1
|
Pais DAM, Mayer JPA, Felderer K, Batalha MB, Eichner T, Santos ST, Kumar R, Silva SD, Kaufmann H. Holistic in silico developability assessment of novel classes of small proteins using publicly available sequence-based predictors. J Comput Aided Mol Des 2024; 38:30. [PMID: 39164492 DOI: 10.1007/s10822-024-00569-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: 01/16/2024] [Accepted: 07/26/2024] [Indexed: 08/22/2024]
Abstract
The development of novel therapeutic proteins is a lengthy and costly process, with an average attrition rate of 91% (Thomas et al. Clinical Development Success Rates and Contributing Factors 2011-2020, 2021). To increase the probability of success and ensure robust drug supply beyond approval, it is essential to assess the developability profile of new potential drug candidates as early and broadly as possible in development (Jain et al. MAbs, 2023. https://doi.org/10.1016/j.copbio.2011.06.002 ). Predicting these properties in silico is expected to be the next leap in innovation as it would enable significantly reduced development timelines combined with broader screens at lower costs. However, developing predictive algorithms typically requires substantial datasets generated under very defined conditions, a limiting factor especially for new classes of therapeutic proteins that hold immense clinical promise. Here we describe a strategy for assessing the developability of a novel class of small therapeutic Anticalin® proteins using machine learning in conjunction with a knowledge-driven approach. The knowledge-driven approach considers developability attributes such as aggregation propensity, charge variants, immunogenicity, specificity, thermal stability, hydrophobicity, and potential post-translational modifications, to calculate a holistic developability score. Based on sequence-derived descriptors as input parameters we established novel statistical models designed to predict the developability scores for Anticalin proteins. The best models yielded low root mean square errors across the entire dataset and were further validated by removing input data from individual screening campaigns and predicting developability scores for those drug candidates. The adoption of the described workflow will enable significantly streamlined preclinical development of Anticalin drug candidates and could potentially be applied to other therapeutic protein scaffolds.
Collapse
Affiliation(s)
- Daniel A M Pais
- Valgenesis Portugal, Lda, R. Castilho 50 4th Floor, 1250-071, Lisbon, Portugal
| | - Jan-Peter A Mayer
- Pieris Pharmaceuticals GmbH, Carl-Zeiss-Ring 15a, 85737, Ismaning, Germany
| | - Karin Felderer
- Pieris Pharmaceuticals GmbH, Carl-Zeiss-Ring 15a, 85737, Ismaning, Germany
| | - Maria B Batalha
- Valgenesis Portugal, Lda, R. Castilho 50 4th Floor, 1250-071, Lisbon, Portugal
| | - Timo Eichner
- Pieris Pharmaceuticals GmbH, Carl-Zeiss-Ring 15a, 85737, Ismaning, Germany
| | - Sofia T Santos
- Valgenesis Portugal, Lda, R. Castilho 50 4th Floor, 1250-071, Lisbon, Portugal
| | - Raman Kumar
- Pieris Pharmaceuticals GmbH, Carl-Zeiss-Ring 15a, 85737, Ismaning, Germany
| | - Sandra D Silva
- Valgenesis Portugal, Lda, R. Castilho 50 4th Floor, 1250-071, Lisbon, Portugal
| | - Hitto Kaufmann
- Pieris Pharmaceuticals GmbH, Carl-Zeiss-Ring 15a, 85737, Ismaning, Germany.
| |
Collapse
|
2
|
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
|
3
|
Gavade A, Nagraj AK, Patel R, Pais R, Dhanure P, Scheele J, Seiz W, Patil J. Understanding the Specific Implications of Amino Acids in the Antibody Development. Protein J 2024; 43:405-424. [PMID: 38724751 DOI: 10.1007/s10930-024-10201-4] [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] [Accepted: 04/21/2024] [Indexed: 06/01/2024]
Abstract
As the demand for immunotherapy to treat and manage cancers, infectious diseases and other disorders grows, a comprehensive understanding of amino acids and their intricate role in antibody engineering has become a prime requirement. Naturally produced antibodies may not have the most suitable amino acids at the complementarity determining regions (CDR) and framework regions, for therapeutic purposes. Therefore, to enhance the binding affinity and therapeutic properties of an antibody, the specific impact of certain amino acids on the antibody's architecture must be thoroughly studied. In antibody engineering, it is crucial to identify the key amino acid residues that significantly contribute to improving antibody properties. Therapeutic antibodies with higher binding affinity and improved functionality can be achieved through modifications or substitutions with highly suitable amino acid residues. Here, we have indicated the frequency of amino acids and their association with the binding free energy in CDRs. The review also analyzes the experimental outcome of two studies that reveal the frequency of amino acids in CDRs and provides their significant correlation between the outcomes. Additionally, it discusses the various bond interactions within the antibody structure and antigen binding. A detailed understanding of these amino acid properties should assist in the analysis of antibody sequences and structures needed for designing and enhancing the overall performance of therapeutic antibodies.
Collapse
Affiliation(s)
- Akshata Gavade
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Anil Kumar Nagraj
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Riya Patel
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Roylan Pais
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Pratiksha Dhanure
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India
| | | | | | - Jaspal Patil
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India.
| |
Collapse
|
4
|
Amash A, Volkers G, Farber P, Griffin D, Davison KS, Goodman A, Tonikian R, Yamniuk A, Barnhart B, Jacobs T. Developability considerations for bispecific and multispecific antibodies. MAbs 2024; 16:2394229. [PMID: 39189686 DOI: 10.1080/19420862.2024.2394229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/08/2024] [Accepted: 08/15/2024] [Indexed: 08/28/2024] Open
Abstract
Bispecific antibodies (bsAb) and multispecific antibodies (msAb) encompass a diverse variety of formats that can concurrently bind multiple epitopes, unlocking mechanisms to address previously difficult-to-treat or incurable diseases. Early assessment of candidate developability enables demotion of antibodies with low potential and promotion of the most promising candidates for further development. Protein-based therapies have a stringent set of developability requirements in order to be competitive (e.g. high-concentration formulation, and long half-life) and their assessment requires a robust toolkit of methods, few of which are validated for interrogating bsAbs/msAbs. Important considerations when assessing the developability of bsAbs/msAbs include their molecular format, likelihood for immunogenicity, specificity, stability, and potential for high-volume production. Here, we summarize the critical aspects of developability assessment, and provide guidance on how to develop a comprehensive plan tailored to a given bsAb/msAb.
Collapse
Affiliation(s)
- Alaa Amash
- AbCellera Biologics Inc, Vancouver, BC, Canada
| | | | | | | | | | | | | | | | | | - Tim Jacobs
- AbCellera Biologics Inc, Vancouver, BC, Canada
| |
Collapse
|
5
|
Arsiccio A, Stratta L, Menzen T. Evaluating the chaos game representation of proteins for applications in machine learning models: prediction of antibody affinity and specificity as a case study. J Mol Model 2023; 29:377. [PMID: 37968495 DOI: 10.1007/s00894-023-05777-0] [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: 09/21/2023] [Accepted: 10/31/2023] [Indexed: 11/17/2023]
Abstract
CONTEXT Machine learning techniques are becoming increasingly important in the selection and optimization of therapeutic molecules, as well as for the selection of formulation components and the prediction of long-term stability. Compared to first-principle models, machine learning techniques are easier to implement, and can identify correlations that would be hard to describe at a mechanistic level, but strongly rely on high-quality input training data. Here, we evaluate the potential of the "chaos game" representation to provide input data for machine learning models. The chaos game is an algorithm originally developed for the production of fractal structures, and later on applied also to the representation of biological sequences, such as genes and proteins. Our results show that the combination of the chaos game representation with convolutional neural networks results in comparable accuracy to other machine learning approaches, thus indicating that chaos game representations could be a valid alternative to existing featurization strategies for machine learning models of biological sequences. METHODS We implement the chaos game in Python 3.8.10, and use it to produce fractal as well as novel expanding representations of protein sequences. We then feed the resulting images to a convolutional neural network, built in Python 3.8.10, using TensorFlow 2.9.1, Keras 2.9.0, and the scikit-learn 1.1.1 packages. We select as case study a recently published dataset for the antibody emibetuzumab, with the objective of co-optimizing antibodies variants with both high affinity and low non-specific binding.
