1
|
Wan J, Wang M, Chen S, Zhang X, Xu W, Wu D, Hu Q, Niu L. Biologically-driven RAFT polymerization-amplified platform for electrochemical detection of antibody drugs. Talanta 2025; 285:127431. [PMID: 39709831 DOI: 10.1016/j.talanta.2024.127431] [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: 10/24/2024] [Revised: 12/11/2024] [Accepted: 12/18/2024] [Indexed: 12/24/2024]
Abstract
The individualized administration and pharmacokinetics profiling are integral to the safe use of antibody drugs in immunotherapy. Here, we propose an electrochemical platform for the highly sensitive and selective detection of antibody drugs, taking advantage of the affinity capture by the peptide mimotopes together with the signal amplification by the biologically-driven RAFT polymerization (BDRP). Briefly, the BDRP-based platform involves the capture of antibody drugs by peptide mimotopes, the labeling of multiple reversible addition-fragmentation chain-transfer (RAFT) agents to the glycan chains of antibody drugs, and the BDRP-enabled controlled recruitment of numerous redox labels. The BDRP-based signal amplification relies on the reduction of RAFT agents by NADH coenzymes into the carbon-centered radicals, which can propagate efficiently into long polymer chains by reacting with the ferrocene-derivated monomers, recruiting numerous redox labels to the glycan chains of antibody drugs. The BDRP is conducted at the physiological temperature (i.e., 37 °C) and in the absence of external stimuli or radical sources, holding the advantages of biological compatibility and desirable simplicity over conventional RAFT polymerization approaches. The developed platform is highly selective and the detection limit in the presence of rituximab as the target was determined to be 0.14 ng/mL. Moreover, the applicability of the BDRP-based platform in the sensitive assay of antibody drugs in serum samples has been validated. In view of the biocompatibility, desirable simplicity, and cost-effectiveness, the BDRP-based platform shows great promise in the quantitative assay of antibody drugs.
Collapse
Affiliation(s)
- Jianwen Wan
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou, 510006, PR China
| | - Mengge Wang
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou, 510006, PR China
| | - Songmin Chen
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou, 510006, PR China
| | - Xiyao Zhang
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou, 510006, PR China
| | - Wenhui Xu
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou, 510006, PR China
| | - Di Wu
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou, 510006, PR China
| | - Qiong Hu
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou, 510006, PR China.
| | - Li Niu
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou, 510006, PR China.
| |
Collapse
|
2
|
Janisse SE, Fellers RT, Raab SA, Goodwin MP, Bowen KP, Lian ZJ, Durbin KR, Senko MW, Compton PD, Kafader JO, Kelleher NL. A Robust and Automated Platform for Charge Detection Mass Spectrometry of Megadalton Biotherapeutics. Anal Chem 2025; 97:4549-4555. [PMID: 39967556 DOI: 10.1021/acs.analchem.4c06397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
Gene therapies based on adeno-associated viruses are an emerging area with high potential to improve human health. Current quality control techniques to assess contaminates and byproducts from the adeno-associated virus (AAV) production pipelines are lacking in robustness and throughput. To address these limitations, we coupled an automated microfluidic device called SampleStream with Orbitrap-based charge detection mass spectrometry (SS-CDMS). We demonstrate that the SS-CDMS workflow performs AAV analysis in under 15 min per sample in a completely autonomous manner. The SS-CDMS workflow enables rapid assessment of key quality control attributes (CQAs), such as of molecular weight and content ratio of AAV formulations with a small sample requirement (<2 × 109 capsids) without being limited by sample concentration. Additionally, this work shows the potential for the SS-CDMS workflow to be implemented at various stages of the production pipeline through effective sample clean up from more complex AAV matrices such as cell culture media.
Collapse
Affiliation(s)
- Samuel E Janisse
- Proteomics Center of Excellence, Northwestern University, Evanston, Illinois 60208, United States
| | - Ryan T Fellers
- Proteomics Center of Excellence, Northwestern University, Evanston, Illinois 60208, United States
- Proteinaceous, Inc, Evanston, Illinois 60201, United States
| | - Shannon A Raab
- Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | | | - Kyle P Bowen
- ThermoFisher Scientific, San Jose, California 95134, United States
| | - Zhirui J Lian
- Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | | | - Michael W Senko
- ThermoFisher Scientific, San Jose, California 95134, United States
| | - Philip D Compton
- Integrated Protein Technologies, Inc, Evanston, Illinois 60201, United States
| | - Jared O Kafader
- Proteomics Center of Excellence, Northwestern University, Evanston, Illinois 60208, United States
| | - Neil L Kelleher
- Proteomics Center of Excellence, Northwestern University, Evanston, Illinois 60208, United States
| |
Collapse
|
3
|
Skinner KA, Fisher TD, Lee A, Su T, Forte E, Sanchez A, Caldwell MA, Kelleher NL. Next-Generation Protein Sequencing and individual ion mass spectrometry enable complementary analysis of interleukin-6. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.637157. [PMID: 39975277 PMCID: PMC11839055 DOI: 10.1101/2025.02.07.637157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The vast complexity of the proteome currently overwhelms any single analytical technology in capturing the full spectrum of proteoform diversity. In this study, we evaluated the complementarity of two cutting-edge proteomic technologies-single-molecule protein sequencing and individual ion mass spectrometry-for analyzing recombinant human IL-6 (rhIL-6) at the amino acid, peptide, and intact proteoform levels. For single-molecule protein sequencing, we employ the recently released Platinum® instrument. Next-Generation Protein Sequencing™ (NGPS™) on Platinum utilizes cycles of N-terminal amino acid recognizer binding and aminopeptidase cleavage to enable parallelized sequencing of single peptide molecules. We found that NGPS produces single amino acid coverage of multiple key regions of IL-6, including two peptides within helices A and C which harbor residues that reportedly impact IL-6 function. For top-down proteoform evaluation, we use individual ion mass spectrometry (I2MS), a highly parallelized orbitrap-based charge detection MS platform. Single ion detection of gas-phase fragmentation products (I2MS2) gives significant sequence coverage in key regions in IL-6, including two regions within helices B and D that are involved in IL-6 signaling. Together, these complementary technologies deliver a combined 52% sequence coverage, offering a more complete view of IL-6 structural and functional diversity than either technology alone. This study highlights the synergy of complementary protein detection methods to more comprehensively cover protein segments relevant to biological interactions.
Collapse
Affiliation(s)
| | - Troy D. Fisher
- Proteomics Center of Excellence, Northwestern University, Evanston, Illinois, United States
| | - Andrew Lee
- Departments of Molecular Biosciences, Chemistry and Chemical and Biological Engineering, Northwestern University, Evanston, IL, United States
| | - Taojunfeng Su
- Departments of Molecular Biosciences, Chemistry and Chemical and Biological Engineering, Northwestern University, Evanston, IL, United States
| | - Eleonora Forte
- Proteomics Center of Excellence, Northwestern University, Evanston, Illinois, United States
- Department of Surgery, Feinberg School of Medicine, Comprehensive Transplant Center, Northwestern University, Chicago, Illinois, United States
| | - Aniel Sanchez
- Proteomics Center of Excellence, Northwestern University, Evanston, Illinois, United States
| | - Michael A. Caldwell
- Proteomics Center of Excellence, Northwestern University, Evanston, Illinois, United States
- Department of Medicine, Division of Hematology Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Neil L. Kelleher
- Proteomics Center of Excellence, Northwestern University, Evanston, Illinois, United States
- Departments of Molecular Biosciences, Chemistry and Chemical and Biological Engineering, Northwestern University, Evanston, IL, United States
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
4
|
Christofi E, O’Hanlon M, Curtis R, Barman A, Keen J, Nagy T, Barran P. Hybrid Mass Spectrometry Applied across the Production of Antibody Biotherapeutics. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2025; 36:44-57. [PMID: 39573914 PMCID: PMC11697328 DOI: 10.1021/jasms.4c00253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/12/2024] [Accepted: 10/01/2024] [Indexed: 01/02/2025]
Abstract
Post expression from the host cells, biotherapeutics undergo downstream processing steps before final formulation. Mass spectrometry and biophysical characterization methods are valuable for examining conformational and stoichiometric changes at these stages, although typically not used in biomanufacturing, where stability is assessed via bulk property studies. Here we apply hybrid MS methods to understand how solution condition changes impact the structural integrity of a biopharmaceutical across the processing pipeline. As an exemplar product, we use the model IgG1 antibody, mAb4. Flexibility, stability, aggregation propensity, and bulk properties are evaluated in relation to perfusion media, purification stages, and formulation solutions. Comparisons with Herceptin, an extensively studied IgG1 antibody, were conducted in a mass spectrometry-compatible solution. Despite presenting similar charge state distributions (CSD) in native MS, mAb4, and Herceptin show distinct unfolding patterns in activated ion mobility mass spectrometry (aIM-MS) and differential scanning fluorimetry (DSF). Herceptin's greater structural stability and aggregation onset temperature (Tagg) are attributed to heavier glycosylation and kappa-class light chains, unlike the lambda-class light chains in mAb4. Hydrogen-deuterium exchange mass spectrometry (HDX-MS) revealed that mAb4 undergoes substantial structural changes during purification, marked by high flexibility, low melting temperature (Tm), and prevalent repulsive protein-protein interactions but transitions to a compact and stable structure in high-salt and formulated environments. Notably, in formulation, the third constant domain (CH3) of the heavy chain retains flexibility and is a region of interest for aggregation. Future work could translate features of interest from comprehensive studies like this to targeted approaches that could be utilized early in the development stage to aid in decision-making regarding targeted mutations or to guide the design space of bioprocesses and formulation choices.
