1
|
Rossmueller G, Mirkina I, Thiele M, Puchol Tarazona A, Rueker F, Kerschbaumer RJ, Schinagl A. Integrating In Silico and In Vitro Tools for Optimized Antibody Development-Design of Therapeutic Anti-oxMIF Antibodies. Antibodies (Basel) 2024; 13:104. [PMID: 39727487 DOI: 10.3390/antib13040104] [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: 11/12/2024] [Revised: 12/06/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Rigorous assessment of antibody developability is crucial for optimizing lead candidates before progressing to clinical studies. Recent advances in predictive tools for protein structures, surface properties, stability, and immunogenicity have streamlined the development of new biologics. However, accurate prediction of the impact of single amino acid substitutions on antibody structures remains challenging, due to the diversity of complementarity-determining regions (CDRs), particularly CDR3s. METHODS In this study, we combined in silico tools with in vitro assessments to engineer improved antibodies against the oxidized isoform of the macrophage migration inhibitory factor (oxMIF), building on the first generation anti-oxMIF antibody imalumab. RESULTS We identified hydrophobic hotspots conferring increased self-interaction and aggregation propensity on imalumab, which unravels its unusually short half-life in humans. By introducing mutations into the variable regions, we addressed these liabilities. Structural prediction tools and molecular dynamics simulations guided the selection of mutations, which were then experimentally validated. The lead candidate antibody, C0083, demonstrated reduced hydrophobicity and self-interaction due to the restructuring of its heavy chain CDR3 loop. Despite these structural changes, C0083 retained target specificity and binding affinity to oxMIF. CONCLUSIONS Altogether, this study shows that a small number of well-selected mutations was sufficient to substantially improve the biophysicochemical properties of imalumab.
Collapse
Affiliation(s)
- Gregor Rossmueller
- OncoOne Research & Development GmbH, Karl-Farkas-Gasse 22, A-1030 Vienna, Austria
| | - Irina Mirkina
- OncoOne Research & Development GmbH, Karl-Farkas-Gasse 22, A-1030 Vienna, Austria
| | - Michael Thiele
- OncoOne Research & Development GmbH, Karl-Farkas-Gasse 22, A-1030 Vienna, Austria
| | | | - Florian Rueker
- Department of Biotechnology, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| | | | - Alexander Schinagl
- OncoOne Research & Development GmbH, Karl-Farkas-Gasse 22, A-1030 Vienna, Austria
| |
Collapse
|
2
|
Wu IE, Kalejaye L, Lai PK. Machine Learning Models for Predicting Monoclonal Antibody Biophysical Properties from Molecular Dynamics Simulations and Deep Learning-Based Surface Descriptors. Mol Pharm 2024. [PMID: 39606945 DOI: 10.1021/acs.molpharmaceut.4c00804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Monoclonal antibodies (mAbs) have found extensive applications and development in treating various diseases. From the pharmaceutical industry's perspective, the journey from the design and development of mAbs to clinical testing and large-scale production is a highly time-consuming and resource-intensive process. During the research and development phase, assessing and optimizing the developability of mAbs is of paramount importance to ensure their success as candidates for therapeutic drugs. The critical factors influencing mAb development are their biophysical properties, such as aggregation propensity, solubility, and viscosity. This study utilized a data set comprising 12 biophysical properties of 137 antibodies from a previous study (Proc Natl Acad Sci USA. 114(5):944-949, 2017). We employed full-length antibody molecular dynamics simulations and machine learning techniques to predict experimental data for these 12 biophysical properties. Additionally, we utilized a newly developed deep learning model called DeepSP, which directly predicts the dynamical and structural properties of spatial aggregation propensity and spatial charge map in different antibody regions from sequences. Our research findings indicate that the machine learning models we developed outperform previous methods in predicting most biophysical properties. Furthermore, the DeepSP model yields similar predictive results compared to molecular dynamic simulations while significantly reducing computational time. The code and parameters are freely available at https://github.com/Lailabcode/AbDev. Also, the webapp, AbDev, for 12 biophysical properties prediction has been developed and provided at https://devpred.onrender.com/AbDev.
Collapse
Affiliation(s)
- I-En Wu
- Department of Chemical Engineering and Materials Science, Stevens Institute of Technology, Hoboken 07030 New Jersey
| | - Lateefat Kalejaye
- Department of Chemical Engineering and Materials Science, Stevens Institute of Technology, Hoboken 07030 New Jersey
| | - Pin-Kuang Lai
- Department of Chemical Engineering and Materials Science, Stevens Institute of Technology, Hoboken 07030 New Jersey
| |
Collapse
|
3
|
Sheehan K, Jeon H, Corr SC, Hayes JM, Mok KH. Antibody Aggregation: A Problem Within the Biopharmaceutical Industry and Its Role in AL Amyloidosis Disease. Protein J 2024:10.1007/s10930-024-10237-6. [PMID: 39527351 DOI: 10.1007/s10930-024-10237-6] [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] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Due to the large size and rapid growth of the global therapeutic antibody market, there is major interest in understanding the aggregation of protein products as it can compromise efficacy, concentration, and safety. Various production and storage conditions have been identified as capable of inducing aggregation of polyclonal and monoclonal antibody (mAb) therapies such as low pH, freezing, light exposure, lyophilisation and increased ionic strength. The addition of stabilising excipients to these therapeutics helps to combat the formation of aggregates with future aggregation inhibition mechanisms involving the introduction of point mutations and glycoengineering within aggregation prone regions (APRs). Antibody aggregation also plays an integral role in the pathogenesis of a condition known as amyloid light chain (AL) amyloidosis which is characterised by the production of improperly folded and amyloidogenic immunoglobulin light chains (LCs). Current diagnostic tools rely heavily on histological staining with their future moving towards amyloid component identification and proteomic analysis. For many years, treatment options designed for multiple myeloma (MM) have been applied to AL amyloidosis patients by depleting plasma cell numbers. More recently, treatment strategies more specific to this condition have been developed with many designed to recognize amyloid fibrils and trigger their degradation without causing systemic plasma cell cytotoxicity. Amyloid fibrils in AL disease and aggregates in antibody therapeutics are both formed through the oligomerisation of misfolded / modified proteins attempting to reach a thermodynamically stable, free energy minimum that is lower than the respective monomers themselves. Although the final morphologies are different, by understanding the principles underlying such aggregation, we expect to find common insights that may contribute to the development of new and effective methods of antibody aggregation and/or amyloidosis management. We envision that this area of research will continue to be very relevant in both industry and clinical settings.
Collapse
Affiliation(s)
- Kate Sheehan
- Trinity Biomedical Sciences Institute (TBSI), School of Biochemistry & Immunology, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland
- School of Genetics & Microbiology, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland
| | - Hyesoo Jeon
- Trinity Biomedical Sciences Institute (TBSI), School of Biochemistry & Immunology, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland
- Lonza Biologics Tuas Pte. Ltd., 35 Tuas South Ave 6, Singapore, 637377, Republic of Singapore
| | - Sinéad C Corr
- School of Genetics & Microbiology, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland
- School of Microbiology and APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Jerrard M Hayes
- Trinity Biomedical Sciences Institute (TBSI), School of Biochemistry & Immunology, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland
| | - K H Mok
- Trinity Biomedical Sciences Institute (TBSI), School of Biochemistry & Immunology, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland.
- Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Trinity College Dublin, The University of Dublin, Dublin 2, Ireland.
| |
Collapse
|
4
|
Wang Y, Williams HD, Dikicioglu D, Dalby PA. Predictive Model Building for Aggregation Kinetics Based on Molecular Dynamics Simulations of an Antibody Fragment. Mol Pharm 2024; 21:5827-5841. [PMID: 39348223 PMCID: PMC11539058 DOI: 10.1021/acs.molpharmaceut.4c00859] [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: 07/31/2024] [Revised: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 10/02/2024]
Abstract
Computational methods including machine learning and molecular dynamics simulations have strong potential to characterize, understand, and ultimately predict the properties of proteins relevant to their stability and function as therapeutics. Such methods would streamline the development pathway by minimizing the current experimental testing required for many protein variants and formulations. The molecular understanding of thermostability and aggregation propensity has advanced significantly along with predictive algorithms based on the sequence-level or structural-level information on a protein. However, these approaches focus largely on a comparison of protein sequence variations to correlate the properties of proteins to their stability, solubility, and aggregation propensity. For therapeutic protein development, it is of equal importance to take into account the impact of the formulation conditions to elucidate and predict the stability of the antibody drugs. At the macroscopic level, changing temperature, pH, ionic strength, and the addition of excipients can significantly alter the kinetics of protein aggregation. The mechanisms controlling aggregation kinetics have been traced back to a combination of molecular features, including conformational stability, partial unfolding to aggregation-prone states, and the colloidal stability governed by surface charges and hydrophobicity. However, very little has been done to evaluate these features in the context of protein dynamics in different formulations. In this work, we have combined a range of molecular features calculated from the Fab A33 protein sequence and molecular dynamics simulations. Using the power of advanced, yet interpretable, statistical tools, it has been possible to uncover greater insights into the mechanisms behind protein stability, validating previous findings, and also develop models that can predict the aggregation kinetics within a range of 49 different solution conditions.
Collapse
Affiliation(s)
- Yuhan Wang
- Department
of Biochemical Engineering, University College
London, London WC1E 6BT, U.K.
| | - Hywel D. Williams
- Biopharmaceutical
Product Development, CSL Ltd., 45 Poplar Road, Parkville 3052, Australia
| | - Duygu Dikicioglu
- Department
of Biochemical Engineering, University College
London, London WC1E 6BT, U.K.
| | - Paul A. Dalby
- Department
of Biochemical Engineering, University College
London, London WC1E 6BT, U.K.
| |
Collapse
|
5
|
Barragan-Galvez JC, Hernandez-Flores A, Lopez-Ortega O, Rodriguez-Alvarez AA, Maravillas-Montero JL, Ortiz-Navarrete V. The constant domain of CRTAM is essential for high-affinity interaction with Nectin-like 2. Biochem Biophys Rep 2024; 39:101813. [PMID: 39263316 PMCID: PMC11388666 DOI: 10.1016/j.bbrep.2024.101813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024] Open
Abstract
CRTAM (Class-I MHC restricted T cell-associated molecule) is a member of the Nectin-like family, composed of two extracellular domains, one constant domain (IgC) and another variable domain (IgV), expressed in activated CD8 T cells, epithelial cells, natural killer (NK) cells, and in a subpopulation of CD4 T cells. CRTAM recognizes the ligand Nectin-like 2 (Necl2) through the IgV domain. However, the role of the IgC domain during this ligand recognition has yet to be understood. In this study, we show the purification of soluble-folded Ig domains of CRTAM, and we demonstrate that the IgC domain forms a homodimer in solution via hydrophobic interactions. By surface plasmon resonance (SPR) analysis, we also demonstrate that CRTAM binds to Necl2 with an affinity of 2.16 nM. In conclusion, CRTAM's IgC is essential for a high-affinity interaction with Necl-2.
Collapse
Affiliation(s)
- Juan Carlos Barragan-Galvez
- Department of Molecular Biomedicine, Center for Research and Advanced Studies (CINVESTAV), Mexico City, Mexico
- Departamento de Farmacia, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Guanajuato, 36200, Mexico
| | | | - Orestes Lopez-Ortega
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, 75015, Paris, France
| | | | - Jose Luis Maravillas-Montero
- Research Support Network, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", Mexico City, Mexico
| | - Vianney Ortiz-Navarrete
- Department of Molecular Biomedicine, Center for Research and Advanced Studies (CINVESTAV), Mexico City, Mexico
| |
Collapse
|
6
|
Ingavat N, Dzulkiflie N, Liew JM, Wang X, Leong E, Loh HP, Ng SK, Yang Y, Zhang W. Investigation on environmental factors contributing to bispecific antibody stability and the reversal of self-associated aggregates. BIORESOUR BIOPROCESS 2024; 11:82. [PMID: 39177850 PMCID: PMC11343937 DOI: 10.1186/s40643-024-00796-y] [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: 06/19/2024] [Accepted: 08/09/2024] [Indexed: 08/24/2024] Open
Abstract
Bispecific antibodies (bsAbs) hold promises for enhanced therapeutic potential surpassing that of their parental monoclonal antibodies. However, bsAbs pose great challenges in their manufacturing, and one of the common reasons is their susceptibility to aggregation. Building on previous studies demonstrating the functionality and potential manufacturability of Fab-scFv format bsAb, this investigation delved into the impact of environmental factors-such as pH, buffer types, ionic strength, protein concentrations, and temperatures-on its stability and the reversal of its self-associated aggregates. Mildly acidic, low-salt conditions were found optimal, ensuring bsAb stability for 30 days even at elevated temperature of 40 °C. Furthermore, these conditions facilitated the reversal of its self-associated aggregates to monomers during the initial 7-day incubation period. Our findings underscore the robustness and resilience of Fab-scFv format bsAb, further confirming its potential manufacturability despite its current absence as commercial products.
