1
|
Okada R, Shibata K, Shibuya R, Torisu T, Uchiyama S. Hierarchical clustering of therapeutic proteins based on agitation-induced aggregation propensity and its relation to physicochemical parameters. Eur J Pharm Sci 2025; 208:107060. [PMID: 40086315 DOI: 10.1016/j.ejps.2025.107060] [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: 10/25/2024] [Revised: 02/05/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Physical stresses such as agitation induce protein aggregation, which causes adverse effects on the immune system of patients, leading to challenges in drug development. Aggregation induced by physical stresses can be minimized by formulation optimization. In this study, 120 combinations of 10 therapeutic proteins and 12 different formulations (4 pH conditions and 3 salt concentrations) were prepared. Subsequently, the agitation-induced aggregation propensity of each protein was investigated by evaluating its monomer recovery (%) using size exclusion chromatography. Hierarchical clustering was applied to categorize each protein according to its aggregation propensity, resulting in two groups of proteins: group A and B. The aggregation propensity of proteins in group A was insensitive to changes in formulation conditions because conformational, colloidal, and interfacial stabilities were minimally affected by changes in the pH and salt concentration and a compensation mechanism existed between conformational and colloidal stabilities. Thus, proteins in group A can be formulated with a relatively high degree of freedom. In contrast, the aggregation propensity of proteins in group B was sensitive to changes in formulation conditions. Multiple regression analysis of the physicochemical parameters and monomer recovery of proteins in group B clarified that changes in conformational stability in response to changes in formulations primarily contributed to the sensitivity of the monomer recovery to changes in formulation conditions. For all antibodies, there was a positive correlation between the monomer recovery after agitation and that after quiescent storage at 40 °C for 1 month, suggesting that a stable formulation can be obtained without the quiescent testing. Therefore, a proposed formulation optimization strategy based on the agitation-induced monomer recovery can improve the efficiency of formulating selected therapeutic proteins. This strategic approach is expected to accelerate the development of therapeutic proteins while reflecting the importance of aggregation factors and quiescent stability in the optimization of therapeutic protein formulations.
Collapse
Affiliation(s)
- Rio Okada
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Japan
| | - Kosei Shibata
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Japan; U-Medico Inc., 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Risa Shibuya
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Japan
| | - Tetsuo Torisu
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Japan
| | - Susumu Uchiyama
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Japan; U-Medico Inc., 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
2
|
Garg R, McCarthy S, Thompson AG, Zhang J, Mattson E, Clabbers A, Acquah A, Xu J, Zhou C, Ali A, Filoti D, Singh R. In vitro Stability Study of a Panel of Commercial Antibodies at Physiological pH and Temperature as a Guide to Screen Biologic Candidate Molecules for the Potential Risk of In vivo Asparagine Deamidation and Activity Loss. Pharm Res 2025; 42:353-363. [PMID: 39979532 PMCID: PMC11880144 DOI: 10.1007/s11095-025-03825-3] [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: 11/11/2024] [Accepted: 01/19/2025] [Indexed: 02/22/2025]
Abstract
OBJECTIVE Biologic drug molecules such as antibodies are exposed to the physiological stress conditions of pH 7.4 and 37°C during their long circulation lifetime in vivo. The stress on biologic molecules in vivo is more severe compared to that under typical storage conditions of low pH formulation and cold temperature. Chemical degradation of critical residues such as asparagine may occur in vivo, leading to potential loss of biological activity. This study describes a physiologically relevant and convenient in vitro PBS stress condition of pH 7.4 and 40°C for pre-clinical stability screening of biologic molecules. METHODS As benchmarks, multiple commercial antibodies (alirocumab, evolocumab, golimumab, ramucirumab, and trastuzumab) were tested in parallel for formulation stability at storage and accelerated temperature conditions and for physiological stability at pH 7.4 and 40°C stress both for 3-4 weeks. The stressed antibodies were monitored for chemical modification and target binding, without requiring affinity purification. RESULTS The major CDR chemical modifications observed in PBS-stressed commercial antibodies were deamidations of asparagine residues. Although slight decreases in target binding were observed for two antibodies, the affinities overall remained strong after PBS stress. CONCLUSIONS This benchmarking study of commercial antibodies would be useful as a guide to screen discovery-stage biologic molecules both for drug product stability at formulation pH under storage and accelerated temperature conditions and for physiological stability under in vivo-mimicking pH and temperature stress condition.
Collapse
Affiliation(s)
- Richa Garg
- Biologics CMC Drug Product Development, Preformulation, AbbVie, Worcester, MA, 01605, USA
| | - Sean McCarthy
- Biologics CMC Analytical Research and Development, Developability, AbbVie, Worcester, MA, 01605, USA
| | - Alayna George Thompson
- Biologics CMC Analytical Research and Development, Developability, AbbVie, North Chicago, IL, 60064, USA
| | - Jiang Zhang
- Analytical Development, Product Development Science & Technology, AbbVie, Worcester, MA, 01605, USA
| | - Emily Mattson
- Biomolecular Interaction Group, Protein Sciences, Biotherapeutics and Genetic Medicine, AbbVie, Worcester, MA, 01605, USA
| | - Anca Clabbers
- Biomolecular Interaction Group, Protein Sciences, Biotherapeutics and Genetic Medicine, AbbVie, Worcester, MA, 01605, USA
| | - Aimalohi Acquah
- Biologics CMC Drug Product Development, Preformulation, AbbVie, Worcester, MA, 01605, USA
| | - Jianwen Xu
- Biologics CMC Drug Product Development, Preformulation, AbbVie, Worcester, MA, 01605, USA
- Kiniksa Pharmaceuticals, Lexington, MA, 02421, USA
| | - Chen Zhou
- Biologics CMC Drug Product Development, Preformulation, AbbVie, Worcester, MA, 01605, USA
| | - Amr Ali
- Analytical Development, Product Development Science & Technology, AbbVie, Worcester, MA, 01605, USA
| | - Dana Filoti
- Biologics CMC Analytical Research and Development, Developability, AbbVie, Worcester, MA, 01605, USA
| | - Rajeeva Singh
- Biologics CMC Drug Product Development, Preformulation, AbbVie, Worcester, MA, 01605, USA.
| |
Collapse
|
3
|
Dykstra AB, Lubinsky TG, Vitrac H, Campuzano IDG, Bondarenko PV, Simone AR. Utilization of Liquid Chromatography-Mass Spectrometry and High-Resolution Ion Mobility-Mass Spectrometry to Characterize Therapeutically Relevant Peptides with Asparagine Deamidation and Isoaspartate. Anal Chem 2025; 97:749-757. [PMID: 39714115 DOI: 10.1021/acs.analchem.4c05246] [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: 12/24/2024]
Abstract
Rapid identification of asparagine (Asn) deamidation and isoaspartate (isoAsp) in proteins remains a challenging analytical task during the development of biological therapeutics. For this study, 46 therapeutically relevant peptides corresponding to 13 peptide families (13 unmodified peptides and 33 modified peptides) were obtained; modified peptides included Asn deamidation and isoAsp. The peptide families were characterized by three methods: reversed-phase ultrahigh performance liquid chromatography-mass spectrometry (RP-UHPLC-MS); flow injection analysis high-resolution ion mobility-mass spectrometry (FIA-HRIM-MS); and shortened gradient RP-UHPLC-HRIM-MS. UHPLC-MS data acquisition was 2 h per injection, in contrast to high-throughput 1 min data acquisition of the FIA-HRIM-MS technique. A rapid 2D peptide map has been demonstrated by combining shortened gradient RP-UHPLC with HRIM, to optimize the resolution of the Asn-, Asp-, and isoAsp-containing peptides, increasing the likelihood of detecting peptides containing these quality attributes with expedited data acquisition. Additionally, this paper provides an ion mobility calibration data set for therapeutically relevant peptides (unmodified and modified) over an ion-neutral collisional cross-section range of 300-800 Å2.
Collapse
Affiliation(s)
- Andrew B Dykstra
- Pre-Pivotal Attribute Sciences, Amgen, Inc, Thousand Oaks, California 91320, United States
| | | | - Heidi Vitrac
- MOBILion Systems, Chadds Ford, Pennsylvania 19317, United States
| | - Iain D G Campuzano
- Molecular Analytics, Amgen, Inc, Thousand Oaks, California 91320, United States
| | - Pavel V Bondarenko
- Pre-Pivotal Attribute Sciences, Amgen, Inc, Thousand Oaks, California 91320, United States
| | - Ashli R Simone
- MOBILion Systems, Chadds Ford, Pennsylvania 19317, United States
| |
Collapse
|
4
|
Yang J, Tan HY, Yuan J, Huang Y, Rosenbaum AI. Detailed Structural Elucidation of Antibody-Drug Conjugate Biotransformation Species Using High Resolution Multiple Reaction Monitoring Mass Spectrometry with Orthogonal Dissociation Methods. ACS Pharmacol Transl Sci 2025; 8:113-123. [PMID: 39816793 PMCID: PMC11729422 DOI: 10.1021/acsptsci.4c00445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 01/18/2025]
Abstract
Antibody-drug conjugates (ADCs) are a promising drug modality substantially expanding in both the discovery space and clinical development. Assessing the biotransformation of ADCs in vitro and in vivo is important in understanding their stability and pharmacokinetic properties. We previously reported biotransformation pathways for the anti-B7H4 topoisomerase I inhibitor ADC, AZD8205, puxitatug samrotecan, that underpin its structural stability in vivo using an intact protein liquid chromatography-high resolution mass spectrometry (LC-HRMS) approach. Herein, we employed a LC-high resolution multiple reaction monitoring (LC-MRMHR) approach using both collision-induced dissociation (CID) and electron-activated dissociation (EAD) methods, confirming our earlier findings. Furthermore, we were able to obtain additional detailed structural information on the biotransformation products expanding on earlier intact analyses. We also highlight the high sensitivity of LC-MRMHR for successfully identifying minor biotransformation products at low concentrations that were not detectable using the intact protein LC-HRMS workflow. Especially, EAD aided in the confirmation of biotransformation species that contain newly formed disulfide bonds due to the preferential dissociation of disulfide bonds using this method. We observed biotransformation reactions that vary between linker-payload (PL) conjugation sites on the antibody. For example, the trend toward constitutional isomerism in thio-succinimide linker hydrolysis, and the resulting positional isomers from thiol adduct formation following linker-PL deconjugation. The reported orthogonal analytical approaches highly complement and fortify the intact protein LC-HRMS data. This study sheds further light on detailed structural characterization of various ADC species and validates the proposed biotransformation pathways explaining the stability of AZD8205 in vivo.
Collapse
Affiliation(s)
| | - Hui Yin Tan
- Integrated Bioanalysis, Clinical
Pharmacology and Safety Sciences, R&D, AstraZeneca, 121 Oyster
Point Blvd, South San Francisco, California 94080, United States
| | - Jiaqi Yuan
- Integrated Bioanalysis, Clinical
Pharmacology and Safety Sciences, R&D, AstraZeneca, 121 Oyster
Point Blvd, South San Francisco, California 94080, United States
| | | | | |
Collapse
|
5
|
Chawathe A, Ahire V, Luthra K, Patil B, Garkhal K, Sharma N. Analytical and drug delivery strategies for short peptides: From manufacturing to market. Anal Biochem 2025; 696:115699. [PMID: 39461693 DOI: 10.1016/j.ab.2024.115699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/17/2024] [Accepted: 10/23/2024] [Indexed: 10/29/2024]
Abstract
In recent times, biopharmaceuticals have gained attention because of their tremendous potential to benefit millions of patients globally by treating widespread diseases such as cancer, diabetes and many rare diseases. Short peptides (SP), also termed as oligopeptides, are one such class of biopharmaceuticals, that are majorly involved in efficient functioning of biological systems. Peptide chains that are 2-20 amino acids long are considered as oligopeptides by researchers and are some of the functionally vital compounds with widespread applications including self-assembly material for drug delivery, targeting ligands for precise/specific targeting and other biological uses. Using functionalised biomacromolecules such as short chained peptides, helps in improving pharmacokinetic properties and biodistribution profile of the drug. Apart from this, functionalised SP are being employed as cell penetrating peptides and prodrug to specifically and selectively target tumor sites. In order to minimize any unwanted interaction and adverse effects, the stability and safety of SP should be ensured throughout its development from manufacturing to market. Formulation development and characterization strategies of these potential molecules are described in the following review along with various applications and details of marketed formulations.
Collapse
Affiliation(s)
- Ashwini Chawathe
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research-Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Vishal Ahire
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research-Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Kshitiz Luthra
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Bhumika Patil
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Kalpna Garkhal
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat, 382355, India.
| | - Nitish Sharma
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research-Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat, 382355, India.
| |
Collapse
|
6
|
Zhang H, Xiong Y, Shi X, Zhu L, Wu Q, Wu T, Du Y. Deamidation analysis of therapeutic drugs using matrix-assisted laser desorption ionization mass spectrometry and a novel algorithm QuanDA. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 17:37-44. [PMID: 39560336 DOI: 10.1039/d4ay01595a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
A robust deamidation quantification method, called QuanDA, was developed to quantify the spontaneous nonenzymatic deamidation of peptides based on the isotopic distribution change of peptides in matrix-assisted laser desorption ionization (MALDI) mass spectra and non-negative least squares calculation. The predictive model of QuanDA using theoretical spectra of pure un-deamidated and deamidated peptides for a series of simulated partial deamidated peptides is satisfying, with a coefficient of determination (R2) and root mean squared error (RMSE) of 0.9914 and 0.03356, respectively. It was applicable in cases where there is a lack of reference standards of un-deamidated and deamidated peptides. The only requirements were the chemical formulae of un-deamidated and deamidated peptides for isotopic pattern calculation. QuanDA provided a rapid, low-cost and easily accessible method for deamidation analysis in therapeutic drugs.
