1
|
Cho YB, Ko IY, Kim HJ, Kim JW, Heo K, Na HJ, Woo JR, Lee S. Functional epitope mapping of cell surface glucose-regulated protein 94: A combinatorial approach for therapeutic targeting. Int J Biol Macromol 2025; 301:140374. [PMID: 39880265 DOI: 10.1016/j.ijbiomac.2025.140374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/14/2025] [Accepted: 01/25/2025] [Indexed: 01/31/2025]
Abstract
Glucose-regulated protein 94 (GRP94) overexpression plays a critical role in tumor cell survival across various cancers. Previously, we developed K101.1, a fully human antibody targeting cell surface GRP94, which effectively inhibits tumor angiogenesis in colorectal cancer (CRC). This study aims to identify K101.1's interaction site and further elucidate GRP94's role in tumor angiogenesis. In an HT29 CRC xenograft mouse model, K101.1 reduced tumor growth and angiogenesis by 30 % and 69 %, respectively, without inducing severe toxicity. Using hydrogen‑deuterium exchange mass spectrometry, we identified the binding site of K101.1 on GRP94, analyzing 225 peptides with 94.5 % sequence coverage and a redundancy score of 4.20. This revealed a linear epitope (Glu26-Ala34) as the specific binding site. The binding was further validated via enzyme-linked immunosorbent assay, and human umbilical vein endothelial cell tube formation assays using a synthetic epitope peptide corresponding to the identified epitope. Our findings suggest that this epitope is functionally involved in GRP94-mediated angiogenesis and is integral to its proangiogenic activity. By integrating epitope profiling with complementary experimental approaches, this study advances the understanding of GRP94's role in CRC angiogenesis and provides new insights for developing targeted cancer therapies and vaccines, offering promising avenues for CRC prevention and treatment.
Collapse
Affiliation(s)
- Yea Bin Cho
- Department of Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - In Young Ko
- New Drug Development Center, KBIOHealth, Cheongju 28160, Republic of Korea
| | - Hyun Jung Kim
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Ji Woong Kim
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Kyun Heo
- Department of Chemistry, Kookmin University, Seoul 02707, Republic of Korea; Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea; Antibody Research Institute, Kookmin University, Seoul 02707, Republic of Korea
| | - Hee Jun Na
- HauulBio, Chuncheon, Gangwon 24398, Republic of Korea
| | - Ju Rang Woo
- New Drug Development Center, KBIOHealth, Cheongju 28160, Republic of Korea.
| | - Sukmook Lee
- Department of Chemistry, Kookmin University, Seoul 02707, Republic of Korea; Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea; Antibody Research Institute, Kookmin University, Seoul 02707, Republic of Korea.
| |
Collapse
|
2
|
Dewaker V, Morya VK, Kim YH, Park ST, Kim HS, Koh YH. Revolutionizing oncology: the role of Artificial Intelligence (AI) as an antibody design, and optimization tools. Biomark Res 2025; 13:52. [PMID: 40155973 PMCID: PMC11954232 DOI: 10.1186/s40364-025-00764-4] [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: 02/25/2025] [Accepted: 03/13/2025] [Indexed: 04/01/2025] Open
Abstract
Antibodies play a crucial role in defending the human body against diseases, including life-threatening conditions like cancer. They mediate immune responses against foreign antigens and, in some cases, self-antigens. Over time, antibody-based technologies have evolved from monoclonal antibodies (mAbs) to chimeric antigen receptor T cells (CAR-T cells), significantly impacting biotechnology, diagnostics, and therapeutics. Although these advancements have enhanced therapeutic interventions, the integration of artificial intelligence (AI) is revolutionizing antibody design and optimization. This review explores recent AI advancements, including large language models (LLMs), diffusion models, and generative AI-based applications, which have transformed antibody discovery by accelerating de novo generation, enhancing immune response precision, and optimizing therapeutic efficacy. Through advanced data analysis, AI enables the prediction and design of antibody sequences, 3D structures, complementarity-determining regions (CDRs), paratopes, epitopes, and antigen-antibody interactions. These AI-powered innovations address longstanding challenges in antibody development, significantly improving speed, specificity, and accuracy in therapeutic design. By integrating computational advancements with biomedical applications, AI is driving next-generation cancer therapies, transforming precision medicine, and enhancing patient outcomes.
Collapse
Affiliation(s)
- Varun Dewaker
- Institute of New Frontier Research Team, Hallym University, Chuncheon-Si, Gangwon-Do, 24252, Republic of Korea
| | - Vivek Kumar Morya
- Department of Orthopedic Surgery, Hallym University Dongtan Sacred Hospital, Hwaseong-Si, 18450, Republic of Korea
| | - Yoo Hee Kim
- Department of Biomedical Gerontology, Ilsong Institute of Life Science, Hallym University, Seoul, 07247, Republic of Korea
| | - Sung Taek Park
- Institute of New Frontier Research Team, Hallym University, Chuncheon-Si, Gangwon-Do, 24252, Republic of Korea
- Department of Obstetrics and Gynecology, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul, 07441, Republic of Korea
- EIONCELL Inc, Chuncheon-Si, 24252, Republic of Korea
| | - Hyeong Su Kim
- Institute of New Frontier Research Team, Hallym University, Chuncheon-Si, Gangwon-Do, 24252, Republic of Korea.
- Department of Internal Medicine, Division of Hemato-Oncology, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul, 07441, Republic of Korea.
- EIONCELL Inc, Chuncheon-Si, 24252, Republic of Korea.
| | - Young Ho Koh
- Department of Biomedical Gerontology, Ilsong Institute of Life Science, Hallym University, Seoul, 07247, Republic of Korea.
| |
Collapse
|
3
|
Duan H, Chi X, Yang X, Pan S, Liu X, Gao P, Zhang F, Zhang X, Dong X, Liao Y, Yang W. Computational design and improvement of a broad influenza virus HA stem targeting antibody. Structure 2025; 33:489-503.e5. [PMID: 39884272 DOI: 10.1016/j.str.2025.01.002] [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: 09/26/2024] [Revised: 11/28/2024] [Accepted: 01/03/2025] [Indexed: 02/01/2025]
Abstract
Broadly neutralizing antibodies (nAbs) are vital therapeutic tools to counteract both pandemic and seasonal influenza threats. Traditional strategies for optimizing nAbs generally rely on labor-intensive, high-throughput mutagenesis screens. Here, we present an innovative structure-based design framework for the optimization of nAbs, which integrates epitope-paratope analysis, computational modeling, and rational design approaches, complemented by comprehensive experimental assessment. This approach was applied to optimize MEDI8852, a nAb targeting the stalk region of influenza A virus hemagglutinin (HA). The resulting variant, M18.1.2.2, shows a marked enhancement in both affinity and neutralizing efficacy, as demonstrated both in vitro and in vivo. Computational modeling reveals that this improvement can be attributed to the fine-tuning of interactions between the antibody's side-chains and the epitope residues that are highly conserved across the influenza A virus HA stalk. Our dry-wet iterative protocol for nAb optimization presented here yielded a promising candidate for influenza intervention.