Collapse
Affiliation(s)
- Andrea Arsiccio
- Coriolis Pharma, Fraunhoferstrasse 18 b, 82152, Martinsried, Germany.
| | - Lorenzo Stratta
- Molecular Engineering Laboratory (molE), Department of Applied Science and Technology, Politecnico di Torino, 24 corso Duca degli Abruzzi, IT-10129, Torino, Italy
| | - Tim Menzen
- Coriolis Pharma, Fraunhoferstrasse 18 b, 82152, Martinsried, Germany
| |
Collapse
|
6
|
Levin I, Štrajbl M, Fastman Y, Baran D, Twito S, Mioduser J, Keren A, Fischman S, Zhenin M, Nimrod G, Levitin N, Mayor MB, Gadrich M, Ofran Y. Accurate profiling of full-length Fv in highly homologous antibody libraries using UMI tagged short reads. Nucleic Acids Res 2023; 51:e61. [PMID: 37014016 PMCID: PMC10287906 DOI: 10.1093/nar/gkad235] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/14/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
Deep parallel sequencing (NGS) is a viable tool for monitoring scFv and Fab library dynamics in many antibody engineering high-throughput screening efforts. Although very useful, the commonly used Illumina NGS platform cannot handle the entire sequence of scFv or Fab in a single read, usually focusing on specific CDRs or resorting to sequencing VH and VL variable domains separately, thus limiting its utility in comprehensive monitoring of selection dynamics. Here we present a simple and robust method for deep sequencing repertoires of full length scFv, Fab and Fv antibody sequences. This process utilizes standard molecular procedures and unique molecular identifiers (UMI) to pair separately sequenced VH and VL. We show that UMI assisted VH-VL matching allows for a comprehensive and highly accurate mapping of full length Fv clonal dynamics in large highly homologous antibody libraries, as well as identification of rare variants. In addition to its utility in synthetic antibody discovery processes, our method can be instrumental in generating large datasets for machine learning (ML) applications, which in the field of antibody engineering has been hampered by conspicuous paucity of large scale full length Fv data.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Adi Keren
- Biolojic Design, Ltd, Rehovot, Israel
| | | | | | | | | | | | | | - Yanay Ofran
- Biolojic Design, Ltd, Rehovot, Israel
- The Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan, Israel
| |
Collapse
|
7
|
Vincent MS, Ezraty B. Methionine oxidation in bacteria: A reversible post-translational modification. Mol Microbiol 2023; 119:143-150. [PMID: 36350090 DOI: 10.1111/mmi.15000] [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: 08/29/2022] [Revised: 11/04/2022] [Accepted: 11/05/2022] [Indexed: 11/10/2022]
Abstract
Methionine is a sulfur-containing residue found in most proteins which are particularly susceptible to oxidation. Although methionine oxidation causes protein damage, it can in some cases activate protein function. Enzymatic systems reducing oxidized methionine have evolved in most bacterial species and methionine oxidation proves to be a reversible post-translational modification regulating protein activity. In this review, we inspect recent examples of methionine oxidation provoking protein loss and gain of function. We further speculate on the role of methionine oxidation as a multilayer endogenous antioxidant system and consider its potential consequences for bacterial virulence.
Collapse
Affiliation(s)
- Maxence S Vincent
- Laboratoire de Chimie Bactérienne, Institut de Microbiologie de la Méditerranée, Aix-Marseille University, CNRS, Marseille, France
| | - Benjamin Ezraty
- Laboratoire de Chimie Bactérienne, Institut de Microbiologie de la Méditerranée, Aix-Marseille University, CNRS, Marseille, France
| |
Collapse
|
8
|
Jain T, Boland T, Vásquez M. Identifying developability risks for clinical progression of antibodies using high-throughput in vitro and in silico approaches. MAbs 2023; 15:2200540. [PMID: 37072706 PMCID: PMC10114995 DOI: 10.1080/19420862.2023.2200540] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/20/2023] Open
Abstract
With the growing significance of antibodies as a therapeutic class, identifying developability risks early during development is of paramount importance. Several high-throughput in vitro assays and in silico approaches have been proposed to de-risk antibodies during early stages of the discovery process. In this review, we have compiled and collectively analyzed published experimental assessments and computational metrics for clinical antibodies. We show that flags assigned based on in vitro measurements of polyspecificity and hydrophobicity are more predictive of clinical progression than their in silico counterparts. Additionally, we assessed the performance of published models for developability predictions on molecules not used during model training. We find that generalization to data outside of those used for training remains a challenge for models. Finally, we highlight the challenges of reproducibility in computed metrics arising from differences in homology modeling, in vitro assessments relying on complex reagents, as well as curation of experimental data often used to assess the utility of high-throughput approaches. We end with a recommendation to enable assay reproducibility by inclusion of controls with disclosed sequences, as well as sharing of structural models to enable the critical assessment and improvement of in silico predictions.
Collapse
Affiliation(s)
| | - Todd Boland
- Computational Biology, Adimab LLC, Lebanon, NH, USA
| | | |
Collapse
|
9
|
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
|
10
|
Tavella D, Ouellette DR, Garofalo R, Zhu K, Xu J, Oloo EO, Negron C, Ihnat PM. A novel method for in silico assessment of Methionine oxidation risk in monoclonal antibodies: Improvement over the 2-shell model. PLoS One 2022; 17:e0279689. [PMID: 36580468 PMCID: PMC9799309 DOI: 10.1371/journal.pone.0279689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 12/13/2022] [Indexed: 12/30/2022] Open
Abstract
Over the past decade, therapeutic monoclonal antibodies (mAbs) have established their role as valuable agents in the treatment of various diseases ranging from cancers to infectious, cardiovascular and autoimmune diseases. Reactive groups of the amino acids within these proteins make them susceptible to many kinds of chemical modifications during manufacturing, storage and in vivo circulation. Among these reactions, the oxidation of methionine residues to their sulfoxide form is a commonly observed chemical modification in mAbs. When the oxidized methionine is in the complementarity-determining region (CDR), this modification can affect antigen binding and thus abrogate biological activity. For these reasons, it is essential to identify oxidation liabilities during the antibody discovery and development phases. Here, we present an in silico method, based on protein modeling and molecular dynamics simulations, to predict the oxidation-liable residues in the variable region of therapeutic antibodies. Previous studies have used the 2-shell water coordination number descriptor (WCN) to identify methionine residues susceptible to oxidation. Although the WCN descriptor successfully predicted oxidation liabilities when the residue was solvent exposed, the method was much less accurate for partially buried methionine residues. Consequently, we introduce a new descriptor, WCN-OH, that improves the accuracy of prediction of methionine oxidation susceptibility by extending the theoretical framework of the water coordination number to incorporate the effects of polar amino acids side chains in close proximity to the methionine of interest.
