significantly reduced in contrast to the monomer, they preserve some fraction of bioactivity, manifested by their IL-13RA2 receptor affinity and selective cytotoxic potency towards the U-251 glioblastoma cell line. These molecular assemblies probably preserve structural integrity and native-like fold, at least to some extent. As our study demonstrated, the dimeric and oligomeric cytotoxin may be an exciting model protein, introducing a new understanding of its monomeric counterpart’s molecular characteristics.
Collapse
|
9
|
van Schaick G, el Hajjouti N, Nicolardi S, den Hartog J, Jansen R, van der Hoeven R, Bijleveld W, Abello N, Wuhrer M, Olsthoorn MMA, Domínguez-Vega E. Native Liquid Chromatography and Mass Spectrometry to Structurally and Functionally Characterize Endo-Xylanase Proteoforms. Int J Mol Sci 2022; 23:1307. [PMID: 35163230 PMCID: PMC8835838 DOI: 10.3390/ijms23031307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 12/16/2022] Open
Abstract
Xylanases are of great value in various industries, including paper, food, and biorefinery. Due to their biotechnological production, these enzymes can contain a variety of post-translational modifications, which may have a profound effect on protein function. Understanding the structure-function relationship can guide the development of products with optimal performance. We have developed a workflow for the structural and functional characterization of an endo-1,4-β-xylanase (ENDO-I) produced by Aspergillus niger with and without applying thermal stress. This workflow relies on orthogonal native separation techniques to resolve proteoforms. Mass spectrometry and activity assays of separated proteoforms permitted the establishment of structure-function relationships. The separation conditions were focus on balancing efficient separation and protein functionality. We employed size exclusion chromatography (SEC) to separate ENDO-I from other co-expressed proteins. Charge variants were investigated with ion exchange chromatography (IEX) and revealed the presence of low abundant glycated variants in the temperature-stressed material. To obtain better insights into the effect on glycation on function, we enriched for these species using boronate affinity chromatography (BAC). The activity measurements showed lower activity of glycated species compared to the non-modified enzyme. Altogether, this workflow allowed in-depth structural and functional characterization of ENDO-I proteoforms.
Collapse
Affiliation(s)
- Guusje van Schaick
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (N.e.H.); (S.N.); (M.W.); (E.D.-V.)
| | - Nadi el Hajjouti
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (N.e.H.); (S.N.); (M.W.); (E.D.-V.)
| | - Simone Nicolardi
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (N.e.H.); (S.N.); (M.W.); (E.D.-V.)
| | - Joost den Hartog
- Center for Analytical Innovation, DSM, Alexander Fleminglaan 1, 2613 AX Delft, The Netherlands; (J.d.H.); (R.J.); (R.v.d.H.); (W.B.); (N.A.); (M.M.A.O.)
| | - Romana Jansen
- Center for Analytical Innovation, DSM, Alexander Fleminglaan 1, 2613 AX Delft, The Netherlands; (J.d.H.); (R.J.); (R.v.d.H.); (W.B.); (N.A.); (M.M.A.O.)
| | - Rob van der Hoeven
- Center for Analytical Innovation, DSM, Alexander Fleminglaan 1, 2613 AX Delft, The Netherlands; (J.d.H.); (R.J.); (R.v.d.H.); (W.B.); (N.A.); (M.M.A.O.)
| | - Wim Bijleveld
- Center for Analytical Innovation, DSM, Alexander Fleminglaan 1, 2613 AX Delft, The Netherlands; (J.d.H.); (R.J.); (R.v.d.H.); (W.B.); (N.A.); (M.M.A.O.)
| | - Nicolas Abello
- Center for Analytical Innovation, DSM, Alexander Fleminglaan 1, 2613 AX Delft, The Netherlands; (J.d.H.); (R.J.); (R.v.d.H.); (W.B.); (N.A.); (M.M.A.O.)