Collapse
Affiliation(s)
- Emilia Christofi
- Michael
Barber Centre for Collaborative Mass Spectrometry, MBCCMS, Princess Street, Manchester M17DN, U.K.
- Manchester
Institute of Biotechnology, University of
Manchester, Princess Street, Manchester M17DN, U.K.
| | - Mark O’Hanlon
- Manchester
Institute of Biotechnology, University of
Manchester, Princess Street, Manchester M17DN, U.K.
| | - Robin Curtis
- Manchester
Institute of Biotechnology, University of
Manchester, Princess Street, Manchester M17DN, U.K.
| | - Arghya Barman
- FUJIFILM
Diosynth Biotechnologies, Belasis Ave, Stockton-on-Tees, Billingham TS23 1LH, U.K.
| | - Jeff Keen
- FUJIFILM
Diosynth Biotechnologies, Belasis Ave, Stockton-on-Tees, Billingham TS23 1LH, U.K.
| | - Tibor Nagy
- FUJIFILM
Diosynth Biotechnologies, Belasis Ave, Stockton-on-Tees, Billingham TS23 1LH, U.K.
| | - Perdita Barran
- Michael
Barber Centre for Collaborative Mass Spectrometry, MBCCMS, Princess Street, Manchester M17DN, U.K.
- Manchester
Institute of Biotechnology, University of
Manchester, Princess Street, Manchester M17DN, U.K.
| |
Collapse
|
5
|
Niu B, Lee B, Chen W, Alberto C, Betancourt Moreira K, Compton P, Homan K, Pinckney J, Zhu Y, Vendel M, Wetterhorn K, Walrond S, Santha E, Horowitz A, Zaubi N, Johnson J. End-To-End Automated Intact Protein Mass Spectrometry for High-Throughput Screening and Characterization of Bispecific and Multispecific Antibodies. Anal Chem 2024; 96:18287-18300. [PMID: 39479787 PMCID: PMC11562946 DOI: 10.1021/acs.analchem.4c04833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 11/13/2024]
Abstract
Bispecific antibodies (bsAbs) and multispecific antibodies (msAbs) represent a promising frontier in therapeutic antibody development, offering unique capabilities not achievable with traditional monoclonal antibodies. Despite their potential, significant challenges remain due to their increased molecular complexity. One prominent challenge is the correct assembly of light and heavy chains, as improper pairing leads to mispaired or incompletely assembled species that lack therapeutic efficacy and possess undesired properties, impairing the developability, manufacturability, and safety. There is a critical need for rapid, sensitive analytical tools to monitor and control these undesired species and ensure the quality assessment of bsAbs and msAbs. To address this need, we present a novel high-throughput, format-agnostic intact mass workflow that significantly enhances the efficiency of detecting and quantifying biotherapeutic products and related impurities. This workflow integrates automated sample preparation, novel high-resolution rapid mass detection powered by SampleStream-MS, and an advanced data analysis pipeline. It offers increased throughput and data quality while substantially reducing analysis turnover time and labor. This was demonstrated in a pilot program where ∼800 multispecific antibodies were processed in 10 working days. The article details the evaluation and validation of our method, demonstrating its repeatability and intermediate precision in terms of measurement accuracy and relative quantification of various product-related species. We underscore the transformative potential of this end-to-end high-throughput workflow in expediting bispecific and multispecific antibody discovery, optimizing production processes, and ensuring high-quality development and manufacturing for therapeutic antibodies.
Collapse
Affiliation(s)
- Ben Niu
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Benjamin Lee
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Wen Chen
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Cristian Alberto
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Karen Betancourt Moreira
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Philip Compton
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Kristoff Homan
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Jason Pinckney
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Yaxing Zhu
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Michelle Vendel
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Karl Wetterhorn
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Shana Walrond
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Esrath Santha
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Amanda Horowitz
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Nicole Zaubi
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| | - Jeffrey Johnson
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, California 92121, United States
| |
Collapse
|
6
|
Des Soye BJ, Melani RD, Hollas MAR, Duan J, Patrie SM, Fisher TD, Mattamana BB, Daud A, Pinelli DF, Ladner DP, Kelleher NL, Forte E. Characterization of the Antibody Response to SARS-CoV-2 Infection in COVID-19 Transplant versus Nontransplant Recipients by Ig-MS. J Proteome Res 2024; 23:3944-3957. [PMID: 39146476 PMCID: PMC11812117 DOI: 10.1021/acs.jproteome.4c00285] [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: 08/17/2024]
Abstract
Solid organ transplant recipients with immunosuppressant regimens to prevent rejection are less able to mount effective immune responses to pathogenic infection. Here, we apply a recently reported mass spectrometry-based serological approach known as Ig-MS to characterize immune responses against infection with SARS-CoV-2 in cohorts of transplant recipients and immunocompetent controls, both at a single early time point following COVID-19 diagnosis as well as over the course of one-month postdiagnosis. We found that the antibody repertoires generated by transplant recipients against SARS-CoV-2 do not differ significantly compared to immunocompetent individuals with regard to repertoire titer, clonality, or glycan composition. Importantly, our study is the first to characterize the evolution of antibody glycan profiles in transplant recipients with COVID-19 disease, presenting evidence that the evolution of glycan composition in these immunocompromised individuals is similar to that in immunocompetent people.
Collapse
Affiliation(s)
- Benjamin J Des Soye
- Proteomics Center of Excellence, Northwestern University, Evanston, Illinois 60208, United States
| | - Rafael D Melani
- Proteomics Center of Excellence, Northwestern University, Evanston, Illinois 60208, United States
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Michael A R Hollas
- Proteomics Center of Excellence, Northwestern University, Evanston, Illinois 60208, United States
| | - Jiana Duan
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Steven M Patrie
- Proteomics Center of Excellence, Northwestern University, Evanston, Illinois 60208, United States
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Troy D Fisher
- Proteomics Center of Excellence, Northwestern University, Evanston, Illinois 60208, United States
| | - Basil Baby Mattamana
- Proteomics Center of Excellence, Northwestern University, Evanston, Illinois 60208, United States
| | - Amna Daud
- Department of Surgery, Feinberg School of Medicine, Comprehensive Transplant Center, Northwestern University, Chicago, Illinois 60611, United States
| | - David F Pinelli
- Department of Surgery, Feinberg School of Medicine, Comprehensive Transplant Center, Northwestern University, Chicago, Illinois 60611, United States
| | - Daniela P Ladner
- Department of Surgery, Feinberg School of Medicine, Comprehensive Transplant Center, Northwestern University, Chicago, Illinois 60611, United States
| | - Neil L Kelleher
- Proteomics Center of Excellence, Northwestern University, Evanston, Illinois 60208, United States
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Eleonora Forte
- Proteomics Center of Excellence, Northwestern University, Evanston, Illinois 60208, United States
- Department of Surgery, Feinberg School of Medicine, Comprehensive Transplant Center, Northwestern University, Chicago, Illinois 60611, United States
| |
Collapse
|
7
|
Wan J, Tian Y, Wu D, Ye Z, Chen S, Hu Q, Wang M, Lv J, Xu W, Zhang X, Han D, Niu L. Site-Directed Electrochemical Grafting for Amplified Detection of Antibody Pharmaceuticals. Anal Chem 2024; 96:9278-9284. [PMID: 38768425 DOI: 10.1021/acs.analchem.4c01798] [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: 05/22/2024]
Abstract
Antibody pharmaceuticals have become the most popular immunotherapeutic drugs and are often administered with low serum drug dosages. Hence, the development of a highly sensitive method for the quantitative assay of antibody levels is of great importance to individualized therapy. On the basis of the dual signal amplification by the glycan-initiated site-directed electrochemical grafting of polymer chains (glyGPC), we report herein a novel strategy for the amplified electrochemical detection of antibody pharmaceuticals. The target of interest was affinity captured by a DNA aptamer ligand, and then the glycans of antibody pharmaceuticals were decorated with the alkyl halide initiators (AHIs) via boronate cross-linking, followed by the electrochemical grafting of the ferrocenyl polymer chains from the glycans of antibody pharmaceuticals through the electrochemically controlled atom transfer radical polymerization (eATRP). As the glycans can be decorated with multiple AHIs and the grafted polymer chains are composed of tens to hundreds of electroactive tags, the glyGPC-based strategy permits the dually amplified electrochemical detection of antibody pharmaceuticals. In the presence of trastuzumab (Herceptin) as the target, the glyGPC-based strategy achieved a detection limit of 71.5 pg/mL. Moreover, the developed method is highly selective, and the results of the quantitative assay of trastuzumab levels in human serum are satisfactory. Owing to its uncomplicated operation and cost-effectiveness, the glyGPC-based strategy shows great promise in the amplified electrochemical detection of antibody pharmaceuticals.