Collapse
Affiliation(s)
- Davide Tavella
- AbbVie Bioresearch Center, Worcester, Massachusetts, United States of America
- * E-mail: (DT); (CN)
| | - David R. Ouellette
- AbbVie Bioresearch Center, Worcester, Massachusetts, United States of America
| | - Raffaella Garofalo
- AbbVie Deutschland GmbH & Co. KG, Analytical Innovation and Mass Spectrometry, Knollstrasse, Ludwigshafen, Germany
| | - Kai Zhu
- Schrödinger, Inc., New York, New York, United States of America
| | - Jianwen Xu
- AbbVie Bioresearch Center, Worcester, Massachusetts, United States of America
| | - Eliud O. Oloo
- Schrödinger, Inc., New York, New York, United States of America
| | - Christopher Negron
- AbbVie Bioresearch Center, Worcester, Massachusetts, United States of America
- * E-mail: (DT); (CN)
| | - Peter M. Ihnat
- Regeneron Pharmaceuticals Inc., Tarrytown, New York, United States of America
| |
Collapse
|
11
|
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
|
12
|
Puranik A, Dandekar P, Jain R. Exploring the potential of machine learning for more efficient development and production of biopharmaceuticals. Biotechnol Prog 2022; 38:e3291. [PMID: 35918873 DOI: 10.1002/btpr.3291] [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: 03/26/2022] [Revised: 06/20/2022] [Accepted: 07/31/2022] [Indexed: 11/10/2022]
Abstract
Principles of Industry 4.0 direct us to predict how pharmaceutical operations and regulations may exist with automation, digitization, artificial intelligence (AI), and real time data acquisition. Machine learning (ML), a sub-discipline of AI, involves the use of statistical tools to extract the desired information either through understanding the underlying patterns in the information or by development of mathematical relationships among the critical process parameters (CPPs) and critical quality attributes (CQAs) of biopharmaceuticals. ML is still in its infancy for directly supporting the quality-by-design based development and manufacturing of biopharmaceuticals. However, adoption of ML-based models in place of conventional multi-variate-data-analysis (MVDA) is increasing with the accumulation of large-scale data. This has been majorly contributed by the real-time monitoring of process variables and quality attributes of products through the implementation of process analytical technology in biopharmaceutical manufacturing. All aspects of healthcare, from drug design to product distribution, are complex and multidimensional. Thus, ML-based approaches are being applied to achieve sophistication, accuracy, flexibility and agility in all these areas. This review discusses the potential of ML for addressing the complex issues in diverse areas of biopharmaceutical development, such as biopharmaceuticals design and assessment of early stage development, upstream and downstream process development, analysis, characterization and prediction of post translational modifications (PTMs), formulation and stability studies. Moreover, the challenges in acquisition, cleaning and structuring the bioprocess data, which is one of the major hurdles in implementation of ML in biopharma industry, have also been discussed. Regulatory perspectives on implementation of AI/ML in the biopharma sector have also been briefly discussed. This article is a bird's eye view on the recent developments and applications of ML in overcoming the challenges for adopting "Industry - 4.0" in the biopharma industry.
Collapse
Affiliation(s)
- Amita Puranik
- Department of Chemical Engineering, Institute of Chemical Technology, Matunga, Mumbai, India
| | - Prajakta Dandekar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Matunga, Mumbai, India
| | - Ratnesh Jain
- Department of Chemical Engineering, Institute of Chemical Technology, Matunga, Mumbai, India
| |
Collapse
|
13
|
Suresh SA, Ethiraj S, Rajnish KN. A systematic review of recent trends in research on therapeutically significant L-asparaginase and acute lymphoblastic leukemia. Mol Biol Rep 2022; 49:11281-11287. [PMID: 35816224 DOI: 10.1007/s11033-022-07688-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/08/2022] [Indexed: 12/01/2022]
Abstract
L-asparaginases are mostly obtained from bacterial sources for their application in the therapy and food industry. Bacterial L-asparaginases are employed in the treatment of Acute Lymphoblastic Leukemia (ALL) and its subtypes, a type of blood and bone marrow cancer that results in the overproduction of immature blood cells. It also plays a role in the food industry in reducing the acrylamide formed during baking, roasting, and frying starchy foods. This importance of the enzyme makes it to be of constant interest to the researchers to isolate novel sources. Presently L-asparaginases from E. coli native and PEGylated form, Dickeya chrysanthemi (Erwinia chrysanthemi) are in the treatment regime. In therapy, the intrinsic glutaminase activity of the enzyme is a major drawback as the patients in treatment experience side effects like fever, skin rashes, anaphylaxis, pancreatitis, steatosis in the liver, and many complications. Its significance in the food industry in mitigating acrylamide is also a major reason. Acrylamide, a potent carcinogen was formed when treating starchy foods at higher temperatures. Acrylamide content in food was analyzed and pre-treatment was considered a valuable option. Immobilization of the enzyme is an advancing and promising technique in the effective delivery of the enzyme than in free form. The concept of machine learning by employing the Artificial Network and Genetic Algorithm has paved the way to optimize the production of L-asparaginase from its sources. Gene-editing tools are gaining momentum in the study of several diseases and this review focuses on the CRISPR-Cas9 gene-editing tool in ALL.
Collapse
Affiliation(s)
| | | | - K N Rajnish
- SRM Institute of Science and Technology, Chennai, Tamil Nadu, India.
| |
Collapse
|
14
|
Jacobitz AW, Rodezno W, Agrawal NJ. Utilizing cross-product prior knowledge to rapidly de-risk chemical liabilities in therapeutic antibody candidates. AAPS OPEN 2022. [DOI: 10.1186/s41120-022-00057-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AbstractThere is considerable pressure in the pharmaceutical industry to advance better molecules faster. One pervasive concern for protein-based therapeutics is the presence of potential chemical liabilities. We have developed a simple methodology for rapidly de-risking specific chemical concerns in antibody-based molecules using prior knowledge of each individual liability at a specific position in the molecule’s sequence. Our methodology hinges on the development of sequence-aligned chemical liability databases of molecules from different stages of commercialization and on sequence-aligned experimental data from prior molecules that have been developed at Amgen. This approach goes beyond the standard practice of simply flagging all instances of each motif that fall in a CDR. Instead, we de-risk motifs that are common at a specific site in commercial mAb-based molecules (and therefore did not previously pose an insurmountable barrier to commercialization) and motifs at specific sites for which we have prior experimental data indicating acceptably low levels of modification. We have used this approach successfully to identify candidates in a discovery phase program with exclusively very low risk potential chemical liabilities. Identifying these candidates in the discovery phase allowed us to bypass protein engineering and accelerate the program’s timeline by 6 months.
Collapse
|
15
|
Protein folding stabilities are a major determinant of oxidation rates for buried methionine residues. J Biol Chem 2022; 298:101872. [PMID: 35346688 PMCID: PMC9062257 DOI: 10.1016/j.jbc.2022.101872] [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: 12/21/2021] [Revised: 03/19/2022] [Accepted: 03/21/2022] [Indexed: 12/20/2022] Open
Abstract
The oxidation of protein-bound methionines to form methionine sulfoxides has a broad range of biological ramifications, making it important to delineate factors that influence methionine oxidation rates within a given protein. This is especially important for biopharmaceuticals, where oxidation can lead to deactivation and degradation. Previously, neighboring residue effects and solvent accessibility have been shown to impact the susceptibility of methionine residues to oxidation. In this study, we provide proteome-wide evidence that oxidation rates of buried methionine residues are also strongly influenced by the thermodynamic folding stability of proteins. We surveyed the Escherichia coli proteome using several proteomic methodologies and globally measured oxidation rates of methionine residues in the presence and absence of tertiary structure, as well as the folding stabilities of methionine-containing domains. These data indicated that buried methionines have a wide range of protection factors against oxidation that correlate strongly with folding stabilities. Consistent with this, we show that in comparison to E. coli, the proteome of the thermophile Thermus thermophilus is significantly more stable and thus more resistant to methionine oxidation. To demonstrate the utility of this correlation, we used native methionine oxidation rates to survey the folding stabilities of E. coli and T. thermophilus proteomes at various temperatures and propose a model that relates the temperature dependence of the folding stabilities of these two species to their optimal growth temperatures. Overall, these results indicate that oxidation rates of buried methionines from the native state of proteins can be used as a metric of folding stability.