Collapse
Affiliation(s)
- Nattha Ingavat
- Downstream Processing Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Nuruljannah Dzulkiflie
- Downstream Processing Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jia Min Liew
- Downstream Processing Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Xinhui Wang
- Downstream Processing Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Eunice Leong
- Animal Cell Bioprocessing Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Han Ping Loh
- Cell Line Development Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Say Kong Ng
- Animal Cell Bioprocessing Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yuansheng Yang
- Cell Line Development Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Wei Zhang
- Downstream Processing Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
| |
Collapse
|
7
|
Armstrong GB, Lewis A, Shah V, Taylor P, Jamieson CJ, Burley GA, Lewis W, Rattray Z. A First Insight into the Developability of an Immunoglobulin G3: A Combined Computational and Experimental Approach. ACS Pharmacol Transl Sci 2024; 7:2439-2451. [PMID: 39144567 PMCID: PMC11320737 DOI: 10.1021/acsptsci.4c00271] [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: 05/08/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 08/16/2024]
Abstract
Immunoglobulin G 3 (IgG3) monoclonal antibodies (mAbs) are high-value scaffolds for developing novel therapies. Despite their wide-ranging therapeutic potential, IgG3 physicochemical properties and developability characteristics remain largely under-characterized. Protein-protein interactions elevate solution viscosity in high-concentration formulations, impacting physicochemical stability, manufacturability, and the injectability of mAbs. Therefore, in this manuscript, the key molecular descriptors and biophysical properties of a model anti-IL-8 IgG1 and its IgG3 ortholog are characterized. A computational and experimental framework was applied to measure molecular descriptors impacting their downstream developability. Findings from this approach underpin a detailed understanding of the molecular characteristics of IgG3 mAbs as potential therapeutic entities. This work is the first report examining the manufacturability of IgG3 for high-concentration mAb formulations. While poorer conformational and colloidal stability and elevated solution viscosity were observed for IgG3, future efforts controlling surface potential through sequence-engineering of solvent-accessible patches can be used to improve biophysical parameters that dictate mAb developability.
Collapse
Affiliation(s)
- Georgina B. Armstrong
- Drug
Substance Development, GlaxoSmithKline, Gunnels Wood Road, Stevenage SG1 2NY, U.K.
- Strathclyde
Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, U.K.
| | - Alan Lewis
- Computational
and Modelling Sciences, GlaxoSmithKline, Gunnels Wood Road, Stevenage SG1 2NY, U.K.
| | - Vidhi Shah
- Large
Molecule Discovery, GlaxoSmithKline, Gunnels Wood Road, Stevenage SG1 2NY, U.K.
| | - Paul Taylor
- Drug
Substance Development, GlaxoSmithKline, Gunnels Wood Road, Stevenage SG1 2NY, U.K.
| | - Craig J. Jamieson
- Department
of Pure and Applied Chemistry, University
of Strathclyde, Glasgow G1 1XL, U.K.
| | - Glenn A. Burley
- Department
of Pure and Applied Chemistry, University
of Strathclyde, Glasgow G1 1XL, U.K.
| | - William Lewis
- Drug
Substance Development, GlaxoSmithKline, Gunnels Wood Road, Stevenage SG1 2NY, U.K.
| | - Zahra Rattray
- Strathclyde
Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, U.K.
| |
Collapse
|
8
|
Zhang Y, Mi J, Wu W, Fei J, Lv B, Yu X, Wen K, Shen J, Wang Z. Investigation of Antibody Tolerance in Methanol for Analytical Purposes: Methanol Effect Patterns and Molecular Mechanisms. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402050. [PMID: 38889249 PMCID: PMC11336977 DOI: 10.1002/advs.202402050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/16/2024] [Indexed: 06/20/2024]
Abstract
The extraction of targets from biological samples for immunoassays using organic solvents, such as methanol, is often necessary. However, high concentrations of organic solvents in extracts invariably lead to instability of the employed antibody, resulting in poor performance of the immunoassay. Evaluating the tolerance ability and exploring the molecular mechanisms of antibody tolerance in organic solvents are essential for the development of robust immunoassays. In this work, 25 monoclonal antibodies and methanol are utilized as models to address these questions. A novel protocol is initially established to precisely and rapidly determine antibody tolerance in methanol, identifying two distinct methanol effect patterns. Through a detailed investigation of the structural basis, a novel hypothesis regarding methanol effect patterns is proposed, termed "folding-aggregation," which is subsequently validated through molecular dynamics simulations. Furthermore, the investigation of sequence basis reveals significant differences in residue types within the complementarity-determining regions and ligand-binding residues, distinguishing the two antibody methanol effect patterns. Moreover, the methanol effect patterns of the antibodies are defined by germline antibodies. This work represents the first exploration of antibody methanol effect patterns and associated molecular mechanisms, with potential implications for the discovery and engineering of tolerant antibodies for the development of robust immunoassays.
Collapse
Affiliation(s)
- Yingjie Zhang
- National Key Laboratory of Veterinary Public Health and SafetyBeijing Key Laboratory of Detection Technology for Animal‐Derived FoodCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193P. R. China
| | - Jiafei Mi
- National Key Laboratory of Veterinary Public Health and SafetyBeijing Key Laboratory of Detection Technology for Animal‐Derived FoodCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193P. R. China
| | - Weilin Wu
- National Key Laboratory of Veterinary Public Health and SafetyBeijing Key Laboratory of Detection Technology for Animal‐Derived FoodCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193P. R. China
| | - Jie Fei
- National Key Laboratory of Veterinary Public Health and SafetyBeijing Key Laboratory of Detection Technology for Animal‐Derived FoodCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193P. R. China
| | - Bochen Lv
- National Key Laboratory of Veterinary Public Health and SafetyBeijing Key Laboratory of Detection Technology for Animal‐Derived FoodCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193P. R. China
| | - Xuezhi Yu
- National Key Laboratory of Veterinary Public Health and SafetyBeijing Key Laboratory of Detection Technology for Animal‐Derived FoodCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193P. R. China
| | - Kai Wen
- National Key Laboratory of Veterinary Public Health and SafetyBeijing Key Laboratory of Detection Technology for Animal‐Derived FoodCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193P. R. China
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health and SafetyBeijing Key Laboratory of Detection Technology for Animal‐Derived FoodCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193P. R. China
| | - Zhanhui Wang
- National Key Laboratory of Veterinary Public Health and SafetyBeijing Key Laboratory of Detection Technology for Animal‐Derived FoodCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193P. R. China
| |
Collapse
|
9
|
Wang S, Zhang W, Yang B, Zhang X, Fang J, Rui H, Chen Z, Gu J, Chen Z, Xu J. A case study of a bispecific antibody manufacturability assessment and optimization during discovery stage and its implications. Antib Ther 2024; 7:189-198. [PMID: 39036070 PMCID: PMC11259756 DOI: 10.1093/abt/tbae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 07/23/2024] Open
Abstract
The manufacturability assessment and optimization of bispecific antibodies (bsAbs) during the discovery stage are crucial for the success of the drug development process, impacting the speed and cost of advancing such therapeutics to the Investigational New Drug (IND) stage and ultimately to the market. The complexity of bsAbs creates challenges in employing effective evaluation methods to detect developability risks in early discovery stage, and poses difficulties in identifying the root causes and implementing subsequent engineering solutions. This study presents a case of engineering a bsAb that displayed a normal solution appearance during the discovery phase but underwent significant precipitation when subjected to agitation stress during 15 L Chemistry, Manufacturing, and Control (CMC) production Leveraging analytical tools, structural analysis, in silico prediction, and wet-lab validations, the key molecular origins responsible for the observed precipitation were identified and addressed. Sequence engineering to reduce protein surface hydrophobicity and enhance conformational stability proved effective in resolving agitation-induced aggregation. The refined bsAb sequences enabled successful mass production in CMC department. The findings of this case study contribute to the understanding of the fundamental mechanism of agitation-induced aggregation and offer a potential protein engineering procedure for addressing similar issues in bsAb. Furthermore, this case study emphasizes the significance of a close partnership between Discovery and CMC teams. Integrating CMC's rigorous evaluation methods with Discovery's engineering capability can facilitate a streamlined development process for bsAb molecules.
Collapse
Affiliation(s)
- Shuang Wang
- Biologics Innovation Discovery, WuXi Biologics, 1951 Huifeng West Road, Fengxian District, Shanghai, 201400, China
| | - Weijie Zhang
- Biologics Innovation Discovery, WuXi Biologics, 1951 Huifeng West Road, Fengxian District, Shanghai, 201400, China
| | - Baotian Yang
- Biologics Innovation Discovery, WuXi Biologics, 1951 Huifeng West Road, Fengxian District, Shanghai, 201400, China
| | - Xudong Zhang
- Downstream Process Development (DSPD), WuXi Biologics, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai, 200131, China
| | - Jing Fang
- Biologics Innovation Discovery, WuXi Biologics, 1951 Huifeng West Road, Fengxian District, Shanghai, 201400, China
| | - Haopeng Rui
- D3 Bio (Wuxi) Co., Ltd., 1101, 11/F, Building 1, No.6, Lane 38, Yuanshen Road, Pudong, Shanghai, 200120, China
| | - Zhijian Chen
- D3 Bio (Wuxi) Co., Ltd., 1101, 11/F, Building 1, No.6, Lane 38, Yuanshen Road, Pudong, Shanghai, 200120, China
| | - Jijie Gu
- Biologics Innovation Discovery, WuXi Biologics, 1951 Huifeng West Road, Fengxian District, Shanghai, 201400, China
| | - Zhiqiang Chen
- D3 Bio (Wuxi) Co., Ltd., 1101, 11/F, Building 1, No.6, Lane 38, Yuanshen Road, Pudong, Shanghai, 200120, China
| | - Jianqing Xu
- Biologics Innovation Discovery, WuXi Biologics, 1951 Huifeng West Road, Fengxian District, Shanghai, 201400, China
| |
Collapse
|
10
|
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
|
11
|
Chis AA, Dobrea CM, Arseniu AM, Frum A, Rus LL, Cormos G, Georgescu C, Morgovan C, Butuca A, Gligor FG, Vonica-Tincu AL. Antibody-Drug Conjugates-Evolution and Perspectives. Int J Mol Sci 2024; 25:6969. [PMID: 39000079 PMCID: PMC11241239 DOI: 10.3390/ijms25136969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Antineoplastic therapy is one of the main research themes of this century. Modern approaches have been implemented to target and heighten the effect of cytostatic drugs on tumors and diminish their general/unspecific toxicity. In this context, antibody-drug conjugates (ADCs) represent a promising and successful strategy. The aim of this review was to assess different aspects regarding ADCs. They were presented from a chemical and a pharmacological perspective and aspects like structure, conjugation and development particularities alongside effects, clinical trials, safety issues and perspectives and challenges for future use of these drugs were discussed. Representative examples include but are not limited to the following main structural components of ADCs: monoclonal antibodies (trastuzumab, brentuximab), linkers (pH-sensitive, reduction-sensitive, peptide-based, phosphate-based, and others), and payloads (doxorubicin, emtansine, ravtansine, calicheamicin). Regarding pharmacotherapy success, the high effectiveness expectation associated with ADC treatment is supported by the large number of ongoing clinical trials. Major aspects such as development strategies are first discussed, advantages and disadvantages, safety and efficacy, offering a retrospective insight on the subject. The second part of the review is prospective, focusing on various plans to overcome the previously identified difficulties.
Collapse
Affiliation(s)
| | | | - Anca Maria Arseniu
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | - Adina Frum
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | - Luca-Liviu Rus
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | - Gabriela Cormos
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | - Cecilia Georgescu
- Faculty of Agriculture Science, Food Industry and Environmental Protection, "Lucian Blaga" University of Sibiu, 550012 Sibiu, Romania
| | - Claudiu Morgovan
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | - Anca Butuca
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | | | | |
Collapse
|
12
|
Liang G, Madhavarao CN, Morris C, O'Connor T, Ashraf M, Yoon S. Effects of process intensification on homogeneity of an IgG1:κ monoclonal antibody during perfusion culture. Appl Microbiol Biotechnol 2024; 108:274. [PMID: 38530495 DOI: 10.1007/s00253-024-13110-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 03/01/2024] [Accepted: 03/13/2024] [Indexed: 03/28/2024]
Abstract
The pharmaceutical industry employs various strategies to improve cell productivity. These strategies include process intensification, culture media improvement, clonal selection, media supplementation and genetic engineering of cells. However, improved cell productivity has inherent risk of impacting product quality attributes (PQA). PQAs may affect the products' efficacy via stability, bioavailability, or in vivo bioactivity. Variations in manufacturing process may introduce heterogeneity in the products by altering the type and extent of N-glycosylation, which is a PQA of therapeutic proteins. We investigated the effect of different cell densities representing increasing process intensification in a perfusion cell culture on the production of an IgG1-κ monoclonal antibody from a CHO-K1 cell line. This antibody is glycosylated both on light chain and heavy chain. Our results showed that the contents of glycosylation of IgG1-κ mAb increased in G0F and fucosylated type glycans as a group, whereas sialylated type glycans decreased, for the mAb whole protein. Overall, significant differences were observed in amounts of G0F, G1F, G0, G2FS1, and G2FS2 type glycans across all process intensification levels. G2FS2 and G2 type N-glycans were predominantly quantifiable from light chain rather than heavy chain. It may be concluded that there is a potential impact to product quality attributes of therapeutic proteins during process intensification via perfusion cell culture that needs to be assessed. Since during perfusion cell culture the product is collected throughout the duration of the process, lot allocation needs careful attention to process parameters, as PQAs are affected by the critical process parameters (CPPs). KEY POINTS: • Molecular integrity may suffer with increasing process intensity. • Galactosylated and sialylated N-glycans may decrease. • Perfusion culture appears to maintain protein charge structure.