Collapse
Affiliation(s)
- Jianwen Wan
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Yiyan Tian
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Di Wu
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Zhuojun Ye
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Songmin Chen
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Qiong Hu
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
- School of Chemistry and Chemical Engineering, Anshun University, Anshun 561000, P. R. China
| | - Mengge Wang
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Junpeng Lv
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Wenhui Xu
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Xiyao Zhang
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Dongxue Han
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Li Niu
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| |
Collapse
|
8
|
Peters-Clarke TM, Coon JJ, Riley NM. Instrumentation at the Leading Edge of Proteomics. Anal Chem 2024; 96:7976-8010. [PMID: 38738990 PMCID: PMC11996003 DOI: 10.1021/acs.analchem.3c04497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Affiliation(s)
- Trenton M. Peters-Clarke
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Joshua J. Coon
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, USA
- Morgridge Institute for Research, Madison, WI, USA
| | | |
Collapse
|
9
|
Thibert S, Reid DJ, Wilson JW, Varikoti R, Maltseva N, Schultz KJ, Kruel A, Babnigg G, Joachimiak A, Kumar N, Zhou M. Native Mass Spectrometry Dissects the Structural Dynamics of an Allosteric Heterodimer of SARS-CoV-2 Nonstructural Proteins. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:912-921. [PMID: 38535992 PMCID: PMC11066969 DOI: 10.1021/jasms.3c00453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/02/2024] [Accepted: 02/07/2024] [Indexed: 05/02/2024]
Abstract
Structure-based drug design, which relies on precise understanding of the target protein and its interaction with the drug candidate, is dramatically expedited by advances in computational methods for candidate prediction. Yet, the accuracy needs to be improved with more structural data from high throughput experiments, which are challenging to generate, especially for dynamic and weak associations. Herein, we applied native mass spectrometry (native MS) to rapidly characterize ligand binding of an allosteric heterodimeric complex of SARS-CoV-2 nonstructural proteins (nsp) nsp10 and nsp16 (nsp10/16), a complex essential for virus survival in the host and thus a desirable drug target. Native MS showed that the dimer is in equilibrium with monomeric states in solution. Consistent with the literature, well characterized small cosubstrate, RNA substrate, and product bind with high specificity and affinity to the dimer but not the free monomers. Unsuccessfully designed ligands bind indiscriminately to all forms. Using neutral gas collision, the nsp16 monomer with bound cosubstrate can be released from the holo dimer complex, confirming the binding to nsp16 as revealed by the crystal structure. However, we observed an unusual migration of the endogenous zinc ions bound to nsp10 to nsp16 after collisional dissociation. The metal migration can be suppressed by using surface collision with reduced precursor charge states, which presumably resulted in minimal gas-phase structural rearrangement and highlighted the importance of complementary techniques. With minimal sample input (∼μg), native MS can rapidly detect ligand binding affinities and locations in dynamic multisubunit protein complexes, demonstrating the potential of an "all-in-one" native MS assay for rapid structural profiling of protein-to-AI-based compound systems to expedite drug discovery.
Collapse
Affiliation(s)
- Stephanie
M. Thibert
- Environmental
Molecular Sciences Laboratory, Pacific Northwest
National Laboratory, Richland, Washington 99354, United States
| | - Deseree J. Reid
- Chemical
and Biological Signature Sciences, Pacific
Northwest National Laboratory, Richland, Washington 99354, United States
| | - Jesse W. Wilson
- Environmental
Molecular Sciences Laboratory, Pacific Northwest
National Laboratory, Richland, Washington 99354, United States
| | - Rohith Varikoti
- Biological
Sciences Division, Pacific Northwest National
Laboratory, Richland, Washington 99354, United States
| | - Natalia Maltseva
- Center
for Structural Biology of Infectious Diseases, Consortium for Advanced
Science and Engineering, University of Chicago, Chicago, Illinois 60637, United States
- Structural
Biology Center, X-ray Science Division, Argonne National Laboratory, Argonne, Illinois 60439, United States
| | - Katherine J. Schultz
- Biological
Sciences Division, Pacific Northwest National
Laboratory, Richland, Washington 99354, United States
| | - Agustin Kruel
- Biological
Sciences Division, Pacific Northwest National
Laboratory, Richland, Washington 99354, United States
| | - Gyorgy Babnigg
- Center
for Structural Biology of Infectious Diseases, Consortium for Advanced
Science and Engineering, University of Chicago, Chicago, Illinois 60637, United States
- Biosciences
Division, Argonne National Laboratory, Argonne, Illinois 60439, United States
| | - Andrzej Joachimiak
- Center
for Structural Biology of Infectious Diseases, Consortium for Advanced
Science and Engineering, University of Chicago, Chicago, Illinois 60637, United States
- Structural
Biology Center, X-ray Science Division, Argonne National Laboratory, Argonne, Illinois 60439, United States
| | - Neeraj Kumar
- Biological
Sciences Division, Pacific Northwest National
Laboratory, Richland, Washington 99354, United States
| | - Mowei Zhou
- Environmental
Molecular Sciences Laboratory, Pacific Northwest
National Laboratory, Richland, Washington 99354, United States
| |
Collapse
|
10
|
Drown BS, Gupta R, McGee JP, Hollas MAR, Hergenrother PJ, Kafader JO, Kelleher NL. Precise Readout of MEK1 Proteoforms upon MAPK Pathway Modulation by Individual Ion Mass Spectrometry. Anal Chem 2024; 96:4455-4462. [PMID: 38458998 PMCID: PMC11008683 DOI: 10.1021/acs.analchem.3c04758] [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] [Indexed: 03/10/2024]
Abstract
The functions of proteins bearing multiple post-translational modifications (PTMs) are modulated by their modification patterns, yet precise characterization of them is difficult. MEK1 (also known as MAP2K1) is one such example that acts as a gatekeeper of the mitogen-activating protein kinase (MAPK) pathway and propagates signals via phosphorylation by upstream kinases. In principle, top-down mass spectrometry can precisely characterize whole MEK1 proteoforms, but fragmentation methods that would enable the site-specific characterization of labile modifications on 43 kDa protein ions result in overly dense tandem mass spectra. By using the charge-detection method called individual ion mass spectrometry, we demonstrate how complex mixtures of phosphoproteoforms and their fragment ions can be reproducibly handled to provide a "bird's eye" view of signaling activity through mapping proteoform landscapes in a pathway. Using this approach, the overall stoichiometry and distribution of 0-4 phosphorylations on MEK1 was determined in a cellular model of drug-resistant metastatic melanoma. This approach can be generalized to other multiply modified proteoforms, for which PTM combinations are key to their function and drug action.
Collapse
Affiliation(s)
- Bryon S Drown
- Proteomics Center of Excellence, Departments of Molecular Biosciences, Chemistry, and the Feinberg School of Medicine, Northwestern University, Evanston, Illinois 60202, United States
| | - Raveena Gupta
- Proteomics Center of Excellence, Departments of Molecular Biosciences, Chemistry, and the Feinberg School of Medicine, Northwestern University, Evanston, Illinois 60202, United States
| | - John P McGee
- Proteomics Center of Excellence, Departments of Molecular Biosciences, Chemistry, and the Feinberg School of Medicine, Northwestern University, Evanston, Illinois 60202, United States
| | - Michael A R Hollas
- Proteomics Center of Excellence, Departments of Molecular Biosciences, Chemistry, and the Feinberg School of Medicine, Northwestern University, Evanston, Illinois 60202, United States
| | - Paul J Hergenrother
- Department of Chemistry, Carl R. Woese Institute for Genomic Biology, Cancer Center at Illinois, University of Illinois at Urbana─Champaign, Urbana, Illinois 61801, United States
| | - Jared O Kafader
- Proteomics Center of Excellence, Departments of Molecular Biosciences, Chemistry, and the Feinberg School of Medicine, Northwestern University, Evanston, Illinois 60202, United States
| | - Neil L Kelleher
- Proteomics Center of Excellence, Departments of Molecular Biosciences, Chemistry, and the Feinberg School of Medicine, Northwestern University, Evanston, Illinois 60202, United States
| |
Collapse
|
11
|
Harvey SR, Gadkari VV, Ruotolo BT, Russell DH, Wysocki VH, Zhou M. Expanding Native Mass Spectrometry to the Masses. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:646-652. [PMID: 38303101 PMCID: PMC11821079 DOI: 10.1021/jasms.3c00352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
At the 33rd ASMS Sanibel Meeting, on Membrane Proteins and Their Complexes, a morning roundtable discussion was held discussing the current challenges facing the field of native mass spectrometry and approaches to expanding the field to nonexperts. This Commentary summarizes the discussion and current initiatives to address these challenges.