Collapse
Affiliation(s)
- Huarui Duan
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaojing Chi
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xuehua Yang
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shengnan Pan
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiuying Liu
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Peixiang Gao
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fangyuan Zhang
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xinhui Zhang
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xuemeng Dong
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yi Liao
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wei Yang
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; State Key Laboratory of Respiratory Health and Multimorbidity, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
4
|
Khairunisa SQ, Rachman BE, Nasronudin, Fahmi M, Dinana IA, Ito M. Designing a multi-epitope vaccine targeting the HIV-1 subtype CRF01_AE in Indonesia. Comput Biol Med 2025; 187:109758. [PMID: 39889449 DOI: 10.1016/j.compbiomed.2025.109758] [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: 08/30/2024] [Revised: 01/24/2025] [Accepted: 01/25/2025] [Indexed: 02/03/2025]
Abstract
HIV has markedly affected millions of people globally, with antiretroviral therapy (ART) transforming acquired immunodeficiency syndrome from a fatal disease to a manageable chronic condition. However, global disparities in ART access persist, particularly in low- and middle-income countries, highlighting the urgent need for affordable HIV vaccines. In this study, we investigated the potential development of a multi-epitope vaccine (MEV) targeting the HIV subtype CRF01_AE, which is prevalent in Indonesia. Using likelihood-based evolutionary inference based on site rates to analyze mutation rates, we identified the Pol and Env proteins as optimal targets. Nine T cell epitopes (five cytotoxic and four helper) were selected based on HLA binding affinity, conservation, antigenicity, and predicted immunogenicity, achieving broad population coverage (∼95 % globally and 99.58 % in Indonesia). The MEV construct incorporated epitopes conjugated to a Vibrio cholerae toxin B subunit adjuvant and a B cell epitope known to induce broadly neutralizing antibodies. In silico characterization, including physicochemical analysis, structural modeling (validated using ProSA-web and Ramachandran plot analysis), and protein-protein docking simulations (using HADDOCK and PRODIGY), demonstrated favorable properties, stable conformation, and high-affinity interaction with antibody fragments (ΔGbind = -10.8 kcal/mol). Molecular dynamics simulations confirmed the formation of a stable complex. Immunogenicity tests revealed a strong antibody and cytokine response. These findings suggest that this MEV construct is a promising and affordable HIV-1 vaccine candidate that warrants further validation.
Collapse
Affiliation(s)
| | - Brian Eka Rachman
- Institute of Tropical Disease, Universitas Airlangga, Surabaya, 60115, East Java, Indonesia; Airlangga Hospital, Universitas Airlangga, Surabaya, 60115, East Java, Indonesia; Faculty of Medicine, Universitas Airlangga, Surabaya, 60132, East Java, Indonesia
| | - Nasronudin
- Institute of Tropical Disease, Universitas Airlangga, Surabaya, 60115, East Java, Indonesia; Airlangga Hospital, Universitas Airlangga, Surabaya, 60115, East Java, Indonesia; Faculty of Medicine, Universitas Airlangga, Surabaya, 60132, East Java, Indonesia
| | - Muhamad Fahmi
- Research Department, Research Institute of Humanity and Nature, Japan
| | - Ichda Arini Dinana
- Department of Bioinformatics, College of Life Sciences, Ritsumeikan University, Kusatsu, Japan
| | - Masahiro Ito
- Department of Bioinformatics, College of Life Sciences, Ritsumeikan University, Kusatsu, Japan
| |
Collapse
|
5
|
Gallo E. Revolutionizing Synthetic Antibody Design: Harnessing Artificial Intelligence and Deep Sequencing Big Data for Unprecedented Advances. Mol Biotechnol 2025; 67:410-424. [PMID: 38308755 DOI: 10.1007/s12033-024-01064-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/02/2024] [Indexed: 02/05/2024]
Abstract
Synthetic antibodies (Abs) represent a category of engineered proteins meticulously crafted to replicate the functions of their natural counterparts. Such Abs are generated in vitro, enabling advanced molecular alterations associated with antigen recognition, paratope site engineering, and biochemical refinements. In a parallel realm, deep sequencing has brought about a paradigm shift in molecular biology. It facilitates the prompt and cost-effective high-throughput sequencing of DNA and RNA molecules, enabling the comprehensive big data analysis of Ab transcriptomes, including specific regions of interest. Significantly, the integration of artificial intelligence (AI), based on machine- and deep- learning approaches, has fundamentally transformed our capacity to discern patterns hidden within deep sequencing big data, including distinctive Ab features and protein folding free energy landscapes. Ultimately, current AI advances can generate approximations of the most stable Ab structural configurations, enabling the prediction of de novo synthetic Abs. As a result, this manuscript comprehensively examines the latest and relevant literature concerning the intersection of deep sequencing big data and AI methodologies for the design and development of synthetic Abs. Together, these advancements have accelerated the exploration of antibody repertoires, contributing to the refinement of synthetic Ab engineering and optimizations, and facilitating advancements in the lead identification process.
Collapse
Affiliation(s)
- Eugenio Gallo
- Avance Biologicals, Department of Medicinal Chemistry, 950 Dupont Street, Toronto, ON, M6H 1Z2, Canada.
- RevivAb, Department of Protein Engineering, Av. Ipiranga, 6681, Partenon, Porto Alegre, RS, 90619-900, Brazil.
| |
Collapse
|
6
|
Yadav AJ, Bhagat K, Sharma A, Padhi AK. Navigating the landscape: A comprehensive overview of computational approaches in therapeutic antibody design and analysis. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2025; 144:33-76. [PMID: 39978970 DOI: 10.1016/bs.apcsb.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Immunotherapy, harnessing components like antibodies, cells, and cytokines, has become a cornerstone in treating diseases such as cancer and autoimmune disorders. Therapeutic antibodies, in particular, have transformed modern medicine, providing a targeted approach that destroys disease-causing cells while sparing healthy tissues, thereby reducing the side effects commonly associated with chemotherapy. Beyond oncology, these antibodies also hold promise in addressing chronic infections where conventional therapeutics may fall short. However, antibodies identified through in vivo or in vitro methods often require extensive engineering to enhance their therapeutic potential. This optimization process, aimed at improving affinity, specificity, and reducing immunogenicity, is both challenging and costly, often involving trade-offs between critical properties. Traditional methods of antibody development, such as hybridoma technology and display techniques, are resource-intensive and time-consuming. In contrast, computational approaches offer a faster, more efficient alternative, enabling the precise design and analysis of therapeutic antibodies. These methods include sequence and structural bioinformatics approaches, next-generation sequencing-based data mining, machine learning algorithms, systems biology, immuno-informatics, and integrative approaches. These approaches are advancing the field by providing new insights and enhancing the accuracy of antibody design and analysis. In conclusion, computational approaches are essential in the development of therapeutic antibodies, significantly improving the precision and speed of discovery, optimization, and validation. Integrating these methods with experimental approaches accelerates therapeutic antibody development, paving the way for innovative strategies and treatments for various diseases ranging from cancers to autoimmune and infectious diseases.
Collapse
Affiliation(s)
- Amar Jeet Yadav
- Laboratory for Computational Biology & Biomolecular Design, School of Biochemical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi, Uttar Pradesh, India
| | - Khushboo Bhagat
- Laboratory for Computational Biology & Biomolecular Design, School of Biochemical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi, Uttar Pradesh, India
| | - Akshit Sharma
- Laboratory for Computational Biology & Biomolecular Design, School of Biochemical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi, Uttar Pradesh, India
| | - Aditya K Padhi
- Laboratory for Computational Biology & Biomolecular Design, School of Biochemical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi, Uttar Pradesh, India.
| |
Collapse
|
7
|
Wang J, Aceves AJ, Friesenhahn NJ, Mayo SL. CDRxAbs: antibody small-molecule conjugates with computationally designed target-binding synergy. Protein Eng Des Sel 2025; 38:gzaf004. [PMID: 40114302 DOI: 10.1093/protein/gzaf004] [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: 12/19/2023] [Revised: 02/05/2025] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
Bioconjugates as therapeutic modalities combine the advantages and offset the disadvantages of their constituent parts to achieve a refined spectrum of action. We combine the concept of bioconjugation with the full atomic simulation capability of computational protein design to define a new class of molecular recognition agents: CDR-extended antibodies, abbreviated as CDRxAbs. A CDRxAb incorporates a covalently attached small molecule into an antibody/target binding interface using computational protein design to create an antibody small-molecule conjugate that binds tighter to the target of the small molecule than the small molecule would alone. CDRxAbs are also expected to increase the target binding specificity of their associated small molecules. In a proof-of-concept study using monomeric streptavidin/biotin pairs at either a nanomolar or micromolar-level initial affinity, we designed nanobody-biotin conjugates that exhibited >20-fold affinity improvement against their protein targets with step-wise optimization of binding kinetics and overall protein stability. The workflow explored through this process promises a novel approach to optimize small-molecule based therapeutics and to explore new chemical and target space for molecular-recognition agents in general.
