1
|
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
|
2
|
Zhang Z, Khanal N, Dykstra AB, Daris K. Stop-Codon Readthrough in Therapeutic Protein Candidates Expressed from Mammalian Cells. J Pharm Sci 2024; 113:1498-1505. [PMID: 38342339 DOI: 10.1016/j.xphs.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/03/2024] [Accepted: 02/04/2024] [Indexed: 02/13/2024]
Abstract
Stop codon readthroughs were examined in 48 recombinant therapeutic protein candidates produced from multiple clones of Chinese hamster ovary cells, using peptide mapping with LC-MS/MS detection. We found that stop codon readthrough is a common phenomenon occurring in most of these candidates, with levels varying from below the detection limit of ∼0.001 % to ∼1 %. The readthrough propensity depends on the stop codon being used, as well as the nucleotides surrounding it. The amino acids misincorporated into the stop position can be well-predicted by a third-base wobble mismatch and a first-base U/G mismatch during codon recognition, i.e., tyrosine or glutamine insertion for the UAA and UAG stop codons, and tryptophan, cysteine or arginine insertion for the UGA stop codon. Data shown in this report demonstrate the importance of optimizing the DNA sequence near the stop codon, and the importance of detecting stop codon readthroughs during the development of a therapeutic product.
Collapse
Affiliation(s)
- Zhongqi Zhang
- Process Development, Amgen Inc. Thousand Oaks, CA 91320, USA.
| | - Neelam Khanal
- Process Development, Amgen Inc. Thousand Oaks, CA 91320, USA
| | | | - Kristi Daris
- Process Development, Amgen Inc. Thousand Oaks, CA 91320, USA
| |
Collapse
|
3
|
Ruppen I, Verscheure L, Vandenheede I, Ortiz A, de Melo IS, Liebig T, Sandra P, Beydon ME, Sandra K. Characterization of mAb size heterogeneity originating from a cysteine to tyrosine substitution using denaturing and native LC-MS. J Pharm Biomed Anal 2023; 236:115743. [PMID: 37757547 DOI: 10.1016/j.jpba.2023.115743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023]
Abstract
Upon assessing the comparability between a biosimilar mAb and its reference product by non-reducing CE-SDS, increased levels of a heavy-heavy-light chain (HHL) variant, present as a low molecular weight (LMW) peak, were observed. RPLC-MS applied at top, middle-up and bottom-up level revealed the existence of Cys-to-Tyr substitutions, predominantly at position HC226 involved in connecting LC and HC, explaining the abundant HHL levels. Antigen binding was not impacted by the presence of this size variant suggesting a non-covalent association of Tyr substituted HHL and LC. The latter complex is not maintained in the denaturing conditions associated with CE-SDS and RPLC-MS. Its existence could, nevertheless, be confirmed by native SEC-MS which preserves non-covalent protein interactions during separation and electrospray ionization. Amino acid analysis furthermore demonstrated a depletion of Cys during the fed-batch process indicating that the observed size/sequence variant is not of genetic but rather of metabolic origin. Native SEC-MS showed that supplementing the cell culture medium with Cys halts misincorporation of Tyr and promotes the formation of the desired mAb structure. To the best of our knowledge, Cys-to-Tyr substitutions preventing interchain disulfide bridge formation have not been described earlier. This observation adds to the impressive structural heterogeneity reported to date for mAbs.
Collapse
Affiliation(s)
- Isabel Ruppen
- mAbxience Research, Manuel Pombo Angulo 28, 28050 Madrid, Spain
| | | | | | - Alexia Ortiz
- RIC group, President Kennedypark 26, 8500 Kortrijk, Belgium
| | | | - Timo Liebig
- mAbxience Research, Manuel Pombo Angulo 28, 28050 Madrid, Spain
| | - Pat Sandra
- RIC group, President Kennedypark 26, 8500 Kortrijk, Belgium
| | | | - Koen Sandra
- RIC group, President Kennedypark 26, 8500 Kortrijk, Belgium.