Collapse
Affiliation(s)
- Sophie R. Harvey
- Department of Chemistry and Biochemistry, Native Mass Spectrometry Guided Structural Biology Center, The Ohio State University, Columbus, OH, USA, 43210
| | - Varun V. Gadkari
- Department of Chemistry, University of Minnesota, Minneapolis, MN, USA, 55455
| | - Brandon T. Ruotolo
- Department of Chemistry, University of Michigan, Ann Arbor, MI, USA, 48109
| | - David H. Russell
- Department of Chemistry, Texas A&M University, College Station, TX, USA, 77844
| | - Vicki H. Wysocki
- Department of Chemistry and Biochemistry, Native Mass Spectrometry Guided Structural Biology Center, The Ohio State University, Columbus, OH, USA, 43210
| | - Mowei Zhou
- Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, China, 310058
| |
Collapse
|
12
|
Yang Y, Rao R, Valliere-Douglass J, Tremintin G. Automated high-throughput buffer exchange platform enhances rapid flow analysis of antibody drug conjugates by high resolution mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1235:124007. [PMID: 38387340 DOI: 10.1016/j.jchromb.2024.124007] [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: 06/14/2023] [Revised: 12/10/2023] [Accepted: 01/06/2024] [Indexed: 02/24/2024]
Abstract
Antibody drug conjugates (ADCs) are an increasingly important therapeutic class of molecules for the treatment of cancer. Average drug-to-antibody ratio (DAR) and drug-load distribution are critical quality attributes of ADCs with the potential to impact efficacy and toxicity of the molecule and need to be analytically characterized and understood. Several platform methods including hydrophobic interaction chromatography (HIC) and native size-exclusion chromatography-mass spectrometry (nSEC-MS) have been developed for that purpose; however, each presents some limitations. In this work, we assessed a new sample preparation and buffer exchange platform coupled with high-resolution mass spectrometry for characterizing the drug-load and distribution of several cysteine-linked ADCs conjugated with a variety of chemotypes. Several criteria were evaluated during the optimization of the buffer exchange-mass spectrometry system performance and the data generated with the system were compared with results from nSEC-MS and HIC. The results indicated that the platform enables automated and high throughput quantitative DAR characterization for antibody-drug conjugates with high reproducibility and offers several key advantages over existing approaches that are used for chemotype-agnostic ADC characterization.
Collapse
Affiliation(s)
- Yun Yang
- Bruker Scientific, LLC., 101 Daggett Drive, San Jose, CA, USA.
| | - Romesh Rao
- Analytical Sciences, Seagen Inc., 21823 30th Drive S.E., Bothell, WA, USA.
| | | | | |
Collapse
|
13
|
Dhenin J, Lafont V, Dupré M, Krick A, Mauriac C, Chamot-Rooke J. Monitoring mAb proteoforms in mouse plasma using an automated immunocapture combined with top-down and middle-down mass spectrometry. Proteomics 2024; 24:e2300069. [PMID: 37480175 DOI: 10.1002/pmic.202300069] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/27/2023] [Accepted: 07/10/2023] [Indexed: 07/23/2023]
Abstract
Monoclonal antibodies (mAbs) have established themselves as the leading biopharmaceutical therapeutic modality. Once the developability of a mAb drug candidate has been assessed, an important step is to check its in vivo stability through pharmacokinetics (PK) studies. The gold standard is ligand-binding assay (LBA) and liquid chromatography-mass spectrometry (LC-MS) performed at the peptide level (bottom-up approach). However, these analytical techniques do not allow to address the different mAb proteoforms that can arise from biotransformation. In recent years, top-down and middle-down mass spectrometry approaches have gained popularity to characterize proteins at the proteoform level but are not yet widely used for PK studies. We propose here a workflow based on an automated immunocapture followed by top-down and middle-down liquid chromatography-tandem mass spectrometry (LC-MS/MS) approaches to characterize mAb proteoforms spiked in mouse plasma. We demonstrate the applicability of our workflow on a large concentration range using pembrolizumab as a model. We also compare the performance of two state-of-the-art Orbitrap platforms (Tribrid Eclipse and Exploris 480) for these studies. The added value of our workflow for an accurate and sensitive characterization of mAb proteoforms in mouse plasma is highlighted.
Collapse
Affiliation(s)
- Jonathan Dhenin
- Institut Pasteur, Université Paris Cité, CNRS UAR2024, Mass Spectrometry for Biology, Paris, France
- Université Paris Cité, Sorbonne Paris Cité, Paris, France
- DMPK, Sanofi R&D, Chilly-Mazarin, France
| | | | | | | | | | - Julia Chamot-Rooke
- Institut Pasteur, Université Paris Cité, CNRS UAR2024, Mass Spectrometry for Biology, Paris, France
| |
Collapse
|
14
|
Shi RL, Dillon MA, Compton PD, Sawyer WS, Thorup JR, Kwong M, Chan P, Chiu CPC, Li R, Yadav R, Lee GY, Gober JG, Li Z, ElSohly AM, Ovacik AM, Koerber JT, Spiess C, Josephs JL, Tran JC. High-Throughput Analyses of Therapeutic Antibodies Using High-Field Asymmetric Waveform Ion Mobility Spectrometry Combined with SampleStream and Intact Protein Mass Spectrometry. Anal Chem 2023; 95:17263-17272. [PMID: 37956201 DOI: 10.1021/acs.analchem.3c03158] [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: 11/15/2023]
Abstract
Intact protein mass spectrometry (MS) coupled with liquid chromatography was applied to characterize the pharmacokinetics and stability profiles of therapeutic proteins. However, limitations from chromatography, including throughput and carryover, result in challenges with handling large sample numbers. Here, we combined intact protein MS with multiple front-end separations, including affinity capture, SampleStream, and high-field asymmetric waveform ion mobility spectrometry (FAIMS), to perform high-throughput and specific mass measurements of a multivalent antibody with one antigen-binding fragment (Fab) fused to an immunoglobulin G1 (IgG1) antibody. Generic affinity capture ensures the retention of both intact species 1Fab-IgG1 and the tentative degradation product IgG1. Subsequently, the analytes were directly loaded into SampleStream, where each injection occurs within ∼30 s. By separating ions prior to MS detection, FAIMS further offered improvement in signal-overnoise by ∼30% for denatured protein MS via employing compensation voltages that were optimized for different antibody species. When enhanced FAIMS transmission of 1Fab-IgG1 was employed, a qualified assay was established for spiked-in serum samples between 0.1 and 25 μg/mL, resulting in ∼10% accuracy bias and precision coefficient of variation. Selective FAIMS transmission of IgG1 as the degradation surrogate product enabled more sensitive detection of clipped species for intact 1Fab-IgG1 at 5 μg/mL in serum, generating an assay to measure 1Fab-IgG1 truncation between 2.5 and 50% with accuracy and precision below 20% bias and coefficient of variation. Our results revealed that the SampleStream-FAIMS-MS platform affords high throughput, selectivity, and sensitivity for characterizing therapeutic antibodies from complex biomatrices qualitatively and quantitatively.