Collapse
Affiliation(s)
- Han Zhang
- School of Chemistry and Molecular Engineering and Research Centre of Analysis and Test, East China University of Science and Technology, Shanghai, 200237, P. R. China.
| | - Yinran Xiong
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Bio-logical Science Research Center, Southwest University, Chongqing, 400715, China
- Chongqing Municipal Key Laboratory of Scientific Utilization of Tobacco Resources, Chongqing, 400060, China
| | - Xiaonan Shi
- School of Chemistry and Molecular Engineering and Research Centre of Analysis and Test, East China University of Science and Technology, Shanghai, 200237, P. R. China.
| | - Lijia Zhu
- School of Chemistry and Molecular Engineering and Research Centre of Analysis and Test, East China University of Science and Technology, Shanghai, 200237, P. R. China.
| | - Qiong Wu
- School of Chemistry and Molecular Engineering and Research Centre of Analysis and Test, East China University of Science and Technology, Shanghai, 200237, P. R. China.
| | - Ting Wu
- School of Chemistry and Molecular Engineering and Research Centre of Analysis and Test, East China University of Science and Technology, Shanghai, 200237, P. R. China.
| | - Yiping Du
- School of Chemistry and Molecular Engineering and Research Centre of Analysis and Test, East China University of Science and Technology, Shanghai, 200237, P. R. China.
| |
Collapse
|
7
|
Sarin D, Kumar S, Rathore AS. Offline Coupling of Hydrophobic Interaction Chromatography-Capillary Zone Electrophoresis for Monitoring Charge-Based Heterogeneity of Recombinant Monoclonal Antibodies. Electrophoresis 2024; 45:1927-1938. [PMID: 39402848 DOI: 10.1002/elps.202400158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/11/2024] [Accepted: 09/23/2024] [Indexed: 12/22/2024]
Abstract
A holistic understanding of the charge heterogeneity in monoclonal antibodies (mAbs) is paramount for ensuring acceptable product quality. Hence, biotherapeutic manufacturers are expected to thoroughly characterize their products via advanced analytical techniques. Recently, two-dimensional liquid chromatography (2DLC) methods have gained popularity for resolving complex charged species. Capillary electrophoresis (CE) is regarded as a sensitive and faster tool for charged species estimation in biotherapeutics. In this study, we aim to combine the separation power of chromatographic and electrophoretic tools (liquid chromatography [LC]-CE) so as to achieve maximum resolution of mAb charge variants. Hydrophobic interaction chromatography (HIC) has been used as the preferred LC mode with CE for achieving successful separation of both charge and hydrophobic variants for two of the mAbs (trastuzumab and rituximab). The standalone HIC and capillary zone electrophoresis (CZE) methods separated 4 hydrophobic variants and 7 charge variants for each mAb, whereas the 2DLC method separated 10 and 11 variants for mAbs A and B. On the other hand, the HIC-CZE-UV method resolved 29 variants in mAb A and 23 variants in mAb B. The reproducibility of the HIC-CZE-UV method was demonstrated by % change in values of retention time (RT) and peak area as <5% (mAb A), <3% (mAb B), and <12% (for both mAbs), respectively. Thus, the utility of the proposed LC-CE method for characterization of mAb charge variants has been displayed.
Collapse
Affiliation(s)
- Deepika Sarin
- Department of Chemical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Sunil Kumar
- Department of Chemical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Anurag S Rathore
- Department of Chemical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| |
Collapse
|
8
|
Niu B, Lee B, Wang L, Chen W, Johnson J. The Accurate Prediction of Antibody Deamidations by Combining High-Throughput Automated Peptide Mapping and Protein Language Model-Based Deep Learning. Antibodies (Basel) 2024; 13:74. [PMID: 39311379 PMCID: PMC11417914 DOI: 10.3390/antib13030074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 08/30/2024] [Accepted: 09/06/2024] [Indexed: 09/26/2024] Open
Abstract
Therapeutic antibodies such as monoclonal antibodies (mAbs), bispecific and multispecific antibodies are pivotal in therapeutic protein development and have transformed disease treatments across various therapeutic areas. The integrity of therapeutic antibodies, however, is compromised by sequence liabilities, notably deamidation, where asparagine (N) and glutamine (Q) residues undergo chemical degradations. Deamidation negatively impacts the efficacy, stability, and safety of diverse classes of antibodies, thus necessitating the critical need for the early and accurate identification of vulnerable sites. In this article, a comprehensive antibody deamidation-specific dataset (n = 2285) of varied modalities was created by using high-throughput automated peptide mapping followed by supervised machine learning to predict the deamidation propensities, as well as the extents, throughout the entire antibody sequences. We propose a novel chimeric deep learning model, integrating protein language model (pLM)-derived embeddings with local sequence information for enhanced deamidation predictions. Remarkably, this model requires only sequence inputs, eliminating the need for laborious feature engineering. Our approach demonstrates state-of-the-art performance, offering a streamlined workflow for high-throughput automated peptide mapping and deamidation prediction, with the potential of broader applicability to other antibody sequence liabilities.
Collapse
Affiliation(s)
- Ben Niu
- Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, CA 92121, USA
| | - Benjamin Lee
- Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, CA 92121, USA
| | - Lili Wang
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Wen Chen
- Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, CA 92121, USA
| | - Jeffrey Johnson
- Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, CA 92121, USA
| |
Collapse
|
9
|
Li W, Lin H, Huang Z, Xie S, Zhou Y, Gong R, Jiang Q, Xiang C, Huang J. DOTAD: A Database of Therapeutic Antibody Developability. Interdiscip Sci 2024; 16:623-634. [PMID: 38530613 DOI: 10.1007/s12539-024-00613-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 01/25/2024] [Accepted: 01/27/2024] [Indexed: 03/28/2024]
Abstract
The development of therapeutic antibodies is an important aspect of new drug discovery pipelines. The assessment of an antibody's developability-its suitability for large-scale production and therapeutic use-is a particularly important step in this process. Given that experimental assays to assess antibody developability in large scale are expensive and time-consuming, computational methods have been a more efficient alternative. However, the antibody research community faces significant challenges due to the scarcity of readily accessible data on antibody developability, which is essential for training and validating computational models. To address this gap, DOTAD (Database Of Therapeutic Antibody Developability) has been built as the first database dedicated exclusively to the curation of therapeutic antibody developability information. DOTAD aggregates all available therapeutic antibody sequence data along with various developability metrics from the scientific literature, offering researchers a robust platform for data storage, retrieval, exploration, and downloading. In addition to serving as a comprehensive repository, DOTAD enhances its utility by integrating a web-based interface that features state-of-the-art tools for the assessment of antibody developability. This ensures that users not only have access to critical data but also have the convenience of analyzing and interpreting this information. The DOTAD database represents a valuable resource for the scientific community, facilitating the advancement of therapeutic antibody research. It is freely accessible at http://i.uestc.edu.cn/DOTAD/ , providing an open data platform that supports the continuous growth and evolution of computational methods in the field of antibody development.
Collapse
Affiliation(s)
- Wenzhen Li
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Hongyan Lin
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Ziru Huang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Shiyang Xie
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Yuwei Zhou
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Rong Gong
- School of Computer Science and Technology, Aba Teachers University, Aba, 623002, China
| | - Qianhu Jiang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - ChangCheng Xiang
- School of Computer Science and Technology, Aba Teachers University, Aba, 623002, China.
| | - Jian Huang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China.
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, 611844, China.
| |
Collapse
|
10
|
Sarin D, Kumar S, Rathore AS. Titer and charge-based heterogeneity multiattribute monitoring of mAbs in cell culture harvest using 2D ProA CEX MS. Talanta 2024; 276:126232. [PMID: 38749159 DOI: 10.1016/j.talanta.2024.126232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/24/2024] [Accepted: 05/07/2024] [Indexed: 06/14/2024]
Abstract
Robust monitoring of heterogeneity in biopharmaceutical development is crucial for producing safe and efficacious biotherapeutic products. Multiattribute monitoring (MAM) has emerged as an efficient tool for monitoring of mAb heterogeneities like deamidation, sialylation, glycosylation, and oxidation. Conventional biopharma analysis during mAb development relies on use of one-dimensional methods for monitoring titer and charge-based heterogeneity using non-volatile solvents without direct coupling with mass spectrometry (MS). This approach requires analysis of mAb harvest by ProA for titer estimation followed by separate cation exchange chromatography (CEX) analysis of the purified sample for estimating charge-based heterogeneity. This can take up to 60-90 min due to the required fraction collection and buffer exchange steps. In this work, a native two-dimensional liquid chromatography (2DLC) mass spectrometry method has been developed with Protein A chromatography in the first dimension for titer estimation and cation exchange chromatography (CEX) in the second dimension for charge variant analysis. The method uses volatile salts for both dimensions and enables easy coupling to MS. The proposed 2DLC method exhibits a charge variant profile that is similar to that observed via the traditional methods and takes only 15 min for mass identification of each variant. A total of six charge variants were separated by the CEX analysis after titer estimation, including linearity assessment from 5 μg to 160 μg of injected mAb sample. The proposed method successfully estimated charge variants for the mAb innovator and 4 of its biosimilars, showcasing its applicability for biosimilarity exercises. Hence, the 2D ProA CEX MS method allows direct titer and charge variant estimation of mAbs in a single workflow.
Collapse
Affiliation(s)
- Deepika Sarin
- Department of Chemical Engineering, Indian Institute of Technology, Delhi, Hauz Khas, New Delhi, 110016, India
| | - Sunil Kumar
- Department of Chemical Engineering, Indian Institute of Technology, Delhi, Hauz Khas, New Delhi, 110016, India
| | - Anurag S Rathore
- Department of Chemical Engineering, Indian Institute of Technology, Delhi, Hauz Khas, New Delhi, 110016, India.
| |
Collapse
|
11
|
Manning MC, Holcomb RE, Payne RW, Stillahn JM, Connolly BD, Katayama DS, Liu H, Matsuura JE, Murphy BM, Henry CS, Crommelin DJA. Stability of Protein Pharmaceuticals: Recent Advances. Pharm Res 2024; 41:1301-1367. [PMID: 38937372 DOI: 10.1007/s11095-024-03726-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/03/2024] [Indexed: 06/29/2024]
Abstract
There have been significant advances in the formulation and stabilization of proteins in the liquid state over the past years since our previous review. Our mechanistic understanding of protein-excipient interactions has increased, allowing one to develop formulations in a more rational fashion. The field has moved towards more complex and challenging formulations, such as high concentration formulations to allow for subcutaneous administration and co-formulation. While much of the published work has focused on mAbs, the principles appear to apply to any therapeutic protein, although mAbs clearly have some distinctive features. In this review, we first discuss chemical degradation reactions. This is followed by a section on physical instability issues. Then, more specific topics are addressed: instability induced by interactions with interfaces, predictive methods for physical stability and interplay between chemical and physical instability. The final parts are devoted to discussions how all the above impacts (co-)formulation strategies, in particular for high protein concentration solutions.'
Collapse
Affiliation(s)
- Mark Cornell Manning
- Legacy BioDesign LLC, Johnstown, CO, USA.
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA.
| | - Ryan E Holcomb
- Legacy BioDesign LLC, Johnstown, CO, USA
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA
| | - Robert W Payne
- Legacy BioDesign LLC, Johnstown, CO, USA
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA
| | - Joshua M Stillahn
- Legacy BioDesign LLC, Johnstown, CO, USA
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA
| | | | | | | | | | | | - Charles S Henry
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA
| | | |
Collapse
|
12
|
Malani H, Shrivastava A, Nupur N, Rathore AS. LC-MS Characterization and Stability Assessment Elucidate Correlation Between Charge Variant Composition and Degradation of Monoclonal Antibody Therapeutics. AAPS J 2024; 26:42. [PMID: 38570351 DOI: 10.1208/s12248-024-00915-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/19/2024] [Indexed: 04/05/2024] Open
Abstract
Aggregation stability of monoclonal antibody (mAb) therapeutics is influenced by many critical quality attributes (CQA) such as charge and hydrophobic variants in addition to environmental factors. In this study, correlation between charge heterogeneity and stability of mAbs for bevacizumab and trastuzumab has been investigated under a variety of stresses including thermal stress at 40 °C, thermal stress at 55 °C, shaking (mechanical), and low pH. Size- and charge-based heterogeneities were monitored using analytical size exclusion chromatography (SEC) and cation exchange chromatography (CEX), respectively, while dynamic light scattering was used to assess changes in hydrodynamic size. CEX analysis revealed an increase in cumulative acidic content for all variants of both mAbs post-stress treatment attributed to increased deamidation. Higher charge heterogeneity was observed in variants eluting close to the main peak than the ones eluting further away (25-fold and 42-fold increase in acidic content for main and B1 of bevacizumab and 19-fold for main of trastuzumab, respectively, under thermal stress; 50-fold increase in acidic for main and B1 of bevacizumab and 10% rise in basic content of main of trastuzumab under pH stress). Conversely, variants eluting far away from main exhibit greater aggregation as compared to close-eluting ones. Aggregation kinetics of variants followed different order for the different stresses for both mAbs (2nd order for thermal and pH stresses and 0th order for shaking stress). Half-life of terminal charge variants of both mAbs was 2- to 8-fold less than main indicating increased degradation propensity.