| |
Collapse
|
4
|
Millán-Martín S, Jakes C, Carillo S, Bones J. Multi-Attribute Method (MAM) Analytical Workflow for Biotherapeutic Protein Characterization from Process Development to QC. Curr Protoc 2023; 3:e927. [PMID: 37929772 DOI: 10.1002/cpz1.927] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
The multi-attribute method (MAM) has emerged significantly in recent years to support biotherapeutic protein characterization from process development to the QC environment. MAM is a liquid chromatography mass spectrometry (LC-MS) based peptide mapping approach, which combines the benefits from liquid chromatography coupled to high resolution accurate mass mass spectrometry (LC-HRAM MS), enabling direct assessment of protein sequence and product quality attributes with site specificity. These product quality attributes may impact efficacy, safety, stability, and process robustness. MAM is intended to replace conventional analytical approaches as it offers a more streamlined strategy for parallel monitoring of multiple attributes in a single analysis with high sensitivity and confidence, and ultimately supports more robust Quality by Design (QbD) approaches and faster decision cycles for biotherapeutic development. MAM consists of three main stages. The first stage is sample digestion, which typically entails proteolytic digestion of the protein. The second stage is reversed-phase chromatographic separation of the generated peptides and detection by HRAM MS in two phases. During MAM Phase I (discovery phase), data-dependent acquisition (DDA) MS/MS is performed to enable confident identification of peaks and development of a peptide workbook. During MAM Phase II (monitoring phase), full MS acquisition is only carried out for the monitoring of predefined product quality attributes (PQAs). The third stage is data processing, which entails analysis and reporting for each of the two phases including evaluation of sequence coverage, assessment of PQAs and peptide workbook creation during phase I, and targeted monitoring of predefined product attributes and new peak detection (NPD) during phase II. The latter is a comparative analysis that uses a base peak alignment algorithm to determine any non-monitored differences between the LC-MS chromatograms of a test sample and a reference standard. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: In-solution sample digestion Alternate Protocol: Automated sample digestion Basic Protocol 2: Reversed-phase chromatographic separation and detection by HRAM-MS (RPLC-HRAM MS) Basic Protocol 3: Data processing and reporting.
Collapse
Affiliation(s)
| | - Craig Jakes
- National Institute for Bioprocessing Research and Training, Dublin, Ireland
| | - Sara Carillo
- National Institute for Bioprocessing Research and Training, Dublin, Ireland
| | - Jonathan Bones
- National Institute for Bioprocessing Research and Training, Dublin, Ireland
- School of Chemical and Bioprocess Engineering, University College Dublin, Dublin, Ireland
| |
Collapse
|
5
|
Millán-Martín S, Jakes C, Carillo S, Bones J. Multi-attribute method (MAM) to assess analytical comparability of adalimumab biosimilars. J Pharm Biomed Anal 2023; 234:115543. [PMID: 37385093 DOI: 10.1016/j.jpba.2023.115543] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/12/2023] [Accepted: 06/20/2023] [Indexed: 07/01/2023]
Abstract
Adalimumab drug product (Humira ®), the first fully human monoclonal antibody (mAb) approved by FDA in 2002, led the top ten list of best-selling mAbs in 2018 and has been the most profitable drug in the world. With the expiration of patent protection in Europe in 2018 and in United States by 2023, the landscape is changing as up to 10 adalimumab biosimilars are expected to enter the market in the US. Biosimilars offer the potential to lower costs on health care systems and increase patient accessibility. The analytical similarity of seven different adalimumab biosimilars was accomplished in the present study using the multi-attribute method (MAM), a LC-MS based peptide mapping technique that allows for primary sequence assessment and evaluation of multiple quality attributes including deamidation, oxidation, succinimide formation, N- and C- terminal composition and detailed N-glycosylation analysis. In the first step, characterization of the most relevant post-translational modifications of a reference product was attained during the discovery phase of MAM. During the second step, as part of the MAM targeted monitoring phase, adalimumab batch-to batch variability was evaluated to define statistical intervals for the establishment of similarity ranges. The third step describes biosimilarity evaluation of predefined quality attributes and new peak detection for the assessment of any new or modified peak compared to the reference product. This study highlights a new perspective of the MAM approach and its underlying power for biotherapeutic comparability exercises in addition to analytical characterization. MAM offers a streamlined comparability assessment workflow based on high-confidence quality attribute analysis using high-resolution accurate mass mass spectrometry (HRAM MS) and the capability to detect any new or modified peak compared to the reference product.
Collapse
Affiliation(s)
- Silvia Millán-Martín
- National Institute for Bioprocessing Research & Training, Fosters Avenue, Mount Merrion, Blackrock, A94 X099 Dublin, Ireland
| | - Craig Jakes
- National Institute for Bioprocessing Research & Training, Fosters Avenue, Mount Merrion, Blackrock, A94 X099 Dublin, Ireland
| | - Sara Carillo
- National Institute for Bioprocessing Research & Training, Fosters Avenue, Mount Merrion, Blackrock, A94 X099 Dublin, Ireland
| | - Jonathan Bones
- National Institute for Bioprocessing Research & Training, Fosters Avenue, Mount Merrion, Blackrock, A94 X099 Dublin, Ireland; School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Dublin 4 D04 V1W8, Ireland.
| |
Collapse
|
6
|
Niu B, Lu Y, Chen X, Xu W. Using New Peak Detection to Solve Sequence Variants Analysis Challenges in Bioprocess Development. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2023; 34:401-408. [PMID: 36705517 DOI: 10.1021/jasms.2c00292] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Recombinant therapeutic proteins have become the major class of drugs to treat various human diseases in recent years. Low levels of protein sequence variants (SVs) have been reported to be present in recombinant therapeutic proteins. The consequences of potential unwanted immune response from SVs of recombinant therapeutic proteins have increasingly drawn attention from regulatory authorities and the biopharmaceutical industry. It is highly desirable to detect low-level SVs during clone selection and early process development as part of the control strategy. Peptide mapping with LC-MS/MS analysis has been applied as a powerful tool to characterize post-translation modifications of therapeutic proteins. Despite the recent advancements in mass spectrometry hardware and software, it is still quite challenging and time-consuming to detect and identify low-level SVs. In this study, we present an optimized approach using new peak detection to detect and identify low level SVs with high confidence and high speed. The new approach makes sequence variants analysis by LC-MS/MS broadly applicable and practical in bioprocess development of therapeutic proteins.