Collapse
Affiliation(s)
- Rachel Liuqing Shi
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California 94080, United States
| | - Michael A Dillon
- Department of Antibody Engineering, Genentech, Inc., South San Francisco, California 94080, United States
| | - Philip D Compton
- Integrated Protein Technologies, Evanston, Illinois 60201, United States
| | - William S Sawyer
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California 94080, United States
| | - John R Thorup
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California 94080, United States
| | - Mandy Kwong
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California 94080, United States
| | - Pamela Chan
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California 94080, United States
| | - Cecilia P C Chiu
- Department of Antibody Engineering, Genentech, Inc., South San Francisco, California 94080, United States
| | - Ran Li
- Department of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc., South San Francisco, California 94080, United States
| | - Rajbharan Yadav
- Department of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc., South San Francisco, California 94080, United States
| | - Genee Y Lee
- Department of Molecular Oncology, Genentech Inc., South San Francisco, California 94080, United States
| | - Joshua G Gober
- Department of Protein Chemistry, Genentech Inc., South San Francisco, California 94080, United States
| | - Zhiyu Li
- The DMPK Service Department, WuXi AppTec Inc., Shanghai 200131, China
| | - Adel M ElSohly
- Department of Protein Chemistry, Genentech Inc., South San Francisco, California 94080, United States
| | - Ayse Meric Ovacik
- Department of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc., South San Francisco, California 94080, United States
| | - James T Koerber
- Department of Antibody Engineering, Genentech, Inc., South San Francisco, California 94080, United States
| | - Christoph Spiess
- Department of Antibody Engineering, Genentech, Inc., South San Francisco, California 94080, United States
| | - Jonathan L Josephs
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California 94080, United States
| | - John C Tran
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California 94080, United States
| |
Collapse
|
15
|
Wan J, Liang Y, Hu Q, Liang Z, Feng W, Tian Y, Li S, Ye Z, Hong M, Han D, Niu L. Amplification-Free Ratiometric Electrochemical Aptasensor for Point-of-Care Detection of Therapeutic Monoclonal Antibodies. Anal Chem 2023; 95:14094-14100. [PMID: 37672684 DOI: 10.1021/acs.analchem.3c03052] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
The rapid quantification of therapeutic monoclonal antibodies (mAbs) is of great significance to their pharmacokinetics/pharmacodynamics (PK/PD) research and the personalized medication for disease treatment. Taking advantage of the direct decoration of tens of redox tags to the target of interest, we illustrate herein an amplification-free ratiometric electrochemical aptasensor for the point-of-care (POC) detection of trace amounts of therapeutic mAbs. The POC detection of therapeutic mAbs involved the use of the methylene blue (MB)-conjugated aptamer as the affinity element and the decoration of therapeutic mAbs with ferrocene (Fc) tags via the boronate crosslinking, in which the MB-derived peak current was used as the reference signal, and the peak current of the Fc tag was used as the output signal. As each therapeutic mAb carries tens of diol sites for the site-specific decoration of the Fc output tags, the boronate crosslinking enabled the amplification-free detection, which is cost-effective and quite simple in operation. In the presence of bevacizumab (BevMab) as the target, the resulting ratiometric signal (i.e., the IFc/IMB value) exhibited a good linear response over the range of 0.025-2.5 μg/mL, and the limit of detection (LOD) of the electrochemical aptasensor was 6.5 ng/mL. Results indicated that the aptamer-based affinity recognition endowed the detection of therapeutic mAbs with high selectivity, while the ratiometric readout exhibited satisfactory reproducibility and robustness. Moreover, the ratiometric electrochemical aptasensor is applicable to the detection of therapeutic mAbs in serum samples. Taking together, the amplification-free ratiometric electrochemical aptasensor holds great promise in the POC detection of therapeutic mAbs.
Collapse
Affiliation(s)
- Jianwen Wan
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Yiyi Liang
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Qiong Hu
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Zhiwen Liang
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Wenxing Feng
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Yiyan Tian
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Shiqi Li
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Zhuojun Ye
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Mingru Hong
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Dongxue Han
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Li Niu
- School of Chemical Engineering and Technology, Sun Yat-sen University, Zhuhai 519082, P. R. China
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials and Devices, Guangdong Engineering Technology Research Center for Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| |
Collapse
|
16
|
Liu C. High-throughput MS for intact protein analysis. Bioanalysis 2023; 15:1017-1019. [PMID: 37584366 DOI: 10.4155/bio-2023-0139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2023] Open
|
17
|
Reid DJ, Thibert S, Zhou M. Dissecting the structural heterogeneity of proteins by native mass spectrometry. Protein Sci 2023; 32:e4612. [PMID: 36851867 PMCID: PMC10031758 DOI: 10.1002/pro.4612] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/01/2023]
Abstract
A single gene yields many forms of proteins via combinations of posttranscriptional/posttranslational modifications. Proteins also fold into higher-order structures and interact with other molecules. The combined molecular diversity leads to the heterogeneity of proteins that manifests as distinct phenotypes. Structural biology has generated vast amounts of data, effectively enabling accurate structural prediction by computational methods. However, structures are often obtained heterologously under homogeneous states in vitro. The lack of native heterogeneity under cellular context creates challenges in precisely connecting the structural data to phenotypes. Mass spectrometry (MS) based proteomics methods can profile proteome composition of complex biological samples. Most MS methods follow the "bottom-up" approach, which denatures and digests proteins into short peptide fragments for ease of detection. Coupled with chemical biology approaches, higher-order structures can be probed via incorporation of covalent labels on native proteins that are maintained at the peptide level. Alternatively, native MS follows the "top-down" approach and directly analyzes intact proteins under nondenaturing conditions. Various tandem MS activation methods can dissect the intact proteins for in-depth structural elucidation. Herein, we review recent native MS applications for characterizing heterogeneous samples, including proteins binding to mixtures of ligands, homo/hetero-complexes with varying stoichiometry, intrinsically disordered proteins with dynamic conformations, glycoprotein complexes with mixed modification states, and active membrane protein complexes in near-native membrane environments. We summarize the benefits, challenges, and ongoing developments in native MS, with the hope to demonstrate an emerging technology that complements other tools by filling the knowledge gaps in understanding the molecular heterogeneity of proteins.
Collapse
Affiliation(s)
- Deseree J. Reid
- Chemical and Biological Signature SciencesPacific Northwest National LaboratoryRichlandWashingtonUSA
| | - Stephanie Thibert
- Environmental Molecular Sciences LaboratoryPacific Northwest National LaboratoryRichlandWashingtonUSA
| | - Mowei Zhou
- Environmental Molecular Sciences LaboratoryPacific Northwest National LaboratoryRichlandWashingtonUSA
| |
Collapse
|
18
|
Nickerson JL, Baghalabadi V, Rajendran SRCK, Jakubec PJ, Said H, McMillen TS, Dang Z, Doucette AA. Recent advances in top-down proteome sample processing ahead of MS analysis. MASS SPECTROMETRY REVIEWS 2023; 42:457-495. [PMID: 34047392 DOI: 10.1002/mas.21706] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/21/2021] [Accepted: 05/06/2021] [Indexed: 06/12/2023]
Abstract
Top-down proteomics is emerging as a preferred approach to investigate biological systems, with objectives ranging from the detailed assessment of a single protein therapeutic, to the complete characterization of every possible protein including their modifications, which define the human proteoform. Given the controlling influence of protein modifications on their biological function, understanding how gene products manifest or respond to disease is most precisely achieved by characterization at the intact protein level. Top-down mass spectrometry (MS) analysis of proteins entails unique challenges associated with processing whole proteins while maintaining their integrity throughout the processes of extraction, enrichment, purification, and fractionation. Recent advances in each of these critical front-end preparation processes, including minimalistic workflows, have greatly expanded the capacity of MS for top-down proteome analysis. Acknowledging the many contributions in MS technology and sample processing, the present review aims to highlight the diverse strategies that have forged a pathway for top-down proteomics. We comprehensively discuss the evolution of front-end workflows that today facilitate optimal characterization of proteoform-driven biology, including a brief description of the clinical applications that have motivated these impactful contributions.
Collapse
Affiliation(s)
| | - Venus Baghalabadi
- Department of Chemistry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Subin R C K Rajendran
- Department of Chemistry, Dalhousie University, Halifax, Nova Scotia, Canada
- Verschuren Centre for Sustainability in Energy and the Environment, Sydney, Nova Scotia, Canada
| | - Philip J Jakubec
- Department of Chemistry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Hammam Said
- Department of Chemistry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Teresa S McMillen
- Department of Chemistry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Ziheng Dang
- Department of Chemistry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Alan A Doucette
- Department of Chemistry, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
19
|
Zacharias AO, Liu C, VanAernum ZL, Covey TR, Bateman KP, Wen X, McLaren DG. Ultrahigh-Throughput Intact Protein Analysis with Acoustic Ejection Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2023; 34:4-9. [PMID: 36468949 DOI: 10.1021/jasms.2c00276] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The need for high-throughput intact protein analysis has been rising as drug discovery increasingly requires the analysis of large sets of covalent modifiers and protein therapeutics. Liquid chromatography-mass spectrometry (LC-MS) is the primary analytical tool used to date to characterize proteins within the biopharmaceutical industry. However, the speed of LC-MS prevents the analysis of large-scale sample sets (>1000 within a day). Acoustic ejection mass spectrometry (AEMS) has recently been established as an electrospray ionization (ESI)-MS based platform with both fast analytical throughput and high data quality. Since its introduction, this technology has been applied in numerous fields with a primary focus on small-molecule analysis in high-throughput drug discovery and development. Here we explore the application of AEMS to high-throughput intact protein analysis for proteins ranging in molecular weight from 17 to 150 kDa on a prototype high-resolution quadrupole time-of-flight (HR QTOF) based AEMS system. Data quality obtained on this platform is comparable to LC-MS, while the analysis speed is significantly improved to one-second-per-sample. This ultrahigh-throughput intact protein analysis platform has the potential to be used broadly in drug discovery.