Collapse
Affiliation(s)
- Himanshu Malani
- Department of Chemical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Anuj Shrivastava
- Department of Chemical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Neh Nupur
- Department of Chemical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Anurag S Rathore
- Department of Chemical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India.
| |
Collapse
|
13
|
Pastrana B, Culyba E, Nieves S, Sazinsky SL, Canto EI, Noda I. Streamlined Multi-Attribute Assessment of an Array of Clinical-Stage Antibodies: Relationship Between Degradation and Stability. APPLIED SPECTROSCOPY 2024; 79:37028241231824. [PMID: 38419510 PMCID: PMC11684140 DOI: 10.1177/00037028241231824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/18/2024] [Indexed: 03/02/2024]
Abstract
Clinical antibodies are an important class of drugs for the treatment of both chronic and acute diseases. Their manufacturability is subject to evaluation to ensure product quality and efficacy. One critical quality attribute is deamidation, a non-enzymatic process that is observed to occur during thermal stress, at low or high pH, or a combination thereof. Deamidation may induce antibody instability and lead to aggregation, which may pose immunogenicity concerns. The introduction of a negative charge via deamidation may impact the desired therapeutic function (i) within the complementarity-determining region, potentially causing loss of efficacy; or (ii) within the fragment crystallizable region, limiting the effector function involving antibody-dependent cellular cytotoxicity. Here we describe a transformative solution that allows for a comparative assessment of deamidation and its impact on stability and aggregation. The innovative streamlined method evaluates the intact protein in its formulation conditions. This breakthrough platform technology is comprised of a quantum cascade laser microscope, a slide cell array that allows for flexibility in the design of experiments, and dedicated software. The enhanced spectral resolution is achieved using two-dimensional correlation, co-distribution, and two-trace two-dimensional correlation spectroscopies that reveal the molecular impact of deamidation. Eight re-engineered immunoglobulin G4 scaffold clinical antibodies under control and forced degradation conditions were evaluated for deamidation and aggregation. We determined the site of deamidation, the overall extent of deamidation, and where applicable, whether the deamidation event led to self-association or aggregation of the clinical antibody and the molecular events that led to the instability. The results were confirmed using orthogonal techniques for four of the samples.
Collapse
Affiliation(s)
- Belinda Pastrana
- Research and Development, Protein Dynamic Solutions, Inc., Wakefield, Massachusetts, USA
| | - Elizabeth Culyba
- Research and Development, Protein Dynamic Solutions, Inc., Wakefield, Massachusetts, USA
- Antibody Discovery, Verseau Therapeutics, Inc., Bedford, Massachusetts, USA
| | - Sherly Nieves
- Research and Development, Protein Dynamic Solutions, Inc., Wakefield, Massachusetts, USA
| | | | - Eduardo I. Canto
- Translational Sciences, Auxilio BioLab, Auxilio Mutuo Hospital, San Juan, Puerto Rico, USA
| | - Isao Noda
- Infectious Disease Research, Department of Materials Sciences and Engineering, University of Delaware, Newark, Delaware, USA
| |
Collapse
|
14
|
Hoffmann D, Bauer J, Kossner M, Henry A, Karow-Zwick AR, Licari G. Predicting deamidation and isomerization sites in therapeutic antibodies using structure-based in silico approaches. MAbs 2024; 16:2333436. [PMID: 38546837 PMCID: PMC10984128 DOI: 10.1080/19420862.2024.2333436] [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: 09/08/2023] [Accepted: 03/18/2024] [Indexed: 04/02/2024] Open
Abstract
Asparagine (Asn) deamidation and aspartic acid (Asp) isomerization are common degradation pathways that affect the stability of therapeutic antibodies. These modifications can pose a significant challenge in the development of biopharmaceuticals. As such, the early engineering and selection of chemically stable monoclonal antibodies (mAbs) can substantially mitigate the risk of subsequent failure. In this study, we introduce a novel in silico approach for predicting deamidation and isomerization sites in therapeutic antibodies by analyzing the structural environment surrounding asparagine and aspartate residues. The resulting quantitative structure-activity relationship (QSAR) model was trained using previously published forced degradation data from 57 clinical-stage mAbs. The predictive accuracy of the model was evaluated for four different states of the protein structure: (1) static homology models, (2) enhancing low-frequency vibrational modes during short molecular dynamics (MD) runs, (3) a combination of (2) with a protonation state reassignment, and (4) conventional full-atomistic MD simulations. The most effective QSAR model considered the accessible surface area (ASA) of the residue, the pKa value of the backbone amide, and the root mean square deviations of both the alpha carbon and the side chain. The accuracy was further enhanced by incorporating the QSAR model into a decision tree, which also includes empirical information about the sequential successor and the position in the protein. The resulting model has been implemented as a plugin named "Forecasting Reactivity of Isomerization and Deamidation in Antibodies" in MOE software, completed with a user-friendly graphical interface to facilitate its use.
Collapse
Affiliation(s)
- David Hoffmann
- Early Stage Pharmaceutical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach/Riss, Germany
- In Silico Team, Boehringer Ingelheim, Biberach/Riss, Germany
| | - Joschka Bauer
- Early Stage Pharmaceutical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach/Riss, Germany
- In Silico Team, Boehringer Ingelheim, Biberach/Riss, Germany
| | - Markus Kossner
- Scientific Services, Chemical Computing Group, Cologne, Germany
| | - Andrew Henry
- Scientific Support, Chemical Computing Group, Cambridge, UK
| | - Anne R. Karow-Zwick
- Early Stage Pharmaceutical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach/Riss, Germany
- In Silico Team, Boehringer Ingelheim, Biberach/Riss, Germany
| | - Giuseppe Licari
- Early Stage Pharmaceutical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach/Riss, Germany
- In Silico Team, Boehringer Ingelheim, Biberach/Riss, Germany
| |
Collapse
|
15
|
De Sciscio M, Nardi AN, Centola F, Rossi M, Guarnera E, D’Abramo M. Molecular Modeling of the Deamidation Reaction in Solution: A Theoretical-Computational Study. J Phys Chem B 2023; 127:9550-9559. [PMID: 37903302 PMCID: PMC10641835 DOI: 10.1021/acs.jpcb.3c04662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/15/2023] [Accepted: 10/06/2023] [Indexed: 11/01/2023]
Abstract
In this work, a theoretical-computational method is applied to study the deamidation reaction, a critical post-translational modification in proteins, using a simple model molecule in solution. The method allows one to comprehensively address the environmental effect, thereby enabling one to accurately derive the kinetic rate constants for the three main steps of the deamidation process. The results presented, in rather good agreement with the available experimental data, underline the necessity for a rigorous treatment of environmental factors and a precise kinetic model to correctly assess the overall kinetics of the deamidation reaction.
Collapse
Affiliation(s)
- Maria
Laura De Sciscio
- Department
of Chemistry, University of Rome, Sapienza, P.le A. Moro 5, 00185 Rome, Italy
| | | | - Fabio Centola
- Global
Analytical Development, Merck Serono S.p.A., 00012 Guidonia Montecelio, Italy
| | - Mara Rossi
- Global
Analytical Development, Merck Serono S.p.A., 00012 Guidonia Montecelio, Italy
| | - Enrico Guarnera
- Global
Analytical Development, Merck Serono S.p.A., 00012 Guidonia Montecelio, Italy
- Antibody
Discovery and Protein Engineering, Merck
Healthcare KGaA, 64293 Darmstadt, Germany
| | - Marco D’Abramo
- Department
of Chemistry, University of Rome, Sapienza, P.le A. Moro 5, 00185 Rome, Italy
| |
Collapse
|
16
|
Li S, Wang S, Fordjour E, Liang Y, Wang X, Ye Y, Bai Z, Yang Y, Chen Y. Development and characterization of anti-IL-5 monoclonal antibody Fab fragment for blocking IL-5/IL-5Rα binding. Int Immunopharmacol 2023; 124:111032. [PMID: 37832239 DOI: 10.1016/j.intimp.2023.111032] [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: 05/22/2023] [Revised: 10/03/2023] [Accepted: 10/04/2023] [Indexed: 10/15/2023]
Abstract
Interleukin-5 (IL-5) is a homodimeric cytokine that is a crucial regulator of the proliferation, activation, and maturation of eosinophils. Anti-IL-5 monoclonal antibodies, which block the binding of IL-5 to the IL-5 receptor subunit alpha (IL-5Rα), have been successfully used to treat eosinophilic (EOS) asthma. The currently marketed monoclonal antibody drugs require repeated injections for administration, which seriously affect patient compliance and high systemic exposure for injectable drug delivery. Here we successfully screened and developed the Fab (fragment of antigen binding), which is 1/3rd the molecular weight of IgG, favoring inhalation-mediated delivery to the lungs, making it more effective for asthma treatment. The 20A12-Fab-H12L3 can bind to IL-5 with a binding constant of 1.236E-09 M while significantly inhibiting the IL-5/IL-5Rα complex formation. We found that the light chain amino acids (S46 and F71) significantly affected the antibody expression during humanization. The 20A12-Fab-H12L3 significantly inhibited the proliferation of TF-1 cells and blocked the IL-5 binding to the IL-5Rα-overexpressing human embryonic kidney (HEK)-293 cells in vitro. Therefore, based on the mutant IL-5 binding with Fab, we explained why antibodies blocked IL-5 binding to IL-5Rα. Thus, this study provided a candidate pharmaceutical antibody for inhalation drug delivery.
Collapse
Affiliation(s)
- Shijie Li
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China; National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China; Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi 214122, China
| | - Shijie Wang
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China; National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China; Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi 214122, China
| | - Eric Fordjour
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China; National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China; Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi 214122, China
| | - Yaoji Liang
- Zhuhai Resproly Pharmaceutical Technology Co., Ltd, Zhuhai, 519040, Guangdong, China
| | - Xuelian Wang
- Zhuhai Resproly Pharmaceutical Technology Co., Ltd, Zhuhai, 519040, Guangdong, China
| | - Yonghao Ye
- Zhuhai Resproly Pharmaceutical Technology Co., Ltd, Zhuhai, 519040, Guangdong, China
| | - Zhonghu Bai
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China; National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China; Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi 214122, China.
| | - Yankun Yang
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China; National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China; Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi 214122, China.
| | - Yongqi Chen
- Zhuhai Resproly Pharmaceutical Technology Co., Ltd, Zhuhai, 519040, Guangdong, China.
| |
Collapse
|
17
|
Cao M, Hussmann GP, Tao Y, O’Connor E, Parthemore C, Zhang-Hulsey D, Liu D, Jiao Y, de Mel N, Prophet M, Korman S, Sonawane J, Grigoriadou C, Huang Y, Umlauf S, Chen X. Atypical Asparagine Deamidation of NW Motif Significantly Attenuates the Biological Activities of an Antibody Drug Conjugate. Antibodies (Basel) 2023; 12:68. [PMID: 37987246 PMCID: PMC10660493 DOI: 10.3390/antib12040068] [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: 09/07/2023] [Revised: 10/12/2023] [Accepted: 10/17/2023] [Indexed: 11/22/2023] Open
Abstract
Asparagine deamidation is a post-translational modification (PTM) that converts asparagine residues into iso-aspartate and/or aspartate. Non-enzymatic asparagine deamidation is observed frequently during the manufacturing, processing, and/or storage of biotherapeutic proteins. Depending on the site of deamidation, this PTM can significantly impact the therapeutic's potency, stability, and/or immunogenicity. Thus, deamidation is routinely monitored as a potential critical quality attribute. The initial evaluation of an asparagine's potential to deamidate begins with identifying sequence liabilities, in which the n + 1 amino acid is of particular interest. NW is one motif that occurs frequently within the complementarity-determining region (CDR) of therapeutic antibodies, but according to the published literature, has a very low risk of deamidating. Here we report an unusual case of this NW motif readily deamidating within the CDR of an antibody drug conjugate (ADC), which greatly impacts the ADC's biological activities. Furthermore, this NW motif solely deamidates into iso-aspartate, rather than the typical mixture of iso-aspartate and aspartate. Interestingly, biological activities are more severely impacted by the conversion of asparagine into iso-aspartate via deamidation than by conversion into aspartate via mutagenesis. Here, we detail the discovery of this unusual NW deamidation occurrence, characterize its impact on biological activities, and utilize structural data and modeling to explain why conversion to iso-aspartate is favored and impacts biological activities more severely.