Collapse
Affiliation(s)
- Ben Niu
- Analytical Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland20878United States
| | - Yali Lu
- Analytical Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland20878United States
| | - Xiaoyu Chen
- Analytical Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland20878United States
| | - Wei Xu
- Analytical Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland20878United States
| |
Collapse
|
7
|
Monoclonal Antibody Sequence Variants Disguised as Fragments: Identification, Characterization, and Their Removal by Purification Process Optimization. J Pharm Sci 2022; 111:3009-3016. [PMID: 35940243 DOI: 10.1016/j.xphs.2022.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/02/2022] [Accepted: 08/02/2022] [Indexed: 12/14/2022]
Abstract
During early stage development of a therapeutic IgG1 monoclonal antibody, high levels of low molecular weight (LMW) peaks were observed by high performance size-exclusion chromatography and capillary electrophoresis. Further characterization of the LMW peak enriched HPSEC fractions using reversed phase liquid chromatography coupled to mass spectrometry showed these LMW species were 47 kDa and 50 kDa in size. However, the measured masses could not be matched to any fragments resulting from peptide bond hydrolysis. To identify these unknown LMW species, molecular characterization methods were employed, including high-throughput sequencing of RNA. Transcriptomic analysis revealed the LMW species were generated by mis-splicing events in the heavy chain transcript, which produced truncated heavy chain products that assembled with the light chain to mimic the appearance of fragments identified by routine purity assays. In an effort to improve product quality, an optimized purification process was developed. Characterization of the process intermediates confirmed removal of both LMW species by the optimized process. Our study demonstrates that deep-dive analytical characterization of biotherapeutics is critical to ensure product quality and inform process development. Transcriptomic analysis tools can help identify the cause of unknown species, and plays a key role in product and process characterization.
Collapse
|
8
|
Lin J, Xie M, Liu D, Gao Z, Zhao X, Ma H, Ding S, Li SM, Li S, Liu Y, Zhou F, Hu H, Chen T, Chen H, Xie M, Yang B, Cheng J, Ma M, Nan Y, Ju D. Characterization of light chain c-terminal extension sequence variant in one bispecific antibody. Front Chem 2022; 10:994472. [PMID: 36204149 PMCID: PMC9530627 DOI: 10.3389/fchem.2022.994472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
Protein modifications such as post-translational modifications (PTMs) and sequence variants (SVs) occur frequently during protein biosynthesis and have received great attention by biopharma industry and regulatory agencies. In this study, an aberrant peak near light chain (LC) was observed in the non-reduced capillary electrophoresis sodium dodecyl sulfate (nrCE-SDS) electrophoretogram during cell line development of one bispecific antibody (BsAb) product, and the detected mass was about 944 Da higher than LC. The corresponding peak was then enriched by denaturing size-exclusion chromatography (SEC-HPLC) and further characterized by nrCE-SDS and peptide mapping analyses. De novo mass spectra/mass spectra (MS/MS) analysis revealed that the aberrant peak was LC related sequence variant, with the truncated C-terminal sequence “SFNR” (“GEC”deleted) linked with downstream SV40 promotor sequence “EAEAASASELFQ”. The unusual sequence was further confirmed by comparing with the direct synthetic peptide “SFNREAEAASASELFQ”. It was demonstrated by mRNA sequencing of the cell pool that the sequence variant was caused by aberrant splicing at the transcription step. The prepared product containing this extension variant maintained well-folded structure and good functional properties though the LC/Heavy chain (HC) inter-chain disulfide was not formed. Several control strategies to mitigate the risk of this LC related sequence variant were also proposed.