Collapse
|
16
|
Sankar K, Trainor K, Blazer L, Adams J, Sidhu S, Day T, Meiering E, Maier J. A Descriptor set for Quantitative Structure-Property Relationship Prediction in Biologics. Mol Inform 2022; 41:e2100240. [PMID: 35277930 DOI: 10.1002/minf.202100240] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 03/11/2022] [Indexed: 11/12/2022]
Abstract
In addition to attaining the desired binding to their targets, a crucial aspect in the development of biotherapeutics is 'developability', which includes several desirable properties such as high solubility, low viscosity and aggregation, physico-chemical stability and low immunogenicity. The lack of any of these properties can lead to significant obstacles in advancing them to clinic; thus in silico methods capable of raising warning flags in earlier stages of development are highly beneficial. We have developed a computational framework based on a large and diverse set of protein specific descriptors ideal for making liability predictions using a machine-learning approach. This set offers a high degree of feature diversity classifiable by sequence, structure and surface patches. We assess the sensitivity and applicability of these descriptors in four dedicated case studies that are believed to be representative of biophysical characterizations commonly employed during the development process. In addition to data sets obtained from public sources, we have validated the descriptors on novel experimental data sets in order to address antibody developability and to generate prospective predictions on Adnectins. The results demonstrate that the descriptors are well suited to assist in the improvement of properties of systems that exhibit poor solubility or aggregation.
Collapse
|
17
|
Khetan R, Curtis R, Deane CM, Hadsund JT, Kar U, Krawczyk K, Kuroda D, Robinson SA, Sormanni P, Tsumoto K, Warwicker J, Martin ACR. Current advances in biopharmaceutical informatics: guidelines, impact and challenges in the computational developability assessment of antibody therapeutics. MAbs 2022; 14:2020082. [PMID: 35104168 PMCID: PMC8812776 DOI: 10.1080/19420862.2021.2020082] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Therapeutic monoclonal antibodies and their derivatives are key components of clinical pipelines in the global biopharmaceutical industry. The availability of large datasets of antibody sequences, structures, and biophysical properties is increasingly enabling the development of predictive models and computational tools for the "developability assessment" of antibody drug candidates. Here, we provide an overview of the antibody informatics tools applicable to the prediction of developability issues such as stability, aggregation, immunogenicity, and chemical degradation. We further evaluate the opportunities and challenges of using biopharmaceutical informatics for drug discovery and optimization. Finally, we discuss the potential of developability guidelines based on in silico metrics that can be used for the assessment of antibody stability and manufacturability.
Collapse
Affiliation(s)
- Rahul Khetan
- Manchester Institute of Biotechnology, University of Manchester, Manchester, UK
| | - Robin Curtis
- Manchester Institute of Biotechnology, University of Manchester, Manchester, UK
| | | | | | - Uddipan Kar
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | | | - Daisuke Kuroda
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan.,Medical Device Development and Regulation Research Center, School of Engineering, The University of Tokyo, Tokyo, Japan.,Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, Japan
| | | | - Pietro Sormanni
- Chemistry of Health, Yusuf Hamied Department of Chemistry, University of Cambridge
| | - Kouhei Tsumoto
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan.,Medical Device Development and Regulation Research Center, School of Engineering, The University of Tokyo, Tokyo, Japan.,Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, Japan.,The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Jim Warwicker
- Manchester Institute of Biotechnology, University of Manchester, Manchester, UK
| | - Andrew C R Martin
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK
| |
Collapse
|
18
|
Abstract
Monoclonal antibodies are susceptible to chemical and enzymatic modifications during manufacturing, storage, and shipping. Deamidation, isomerization, and oxidation can compromise the potency, efficacy, and safety of therapeutic antibodies. Recently, in silico tools have been used to identify liable residues and engineer antibodies with better chemical stability. Computational approaches for predicting deamidation, isomerization, oxidation, glycation, carbonylation, sulfation, and hydroxylation are reviewed here. Although liable motifs have been used to improve the chemical stability of antibodies, the accuracy of in silico predictions can be improved using machine learning and molecular dynamic simulations. In addition, there are opportunities to improve predictions for specific stress conditions, develop in silico prediction of novel modifications in antibodies, and predict the impact of modifications on physical stability and antigen-binding.
Collapse
Affiliation(s)
- Shabdita Vatsa
- Development Services, Lonza Biologics, Singapore, Singapore
| |
Collapse
|
19
|
Han X, Shih J, Lin Y, Chai Q, Cramer SM. Development of QSAR models for in silico screening of antibody solubility. MAbs 2022; 14:2062807. [PMID: 35442164 PMCID: PMC9037471 DOI: 10.1080/19420862.2022.2062807] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Although monoclonal antibodies (mAbs) have been shown to be extremely effective in treating a number of diseases, they often suffer from poor developability attributes, such as high viscosity and low solubility at elevated concentrations. Since experimental candidate screening is often materials and labor intensive, there is substantial interest in developing in silico tools for expediting mAb design. Here, we present a strategy using machine learning-based QSAR models for the a priori estimation of mAb solubility. The extrapolated protein solubilities of a set of 111 antibodies in a histidine buffer were determined using a high throughput PEG precipitation assay. 3D homology models of the antibodies were determined, and a large set of in house and commercially available molecular descriptors were then calculated. The resulting experimental and descriptor data were then used for the development of QSAR models of mAb solubilities. After feature selection and training with different machine learning algorithms, the models were evaluated with external test sets. The resulting regression models were able to estimate the solubility values of external test set data with R2 of 0.81 and 0.85 for the two regression models developed. In addition, three class and binary classification models were developed and shown to be good estimators of mAb solubility behavior, with overall test set accuracies of 0.70 and 0.95, respectively. The analysis of the selected molecular descriptors in these models was also found to be informative and suggested that several charge-based descriptors and isotype may play important roles in mAb solubility. The combination of high throughput relative solubility experimental techniques in concert with efficient machine learning QSAR models offers an opportunity to rapidly screen potential mAb candidates and to design therapeutics with improved solubility characteristics.
Collapse
Affiliation(s)
- Xuan Han
- Department of Chemical and Biological Engineering and Center for Biotechnology and interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - James Shih
- Biotechnology Discovery Research, Eli Lilly Biotechnology Center, San Diego, California, USA
| | - Yuhao Lin
- Research Information & Digital Solutions, Eli Lilly Biotechnology Center, San Diego, California, USA
| | - Qing Chai
- Biotechnology Discovery Research, Eli Lilly Biotechnology Center, San Diego, California, USA
| | - Steven M Cramer
- Department of Chemical and Biological Engineering and Center for Biotechnology and interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA
| |
Collapse
|
20
|
Gupta S, Jiskoot W, Schöneich C, Rathore AS. Oxidation and Deamidation of Monoclonal Antibody Products: Potential Impact on Stability, Biological Activity, and Efficacy. J Pharm Sci 2021; 111:903-918. [PMID: 34890632 DOI: 10.1016/j.xphs.2021.11.024] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/24/2021] [Accepted: 11/26/2021] [Indexed: 12/25/2022]
Abstract
The role in human health of therapeutic proteins in general, and monoclonal antibodies (mAbs) in particular, has been significant and is continuously evolving. A considerable amount of time and resources are invested first in mAb product development and then in clinical examination of the product. Physical and chemical degradation can occur during manufacturing, processing, storage, handling, and administration. Therapeutic proteins may undergo various chemical degradation processes, including oxidation, deamidation, isomerization, hydrolysis, deglycosylation, racemization, disulfide bond breakage and formation, Maillard reaction, and β-elimination. Oxidation and deamidation are the most common chemical degradation processes of mAbs, which may result in changes in physical properties, such as hydrophobicity, charge, secondary or/and tertiary structure, and may lower the thermodynamic or kinetic barrier to unfold. This may predispose the product to aggregation and other chemical modifications, which can alter the binding affinity, half-life, and efficacy of the product. This review summarizes major findings from the past decade on the impact of oxidation and deamidation on the stability, biological activity, and efficacy of mAb products. Mechanisms of action, influencing factors, characterization tools, clinical impact, and risk mitigation strategies have been addressed.