Collapse
Affiliation(s)
- Adway O Zacharias
- Merck & Co., Inc., 126 East Lincoln Ave. Rahway, New Jersey07065, United States
| | - Chang Liu
- SCIEX, 71 Four Valley Drive, Concord, OntarioL4K 4V8, Canada
| | - Zachary L VanAernum
- Merck & Co., Inc., 126 East Lincoln Ave. Rahway, New Jersey07065, United States
| | - Thomas R Covey
- SCIEX, 71 Four Valley Drive, Concord, OntarioL4K 4V8, Canada
| | - Kevin P Bateman
- Merck & Co., Inc., 126 East Lincoln Ave. Rahway, New Jersey07065, United States
| | - Xiujuan Wen
- Merck & Co., Inc., 126 East Lincoln Ave. Rahway, New Jersey07065, United States
| | - David G McLaren
- Merck & Co., Inc., 126 East Lincoln Ave. Rahway, New Jersey07065, United States
| |
Collapse
|
20
|
Shao X, Huang Y, Wang G. Microfluidic devices for protein analysis using intact and top‐down mass spectrometry. VIEW 2022. [DOI: 10.1002/viw.20220032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Xinyang Shao
- Institute for Cell Analysis Shenzhen Bay Laboratory Shenzhen China
- Biomedical Pioneering Innovation Center Peking University Beijing China
- Peking‐Tsinghua Center for Life Sciences Peking University Beijing China
| | - Yanyi Huang
- Institute for Cell Analysis Shenzhen Bay Laboratory Shenzhen China
- Biomedical Pioneering Innovation Center Peking University Beijing China
- Peking‐Tsinghua Center for Life Sciences Peking University Beijing China
- College of Chemistry and Molecular Engineering and Beijing National Laboratory for Molecular Sciences Peking University Beijing China
| | - Guanbo Wang
- Institute for Cell Analysis Shenzhen Bay Laboratory Shenzhen China
- Biomedical Pioneering Innovation Center Peking University Beijing China
| |
Collapse
|
21
|
McGee JP, Senko MW, Jooß K, Des Soye BJ, Compton PD, Kelleher NL, Kafader JO. Automated Control of Injection Times for Unattended Acquisition of Multiplexed Individual Ion Mass Spectra. Anal Chem 2022; 94:16543-16548. [PMID: 36416365 PMCID: PMC9969899 DOI: 10.1021/acs.analchem.2c03495] [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] [Indexed: 11/24/2022]
Abstract
Charge detection mass spectrometry (CDMS) provides mass domain spectra of large and highly heterogeneous analytes. Over the past few years, we have multiplexed CDMS on Orbitrap instruments, an approach termed Individual Ion Mass Spectrometry (I2MS). Until now, I2MS required manual adjustment of injection times to collect spectra in the individual ion regime. To increase sample adaptability, enable online separations, and reduce the barrier for entry, we report an automated method for adjusting ion injection times in I2MS for image current detectors like the Orbitrap. Automatic Ion Control (AIC) utilizes the density of signals in the m/z domain to adjust an ensemble of ions down to the individual ion regime in real-time. The AIC technique was applied to both denatured and native proteins yielding high quality data without human intervention directly in the mass domain.
Collapse
Affiliation(s)
- John P McGee
- Departments of Chemistry and Molecular Biosciences, Department of Chemical and Biological Engineering, the Chemistry of Life Processes Institute, the Proteomics Center of Excellence at Northwestern University, Evanston, Illinois60208, United States
| | - Michael W Senko
- Thermo Fisher Scientific, San Jose, California95134, United States
| | - Kevin Jooß
- Departments of Chemistry and Molecular Biosciences, Department of Chemical and Biological Engineering, the Chemistry of Life Processes Institute, the Proteomics Center of Excellence at Northwestern University, Evanston, Illinois60208, United States
| | - Benjamin J Des Soye
- Departments of Chemistry and Molecular Biosciences, Department of Chemical and Biological Engineering, the Chemistry of Life Processes Institute, the Proteomics Center of Excellence at Northwestern University, Evanston, Illinois60208, United States
| | - Philip D Compton
- Integrated Protein Technologies, Inc., Evanston, Illinois60201, United States
| | - Neil L Kelleher
- Departments of Chemistry and Molecular Biosciences, Department of Chemical and Biological Engineering, the Chemistry of Life Processes Institute, the Proteomics Center of Excellence at Northwestern University, Evanston, Illinois60208, United States
| | - Jared O Kafader
- Departments of Chemistry and Molecular Biosciences, Department of Chemical and Biological Engineering, the Chemistry of Life Processes Institute, the Proteomics Center of Excellence at Northwestern University, Evanston, Illinois60208, United States
| |
Collapse
|
22
|
Seeing the complete picture: proteins in top-down mass spectrometry. Essays Biochem 2022; 67:283-300. [PMID: 36468679 DOI: 10.1042/ebc20220098] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022]
Abstract
Abstract
Top-down protein mass spectrometry can provide unique insights into protein sequence and structure, including precise proteoform identification and study of protein–ligand and protein–protein interactions. In contrast with the commonly applied bottom-up approach, top-down approaches do not include digestion of the protein of interest into small peptides, but instead rely on the ionization and subsequent fragmentation of intact proteins. As such, it is fundamentally the only way to fully characterize the composition of a proteoform. Here, we provide an overview of how a top-down protein mass spectrometry experiment is performed and point out recent applications from the literature to the reader. While some parts of the top-down workflow are broadly applicable, different research questions are best addressed with specific experimental designs. The most important divide is between studies that prioritize sequence information (i.e., proteoform identification) versus structural information (e.g., conformational studies, or mapping protein–protein or protein–ligand interactions). Another important consideration is whether to work under native or denaturing solution conditions, and the overall complexity of the sample also needs to be taken into account, as it determines whether (chromatographic) separation is required prior to MS analysis. In this review, we aim to provide enough information to support both newcomers and more experienced readers in the decision process of how to answer a potential research question most efficiently and to provide an overview of the methods that exist to answer these questions.
Collapse
|
23
|
Forte E, Des Soye BJ, Melani RD, Hollas MAR, Kafader JO, Sha BE, Schneider JR, Kelleher NL. Divergent Antibody Repertoires Found for Omicron versus Wuhan SARS-CoV-2 Strains Using Ig-MS. J Proteome Res 2022; 21:2987-2997. [PMID: 36343328 PMCID: PMC9668041 DOI: 10.1021/acs.jproteome.2c00514] [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: 08/24/2022] [Indexed: 11/09/2022]
Abstract
SARS-CoV-2 Omicron (B.1.1.529) and its subvariants are currently the most common variants of concern worldwide, featuring numerous mutations in the spike protein and elsewhere that collectively make Omicron variants more transmissible and more resistant to antibody-mediated neutralization provided by vaccination, previous infections, and monoclonal antibody therapies than their predecessors. We recently reported the creation and characterization of Ig-MS, a new mass spectrometry-based serology platform that can define the repertoire of antibodies against an antigen of interest at single proteoform resolution. Here, we applied Ig-MS to investigate the evolution of plasma antibody repertoires against the receptor-binding domain (RBD) of SARS-CoV-2 in response to the booster shot and natural viral infection. We also assessed the capacity for antibody repertoires generated in response to vaccination and/or infection with the Omicron variant to bind to both Wuhan- and Omicron-RBDs. Our results show that (1) the booster increases antibody titers against both Wuhan- and Omicron- RBDs and elicits an Omicron-specific response and (2) vaccination and infection act synergistically in generating anti-RBD antibody repertoires able to bind both Wuhan- and Omicron-RBDs with variant-specific antibodies.
Collapse
Affiliation(s)
- Eleonora Forte
- Proteomics Center of Excellence, Evanston, IL, 60208, USA
- Department of Surgery, Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | | | - Rafael D. Melani
- Proteomics Center of Excellence, Evanston, IL, 60208, USA
- Department of Molecular Biosciences, Chemistry, Northwestern University, Evanston, IL, 60208, USA
| | | | | | - Beverly E. Sha
- Division of Infectious Diseases, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Jeffrey R. Schneider
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Neil L. Kelleher
- Proteomics Center of Excellence, Evanston, IL, 60208, USA
- Department of Surgery, Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| |
Collapse
|
24
|
Hecht ES, Obiorah EC, Liu X, Morrison L, Shion H, Lauber M. Microflow size exclusion chromatography to preserve micromolar affinity complexes and achieve subunit separations for native state mass spectrometry. J Chromatogr A 2022; 1685:463638. [DOI: 10.1016/j.chroma.2022.463638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022]
|
25
|
Pu F, Ugrin SA, Radosevich AJ, Chang-Yen D, Sawicki JW, Talaty NN, Elsen NL, Williams JD. High-Throughput Intact Protein Analysis for Drug Discovery Using Infrared Matrix-Assisted Laser Desorption Electrospray Ionization Mass Spectrometry. Anal Chem 2022; 94:13566-13574. [PMID: 36129783 DOI: 10.1021/acs.analchem.2c03211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Mass spectrometry (MS) is the primary analytical tool used to characterize proteins within the biopharmaceutical industry. Electrospray ionization (ESI) coupled to liquid chromatography (LC) is the current gold standard for intact protein analysis. However, inherent speed limitations of LC/MS prevent analysis of large sample numbers (>1000) in a day. Infrared matrix-assisted laser desorption electrospray ionization (IR-MALDESI-MS), an ambient ionization MS technology, has recently been established as a platform for high-throughput small molecule analysis. Here, we report the applications of such a system for the analysis of intact proteins commonly performed within the drug discovery process. A wide molecular weight range of proteins 10-150 kDa was detected on the system with improved tolerance to salts and buffers compared to ESI. With high concentrations and model proteins, a sample rate of up to 22 Hz was obtained. For proteins at low concentrations and in buffers used in commonly employed assays, robust data at a sample rate of 1.5 Hz were achieved, which is ∼22× faster than current technologies used for high-throughput ESI-MS-based protein assays. In addition, two multiplexed plate-based high-throughput sample cleanup methods were coupled to IR-MALDESI-MS to enable analysis of samples containing excessive amounts of salts and buffers without fully compromising productivity. Example experiments, which leverage the speed of the IR-MALDESI-MS system to monitor NISTmAb reduction, protein autophosphorylation, and compound binding kinetics in near real time, are demonstrated.