Collapse
Affiliation(s)
- Jun Lin
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai, China
- Genor Biopharma Co., Ltd., Shanghai, China
| | - Mengyu Xie
- Genor Biopharma Co., Ltd., Shanghai, China
| | - Dan Liu
- Genor Biopharma Co., Ltd., Shanghai, China
| | - Zhen Gao
- Genor Biopharma Co., Ltd., Shanghai, China
| | | | - Hongxia Ma
- Genor Biopharma Co., Ltd., Shanghai, China
| | - Sheng Ding
- Genor Biopharma Co., Ltd., Shanghai, China
| | - Shu mei Li
- Genor Biopharma Co., Ltd., Shanghai, China
| | - Song Li
- Genor Biopharma Co., Ltd., Shanghai, China
| | | | - Fang Zhou
- Genor Biopharma Co., Ltd., Shanghai, China
| | - Hao Hu
- Genor Biopharma Co., Ltd., Shanghai, China
| | - Tao Chen
- Genor Biopharma Co., Ltd., Shanghai, China
| | - He Chen
- Genor Biopharma Co., Ltd., Shanghai, China
| | - Min Xie
- Genor Biopharma Co., Ltd., Shanghai, China
| | - Bo Yang
- Genor Biopharma Co., Ltd., Shanghai, China
| | - Jun Cheng
- Genor Biopharma Co., Ltd., Shanghai, China
| | - Mingjun Ma
- Genor Biopharma Co., Ltd., Shanghai, China
| | - Yanyang Nan
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai, China
| | - Dianwen Ju
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai, China
- *Correspondence: Dianwen Ju,
| |
Collapse
|
9
|
Zhang Z, Shah B. Limited Proteolysis Coupled with Mass Spectrometry for Simultaneous Evaluation of a Large Number of Protein Variants for Their Impact on Conformational Stability. Anal Chem 2021; 93:14263-14271. [PMID: 34637272 DOI: 10.1021/acs.analchem.1c03335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A stable molecular structure is important in the development of a protein candidate into a therapeutic product. A therapeutic protein often contains many different variants; some of them may have an impact on the conformational stability of the protein. Conventionally, to evaluate the impact of a variant on stability, the variant must be enriched to a reasonable purity, and then its stability characterized by chromatographic or biophysical techniques. However, it is often impractical to purify and characterize each variant in a therapeutic protein. A workflow, based on limited proteolysis followed by MS detection, was established to simultaneously assess the impact of a large number of variants on conformational stability without enrichment. Because a less stable domain is more susceptible to proteolytic degradation, conformational stability of the domain can be reported from the release rate of a proteolytic peptide. A kinetic model is established to quantitatively determine the extent of domain stabilization/destabilization of different variants. The methodology is demonstrated by examining variants known to affect the stability of immunoglobulin domains, such as different N-glycoforms, methionine oxidations, and sequence variants. With this methodology, near 100 variants may be evaluated within 2 days in a single experiment. Insights into the sequence-stability relationship will be obtained by monitoring the large number of low-level sequence variants, facilitating engineering of more stable molecules.
Collapse
Affiliation(s)
- Zhongqi Zhang
- Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320 United States
| | - Bhavana Shah
- Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320 United States
| |
Collapse
|
10
|
Apostol I, Bondarenko PV, Ren D, Semin DJ, Wu CH, Zhang Z, Goudar CT. Enabling development, manufacturing, and regulatory approval of biotherapeutics through advances in mass spectrometry. Curr Opin Biotechnol 2021; 71:206-215. [PMID: 34508981 DOI: 10.1016/j.copbio.2021.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 07/20/2021] [Accepted: 08/04/2021] [Indexed: 10/20/2022]
Abstract
Rapid technological advances have significantly improved the capability, versatility, and robustness of mass spectrometers which has led to them playing a central role in the development, characterization, and regulatory filings of biopharmaceuticals. Their application spans the entire continuum of drug development, starting with discovery research through product development, characterization, and marketing authorization and continues well into product life cycle management. The scope of application extends beyond traditional protein characterization and includes elements like clone selection, cell culture physiology and bioprocess optimization, investigation support, and process analytical technology. More recently, advances in the MS-based multi-attribute method are enabling the introduction of MS in a cGMP environment for routine release and stability testing. While most applications of MS to date have been for monoclonal antibodies, the successes and learnings should translate to the characterization of next-gen biotherapeutics where modalities like multispecifics could be more prevalent. In this review, we describe the most significant advances in MS and correlate them to the broad spectrum of applications to biotherapeutic development. We anticipate rapid technological improvements to continue that will further accelerate widespread deployment of MS, thereby elevating our overall understanding of product quality and enabling attribute-focused product development.
Collapse
Affiliation(s)
- Izydor Apostol
- Attribute Sciences, Process Development, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, CA 91320, United States
| | - Pavel V Bondarenko
- Attribute Sciences, Process Development, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, CA 91320, United States
| | - Da Ren
- Attribute Sciences, Process Development, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, CA 91320, United States
| | - David J Semin
- Attribute Sciences, Process Development, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, CA 91320, United States
| | - Chao-Hsiang Wu
- Attribute Sciences, Process Development, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, CA 91320, United States
| | - Zhongqi Zhang
- Attribute Sciences, Process Development, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, CA 91320, United States
| | - Chetan T Goudar
- Attribute Sciences, Process Development, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, CA 91320, United States.