Collapse
Affiliation(s)
- Surbhi Gupta
- Department of Chemical Engineering, Indian Institute of Technology Delhi, New Delhi-110016, India
| | - Wim Jiskoot
- Division of BioTherapeutics, Leiden Centre for Drug Research (LACDR), Leiden University, Leiden, the Netherlands
| | | | - Anurag S Rathore
- Department of Chemical Engineering, Indian Institute of Technology Delhi, New Delhi-110016, India.
| |
Collapse
|
21
|
Kadaoluwa Pathirannahalage SP, Meftahi N, Elbourne A, Weiss ACG, McConville CF, Padua A, Winkler DA, Costa Gomes M, Greaves TL, Le TC, Besford QA, Christofferson AJ. Systematic Comparison of the Structural and Dynamic Properties of Commonly Used Water Models for Molecular Dynamics Simulations. J Chem Inf Model 2021; 61:4521-4536. [PMID: 34406000 DOI: 10.1021/acs.jcim.1c00794] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Water is a unique solvent that is ubiquitous in biology and present in a variety of solutions, mixtures, and materials settings. It therefore forms the basis for all molecular dynamics simulations of biological phenomena, as well as for many chemical, industrial, and materials investigations. Over the years, many water models have been developed, and it remains a challenge to find a single water model that accurately reproduces all experimental properties of water simultaneously. Here, we report a comprehensive comparison of structural and dynamic properties of 30 commonly used 3-point, 4-point, 5-point, and polarizable water models simulated using consistent settings and analysis methods. For the properties of density, coordination number, surface tension, dielectric constant, self-diffusion coefficient, and solvation free energy of methane, models published within the past two decades consistently show better agreement with experimental values compared to models published earlier, albeit with some notable exceptions. However, no single model reproduced all experimental values exactly, highlighting the need to carefully choose a water model for a particular study, depending on the phenomena of interest. Finally, machine learning algorithms quantified the relationship between the water model force field parameters and the resulting bulk properties, providing insight into the parameter-property relationship and illustrating the challenges of developing a water model that can accurately reproduce all properties of water simultaneously.
Collapse
Affiliation(s)
- Sachini P Kadaoluwa Pathirannahalage
- School of Science, RMIT University, Melbourne, Victoria 3000, Australia.,Laboratoire de Chimie, Ecole Normale Supérieure de Lyon, CNRS, Lyon 69342, France
| | - Nastaran Meftahi
- ARC Centre of Excellence in Exciton Science, School of Science, RMIT University, Melbourne, Victoria 3000, Australia
| | - Aaron Elbourne
- School of Science, RMIT University, Melbourne, Victoria 3000, Australia
| | - Alessia C G Weiss
- Leibniz-Institut für Polymerforschung e.V., Hohe Straße 6, 01069 Dresden, Germany
| | - Chris F McConville
- School of Science, RMIT University, Melbourne, Victoria 3000, Australia.,Institute for Frontier Materials, Deakin University, Geelong, Victoria 3220, Australia
| | - Agilio Padua
- Laboratoire de Chimie, Ecole Normale Supérieure de Lyon, CNRS, Lyon 69342, France
| | - David A Winkler
- School of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3086, Australia.,Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.,School of Pharmacy, University of Nottingham, Nottingham NG7 2QL, U.K
| | | | - Tamar L Greaves
- School of Science, RMIT University, Melbourne, Victoria 3000, Australia
| | - Tu C Le
- School of Engineering, RMIT University, Melbourne, Victoria 3001, Australia
| | - Quinn A Besford
- Leibniz-Institut für Polymerforschung e.V., Hohe Straße 6, 01069 Dresden, Germany
| | - Andrew J Christofferson
- School of Science, RMIT University, Melbourne, Victoria 3000, Australia.,ARC Centre of Excellence in Exciton Science, School of Science, RMIT University, Melbourne, Victoria 3000, Australia
| |
Collapse
|
22
|
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
|
23
|
Delmar JA, Buehler E, Chetty AK, Das A, Quesada GM, Wang J, Chen X. Machine learning prediction of methionine and tryptophan photooxidation susceptibility. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:466-477. [PMID: 33898635 PMCID: PMC8060516 DOI: 10.1016/j.omtm.2021.03.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 03/26/2021] [Indexed: 12/01/2022]
Abstract
Photooxidation of methionine (Met) and tryptophan (Trp) residues is common and includes major degradation pathways that often pose a serious threat to the success of therapeutic proteins. Oxidation impacts all steps of protein production, manufacturing, and shelf life. Prediction of oxidation liability as early as possible in development is important because many more candidate drugs are discovered than can be tested experimentally. Undetected oxidation liabilities necessitate expensive and time-consuming remediation strategies in development and may lead to good drugs reaching patients slowly. Conversely, sites mischaracterized as oxidation liabilities could result in overengineering and lead to good drugs never reaching patients. To our knowledge, no predictive model for photooxidation of Met or Trp is currently available. We applied the random forest machine learning algorithm to in-house liquid chromatography-tandem mass spectrometry (LC-MS/MS) datasets (Met, n = 421; Trp, n = 342) of tryptic therapeutic protein peptides to create computational models for Met and Trp photooxidation. We show that our machine learning models predict Met and Trp photooxidation likelihood with 0.926 and 0.860 area under the curve (AUC), respectively, and Met photooxidation rate with a correlation coefficient (Q2) of 0.511 and root-mean-square error (RMSE) of 10.9%. We further identify important physical, chemical, and formulation parameters that influence photooxidation. Improvement of biopharmaceutical liability predictions will result in better, more stable drugs, increasing development throughput, product quality, and likelihood of clinical success.