Collapse
Affiliation(s)
- Mingyan Cao
- Department of Process and Analytical Sciences, Biopharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, One Medimmune Way, Gaithersburg, MD 20878, USA; (M.C.); (Y.J.); (N.d.M.); (C.G.)
| | - G. Patrick Hussmann
- Department of Process and Analytical Sciences, Biopharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, One Medimmune Way, Gaithersburg, MD 20878, USA; (M.C.); (Y.J.); (N.d.M.); (C.G.)
| | - Yeqing Tao
- Department of Process and Analytical Sciences, Biopharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, One Medimmune Way, Gaithersburg, MD 20878, USA; (M.C.); (Y.J.); (N.d.M.); (C.G.)
| | - Ellen O’Connor
- Department of Process and Analytical Sciences, Biopharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, One Medimmune Way, Gaithersburg, MD 20878, USA; (M.C.); (Y.J.); (N.d.M.); (C.G.)
| | - Conner Parthemore
- Department of Process and Analytical Sciences, Biopharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, One Medimmune Way, Gaithersburg, MD 20878, USA; (M.C.); (Y.J.); (N.d.M.); (C.G.)
| | - Diana Zhang-Hulsey
- Department of Process and Analytical Sciences, Biopharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, One Medimmune Way, Gaithersburg, MD 20878, USA; (M.C.); (Y.J.); (N.d.M.); (C.G.)
| | - Dengfeng Liu
- Department of Process and Analytical Sciences, Biopharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, One Medimmune Way, Gaithersburg, MD 20878, USA; (M.C.); (Y.J.); (N.d.M.); (C.G.)
| | - Yang Jiao
- Department of Process and Analytical Sciences, Biopharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, One Medimmune Way, Gaithersburg, MD 20878, USA; (M.C.); (Y.J.); (N.d.M.); (C.G.)
| | - Niluka de Mel
- Department of Process and Analytical Sciences, Biopharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, One Medimmune Way, Gaithersburg, MD 20878, USA; (M.C.); (Y.J.); (N.d.M.); (C.G.)
| | - Meagan Prophet
- Department of Process and Analytical Sciences, Biopharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, One Medimmune Way, Gaithersburg, MD 20878, USA; (M.C.); (Y.J.); (N.d.M.); (C.G.)
| | - Samuel Korman
- Department of Process and Analytical Sciences, Biopharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, One Medimmune Way, Gaithersburg, MD 20878, USA; (M.C.); (Y.J.); (N.d.M.); (C.G.)
| | - Jaytee Sonawane
- Department of Process and Analytical Sciences, Biopharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, One Medimmune Way, Gaithersburg, MD 20878, USA; (M.C.); (Y.J.); (N.d.M.); (C.G.)
| | - Christina Grigoriadou
- Department of Process and Analytical Sciences, Biopharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, One Medimmune Way, Gaithersburg, MD 20878, USA; (M.C.); (Y.J.); (N.d.M.); (C.G.)
| | - Yue Huang
- Department of Integrated Bioanalysis, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, 121 Oyster Point Boulevard, South San Francisco, CA 94080, USA
| | - Scott Umlauf
- Department of Process and Analytical Sciences, Biopharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, One Medimmune Way, Gaithersburg, MD 20878, USA; (M.C.); (Y.J.); (N.d.M.); (C.G.)
| | - Xiaoyu Chen
- Department of Process and Analytical Sciences, Biopharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, One Medimmune Way, Gaithersburg, MD 20878, USA; (M.C.); (Y.J.); (N.d.M.); (C.G.)
| |
Collapse
|
18
|
Huang Y, Yuan J, Mu R, Kubiak RJ, Ball K, Cao M, Hussmann GP, de Mel N, Liu D, Roskos LK, Liang M, Rosenbaum AI. Multiplex Bioanalytical Methods for Comprehensive Characterization and Quantification of the Unique Complementarity-Determining-Region Deamidation of MEDI7247, an Anti-ASCT2 Pyrrolobenzodiazepine Antibody-Drug Conjugate. Antibodies (Basel) 2023; 12:66. [PMID: 37873863 PMCID: PMC10594446 DOI: 10.3390/antib12040066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 09/23/2023] [Accepted: 09/29/2023] [Indexed: 10/25/2023] Open
Abstract
Deamidation, a common post-translational modification, may impact multiple physiochemical properties of a therapeutic protein. MEDI7247, a pyrrolobenzodiazepine (PBD) antibody-drug conjugate (ADC), contains a unique deamidation site, N102, located within the complementarity-determining region (CDR), impacting the affinity of MEDI7247 to its target. Therefore, it was necessary to monitor MEDI7247 deamidation status in vivo. Due to the low dose, a sensitive absolute quantification method using immunocapture coupled with liquid chromatography-tandem mass spectrometry (LBA-LC-MS/MS) was developed and qualified. We characterized the isomerization via Electron-Activated Dissociation (EAD), revealing that deamidation resulted in iso-aspartic acid. The absolute quantification of deamidation requires careful assay optimization in order not to perturb the balance of the deamidated and nondeamidated forms. Moreover, the selection of capture reagents essential for the correct quantitative assessment of deamidation was evaluated. The final assay was qualified with 50 ng/mL LLOQ for ADC for total and nondeamidated antibody quantification, with qualitative monitoring of the deamidated antibody. The impact of deamidation on the pharmacokinetic characteristics of MEDI7247 from clinical trial NCT03106428 was analyzed, revealing a gradual reduction in the nondeamidated form of MEDI7247 in vivo. Careful quantitative biotransformation analyses of complex biotherapeutic conjugates help us understand changes in product PTMs after administration, thus providing a more complete view of in vivo pharmacology.
Collapse
Affiliation(s)
- Yue Huang
- Integrated Bioanalysis, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, 121 Oyster Point Boulevard, South San Francisco, CA 94080, USA; (Y.H.); (J.Y.); (R.M.); (M.L.)
| | - Jiaqi Yuan
- Integrated Bioanalysis, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, 121 Oyster Point Boulevard, South San Francisco, CA 94080, USA; (Y.H.); (J.Y.); (R.M.); (M.L.)
| | - Ruipeng Mu
- Integrated Bioanalysis, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, 121 Oyster Point Boulevard, South San Francisco, CA 94080, USA; (Y.H.); (J.Y.); (R.M.); (M.L.)
| | - Robert J. Kubiak
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878, USA; (R.J.K.); (L.K.R.)
| | - Kathryn Ball
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Granta Park, Cambridge CB21 6GH, UK;
| | - Mingyan Cao
- Department of Analytical Sciences, Biopharmaceutical Development, R&D, AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878, USA; (M.C.); (G.P.H.); (N.d.M.); (D.L.)
| | - G. Patrick Hussmann
- Department of Analytical Sciences, Biopharmaceutical Development, R&D, AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878, USA; (M.C.); (G.P.H.); (N.d.M.); (D.L.)
| | - Niluka de Mel
- Department of Analytical Sciences, Biopharmaceutical Development, R&D, AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878, USA; (M.C.); (G.P.H.); (N.d.M.); (D.L.)
| | - Dengfeng Liu
- Department of Analytical Sciences, Biopharmaceutical Development, R&D, AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878, USA; (M.C.); (G.P.H.); (N.d.M.); (D.L.)
| | - Lorin K. Roskos
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878, USA; (R.J.K.); (L.K.R.)
| | - Meina Liang
- Integrated Bioanalysis, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, 121 Oyster Point Boulevard, South San Francisco, CA 94080, USA; (Y.H.); (J.Y.); (R.M.); (M.L.)
| | - Anton I. Rosenbaum
- Integrated Bioanalysis, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, 121 Oyster Point Boulevard, South San Francisco, CA 94080, USA; (Y.H.); (J.Y.); (R.M.); (M.L.)
| |
Collapse
|
19
|
Lippold S, Mistry K, Lenka S, Whang K, Liu P, Pitschi S, Kuhne F, Reusch D, Cadang L, Knaupp A, Izadi S, Dunkle A, Yang F, Schlothauer T. Function-structure approach reveals novel insights on the interplay of Immunoglobulin G 1 proteoforms and Fc gamma receptor IIa allotypes. Front Immunol 2023; 14:1260446. [PMID: 37790943 PMCID: PMC10544997 DOI: 10.3389/fimmu.2023.1260446] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 08/30/2023] [Indexed: 10/05/2023] Open
Abstract
Human Fc gamma receptor IIa (FcγRIIa) or CD32a has two major allotypes with a single amino acid difference at position 131 (histidine or arginine). Differences in FcγRIIa allotypes are known to impact immunological responses such as the clinical outcome of therapeutic monoclonal antibodies (mAbs). FcγRIIa is involved in antibody-dependent cellular phagocytosis (ADCP), which is an important contributor to the mechanism-of-action of mAbs by driving phagocytic clearance of cancer cells. Hence, understanding the impact of individual mAb proteoforms on the binding to FcγRIIa, and its different allotypes, is crucial for defining meaningful critical quality attributes (CQAs). Here, we report a function-structure based approach guided by novel FcγRIIa affinity chromatography-mass spectrometry (AC-MS) assays to assess individual IgG1 proteoforms. This allowed to unravel allotype-specific differences of IgG1 proteoforms on FcγRIIa binding. FcγRIIa AC-MS confirmed and refined structure-function relationships of IgG1 glycoform interactions. For example, the positive impact of afucosylation was higher than galactosylation for FcγRIIa Arg compared to FcγRIIa His. Moreover, we observed FcγRIIa allotype-opposing and IgG1 proteoform integrity-dependent differences in the binding response of stress-induced IgG1 proteoforms comprising asparagine 325 deamidation. The FcγRIIa-allotype dependent binding differences resolved by AC-MS were in line with functional ADCP-surrogate bioassay models. The molecular basis of the observed allotype specificity and proteoform selectivity upon asparagine 325 deamidation was elucidated using molecular dynamics. The observed differences were attributed to the contributions of an inter-molecular salt bridge between IgG1 and FcγRIIa Arg and the contribution of an intra-molecular hydrophobic pocket in IgG1. Our work highlights the unprecedented structural and functional resolution of AC-MS approaches along with predictive biological significance of observed affinity differences within relevant cell-based methods. This makes FcγRIIa AC-MS an invaluable tool to streamline the CQA assessment of therapeutic mAbs.
Collapse
Affiliation(s)
- Steffen Lippold
- Protein Analytical Chemistry, Genentech, A Member of the Roche Group, South San Francisco, CA, United States
| | - Karishma Mistry
- Biological Technologies, Genentech, A Member of the Roche Group, South San Francisco, CA, United States
| | - Sunidhi Lenka
- Pharmaceutical Development, Genentech, A Member of The Roche Group, South San Francisco, CA, United States
| | - Kevin Whang
- Biological Technologies, Genentech, A Member of the Roche Group, South San Francisco, CA, United States
| | - Peilu Liu
- Protein Analytical Chemistry, Genentech, A Member of the Roche Group, South San Francisco, CA, United States
| | - Sebastian Pitschi
- Pharma Technical Development Europe, Roche Diagnostics GmbH, Penzberg, Germany
| | - Felix Kuhne
- Pharma Technical Development Europe, Roche Diagnostics GmbH, Penzberg, Germany
| | - Dietmar Reusch
- Pharma Technical Development Europe, Roche Diagnostics GmbH, Penzberg, Germany
| | - Lance Cadang
- Protein Analytical Chemistry, Genentech, A Member of the Roche Group, South San Francisco, CA, United States
| | - Alexander Knaupp
- Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Saeed Izadi
- Pharmaceutical Development, Genentech, A Member of The Roche Group, South San Francisco, CA, United States
| | - Alexis Dunkle
- Biological Technologies, Genentech, A Member of the Roche Group, South San Francisco, CA, United States
| | - Feng Yang
- Protein Analytical Chemistry, Genentech, A Member of the Roche Group, South San Francisco, CA, United States
| | - Tilman Schlothauer
- Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| |
Collapse
|
20
|
Beaudoin CA, Petsolari E, Hamaia SW, Hala S, Alofi FS, Pandurangan AP, Blundell TL, Chaitanya Vedithi S, Huang CLH, Jackson AP. SARS-CoV-2 Omicron subvariant spike N405 unlikely to rapidly deamidate. Biochem Biophys Res Commun 2023; 666:61-67. [PMID: 37178506 PMCID: PMC10152834 DOI: 10.1016/j.bbrc.2023.04.088] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023]
Abstract
The RGD motif on the SARS-CoV-2 spike protein has been suggested to interact with RGD-binding integrins αVβ3 and α5β1 to enhance viral cell entry and alter downstream signaling cascades. The D405N mutation on the Omicron subvariant spike proteins, resulting in an RGN motif, has recently been shown to inhibit binding to integrin αVβ3. Deamidation of asparagines in protein ligand RGN motifs has been demonstrated to generate RGD and RGisoD motifs that permit binding to RGD-binding integrins. Two asparagines, N481 and N501, on the Wild-type spike receptor-binding domain have been previously shown to have deamidation half-lives of 16.5 and 123 days, respectively, which may occur during the viral life cycle. Deamidation of Omicron subvariant N405 may recover the ability to interact with RGD-binding integrins. Thus, herein, all-atom molecular dynamics simulations of the Wild-type and Omicron subvariant spike protein receptor-binding domains were conducted to investigate the potential for asparagines, the Omicron subvariant N405 in particular, to assume the optimized geometry for deamidation to occur. In summary, the Omicron subvariant N405 was primarily found to be stabilized in a state unfavourable for deamidation after hydrogen bonding with downstream E406. Nevertheless, a small number of RGD or RGisoD motifs on the Omicron subvariant spike proteins may restore the ability to interact with RGD-binding integrins. The simulations also provided structural clarification regarding the deamidation rates of Wild-type N481 and N501 and highlighted the utility of tertiary structure dynamics information in predicting asparagine deamidation. Further work is needed to characterize the effects of deamidation on spike-integrin interactions.
Collapse
Affiliation(s)
- Christopher A Beaudoin
- Department of Biochemistry, Hopkins Building, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QW, United Kingdom.
| | - Emmanouela Petsolari
- Department of Biochemistry, Sanger Building, University of Cambridge, Tennis Court Road, Cambridge, CB2 1GA, United Kingdom
| | - Samir W Hamaia
- Department of Biochemistry, Hopkins Building, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QW, United Kingdom
| | - Sharif Hala
- Infectious Disease Research Department, King Abdullah International Medical Research Centre, Ministry of National Guard Health Affairs, Jeddah, Saudi Arabia; King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Jeddah, Saudi Arabia
| | - Fadwa S Alofi
- Infectious Disease Research Department, King Abdullah International Medical Research Centre, Ministry of National Guard Health Affairs, Jeddah, Saudi Arabia
| | - Arun P Pandurangan
- Heart and Lung Research Institute, University of Cambridge, Papworth Road, Cambridge, CB2 0BB, United Kingdom
| | - Tom L Blundell
- Heart and Lung Research Institute, University of Cambridge, Papworth Road, Cambridge, CB2 0BB, United Kingdom
| | - Sundeep Chaitanya Vedithi
- Heart and Lung Research Institute, University of Cambridge, Papworth Road, Cambridge, CB2 0BB, United Kingdom
| | - Christopher L-H Huang
- Department of Biochemistry, Hopkins Building, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QW, United Kingdom; Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom
| | - Antony P Jackson
- Department of Biochemistry, Hopkins Building, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QW, United Kingdom.
| |
Collapse
|
21
|
Creutznacher R, Schulze-Niemand E, König P, Stanojlovic V, Mallagaray A, Peters T, Stein M, Schubert M. Conformational Control of Fast Asparagine Deamidation in a Norovirus Capsid Protein. Biochemistry 2023; 62:1032-1043. [PMID: 36808948 PMCID: PMC9996831 DOI: 10.1021/acs.biochem.2c00656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Accelerated spontaneous deamidation of asparagine 373 and subsequent conversion into an isoaspartate has been shown to attenuate the binding of histo blood group antigens (HBGAs) to the protruding domain (P-domain) of the capsid protein of a prevalent norovirus strain (GII.4). Here, we link an unusual backbone conformation of asparagine 373 to its fast site-specific deamidation. NMR spectroscopy and ion exchange chromatography have been used to monitor the deamidation reaction of P-domains of two closely related GII.4 norovirus strains, specific point mutants, and control peptides. MD simulations over several microseconds have been instrumental to rationalize the experimental findings. While conventional descriptors such as available surface area, root-mean-square fluctuations, or nucleophilic attack distance fail as explanations, the population of a rare syn-backbone conformation distinguishes asparagine 373 from all other asparagine residues. We suggest that stabilization of this unusual conformation enhances the nucleophilicity of the backbone nitrogen of aspartate 374, in turn accelerating the deamidation of asparagine 373. This finding should be relevant to the development of reliable prediction algorithms for sites of rapid asparagine deamidation in proteins.