| |
Collapse
|
11
|
Zhang A, Chen Z, Li M, Qiu H, Lawrence S, Bak H, Li N. A general evidence-based sequence variant control limit for recombinant therapeutic protein development. MAbs 2021; 12:1791399. [PMID: 32744138 PMCID: PMC7531532 DOI: 10.1080/19420862.2020.1791399] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Sequence variants (SVs) resulting from unintended amino acid substitutions in recombinant therapeutic proteins have increasingly gained attention from both regulatory agencies and the biopharmaceutical industry given their potential impact on efficacy and safety. With well-optimized production systems, such sequence variants usually exist at very low levels in the final protein products due to the high fidelity of DNA replication and protein biosynthesis process in mammalian expression systems such as Chinese hamster ovary cell lines. However, their levels can be significantly elevated in cases where the selected production cell line has unexpected DNA mutations or the manufacturing process is not fully optimized, for example, if depletion of certain amino acids occurs in the cell culture media in bioreactors. Therefore, it is important to design and implement an effective monitoring and control strategy to prevent or minimize the possible risks of SVs during the early stage of product and process development. However, there is no well-established guidance from the regulatory agencies or consensus across the industry to assess and manage SV risks. A question frequently asked is: What levels of SVs can be considered acceptable during product and process development, but also have no negative effects on drug safety and efficacy in patients? To address this critical question, we have taken a holistic approach and conducted a comprehensive sequence variant analysis. To guide biologic development, a general SV control limit of 0.1% at individual amino acid sites was proposed and properly justified based on extensive literature review, SV benchmark survey of approved therapeutic proteins, and accumulated experience on SV control practice at Regeneron.
Collapse
Affiliation(s)
- Aming Zhang
- Analytical Chemistry, Regeneron Pharmaceuticals, Inc ., Tarrytown, New York, USA
| | - Zhengwei Chen
- Analytical Chemistry, Regeneron Pharmaceuticals, Inc ., Tarrytown, New York, USA
| | - Meinuo Li
- Analytical Chemistry, Regeneron Pharmaceuticals, Inc ., Tarrytown, New York, USA
| | - Haibo Qiu
- Analytical Chemistry, Regeneron Pharmaceuticals, Inc ., Tarrytown, New York, USA
| | - Shawn Lawrence
- Preclinical Manufacturing and Process Development , Tarrytown, New York, USA
| | - Hanne Bak
- Analytical Chemistry, Regeneron Pharmaceuticals, Inc ., Tarrytown, New York, USA
| | - Ning Li
- Analytical Chemistry, Regeneron Pharmaceuticals, Inc ., Tarrytown, New York, USA
| |
Collapse
|
12
|
Zhang Z, Chan PK, Richardson J, Shah B. An evaluation of instrument types for mass spectrometry-based multi-attribute analysis of biotherapeutics. MAbs 2021; 12:1783062. [PMID: 32643531 PMCID: PMC7531562 DOI: 10.1080/19420862.2020.1783062] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Multi-attribute methods (MAM), based on proteolytic digestion followed by liquid chromatography-mass spectrometry analysis of proteolytic peptides, have gained substantial attention in the biopharmaceutical industry for quantifying a variety of quality attributes for therapeutic proteins. Most MAM developed so far have been based on high-resolution mass spectrometers, due to their superb resolving power to distinguish analyte signals from interferences. Lower-resolution instruments, if demonstrated suitable, may further promote the adoption of the technology due to their low cost, small footprint, and ease of use. In this work, we compared the performance of a high-resolution instrument with a few low-resolution quadrupole-type instruments in quantifying a diverse set of quality attributes in a monoclonal antibody product. Different modes of operation for the quadrupole instruments, including scan mode, selected-ion monitoring and multiple-reaction monitoring, were evaluated. The high-resolution instrument has superb performance, with a quantitation limit of 0.002%. Single-quadrupole instruments in scan mode, on the other hand, provide a quantitation limit of about 1%, which may be fit-for-purpose for many routine MAM applications.
Collapse
Affiliation(s)
- Zhongqi Zhang
- Process Development, Amgen, Inc ., Thousand Oaks, CA, USA
| | - Pik K Chan
- Process Development, Amgen, Inc ., Thousand Oaks, CA, USA
| | | | - Bhavana Shah
- Process Development, Amgen, Inc ., Thousand Oaks, CA, USA
| |
Collapse
|
13
|
Identification, characterization and control of a sequence variant in monoclonal antibody drug product: a case study. Sci Rep 2021; 11:13233. [PMID: 34168178 PMCID: PMC8225904 DOI: 10.1038/s41598-021-92338-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 02/22/2021] [Indexed: 02/06/2023] Open
Abstract
Sequence variants (SV) in protein bio therapeutics can be categorized as unwanted impurities and may raise serious concerns in efficacy and safety of the product. Early detection of specific sequence modifications, that can result in altered physicochemical and or biological properties, is therefore desirable in product manufacturing. Because of their low abundance, and finite resolving power of conventional analytical techniques, they are often overlooked in early drug development. Here, we present a case study where trace amount of a sequence variant is identified in a monoclonal antibody (mAb) based therapeutic protein by LC-MS/MS and the structural and functional features of the SV containing mAb is assessed using appropriate analytical techniques. Further, a very sensitive selected reaction monitoring (SRM) technique is developed to quantify the SV which revealed both prominent and inconspicuous nature of the variant in process chromatography. We present the extensive characterization of a sequence variant in protein biopharmaceutical and first report on control of sequence variants to < 0.05% in final drug product by utilizing SRM based mass spectrometry method during the purification steps.