Collapse
Affiliation(s)
- Jingzhou Wang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, United States
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, United States
- Merck Research Laboratories, 213 E Grand Ave, South San Francisco, CA 94080, USA
| | - Aiden J Aceves
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, United States
- Insight Partners, 1114 Avenue of the Americas, 36th Floor, New York, NY 10036, USA
| | - Nicholas J Friesenhahn
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, United States
| | - Stephen L Mayo
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, United States
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, United States
| |
Collapse
|
8
|
Xiang S, Zhu C, Zhou Y, Wu W, Zhang Y, Chen C, Wang F. Facile Generation of Neutralizing Antibodies on Tyrosine Phosphorylated IRS1 by Epitope-Directed Elicitation. ACS Chem Biol 2024; 19:2050-2059. [PMID: 39137393 DOI: 10.1021/acschembio.4c00382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Generating antibodies specific to the functional epitope containing phosphotyrosine remains highly challenging. Here, we create an "epitope-directed immunogen" by incorporating fluorosulfate-l-tyrosine (FSY) with cross-linking activities into a specific tyrosine phosphorylation site of insulin receptor substrate 1 (IRS1) and immunizing mice to elicit site-specific antibody responses. By taking advantage of antibody clonal selection and evolution in vivo, we efficiently identified antibodies that target the IRS1 Y612 epitope and are capable of neutralizing the binding interactions between IRS1 and p85α mediated by the phosphorylation of Y612. This epitope-directed antibody elicitation by encoding the cross-linking reactivity in the immunogen potentially enables a general method for facile generation of neutralizing antibodies to protein tyrosine phosphorylation sites.
Collapse
Affiliation(s)
- Shuqin Xiang
- Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing 100101, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100408, China
| | - Chaoyang Zhu
- Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing 100101, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100408, China
| | - Yinjian Zhou
- Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing 100101, China
| | - Weiping Wu
- Suzhou Institute for Biomedical Research, Suzhou 215028, Jiangsu, China
| | - Yuhan Zhang
- Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing 100101, China
| | - Chen Chen
- Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing 100101, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100408, China
| | - Feng Wang
- Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing 100101, China
- Suzhou Institute for Biomedical Research, Suzhou 215028, Jiangsu, China
- Beijing Translational Center for Biopharmaceuticals, Beijing 100101, China
| |
Collapse
|
9
|
Hutchings CJ, Sato AK. Phage display technology and its impact in the discovery of novel protein-based drugs. Expert Opin Drug Discov 2024; 19:887-915. [PMID: 39074492 DOI: 10.1080/17460441.2024.2367023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 06/07/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION Phage display technology is a well-established versatile in vitro display technology that has been used for over 35 years to identify peptides and antibodies for use as reagents and therapeutics, as well as exploring the diversity of alternative scaffolds as another option to conventional therapeutic antibody discovery. Such successes have been responsible for spawning a range of biotechnology companies, as well as many complementary technologies devised to expedite the drug discovery process and resolve bottlenecks in the discovery workflow. AREAS COVERED In this perspective, the authors summarize the application of phage display for drug discovery and provide examples of protein-based drugs that have either been approved or are being developed in the clinic. The amenability of phage display to generate functional protein molecules to challenging targets and recent developments of strategies and techniques designed to harness the power of sampling diverse repertoires are highlighted. EXPERT OPINION Phage display is now routinely combined with cutting-edge technologies to deep-mine antibody-based repertoires, peptide, or alternative scaffold libraries generating a wealth of data that can be leveraged, e.g. via artificial intelligence, to enable the potential for clinical success in the discovery and development of protein-based therapeutics.
Collapse
|
10
|
Song B, Wang K, Na S, Yao J, Fattah FJ, von Itzstein MS, Yang DM, Liu J, Xue Y, Liang C, Guo Y, Raman I, Zhu C, Dowell JE, Homsi J, Rashdan S, Yang S, Gwin ME, Hsiehchen D, Gloria-McCutchen Y, Raj P, Bai X, Wang J, Conejo-Garcia J, Xie Y, Gerber DE, Huang J, Wang T. Cmai: Predicting Antigen-Antibody Interactions from Massive Sequencing Data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.601035. [PMID: 39005456 PMCID: PMC11244862 DOI: 10.1101/2024.06.27.601035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
The interaction between antigens and antibodies (B cell receptors, BCRs) is the key step underlying the function of the humoral immune system in various biological contexts. The capability to profile the landscape of antigen-binding affinity of a vast number of BCRs will provide a powerful tool to reveal novel insights at unprecedented levels and will yield powerful tools for translational development. However, current experimental approaches for profiling antibody-antigen interactions are costly and time-consuming, and can only achieve low-to-mid throughput. On the other hand, bioinformatics tools in the field of antibody informatics mostly focus on optimization of antibodies given known binding antigens, which is a very different research question and of limited scope. In this work, we developed an innovative Artificial Intelligence tool, Cmai, to address the prediction of the binding between antibodies and antigens that can be scaled to high-throughput sequencing data. Cmai achieved an AUROC of 0.91 in our validation cohort. We devised a biomarker metric based on the output from Cmai applied to high-throughput BCR sequencing data. We found that, during immune-related adverse events (irAEs) caused by immune-checkpoint inhibitor (ICI) treatment, the humoral immunity is preferentially responsive to intracellular antigens from the organs affected by the irAEs. In contrast, extracellular antigens on malignant tumor cells are inducing B cell infiltrations, and the infiltrating B cells have a greater tendency to co-localize with tumor cells expressing these antigens. We further found that the abundance of tumor antigen-targeting antibodies is predictive of ICI treatment response. Overall, Cmai and our biomarker approach filled in a gap that is not addressed by current antibody optimization works nor works such as AlphaFold3 that predict the structures of complexes of proteins that are known to bind.
Collapse
|
11
|
Martarelli N, Capurro M, Mansour G, Jahromi RV, Stella A, Rossi R, Longetti E, Bigerna B, Gentili M, Rosseto A, Rossi R, Cencini C, Emiliani C, Martino S, Beeg M, Gobbi M, Tiacci E, Falini B, Morena F, Perriello VM. Artificial Intelligence-Powered Molecular Docking and Steered Molecular Dynamics for Accurate scFv Selection of Anti-CD30 Chimeric Antigen Receptors. Int J Mol Sci 2024; 25:7231. [PMID: 39000338 PMCID: PMC11242627 DOI: 10.3390/ijms25137231] [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: 06/12/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cells represent a revolutionary immunotherapy that allows specific tumor recognition by a unique single-chain fragment variable (scFv) derived from monoclonal antibodies (mAbs). scFv selection is consequently a fundamental step for CAR construction, to ensure accurate and effective CAR signaling toward tumor antigen binding. However, conventional in vitro and in vivo biological approaches to compare different scFv-derived CARs are expensive and labor-intensive. With the aim to predict the finest scFv binding before CAR-T cell engineering, we performed artificial intelligence (AI)-guided molecular docking and steered molecular dynamics analysis of different anti-CD30 mAb clones. Virtual computational scFv screening showed comparable results to surface plasmon resonance (SPR) and functional CAR-T cell in vitro and in vivo assays, respectively, in terms of binding capacity and anti-tumor efficacy. The proposed fast and low-cost in silico analysis has the potential to advance the development of novel CAR constructs, with a substantial impact on reducing time, costs, and the need for laboratory animal use.