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (N.e.H.); (S.N.); (M.W.); (E.D.-V.)
| | - Maurien M. A. Olsthoorn
- Center for Analytical Innovation, DSM, Alexander Fleminglaan 1, 2613 AX Delft, The Netherlands; (J.d.H.); (R.J.); (R.v.d.H.); (W.B.); (N.A.); (M.M.A.O.)
| | - Elena Domínguez-Vega
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (N.e.H.); (S.N.); (M.W.); (E.D.-V.)
| |
Collapse
|
10
|
Degterev M, Smolov M, Shukurov R. Improvement of the Degradation Profiling of Eculizumab and Omalizumab Monoclonal Antibodies by Liquid Chromatography–Mass Spectrometry. JOURNAL OF ANALYTICAL CHEMISTRY 2021. [DOI: 10.1134/s1061934821140033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
11
|
Yan Y, Xing T, Liu AP, Zhang Z, Wang S, Li N. Post-Column Denaturation-Assisted Native Size-Exclusion Chromatography-Mass Spectrometry for Rapid and In-Depth Characterization of High Molecular Weight Variants in Therapeutic Monoclonal Antibodies. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2021; 32:2885-2894. [PMID: 34786946 DOI: 10.1021/jasms.1c00289] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The high molecular weight (HMW) size variants present in therapeutic monoclonal antibody (mAb) samples need to be closely monitored and characterized due to their impact on product safety and efficacy. Because of the complexity and often low abundances in final drug substance (DS) samples, characterization of such HMW species is challenging and traditionally requires offline enrichment of the HMW species followed by analysis using various analytical tools. Here, we report the development of a postcolumn denaturation-assisted native SEC-MS method that allows rapid and in-depth characterization of mAb HMW species directly from unfractionated DS samples. This method not only provides high-confidence identification of HMW complexes based on accurate mass measurement of both the intact assembly and the constituent subunits but also allows in-depth analysis of the interaction nature and location. In addition, using the extracted ion chromatograms, derived from high-quality, native-like mass spectra, the elution profiles of each noncovalent and/or nondissociable complex can be readily reconstructed, facilitating the comprehension of a complex HMW profile. The utility of this novel method was demonstrated in different applications, ranging from enriched HMW characterization at late stage development, comparability assessment due to process changes, and forced degradation study of coformulated mAbs. As this method does not require prior enrichment, it is thus desirable for providing both rapid and in-depth characterization of HMW species during the development of therapeutic mAbs.
Collapse
Affiliation(s)
- Yuetian Yan
- Analytical Chemistry Group, Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Tao Xing
- Analytical Chemistry Group, Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Anita P Liu
- Analytical Chemistry Group, Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Zhengqi Zhang
- Analytical Chemistry Group, Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Shunhai Wang
- Analytical Chemistry Group, Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Ning Li
- Analytical Chemistry Group, Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| |
Collapse
|
12
|
Cao M, Jiao Y, Parthemore C, Korman S, Ma J, Hunter A, Kilby G, Chen X. Identification of a CE-SDS shoulder peak as disulfide-linked fragments from common C H2 cleavages in IgGs and IgG-like bispecific antibodies. MAbs 2021; 13:1981806. [PMID: 34719342 PMCID: PMC8565840 DOI: 10.1080/19420862.2021.1981806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Fragmentation is a well-characterized degradation pathway of therapeutic antibodies and is usually monitored by capillary electrophoresis–sodium dodecyl sulfate (CE-SDS). Although fragments due to cleavage in CH2 domains linked by intrachain disulfide bonds are common and can be detected by reduced reversed-phase – liquid chromatography mass spectrometry (RP-LCMS) and reduced CE-SDS methods, their separation in nonreduced CE-SDS (nrCE-SDS) has not been reported but speculated as comigrating with intact IgG. A shoulder peak in nrCE-SDS was observed in the stability samples of an IgG-like bispecific antibody and was determined to be mainly caused by fragments from clipping at the C-terminus of leucine (L)306 or L309 (EU numbering) in the CH2 domain of both heavy chains (HCs) and, to a lesser degree, at the C-terminus of L182 in the CH1 domain of the knob HC. Subunit LCMS analysis verified that the crystallizable fragment contained variants with one or multiple mass additions of ~18 Da due to clipping. Further investigation revealed that CH2 clippings at L306 and L309 were largely due to proteolytic activity, and cleavages were present at various levels in all in-house IgG1 and IgG4 molecules studied. Our study shows that CH2 domain cleavages, with complementary fragments still linked by intrachain disulfide, can be electrophoretically resolved as a front shoulder of the main peak in nrCE-SDS. Given the high occurrence of CH2 cleavages in antibodies, these findings will have broad applicability and could help manufacturers of therapeutic antibodies in process improvement, product characterization, investigations, formulation stability, and stability comparability studies.