Collapse
|
14
|
Garofalo R, Wohlgemuth I, Pearson M, Lenz C, Urlaub H, Rodnina MV. Broad range of missense error frequencies in cellular proteins. Nucleic Acids Res 2019; 47:2932-2945. [PMID: 30649420 PMCID: PMC6451103 DOI: 10.1093/nar/gky1319] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/21/2018] [Accepted: 12/30/2018] [Indexed: 12/25/2022] Open
Abstract
Assessment of the fidelity of gene expression is crucial to understand cell homeostasis. Here we present a highly sensitive method for the systematic Quantification of Rare Amino acid Substitutions (QRAS) using absolute quantification by targeted mass spectrometry after chromatographic enrichment of peptides with missense amino acid substitutions. By analyzing incorporation of near- and non-cognate amino acids in a model protein EF-Tu, we show that most of missense errors are too rare to detect by conventional methods, such as DDA, and are estimated to be between <10−7–10-5 by QRAS. We also observe error hotspots of up to 10−3 for some types of mismatches, including the G-U mismatch. The error frequency depends on the expression level of EF-Tu and, surprisingly, the amino acid position in the protein. QRAS is not restricted to any particular miscoding event, organism, strain or model protein and is a reliable tool to analyze very rare proteogenomic events.
Collapse
Affiliation(s)
- Raffaella Garofalo
- Department of Physical Biochemistry, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Goettingen, Germany
| | - Ingo Wohlgemuth
- Department of Physical Biochemistry, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Goettingen, Germany
| | - Michael Pearson
- Department of Physical Biochemistry, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Goettingen, Germany
| | - Christof Lenz
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Goettingen, Germany.,Department of Clinical Chemistry, Bioanalytics, University Medical Center Goettingen, Robert-Koch-Straße 40, 37075 Goettingen, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Goettingen, Germany.,Department of Clinical Chemistry, Bioanalytics, University Medical Center Goettingen, Robert-Koch-Straße 40, 37075 Goettingen, Germany
| | - Marina V Rodnina
- Department of Physical Biochemistry, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Goettingen, Germany
| |
Collapse
|
15
|
Harris C, Xu W, Grassi L, Wang C, Markle A, Hardman C, Stevens R, Miro-Quesada G, Hatton D, Wang J. Identification and characterization of an IgG sequence variant with an 11 kDa heavy chain C-terminal extension using a combination of mass spectrometry and high-throughput sequencing analysis. MAbs 2019; 11:1452-1463. [PMID: 31570042 PMCID: PMC6816433 DOI: 10.1080/19420862.2019.1667740] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Protein primary structure is a potential critical quality attribute for biotherapeutics. Identifying and characterizing any sequence variants present is essential for product development. A sequence variant ~11 kDa larger than the expected IgG mass was observed by size-exclusion chromatography and two-dimensional liquid chromatography coupled with online mass spectrometry. Further characterization indicated that the 11 kDa was added to the heavy chain (HC) Fc domain. Despite the relatively large mass addition, only one unknown peptide was detected by peptide mapping. To decipher the sequence, the transcriptome of the manufacturing cell line was characterized by Illumina RNA-seq. Transcriptome reconstruction detected an aberrant fusion transcript, where the light chain (LC) constant domain sequence was fused to the 3ʹ end of the HC transcript. Translation of this fusion transcript generated an extended peptide sequence at the HC C-terminus corresponding to the observed 11 kDa mass addition. Nanopore-based genome sequencing showed multiple copies of the plasmid had integrated in tandem with one copy missing the 5ʹ end of the plasmid, deleting the LC variable domain. The fusion transcript was due to read-through of the HC terminator sequence into the adjacent partial LC gene and an unexpected splicing event between a cryptic splice-donor site at the 3ʹ end of the HC and the splice acceptor site at the 5ʹ end of the LC constant domain. Our study demonstrates that combining protein physicochemical characterization with genomic and transcriptomic analysis of the manufacturing cell line greatly improves the identification of sequence variants and understanding of the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Claire Harris
- Biopharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca , Cambridge , UK
| | - Weichen Xu
- Biopharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca , Gaithersburg , MD , USA
| | - Luigi Grassi
- Biopharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca , Cambridge , UK
| | - Chunlei Wang
- Biopharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca , Gaithersburg , MD , USA
| | - Abigail Markle
- Biopharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca , Gaithersburg , MD , USA