Collapse
Affiliation(s)
- Nico Martarelli
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Michela Capurro
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Gizem Mansour
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milano, Italy; (G.M.); (M.B.); (M.G.)
| | - Ramina Vossoughi Jahromi
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Arianna Stella
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Roberta Rossi
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Emanuele Longetti
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Barbara Bigerna
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Marco Gentili
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Ariele Rosseto
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Riccardo Rossi
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Chiara Cencini
- Department of Chemistry, Biology, and Biotechnologies, Via del Giochetto, University of Perugia, 06122 Perugia, Italy; (C.C.); (C.E.); (S.M.)
| | - Carla Emiliani
- Department of Chemistry, Biology, and Biotechnologies, Via del Giochetto, University of Perugia, 06122 Perugia, Italy; (C.C.); (C.E.); (S.M.)
| | - Sabata Martino
- Department of Chemistry, Biology, and Biotechnologies, Via del Giochetto, University of Perugia, 06122 Perugia, Italy; (C.C.); (C.E.); (S.M.)
| | - Marten Beeg
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milano, Italy; (G.M.); (M.B.); (M.G.)
| | - Marco Gobbi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milano, Italy; (G.M.); (M.B.); (M.G.)
| | - Enrico Tiacci
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Brunangelo Falini
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Francesco Morena
- Department of Chemistry, Biology, and Biotechnologies, Via del Giochetto, University of Perugia, 06122 Perugia, Italy; (C.C.); (C.E.); (S.M.)
| | - Vincenzo Maria Perriello
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| |
Collapse
|
12
|
Fawcett C, Tickle JR, Coles CH. Facilitating high throughput bispecific antibody production and potential applications within biopharmaceutical discovery workflows. MAbs 2024; 16:2311992. [PMID: 39674918 DOI: 10.1080/19420862.2024.2311992] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/08/2024] [Accepted: 01/25/2024] [Indexed: 12/17/2024] Open
Abstract
A major driver for the recent investment surge in bispecific antibody (bsAb) platforms and products is the multitude of distinct mechanisms of action that bsAbs offer compared to a combination of two monoclonal antibodies. Four bsAb products were granted first regulatory approvals in the US or EU during 2023 and the biopharmaceutical industry pipeline is brimming with bsAb candidates across a broad range of therapeutic applications. In previously reported bsAb discovery campaigns, following a hypothesis-based choice of two specific target proteins, selections and screening activities have often been performed in mono-specific formats. The conversion to bispecific modalities has usually been positioned toward the end of the discovery process and has involved small numbers of lead molecules, largely due to challenges in expressing, purifying, and analyzing large numbers of bsAbs. In this review, we discuss emerging strategies to facilitate the production of expanded bsAb panels, focusing particularly upon combinatorial methods to generate bsAb matrices. Such technologies will enable screening in. bispecific formats at earlier stages of discovery campaigns, not only widening the accessible protein space to maximize chances of success, but also advancing empirical bi-target validation activities to assess initial target selection hypotheses.
Collapse
Affiliation(s)
- Caitlin Fawcett
- Large Molecule Discovery, GSK, GSK Medicines Research Centre, Stevenage, UK
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - Joseph R Tickle
- Large Molecule Discovery, GSK, GSK Medicines Research Centre, Stevenage, UK
| | - Charlotte H Coles
- Large Molecule Discovery, GSK, GSK Medicines Research Centre, Stevenage, UK
| |
Collapse
|
13
|
Fischman S, Levin I, Rondeau JM, Štrajbl M, Lehmann S, Huber T, Nimrod G, Cebe R, Omer D, Kovarik J, Bernstein S, Sasson Y, Demishtein A, Shlamkovich T, Bluvshtein O, Grossman N, Barak-Fuchs R, Zhenin M, Fastman Y, Twito S, Vana T, Zur N, Ofran Y. "Redirecting an anti-IL-1β antibody to bind a new, unrelated and computationally predicted epitope on hIL-17A". Commun Biol 2023; 6:997. [PMID: 37773269 PMCID: PMC10542344 DOI: 10.1038/s42003-023-05369-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: 01/16/2023] [Accepted: 09/18/2023] [Indexed: 10/01/2023] Open
Abstract
Antibody engineering technology is at the forefront of therapeutic antibody development. The primary goal for engineering a therapeutic antibody is the generation of an antibody with a desired specificity, affinity, function, and developability profile. Mature antibodies are considered antigen specific, which may preclude their use as a starting point for antibody engineering. Here, we explore the plasticity of mature antibodies by engineering novel specificity and function to a pre-selected antibody template. Using a small, focused library, we engineered AAL160, an anti-IL-1β antibody, to bind the unrelated antigen IL-17A, with the introduction of seven mutations. The final redesigned antibody, 11.003, retains favorable biophysical properties, binds IL-17A with sub-nanomolar affinity, inhibits IL-17A binding to its cognate receptor and is functional in a cell-based assay. The epitope of the engineered antibody can be computationally predicted based on the sequence of the template antibody, as is confirmed by the crystal structure of the 11.003/IL-17A complex. The structures of the 11.003/IL-17A and the AAL160/IL-1β complexes highlight the contribution of germline residues to the paratopes of both the template and re-designed antibody. This case study suggests that the inherent plasticity of antibodies allows for re-engineering of mature antibodies to new targets, while maintaining desirable developability profiles.
Collapse
Affiliation(s)
| | - Itay Levin
- Biolojic Design LTD, Rehovot, Israel
- Enzymit LTD, Ness Ziona, Israel
| | | | | | - Sylvie Lehmann
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Thomas Huber
- Novartis Institutes for Biomedical Research, Basel, Switzerland
- Ridgelinediscovery, Basel, Switzerland
| | | | - Régis Cebe
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Dotan Omer
- Biolojic Design LTD, Rehovot, Israel
- EmendoBio Inc., Rehovot, Israel
| | - Jiri Kovarik
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | | | | | - Alik Demishtein
- Biolojic Design LTD, Rehovot, Israel
- Anima Biotech, Ramat-Gan, Israel
| | | | - Olga Bluvshtein
- Biolojic Design LTD, Rehovot, Israel
- Enzymit LTD, Ness Ziona, Israel
| | | | | | | | | | - Shir Twito
- Biolojic Design LTD, Rehovot, Israel
- Enzymit LTD, Ness Ziona, Israel
| | - Tal Vana
- Biolojic Design LTD, Rehovot, Israel
| | - Nevet Zur
- Biolojic Design LTD, Rehovot, Israel
| | - Yanay Ofran
- Biolojic Design LTD, Rehovot, Israel
- The Goodman Faculty of Life Sciences, Nanotechnology Building, Bar Ilan University, Ramat Gan, Israel
| |
Collapse
|
14
|
Bauer J, Rajagopal N, Gupta P, Gupta P, Nixon AE, Kumar S. How can we discover developable antibody-based biotherapeutics? Front Mol Biosci 2023; 10:1221626. [PMID: 37609373 PMCID: PMC10441133 DOI: 10.3389/fmolb.2023.1221626] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/10/2023] [Indexed: 08/24/2023] Open
Abstract
Antibody-based biotherapeutics have emerged as a successful class of pharmaceuticals despite significant challenges and risks to their discovery and development. This review discusses the most frequently encountered hurdles in the research and development (R&D) of antibody-based biotherapeutics and proposes a conceptual framework called biopharmaceutical informatics. Our vision advocates for the syncretic use of computation and experimentation at every stage of biologic drug discovery, considering developability (manufacturability, safety, efficacy, and pharmacology) of potential drug candidates from the earliest stages of the drug discovery phase. The computational advances in recent years allow for more precise formulation of disease concepts, rapid identification, and validation of targets suitable for therapeutic intervention and discovery of potential biotherapeutics that can agonize or antagonize them. Furthermore, computational methods for de novo and epitope-specific antibody design are increasingly being developed, opening novel computationally driven opportunities for biologic drug discovery. Here, we review the opportunities and limitations of emerging computational approaches for optimizing antigens to generate robust immune responses, in silico generation of antibody sequences, discovery of potential antibody binders through virtual screening, assessment of hits, identification of lead drug candidates and their affinity maturation, and optimization for developability. The adoption of biopharmaceutical informatics across all aspects of drug discovery and development cycles should help bring affordable and effective biotherapeutics to patients more quickly.