Collapse
Affiliation(s)
- Jared A Delmar
- Biopharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Eugen Buehler
- Data Sciences and AI, R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Ashwin K Chetty
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA
| | - Agastya Das
- Khoury College of Computer Sciences, Northeastern University, Boston, MA 02115, USA
| | | | - Jihong Wang
- Biopharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Xiaoyu Chen
- Biopharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| |
Collapse
|
24
|
Narayanan H, Dingfelder F, Butté A, Lorenzen N, Sokolov M, Arosio P. Machine Learning for Biologics: Opportunities for Protein Engineering, Developability, and Formulation. Trends Pharmacol Sci 2021; 42:151-165. [DOI: 10.1016/j.tips.2020.12.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/10/2020] [Accepted: 12/16/2020] [Indexed: 12/19/2022]
|
25
|
Bailly M, Mieczkowski C, Juan V, Metwally E, Tomazela D, Baker J, Uchida M, Kofman E, Raoufi F, Motlagh S, Yu Y, Park J, Raghava S, Welsh J, Rauscher M, Raghunathan G, Hsieh M, Chen YL, Nguyen HT, Nguyen N, Cipriano D, Fayadat-Dilman L. Predicting Antibody Developability Profiles Through Early Stage Discovery Screening. MAbs 2021; 12:1743053. [PMID: 32249670 PMCID: PMC7153844 DOI: 10.1080/19420862.2020.1743053] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Monoclonal antibodies play an increasingly important role for the development of new drugs across multiple therapy areas. The term 'developability' encompasses the feasibility of molecules to successfully progress from discovery to development via evaluation of their physicochemical properties. These properties include the tendency for self-interaction and aggregation, thermal stability, colloidal stability, and optimization of their properties through sequence engineering. Selection of the best antibody molecule based on biological function, efficacy, safety, and developability allows for a streamlined and successful CMC phase. An efficient and practical high-throughput developability workflow (100 s-1,000 s of molecules) implemented during early antibody generation and screening is crucial to select the best lead candidates. This involves careful assessment of critical developability parameters, combined with binding affinity and biological properties evaluation using small amounts of purified material (<1 mg), as well as an efficient data management and database system. Herein, a panel of 152 various human or humanized monoclonal antibodies was analyzed in biophysical property assays. Correlations between assays for different sets of properties were established. We demonstrated in two case studies that physicochemical properties and key assay endpoints correlate with key downstream process parameters. The workflow allows the elimination of antibodies with suboptimal properties and a rank ordering of molecules for further evaluation early in the candidate selection process. This enables any further engineering for problematic sequence attributes without affecting program timelines.
Collapse
Affiliation(s)
- Marc Bailly
- Discovery Biologics, Protein Sciences, South San Francisco, CA, USA
| | - Carl Mieczkowski
- Discovery Biologics, Protein Sciences, South San Francisco, CA, USA
| | - Veronica Juan
- Discovery Biologics, Protein Sciences, South San Francisco, CA, USA
| | - Essam Metwally
- Computation and Structural Chemistry, South San Francisco, CA, USA
| | - Daniela Tomazela
- Discovery Biologics, Protein Sciences, South San Francisco, CA, USA
| | - Jeanne Baker
- Discovery Biologics, Protein Sciences, South San Francisco, CA, USA
| | - Makiko Uchida
- Discovery Biologics, Protein Sciences, South San Francisco, CA, USA
| | - Ester Kofman
- Discovery Biologics, Protein Sciences, South San Francisco, CA, USA
| | - Fahimeh Raoufi
- Discovery Biologics, Protein Sciences, South San Francisco, CA, USA
| | - Soha Motlagh
- Discovery Biologics, Protein Sciences, South San Francisco, CA, USA
| | - Yao Yu
- Discovery Biologics, Protein Sciences, South San Francisco, CA, USA
| | - Jihea Park
- Discovery Biologics, Protein Sciences, South San Francisco, CA, USA
| | - Smita Raghava
- Pharmaceutical Sciences, Sterile FormulationSciences, Kenilworth, NJ, USA
| | - John Welsh
- Downstream Process Development andEngineering, Kenilworth, NJ, USA
| | - Michael Rauscher
- Downstream Process Development andEngineering, Kenilworth, NJ, USA
| | | | - Mark Hsieh
- Discovery Biologics, Protein Sciences, South San Francisco, CA, USA
| | - Yi-Ling Chen
- Discovery Biologics, Protein Sciences, South San Francisco, CA, USA
| | - Hang Thu Nguyen
- Discovery Biologics, Protein Sciences, South San Francisco, CA, USA
| | - Nhung Nguyen
- Discovery Biologics, Protein Sciences, South San Francisco, CA, USA
| | - Dan Cipriano
- Discovery Biologics, Protein Sciences, South San Francisco, CA, USA
| | | |
Collapse
|
26
|
Carrara SC, Ulitzka M, Grzeschik J, Kornmann H, Hock B, Kolmar H. From cell line development to the formulated drug product: The art of manufacturing therapeutic monoclonal antibodies. Int J Pharm 2020; 594:120164. [PMID: 33309833 DOI: 10.1016/j.ijpharm.2020.120164] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/23/2020] [Accepted: 12/06/2020] [Indexed: 02/07/2023]
Abstract
Therapeutic monoclonal antibodies and related products have steadily grown to become the dominant product class within the biopharmaceutical market. Production of antibodies requires special precautions to ensure safety and efficacy of the product. In particular, minimizing antibody product heterogeneity is crucial as drug substance variants may impair the activity, efficacy, safety, and pharmacokinetic properties of an antibody, consequently resulting in the failure of a product in pre-clinical and clinical development. This review will cover the manufacturing and formulation challenges and advances of therapeutic monoclonal antibodies, focusing on improved processes to minimize variants and ensure batch-to-batch consistency. Processes put in place by regulatory agencies, such as Quality-by-Design (QbD) and current Good Manufacturing Practices (cGMP), and how their implementation has aided drug development in pharmaceutical companies will be reviewed. Advances in formulation and considerations on the intended use of a therapeutic antibody, including the route of administration and patient compliance, will be discussed.
Collapse
Affiliation(s)
- Stefania C Carrara
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany; Ferring Darmstadt Laboratory, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| | - Michael Ulitzka
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany; Ferring Darmstadt Laboratory, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| | - Julius Grzeschik
- Ferring Darmstadt Laboratory, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| | - Henri Kornmann
- Ferring International Center SA, CH-1162 Saint-Prex, Switzerland
| | - Björn Hock
- Ferring International Center SA, CH-1162 Saint-Prex, Switzerland.
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany.
| |
Collapse
|
27
|
Kamerzell TJ, Middaugh CR. Prediction Machines: Applied Machine Learning for Therapeutic Protein Design and Development. J Pharm Sci 2020; 110:665-681. [PMID: 33278409 DOI: 10.1016/j.xphs.2020.11.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/27/2020] [Accepted: 11/27/2020] [Indexed: 12/11/2022]
Abstract
The rapid growth in technological advances and quantity of scientific data over the past decade has led to several challenges including data storage and analysis. Accurate models of complex datasets were previously difficult to develop and interpret. However, improvements in machine learning algorithms have since enabled unparalleled classification and prediction capabilities. The application of machine learning can be seen throughout diverse industries due to their ease of use and interpretability. In this review, we describe popular machine learning algorithms and highlight their application in pharmaceutical protein development. Machine learning models have now been applied to better understand the nonlinear concentration dependent viscosity of protein solutions, predict protein oxidation and deamidation rates, classify sub-visible particles and compare the physical stability of proteins. We also applied several machine learning algorithms using previously published data and describe models with improved predictions and classification. The authors hope that this review can be used as a resource to others and encourage continued application of machine learning algorithms to problems in pharmaceutical protein development.