| | - Colin Hardman
- Data Science & Artificial Intelligence, BioPharmaceuticals R&D, AstraZeneca , Cambridge , UK
| | - Richard Stevens
- Antibody Discovery and Protein Engineering, BioPharmaceuticals R&D, AstraZeneca , Cambridge , UK
| | - Guillermo Miro-Quesada
- Data & Quantitative Sciences, Biopharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca , Gaithersburg , MD , USA
| | - Diane Hatton
- Biopharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca , Cambridge , UK
| | - Jihong Wang
- Biopharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca , Gaithersburg , MD , USA
| |
Collapse
|
16
|
Sequence Variant and Posttranslational Modification Analysis During Cell Line Selection via High-Throughput Peptide Mapping. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019. [PMID: 31347050 DOI: 10.1007/978-3-030-15950-4_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register]
Abstract
Selection of high-producing lead and backup cell lines with high-fidelity primary structure is a major goal of cell line development of protein therapeutics. Conventional techniques for sequence variant analysis, such as mass spectrometry (MS) and next-generation sequencing (NGS) have limitations on the sample number and turnaround time, thus often are only applied at the final stages of development, where an undesired lead or backup clone could cause a significant delay in project timeline. Here we presented a high-throughput (HT) peptide mapping workflow which can be applied at early stages of cell line selection for testing of a batch of 30-40 clones within 2-week turnaround while reporting valuable information on sequence variants and posttranslational modifications (PTMs). The successful application of this workflow was demonstrated for two mAb programs. Multiple clones were removed from a total of 33 mAb-1 clones using various criteria: nine clones contained at least one >1% upregulated unknown peptide ions, 11 clones contained at least eight >0.1% upregulated unknowns, and six clones contained upregulated critical PTMs. For mAb-2, light chain (LC) sequence extension of approximately 30 amino acids were detected in 6 out of 36 clones at levels up to 11%. Besides, a Q to H mutation at ~30% was detected in the heavy chain (HC) of a single clone. Q to H mutation has mass change of 9.00 Da and failed to be detected by intact mass analysis. Rapid PTM quantitation also facilitated the selection of clones with desirable quality attributes, such as N-glycan profile. Hence, we demonstrated a risk-reducing strategy where abnormal clones could be detected at earlier stages of cell line selection, which should result in reduced and more predictable timeline of cell line development.
Collapse
|
17
|
|
18
|
Wong HE, Huang CJ, Zhang Z. Amino Acid Misincorporation Propensities Revealed through Systematic Amino Acid Starvation. Biochemistry 2018; 57:6767-6779. [DOI: 10.1021/acs.biochem.8b00976] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- H. Edward Wong
- Process Development, Amgen, Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Chung-Jr Huang
- Process Development, Amgen, Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Zhongqi Zhang
- Process Development, Amgen, Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320, United States
| |
Collapse
|
19
|
Rehder DS, Wisniewski CJ, Liu D, Ren D, Farnan D, Schenauer MR. Expression vector-derived heterogeneity in a therapeutic IgG4 monoclonal antibody. MAbs 2018; 11:145-152. [PMID: 30365358 DOI: 10.1080/19420862.2018.1540254] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
While characterizing a therapeutic IgG4 monoclonal antibody (mAb), we observed a variant with a mass 1177 Da larger than the predominant mAb form that could not be ascribed to previously described modifications. Through successive rounds of experimentation, we localized the mass addition to the C-terminus of the heavy chain (HC). During this process we observed that when the mAb was broken down into separate domains, the Fc and the 1177 Da-modified Fc could be chromatographically separated. Separation allowed collection of native and modified Fc fractions for LC/MS peptide mapping. A unique peptide present in the modified fraction was de novo sequenced and demonstrated to be a modified form of the HC C-terminus lacking two native residues (GK) and gaining twelve additional non-native residues (EAEAASASELFQ). Aware of other mAb variants with genetic origins, we sought to understand whether this modification too had a genetic basis. In silico translation of the expression vector encoding the mAb demonstrated that a normally non-coding section of nucleotides in the + 1 reading frame relative to the HC C-terminal coding region could have led to a transcript with the non-native C-terminal extension. Two potential mechanisms for how this nucleotide sequence might have fused to the native HC coding region and led to expression of the extension product are presented.