Collapse
Affiliation(s)
- Robert Creutznacher
- Institute of Chemistry and Metabolomics, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Eric Schulze-Niemand
- Molecular Simulations and Design Group, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstrasse 1, 39106 Magdeburg, Germany
| | - Patrick König
- Institute of Chemistry and Metabolomics, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Vesna Stanojlovic
- Department of Biosciences and Medical Biology, University of Salzburg, Hellbrunnerstrasse 34, 5020 Salzburg, Austria
| | - Alvaro Mallagaray
- Institute of Chemistry and Metabolomics, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Thomas Peters
- Institute of Chemistry and Metabolomics, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Matthias Stein
- Molecular Simulations and Design Group, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstrasse 1, 39106 Magdeburg, Germany
| | - Mario Schubert
- Department of Biosciences and Medical Biology, University of Salzburg, Hellbrunnerstrasse 34, 5020 Salzburg, Austria
| |
Collapse
|
22
|
Spanov B, Olaleye O, Mesurado T, Govorukhina N, Jungbauer A, van de Merbel NC, Lingg N, Bischoff R. Pertuzumab Charge Variant Analysis and Complementarity-Determining Region Stability Assessment to Deamidation. Anal Chem 2023; 95:3951-3958. [PMID: 36795375 PMCID: PMC9979147 DOI: 10.1021/acs.analchem.2c03275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Pertuzumab is a monoclonal antibody used for the treatment of HER2-positive breast cancer in combination with trastuzumab. Charge variants of trastuzumab have been extensively described in the literature; however, little is known about the charge heterogeneity of pertuzumab. Here, changes in the ion-exchange profile of pertuzumab were evaluated by pH gradient cation-exchange chromatography after stressing it for up to 3 weeks at physiological and elevated pH and 37 °C. Isolated charge variants arising under stress conditions were characterized by peptide mapping. The results of peptide mapping showed that deamidation in the Fc domain and N-terminal pyroglutamate formation in the heavy chain are the main contributors to charge heterogeneity. The heavy chain CDR2, which is the only CDR containing asparagine residues, was quite resistant to deamidation under stress conditions according to peptide mapping results. Using surface plasmon resonance, it was shown that the affinity of pertuzumab for the HER2 target receptor does not change under stress conditions. Peptide mapping analysis of clinical samples showed an average of 2-3% deamidation in the heavy chain CDR2, 20-25% deamidation in the Fc domain, and 10-15% N-terminal pyroglutamate formation in the heavy chain. These findings suggest that in vitro stress studies are able to predict in vivo modifications.
Collapse
Affiliation(s)
- Baubek Spanov
- Department
of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, A Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Oladapo Olaleye
- Department
of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, A Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Tomés Mesurado
- Department
of Biotechnology, Institute of Bioprocess Science and Engineering, University of Natural Resources and Life Sciences,
Vienna, Muthgasse 18, Vienna 1190, Austria
| | - Natalia Govorukhina
- Department
of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, A Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Alois Jungbauer
- Department
of Biotechnology, Institute of Bioprocess Science and Engineering, University of Natural Resources and Life Sciences,
Vienna, Muthgasse 18, Vienna 1190, Austria
| | - Nico C. van de Merbel
- Department
of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, A Deusinglaan 1, 9713 AV Groningen, The Netherlands,Bioanalytical
Laboratory, ICON, Amerikaweg 18, 9407 TK Assen, The Netherlands
| | - Nico Lingg
- Department
of Biotechnology, Institute of Bioprocess Science and Engineering, University of Natural Resources and Life Sciences,
Vienna, Muthgasse 18, Vienna 1190, Austria
| | - Rainer Bischoff
- Department
of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, A Deusinglaan 1, 9713 AV Groningen, The Netherlands,
| |
Collapse
|
23
|
Cain P, Huang L, Tang Y, Anguiano V, Feng Y. Impact of IgG subclass on monoclonal antibody developability. MAbs 2023; 15:2191302. [PMID: 36945111 PMCID: PMC10038059 DOI: 10.1080/19420862.2023.2191302] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023] Open
Abstract
IgG-based monoclonal antibody therapeutics, which are mainly IgG1, IgG2, and IgG4 subclasses or related variants, have dominated the biotherapeutics field for decades. Multiple laboratories have reported that the IgG subclasses possess different molecular characteristics that can affect their developability. For example, IgG1, the most popular IgG subclass for therapeutics, is known to have a characteristic degradation pathway related to its hinge fragility. However, there remains a paucity of studies that systematically evaluate the IgG subclasses on manufacturability and long-term stability. We thus conducted a systematic study of 12 mAbs derived from three sets of unrelated variable regions, each cloned into IgG1, an IgG1 variant with diminished effector functions, IgG2, and a stabilized IgG4 variant with further reduced FcγR interaction, to evaluate the impact of IgG subclass on manufacturability and high concentration stability in a common formulation buffer matrix. Our evaluation included Chinese hamster ovary cell productivity, host cell protein removal efficiency, N-linked glycan structure at the conserved N297 Fc position, solution appearance at high concentration, and aggregate growth, fragmentation, charge variant profile change, and post-translational modification upon thermal stress conditions or long-term storage at refrigerated temperature. Our results elucidated molecular attributes that are common to all IgG subclasses, as well as those that are unique to certain Fc domains, providing new insight into the effects of IgG subclass on antibody manufacturability and stability. These learnings can be used to enable a balanced decision on IgG subclass selection for therapeutic antibodies and aid in acceleration of their product development process.
Collapse
Affiliation(s)
- Paul Cain
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Technology Center North, Indianapolis, IN, USA
| | - Lihua Huang
- Bioproduct Research & Development, Lilly Research Laboratories, Lilly Technology Center North, Indianapolis, IN, USA
| | - Yu Tang
- Pharmaceutical Development and Manufacturing, Syndax Pharmaceuticals, Waltham, MA, USA
| | - Victor Anguiano
- Bioproduct Research & Development, Lilly Research Laboratories, Lilly Technology Center North, Indianapolis, IN, USA
| | - Yiqing Feng
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Technology Center North, Indianapolis, IN, USA
| |
Collapse
|
24
|
Thorsteinson N, Comeau SR, Kumar S. Structure-Based Optimization of Antibody-Based Biotherapeutics for Improved Developability: A Practical Guide for Molecular Modelers. Methods Mol Biol 2023; 2552:219-235. [PMID: 36346594 DOI: 10.1007/978-1-0716-2609-2_11] [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] [Indexed: 06/16/2023]
Abstract
A great effort to avoid known developability risks is now more often being made earlier during the lead candidate discovery and optimization phase of biotherapeutic drug development. Predictive computational strategies, used in the early stages of antibody discovery and development, to mitigate the risk of late-stage failure of antibody candidates, are highly valuable. Various structure-based methods exist for accurately predicting properties critical to developability, and, in this chapter, we discuss the history of their development and demonstrate how they can be used to filter large sets of candidates arising from target affinity screening and to optimize lead candidates for developability. Methods for modeling antibody structures from sequence and detecting post-translational modifications and chemical degradation liabilities are also discussed.
Collapse
Affiliation(s)
- Nels Thorsteinson
- Scientific Services Manager, Biologics, Chemical Computing Group ULC, Montreal, QC, Canada
| | - Stephen R Comeau
- Computational Biochemistry and Bioinformatics Group, Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| | - Sandeep Kumar
- Computational Biochemistry and Bioinformatics Group, Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA.
| |
Collapse
|
25
|
Dippel A, Gallegos A, Aleti V, Barnes A, Chen X, Christian E, Delmar J, Du Q, Esfandiary R, Farmer E, Garcia A, Li Q, Lin J, Liu W, Machiesky L, Mody N, Parupudi A, Prophet M, Rickert K, Rosenthal K, Ren S, Shandilya H, Varkey R, Wons K, Wu Y, Loo YM, Esser MT, Kallewaard NL, Rajan S, Damschroder M, Xu W, Kaplan G. Developability profiling of a panel of Fc engineered SARS-CoV-2 neutralizing antibodies. MAbs 2023; 15:2152526. [PMID: 36476037 PMCID: PMC9733695 DOI: 10.1080/19420862.2022.2152526] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
To combat the COVID-19 pandemic, potential therapies have been developed and moved into clinical trials at an unprecedented pace. Some of the most promising therapies are neutralizing antibodies against SARS-CoV-2. In order to maximize the therapeutic effectiveness of such neutralizing antibodies, Fc engineering to modulate effector functions and to extend half-life is desirable. However, it is critical that Fc engineering does not negatively impact the developability properties of the antibodies, as these properties play a key role in ensuring rapid development, successful manufacturing, and improved overall chances of clinical success. In this study, we describe the biophysical characterization of a panel of Fc engineered ("TM-YTE") SARS-CoV-2 neutralizing antibodies, the same Fc modifications as those found in AstraZeneca's Evusheld (AZD7442; tixagevimab and cilgavimab), in which the TM modification (L234F/L235E/P331S) reduce binding to FcγR and C1q and the YTE modification (M252Y/S254T/T256E) extends serum half-life. We have previously shown that combining both the TM and YTE Fc modifications can reduce the thermal stability of the CH2 domain and possibly lead to developability challenges. Here we show, using a diverse panel of TM-YTE SARS-CoV-2 neutralizing antibodies, that despite lowering the thermal stability of the Fc CH2 domain, the TM-YTE platform does not have any inherent developability liabilities and shows an in vivo pharmacokinetic profile in human FcRn transgenic mice similar to the well-characterized YTE platform. The TM-YTE is therefore a developable, effector function reduced, half-life extended antibody platform.
Collapse
Affiliation(s)
- Andrew Dippel
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Austin Gallegos
- Biopharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Vineela Aleti
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Arnita Barnes
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Xiaoru Chen
- Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | - Jared Delmar
- Biopharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Qun Du
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Reza Esfandiary
- Biopharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Erika Farmer
- Biopharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Andrew Garcia
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Qing Li
- Hansoh Bio, Rockville, MD, USA,Biologics Engineering, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Jia Lin
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Weiyi Liu
- Pfizer, La Jolla, CA, USA,Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - LeeAnn Machiesky
- Biopharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Neil Mody
- Biopharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Arun Parupudi
- Biopharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Meagan Prophet
- Biopharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Keith Rickert
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Kim Rosenthal
- Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Song Ren
- Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | - Reena Varkey
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Kevin Wons
- Biopharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Yuling Wu
- Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Yueh-Ming Loo
- Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Mark T. Esser
- Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Nicole L. Kallewaard
- Eli Lilly, Indianapolis, IN, USA,Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Sarav Rajan
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | - Weichen Xu
- Biopharmaceutical Development, MacroGenics, Rockville, MD, USA,Biopharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Gilad Kaplan
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, MD, USA,CONTACT Gilad Kaplan AstraZeneca, Gaithersburg, MD20878
| |
Collapse
|
26
|
Sarin D, Kumar S, Rathore AS. Multiattribute Monitoring of Charge-Based Heterogeneity of Recombinant Monoclonal Antibodies Using 2D HIC-WCX-MS. Anal Chem 2022; 94:15018-15026. [PMID: 36260865 DOI: 10.1021/acs.analchem.2c02931] [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
Charged heterogeneity of monoclonal antibody (mAb) products is regarded as a critical quality attribute (CQA) depending on its impact on the safety and efficacy profile of the product. Hence, manufacturers are expected to perform a comprehensive characterization of the charge heterogeneity to ensure that the manufactured product meets its specifications. Further, monitoring is also expected during the product lifecycle to demonstrate consistency in product quality. However, conventional analytical methods for characterization of hydrophobic and charge variants are nonvolatile salt-based and require manual fraction collection and desalting steps before analysis through mass spectrometry can be performed. In the present study, a workflow of a two-dimensional liquid chromatography method using mass spectrometry (MS)-compatible buffers coupled with native mass spectrometry was performed to characterize hydrophobic variants in the first dimension and charge variants in the second dimension without any need for manual fractionation. This novel two-dimensional (2D) hydrophobic interaction chromatography (HIC)-weak cation-exchange chromatography (WCX)-MS workflow identified 10 variants in mAb A, out of which 2 variants are exclusive to the 2D orthogonal method. Similarly, for mAb B, a total of 11 variants are identified, including 5 variants exclusive to the 2D orthogonal workflow. When compared to stand-alone, HIC resolved only 4 variants for both mAbs and WCX resolved 7 variants for mAb A and 6 variants for mAb B. In addition, the proposed method allows direct characterization of hydrophobic/charge variant peaks through native mass spectrometry in a single-run workflow.