Collapse
Affiliation(s)
- George Liang
- Division of Product Quality Research, OTR/OPQ, CDER/FDA, Silver Spring, MD, USA
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, MA, USA
| | | | - Caitlin Morris
- Division of Product Quality Research, OTR/OPQ, CDER/FDA, Silver Spring, MD, USA
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, MA, USA
| | - Thomas O'Connor
- Division of Product Quality Research, OTR/OPQ, CDER/FDA, Silver Spring, MD, USA
| | - Muhammad Ashraf
- Division of Product Quality Research, OTR/OPQ, CDER/FDA, Silver Spring, MD, USA
| | - Seongkyu Yoon
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, MA, USA
| |
Collapse
|
13
|
Carpanese D, Rossi V, Di Paolo V, Quintieri L, Penna A, Zuccolotto G, Sebellin J, Saran C, Pipitone F, Miolo G, De Diana E, Realdon N, Rigamonti N, Di Sarra F, Coppola M, Rosato A. Prolonging the stability of cetuximab (Erbitux®) and panitumumab (Vectibix®): An orthogonal assessment of physicochemical, biological and microbiological properties of original opened glass vials and diluted saline preparations. Int J Pharm 2024; 649:123643. [PMID: 38040395 DOI: 10.1016/j.ijpharm.2023.123643] [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: 09/19/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/03/2023]
Abstract
The two anti-epidermal growth factor receptor monoclonal antibodies (mAbs) cetuximab and panitumumab are the pillars for the treatment of EGFR-positive, KRAS wild-type metastatic colorectal cancers. However, stability data of these mAbs are generally missing or incomplete. Here, we report for the first time an orthogonal analysis of the stability of cetuximab (Erbitux®) and panitumumab (Vectibix®), either undiluted vial leftovers or saline dilutions in polyolefin/polyamide infusion bags. All samples were stored at 2-8 °C protected from light, according to their summary of product characteristics (SmPCs). Alternatively, opened vials and preparations were maintained at 25 °C for 15 h, and then stored again at 2-8 °C protected from light to mimic a temporary interruption of the cold chain. Vial leftovers proved stable up to 180 days when stored according to their SmPCs, while compounded preparations in infusion bags maintained their physiochemical, biological and microbiological stability up to 30 days. Additionally, no changes were detected up to 30 days for the same samples undergoing a thermal excursion. Our results provide additional rationale to the SmPCs, crucial especially in the case of reassignment and pre-preparation of bags. This information will allow hospitals to achieve significant cost savings, and better organization of the entire therapeutic process.
Collapse
Affiliation(s)
- Debora Carpanese
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Valentina Rossi
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy.
| | - Veronica Di Paolo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Luigi Quintieri
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Alessandro Penna
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Gaia Zuccolotto
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Jessica Sebellin
- Pharmacy Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Camilla Saran
- Pharmacy Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | | | - Giorgia Miolo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Elisabetta De Diana
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Nicola Realdon
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | | | | | - Marina Coppola
- Pharmacy Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy.
| |
Collapse
|
14
|
Brudar S, Breydo L, Chung E, Dill KA, Ehterami N, Phadnis K, Senapati S, Shameem M, Tang X, Tayyab M, Hribar-Lee B. Antibody association in solution: cluster distributions and mechanisms. MAbs 2024; 16:2339582. [PMID: 38666507 PMCID: PMC11057677 DOI: 10.1080/19420862.2024.2339582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/02/2024] [Indexed: 05/01/2024] Open
Abstract
Understanding factors that affect the clustering and association of antibodies molecules in solution is critical to their development as therapeutics. For 19 different monoclonal antibody (mAb) solutions, we measured the viscosities, the second virial coefficients, the Kirkwood-Buff integrals, and the cluster distributions of the antibody molecules as functions of protein concentration. Solutions were modeled using the statistical-physics Wertheim liquid-solution theory, representing antibodies as Y-shaped molecular structures of seven beads each. We found that high-viscosity solutions result from more antibody molecules per cluster. Multi-body properties such as viscosity are well predicted experimentally by the 2-body Kirkwood-Buff quantity, G22, but not by the second virial coefficient, B22, and well-predicted theoretically from the Wertheim protein-protein sticking energy. Weakly interacting antibodies are rate-limited by nucleation; strongly interacting ones by propagation. This approach gives a way to relate micro to macro properties of solutions of associating proteins.
Collapse
Affiliation(s)
- Sandi Brudar
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Leonid Breydo
- Formulation Development Group, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Elisha Chung
- Formulation Development Group, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Ken A. Dill
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, USA
- Department of Chemistry and Department of Physics and Astronomy, Stony Brook University, Stony Brook, NY, USA
| | - Nasim Ehterami
- Formulation Development Group, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Ketan Phadnis
- Formulation Development Group, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Samir Senapati
- Formulation Development Group, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Mohammed Shameem
- Formulation Development Group, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Xiaolin Tang
- Formulation Development Group, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Muhammmad Tayyab
- Formulation Development Group, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Barbara Hribar-Lee
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
15
|
Li Z, Du X, Wang YMC. A survey of FDA Approved Monoclonal Antibodies and Fc-fusion Proteins for Manufacturing Changes and Comparability Assessment. Pharm Res 2024; 41:13-27. [PMID: 37910341 DOI: 10.1007/s11095-023-03627-5] [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: 08/07/2023] [Accepted: 10/16/2023] [Indexed: 11/03/2023]
Abstract
OBJECTIVE Manufacturing changes occur commonly throughout stages of biologics development and may result in product quality attribute changes. As changes in critical quality attributes have the potential to affect clinical safety and efficacy of products, it is imperative to ensure the quality and clinical performance before introducing the after-change products. Thus, we embarked on this project to understand what data have supported the manufacturing changes for licensed products with pre- and post-approval changes. METHODS We surveyed the manufacturing changes of 85 monoclonal antibodies and 10 Fc fusion proteins approved by the Food and Drug Administration as of December 25, 2021. After collecting the type and timing of changes for these products, we investigated the approaches that provided supporting data for the changes. The source documents included reports submitted by applicants and FDA's regulatory reviews. RESULTS Analytical comparability was assessed to support all identified manufacturing changes. Supporting clinical data were available in 92% of these manufacturing changes; including data from pharmacokinetic comparability studies alone (3%), other studies on efficacy or safety (70%) and a combination of both (19%). Clinical pharmacokinetic comparability data contributed to supporting substantial changes, such as host cell type or master cell bank changes, concentration or formulation changes, and changes from pre-filled syringes to autoinjectors, especially when introduced after completing pivotal studies. CONCLUSION Our comprehensive retrospective analysis provides an understanding of the regulatory experience and industry practice, which could facilitate developing appropriate comparability approaches to support manufacturing changes in the future.
Collapse
Affiliation(s)
- Zhe Li
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration (OCP/CDER/FDA), 10903 New Hampshire Avenue, Silver Spring, MD, USA.
| | - Xiulian Du
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration (OCP/CDER/FDA), 10903 New Hampshire Avenue, Silver Spring, MD, USA
| | - Yow-Ming C Wang
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration (OCP/CDER/FDA), 10903 New Hampshire Avenue, Silver Spring, MD, USA
| |
Collapse
|
16
|
Prass TM, Garidel P, Schäfer LV, Blech M. Residue-resolved insights into the stabilization of therapeutic proteins by excipients: A case study of two monoclonal antibodies with arginine and glutamate. MAbs 2024; 16:2427771. [PMID: 39540607 PMCID: PMC11572152 DOI: 10.1080/19420862.2024.2427771] [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: 05/15/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Protein formulation development relies on the selection of excipients that inhibit protein-protein interactions preventing aggregation. Empirical strategies involve screening many excipient and buffer combinations by physicochemical characterization using forced degradation or temperature-induced stress, mostly under accelerated conditions. Such methods do not readily provide information on the inter- and intramolecular interactions responsible for the effects of excipients. Here, we describe a combined experimental and computational approach for investigating the effect of protein-excipient interactions on formulation stability, which allows the identification of preferential interaction sites and thus can aid in the selection of excipients to be experimentally screened. Model systems composed of two marketed therapeutic IgG1 monoclonal antibodies with identical Fc domain sequences, trastuzumab and omalizumab, were investigated with commonly used excipients arginine, glutamate, and equimolar arginine/glutamate mixtures. Protein-excipient interactions were studied using all-atom molecular dynamics (MD) simulations, which show accumulation of the excipients at specific antibody regions. Preferential excipient-interaction sites were particularly found for charged and aromatic residues and in the complementary-determining regions, with more pronounced arginine contacts for omalizumab than trastuzumab. These computational findings are in line with the more pronounced stabilizing effects of arginine observed in the long-term storage stability study. Furthermore, the aggregation and solubility propensity predicted by commonly used in silico tools do not align with the preferential excipient-interaction sites identified by the MD simulations, suggesting that different physicochemical mechanisms are at play.
Collapse
Affiliation(s)
- Tobias M. Prass
- Center for Theoretical Chemistry, Ruhr University Bochum, Bochum, Germany
| | - Patrick Garidel
- Pharmaceutical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, Biberach and der Riss, Germany
| | - Lars V. Schäfer
- Center for Theoretical Chemistry, Ruhr University Bochum, Bochum, Germany
| | - Michaela Blech
- Pharmaceutical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, Biberach and der Riss, Germany
| |
Collapse
|
17
|
Ingavat N, Wang X, Liew JM, Mahfut FB, But KP, Kok YJ, Bi X, Yang Y, Shintaro K, Tsoumpra M, Zhang W. Harnessing ceramic hydroxyapatite as an effective polishing strategy to remove product- and process-related impurities in bispecific antibody purification. BIORESOUR BIOPROCESS 2023; 10:93. [PMID: 38647984 PMCID: PMC10992335 DOI: 10.1186/s40643-023-00713-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 12/06/2023] [Indexed: 04/25/2024] Open
Abstract
Bispecific antibody (bsAb), a novel therapeutic modality, provides excellent treatment efficacy, yet poses numerous challenges to downstream process development, which are mainly due to the intricate diversity of bsAb structures and impurity profiles. Ceramic hydroxyapatite (CHT), a mixed-mode medium, allows proteins to interact with its calcium sites (C-sites) through metal affinity and/or its phosphate sites (P-sites) through cation exchange interactions. This dual-binding capability potentially offers unique bind and elute behaviours for different proteins of interest, resulting in optimal product purity when suitable elution conditions are employed. In this study, the effectiveness of CHT as a polishing step for bsAb purification was investigated across three model molecules and benchmarked against the traditional cation exchange chromatography (CEX). For both asymmetric and symmetric IgG-like bsAb post Protein A eluates, at least 97% product purity was achieved after CHT polishing. CHT delivered a superior aggregate clearance to CEX, resulting in low high molecular weight (HMW) impurities (0.5%) and low process-related impurities in the product pools. Moreover, CHT significantly mitigated "chromatography-induced aggregation" whereas eightfold more HMW was generated by CEX. This study illustrated the developability of CHT in effectively eliminating low molecular weight (LMW) impurities through post-load-wash (PLW) optimization, resulting in an additional reduction of up to 48% in LMW impurities. A mechanistic explanation regarding the performance of impurity removal and mitigation of the chromatography-induced aggregation by CHT was proposed, illustrating unique CHT capability is potentially driven by C-site cooperation, of which effectiveness could depend on the bsAb composition and size.