Collapse
Affiliation(s)
- Joschka Bauer
- Early Stage Pharmaceutical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach/Riss, Germany
- In Silico Team, Boehringer Ingelheim, Hannover, Germany
| | - Nandhini Rajagopal
- In Silico Team, Boehringer Ingelheim, Hannover, Germany
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, United States
| | - Priyanka Gupta
- In Silico Team, Boehringer Ingelheim, Hannover, Germany
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, United States
| | - Pankaj Gupta
- In Silico Team, Boehringer Ingelheim, Hannover, Germany
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, United States
| | - Andrew E. Nixon
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, United States
| | - Sandeep Kumar
- In Silico Team, Boehringer Ingelheim, Hannover, Germany
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, United States
| |
Collapse
|
15
|
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
|
16
|
Chen Z, Wang X, Chen X, Huang J, Wang C, Wang J, Wang Z. Accelerating therapeutic protein design with computational approaches toward the clinical stage. Comput Struct Biotechnol J 2023; 21:2909-2926. [PMID: 38213894 PMCID: PMC10781723 DOI: 10.1016/j.csbj.2023.04.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/11/2023] [Accepted: 04/27/2023] [Indexed: 01/13/2024] Open
Abstract
Therapeutic protein, represented by antibodies, is of increasing interest in human medicine. However, clinical translation of therapeutic protein is still largely hindered by different aspects of developability, including affinity and selectivity, stability and aggregation prevention, solubility and viscosity reduction, and deimmunization. Conventional optimization of the developability with widely used methods, like display technologies and library screening approaches, is a time and cost-intensive endeavor, and the efficiency in finding suitable solutions is still not enough to meet clinical needs. In recent years, the accelerated advancement of computational methodologies has ushered in a transformative era in the field of therapeutic protein design. Owing to their remarkable capabilities in feature extraction and modeling, the integration of cutting-edge computational strategies with conventional techniques presents a promising avenue to accelerate the progression of therapeutic protein design and optimization toward clinical implementation. Here, we compared the differences between therapeutic protein and small molecules in developability and provided an overview of the computational approaches applicable to the design or optimization of therapeutic protein in several developability issues.
Collapse
Affiliation(s)
- Zhidong Chen
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Xinpei Wang
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Xu Chen
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Juyang Huang
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Chenglin Wang
- Shenzhen Qiyu Biotechnology Co., Ltd, Shenzhen 518107, China
| | - Junqing Wang
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Zhe Wang
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| |
Collapse
|
17
|
Duarte AC, Costa EC, Filipe HAL, Saraiva SM, Jacinto T, Miguel SP, Ribeiro MP, Coutinho P. Animal-derived products in science and current alternatives. BIOMATERIALS ADVANCES 2023; 151:213428. [PMID: 37146527 DOI: 10.1016/j.bioadv.2023.213428] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 04/08/2023] [Accepted: 04/11/2023] [Indexed: 05/07/2023]
Abstract
More than fifty years after the 3Rs definition and despite the continuous implementation of regulatory measures, animals continue to be widely used in basic research. Their use comprises not only in vivo experiments with animal models, but also the production of a variety of supplements and products of animal origin for cell and tissue culture, cell-based assays, and therapeutics. The animal-derived products most used in basic research are fetal bovine serum (FBS), extracellular matrix proteins such as Matrigel™, and antibodies. However, their production raises several ethical issues regarding animal welfare. Additionally, their biological origin is associated with a high risk of contamination, resulting, frequently, in poor scientific data for clinical translation. These issues support the search for new animal-free products able to replace FBS, Matrigel™, and antibodies in basic research. In addition, in silico methodologies play an important role in the reduction of animal use in research by refining the data previously to in vitro and in vivo experiments. In this review, we depicted the current available animal-free alternatives in in vitro research.
Collapse
Affiliation(s)
- Ana C Duarte
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal; CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| | - Elisabete C Costa
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal
| | - Hugo A L Filipe
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal
| | - Sofia M Saraiva
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal
| | - Telma Jacinto
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal
| | - Sónia P Miguel
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal; CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| | - Maximiano P Ribeiro
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal; CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| | - Paula Coutinho
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal; CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal.
| |
Collapse
|
18
|
Computational and artificial intelligence-based methods for antibody development. Trends Pharmacol Sci 2023; 44:175-189. [PMID: 36669976 DOI: 10.1016/j.tips.2022.12.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 01/19/2023]
Abstract
Due to their high target specificity and binding affinity, therapeutic antibodies are currently the largest class of biotherapeutics. The traditional largely empirical antibody development process is, while mature and robust, cumbersome and has significant limitations. Substantial recent advances in computational and artificial intelligence (AI) technologies are now starting to overcome many of these limitations and are increasingly integrated into development pipelines. Here, we provide an overview of AI methods relevant for antibody development, including databases, computational predictors of antibody properties and structure, and computational antibody design methods with an emphasis on machine learning (ML) models, and the design of complementarity-determining region (CDR) loops, antibody structural components critical for binding.
Collapse
|
19
|
Khan A, Cowen-Rivers AI, Grosnit A, Deik DGX, Robert PA, Greiff V, Smorodina E, Rawat P, Akbar R, Dreczkowski K, Tutunov R, Bou-Ammar D, Wang J, Storkey A, Bou-Ammar H. Toward real-world automated antibody design with combinatorial Bayesian optimization. CELL REPORTS METHODS 2023; 3:100374. [PMID: 36814835 PMCID: PMC9939385 DOI: 10.1016/j.crmeth.2022.100374] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 10/08/2022] [Accepted: 12/07/2022] [Indexed: 06/14/2023]
Abstract
Antibodies are multimeric proteins capable of highly specific molecular recognition. The complementarity determining region 3 of the antibody variable heavy chain (CDRH3) often dominates antigen-binding specificity. Hence, it is a priority to design optimal antigen-specific CDRH3 to develop therapeutic antibodies. The combinatorial structure of CDRH3 sequences makes it impossible to query binding-affinity oracles exhaustively. Moreover, antibodies are expected to have high target specificity and developability. Here, we present AntBO, a combinatorial Bayesian optimization framework utilizing a CDRH3 trust region for an in silico design of antibodies with favorable developability scores. The in silico experiments on 159 antigens demonstrate that AntBO is a step toward practically viable in vitro antibody design. In under 200 calls to the oracle, AntBO suggests antibodies outperforming the best binding sequence from 6.9 million experimentally obtained CDRH3s. Additionally, AntBO finds very-high-affinity CDRH3 in only 38 protein designs while requiring no domain knowledge.
Collapse
Affiliation(s)
- Asif Khan
- School of Informatics, University of Edinburgh, Edinburgh EH8 9YL, UK
| | | | | | | | - Philippe A. Robert
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo 0315, Norway
| | - Victor Greiff
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo 0315, Norway
| | - Eva Smorodina
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo 0315, Norway
| | - Puneet Rawat
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo 0315, Norway
| | - Rahmad Akbar
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo 0315, Norway
| | | | | | - Dany Bou-Ammar
- American University of Beirut Medical Centre, Beirut 11-0236, Lebanon
| | - Jun Wang
- Huawei Noah’s Ark Lab, London N1C 4AG, UK
- University College London, London WC1E 6BT, UK
| | - Amos Storkey
- School of Informatics, University of Edinburgh, Edinburgh EH8 9YL, UK
| | - Haitham Bou-Ammar
- Huawei Noah’s Ark Lab, London N1C 4AG, UK
- University College London, London WC1E 6BT, UK
| |
Collapse
|
20
|
Ge Q, Teng M, Li X, Guo Q, Tao Y. An epitope-directed selection strategy facilitating the identification of Frizzled receptor selective antibodies. Structure 2023; 31:33-43.e5. [PMID: 36513066 DOI: 10.1016/j.str.2022.11.009] [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: 04/01/2022] [Revised: 09/27/2022] [Accepted: 11/17/2022] [Indexed: 12/15/2022]
Abstract
The lack of incorporating epitope information into the selection process makes the conventional antibody screening method less effective in identifying antibodies with desired functions. Here, we developed an epitope-directed antibody selection method by designing a directed library favoring the target epitope and a precise "counter" antigen for clearing irrelevant binders in the library. With this method, we successfully isolated an antibody, pF7_A5, that targets the less conserved region on the FZD2/7 CRD as designed. Guided by the structure of pF7_A5-FZD2CRD, a further round of evolution was conducted together with the "counter" antigen selection strategy, and ultimately, an FZD2-specific antibody and an FZD7-preferred antibody were obtained. Because of targeting the predefined functional site, all these antibodies exhibited the expected modulatory activity on the Wnt pathway. Together, the method developed here will be useful in antibody drug discovery, and the identified FZD antibodies will have clinical potential in FZD-related cancer therapy.