Collapse
Affiliation(s)
- Fan Pu
- AbbVie Inc, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Scott A Ugrin
- AbbVie Inc, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Andrew J Radosevich
- AbbVie Inc, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - David Chang-Yen
- AbbVie Inc, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - James W Sawicki
- AbbVie Inc, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Nari N Talaty
- AbbVie Inc, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Nathaniel L Elsen
- AbbVie Inc, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Jon D Williams
- AbbVie Inc, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| |
Collapse
|
26
|
Liu R, Xia S, Li H. Native top-down mass spectrometry for higher-order structural characterization of proteins and complexes. MASS SPECTROMETRY REVIEWS 2022:e21793. [PMID: 35757976 DOI: 10.1002/mas.21793] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 06/15/2023]
Abstract
Progress in structural biology research has led to a high demand for powerful and yet complementary analytical tools for structural characterization of proteins and protein complexes. This demand has significantly increased interest in native mass spectrometry (nMS), particularly native top-down mass spectrometry (nTDMS) in the past decade. This review highlights recent advances in nTDMS for structural research of biological assemblies, with a particular focus on the extra multi-layers of information enabled by TDMS. We include a short introduction of sample preparation and ionization to nMS, tandem fragmentation techniques as well as mass analyzers and software/analysis pipelines used for nTDMS. We highlight unique structural information offered by nTDMS and examples of its broad range of applications in proteins, protein-ligand interactions (metal, cofactor/drug, DNA/RNA, and protein), therapeutic antibodies and antigen-antibody complexes, membrane proteins, macromolecular machineries (ribosome, nucleosome, proteosome, and viruses), to endogenous protein complexes. The challenges, potential, along with perspectives of nTDMS methods for the analysis of proteins and protein assemblies in recombinant and biological samples are discussed.
Collapse
Affiliation(s)
- Ruijie Liu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Shujun Xia
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Huilin Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
27
|
Melani RD, Des Soye BJ, Kafader JO, Forte E, Hollas M, Blagojevic V, Negrão F, McGee JP, Drown B, Lloyd-Jones C, Seckler HS, Camarillo JM, Compton PD, LeDuc RD, Early B, Fellers RT, Cho BK, Mattamana BB, Goo YA, Thomas PM, Ash MK, Bhimalli PP, Al-Harthi L, Sha BE, Schneider JR, Kelleher NL. Next-Generation Serology by Mass Spectrometry: Readout of the SARS-CoV-2 Antibody Repertoire. J Proteome Res 2022; 21:274-288. [PMID: 34878788 PMCID: PMC8673472 DOI: 10.1021/acs.jproteome.1c00882] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Indexed: 01/03/2023]
Abstract
Methods of antibody detection are used to assess exposure or immunity to a pathogen. Here, we present Ig-MS, a novel serological readout that captures the immunoglobulin (Ig) repertoire at molecular resolution, including entire variable regions in Ig light and heavy chains. Ig-MS uses recent advances in protein mass spectrometry (MS) for multiparametric readout of antibodies, with new metrics like Ion Titer (IT) and Degree of Clonality (DoC) capturing the heterogeneity and relative abundance of individual clones without sequencing of B cells. We applied Ig-MS to plasma from subjects with severe and mild COVID-19 and immunized subjects after two vaccine doses, using the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2 as the bait for antibody capture. Importantly, we report a new data type for human serology, that could use other antigens of interest to gauge immune responses to vaccination, pathogens, or autoimmune disorders.
Collapse
Affiliation(s)
- Rafael D. Melani
- Departments of Molecular Biosciences, Chemistry, Northwestern University, Evanston, IL, 60208, USA
| | - Benjamin J. Des Soye
- Departments of Molecular Biosciences, Chemistry, Northwestern University, Evanston, IL, 60208, USA
- Proteomics Center of Excellence, Evanston, IL, 60208, USA
| | - Jared O. Kafader
- Departments of Molecular Biosciences, Chemistry, Northwestern University, Evanston, IL, 60208, USA
| | - Eleonora Forte
- Proteomics Center of Excellence, Evanston, IL, 60208, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Michael Hollas
- Departments of Molecular Biosciences, Chemistry, Northwestern University, Evanston, IL, 60208, USA
| | - Voislav Blagojevic
- Departments of Molecular Biosciences, Chemistry, Northwestern University, Evanston, IL, 60208, USA
| | - Fernanda Negrão
- Departments of Molecular Biosciences, Chemistry, Northwestern University, Evanston, IL, 60208, USA
| | - John P. McGee
- Departments of Molecular Biosciences, Chemistry, Northwestern University, Evanston, IL, 60208, USA
| | - Bryon Drown
- Departments of Molecular Biosciences, Chemistry, Northwestern University, Evanston, IL, 60208, USA
| | - Cameron Lloyd-Jones
- Departments of Molecular Biosciences, Chemistry, Northwestern University, Evanston, IL, 60208, USA
| | - Henrique S. Seckler
- Departments of Molecular Biosciences, Chemistry, Northwestern University, Evanston, IL, 60208, USA
| | - Jeannie M. Camarillo
- Departments of Molecular Biosciences, Chemistry, Northwestern University, Evanston, IL, 60208, USA
| | - Philip D. Compton
- Departments of Molecular Biosciences, Chemistry, Northwestern University, Evanston, IL, 60208, USA
- Integrated Protein Technologies, Evanston, IL, 60201, USA
| | - Richard D. LeDuc
- Departments of Molecular Biosciences, Chemistry, Northwestern University, Evanston, IL, 60208, USA
| | - Bryan Early
- Departments of Molecular Biosciences, Chemistry, Northwestern University, Evanston, IL, 60208, USA
| | - Ryan T. Fellers
- Departments of Molecular Biosciences, Chemistry, Northwestern University, Evanston, IL, 60208, USA
| | - Byoung-Kyu Cho
- Proteomics Center of Excellence, Evanston, IL, 60208, USA
| | | | - Young Ah Goo
- Departments of Molecular Biosciences, Chemistry, Northwestern University, Evanston, IL, 60208, USA
- Proteomics Center of Excellence, Evanston, IL, 60208, USA
| | - Paul M. Thomas
- Departments of Molecular Biosciences, Chemistry, Northwestern University, Evanston, IL, 60208, USA
- Proteomics Center of Excellence, Evanston, IL, 60208, USA
| | - Michelle K. Ash
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Pavan P. Bhimalli
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Lena Al-Harthi
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Beverly E. Sha
- Division of Infectious Diseases, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Jeffrey R. Schneider
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Neil L. Kelleher
- Departments of Molecular Biosciences, Chemistry, Northwestern University, Evanston, IL, 60208, USA
- Proteomics Center of Excellence, Evanston, IL, 60208, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611 USA
| |
Collapse
|
28
|
Bennett JL, Nguyen GTH, Donald WA. Protein-Small Molecule Interactions in Native Mass Spectrometry. Chem Rev 2021; 122:7327-7385. [PMID: 34449207 DOI: 10.1021/acs.chemrev.1c00293] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Small molecule drug discovery has been propelled by the continual development of novel scientific methodologies to occasion therapeutic advances. Although established biophysical methods can be used to obtain information regarding the molecular mechanisms underlying drug action, these approaches are often inefficient, low throughput, and ineffective in the analysis of heterogeneous systems including dynamic oligomeric assemblies and proteins that have undergone extensive post-translational modification. Native mass spectrometry can be used to probe protein-small molecule interactions with unprecedented speed and sensitivity, providing unique insights into polydisperse biomolecular systems that are commonly encountered during the drug discovery process. In this review, we describe potential and proven applications of native MS in the study of interactions between small, drug-like molecules and proteins, including large multiprotein complexes and membrane proteins. Approaches to quantify the thermodynamic and kinetic properties of ligand binding are discussed, alongside a summary of gas-phase ion activation techniques that have been used to interrogate the structure of protein-small molecule complexes. We additionally highlight some of the key areas in modern drug design for which native mass spectrometry has elicited significant advances. Future developments and applications of native mass spectrometry in drug discovery workflows are identified, including potential pathways toward studying protein-small molecule interactions on a whole-proteome scale.