Collapse
Affiliation(s)
- Nattha Ingavat
- Downstream Processing Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Xinhui Wang
- Downstream Processing Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jia Min Liew
- Downstream Processing Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Farouq Bin Mahfut
- Cell Line Development Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Ka Pui But
- Protein Analytics Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yee Jiun Kok
- Protein Analytics Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Xuezhi Bi
- Protein Analytics Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yuansheng Yang
- Cell Line Development Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Kobayashi Shintaro
- Chromatography Media Business Division, HOYA Technosurgical Corporation, Singapore Branch, Singapore
| | - Maria Tsoumpra
- Chromatography Media Business Division, HOYA Technosurgical Corporation, Singapore Branch, Singapore
| | - Wei Zhang
- Downstream Processing Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
| |
Collapse
|
18
|
Prass T, Garidel P, Blech M, Schäfer LV. Viscosity Prediction of High-Concentration Antibody Solutions with Atomistic Simulations. J Chem Inf Model 2023; 63:6129-6140. [PMID: 37757589 PMCID: PMC10565822 DOI: 10.1021/acs.jcim.3c00947] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Indexed: 09/29/2023]
Abstract
The computational prediction of the viscosity of dense protein solutions is highly desirable, for example, in the early development phase of high-concentration biopharmaceutical formulations where the material needed for experimental determination is typically limited. Here, we use large-scale atomistic molecular dynamics (MD) simulations with explicit solvation to de novo predict the dynamic viscosities of solutions of a monoclonal IgG1 antibody (mAb) from the pressure fluctuations using a Green-Kubo approach. The viscosities at simulated mAb concentrations of 200 and 250 mg/mL are compared to the experimental values, which we measured with rotational rheometry. The computational viscosity of 24 mPa·s at the mAb concentration of 250 mg/mL matches the experimental value of 23 mPa·s obtained at a concentration of 213 mg/mL, indicating slightly different effective concentrations (or activities) in the MD simulations and in the experiments. This difference is assigned to a slight underestimation of the effective mAb-mAb interactions in the simulations, leading to a too loose dynamic mAb network that governs the viscosity. Taken together, this study demonstrates the feasibility of all-atom MD simulations for predicting the properties of dense mAb solutions and provides detailed microscopic insights into the underlying molecular interactions. At the same time, it also shows that there is room for further improvements and highlights challenges, such as the massive sampling required for computing collective properties of dense biomolecular solutions in the high-viscosity regime with reasonable statistical precision.
Collapse
Affiliation(s)
- Tobias
M. Prass
- Center
for Theoretical Chemistry, Ruhr University
Bochum, D-44780 Bochum, Germany
| | - Patrick Garidel
- Boehringer
Ingelheim Pharma GmbH & Co. KG, Innovation Unit, PDB, D-88397 Biberach
an der Riss, Germany
| | - Michaela Blech
- Boehringer
Ingelheim Pharma GmbH & Co. KG, Innovation Unit, PDB, D-88397 Biberach
an der Riss, Germany
| | - Lars V. Schäfer
- Center
for Theoretical Chemistry, Ruhr University
Bochum, D-44780 Bochum, Germany
| |
Collapse
|
19
|
Pang KT, Yang YS, Zhang W, Ho YS, Sormanni P, Michaels TCT, Walsh I, Chia S. Understanding and controlling the molecular mechanisms of protein aggregation in mAb therapeutics. Biotechnol Adv 2023; 67:108192. [PMID: 37290583 DOI: 10.1016/j.biotechadv.2023.108192] [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: 02/16/2023] [Revised: 05/09/2023] [Accepted: 06/01/2023] [Indexed: 06/10/2023]
Abstract
In antibody development and manufacturing, protein aggregation is a common challenge that can lead to serious efficacy and safety issues. To mitigate this problem, it is important to investigate its molecular origins. This review discusses (1) our current molecular understanding and theoretical models of antibody aggregation, (2) how various stress conditions related to antibody upstream and downstream bioprocesses can trigger aggregation, and (3) current mitigation strategies employed towards inhibiting aggregation. We discuss the relevance of the aggregation phenomenon in the context of novel antibody modalities and highlight how in silico approaches can be exploited to mitigate it.
Collapse
Affiliation(s)
- Kuin Tian Pang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore; School of Chemistry, Chemical Engineering, and Biotechnology, Nanyang Technology University, Singapore
| | - Yuan Sheng Yang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Wei Zhang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Ying Swan Ho
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Pietro Sormanni
- Chemistry of Health, Yusuf Hamied Department of Chemistry, University of Cambridge, United Kingdom
| | - Thomas C T Michaels
- Department of Biology, Institute of Biochemistry, ETH Zurich, Otto-Stern-Weg 3, 8093 Zurich, Switzerland; Bringing Materials to Life Initiative, ETH Zurich, Switzerland
| | - Ian Walsh
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore.
| | - Sean Chia
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore.
| |
Collapse
|
20
|
Roel-Touris J, Nadal M, Marcos E. Single-chain dimers from de novo immunoglobulins as robust scaffolds for multiple binding loops. Nat Commun 2023; 14:5939. [PMID: 37741853 PMCID: PMC10517939 DOI: 10.1038/s41467-023-41717-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 09/15/2023] [Indexed: 09/25/2023] Open
Abstract
Antibody derivatives have sought to recapitulate the antigen binding properties of antibodies, but with improved biophysical attributes convenient for therapeutic, diagnostic and research applications. However, their success has been limited by the naturally occurring structure of the immunoglobulin dimer displaying hypervariable binding loops, which is hard to modify by traditional engineering approaches. Here, we devise geometrical principles for de novo designing single-chain immunoglobulin dimers, as a tunable two-domain architecture that optimizes biophysical properties through more favorable dimer interfaces. Guided by these principles, we computationally designed protein scaffolds that were hyperstable, structurally accurate and robust for accommodating multiple functional loops, both individually and in combination, as confirmed through biochemical assays and X-ray crystallography. We showcase the modularity of this architecture by deep-learning-based diversification, opening up the possibility for tailoring the number, positioning, and relative orientation of ligand-binding loops targeting one or two distal epitopes. Our results provide a route to custom-design robust protein scaffolds for harboring multiple functional loops.
Collapse
Affiliation(s)
- Jorge Roel-Touris
- Protein Design and Modeling Lab, Department of Structural and Molecular Biology, Molecular Biology Institute of Barcelona (IBMB), CSIC, Baldiri Reixac 10, 08028, Barcelona, Spain
| | - Marta Nadal
- Protein Design and Modeling Lab, Department of Structural and Molecular Biology, Molecular Biology Institute of Barcelona (IBMB), CSIC, Baldiri Reixac 10, 08028, Barcelona, Spain
| | - Enrique Marcos
- Protein Design and Modeling Lab, Department of Structural and Molecular Biology, Molecular Biology Institute of Barcelona (IBMB), CSIC, Baldiri Reixac 10, 08028, Barcelona, Spain.
| |
Collapse
|
21
|
Zeven K, De Groof TW, Ceuppens H, Awad RM, Ertveldt T, de Mey W, Meeus F, Raes G, Breckpot K, Devoogdt N. Development and evaluation of nanobody tracers for noninvasive nuclear imaging of the immune-checkpoint TIGIT. Front Immunol 2023; 14:1268900. [PMID: 37799715 PMCID: PMC10548220 DOI: 10.3389/fimmu.2023.1268900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/04/2023] [Indexed: 10/07/2023] Open
Abstract
Introduction T cell Ig and ITIM domain receptor (TIGIT) is a next-generation immune checkpoint predominantly expressed on activated T cells and NK cells, exhibiting an unfavorable prognostic association with various malignancies. Despite the emergence of multiple TIGIT-blocking agents entering clinical trials, only a fraction of patients responded positively to anti-TIGIT therapy. Consequently, an urgent demand arises for noninvasive techniques to quantify and monitor TIGIT expression, facilitating patient stratification and enhancing therapeutic outcomes. Small antigen binding moieties such as nanobodies, are promising candidates for such tracer development. Methods We generated a panel of anti-human or anti-mouse TIGIT nanobodies from immunized llamas. In addition, we designed a single-chain variable fragment derived from the clinically tested monoclonal antibody Vibostolimab targeting TIGIT, and assessed its performance alongside the nanobodies. In vitro characterization studies were performed, including binding ability and affinity to cell expressed or recombinant TIGIT. After Technetium-99m labeling, the nanobodies and the single-chain variable fragment were evaluated in vivo for their ability to detect TIGIT expression using SPECT/CT imaging, followed by ex vivo biodistribution analysis. Results Nine nanobodies were selected for binding to recombinant and cell expressed TIGIT with low sub-nanomolar affinities and are thermostable. A six-fold higher uptake in TIGIT-overexpressing tumor was demonstrated one hour post- injection with Technetium-99m labeled nanobodies compared to an irrelevant control nanobody. Though the single-chain variable fragment exhibited superior binding to TIGIT-expressing peripheral blood mononuclear cells in vitro, its in vivo behavior yielded lower tumor-to-background ratios at one hour post- injection, indicating that nanobodies are better suited for in vivo imaging than the single-chain variable fragment. Despite the good affinity, high specificity and on-target uptake in mice in this setting, imaging of TIGIT expression on tumor- infiltrating lymphocytes within MC38 tumors remained elusive. This is likely due to the low expression levels of TIGIT in this model. Discussion The excellent affinity, high specificity and rapid on-target uptake in mice bearing TIGIT- overexpressing tumors showed the promising diagnostic potential of nanobodies to noninvasively image high TIGIT expression within the tumor. These findings hold promise for clinical translation to aid patient selection and improve therapy response.
Collapse
Affiliation(s)
- Katty Zeven
- Laboratory of Molecular Imaging and Therapy (MITH), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Timo W.M. De Groof
- Laboratory of Molecular Imaging and Therapy (MITH), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Hannelore Ceuppens
- Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Thomas Ertveldt
- Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Wout de Mey
- Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Fien Meeus
- Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Geert Raes
- Laboratory for Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Myeloid Cell Immunology Lab, Vlaams Instituut voor Biotechnologie (VIB) Center for Inflammation Research, Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Nick Devoogdt
- Laboratory of Molecular Imaging and Therapy (MITH), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|
22
|
Liu GY, Zhang Z, Yan Y, Wang S, Li N. Discovery and Characterization of an Acid-Labile Serine-Lysine Cross-Link in Antibody High-Molecular-Weight Species Using a Multipronged Mass Spectrometry Approach. Anal Chem 2023; 95:13813-13821. [PMID: 37674418 PMCID: PMC10515106 DOI: 10.1021/acs.analchem.3c01602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 08/28/2023] [Indexed: 09/08/2023]
Abstract
Characterizing the cross-links responsible for the covalent high-molecular-weight (HMW) species in therapeutic monoclonal antibodies (mAbs) is of great importance as it not only provides a framework for risk assessment but also offers insights for process improvement. However, owing to the complexity and low abundance, identification of novel and unknown cross-links in mAb products can be very challenging. Here, applying a multipronged MS-based approach, we report the discovery of a novel covalent cross-link formed via an imine bond between lysine and serine residues. In particular, this Ser-Lys cross-link was found to be acid-labile and can be easily overlooked by conventional LC-MS techniques operated at low pH. It is worth noting that although imine-based cross-link has been previously reported in collagen protein cross-linking, this is the first time that a Ser-Lys cross-link has been found in a mAb product that contributes to covalent HMW species formation.
Collapse
Affiliation(s)
- Gao-Yuan Liu
- Analytical Chemistry, Regeneron
Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Zhengqi Zhang
- Analytical Chemistry, Regeneron
Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Yuetian Yan
- Analytical Chemistry, Regeneron
Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Shunhai Wang
- Analytical Chemistry, Regeneron
Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Ning Li
- Analytical Chemistry, Regeneron
Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| |
Collapse
|
23
|
Basha S, Mukunda DC, Rodrigues J, Gail D'Souza M, Gangadharan G, Pai AR, Mahato KK. A comprehensive review of protein misfolding disorders, underlying mechanism, clinical diagnosis, and therapeutic strategies. Ageing Res Rev 2023; 90:102017. [PMID: 37468112 DOI: 10.1016/j.arr.2023.102017] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/14/2023] [Accepted: 07/14/2023] [Indexed: 07/21/2023]
Abstract
INTRODUCTION Proteins are the most common biological macromolecules in living system and are building blocks of life. They are extremely dynamic in structure and functions. Due to several modifications, proteins undergo misfolding, leading to aggregation and thereby developing neurodegenerative and systemic diseases. Understanding the pathology of these diseases and the techniques used to diagnose them is therefore crucial for their effective management . There are several techniques, currently being in use to diagnose them and those will be discussed in this review. AIM/OBJECTIVES Current review aims to discuss an overview of protein aggregation and the underlying mechanisms linked to neurodegeneration and systemic diseases. Also, the review highlights protein misfolding disorders, their clinical diagnosis, and treatment strategies. METHODOLOGY Literature related to neurodegenerative and systemic diseases was explored through PubMed, Google Scholar, Scopus, and Medline databases. The keywords used for literature survey and analysis are protein aggregation, neurodegenerative disorders, Alzheimer's disease, Parkinson's disease, systemic diseases, protein aggregation mechanisms, etc. DISCUSSION /CONCLUSION: This review summarises the pathogenesis of neurodegenerative and systemic disorders caused by protein misfolding and aggregation. The clinical diagnosis and therapeutic strategies adopted for the management of these diseases are also discussed to aid in a better understanding of protein misfolding disorders. Many significant concerns about the role, characteristics, and consequences of protein aggregates in neurodegenerative and systemic diseases are not clearly understood to date. Regardless of technological advancements, there are still great difficulties in the management and cure of these diseases. Therefore, for better understanding, diagnosis, and treatment of neurodegenerative and systemic diseases, more studies to identify novel drugs that may aid in their treatment and management are required.