Collapse
Affiliation(s)
- Qiangqiang Ge
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027 Hefei, P.R. China
| | - Maikun Teng
- Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027 Hefei, P.R. China.
| | - Xu Li
- Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027 Hefei, P.R. China.
| | - Qiong Guo
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027 Hefei, P.R. China.
| | - Yuyong Tao
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027 Hefei, P.R. China; Joint Laboratory of Innovation in Life Sciences University of Science and Technology of China (USTC) and Changchun Zhuoyi Biological Co. Ltd., 130616 Changchun, P.R. China.
| |
Collapse
|
21
|
Svilenov HL, Arosio P, Menzen T, Tessier P, Sormanni P. Approaches to expand the conventional toolbox for discovery and selection of antibodies with drug-like physicochemical properties. MAbs 2023; 15:2164459. [PMID: 36629855 PMCID: PMC9839375 DOI: 10.1080/19420862.2022.2164459] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/22/2022] [Accepted: 12/29/2022] [Indexed: 01/12/2023] Open
Abstract
Antibody drugs should exhibit not only high-binding affinity for their target antigens but also favorable physicochemical drug-like properties. Such drug-like biophysical properties are essential for the successful development of antibody drug products. The traditional approaches used in antibody drug development require significant experimentation to produce, optimize, and characterize many candidates. Therefore, it is attractive to integrate new methods that can optimize the process of selecting antibodies with both desired target-binding and drug-like biophysical properties. Here, we summarize a selection of techniques that can complement the conventional toolbox used to de-risk antibody drug development. These techniques can be integrated at different stages of the antibody development process to reduce the frequency of physicochemical liabilities in antibody libraries during initial discovery and to co-optimize multiple antibody features during early-stage antibody engineering and affinity maturation. Moreover, we highlight biophysical and computational approaches that can be used to predict physical degradation pathways relevant for long-term storage and in-use stability to reduce the need for extensive experimentation.
Collapse
Affiliation(s)
- Hristo L. Svilenov
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Gent, Belgium
| | - Paolo Arosio
- Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich, Switzerland
| | - Tim Menzen
- Coriolis Pharma Research GmbH, Martinsried, 82152, Germany
| | - Peter Tessier
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Pietro Sormanni
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
22
|
Abstract
The immune systems protect vertebrates from foreign molecules or antigens, and antibodies are important mediators of this system. The sequences and structural features of antibodies vary depending on species. Many of antibodies from vertebrates, including camelids, have both heavy and light chain variable domains, but camelids also have antibodies that lack the light chains. In antibodies that lack light chains, the C-terminal variable region is called the VHH domain. Antibodies recognize antigens through six complementarity-determining regions (CDRs). The third CDR of the heavy chain (CDR-H3) is at the center of the antigen-binding site and is diverse in terms of sequence and structure. Due to the importance of antibodies in basic science as well as in medical applications, there have been many studies of CDR-H3s of antibodies that possess both light and heavy chains. However, nature of CDR-H3s of single-domain VHH antibodies is less well studied. In this chapter, we describe current knowledge of sequence-structure-function correlations of single-domain VHH antibodies with emphasis on CDR-H3. Based on the 370 crystal structures in the Protein Data Bank, we also attempt structural classification of CDR-H3 in single-domain VHH antibodies and discuss lessons learned from the ever-increasing number of the structures.
Collapse
Affiliation(s)
- Daisuke Kuroda
- Medical Device Development and Regulation Research Center, School of Engineering, The University of Tokyo, Tokyo, Japan.
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan.
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, Japan.
| | - Kouhei Tsumoto
- Medical Device Development and Regulation Research Center, School of Engineering, The University of Tokyo, Tokyo, Japan.
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan.
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, Japan.
- Laboratory of Medical Proteomics, Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
23
|
Zhang C, Dalby PA. Assessing and Engineering Antibody Stability Using Experimental and Computational Methods. Methods Mol Biol 2023; 2552:165-197. [PMID: 36346592 DOI: 10.1007/978-1-0716-2609-2_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Engineering increased stability into antibodies can improve their developability. While a range of properties need to be optimized, thermal stability and aggregation are two key factors that affect the antibody yield, purity, and specificity throughout the development and manufacturing pipeline. Therefore, an ideal goal would be to apply protein engineering methods early-on, such as in parallel to affinity maturation, to screen out potential drug molecules with the desired conformational and colloidal stability. This chapter introduces our methods to computationally characterize an antibody Fab fragment, propose stabilizing variants, and then experimentally verify these predictions.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Biochemical Engineering, University College London, London, UK
| | - Paul Anthony Dalby
- Department of Biochemical Engineering, University College London, London, UK.
| |
Collapse
|
24
|
Comeau SR, Thorsteinson N, Kumar S. Structural Considerations in Affinity Maturation of Antibody-Based Biotherapeutic Candidates. Methods Mol Biol 2023; 2552:309-321. [PMID: 36346600 DOI: 10.1007/978-1-0716-2609-2_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Affinity maturation is an important stage in biologic drug discovery as is the natural process of generating an immune response inside the human body. In this chapter, we describe in silico approaches to affinity maturation via a worked example. Both advantages and limitations of the computational methods used are critically examined. Furthermore, construction of affinity maturation libraries and how their outputs might be implemented in an experimental setting are also described. It should be noted that structure-based design of biologic drugs is an emerging field and the tools currently available require further development. Furthermore, there are no standardized structure-based strategies yet for antibody affinity maturation as this research relies heavily on scientific logic as well as creative intuition.
Collapse
Affiliation(s)
- Stephen R Comeau
- Computational Biochemistry and Bioinformatics Group, Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Nels Thorsteinson
- Scientific Services Manager, Biologics, Chemical Computing Group ULC, Montreal, QC, Canada
| | - Sandeep Kumar
- Computational Biochemistry and Bioinformatics Group, Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA.
| |
Collapse
|
25
|
Robert PA, Akbar R, Frank R, Pavlović M, Widrich M, Snapkov I, Slabodkin A, Chernigovskaya M, Scheffer L, Smorodina E, Rawat P, Mehta BB, Vu MH, Mathisen IF, Prósz A, Abram K, Olar A, Miho E, Haug DTT, Lund-Johansen F, Hochreiter S, Haff IH, Klambauer G, Sandve GK, Greiff V. Unconstrained generation of synthetic antibody-antigen structures to guide machine learning methodology for antibody specificity prediction. NATURE COMPUTATIONAL SCIENCE 2022; 2:845-865. [PMID: 38177393 DOI: 10.1038/s43588-022-00372-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 11/09/2022] [Indexed: 01/06/2024]
Abstract
Machine learning (ML) is a key technology for accurate prediction of antibody-antigen binding. Two orthogonal problems hinder the application of ML to antibody-specificity prediction and the benchmarking thereof: the lack of a unified ML formalization of immunological antibody-specificity prediction problems and the unavailability of large-scale synthetic datasets to benchmark real-world relevant ML methods and dataset design. Here we developed the Absolut! software suite that enables parameter-based unconstrained generation of synthetic lattice-based three-dimensional antibody-antigen-binding structures with ground-truth access to conformational paratope, epitope and affinity. We formalized common immunological antibody-specificity prediction problems as ML tasks and confirmed that for both sequence- and structure-based tasks, accuracy-based rankings of ML methods trained on experimental data hold for ML methods trained on Absolut!-generated data. The Absolut! framework has the potential to enable real-world relevant development and benchmarking of ML strategies for biotherapeutics design.