Collapse
Affiliation(s)
- Jack L Bennett
- School of Chemistry, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Giang T H Nguyen
- School of Chemistry, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - William A Donald
- School of Chemistry, University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
29
|
Santos Seckler HD, Park HM, Lloyd-Jones CM, Melani RD, Camarillo JM, Wilkins JT, Compton PD, Kelleher NL. New Interface for Faster Proteoform Analysis: Immunoprecipitation Coupled with SampleStream-Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2021; 32:1659-1670. [PMID: 34043341 PMCID: PMC8530194 DOI: 10.1021/jasms.1c00026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Different proteoform products of the same gene can exhibit differing associations with health and disease, and their patterns of modifications may offer more precise markers of phenotypic differences between individuals. However, currently employed protein-biomarker discovery and quantification tools, such as bottom-up proteomics and ELISAs, are mostly proteoform-unaware. Moreover, the current throughput for proteoform-level analyses by liquid chromatography mass spectrometry (LCMS) for quantitative top-down proteomics is incompatible with population-level biomarker surveys requiring robust, faster proteoform analysis. To this end, we developed immunoprecipitation coupled to SampleStream mass spectrometry (IP-SampleStream-MS) as a high-throughput, automated technique for the targeted quantification of proteoforms. We applied IP-SampleStream-MS to serum samples of 25 individuals to assess the proteoform abundances of apolipoproteins A-I (ApoA-I) and C-III (ApoC-III). The results for ApoA-I were compared to those of LCMS for these individuals, with IP-SampleStream-MS showing a >7-fold higher throughput with >50% better analytical variation. Proteoform abundances measured by IP-SampleStream-MS correlated strongly to LCMS-based values (R2 = 0.6-0.9) and produced convergent proteoform-to-phenotype associations, namely, the abundance of canonical ApoA-I was associated with lower HDL-C (R = 0.5) and glycated ApoA-I with higher fasting glucose (R = 0.6). We also observed proteoform-to-phenotype associations for ApoC-III, 22 glycoproteoforms of which were characterized in this study. The abundance of ApoC-III modified by a single N-acetyl hexosamine (HexNAc) was associated with indices of obesity, such as BMI, weight, and waist circumference (R ∼ 0.7). These data show IP-SampleStream-MS to be a robust, scalable workflow for high-throughput associations of proteoforms to phenotypes.
Collapse
Affiliation(s)
- Henrique Dos Santos Seckler
- Department of Chemistry, Chemistry of Life Processes Institute and Proteomics Center of Excellence, Northwestern University, Evanston, Illinois 60208, United States
| | - Hae-Min Park
- Department of Chemical Engineering and Applied Chemistry, Chungnam National University, Daejeon 34134, South Korea
| | - Cameron M Lloyd-Jones
- Department of Chemistry, Chemistry of Life Processes Institute and Proteomics Center of Excellence, Northwestern University, Evanston, Illinois 60208, United States
| | - Rafael D Melani
- Department of Chemistry, Chemistry of Life Processes Institute and Proteomics Center of Excellence, Northwestern University, Evanston, Illinois 60208, United States
| | - Jeannie M Camarillo
- Department of Chemistry, Chemistry of Life Processes Institute and Proteomics Center of Excellence, Northwestern University, Evanston, Illinois 60208, United States
| | - John T Wilkins
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Philip D Compton
- Department of Chemistry, Chemistry of Life Processes Institute and Proteomics Center of Excellence, Northwestern University, Evanston, Illinois 60208, United States
- Integrated Protein Technologies, Inc., Evanston, Illinois 60646, United States
| | - Neil L Kelleher
- Department of Chemistry, Chemistry of Life Processes Institute and Proteomics Center of Excellence, Northwestern University, Evanston, Illinois 60208, United States
| |
Collapse
|
30
|
Melani RD, Soye BJD, Kafader JO, Forte E, Hollas M, Blagojevic V, Negrão F, McGee JP, Drown B, Lloyd-Jones C, Seckler HS, Camarillo JM, Compton PD, LeDuc RD, Early B, Fellers RT, Cho BK, Mattamana BB, Goo YA, Thomas PM, Ash MK, Bhimalli PP, Al-Harthi L, Sha BE, Schneider JR, Kelleher NL. Next-generation Serology by Mass Spectrometry: Readout of the SARS-CoV-2 Antibody Repertoire. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021. [PMID: 34268518 DOI: 10.1101/2021.07.06.21259226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Methods of antibody detection are used to assess exposure or immunity to a pathogen. Here, we present Ig-MS , a novel serological readout that captures the immunoglobulin (Ig) repertoire at molecular resolution, including entire variable regions in Ig light and heavy chains. Ig-MS uses recent advances in protein mass spectrometry (MS) for multi-parametric readout of antibodies, with new metrics like Ion Titer (IT) and Degree of Clonality (DoC) capturing the heterogeneity and relative abundance of individual clones without sequencing of B cells. We apply Ig-MS to plasma from subjects with severe & mild COVID-19, using the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2 as the bait for antibody capture. Importantly, we report a new data type for human serology, with compatibility to any recombinant antigen to gauge our immune responses to vaccination, pathogens, or autoimmune disorders.
Collapse
|
31
|
Vervoort N, Goossens K, Baeten M, Chen Q. Recent advances in analytical techniques for high throughput experimentation. ANALYTICAL SCIENCE ADVANCES 2021; 2:109-127. [PMID: 38716456 PMCID: PMC10989611 DOI: 10.1002/ansa.202000155] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 11/17/2024]
Abstract
High throughput experimentation is a growing and evolving field that allows to execute dozens to several thousands of experiments per day with relatively limited resources. Through miniaturization, typically a high degree of automation and the use of digital data tools, many parallel reactions or experiments at a time can be run in such workflows. High throughput experimentation also requires fast analytical techniques capable of generating critically important analytical data in line with the increased rate of experimentation. As traditional techniques usually do not deliver the speed required, some unique approaches are required to enable workflows to function as designed. This review covers the recent developments (2019-2020) in this field and was intended to give a comprehensive overview of the current "state-of-the-art."
Collapse
Affiliation(s)
- Nico Vervoort
- Chemical Process R&DProcess Analytical ResearchJanssen R&DBeerseBelgium
| | - Karel Goossens
- Chemical Process R&DProcess Analytical ResearchJanssen R&DBeerseBelgium
| | - Mattijs Baeten
- Chemical Process R&DProcess Analytical ResearchJanssen R&DBeerseBelgium
| | - Qinghao Chen
- Chemical Process R&DHigh Throughput ExperimentationJanssen R&DBeerseBelgium
| |
Collapse
|
32
|
High-throughput antibody screening from complex matrices using intact protein electrospray mass spectrometry. Proc Natl Acad Sci U S A 2020; 117:9851-9856. [PMID: 32327606 PMCID: PMC7211930 DOI: 10.1073/pnas.1917383117] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
While electrospray ionization–mass spectrometry (ESI-MS) provides higher resolution for larger proteins, the conventional liquid chromatography (LC)-MS method suffers from low throughput. Our described RapidFire-MS workflow demonstrated unprecedented screening throughput as fast as 15 s/sample, a 10-fold improvement over conventional LC-MS approaches. The screening enabled selection of clones with the highest purity of bispecific antibody production with intact masses as accurate as 7 ppm with baseline resolution at the glycoform level in samples as complex as plasma sample. The utility of the method can be expanded to many other applications that can exploit the advantages of high-throughput intact protein MS analyses including but not limited to pharmacokinetic analyses, enzymatic screening, biotransformation characterization, and quality control screening. Toward the goal of increasing the throughput of high-resolution mass characterization of intact antibodies, we developed a RapidFire–mass spectrometry (MS) assay using electrospray ionization. We achieved unprecedented screening throughput as fast as 15 s/sample, which is an order of magnitude improvement over conventional liquid chromatography (LC)-MS approaches. The screening enabled intact mass determination as accurate as 7 ppm with baseline resolution at the glycoform level for intact antibodies. We utilized this assay to characterize and perform relative quantitation of antibody species from 248 samples of 62 different cell line clones at four time points in 2 h using RapidFire–time-of-flight MS screening. The screening enabled selection of clones with the highest purity of bispecific antibody production and the results significantly correlated with conventional LC-MS results. In addition, analyzing antibodies from a complex plasma sample using affinity-RapidFire-MS was also demonstrated and qualified. In summary, the platform affords high-throughput analyses of antibodies, including bispecific antibodies and potential mispaired side products, in cell culture media, or other complex matrices.
Collapse
|