Collapse
Affiliation(s)
- Shaik Basha
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | | | - Jackson Rodrigues
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Meagan Gail D'Souza
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Gireesh Gangadharan
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Aparna Ramakrishna Pai
- Department of Neurology, Kasturba Medical College - Manipal, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Krishna Kishore Mahato
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| |
Collapse
|
24
|
Wu Q, Cao C, Wei S, He H, Chen K, Su L, Liu Q, Li S, Lai Y, Li J. Decreasing hydrophobicity or shielding hydrophobic areas of CH2 attenuates low pH-induced IgG4 aggregation. Front Bioeng Biotechnol 2023; 11:1257665. [PMID: 37711444 PMCID: PMC10497874 DOI: 10.3389/fbioe.2023.1257665] [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: 07/12/2023] [Accepted: 08/18/2023] [Indexed: 09/16/2023] Open
Abstract
Protein aggregation is a major challenge in the development of therapeutic monoclonal antibodies (mAbs). Several stressors can cause protein aggregation, including temperature shifts, mechanical forces, freezing-thawing cycles, oxidants, reductants, and extreme pH. When antibodies are exposed to low pH conditions, aggregation increases dramatically. However, low pH treatment is widely used in protein A affinity chromatography and low pH viral inactivation procedures. In the development of an IgG4 subclass antibody, mAb1-IgG4 showed a strong tendency to aggregate when temporarily exposed to low pH conditions. Our findings showed that the aggregation of mAb1-IgG4 under low pH conditions is determined by the stability of the Fc. The CH2 domain is the least stable domain in mAb1-IgG4. The L309E, Q311D, and Q311E mutations in the CH2 domain significantly reduced the aggregation propensity, which could be attributed to a reduction in the hydrophobicity of the CH2 domain. Protein stabilizers, such as sucrose and mannose, could also attenuate low pH-induced mAb1-IgG4 aggregation by shielding hydrophobic areas and increasing protein stability. Our findings provide valuable strategies for managing the aggregation of protein therapeutics with a human IgG4 backbone.
Collapse
Affiliation(s)
- Qiang Wu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
- Zhuhai United Laboratories Co., Ltd., Zhuhai, Guangdong, China
| | - Chunlai Cao
- Zhuhai United Laboratories Co., Ltd., Zhuhai, Guangdong, China
- The United Biotechnology (Zhuhai Hengqin) Co., Ltd., Zhuhai, Guangdong, China
| | - Suzhen Wei
- The United Biotechnology (Zhuhai Hengqin) Co., Ltd., Zhuhai, Guangdong, China
| | - Hua He
- The United Biotechnology (Zhuhai Hengqin) Co., Ltd., Zhuhai, Guangdong, China
| | - Kangyue Chen
- Zhuhai United Laboratories Co., Ltd., Zhuhai, Guangdong, China
| | - Lijuan Su
- Zhuhai United Laboratories Co., Ltd., Zhuhai, Guangdong, China
| | - Qiulian Liu
- Zhuhai United Laboratories Co., Ltd., Zhuhai, Guangdong, China
| | - Shuang Li
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Yongjie Lai
- Department of Microbiology and Immunology, Zunyi Medical University (Zhuhai Campus), Zhuhai, Guangdong, China
| | - Jing Li
- Zhuhai United Laboratories Co., Ltd., Zhuhai, Guangdong, China
| |
Collapse
|
25
|
Calcium-dependent affinity ligands for the purification of antibody fragments at neutral pH. J Chromatogr A 2023; 1694:463902. [PMID: 36871527 DOI: 10.1016/j.chroma.2023.463902] [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: 12/27/2022] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 02/27/2023]
Abstract
The emerging formats of antibody fragments for biotherapeutics suffer from inadequate purification methods, delaying the advances of innovative therapies. One of the top therapeutic candidates, the single-chain variable fragment (scFv), requires the development of individual purification protocols dependent on the type of scFv. The available approaches that are based on selective affinity chromatography but do not involve the use of a purification tag, such as Protein L and Protein A chromatography, require acidic elution buffers. These elution conditions can cause the formation of aggregates and thereby greatly compromise the yield, which can be a major problem for scFvs that are generally unstable molecules. Due to the costly and time-consuming production of biological drugs, like antibody fragments, we have engineered novel purification ligands that elute the scFvs in a calcium-dependent manner. The developed ligands are equipped with new, selective binding surfaces and were shown to efficiently elute all captured scFv at neutral pH with the use of a calcium chelator. Further, two of three ligands were proven not to bind to the CDRs of the scFv, indicating potential for use as generic affinity ligands to a range of different scFvs. Multimerization and optimization of the most promising ligand led to a 3-fold increase in binding capacity for the hexamer compared to the monomer, in addition to highly selective and efficient purification of a scFv with >95% purity in a single purification step. This calcium-dependent ligand could revolutionize the scFv industry, greatly facilitating the purification procedure and improving the quality of the final product.
Collapse
|
26
|
Ko S, Ju MS, Ahn HM, Na JH, Ko WH, Jo M, Kyung M, Lim CS, Ko BJ, Lee WK, Kim YJ, Jung ST. Engineered Human Antibody with Improved Endothelin Receptor Type A Binding Affinity, Developability, and Serum Persistence Exhibits Excellent Antitumor Potency. Mol Pharm 2023; 20:1247-1255. [PMID: 36563318 DOI: 10.1021/acs.molpharmaceut.2c00923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Endothelin receptor A (ETA), a class A G protein-coupled receptor (GPCR), is a promising tumor-associated antigen due to its close association with the progression and metastasis of many types of cancer, such as colorectal, breast, lung, ovarian, and prostate cancer. However, only small-molecule drugs have been developed as ETA antagonists with anticancer effects. In a previous study, we identified an antibody (AG8) with highly selective binding to human ETA through screening of a human naïve immune antibody library. Although both in vitro and in vivo experiments indicated that the identified AG8 had anticancer effects, there is a need for improvement in biochemical and physicochemical properties such as the ETA binding affinity, thermostability, and productivity. In this study, we engineered the framework regions of AG8 and isolated an anti-ETA antibody (MJF1) exhibiting significantly improved thermostability and ETA binding affinity. Subsequently, our previously isolated PFc29, an Fc variant with an enhanced pH-dependent human FcRn binding profile, was introduced to MJF1, and the resulting Fc-engineered anti-ETA antibody (MJF1-PFc29) inhibited the proliferation of tumor cells comparably to MJF1 and showed a 4.2-fold increased serum half-life in human FcRn transgenic mice. Moreover, MJF1-PFc29 elicited higher tumor growth inhibition in colorectal cancer xenograft mice compared to MJF1. Our results demonstrate that the engineered human anti-ETA antibody MJF1-PFc29 has great therapeutic potential and high antitumor potency against various types of cancers including colorectal cancer.
Collapse
Affiliation(s)
- Sanghwan Ko
- Department of Biomedical Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea.,Institute of Human Genetics, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Man-Seok Ju
- Department of Biomedical Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea.,Institute of Human Genetics, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Hye-Mi Ahn
- Targeted Therapy Branch, Division of Rare and Refractory Cancer, Research Institute, National Cancer Center, Goyang, Gyeonggi-do 10408, Republic of Korea
| | - Jung-Hyun Na
- Department of Pharmaceutical Engineering, Sangji University, Wonju, Gangwon-do 26339, Republic of Korea
| | - Woo Hyung Ko
- Department of Biomedical Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea.,BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Migyeong Jo
- Department of Biomedical Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea.,BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Munsu Kyung
- Department of Biomedical Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea.,BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Chung Su Lim
- New Drug Development Center, Osong Medical Innovation Foundation, Cheongju, Chungcheongbuk-do 28160, Republic of Korea
| | - Byoung Joon Ko
- School of Biopharmaceutical and Medical Sciences, Sungshin Women's University, Seoul 02844, Republic of Korea
| | - Won-Kyu Lee
- New Drug Development Center, Osong Medical Innovation Foundation, Cheongju, Chungcheongbuk-do 28160, Republic of Korea
| | - Youn-Jae Kim
- Targeted Therapy Branch, Division of Rare and Refractory Cancer, Research Institute, National Cancer Center, Goyang, Gyeonggi-do 10408, Republic of Korea
| | - Sang Taek Jung
- Department of Biomedical Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea.,Institute of Human Genetics, Korea University College of Medicine, Seoul 02841, Republic of Korea.,BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| |
Collapse
|
27
|
Fukuhara D, Yamauchi M, Itoh SG, Okumura H. Ingenuity in performing replica permutation: How to order the state labels for improving sampling efficiency. J Comput Chem 2023; 44:534-545. [PMID: 36346137 PMCID: PMC10099539 DOI: 10.1002/jcc.27020] [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: 07/22/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 11/11/2022]
Abstract
In the replica-permutation method, an advanced version of the replica-exchange method, all combinations of replicas and parameters are considered for parameter permutation, and a list of all the combinations is prepared. Here, we report that the temperature transition probability depends on how the list is created, especially in replica permutation with solute tempering (RPST). We found that the transition probabilities decrease at large replica indices when the combinations are sequentially assigned to the state labels as in the originally proposed list. To solve this problem, we propose to modify the list by randomly assigning the combinations to the state labels. We performed molecular dynamics simulations of amyloid-β(16-22) peptides using RPST with the "randomly assigned" list (RPST-RA) and RPST with the "sequentially assigned" list (RPST-SA). The results show the decreases in the transition probabilities in RPST-SA are eliminated, and the sampling efficiency is improved in RPST-RA.
Collapse
Affiliation(s)
- Daiki Fukuhara
- Department of Structural Molecular Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Japan.,Department of Theoretical and Computational Molecular Science, Institute for Molecular Science, National Institutes of Natural Sciences, Okazaki, Japan
| | - Masataka Yamauchi
- Department of Structural Molecular Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Japan.,Department of Theoretical and Computational Molecular Science, Institute for Molecular Science, National Institutes of Natural Sciences, Okazaki, Japan
| | - Satoru G Itoh
- Department of Structural Molecular Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Japan.,Department of Theoretical and Computational Molecular Science, Institute for Molecular Science, National Institutes of Natural Sciences, Okazaki, Japan.,Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Japan
| | - Hisashi Okumura
- Department of Structural Molecular Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Japan.,Department of Theoretical and Computational Molecular Science, Institute for Molecular Science, National Institutes of Natural Sciences, Okazaki, Japan.,Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Japan
| |
Collapse
|
28
|
Desai PG, Garidel P, Gbormittah FO, Kamen DE, Mills BJ, Narasimhan CN, Singh S, Stokes ESE, Walsh ER. An Intercompany Perspective on Practical Experiences of Predicting, Optimizing and Analyzing High Concentration Biologic Therapeutic Formulations. J Pharm Sci 2023; 112:359-369. [PMID: 36442683 DOI: 10.1016/j.xphs.2022.11.020] [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: 09/02/2022] [Revised: 11/18/2022] [Accepted: 11/18/2022] [Indexed: 11/27/2022]
Abstract
Developing high-dose biologic drugs for subcutaneous injection often requires high-concentration formulations and optimizing viscosity, solubility, and stability while overcoming analytical, manufacturing, and administration challenges. To understand industry approaches for developing high-concentration formulations, the Formulation Workstream of the BioPhorum Development Group, an industry-wide consortium, conducted an inter-company collaborative exercise which included several surveys. This collaboration provided an industry perspective, experience, and insight into the practicalities for developing high-concentration biologics. To understand solubility and viscosity, companies desire predictive tools, but experience indicates that these are not reliable and experimental strategies are best. Similarly, most companies prefer accelerated and stress stability studies to in-silico or biophysical-based prediction methods to assess aggregation. In addition, optimization of primary container-closure and devices are pursued to mitigate challenges associated with high viscosity of the formulation. Formulation strategies including excipient selection and application of studies at low concentration to high-concentration formulations are reported. Finally, analytical approaches to high concentration formulations are presented. The survey suggests that although prediction of viscosity, solubility, and long-term stability is desirable, the outcome can be inconsistent and molecule dependent. Significant experimental studies are required to confirm robust product definition as modeling at low protein concentrations will not necessarily extrapolate to high concentration formulations.