Collapse
Affiliation(s)
- Philippe A Robert
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway.
| | - Rahmad Akbar
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Robert Frank
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | | | - Michael Widrich
- ELLIS Unit Linz and LIT AI Lab, Institute for Machine Learning, Johannes Kepler University Linz, Linz, Austria
| | - Igor Snapkov
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Andrei Slabodkin
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Maria Chernigovskaya
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | | | - Eva Smorodina
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Puneet Rawat
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Brij Bhushan Mehta
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Mai Ha Vu
- Department of Linguistics and Scandinavian Studies, University of Oslo, Oslo, Norway
| | | | - Aurél Prósz
- Danish Cancer Society Research Center, Translational Cancer Genomics, Copenhagen, Denmark
| | - Krzysztof Abram
- The Novo Nordisk Foundation Center for Biosustainability, Autoflow, DTU Biosustain and IT University of Copenhagen, Copenhagen, Denmark
| | - Alex Olar
- Department of Complex Systems in Physics, Eötvös Loránd University, Budapest, Hungary
| | - Enkelejda Miho
- Institute of Medical Engineering and Medical Informatics, School of Life Sciences, FHNW University of Applied Sciences and Arts Northwestern Switzerland, Muttenz, Switzerland
- aiNET GmbH, Basel, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | | | | | - Sepp Hochreiter
- ELLIS Unit Linz and LIT AI Lab, Institute for Machine Learning, Johannes Kepler University Linz, Linz, Austria
- Institute of Advanced Research in Artificial Intelligence (IARAI), Vienna, Austria
| | | | - Günter Klambauer
- ELLIS Unit Linz and LIT AI Lab, Institute for Machine Learning, Johannes Kepler University Linz, Linz, Austria
| | | | - Victor Greiff
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
26
|
Two Epitope Regions Revealed in the Complex of IL-17A and Anti-IL-17A V HH Domain. Int J Mol Sci 2022; 23:ijms232314904. [PMID: 36499233 PMCID: PMC9738047 DOI: 10.3390/ijms232314904] [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: 10/11/2022] [Revised: 11/22/2022] [Accepted: 11/25/2022] [Indexed: 11/30/2022] Open
Abstract
Interleukin-17 (IL-17) is a cytokine produced by the Th17 cells. It is involved in chronic inflammation in patients with autoimmune diseases, such as rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, and psoriasis. The antibodies targeting IL-17 and/or IL-17R are therapy tools for these diseases. Netakimab is an IL-17A-specific antibody containing a Lama glama VHH derivative domain and a VL variable domain. We have determined the crystal structure of the IL-17A-specific VHH domain in complex with IL-17A at 2.85 Å resolution. Certain amino acid residues of the three complementary-determining regions of the VHH domain form a network of solvent-inaccessible hydrogen bonds with two epitope regions of IL-17A. The β-turn of IL-17A, which forms the so-called epitope-1, appears to be the main region of IL-17A interaction with the antibody. Contacts formed by the IL-17A mobile C-terminal region residues (epitope-2) further stabilize the antibody-antigen complex.
Collapse
|
27
|
Aguilar Rangel M, Bedwell A, Costanzi E, Taylor RJ, Russo R, Bernardes GJL, Ricagno S, Frydman J, Vendruscolo M, Sormanni P. Fragment-based computational design of antibodies targeting structured epitopes. SCIENCE ADVANCES 2022; 8:eabp9540. [PMID: 36367941 PMCID: PMC9651861 DOI: 10.1126/sciadv.abp9540] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 09/23/2022] [Indexed: 06/16/2023]
Abstract
De novo design methods hold the promise of reducing the time and cost of antibody discovery while enabling the facile and precise targeting of predetermined epitopes. Here, we describe a fragment-based method for the combinatorial design of antibody binding loops and their grafting onto antibody scaffolds. We designed and tested six single-domain antibodies targeting different epitopes on three antigens, including the receptor-binding domain of the SARS-CoV-2 spike protein. Biophysical characterization showed that all designs are stable and bind their intended targets with affinities in the nanomolar range without in vitro affinity maturation. We further discuss how a high-resolution input antigen structure is not required, as similar predictions are obtained when the input is a crystal structure or a computer-generated model. This computational procedure, which readily runs on a laptop, provides a starting point for the rapid generation of lead antibodies binding to preselected epitopes.
Collapse
Affiliation(s)
- Mauricio Aguilar Rangel
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Alice Bedwell
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Elisa Costanzi
- Department of Bioscience, Università degli Studi di Milano, Milano 20133, Italy
| | - Ross J. Taylor
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Rosaria Russo
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milano 20122, Italy
| | - Gonçalo J. L. Bernardes
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Stefano Ricagno
- Department of Bioscience, Università degli Studi di Milano, Milano 20133, Italy
- Institute of Molecular and Translational Cardiology, IRCCS Policlinico San Donato, Milan 20097, Italy
| | - Judith Frydman
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Pietro Sormanni
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| |
Collapse
|
28
|
Chauhan VM, Pantazes RJ. MutDock: A computational docking approach for fixed-backbone protein scaffold design. Front Mol Biosci 2022; 9:933400. [PMID: 36106019 PMCID: PMC9465448 DOI: 10.3389/fmolb.2022.933400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Despite the successes of antibodies as therapeutic binding proteins, they still face production and design challenges. Alternative binding scaffolds of smaller size have been developed to overcome these issues. A subset of these alternative scaffolds recognizes target molecules through mutations to a set of surface resides, which does not alter their backbone structures. While the computational design of antibodies for target epitopes has been explored in depth, the same has not been done for alternative scaffolds. The commonly used dock-and-mutate approach for binding proteins, including antibodies, is limited because it uses a constant sequence and structure representation of the scaffold. Docking fixed-backbone scaffolds with a varied group of surface amino acids increases the chances of identifying superior starting poses that can be improved with subsequent mutations. In this work, we have developed MutDock, a novel computational approach that simultaneously docks and mutates fixed backbone scaffolds for binding a target epitope by identifying a minimum number of hydrogen bonds. The approach is broadly divided into two steps. The first step uses pairwise distance alignment of hydrogen bond-forming areas of scaffold residues and compatible epitope atoms. This step considers both native and mutated rotamers of scaffold residues. The second step mutates clashing variable interface residues and thermodynamically unfavorable residues to create additional strong interactions. MutDock was used to dock two scaffolds, namely, Affibodies and DARPins, with ten randomly selected antigens. The energies of the docked poses were minimized and binding energies were compared with docked poses from ZDOCK and HADDOCK. The top MutDock poses consisted of higher and comparable binding energies than the top ZDOCK and HADDOCK poses, respectively. This work contributes to the discovery of novel binders based on smaller-sized, fixed-backbone protein scaffolds.
Collapse
|
29
|
Ahmad B, Choi S. Unraveling the Tomaralimab Epitope on the Toll-like Receptor 2 via Molecular Dynamics and Deep Learning. ACS OMEGA 2022; 7:28226-28237. [PMID: 35990491 PMCID: PMC9386714 DOI: 10.1021/acsomega.2c02559] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/22/2022] [Indexed: 06/15/2023]
Abstract
Tomaralimab (OPN-305) is the first humanized immunoglobulin G4 monoclonal antibody against TLR2 and is designed to prevent inflammation that is driven by inappropriate or excessive activation of innate immune pathways. Here, we constructed a homology model of Tomaralimab and its complex with TLR2 at different mapped epitopes and unraveled their behavior at the atomistic level. Furthermore, we predicted a novel epitope (leucine-rich region 9-12) near the lipopeptide-binding site that can be targeted and studied for the utility of therapeutic antibodies. A geometric deep learning algorithm was used to envisage Tomaralimab binding affinity changes upon mutation. There was a significant difference in binding affinity for Tomaralimab following epitope-mutated alanine substitutions of Val266, Pro294, Arg295, Asn319, Pro326, and His372. Using deep learning-based ΔΔG prediction, we computationally contrasted human TLR2-TLR2, TLR2-TLR1, and TLR2-TLR6 dimerization. These results reveal the mechanism that underlies Tomaralimab binding to TLR2 and should help to design structure-based mimics or bispecific antibodies that can be used to inhibit both lipopeptide-binding and TLR2 dimerization.