Collapse
Affiliation(s)
- Deepika Sarin
- Department of Chemical Engineering, IIT Delhi, Hauz Khas, New Delhi, Delhi 110016, India
| | - Sunil Kumar
- Department of Chemical Engineering, IIT Delhi, Hauz Khas, New Delhi, Delhi 110016, India
| | - Anurag S Rathore
- Department of Chemical Engineering, IIT Delhi, Hauz Khas, New Delhi, Delhi 110016, India
| |
Collapse
|
27
|
An insight on lipid nanoparticles for therapeutic proteins delivery. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
28
|
Suresh SA, Ethiraj S, Rajnish KN. A systematic review of recent trends in research on therapeutically significant L-asparaginase and acute lymphoblastic leukemia. Mol Biol Rep 2022; 49:11281-11287. [PMID: 35816224 DOI: 10.1007/s11033-022-07688-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/08/2022] [Indexed: 12/01/2022]
Abstract
L-asparaginases are mostly obtained from bacterial sources for their application in the therapy and food industry. Bacterial L-asparaginases are employed in the treatment of Acute Lymphoblastic Leukemia (ALL) and its subtypes, a type of blood and bone marrow cancer that results in the overproduction of immature blood cells. It also plays a role in the food industry in reducing the acrylamide formed during baking, roasting, and frying starchy foods. This importance of the enzyme makes it to be of constant interest to the researchers to isolate novel sources. Presently L-asparaginases from E. coli native and PEGylated form, Dickeya chrysanthemi (Erwinia chrysanthemi) are in the treatment regime. In therapy, the intrinsic glutaminase activity of the enzyme is a major drawback as the patients in treatment experience side effects like fever, skin rashes, anaphylaxis, pancreatitis, steatosis in the liver, and many complications. Its significance in the food industry in mitigating acrylamide is also a major reason. Acrylamide, a potent carcinogen was formed when treating starchy foods at higher temperatures. Acrylamide content in food was analyzed and pre-treatment was considered a valuable option. Immobilization of the enzyme is an advancing and promising technique in the effective delivery of the enzyme than in free form. The concept of machine learning by employing the Artificial Network and Genetic Algorithm has paved the way to optimize the production of L-asparaginase from its sources. Gene-editing tools are gaining momentum in the study of several diseases and this review focuses on the CRISPR-Cas9 gene-editing tool in ALL.
Collapse
Affiliation(s)
| | | | - K N Rajnish
- SRM Institute of Science and Technology, Chennai, Tamil Nadu, India.
| |
Collapse
|
29
|
Tong Q, Liu H, Qi Q, Dai C, Yang T, Qian F. Development of a fully human anti-GITR antibody with potent antitumor activity using H2L2 mice. FEBS Open Bio 2022; 12:1542-1557. [PMID: 35674216 PMCID: PMC9340783 DOI: 10.1002/2211-5463.13451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/17/2022] [Accepted: 06/07/2022] [Indexed: 11/28/2022] Open
Abstract
Glucocorticoid‐induced TNF receptor‐related (GITR) can act as a co‐stimulatory receptor, representing a potential target for safely enhancing immunotherapy efficacy. GITR is triggered by a GITR ligand or an agonist antibody and activates CD8+ and CD4+ effector T cells, reducing tumor‐infiltrating Treg numbers and resulting in activation of immune responses and tumor cell destruction by effector T cells. GITR is an attractive target for immunotherapy, especially in combination therapy with immune checkpoint inhibitors, as is being explored in clinical trials. Using H2L2 transgenic mice encoding the human immunoglobulin variable region and hybridoma technology, we generated a panel of fully human antibodies that showed excellent specific affinity and strong activation of human T cells. After conversion to fully human antibodies and engineering modification, we obtained an anti‐GITR antibody hab019e2 with enhanced antitumor activity in a B‐hGITR MC38 mouse model compared to Tab9H6V3, an anti‐GITR antibody that activates T cells and inhibits Treg suppression from XenoMouse. As a fully human antibody with its posttranslational modification hot spot removed, the hab019e2 antibody exerted more potent therapeutic effects, and may have potential as a novel and developable antibody targeting GITR for follow‐up drug studies.
Collapse
Affiliation(s)
- Qiuli Tong
- Shanghai Public Health Clinical Center, Human Phenome Institute and School of Life Sciences, Fudan University, Shanghai, China.,Shanghai Chempartner Co., Ltd, China
| | - Hu Liu
- Shanghai Chempartner Co., Ltd, China
| | | | | | | | - Feng Qian
- Shanghai Public Health Clinical Center, Human Phenome Institute and School of Life Sciences, Fudan University, Shanghai, China
| |
Collapse
|
30
|
Liu AP, Yan Y, Wang S, Li N. Coupling Anion Exchange Chromatography with Native Mass Spectrometry for Charge Heterogeneity Characterization of Monoclonal Antibodies. Anal Chem 2022; 94:6355-6362. [PMID: 35420790 PMCID: PMC9047409 DOI: 10.1021/acs.analchem.2c00707] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Despite the recent success of coupling anion exchange chromatography with native mass spectrometry (AEX-MS) to study anionic proteins, the utility of AEX-MS methods in therapeutic monoclonal antibody (mAb) characterization has been limited. In this work, we developed and optimized a salt gradient-based AEX-MS method and explored its utility in charge variant analysis of therapeutic mAbs. We demonstrated that, although the developed AEX-MS method is less useful for IgG1 molecules that have higher isoelectric points (pIs), it is an attractive alternative for charge variant analysis of IgG4 molecules. By elevating the column temperature and lowering the mAb pI through PNGase F-mediated deglycosylation, the chromatographical resolution from AEX separation can be significantly improved. We also demonstrated that, after PNGase F and IdeS digestion, the AEX-MS method exhibited excellent resolving power for multiple attributes in the IgG4 Fc region, including unprocessed C-terminal Lys, N-glycosylation occupancy, and several conserved Fc deamidations, making it ideally suited for multiple attribute monitoring (MAM). Through fractionation and peptide mapping analysis, we also demonstrated that the developed AEX-MS method can provide site-specific and isoform-resolved separation of Fc deamidation products, allowing rapid and artifact-free quantitation of these modifications without performing bottom-up analysis.
Collapse
Affiliation(s)
- Anita P Liu
- Analytical Chemistry Group, Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Yuetian Yan
- Analytical Chemistry Group, Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Shunhai Wang
- Analytical Chemistry Group, Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Ning Li
- Analytical Chemistry Group, Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| |
Collapse
|
31
|
Ogata Y, Quizon PM, Nightlinger NS, Sitasuwan P, Snodgrass C, Lee LA, Meyer JD, Rogers RS. Automated multi-attribute method sample preparation using high-throughput buffer exchange tips. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2022; 36:e9222. [PMID: 34783086 PMCID: PMC9286584 DOI: 10.1002/rcm.9222] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/02/2021] [Accepted: 11/07/2021] [Indexed: 05/23/2023]
Abstract
RATIONALE The multi-attribute method (MAM) has become a valuable mass spectrometry (MS)-based tool that can identify and quantify the site-specific product attributes and purity information for biotherapeutics such as monoclonal antibodies (mAbs) and fusion molecules in recent years. As we expand the use of the MAM at various stages of drug development, it is critical to enhance the sample preparation throughput without additional chemical modifications and variability. METHODS In this study, a fully automated MAM sample preparation protocol is presented, where rapid desalting in less than 15 minutes is achieved using miniaturized size-exclusion chromatography columns in pipette tips on an automated liquid handler. The peptide samples were analyzed using an electrospray ionization (ESI) orbitrap mass spectrometer coupled to an ultra-high-performance liquid chromatography (UHPLC) system with a dual column switching system. RESULTS No significant change was observed in product attributes and their quantities compared with manual, low-artifact sample preparation. The sample recovery using the buffer exchange tips was greatly enhanced over the manual spin cartridges while still demonstrating excellent reproducibility for a wide variety of starting sample concentrations. Unlike a plate desalting system, the individual columns provide flexibility in the number of samples prepared at a time and sample locations within plates. CONCLUSIONS This automated protocol enables the preparation of up to 96 samples with less "at-bench" time than the manual preparation of a smaller batch of samples, thereby greatly facilitating support of process development and the use of the MAM in quality control.
Collapse
Affiliation(s)
| | | | | | - Pongkwan Sitasuwan
- Integrated Micro‐Chromatography Systems (IMCS), IrmoSCUSA
- 3M CompanySt. PaulMNUSA
| | - Casey Snodgrass
- Hamilton CompanyRenoNVUSA
- Mammoth BiosciencesSan FranciscoCAUSA
| | - L. Andrew Lee
- Integrated Micro‐Chromatography Systems (IMCS), IrmoSCUSA
| | | | | |
Collapse
|
32
|
Abstract
Monoclonal antibodies are susceptible to chemical and enzymatic modifications during manufacturing, storage, and shipping. Deamidation, isomerization, and oxidation can compromise the potency, efficacy, and safety of therapeutic antibodies. Recently, in silico tools have been used to identify liable residues and engineer antibodies with better chemical stability. Computational approaches for predicting deamidation, isomerization, oxidation, glycation, carbonylation, sulfation, and hydroxylation are reviewed here. Although liable motifs have been used to improve the chemical stability of antibodies, the accuracy of in silico predictions can be improved using machine learning and molecular dynamic simulations. In addition, there are opportunities to improve predictions for specific stress conditions, develop in silico prediction of novel modifications in antibodies, and predict the impact of modifications on physical stability and antigen-binding.
Collapse
Affiliation(s)
- Shabdita Vatsa
- Development Services, Lonza Biologics, Singapore, Singapore
| |
Collapse
|
33
|
de Graaf SC, Hoek M, Tamara S, Heck AJR. A perspective toward mass spectrometry-based de novo sequencing of endogenous antibodies. MAbs 2022; 14:2079449. [PMID: 35699511 PMCID: PMC9225641 DOI: 10.1080/19420862.2022.2079449] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
A key step in therapeutic and endogenous humoral antibody characterization is identifying the amino acid sequence. So far, this task has been mainly tackled through sequencing of B-cell receptor (BCR) repertoires at the nucleotide level. Mass spectrometry (MS) has emerged as an alternative tool for obtaining sequence information directly at the – most relevant – protein level. Although several MS methods are now well established, analysis of recombinant and endogenous antibodies comes with a specific set of challenges, requiring approaches beyond the conventional proteomics workflows. Here, we review the challenges in MS-based sequencing of both recombinant as well as endogenous humoral antibodies and outline state-of-the-art methods attempting to overcome these obstacles. We highlight recent examples and discuss remaining challenges. We foresee a great future for these approaches making de novo antibody sequencing and discovery by MS-based techniques feasible, even for complex clinical samples from endogenous sources such as serum and other liquid biopsies.
Collapse
Affiliation(s)
- Sebastiaan C de Graaf
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands.,Netherlands Proteomics Center, Utrecht, Netherlands
| | - Max Hoek
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands.,Netherlands Proteomics Center, Utrecht, Netherlands
| | - Sem Tamara
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands.,Netherlands Proteomics Center, Utrecht, Netherlands
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands.,Netherlands Proteomics Center, Utrecht, Netherlands
| |
Collapse
|
34
|
Gupta S, Jiskoot W, Schöneich C, Rathore AS. Oxidation and Deamidation of Monoclonal Antibody Products: Potential Impact on Stability, Biological Activity, and Efficacy. J Pharm Sci 2021; 111:903-918. [PMID: 34890632 DOI: 10.1016/j.xphs.2021.11.024] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/24/2021] [Accepted: 11/26/2021] [Indexed: 12/25/2022]
Abstract
The role in human health of therapeutic proteins in general, and monoclonal antibodies (mAbs) in particular, has been significant and is continuously evolving. A considerable amount of time and resources are invested first in mAb product development and then in clinical examination of the product. Physical and chemical degradation can occur during manufacturing, processing, storage, handling, and administration. Therapeutic proteins may undergo various chemical degradation processes, including oxidation, deamidation, isomerization, hydrolysis, deglycosylation, racemization, disulfide bond breakage and formation, Maillard reaction, and β-elimination. Oxidation and deamidation are the most common chemical degradation processes of mAbs, which may result in changes in physical properties, such as hydrophobicity, charge, secondary or/and tertiary structure, and may lower the thermodynamic or kinetic barrier to unfold. This may predispose the product to aggregation and other chemical modifications, which can alter the binding affinity, half-life, and efficacy of the product. This review summarizes major findings from the past decade on the impact of oxidation and deamidation on the stability, biological activity, and efficacy of mAb products. Mechanisms of action, influencing factors, characterization tools, clinical impact, and risk mitigation strategies have been addressed.
Collapse
Affiliation(s)
- Surbhi Gupta
- Department of Chemical Engineering, Indian Institute of Technology Delhi, New Delhi-110016, India
| | - Wim Jiskoot
- Division of BioTherapeutics, Leiden Centre for Drug Research (LACDR), Leiden University, Leiden, the Netherlands
| | | | - Anurag S Rathore
- Department of Chemical Engineering, Indian Institute of Technology Delhi, New Delhi-110016, India.
| |
Collapse
|
35
|
Boudier-Lemosquet A, Mahler A, Bobo C, Dufossée M, Priault M. Introducing protein deamidation: Landmark discoveries, societal outreach, and tentative priming workflow to address deamidation. Methods 2021; 200:3-14. [PMID: 34843979 DOI: 10.1016/j.ymeth.2021.11.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 11/15/2021] [Accepted: 11/18/2021] [Indexed: 12/17/2022] Open
Abstract
Our current knowledge on protein deamidation results from a journey that started almost 100 years ago, when a handful of researchers first described the non-enzymatic "desamidation" of glutamine, and the effect of different anions on the catalytic rate of the reaction. Since then, the field has tremendously expended and now finds outreach in very diverse areas. In light of all the recent articles published in these areas, it seemed timely to propose an integrated review on the subject, including a short historical overview of the landmark discoveries in the field, highlighting the current global positioning of protein deamidation in biology and non-biology fields, and concluding with a workflow for those asking if a protein can deamidate, and identify the residues involved. This review is essentially intended to provide newcomers in the field with an overview of how deamidation has penetrated our society and what tools are currently at hand to identify and quantify protein deamidation.