Collapse
Affiliation(s)
- Preeti G Desai
- Bristol Myers Squibb, Sterile Product Development, 556 Morris Avenue, Summit, NJ 07901, USA
| | - Patrick Garidel
- Boehringer Ingelheim Pharma GmbH Co KG, Innovation Unit, PDB-TIP, 88397 Biberach an der Riss, Germany
| | - Francisca O Gbormittah
- GlaxoSmithKline, Strategic External Development, 1000 Winter Street North, Waltham, MA 02451, USA
| | - Douglas E Kamen
- Regeneron Pharmaceuticals Inc., Formulation Development, 777 Old Saw Mill River Road, Tarrytown, NY 10591, USA
| | - Brittney J Mills
- AbbVie, NBE Drug Product Development, 1 N Waukegan Road, North Chicago, IL 60064, USA
| | | | - Shubhadra Singh
- GlaxoSmithKline R&D, Biopharmaceutical Product Sciences, Collegeville, PA 19426, USA
| | - Elaine S E Stokes
- BioPhorum, The Gridiron Building, 1 Pancras Square, London N1C 4AG UK.
| | - Erika R Walsh
- Merck & Co., Inc., Sterile and Specialty Products, Rahway, NJ, USA
| |
Collapse
|
29
|
Erreni M, D'Autilia F, Avigni R, Bolli E, Arnouk SM, Movahedi K, Debie P, Anselmo A, Parente R, Vincke C, van Leeuwen FW, Allavena P, Garlanda C, Mantovani A, Doni A, Hernot S, Van Ginderachter JA. Size-advantage of monovalent nanobodies against the macrophage mannose receptor for deep tumor penetration and tumor-associated macrophage targeting. Theranostics 2023; 13:355-373. [PMID: 36593955 PMCID: PMC9800720 DOI: 10.7150/thno.77560] [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: 07/28/2022] [Accepted: 11/20/2022] [Indexed: 12/23/2022] Open
Abstract
Rationale: Nanobodies (Nbs) have emerged as an elegant alternative to the use of conventional monoclonal antibodies in cancer therapy, but a detailed microscopic insight into the in vivo pharmacokinetics of different Nb formats in tumor-bearers is lacking. This is especially relevant for the recognition and targeting of pro-tumoral tumor-associated macrophages (TAMs), which may be located in less penetrable tumor regions. Methods: We employed anti-Macrophage Mannose Receptor (MMR) Nbs, in a monovalent (m) or bivalent (biv) format, to assess in vivo TAM targeting. Intravital and confocal microscopy were used to analyse the blood clearance rate and targeting kinetics of anti-MMR Nbs in tumor tissue, healthy muscle tissue and liver. Fluorescence Molecular Tomography was applied to confirm anti-MMR Nb accumulation in the primary tumor and in metastatic lesions. Results: Intravital microscopy demonstrated significant differences in the blood clearance rate and macrophage targeting kinetics of (m) and (biv)anti-MMR Nbs, both in tumoral and extra-tumoral tissue. Importantly, (m)anti-MMR Nbs are superior in reaching tissue macrophages, an advantage that is especially prominent in tumor tissue. The administration of a molar excess of unlabelled (biv)anti-MMR Nbs increased the (m)anti-MMR Nb bioavailability and impacted on its macrophage targeting kinetics, preventing their accumulation in extra-tumoral tissue (especially in the liver) but only partially influencing their interaction with TAMs. Finally, anti-MMR Nb administration not only allowed the visualization of TAMs in primary tumors, but also at a distant metastatic site. Conclusions: These data describe, for the first time, a microscopic analysis of (m) and (biv)anti-MMR Nb pharmacokinetics in tumor and healthy tissues. The concepts proposed in this study provide important knowledge for the future use of Nbs as diagnostic and therapeutic agents, especially for the targeting of tumor-infiltrating immune cells.
Collapse
Affiliation(s)
- Marco Erreni
- Unit of Advanced Optical Microscopy, IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy.,✉ Corresponding authors: Marco Erreni, ; Jo A. Van Ginderachter,
| | - Francesca D'Autilia
- Unit of Advanced Optical Microscopy, IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Roberta Avigni
- IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Evangelia Bolli
- Cellular and Molecular Immunology Lab, Vrije Universiteit Brussel, Brussels, Belgium (Pleinlaan 2, 1050 Brussels).,Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Sana M. Arnouk
- Cellular and Molecular Immunology Lab, Vrije Universiteit Brussel, Brussels, Belgium (Pleinlaan 2, 1050 Brussels).,Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Kiavash Movahedi
- Cellular and Molecular Immunology Lab, Vrije Universiteit Brussel, Brussels, Belgium (Pleinlaan 2, 1050 Brussels).,Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Pieterjan Debie
- Laboratory for In vivo Cellular and Molecular Imaging (ICMI-BEFY/MIMA), Vrije Universiteit Brussel, Brussels, Belgium (Laarbeeklaan 103, 1090 Brussels)
| | - Achille Anselmo
- IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Raffaella Parente
- Unit of Advanced Optical Microscopy, IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Cécile Vincke
- Cellular and Molecular Immunology Lab, Vrije Universiteit Brussel, Brussels, Belgium (Pleinlaan 2, 1050 Brussels).,Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Fijs W.B. van Leeuwen
- Leiden University Medical Center, Interventional Molecular Imaging Laboratory, Albinusdreef 2 2333 ZA Leiden
| | - Paola Allavena
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy.,IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Cecilia Garlanda
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy.,IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Alberto Mantovani
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy.,IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy.,The William Harvey Research Institute, Queen Mary University of London, London EC1M6BQ, UK
| | - Andrea Doni
- Unit of Advanced Optical Microscopy, IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Sophie Hernot
- Laboratory for In vivo Cellular and Molecular Imaging (ICMI-BEFY/MIMA), Vrije Universiteit Brussel, Brussels, Belgium (Laarbeeklaan 103, 1090 Brussels)
| | - Jo A. Van Ginderachter
- Cellular and Molecular Immunology Lab, Vrije Universiteit Brussel, Brussels, Belgium (Pleinlaan 2, 1050 Brussels).,Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium.,✉ Corresponding authors: Marco Erreni, ; Jo A. Van Ginderachter,
| |
Collapse
|
30
|
Mieczkowski C, Zhang X, Lee D, Nguyen K, Lv W, Wang Y, Zhang Y, Way J, Gries JM. Blueprint for antibody biologics developability. MAbs 2023; 15:2185924. [PMID: 36880643 PMCID: PMC10012935 DOI: 10.1080/19420862.2023.2185924] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/24/2023] [Indexed: 03/08/2023] Open
Abstract
Large-molecule antibody biologics have revolutionized medicine owing to their superior target specificity, pharmacokinetic and pharmacodynamic properties, safety and toxicity profiles, and amenability to versatile engineering. In this review, we focus on preclinical antibody developability, including its definition, scope, and key activities from hit to lead optimization and selection. This includes generation, computational and in silico approaches, molecular engineering, production, analytical and biophysical characterization, stability and forced degradation studies, and process and formulation assessments. More recently, it is apparent these activities not only affect lead selection and manufacturability, but ultimately correlate with clinical progression and success. Emerging developability workflows and strategies are explored as part of a blueprint for developability success that includes an overview of the four major molecular properties that affect all developability outcomes: 1) conformational, 2) chemical, 3) colloidal, and 4) other interactions. We also examine risk assessment and mitigation strategies that increase the likelihood of success for moving the right candidate into the clinic.
Collapse
Affiliation(s)
- Carl Mieczkowski
- Department of Protein Sciences, Hengenix Biotech, Inc, Milpitas, CA, USA
| | - Xuejin Zhang
- Department of Protein Sciences, Hengenix Biotech, Inc, Milpitas, CA, USA
| | - Dana Lee
- Department of Protein Sciences, Hengenix Biotech, Inc, Milpitas, CA, USA
| | - Khanh Nguyen
- Department of Protein Sciences, Hengenix Biotech, Inc, Milpitas, CA, USA
| | - Wei Lv
- Department of Protein Sciences, Hengenix Biotech, Inc, Milpitas, CA, USA
| | - Yanling Wang
- Department of Protein Sciences, Hengenix Biotech, Inc, Milpitas, CA, USA
| | - Yue Zhang
- Department of Protein Sciences, Hengenix Biotech, Inc, Milpitas, CA, USA
| | - Jackie Way
- Department of Protein Sciences, Hengenix Biotech, Inc, Milpitas, CA, USA
| | - Jean-Michel Gries
- President, Discovery Research, Hengenix Biotech, Inc, Milpitas, CA, USA
| |
Collapse
|
31
|
Zhang W, Wang H, Feng N, Li Y, Gu J, Wang Z. Developability assessment at early-stage discovery to enable development of antibody-derived therapeutics. Antib Ther 2022; 6:13-29. [PMID: 36683767 PMCID: PMC9847343 DOI: 10.1093/abt/tbac029] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/13/2022] Open
Abstract
Developability refers to the likelihood that an antibody candidate will become a manufacturable, safe and efficacious drug. Although the safety and efficacy of a drug candidate will be well considered by sponsors and regulatory agencies, developability in the narrow sense can be defined as the likelihood that an antibody candidate will go smoothly through the chemistry, manufacturing and control (CMC) process at a reasonable cost and within a reasonable timeline. Developability in this sense is the focus of this review. To lower the risk that an antibody candidate with poor developability will move to the CMC stage, the candidate's developability-related properties should be screened, assessed and optimized as early as possible. Assessment of developability at the early discovery stage should be performed in a rapid and high-throughput manner while consuming small amounts of testing materials. In addition to monoclonal antibodies, bispecific antibodies, multispecific antibodies and antibody-drug conjugates, as the derivatives of monoclonal antibodies, should also be assessed for developability. Moreover, we propose that the criterion of developability is relative: expected clinical indication, and the dosage and administration route of the antibody could affect this criterion. We also recommend a general screening process during the early discovery stage of antibody-derived therapeutics. With the advance of artificial intelligence-aided prediction of protein structures and features, computational tools can be used to predict, screen and optimize the developability of antibody candidates and greatly reduce the risk of moving a suboptimal candidate to the development stage.
Collapse
Affiliation(s)
- Weijie Zhang
- Biologicals Innovation and Discovery, WuXi Biologicals, 1951 Huifeng West Road, Fengxian District, Shanghai 201400, China
| | - Hao Wang
- Biologicals Innovation and Discovery, WuXi Biologicals, 1951 Huifeng West Road, Fengxian District, Shanghai 201400, China
| | - Nan Feng
- Biologicals Innovation and Discovery, WuXi Biologicals, 1951 Huifeng West Road, Fengxian District, Shanghai 201400, China
| | - Yifeng Li
- Technology and Process Development, WuXi Biologicals, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Jijie Gu
- Biologicals Innovation and Discovery, WuXi Biologicals, 1951 Huifeng West Road, Fengxian District, Shanghai 201400, China
| | - Zhuozhi Wang
- To whom correspondence should be addressed. Biologics Innovation and Discovery, WuXi Biologicals, 1951 Huifeng West Road, Fengxian District, Shanghai 201400, China, Phone number: +86-21-50518899
| |
Collapse
|
32
|
Bana AAK, Mehta P, Ramnani KAK. Physical Instabilities of Therapeutic Monoclonal Antibodies: A Critical Review. Curr Drug Discov Technol 2022; 19:e240622206367. [PMID: 35748546 DOI: 10.2174/1570163819666220624092622] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/26/2022] [Accepted: 03/30/2022] [Indexed: 01/27/2023]
Abstract
The proteinaceous nature of monoclonal antibodies (mAbs) makes them highly sensitive to various physical and chemical conditions, thus leading to instabilities that are classified as physical and chemical instabilities. In this review, we are discussing in detail the physical instability of mAbs because a large number of articles previously published solely focus on the chemical aspect of the instability with little coverage on the physical side. The physical instabilities of mAbs are classified into denaturation and aggregation (precipitation, visible and subvisible particles). The mechanism involved in their formation is discussed in the article, along with the pathways correlating the denaturation of mAb or the formation of aggregates to immunogenicity. Further equations like Gibbs-Helmholtz involved in detecting and quantifying denaturation are discussed, along with various factors causing the denaturation. Moreover, questions related to aggregation like the types of aggregates and the pathway involved in their formation are answered in this article. Factors influencing the physical stability of the mAbs by causing denaturation or formation of aggregates involving the structure of the protein, concentration of mAbs, pH of the protein and the formulations, excipients involved in the formulations, salts added to the formulations, storage temperature, light and UV radiation exposure and processing factors are mentioned in this article. Finally, the analytical approaches used for detecting and quantifying the physical instability of mAbs at all levels of structural conformation like far and near UV, infrared spectroscopy, capillary electrophoresis, LC-MS, microflow imagining, circular dichroism and peptide mapping are discussed.
Collapse
Affiliation(s)
- Arpit Arun K Bana
- Department of Pharmaceutical Analysis, Institute of Pharmacy, Nirma University, Ahmedabad 382481, India
| | - Priti Mehta
- Department of Pharmaceutical Analysis, Institute of Pharmacy, Nirma University, Ahmedabad 382481, India
| | | |
Collapse
|
33
|
Abstract
The aggregation propensity of monoclonal antibodies can be modified by adding different cosolutes into the solution. A simple coarse-grained model in the combination with the thermodynamic perturbation theory was used to predict cluster distribution and viscosity of the solutions of IgG4 monoclonal anibody in the presence of L-Arginine Hydrochloride. The data were analysed using binding polynomial to describe the binding of cosolute (Arginine) to the antibody molecule. The results show that by binding to the antibody molecule the cosolute occupies some of the binding sites of the antibody, and in this way reduces the amount of binding sites available to other antibody molecules. The aggregation propensity of the antibody molecules is therefore reduced.