Collapse
Affiliation(s)
- Tim J Kamerzell
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS, USA; Division of Internal Medicine, HCA MidWest Health, Overland Park, KS, USA.
| | - C Russell Middaugh
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS, USA
| |
Collapse
|
28
|
Predicting antibody affinity changes upon mutations by combining multiple predictors. Sci Rep 2020; 10:19533. [PMID: 33177627 PMCID: PMC7658247 DOI: 10.1038/s41598-020-76369-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023] Open
Abstract
Antibodies are proteins working in our immune system with high affinity and specificity for target antigens, making them excellent tools for both biotherapeutic and bioengineering applications. The prediction of antibody affinity changes upon mutations (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${{\Delta \Delta {\mathrm{G}}}}_{\mathrm{binding}}$$\end{document}ΔΔGbinding) is important for antibody engineering. Numerous computational methods have been proposed based on different approaches including molecular mechanics and machine learning. However, the accuracy by each individual predictor is not enough for efficient antibody development. In this study, we develop a new prediction method by combining multiple predictors based on machine learning. Our method was tested on the SiPMAB database, evaluating the Pearson’s correlation coefficient between predicted and experimental \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${{\Delta \Delta {\mathrm{G}}}}_{\mathrm{binding}}$$\end{document}ΔΔGbinding. Our method achieved higher accuracy (R = 0.69) than previous molecular mechanics or machine-learning based methods (R = 0.59) and the previous method using the average of multiple predictors (R = 0.64). Feature importance analysis indicated that the improved accuracy was obtained by combining predictors with different importance, which have different protocols for calculating energies and for generating mutant and unbound state structures. This study demonstrates that machine learning is a powerful framework for combining different approaches to predict antibody affinity changes.
Collapse
|
29
|
Karlberg M, de Souza JV, Fan L, Kizhedath A, Bronowska AK, Glassey J. QSAR Implementation for HIC Retention Time Prediction of mAbs Using Fab Structure: A Comparison between Structural Representations. Int J Mol Sci 2020; 21:ijms21218037. [PMID: 33126648 PMCID: PMC7663183 DOI: 10.3390/ijms21218037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/22/2020] [Accepted: 10/27/2020] [Indexed: 12/19/2022] Open
Abstract
Monoclonal antibodies (mAbs) constitute a rapidly growing biopharmaceutical sector. However, their growth is impeded by high failure rates originating from failed clinical trials and developability issues in process development. There is, therefore, a growing need for better in silico tools to aid in risk assessment of mAb candidates to promote early-stage screening of potentially problematic mAb candidates. In this study, a quantitative structure–activity relationship (QSAR) modelling workflow was designed for the prediction of hydrophobic interaction chromatography (HIC) retention times of mAbs. Three novel descriptor sets derived from primary sequence, homology modelling, and atomistic molecular dynamics (MD) simulations were developed and assessed to determine the necessary level of structural resolution needed to accurately capture the relationship between mAb structures and HIC retention times. The results showed that descriptors derived from 3D structures obtained after MD simulations were the most suitable for HIC retention time prediction with a R2 = 0.63 in an external test set. It was found that when using homology modelling, the resulting 3D structures became biased towards the used structural template. Performing an MD simulation therefore proved to be a necessary post-processing step for the mAb structures in order to relax the structures and allow them to attain a more natural conformation. Based on the results, the proposed workflow in this paper could therefore potentially contribute to aid in risk assessment of mAb candidates in early development.
Collapse
Affiliation(s)
- Micael Karlberg
- School of Engineering, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; (M.K.); (L.F.); (A.K.)
| | - João Victor de Souza
- Chemistry—School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; (J.V.d.S.); (A.K.B.)
| | - Lanyu Fan
- School of Engineering, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; (M.K.); (L.F.); (A.K.)
- Chemistry—School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; (J.V.d.S.); (A.K.B.)
| | - Arathi Kizhedath
- School of Engineering, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; (M.K.); (L.F.); (A.K.)
| | - Agnieszka K. Bronowska
- Chemistry—School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; (J.V.d.S.); (A.K.B.)
| | - Jarka Glassey
- School of Engineering, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; (M.K.); (L.F.); (A.K.)
- Correspondence:
| |
Collapse
|
30
|
Kuroda D, Tsumoto K. Engineering Stability, Viscosity, and Immunogenicity of Antibodies by Computational Design. J Pharm Sci 2020; 109:1631-1651. [DOI: 10.1016/j.xphs.2020.01.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/25/2019] [Accepted: 01/10/2020] [Indexed: 12/18/2022]
|
31
|
Yoo DK, Lee SR, Jung Y, Han H, Lee HK, Han J, Kim S, Chae J, Ryu T, Chung J. Machine Learning-Guided Prediction of Antigen-Reactive In Silico Clonotypes Based on Changes in Clonal Abundance through Bio-Panning. Biomolecules 2020; 10:E421. [PMID: 32182714 PMCID: PMC7175295 DOI: 10.3390/biom10030421] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/04/2020] [Accepted: 03/06/2020] [Indexed: 02/07/2023] Open
Abstract
c-Met is a promising target in cancer therapy for its intrinsic oncogenic properties. However, there are currently no c-Met-specific inhibitors available in the clinic. Antibodies blocking the interaction with its only known ligand, hepatocyte growth factor, and/or inducing receptor internalization have been clinically tested. To explore other therapeutic antibody mechanisms like Fc-mediated effector function, bispecific T cell engagement, and chimeric antigen T cell receptors, a diverse panel of antibodies is essential. We prepared a chicken immune scFv library, performed four rounds of bio-panning, obtained 641 clones using a high-throughput clonal retrieval system (TrueRepertoireTM, TR), and found 149 antigen-reactive scFv clones. We also prepared phagemid DNA before the start of bio-panning (round 0) and, after each round of bio-panning (round 1-4), performed next-generation sequencing of these five sets of phagemid DNA, and identified 860,207 HCDR3 clonotypes and 443,292 LCDR3 clonotypes along with their clonal abundance data. We then established a TR data set consisting of antigen reactivity for scFv clones found in TR analysis and the clonal abundance of their HCDR3 and LCDR3 clonotypes in five sets of phagemid DNA. Using the TR data set, a random forest machine learning algorithm was trained to predict the binding properties of in silico HCDR3 and LCDR3 clonotypes. Subsequently, we synthesized 40 HCDR3 and 40 LCDR3 clonotypes predicted to be antigen reactive (AR) and constructed a phage-displayed scFv library called the AR library. In parallel, we also prepared an antigen non-reactive (NR) library using 10 HCDR3 and 10 LCDR3 clonotypes predicted to be NR. After a single round of bio-panning, we screened 96 randomly-selected phage clones from the AR library and found out 14 AR scFv clones consisting of 5 HCDR3 and 11 LCDR3 AR clonotypes. We also screened 96 randomly-selected phage clones from the NR library, but did not identify any AR clones. In summary, machine learning algorithms can provide a method for identifying AR antibodies, which allows for the characterization of diverse antibody libraries inaccessible by traditional methods.
Collapse
Affiliation(s)
- Duck Kyun Yoo
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Seung Ryul Lee
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Yushin Jung
- Celemics, Inc., 131 Gasandigital 1-ro, Geumcheon-gu, Seoul 08506, Korea
| | - Haejun Han
- Celemics, Inc., 131 Gasandigital 1-ro, Geumcheon-gu, Seoul 08506, Korea
| | - Hwa Kyoung Lee
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jerome Han
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Soohyun Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jisu Chae
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Taehoon Ryu
- Celemics, Inc., 131 Gasandigital 1-ro, Geumcheon-gu, Seoul 08506, Korea
| | - Junho Chung
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul 03080, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
32
|
Perdomo-Abúndez FC, Vallejo-Castillo L, Vázquez-Leyva S, López-Morales CA, Velasco-Velázquez M, Pavón L, Pérez-Tapia SM, Medina-Rivero E. Development and validation of a mass spectrometric method to determine the identity of rituximab based on its microheterogeneity profile. J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1139:121885. [PMID: 31806401 DOI: 10.1016/j.jchromb.2019.121885] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 11/10/2019] [Accepted: 11/14/2019] [Indexed: 11/19/2022]
Abstract
Analytical methods have been considered the "eyes" for development, characterization and batch releasing of biotherapeutics over the past 40 years. One of the most powerful analytical platform for biotherapeutic analysis is mass spectrometry coupled to liquid chromatography (LC-MS). Due to its wide flexibility and instrumental configurations, LC-MS can determine different physicochemical attributes of proteins, e.g. molecular mass, primary sequence, and posttranslational modifications. Intact molecular mass analysis of therapeutic proteins is essential to confirm their identity. Analytical methods must be validated to support drug quality information during its approval process. Although there are international guidelines that provide general information on validation of analytical methods, practical examples about the design, selection of validation attributes and acceptance criteria of identity LC-MS methods are scarce. Here, according to the recommendations of Q2R1 ICH guideline, we showcase the validation of an LC-MS-TOF method to identity rituximab by determining its intact and deglycosylated molecular mass profiles. The proposed method specifically identified the m/z profile and deconvoluted mass profile of rituximab from deglycosylated rituximab and from excipient blank (specificity) with a maximum error of 76.63 ppm (accuracy) and a maximum Relative Standard Deviation (RSD) of 0.00315% (precision). Besides, the system suitability test, which was based on the expected mass value of the mass calibrator, confirmed the reliability of the analytical results. In summary, validation showed that the proposed method is suitable for identifying rituximab based on its glycosylated (intact) and deglycosylated mass profile.