Collapse
Affiliation(s)
- Douglas S Rehder
- a Biologics Analytical Operations , Gilead Sciences , Oceanside , CA , USA
| | - Chris J Wisniewski
- a Biologics Analytical Operations , Gilead Sciences , Oceanside , CA , USA
| | - Denfeng Liu
- a Biologics Analytical Operations , Gilead Sciences , Oceanside , CA , USA
| | - Diya Ren
- a Biologics Analytical Operations , Gilead Sciences , Oceanside , CA , USA
| | - Dell Farnan
- a Biologics Analytical Operations , Gilead Sciences , Oceanside , CA , USA
| | | |
Collapse
|
20
|
Lin TJ, Beal KM, Brown PW, DeGruttola HS, Ly M, Wang W, Chu CH, Dufield RL, Casperson GF, Carroll JA, Friese OV, Figueroa B, Marzilli LA, Anderson K, Rouse JC. Evolution of a comprehensive, orthogonal approach to sequence variant analysis for biotherapeutics. MAbs 2018; 11:1-12. [PMID: 30303443 PMCID: PMC6343769 DOI: 10.1080/19420862.2018.1531965] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Amino acid sequence variation in protein therapeutics requires close monitoring during cell line and cell culture process development. A cross-functional team of Pfizer colleagues from the Analytical and Bioprocess Development departments worked closely together for over 6 years to formulate and communicate a practical, reliable sequence variant (SV) testing strategy with state-of-the-art techniques that did not necessitate more resources or lengthen project timelines. The final Pfizer SV screening strategy relies on next-generation sequencing (NGS) and amino acid analysis (AAA) as frontline techniques to identify mammalian cell clones with genetic mutations and recognize cell culture process media/feed conditions that induce misincorporations, respectively. Mass spectrometry (MS)-based techniques had previously been used to monitor secreted therapeutic products for SVs, but we found NGS and AAA to be equally informative, faster, less cumbersome screening approaches. MS resources could then be used for other purposes, such as the in-depth characterization of product quality in the final stages of commercial-ready cell line and culture process development. Once an industry-wide challenge, sequence variation is now routinely monitored and controlled at Pfizer (and other biopharmaceutical companies) through increased awareness, dedicated cross-line efforts, smart comprehensive strategies, and advances in instrumentation/software, resulting in even higher product quality standards for biopharmaceutical products.
Collapse
Affiliation(s)
- T Jennifer Lin
- a Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Andover , MA , USA
| | - Kathryn M Beal
- a Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Andover , MA , USA
| | - Paul W Brown
- b Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Chesterfield , MO , USA
| | | | - Mellisa Ly
- a Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Andover , MA , USA
| | - Wenge Wang
- a Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Andover , MA , USA
| | - Chia H Chu
- b Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Chesterfield , MO , USA
| | - Robert L Dufield
- b Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Chesterfield , MO , USA
| | - Gerald F Casperson
- b Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Chesterfield , MO , USA
| | - James A Carroll
- b Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Chesterfield , MO , USA
| | - Olga V Friese
- b Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Chesterfield , MO , USA
| | - Bruno Figueroa
- a Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Andover , MA , USA
| | - Lisa A Marzilli
- a Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Andover , MA , USA
| | - Karin Anderson
- a Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Andover , MA , USA
| | - Jason C Rouse
- a Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Andover , MA , USA
| |
Collapse
|
21
|
Háda V, Bagdi A, Bihari Z, Timári SB, Fizil Á, Szántay C. Recent advancements, challenges, and practical considerations in the mass spectrometry-based analytics of protein biotherapeutics: A viewpoint from the biosimilar industry. J Pharm Biomed Anal 2018; 161:214-238. [PMID: 30205300 DOI: 10.1016/j.jpba.2018.08.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/08/2018] [Accepted: 08/10/2018] [Indexed: 01/22/2023]
Abstract
The extensive analytical characterization of protein biotherapeutics, especially of biosimilars, is a critical part of the product development and registration. High-resolution mass spectrometry became the primary analytical tool used for the structural characterization of biotherapeutics. Its high instrumental sensitivity and methodological versatility made it possible to use this technique to characterize both the primary and higher-order structure of these proteins. However, even by using high-end instrumentation, analysts face several challenges with regard to how to cope with industrial and regulatory requirements, that is, how to obtain accurate and reliable analytical data in a time- and cost-efficient way. New sample preparation approaches, measurement techniques and data evaluation strategies are available to meet those requirements. The practical considerations of these methods are discussed in the present review article focusing on hot topics, such as reliable and efficient sequencing strategies, minimization of artefact formation during sample preparation, quantitative peptide mapping, the potential of multi-attribute methodology, the increasing role of mass spectrometry in higher-order structure characterization and the challenges of MS-based identification of host cell proteins. On the basis of the opportunities in new instrumental techniques, methodological advancements and software-driven data evaluation approaches, for the future one can envision an even wider application area for mass spectrometry in the biopharmaceutical industry.
Collapse
Affiliation(s)
- Viktor Háda
- Analytical Department of Biotechnology, Gedeon Richter Plc, Hungary.
| | - Attila Bagdi
- Analytical Department of Biotechnology, Gedeon Richter Plc, Hungary
| | - Zsolt Bihari
- Analytical Department of Biotechnology, Gedeon Richter Plc, Hungary
| | | | - Ádám Fizil
- Analytical Department of Biotechnology, Gedeon Richter Plc, Hungary
| | - Csaba Szántay
- Spectroscopic Research Department, Gedeon Richter Plc, Hungary.
| |
Collapse
|