Collapse
Affiliation(s)
- Mingyan Cao
- Analytical Sciences, Biopharmaceutical Development, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Yang Jiao
- Analytical Sciences, Biopharmaceutical Development, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Conner Parthemore
- Analytical Sciences, Biopharmaceutical Development, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Samuel Korman
- Analytical Sciences, Biopharmaceutical Development, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Jiao Ma
- Analytical Sciences, Biopharmaceutical Development, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Alan Hunter
- Purification Process Sciences, Biopharmaceutical Development, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Greg Kilby
- Analytical Sciences, Biopharmaceutical Development, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Xiaoyu Chen
- Analytical Sciences, Biopharmaceutical Development, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| |
Collapse
|
13
|
Kumar R, Guttman A, Rathore AS. Applications of capillary electrophoresis for biopharmaceutical product characterization. Electrophoresis 2021; 43:143-166. [PMID: 34591322 DOI: 10.1002/elps.202100182] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/07/2021] [Accepted: 09/23/2021] [Indexed: 12/16/2022]
Abstract
Capillary electrophoresis (CE), after being introduced several decades ago, has carved out a niche for itself in the field of analytical characterization of biopharmaceutical products. It does not only offer fast separation, high resolution in miniaturized format, but equally importantly represents an orthogonal separation mechanism to high-performance liquid chromatography. Therefore, it is not surprising that CE-based methods can be found in all major pharmacopoeias and are recommended for the analysis of biopharmaceutical products during process development, characterization, quality control, and release testing. Different separation formats of CE, such as capillary gel electrophoresis, capillary isoelectric focusing, and capillary zone electrophoresis are widely used for size and charge heterogeneity characterization as well as purity and stability testing of therapeutic proteins. Hyphenation of CE with MS is emerging as a promising bioanalytical tool to assess the primary structure of therapeutic proteins along with any impurities. In this review, we confer the latest developments in capillary electrophoresis, used for the characterization of critical quality attributes of biopharmaceutical products covering the past 6 years (2015-2021). Monoclonal antibodies, due to their significant share in the market, have been given prioritized coverage.
Collapse
Affiliation(s)
- Ramesh Kumar
- Department of Chemical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, India
| | - Andras Guttman
- Horváth Csaba Memorial Laboratories of Bioseparation Sciences, Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Translational Glycomics Group, Research Institute of Biomolecular and Chemical Engineering, University of Pannonia, Veszprem, Hungary
| | - Anurag S Rathore
- Department of Chemical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, India
| |
Collapse
|
14
|
Chen H, Chen JS, Paerhati P, Jakos T, Bai SY, Zhu JW, Yuan YS. Strategies and Applications of Antigen-Binding Fragment (Fab) Production in Escherichia coli. PHARMACEUTICAL FRONTS 2021. [DOI: 10.1055/s-0041-1735145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
AbstractWith the advancement of genetic engineering, monoclonal antibodies (mAbs) have made far-reaching progress in the treatment of various human diseases. However, due to the high cost of production, the increasing demands for antibody-based therapies have not been fully met. Currently, mAb-derived alternatives, such as antigen-binding fragments (Fab), single-chain variable fragments, bispecifics, nanobodies, and conjugated mAbs have emerged as promising new therapeutic modalities. They can be readily prepared in bacterial systems with well-established fermentation technology and ease of manipulation, leading to the reduction of overall cost. This review aims to shed light on the strategies to improve the expression, purification, and yield of Fab fragments in Escherichia coli expression systems, as well as current advances in the applications of Fab fragments.