Collapse
Affiliation(s)
| | - Adrien Mahler
- Univ. Bordeaux, CNRS, IBGC, UMR 5095, F-33000 Bordeaux, France
| | - Claude Bobo
- Univ. Bordeaux, CNRS, IBGC, UMR 5095, F-33000 Bordeaux, France
| | - Mélody Dufossée
- Univ. Bordeaux, CNRS, IBGC, UMR 5095, F-33000 Bordeaux, France
| | - Muriel Priault
- Univ. Bordeaux, CNRS, IBGC, UMR 5095, F-33000 Bordeaux, France.
| |
Collapse
|
36
|
The Impact of Glycerol on an Affibody Conformation and Its Correlation to Chemical Degradation. Pharmaceutics 2021; 13:pharmaceutics13111853. [PMID: 34834267 PMCID: PMC8618440 DOI: 10.3390/pharmaceutics13111853] [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] [Received: 09/20/2021] [Revised: 10/27/2021] [Accepted: 10/31/2021] [Indexed: 11/21/2022] Open
Abstract
The addition of glycerol to protein solutions is often used to hinder the aggregation and denaturation of proteins. However, it is not a generalised practice against chemical degradation reactions. The chemical degradation of proteins, such as deamidation and isomerisation, is an important deteriorative mechanism that leads to a loss of functionality of pharmaceutical proteins. Here, the influence of glycerol on the chemical degradation of a protein and its correlation to glycerol-induced conformational changes is presented. The time-dependent chemical degradation of a pharmaceutical protein, GA-Z, in the absence and presence of glycerol was investigated in a stability study. The effect of glycerol on protein conformation and oligomerisation was characterised using asymmetric field-flow fractionation and small-angle neutron scattering in a wide glycerol concentration range of 0–90% v/v. The results from the stability study were connected to the observed glycerol-induced conformational changes in the protein. A correlation between protein conformation and the protective effect of glycerol against the degradation reactions deamidation, isomerisation, and hydrolysis was found. The study reveals that glycerol induces conformational changes of the protein, which favour a more compact and chemically stable state. It is also shown that the conformation can be changed by other system properties, e.g., protein concentration, leading to increased chemical stability.
Collapse
|
37
|
Lorenzo R, Defelipe LA, Aliperti L, Niebling S, Custódio TF, Löw C, Schwarz JJ, Remans K, Craig PO, Otero LH, Klinke S, García-Alai M, Sánchez IE, Alonso LG. Deamidation drives molecular aging of the SARS-CoV-2 spike protein receptor-binding motif. J Biol Chem 2021; 297:101175. [PMID: 34499924 PMCID: PMC8421091 DOI: 10.1016/j.jbc.2021.101175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/03/2021] [Accepted: 09/03/2021] [Indexed: 01/22/2023] Open
Abstract
The spike protein is the main protein component of the SARS-CoV-2 virion surface. The spike receptor-binding motif mediates recognition of the human angiotensin-converting enzyme 2 receptor, a critical step in infection, and is the preferential target for spike-neutralizing antibodies. Posttranslational modifications of the spike receptor-binding motif have been shown to modulate viral infectivity and host immune response, but these modifications are still being explored. Here we studied asparagine deamidation of the spike protein, a spontaneous event that leads to the appearance of aspartic and isoaspartic residues, which affect both the protein backbone and its charge. We used computational prediction and biochemical experiments to identify five deamidation hotspots in the SARS-CoV-2 spike protein. Asparagine residues 481 and 501 in the receptor-binding motif deamidate with a half-life of 16.5 and 123 days at 37 °C, respectively. Deamidation is significantly slowed at 4 °C, indicating a strong dependence of spike protein molecular aging on environmental conditions. Deamidation of the spike receptor-binding motif decreases the equilibrium constant for binding to the human angiotensin-converting enzyme 2 receptor more than 3.5-fold, yet its high conservation pattern suggests some positive effect on viral fitness. We propose a model for deamidation of the full SARS-CoV-2 virion illustrating how deamidation of the spike receptor-binding motif could lead to the accumulation on the virion surface of a nonnegligible chemically diverse spike population in a timescale of days. Our findings provide a potential mechanism for molecular aging of the spike protein with significant consequences for understanding virus infectivity and vaccine development.
Collapse
Affiliation(s)
- Ramiro Lorenzo
- Centro de Investigación Veterinaria de Tandil (CIVETAN), CONICET-CICPBA-UNCPBA, Facultad de Ciencias Veterinarias, Universidad Nacional del Centro (FCV-UNCPBA), Tandil, Argentina
| | - Lucas A Defelipe
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany
| | - Lucio Aliperti
- Laboratorio de Fisiología de Proteínas, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Stephan Niebling
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany; Centre for Structural Systems Biology, Hamburg, Germany
| | - Tânia F Custódio
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany; Centre for Structural Systems Biology, Hamburg, Germany
| | - Christian Löw
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany; Centre for Structural Systems Biology, Hamburg, Germany
| | | | - Kim Remans
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Patricio O Craig
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Lisandro H Otero
- Fundación Instituto Leloir, IIBBA-CONICET, and Plataforma Argentina de Biología Estructural y Metabolómica PLABEM, Buenos Aires, Argentina
| | - Sebastián Klinke
- Fundación Instituto Leloir, IIBBA-CONICET, and Plataforma Argentina de Biología Estructural y Metabolómica PLABEM, Buenos Aires, Argentina
| | - María García-Alai
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany; Centre for Structural Systems Biology, Hamburg, Germany
| | - Ignacio E Sánchez
- Laboratorio de Fisiología de Proteínas, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Leonardo G Alonso
- Instituto de Nanobiotecnologıa (NANOBIOTEC), UBA-CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
38
|
Trends in deamidation across archaeological bones, ceramics and dental calculus. Methods 2021; 200:67-79. [PMID: 34450289 DOI: 10.1016/j.ymeth.2021.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 06/04/2021] [Accepted: 08/14/2021] [Indexed: 11/23/2022] Open
Abstract
The accumulation of post-translational modifications (PTMs) in proteins throughout the lifecycle has been studied for decades, particularly more so with the advent of soft-ionization mass spectrometry-based proteomic techniques. However, particular PTMs, such as the deamidations of asparagine and glutamine residues, continue to accumulate in proteins that remain into the forensic, archaeological, and palaeontological records. The accurate measurement of these ancient 'molecular timers' has been proposed as a method to not only differentiate between exogenous and endogenous proteins within complex mixtures (i.e., contamination), but also as a method of providing relative age estimations into geological time. In this study we explored the extent to which deamidation varies with chronological age across different proteins in bones, as well as investigated differences between proteins across dental calculus and archaeological ceramics. We also analysed the relationships between the observed extent of deamidation and the protein primary structure. We found that collagen obtained from archaeological bones showed a chronological dependence on the extent of deamidation observed, but only when they were from similar environments, supporting prior suggestions about 'thermal age' being a major influence on the deamidation observed. Our study on non-collagenous proteins (NCPs) in archaeological bones showed that while biglycan, and to a lesser extent chondroadherin, showed positive correlations between geological age and the extent of deamidation, others including fetuin-A and serum albumin did not. However, despite the well-known dependence of deamidation on the three-dimensional structure of the peptides, we were unable to find any clear correlation between the structural motifs of the peptides in archaeological bones and the extent of deamidation observed. Our analysis of a set of food proteins obtained from Neolithic archaeological ceramics in Çatalhöyük also showed similar deamidation levels irrespective of the protein structure. Overall, our results suggest that deamidation in archaeological samples could be useful for obtaining additional information beyond identification of species and tissue type, be that as a measure of protein endogeneity and potential contamination, or a measure of protein degradation, or as an indicator of thermal age and for relative dating; however, further research needs to be undertaken to understand why particular proteins are better for this than others, going beyond simple consideration of their secondary structure.
Collapse
|
39
|
Nakayoshi T, Wanita K, Kato K, Kurimoto E, Oda A. Computational analysis of nonenzymatic deamidation of asparagine residues catalysed by acetic acid. Mol Phys 2021. [DOI: 10.1080/00268976.2020.1827176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Tomoki Nakayoshi
- Graduate School of Pharmacy, Meijo University, Nagoya, Japan
- Institute of Medical, Pharmaceutical and Health Science, Kanazawa University, Kanazawa, Japan
| | - Kota Wanita
- Graduate School of Pharmacy, Meijo University, Nagoya, Japan
| | - Koichi Kato
- Graduate School of Pharmacy, Meijo University, Nagoya, Japan
- Department of Pharmacy, Kinjo Gakuin University, Nagoya, Japan
| | - Eiji Kurimoto
- Graduate School of Pharmacy, Meijo University, Nagoya, Japan
| | - Akifumi Oda
- Graduate School of Pharmacy, Meijo University, Nagoya, Japan
- Institute of Medical, Pharmaceutical and Health Science, Kanazawa University, Kanazawa, Japan
- Institute for Protein Research, Suita, Japan
| |
Collapse
|
40
|
Song YE, Dubois H, Hoffmann M, D́Eri S, Fromentin Y, Wiesner J, Pfenninger A, Clavier S, Pieper A, Duhau L, Roth U. Automated mass spectrometry multi-attribute method analyses for process development and characterization of mAbs. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1166:122540. [DOI: 10.1016/j.jchromb.2021.122540] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 10/22/2022]
|
41
|
Kuang J, Tao Y, Song Y, Chemmalil L, Mussa N, Ding J, Li ZJ. Understanding the pathway and kinetics of aspartic acid isomerization in peptide mapping methods for monoclonal antibodies. Anal Bioanal Chem 2021; 413:2113-2123. [PMID: 33543314 DOI: 10.1007/s00216-021-03176-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/28/2020] [Accepted: 01/13/2021] [Indexed: 11/26/2022]
Abstract
Isomerization of aspartic acid (Asp) in therapeutic proteins could lead to safety and efficacy concerns. Thus, accurate quantitation of various Asp isomerization along with kinetic understanding of the variant formations is needed to ensure optimal process development and sufficient product quality control. In this study, we first observed Asp-succinimide conversion in complementarity-determining regions (CDRs) Asp-Gly motif of a recombinant mAb through ion exchange chromatography, intact protein analysis by mass spectrometry, and LC-MS/MS. Then, we developed a specific peptide mapping method, with optimized sample digestion conditions, to accurately quantitate Asp-succinimide-isoAsp variants at peptide level without method-induced isomerization. Various kinetics of Asp-succinimide-isoAsp isomerization pathways were elucidated using 18O labeling followed by LC-MS analysis. Molecular modeling and molecular dynamic simulation provide additional insight on the kinetics of Asp-succinimide formation and stability of succinimide intermediate. Findings of this work shed light on the molecular construct and the kinetics of the formation of isoAsp and succinimide in peptides and proteins, which facilitates analytical method development, protein engineering, and late phase development for commercialization of therapeutic proteins.
Collapse
Affiliation(s)
- June Kuang
- Biologics Development Organization, Bristol-Myers Squibb Company, Devens, MA, 01434, USA
| | - Yuanqi Tao
- Biologics Development Organization, Bristol-Myers Squibb Company, Devens, MA, 01434, USA
- Analytical Science Biologics, Takeda Pharmaceutical Company, Lexington, MA, 02421, USA
| | - Yuanli Song
- Biologics Development Organization, Bristol-Myers Squibb Company, Devens, MA, 01434, USA
- Process Development & Manufacture Operations, GSK, MA, 02451, Waltham, USA
| | - Letha Chemmalil
- Biologics Development Organization, Bristol-Myers Squibb Company, Devens, MA, 01434, USA
| | - Nesredin Mussa
- Biologics Development Organization, Bristol-Myers Squibb Company, Devens, MA, 01434, USA
- Ultragenyx, CA, 94005, Brisbane, USA
| | - Julia Ding
- Biologics Development Organization, Bristol-Myers Squibb Company, Devens, MA, 01434, USA.
| | - Zheng Jian Li
- Biologics Development Organization, Bristol-Myers Squibb Company, Devens, MA, 01434, USA
| |
Collapse
|
42
|
Zhu HJ, Liu D, Tran VP, Wu Z, Jiang K, Zhu H, Zhang J, Gibbons C, Xue B, Shi H, Wang PG. N-Linked Glycosylation Prevents Deamidation of Glycopeptide and Glycoprotein. ACS Chem Biol 2020; 15:3197-3205. [PMID: 33270417 DOI: 10.1021/acschembio.0c00734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Deamidation has been recognized as a common spontaneous pathway of protein degradation and a prevalent concern in the pharmaceutical industry; deamidation caused the reduction of protein/peptide drug efficacy and shelf life in several cases. More importantly, deamidation of physiological proteins is related to several human diseases and considered a "timer" for the diseases. N-linked glycosylation has a variety of significant biological functions, and it interestingly occurs right on the deamidation site-asparagine. It has been perceived that N-glycosylation could prevent deamidation, but experimental support is still lacking for clearly understanding the role of N-glycosylation on deamidation. Our results presented that deamidation is prevented by naturally occurring N-linked glycosylation. Glycopeptides and corresponding nonglycosylated peptides were used to compare their deamidation rates. All the nonglycosylated peptides have different half-lives ranging from one to 20 days, for the corresponding glycosylated peptides; all the results showed that the deamidation reaction was significantly reduced by the introduction of N-linked glycosylation. A glycoprotein, RNase B, also showed a significantly elongated deamidation half-life compared to nonglycosylated protein RNase A. At last, N-linked glycosylation on INGAP-P, a therapeutic peptide, increased the deamidation half-life of INGAP-P as well as its therapeutic potency.