Collapse
|
34
|
Damerow H, Cheng X, von Kiedrowski V, Schirrmacher R, Wängler B, Fricker G, Wängler C. Toward Optimized 89Zr-Immuno-PET: Side-by-Side Comparison of [ 89Zr]Zr-DFO-, [ 89Zr]Zr-3,4,3-(LI-1,2-HOPO)- and [ 89Zr]Zr-DFO*-Cetuximab for Tumor Imaging: Which Chelator Is the Most Suitable? Pharmaceutics 2022; 14:pharmaceutics14102114. [PMID: 36297549 PMCID: PMC9611803 DOI: 10.3390/pharmaceutics14102114] [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: 09/16/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/21/2022] Open
Abstract
89Zr represents a highly favorable positron emitter for application in immuno-PET (Positron Emission Tomography) imaging. Clinically, the 89Zr4+ ion is introduced into antibodies by complexation with desferrioxamine B. However, producing complexes of limited kinetic inertness. Therefore, several new chelators for 89Zr introduction have been developed over the last years. Of these, the direct comparison of the most relevant ones for clinical translation, DFO* and 3,4,3-(LI-1,2-HOPO), is still missing. Thus, we directly compared DFO with DFO* and 3,4,3-(LI-1,2-HOPO) immunoconjugates to identify the most suitable agent stable 89Zr-complexation. The chelators were introduced into cetuximab, and an optical analysis method was developed, enabling the efficient quantification of derivatization sites per protein. The cetuximab conjugates were efficiently obtained and radiolabeled with 89Zr at 37 °C within 30 min, giving the [89Zr]Zr-cetuximab derivatives in high radiochemical yields and purities of >99% as well as specific activities of 50 MBq/mg. The immunoreactive fraction of all 89Zr-labeled cetuximab derivatives was determined to be in the range of 86.5−88.1%. In vivo PET imaging and ex vivo biodistribution studies in tumor-bearing animals revealed a comparable and significantly higher kinetic inertness for both [89Zr]Zr-3,4,3-(LI-1,2-HOPO)-cetuximab and [89Zr]Zr-DFO*-cetuximab, compared to [89Zr]Zr-DFO-cetuximab. Of these, [89Zr]Zr-DFO*-cetuximab showed a considerably more favorable pharmacokinetic profile with significantly lower liver and spleen retention than [89Zr]Zr-3,4,3-(LI-1,2-HOPO)-cetuximab. Since [89Zr]Zr-DFO* demonstrates a very high kinetic inertness, paired with a highly favorable pharmacokinetic profile of the resulting antibody conjugate, DFO* currently represents the most suitable chelator candidate for stable 89Zr-radiolabeling of antibodies and clinical translation.
Collapse
Affiliation(s)
- Helen Damerow
- Biomedical Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Xia Cheng
- Molecular Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Valeska von Kiedrowski
- Molecular Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Ralf Schirrmacher
- Division of Oncologic Imaging, Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Björn Wängler
- Molecular Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Carmen Wängler
- Biomedical Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Correspondence:
| |
Collapse
|
35
|
Conner CG, McAndrew J, Menegatti S, Velev OD. An accelerated antibody aggregation test based on time sequenced dynamic light scattering. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.129833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
36
|
Yin Y, Romei MG, Sankar K, Pal LR, Hon Hoi K, Yang Y, Leonard B, De Leon Boenig G, Kumar N, Matsumoto M, Payandeh J, Harris SF, Moult J, Lazar GA. Antibody Interfaces Revealed Through Structural Mining. Comput Struct Biotechnol J 2022; 20:4952-4968. [PMID: 36147680 PMCID: PMC9474289 DOI: 10.1016/j.csbj.2022.08.048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 11/15/2022] Open
Abstract
Antibodies are fundamental effectors of humoral immunity, and have become a highly successful class of therapeutics. There is increasing evidence that antibodies utilize transient homotypic interactions to enhance function, and elucidation of such interactions can provide insights into their biology and new opportunities for their optimization as drugs. Yet the transitory nature of weak interactions makes them difficult to investigate. Capitalizing on their rich structural data and high conservation, we have characterized all the ways that antibody fragment antigen-binding (Fab) regions interact crystallographically. This approach led to the discovery of previously unrealized interfaces between antibodies. While diverse interactions exist, β-sheet dimers and variable-constant elbow dimers are recurrent motifs. Disulfide engineering enabled interactions to be trapped and investigated structurally and functionally, providing experimental validation of the interfaces and illustrating their potential for optimization. This work provides first insight into previously undiscovered oligomeric interactions between antibodies, and enables new opportunities for their biotherapeutic optimization.
Collapse
|
37
|
Poly (ethylene) glycol (PEG) precipitation of glycosylated and non-glycosylated monoclonal antibodies. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.07.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
38
|
Xu T, Zhang J, Wang T, Wang X. Recombinant antibodies aggregation and overcoming strategies in CHO cells. Appl Microbiol Biotechnol 2022; 106:3913-3922. [PMID: 35608667 DOI: 10.1007/s00253-022-11977-0] [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: 02/01/2022] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 11/27/2022]
Abstract
Mammalian cell lines are frequently used as the preferred host cells for producing recombinant therapeutic proteins (RTPs) having post-translational modified modifications similar to those observed in proteins produced by human cells. Nowadays, most RTPs approved for marketing are produced in Chinese hamster ovary (CHO) cells. Recombinant therapeutic antibodies (RTAs) are among the most important and promising RTPs for biomedical applications. A major limitation associated with the use of RTAs is their aggregation, which can be caused by a variety of factors; this results in a reduction of quality. RTA aggregations are especially concerning as they can trigger human immune responses in humans and may be fatal. Therefore, the mechanisms underlying RTA aggregation and measures for avoiding aggregation are interesting topics in RTAs research. In this review, we discuss recent progress in the field of RTAs aggregation, with a focus on factors that cause aggregation during RTA production and the development of strategies for overcoming RTA aggregation. KEY POINTS: • The recombinant antibody aggregation in mammalian cell systems is reviewed. • Intracellular environment and extracellular parameters influence recombinant antibody aggregation. • Reducing the aggregations can improve the quality of recombinant antibodies.
Collapse
Affiliation(s)
- Tingting Xu
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, Henan, China.,The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, Henan, China
| | - Jihong Zhang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, Henan, China.,School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Tianyun Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, Henan, China. .,School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
| | - Xiaoyin Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, Henan, China. .,School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
| |
Collapse
|
39
|
Sun Z, Li W, Mellors JW, Orentas R, Dimitrov DS. Construction of a Large Size Human Immunoglobulin Heavy Chain Variable (VH) Domain Library, Isolation and Characterization of Novel Human Antibody VH Domains Targeting PD-L1 and CD22. Front Immunol 2022; 13:869825. [PMID: 35464476 PMCID: PMC9019674 DOI: 10.3389/fimmu.2022.869825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 03/16/2022] [Indexed: 12/03/2022] Open
Abstract
Phage display is a well-established technology for in vitro selection of monoclonal antibodies (mAb), and more than 12 antibodies isolated from phage displayed libraries of different formats have been approved for therapy. We have constructed a large size (10^11) human antibody VH domain library based on thermo-stable, aggregation-resistant scaffolds. This diversity was obtained by grafting naturally occurring CDR2s and CDR3s from healthy donors with optimized primers into the VH library. This phage-displayed library was used for bio-panning against various antigens. So far, panels of binders have been isolated against different viral and tumor targets, including the SARS-CoV-2 RBD, HIV-1 ENV protein, mesothelin and FLT3. In the present study, we discuss domain library construction, characterize novel VH binders against human CD22 and PD-L1, and define our design process for antibody domain drug conjugation (DDC) as tumoricidal reagents. Our study provides examples for the potential applications of antibody domains derived from library screens in therapeutics and provides key information for large size human antibody domain library construction.
Collapse
Affiliation(s)
- Zehua Sun
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, Pittsburgh, PA, United States
| | - Wei Li
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, Pittsburgh, PA, United States
| | - John W. Mellors
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, Pittsburgh, PA, United States
- Abound Bio, Pittsburgh, PA, United States
| | - Rimas Orentas
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA, United States
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
| | - Dimiter S. Dimitrov
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, Pittsburgh, PA, United States
- Abound Bio, Pittsburgh, PA, United States
| |
Collapse
|
40
|
Process- and Product-Related Foulants in Virus Filtration. Bioengineering (Basel) 2022; 9:bioengineering9040155. [PMID: 35447715 PMCID: PMC9030149 DOI: 10.3390/bioengineering9040155] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/01/2022] [Accepted: 04/01/2022] [Indexed: 11/16/2022] Open
Abstract
Regulatory authorities place stringent guidelines on the removal of contaminants during the manufacture of biopharmaceutical products. Monoclonal antibodies, Fc-fusion proteins, and other mammalian cell-derived biotherapeutics are heterogeneous molecules that are validated based on the production process and not on molecular homogeneity. Validation of clearance of potential contamination by viruses is a major challenge during the downstream purification of these therapeutics. Virus filtration is a single-use, size-based separation process in which the contaminating virus particles are retained while the therapeutic molecules pass through the membrane pores. Virus filtration is routinely used as part of the overall virus clearance strategy. Compromised performance of virus filters due to membrane fouling, low throughput and reduced viral clearance, is of considerable industrial significance and is frequently a major challenge. This review shows how components generated during cell culture, contaminants, and product variants can affect virus filtration of mammalian cell-derived biologics. Cell culture-derived foulants include host cell proteins, proteases, and endotoxins. We also provide mitigation measures for each potential foulant.
Collapse
|
41
|
Bhojane PP, Joshi S, Sahoo SJ, Rathore AS. Unexplored Excipients in Biotherapeutic Formulations: Natural Osmolytes as Potential Stabilizers Against Thermally Induced Aggregation of IgG1 Biotherapeutics. AAPS PharmSciTech 2021; 23:26. [PMID: 34907498 PMCID: PMC8670780 DOI: 10.1208/s12249-021-02183-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/17/2021] [Indexed: 11/30/2022] Open
Abstract
Monoclonal antibodies (mAbs), while incredibly successful, are prone to a variety of degradation pathways, the most significant of which is aggregation. One of the most commonly used strategy to overcome protein aggregation is addition of excipients to the formulation. Osmolytes such as trehalose, sucrose, and glycine are widely used. In this paper, we explore potential use of naturally occurring osmolytes such as betaine, sarcosine, ectoine, and hydroxyectoine for reducing aggregation of mAb therapeutics. Experimentation has been performed on two IgG1 mAbs via accelerated stability studies. A variety of analytical tools have been used for monitoring the impact, dynamic light scattering (DLS) for colloidal stability, Fourier transform infrared (FTIR) spectroscopy and fluorescence spectroscopy for conformational stability and the higher order structure (HOS), and differential scanning calorimetry (DSC) for thermal stability. No significant impact of osmolyte addition was observed on protein structure, on comparative Fc receptor (FcRn) binding, and on biocompatibility as per our hemolytic assay. Our results rank the osmolytes’ stabilizing trend to be sarcosine > betaine > hydroxyectoine > ectoine. Sarcosine emerged as the most successful osmolyte rendering highest degree of protection against aggregation. Our data support the prospect of using these osmolytes as successful excipients for mAb formulations.
Collapse
|
42
|
Abstract
The aggregation of therapeutic antibodies is a major issue for the pharmaceutical industry leading to loss of drug quality, increased dosage, and unwanted immune responses such as the production of anti-drug antibodies (ADA). As aggregation can occur at various stages of development and storage, much work has been performed to reduce or eliminate it. In this report we analyzed four antibodies available in the PDB (1IGT, 1IGY, 1HZH, and 5DK3) using the online software UCSF Chimera to study the structural features of the proteins and the associated N-linked glycans in the CH2 domains of the Fc region. To study antibody aggregation in silico we used the online software TANGO and AGGRESCAN to identify aggregation prone regions (APR) in the antibodies and the influence of the Fc glycans on hydrophobic and aromatic residues present in the APRs. In the 3D structures of 1IGT and 1IGY the glycan chains are in close enough proximity to influence and protect these hydrophobic regions. However, in the 3D structures of 1HZH and 5DK3 the glycans do not appear to influence the likely APRs of the antibodies. Therefore, in these structures we modified the Fc glycan regions by adjusting the glycosylated asparagine side chains and glycosidic bonds. We successfully adjusted the glycan chains of 1HZH and 5DK3 and reduced the distance between them and the APRs to show potential influence on aggregation. However, similar to 5DK3, the influence of glycosylation on the APRs of the antibody was limited due to the size of the glycans present in the 3D structure. This report is based on in silico studies to show how antibody glycans can influence aggregation.