Collapse
Affiliation(s)
- Francisco C Perdomo-Abúndez
- Unidad de Desarrollo e Investigación en Bioprocesos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico.
| | - Luis Vallejo-Castillo
- Unidad de Desarrollo e Investigación en Bioprocesos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico.
| | - Said Vázquez-Leyva
- Unidad de Desarrollo e Investigación en Bioprocesos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico.
| | - Carlos A López-Morales
- Unidad de Desarrollo e Investigación en Bioprocesos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico.
| | - Marco Velasco-Velázquez
- Departamento de Farmacología y Unidad Periférica de Investigación en Biomedicina Translacional (CMN 20 de noviembre, ISSSTE), Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, Mexico.
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias del Instituto Nacional de Psiquiatría Ramón de la Fuente, Ciudad de México 14370, Mexico.
| | - Sonia Mayra Pérez-Tapia
- Unidad de Desarrollo e Investigación en Bioprocesos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico; Laboratorio Nacional para Servicios Especializados de Investigación, Desarrollo e Innovación (I+D+i) para Farmoquímicos y Biotecnológicos, LANSEIDI-FarBiotec-CONACyT, Ciudad de México 11340, Mexico; Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico.
| | - Emilio Medina-Rivero
- Unidad de Desarrollo e Investigación en Bioprocesos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico.
| |
Collapse
|
33
|
Tyshchuk O, Gstöttner C, Funk D, Nicolardi S, Frost S, Klostermann S, Becker T, Jolkver E, Schumacher F, Koller CF, Völger HR, Wuhrer M, Bulau P, Mølhøj M. Characterization and prediction of positional 4-hydroxyproline and sulfotyrosine, two post-translational modifications that can occur at substantial levels in CHO cells-expressed biotherapeutics. MAbs 2019; 11:1219-1232. [PMID: 31339437 PMCID: PMC6748591 DOI: 10.1080/19420862.2019.1635865] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/07/2019] [Accepted: 06/21/2019] [Indexed: 02/06/2023] Open
Abstract
Biotherapeutics may contain a multitude of different post-translational modifications (PTMs) that need to be assessed and possibly monitored and controlled to ensure reproducible product quality. During early development of biotherapeutics, unexpected PTMs might be prevented by in silico identification and characterization together with further molecular engineering. Mass determinations of a human IgG1 (mAb1) and a bispecific IgG-ligand fusion protein (BsAbA) demonstrated the presence of unusual PTMs resulting in major +80 Da, and +16/+32 Da chain variants, respectively. For mAb1, analytical cation exchange chromatography demonstrated the presence of an acidic peak accounting for 20%. A + 79.957 Da modification was localized within the light chain complementarity-determining region-2 and identified as a sulfation based on accurate mass, isotopic distribution, and a complete neutral loss reaction upon collision-induced dissociation. Top-down ultrahigh resolution MALDI-ISD FT-ICR MS of modified and unmodified Fabs allowed the allocation of the sulfation to a specific Tyr residue. An aspartate in amino-terminal position-3 relative to the affected Tyr was found to play a key role in determining the sulfation. For BsAbA, a + 15.995 Da modification was observed and localized to three specific Pro residues explaining the +16 Da chain A, and +16 Da and +32 Da chain B variants. The BsAbA modifications were verified as 4-hydroxyproline and not 3-hydroxyproline in a tryptic peptide map via co-chromatography with synthetic peptides containing the two isomeric forms. Finally, our approach for an alert system based on in-house in silico predictors is presented. This system is designed to prevent these PTMs by molecular design and engineering during early biotherapeutic development.
Collapse
Affiliation(s)
- Oksana Tyshchuk
- Roche Pharma Research and Early Development, Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| | - Christoph Gstöttner
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Dennis Funk
- Roche Pharma Research and Early Development, Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| | - Simone Nicolardi
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Stefan Frost
- Roche Pharma Research and Early Development, Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| | - Stefan Klostermann
- Roche Pharma Research and Early Development Informatics, Roche Innovation Center Munich, Penzberg, Germany
| | | | | | - Felix Schumacher
- Roche Pharma Research and Early Development, Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| | - Claudia Ferrara Koller
- Roche Pharma Research and Early Development, Large Molecule Research, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Hans Rainer Völger
- Roche Pharma Research and Early Development, Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Patrick Bulau
- Roche Pharma Technical Development Penzberg, Penzberg, Germany
| | - Michael Mølhøj
- Roche Pharma Research and Early Development, Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| |
Collapse
|
34
|
Delmar JA, Wang J, Choi SW, Martins JA, Mikhail JP. Machine Learning Enables Accurate Prediction of Asparagine Deamidation Probability and Rate. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 15:264-274. [PMID: 31890727 PMCID: PMC6923510 DOI: 10.1016/j.omtm.2019.09.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 09/16/2019] [Indexed: 12/20/2022]
Abstract
The spontaneous conversion of asparagine residues to aspartic acid or iso-aspartic acid, via deamidation, is a major pathway of protein degradation and is often seriously disruptive to biological systems. Deamidation has been shown to negatively affect both in vitro stability and in vivo biological function of diverse classes of proteins. During protein therapeutics development, deamidation liabilities that are overlooked necessitate expensive and time-consuming remediation strategies, sometimes leading to termination of the project. In this paper, we apply machine learning to a large (n = 776) liquid chromatography-tandem mass spectrometry (LC-MS/MS) dataset of monoclonal antibody peptides to create computational models for the post-translational modification asparagine deamidation, using the random decision forest method. We show that our categorical model predicts antibody deamidation with nearly 5% increased accuracy and 0.2 MCC over the best currently available models. Surprisingly, our model also paces or outperforms advanced and conventional models on an independent non-antibody dataset. In addition to deamidation probability, we are able to accurately predict deamidation rate (R2 = 0.963 and Q2 = 0.822), a capability with no peer in current models. This method should enable significant improvement in protein candidate selection, especially in biopharmaceutical development, and can be applied with similar accuracy to enzymes, monoclonal antibodies, next-generation formats, vaccine component antigens, and gene therapy vectors such as adeno-associated virus.
Collapse
Affiliation(s)
- Jared A Delmar
- Analytical Sciences, Biopharmaceutical Development, AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Jihong Wang
- Analytical Sciences, Biopharmaceutical Development, AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Seo Woo Choi
- David H. Koch School of Chemical Engineering Practice, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jason A Martins
- David H. Koch School of Chemical Engineering Practice, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - John P Mikhail
- David H. Koch School of Chemical Engineering Practice, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|