Collapse
Affiliation(s)
- Hui Chen
- Engineering Research Center of Cell & Therapeutic Antibody, Shanghai Jiao Tong University College of Pharmacy, Ministry of Education, Shanghai, People's Republic of China
| | - Jun-Sheng Chen
- Engineering Research Center of Cell & Therapeutic Antibody, Shanghai Jiao Tong University College of Pharmacy, Ministry of Education, Shanghai, People's Republic of China
| | - Pameila Paerhati
- Engineering Research Center of Cell & Therapeutic Antibody, Shanghai Jiao Tong University College of Pharmacy, Ministry of Education, Shanghai, People's Republic of China
| | - Tanja Jakos
- Engineering Research Center of Cell & Therapeutic Antibody, Shanghai Jiao Tong University College of Pharmacy, Ministry of Education, Shanghai, People's Republic of China
| | - Si-Yi Bai
- Engineering Research Center of Cell & Therapeutic Antibody, Shanghai Jiao Tong University College of Pharmacy, Ministry of Education, Shanghai, People's Republic of China
| | - Jian-Wei Zhu
- Engineering Research Center of Cell & Therapeutic Antibody, Shanghai Jiao Tong University College of Pharmacy, Ministry of Education, Shanghai, People's Republic of China
| | - Yun-Sheng Yuan
- Engineering Research Center of Cell & Therapeutic Antibody, Shanghai Jiao Tong University College of Pharmacy, Ministry of Education, Shanghai, People's Republic of China
| |
Collapse
|
15
|
Verscheure L, Oosterlynck M, Cerdobbel A, Sandra P, Lynen F, Sandra K. Middle-up characterization of monoclonal antibodies by online reduction liquid chromatography-mass spectrometry. J Chromatogr A 2020; 1637:461808. [PMID: 33385741 DOI: 10.1016/j.chroma.2020.461808] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023]
Abstract
This study describes the fully automated middle-up characterization of monoclonal antibodies (mAbs) and next-generation variants by online reduction liquid chromatography-mass spectrometry (LC-MS). Proteins were trapped on-column and subjected to online desalting, denaturation and reduction prior to reversed phase elution of the created subunits in the MS. The evaluation of more than 20 different therapeutic proteins including full length mAbs (subclasses IgG1, IgG2 and IgG4), bispecific antibodies, antibody fragments, fusion proteins and antibody-drug conjugates (ADC) revealed that the online reduction method is as powerful as the widely applied offline sample preparation with dithiothreitol (DTT) as reducing agent and guanidine hydrochloride (Gnd.HCl) as denaturant and tackles some major disadvantages associated with the latter method, i.e. corrosion of stainless steel components, adduct formation impacting spectral quality and sample stability. The value of the online reduction LC-MS method is also enforced by its ability to reveal unstable antibody variants such as succinimide intermediates of asparagine deamidation and aspartic acid isomerization which are often lost when using the offline sample preparation method. The performance of the online reduction LC-MS set-up was verified and it was revealed that the method is precise with RSD values below 0.25% and 3.0% for retention time and area, respectively. Carry-over is within acceptable limits (< 0.5%) and the reducing buffer is stable up to 24 hours.
Collapse
Affiliation(s)
- Liesa Verscheure
- Research Institute for Chromatography (RIC), President Kennedypark 26, 8500 Kortrijk, Belgium; Separation Science Group, Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281-S4, B-9000 Ghent, Belgium
| | - Marie Oosterlynck
- Chemistry Department, KU Leuven, Celestijnenlaan 200F, bus 2404, 3001 Leuven, Belgium
| | - An Cerdobbel
- Research Institute for Chromatography (RIC), President Kennedypark 26, 8500 Kortrijk, Belgium
| | - Pat Sandra
- Research Institute for Chromatography (RIC), President Kennedypark 26, 8500 Kortrijk, Belgium; Separation Science Group, Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281-S4, B-9000 Ghent, Belgium
| | - Frederic Lynen
- Separation Science Group, Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281-S4, B-9000 Ghent, Belgium
| | - Koen Sandra
- Research Institute for Chromatography (RIC), President Kennedypark 26, 8500 Kortrijk, Belgium; Separation Science Group, Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281-S4, B-9000 Ghent, Belgium.
| |
Collapse
|