Collapse
|
43
|
Hirschman J, Venkataramani D, Murphy MI, Patel SM, Du J, Amin S. Application of thin gap rheometry for high shear rate viscosity measurement in monoclonal antibody formulations. Colloids Surf A Physicochem Eng Asp 2021. [DOI: 10.1016/j.colsurfa.2021.127018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
44
|
Yeoh SG, Sum JS, Lai JY, W Isa WYH, Lim TS. Potential of Phage Display Antibody Technology for Cardiovascular Disease Immunotherapy. J Cardiovasc Transl Res 2021; 15:360-380. [PMID: 34467463 DOI: 10.1007/s12265-021-10169-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/22/2021] [Indexed: 11/26/2022]
Abstract
Cardiovascular disease (CVD) is one of the leading causes of death worldwide. CVD includes coronary artery diseases such as angina, myocardial infarction, and stroke. "Lipid hypothesis" which is also known as the cholesterol hypothesis proposes the linkage of plasma cholesterol level with the risk of developing CVD. Conventional management involves the use of statins to reduce the serum cholesterol levels as means for CVD prevention or treatment. The regulation of serum cholesterol levels can potentially be regulated with biological interventions like monoclonal antibodies. Phage display is a powerful tool for the development of therapeutic antibodies with successes over the recent decade. Although mainly for oncology, the application of monoclonal antibodies as immunotherapeutic agents could potentially be expanded to CVD. This review focuses on the concept of phage display for antibody development and discusses the potential target antigens that could potentially be beneficial for serum cholesterol management.
Collapse
Affiliation(s)
- Soo Ghee Yeoh
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Jia Siang Sum
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Jing Yi Lai
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - W Y Haniff W Isa
- School of Medical Sciences, Department of Medicine, Universiti Sains Malaysia, Kubang Kerian, 16150, Kelantan, Malaysia
| | - Theam Soon Lim
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800, Penang, Malaysia.
- Analytical Biochemistry Research Centre, Universiti Sains Malaysia, 11800, Penang, Malaysia.
| |
Collapse
|
45
|
Lee AP, Kok YJ, Lakshmanan M, Leong D, Zheng L, Lim HL, Chen S, Mak SY, Ang KS, Templeton N, Salim T, Wei X, Gifford E, Tan AHM, Bi X, Ng SK, Lee DY, Ling WLW, Ho YS. Multi-omics profiling of a CHO cell culture system unravels the effect of culture pH on cell growth, antibody titer, and product quality. Biotechnol Bioeng 2021; 118:4305-4316. [PMID: 34289087 DOI: 10.1002/bit.27899] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 06/23/2021] [Accepted: 07/12/2021] [Indexed: 12/16/2022]
Abstract
A robust monoclonal antibody (mAb) bioprocess requires physiological parameters such as temperature, pH, or dissolved oxygen to be well-controlled as even small variations in them could potentially impact the final product quality. For instance, pH substantially affects N-glycosylation, protein aggregation, and charge variant profiles, as well as mAb productivity. However, relatively less is known about how pH jointly influences product quality and titer. In this study, we investigated the effect of pH on culture performance, product titer, and quality profiles by applying longitudinal multi-omics profiling, including transcriptomics, proteomics, metabolomics, and glycomics, at three different culture pH set points. The subsequent systematic analysis of multi-omics data showed that pH set points differentially regulated various intracellular pathways including intracellular vesicular trafficking, cell cycle, and apoptosis, thereby resulting in differences in specific productivity, product titer, and quality profiles. In addition, a time-dependent variation in mAb N-glycosylation profiles, independent of pH, was identified to be mainly due to the accumulation of mAb proteins in the endoplasmic reticulum disrupting cellular homeostasis over culture time. Overall, this multi-omics-based study provides an in-depth understanding of the intracellular processes in mAb-producing CHO cell line under varied pH conditions, and could serve as a baseline for enabling the quality optimization and control of mAb production.
Collapse
Affiliation(s)
- Alison P Lee
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yee Jiun Kok
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Meiyappan Lakshmanan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Dawn Leong
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Lu Zheng
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Hsueh Lee Lim
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Shuwen Chen
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Shi Ya Mak
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Kok Siong Ang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Neil Templeton
- Process Research and Development, Merck & Co. Inc., West Point, Pennsylvania, USA
| | - Taha Salim
- Process Research and Development, Merck & Co. Inc., Kenilworth, New Jersey, USA
| | - Xiaona Wei
- Scientific Informatics, MSD International GmbH (Singapore Branch), Singapore, Singapore
| | - Eric Gifford
- Scientific Informatics, MSD International GmbH (Singapore Branch), Singapore, Singapore
| | - Andy Hee-Meng Tan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Xuezhi Bi
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Say Kong Ng
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Dong-Yup Lee
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.,School of Chemical Engineering, Sungkyunkwan University, Seoul, Gyeonggi-do, Republic of Korea
| | - Wai Lam W Ling
- Process Research and Development, Merck & Co. Inc., Kenilworth, New Jersey, USA
| | - Ying Swan Ho
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
46
|
Sadeghian I, Hemmati S. Characterization of a Stable Form of Carboxypeptidase G2 (Glucarpidase), a Potential Biobetter Variant, From Acinetobacter sp. 263903-1. Mol Biotechnol 2021; 63:1155-1168. [PMID: 34268672 DOI: 10.1007/s12033-021-00370-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/08/2021] [Indexed: 01/14/2023]
Abstract
Carboxypeptidase G2 (CPG2) is a bacterial enzyme widely used to detoxify methotrexate (MTX) and in enzyme/prodrug therapy for cancer treatment. However, several drawbacks, such as instability, have limited its efficiency. Herein, we have evaluated the properties of a putative CPG2 from Acinetobacter sp. 263903-1 (AcCPG2). AcCPG2 is compared with a CPG2 derived from Pseudomonas sp. strain RS-16 (PsCPG2), available as an FDA-approved medication called glucarpidase. After modeling AcCPG2 using the I-TASSER program, the refined model was validated by PROCHECK, VERIFY 3D and according to the Z score of the model. Using computational analyses, AcCPG2 displayed higher thermodynamic stability and a lower aggregation propensity than PsCPG2. AcCPG2 showed an optimum pH of 7.5 against MTX and was stable over a pH range of 5-10. AcCPG2 exhibited optimum activity at 50 °C and higher thermal stability at a temperature range of 20-70 °C compared to PsCPG2. The Km value of the purified AcCPG2 toward folate and MTX was 31.36 µM and 44.99 µM, respectively. The Vmax value of AcCPG2 for folate and MTX was 125.80 µmol/min/mg and 48.90 µmol/min/mg, respectively. Accordingly, thermostability and pH versatility makes AcCPG2 a potential biobetter variant for therapeutic applications.
Collapse
Affiliation(s)
- Issa Sadeghian
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shiva Hemmati
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
47
|
A structural perspective on the design of decoy immune modulators. Pharmacol Res 2021; 170:105735. [PMID: 34146695 DOI: 10.1016/j.phrs.2021.105735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/23/2021] [Accepted: 06/15/2021] [Indexed: 11/22/2022]
Abstract
Therapeutic mAbs have dominated the class of immunotherapeutics in general and immune checkpoint inhibitors in particular. The high specificity of mAbs to the target molecule as well as their extended half-life and (or) the effector functions raised by the Fc part are some of the important aspects that contribute to the success of this class of therapeutics. Equally potential candidates are decoys and their fusions that can address some of the inherent limitations of mAbs, like immunogenicity, resistance development, low bio-availability and so on, besides maintaining the advantages of mAbs. The decoys are molecules that trap the ligands and prevent them from interacting with the signaling receptors. Although a few FDA-approved decoy immune modulators are very successful, the potential of this class of drugs is yet to be fully realized. Here, we review various strategies employed in fusion protein therapeutics with a focus on the design of decoy immunomodulators from the structural perspective and discuss how the information on protein structure and function can strategically guide the development of next-generation immune modulators.
Collapse
|
48
|
Ren T, Tan Z, Ehamparanathan V, Lewandowski A, Ghose S, Li ZJ. Antibody disulfide bond reduction and recovery during biopharmaceutical process development-A review. Biotechnol Bioeng 2021; 118:2829-2844. [PMID: 33844277 DOI: 10.1002/bit.27790] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 12/29/2022]
Abstract
Antibody disulfide bond reduction has been a challenging issue in monoclonal antibody manufacturing. It could lead to a decrease of product purity and failure to meet the targeted product profile and/or specifications. More importantly, disulfide bond reduction could also impact drug safety and efficacy. Scientists across the industry have been examining the root causes and developing mitigation strategies to address the challenge. In recent years, with the development of high titer mammalian cell culture processes to meet the rapidly growing demand for antibody biopharmaceuticals, disulfide bond reduction has been observed more frequently. Thus, it is necessary to continue evolving the disulfide reduction mitigation strategies and developing novel approaches to maintain high product quality. Additionally, in recent years as more complex molecules (such as bispecific and trispecific antibodies) emerge, the molecular heterogeneity due to incomplete formation of the interchain disulfide bonds becomes a more imperative challenging issue. Given the disulfide reduction challenges that biotech industry is facing, in this review, we provide a comprehensive scientific summary of the root cause analysis of disulfide reduction during process development of antibody therapeutics, mitigation strategies and its potential remediated recovery based on published papers. First, this paper intends to highlight different aspects of the root cause for disulfide reduction. Secondly, to provide a broader understanding of the disulfide bond reduction in downstream process, this paper discusses disulfide bond reduction impact on product stability, associated analytical methods for disulfide bond reduction detection and characterization, process control strategies as well as their manufacturing implementation. In addition, brief perspectives on the development of future mitigation strategies are also reviewed, including platform alignment, mitigation strategy application for the emerging new modalities such as bispecific and trispecific antibodies as well as using machine learning to identify molecule susceptibility of disulfide bond reduction. The data in this review are originated from the published papers.
Collapse
Affiliation(s)
- Tingwei Ren
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, Massachusetts
| | - Zhijun Tan
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, Massachusetts
| | - Vivekh Ehamparanathan
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, Massachusetts
| | - Angela Lewandowski
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, Massachusetts
| | - Sanchayita Ghose
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, Massachusetts
| | - Zheng Jian Li
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, Massachusetts
| |
Collapse
|
49
|
Carrara SC, Fiebig D, Bogen JP, Grzeschik J, Hock B, Kolmar H. Recombinant Antibody Production Using a Dual-Promoter Single Plasmid System. ANTIBODIES (BASEL, SWITZERLAND) 2021; 10:antib10020018. [PMID: 34068440 PMCID: PMC8161450 DOI: 10.3390/antib10020018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/29/2021] [Accepted: 05/10/2021] [Indexed: 01/21/2023]
Abstract
Monoclonal antibodies (mAbs) have demonstrated tremendous effects on the treatment of various disease indications and remain the fastest growing class of therapeutics. Production of recombinant antibodies is performed using mammalian expression systems to facilitate native antibody folding and post-translational modifications. Generally, mAb expression systems utilize co-transfection of heavy chain (hc) and light chain (lc) genes encoded on separate plasmids. In this study, we examine the production of two FDA-approved antibodies using a bidirectional (BiDi) vector encoding both hc and lc with mirrored promoter and enhancer elements on a single plasmid, by analysing the individual hc and lc mRNA expression levels and subsequent quantification of fully-folded IgGs on the protein level. From the assessment of different promoter combinations, we have developed a generic expression vector comprised of mirrored enhanced CMV (eCMV) promoters showing comparable mAb yields to a two-plasmid reference. This study paves the way to facilitate small-scale mAb production by transient cell transfection with a single vector in a cost- and time-efficient manner.
Collapse
Affiliation(s)
- Stefania C. Carrara
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Str. 4, D-64287 Darmstadt, Germany; (S.C.C.); (D.F.); (J.P.B.)
- Ferring Darmstadt Laboratories, Alarich-Weiss-Str. 4, D-64287 Darmstadt, Germany;
| | - David Fiebig
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Str. 4, D-64287 Darmstadt, Germany; (S.C.C.); (D.F.); (J.P.B.)
- Ferring Darmstadt Laboratories, Alarich-Weiss-Str. 4, D-64287 Darmstadt, Germany;
| | - Jan P. Bogen
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Str. 4, D-64287 Darmstadt, Germany; (S.C.C.); (D.F.); (J.P.B.)
- Ferring Darmstadt Laboratories, Alarich-Weiss-Str. 4, D-64287 Darmstadt, Germany;
| | - Julius Grzeschik
- Ferring Darmstadt Laboratories, Alarich-Weiss-Str. 4, D-64287 Darmstadt, Germany;
| | - Björn Hock
- Ferring International Center S.A, Chemin de la Vergognausaz 50, CH-1162 Saint Prex, Switzerland;
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Str. 4, D-64287 Darmstadt, Germany; (S.C.C.); (D.F.); (J.P.B.)
- Correspondence:
| |
Collapse
|
50
|
Ashford MB, England RM, Akhtar N. Highway to Success—Developing Advanced Polymer Therapeutics. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000285] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Marianne B. Ashford
- Advanced Drug Delivery Pharmaceutical Sciences, R&D, AstraZeneca Macclesfield SK10 2NA UK
| | - Richard M. England
- Advanced Drug Delivery Pharmaceutical Sciences, R&D, AstraZeneca Macclesfield SK10 2NA UK
| | - Nadim Akhtar
- New Modalities & Parenteral Development Pharmaceutical Technology & Development, Operations, AstraZeneca Macclesfield SK10 2NA UK
| |
Collapse
|