Collapse
Affiliation(s)
- Hailiang Joshua Zhu
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
| | - Ding Liu
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
| | - Vy P. Tran
- Department of Biology, Georgia State University, Atlanta, Georgia 30303, United States
| | - Zhigang Wu
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
| | - Kuan Jiang
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
| | - He Zhu
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
| | - Jiabin Zhang
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
| | - Christopher Gibbons
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
| | - Bingzhong Xue
- Department of Biology, Georgia State University, Atlanta, Georgia 30303, United States
| | - Hang Shi
- Department of Biology, Georgia State University, Atlanta, Georgia 30303, United States
| | - Peng George Wang
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
| |
Collapse
|
43
|
Carrara SC, Ulitzka M, Grzeschik J, Kornmann H, Hock B, Kolmar H. From cell line development to the formulated drug product: The art of manufacturing therapeutic monoclonal antibodies. Int J Pharm 2020; 594:120164. [PMID: 33309833 DOI: 10.1016/j.ijpharm.2020.120164] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/23/2020] [Accepted: 12/06/2020] [Indexed: 02/07/2023]
Abstract
Therapeutic monoclonal antibodies and related products have steadily grown to become the dominant product class within the biopharmaceutical market. Production of antibodies requires special precautions to ensure safety and efficacy of the product. In particular, minimizing antibody product heterogeneity is crucial as drug substance variants may impair the activity, efficacy, safety, and pharmacokinetic properties of an antibody, consequently resulting in the failure of a product in pre-clinical and clinical development. This review will cover the manufacturing and formulation challenges and advances of therapeutic monoclonal antibodies, focusing on improved processes to minimize variants and ensure batch-to-batch consistency. Processes put in place by regulatory agencies, such as Quality-by-Design (QbD) and current Good Manufacturing Practices (cGMP), and how their implementation has aided drug development in pharmaceutical companies will be reviewed. Advances in formulation and considerations on the intended use of a therapeutic antibody, including the route of administration and patient compliance, will be discussed.
Collapse
Affiliation(s)
- Stefania C Carrara
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany; Ferring Darmstadt Laboratory, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| | - Michael Ulitzka
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany; Ferring Darmstadt Laboratory, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| | - Julius Grzeschik
- Ferring Darmstadt Laboratory, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| | - Henri Kornmann
- Ferring International Center SA, CH-1162 Saint-Prex, Switzerland
| | - Björn Hock
- Ferring International Center SA, CH-1162 Saint-Prex, Switzerland.
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany.
| |
Collapse
|
44
|
Kamerzell TJ, Middaugh CR. Prediction Machines: Applied Machine Learning for Therapeutic Protein Design and Development. J Pharm Sci 2020; 110:665-681. [PMID: 33278409 DOI: 10.1016/j.xphs.2020.11.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/27/2020] [Accepted: 11/27/2020] [Indexed: 12/11/2022]
Abstract
The rapid growth in technological advances and quantity of scientific data over the past decade has led to several challenges including data storage and analysis. Accurate models of complex datasets were previously difficult to develop and interpret. However, improvements in machine learning algorithms have since enabled unparalleled classification and prediction capabilities. The application of machine learning can be seen throughout diverse industries due to their ease of use and interpretability. In this review, we describe popular machine learning algorithms and highlight their application in pharmaceutical protein development. Machine learning models have now been applied to better understand the nonlinear concentration dependent viscosity of protein solutions, predict protein oxidation and deamidation rates, classify sub-visible particles and compare the physical stability of proteins. We also applied several machine learning algorithms using previously published data and describe models with improved predictions and classification. The authors hope that this review can be used as a resource to others and encourage continued application of machine learning algorithms to problems in pharmaceutical protein development.
Collapse
Affiliation(s)
- Tim J Kamerzell
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS, USA; Division of Internal Medicine, HCA MidWest Health, Overland Park, KS, USA.
| | - C Russell Middaugh
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS, USA
| |
Collapse
|
45
|
Butreddy A, Janga KY, Ajjarapu S, Sarabu S, Dudhipala N. Instability of therapeutic proteins - An overview of stresses, stabilization mechanisms and analytical techniques involved in lyophilized proteins. Int J Biol Macromol 2020; 167:309-325. [PMID: 33275971 DOI: 10.1016/j.ijbiomac.2020.11.188] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 01/06/2023]
Abstract
Solid-state is the preferred choice for storage of protein therapeutics to improve stability and preserve the biological activity by decreasing the physical and chemical degradation associated with liquid protein formulations. Lyophilization or freeze-drying is an effective drying method to overcome the instability problems of proteins. However, the processing steps (freezing, primary drying and secondary drying) involved in the lyophilization process can expose the proteins to various stress and harsh conditions, leading to denaturation, aggregation often a loss in activity of protein therapeutics. Stabilizers such as sugars and surfactants are often added to protect the proteins against physical stress associated with lyophilization process and storage conditions. Another way to curtail the degradation of proteins due to process related stress is by modification of the lyophilization process. Slow freezing, high nucleation temperature, decreasing the extent of supercooling, and annealing can minimize the formation of the interface (ice-water) by producing large ice crystals with less surface area, thereby preserving the native structure and stability of the proteins. Hence, a thorough understanding of formulation composition, lyophilization process parameters and the choice of analytical methods to characterize and monitor the protein instability is crucial for development of stable therapeutic protein products. This review provides an overview of various stress conditions that proteins might encounter during lyophilization process, mechanisms to improve the stability and analytical techniques to tackle the proteins instability during both freeze-drying and storage.
Collapse
Affiliation(s)
- Arun Butreddy
- Formulation R&D, Biological E. Limited, IKP Knowledge Park, Shameerpet, Hyderabad, Telangana State 500078, India; Laboratory of Nanotechnology, University College of Pharmaceutical Sciences, Kakatiya University, Warangal, Telangana State 506009, India
| | - Karthik Yadav Janga
- Laboratory of Nanotechnology, University College of Pharmaceutical Sciences, Kakatiya University, Warangal, Telangana State 506009, India
| | - Srinivas Ajjarapu
- Industrial Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Pilani 333031, India
| | - Sandeep Sarabu
- Laboratory of Nanotechnology, University College of Pharmaceutical Sciences, Kakatiya University, Warangal, Telangana State 506009, India
| | - Narendar Dudhipala
- Laboratory of Nanotechnology, University College of Pharmaceutical Sciences, Kakatiya University, Warangal, Telangana State 506009, India; Department of Pharmaceutics, Vaagdevi College of Pharmacy, Warangal, Telangana State 506 005, India..
| |
Collapse
|
46
|
Lorenzo JR, Leonetti CO, Alonso LG, Sánchez IE. NGOME-Lite: Proteome-wide prediction of spontaneous protein deamidation highlights differences between taxa. Methods 2020; 200:15-22. [PMID: 33189829 DOI: 10.1016/j.ymeth.2020.11.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/01/2020] [Accepted: 11/10/2020] [Indexed: 12/31/2022] Open
Abstract
Asparagines in proteins deamidate spontaneously, which changes the chemical structure of a protein and often affects its function. Current prediction algorithms for asparagine deamidation require a structure as an input or are too slow to be applied at a proteomic scale. We present NGOME-Lite, a new version of our sequence-based predictor for spontaneous asparagine deamidation that is faster by over two orders of magnitude at a similar degree of accuracy. The algorithm takes into account intrinsic sequence propensities and slowing down of deamidation by local structure. NGOME-Lite can run in a proteomic analysis mode that provides the half-time of the intact form of each protein, predicted by taking into account sequence propensities and structural protection or sequence propensities only, and a structure protection factor. The detailed analysis mode also provides graphical output for all Asn residues in the query sequence. We applied NGOME-Lite to over 257,000 sequences in 38 proteomes and found that different taxa differ in their predicted deamidation dynamics. Spontaneous protein deamidation is faster in Eukarya than in Bacteria because of a higher degree of structural protection in the latter. Predicted protein deamidation half-lifes correlate with protein turnover in human, mouse, rat, C. elegans and budding yeast but not in two plants and two bacteria. NGOME-Lite is implemented in a docker container available at https://ngome.proteinphysiologylab.org.
Collapse
Affiliation(s)
- Juan R Lorenzo
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Laboratorio de Fisiología de Proteínas, Buenos Aires, Argentina; Centro de Investigación Veterinaria de Tandil (CIVETAN), CONICET-CICPBA-UNCPBA, Facultad de Ciencias Veterinarias, Universidad Nacional del Centro (FCV-UNCPBA), Tandil, Argentina
| | - César O Leonetti
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Laboratorio de Fisiología de Proteínas, Buenos Aires, Argentina
| | - Leonardo G Alonso
- Instituto de Nanobiotecnología (NANOBIOTEC), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina.
| | - Ignacio E Sánchez
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Laboratorio de Fisiología de Proteínas, Buenos Aires, Argentina.
| |
Collapse
|
47
|
Kato K, Nakayoshi T, Kurimoto E, Oda A. Mechanisms of Deamidation of Asparagine Residues and Effects of Main-Chain Conformation on Activation Energy. Int J Mol Sci 2020; 21:ijms21197035. [PMID: 32987875 PMCID: PMC7582646 DOI: 10.3390/ijms21197035] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/19/2020] [Accepted: 09/22/2020] [Indexed: 12/14/2022] Open
Abstract
Deamidation of asparagine (Asn) residues is a nonenzymatic post-translational modification of proteins. Asn deamidation is associated with pathogenesis of age-related diseases and hypofunction of monoclonal antibodies. Deamidation rate is known to be affected by the residue following Asn on the carboxyl side and by secondary structure. Information about main-chain conformation of Asn residues is necessary to accurately predict deamidation rate. In this study, the effect of main-chain conformation of Asn residues on deamidation rate was computationally investigated using molecular dynamics (MD) simulations and quantum chemical calculations. The results of MD simulations for γS-crystallin suggested that frequently deamidated Asn residues have common main-chain conformations on the N-terminal side. Based on the simulated structure, initial structures for the quantum chemical calculations were constructed and optimized geometries were obtained using the B3LYP density functional method. Structures that were frequently deamidated had a lower activation energy barrier than that of the little deamidated structure. We also showed that dihydrogen phosphate and bicarbonate ions are important catalysts for deamidation of Asn residues.
Collapse
Affiliation(s)
- Koichi Kato
- College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori, Moriyama-ku, Nagoya, Aichi 463-8521, Japan
- Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya, Aichi 468-8503, Japan; (T.N.); (E.K.); (A.O.)
- Correspondence: ; Tel.: +81-527-980-180
| | - Tomoki Nakayoshi
- Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya, Aichi 468-8503, Japan; (T.N.); (E.K.); (A.O.)
| | - Eiji Kurimoto
- Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya, Aichi 468-8503, Japan; (T.N.); (E.K.); (A.O.)
| | - Akifumi Oda
- Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya, Aichi 468-8503, Japan; (T.N.); (E.K.); (A.O.)
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
48
|
Pham NB, Meng WS. Protein aggregation and immunogenicity of biotherapeutics. Int J Pharm 2020; 585:119523. [PMID: 32531452 DOI: 10.1016/j.ijpharm.2020.119523] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/01/2020] [Accepted: 06/06/2020] [Indexed: 12/19/2022]
Abstract
Recombinant proteins are the mainstay of biopharmaceuticals. A key challenge in the manufacturing and formulation of protein biologic products is the tendency for the active pharmaceutical ingredients to aggregate, resulting in irreversible drug loss, and an increase in immunogenicity risk. While the molecular mechanisms of protein aggregation have been discussed extensively in the literature, knowledge gaps remain in connecting the phenomenon in the context of immunogenicity of biotherapeutics. In this review, we discussed factors that drive aggregation of pharmaceutical recombinant proteins, and highlighted methods of prediction and mitigation that can be deployed through the development stages, from formulation to bioproduction. The purpose is to stimulate new dialogs that would bridge the interface between physical characterizations of protein aggregates in biotherapeutics and the functional attributes of the immune system.
Collapse
Affiliation(s)
- Ngoc B Pham
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, United States
| | - Wilson S Meng
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA 15219, United States.
| |
Collapse
|
49
|
Kuroda D, Tsumoto K. Engineering Stability, Viscosity, and Immunogenicity of Antibodies by Computational Design. J Pharm Sci 2020; 109:1631-1651. [DOI: 10.1016/j.xphs.2020.01.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/25/2019] [Accepted: 01/10/2020] [Indexed: 12/18/2022]
|
50
|
Characterization of IgG1 Fc Deamidation at Asparagine 325 and Its Impact on Antibody-dependent Cell-mediated Cytotoxicity and FcγRIIIa Binding. Sci Rep 2020; 10:383. [PMID: 31941950 PMCID: PMC6962426 DOI: 10.1038/s41598-019-57184-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 12/21/2019] [Indexed: 02/06/2023] Open
Abstract
Antibody-dependent cell-mediated cytotoxicity (ADCC) is an important mechanism of action for many therapeutic antibodies. A therapeutic immunoglobulin (Ig) G1 monoclonal antibody lost more than half of its ADCC activity after heat stress at 40 °C for 4 months. Size-exclusion and ion-exchange chromatography were used to fractionate various size and charge variants from the stressed IgG1. Physicochemical characterization of these fractions revealed that a rarely seen crystallizable fragment (Fc) modification, N325 deamidation, exhibited a positive correlation with the loss of ADCC activity. A further surface plasmon resonance study showed that this modification disrupted the binding between the IgG1 Fc and Fcγ receptor IIIa, resulting in decreased ADCC activity of the IgG1 antibody. Mutants of N325/D and N325/Q were made to confirm the effect of N325 deamidation on ADCC. We hypothesize that N325 deamidation altered the local three-dimensional structure, which might interfere with the binding and interaction with the effector cell. Because of its impact on biological activity, N325 deamidation is a critical quality attribute for products whose mechanism of action includes ADCC. A thorough understanding of the criticality of N325 deamidation and appropriate monitoring can help ensure the safety and efficacy of IgG1 or Fc-fusion products.
Collapse
|