Collapse
Affiliation(s)
- Bilal Ahmad
- Department
of Molecular Science and Technology, Ajou
University, Suwon 16499, Korea
- S&K
Therapeutics, Ajou University
Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon 16502, Korea
| | - Sangdun Choi
- Department
of Molecular Science and Technology, Ajou
University, Suwon 16499, Korea
- S&K
Therapeutics, Ajou University
Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon 16502, Korea
| |
Collapse
|
30
|
Hsueh SCC, Aina A, Roman AY, Cashman NR, Peng X, Plotkin SS. Optimizing Epitope Conformational Ensembles Using α-Synuclein Cyclic Peptide "Glycindel" Scaffolds: A Customized Immunogen Method for Generating Oligomer-Selective Antibodies for Parkinson's Disease. ACS Chem Neurosci 2022; 13:2261-2280. [PMID: 35840132 DOI: 10.1021/acschemneuro.1c00567] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Effectively presenting epitopes on immunogens, in order to raise conformationally selective antibodies through active immunization, is a central problem in treating protein misfolding diseases, particularly neurodegenerative diseases such as Alzheimer's disease or Parkinson's disease. We seek to selectively target conformations enriched in toxic, oligomeric propagating species while sparing the healthy forms of the protein that are often more abundant. To this end, we computationally modeled scaffolded epitopes in cyclic peptides by inserting/deleting a variable number of flanking glycines ("glycindels") to best mimic a misfolding-specific conformation of an epitope of α-synuclein enriched in the oligomer ensemble, as characterized by a region most readily disordered and solvent-exposed in a stressed, partially denatured protofibril. We screen and rank the cyclic peptide scaffolds of α-synuclein in silico based on their ensemble overlap properties with the fibril, oligomer-model and isolated monomer ensembles. We present experimental data of seeded aggregation that support nucleation rates consistent with computationally predicted cyclic peptide conformational similarity. We also introduce a method for screening against structured off-pathway targets in the human proteome by selecting scaffolds with minimal conformational similarity between their epitope and the same solvent-exposed primary sequence in structured human proteins. Different cyclic peptide scaffolds with variable numbers of glycines are predicted computationally to have markedly different conformational ensembles. Ensemble comparison and overlap were quantified by the Jensen-Shannon divergence and a new measure introduced here, the embedding depth, which determines the extent to which a given ensemble is subsumed by another ensemble and which may be a more useful measure in developing immunogens that confer conformational selectivity to an antibody.
Collapse
Affiliation(s)
- Shawn C C Hsueh
- Department of Physics and Astronomy, The University of British Columbia, Vancouver, BC V6T 1Z1, Canada
| | - Adekunle Aina
- Department of Physics and Astronomy, The University of British Columbia, Vancouver, BC V6T 1Z1, Canada
| | - Andrei Yu Roman
- Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, BC V6T 2B5, Canada
| | - Neil R Cashman
- Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, BC V6T 2B5, Canada
| | - Xubiao Peng
- Department of Physics and Astronomy, The University of British Columbia, Vancouver, BC V6T 1Z1, Canada
| | - Steven S Plotkin
- Department of Physics and Astronomy, The University of British Columbia, Vancouver, BC V6T 1Z1, Canada.,Genome Science and Technology Program, The University of British Columbia, Vancouver, BC V6T 1Z1, Canada
| |
Collapse
|
31
|
Wong MTY, Kelm S, Liu X, Taylor RD, Baker T, Essex JW. Higher Affinity Antibodies Bind With Lower Hydration and Flexibility in Large Scale Simulations. Front Immunol 2022; 13:884110. [PMID: 35707541 PMCID: PMC9190259 DOI: 10.3389/fimmu.2022.884110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/22/2022] [Indexed: 11/25/2022] Open
Abstract
We have carried out a long-timescale simulation study on crystal structures of nine antibody-antigen pairs, in antigen-bound and antibody-only forms, using molecular dynamics with enhanced sampling and an explicit water model to explore interface conformation and hydration. By combining atomic level simulation and replica exchange to enable full protein flexibility, we find significant numbers of bridging water molecules at the antibody-antigen interface. Additionally, a higher proportion of interactions excluding bulk waters and a lower degree of antigen bound CDR conformational sampling are correlated with higher antibody affinity. The CDR sampling supports enthalpically driven antibody binding, as opposed to entropically driven, in that the difference between antigen bound and unbound conformations do not correlate with affinity. We thus propose that interactions with waters and CDR sampling are aspects of the interface that may moderate antibody-antigen binding, and that explicit hydration and CDR flexibility should be considered to improve antibody affinity prediction and computational design workflows.
Collapse
Affiliation(s)
- Mabel T. Y. Wong
- School of Chemistry, University of Southampton, Southampton, United Kingdom
| | | | | | | | | | - Jonathan W. Essex
- School of Chemistry, University of Southampton, Southampton, United Kingdom
- *Correspondence: Jonathan W. Essex,
| |
Collapse
|
32
|
Hummer AM, Abanades B, Deane CM. Advances in computational structure-based antibody design. Curr Opin Struct Biol 2022; 74:102379. [PMID: 35490649 DOI: 10.1016/j.sbi.2022.102379] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/28/2022] [Accepted: 03/17/2022] [Indexed: 12/12/2022]
Abstract
Antibodies are currently the most important class of biotherapeutics and are used to treat numerous diseases. Recent advances in computational methods are ushering in a new era of antibody design, driven in part by accurate structure prediction. Previously, structure-based antibody design has been limited to a relatively small number of cases where accurate structures or models of both the target antigen and antibody were available. As we move towards a time where it is possible to accurately model most antibodies and antigens, and to reliably predict their binding site, there is vast potential for true computational antibody design. In this review, we describe the latest methods that promise to launch a paradigm shift towards entirely in silico structure-based antibody design.
Collapse
Affiliation(s)
- Alissa M Hummer
- Oxford Protein Informatics Group, Department of Statistics, University of Oxford, Oxford OX1 3LB, UK. https://twitter.com/@AlissaHummer
| | - Brennan Abanades
- Oxford Protein Informatics Group, Department of Statistics, University of Oxford, Oxford OX1 3LB, UK. https://twitter.com/@brennanaba
| | - Charlotte M Deane
- Oxford Protein Informatics Group, Department of Statistics, University of Oxford, Oxford OX1 3LB, UK.
| |
Collapse
|
33
|
Sepahdar Z, Saghiri R, Miroliaei M, Salimi M. In silico approach to probe the binding affinity between OMVs harboring the Z EGFR affibody and the EGF receptor. J Mol Model 2022; 28:113. [PMID: 35381900 DOI: 10.1007/s00894-022-05043-9] [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: 09/26/2021] [Accepted: 01/25/2022] [Indexed: 11/27/2022]
Abstract
There is a growing interest in designing a nanocarrier containing an EGFR targeting affibody to direct toward cancer cells. Here, cytolysin A was cloned at the N-terminus of ZEGFR:1907 affibody to guarantee its surface presentation on the OMVs while targeting the epidermal growth factor receptors (EGFRs). A separate construct including a fusogenic peptide (GALA) was also designed for the endosomal escape of the nanocarrier. Binding of the two constructs ClyA-affiEGFR and ClyA-affiEGFR-GALA to domain III of EGFR was investigated using molecular docking and molecular dynamic simulations. The higher stability of the ClyA-affiEGFR-GALA/EGFR as compared to the ClyA-affiEGFR/EGFR complex was evident. The ClyA-affiEGFR-GALA structure showed a higher RMSD during the first half of the simulation time implying a much less stable behavior. Plateau state of the radius of gyration plot of ClyA-affiEGFR-GALA confirmed a well-folded structure in the presence of the GALA sequence. Solvent accessible surface area for both proteins was in the same range. The data obtained from hydrogen bond analysis revealed a more equilibrated and stable form of the ClyA-affiEGFR-GALA structure upon interaction with EGFR. The data provided here was a requisite for our biological evaluation of the synthesized constructs as a component of a novel drug delivery system.
Collapse
Affiliation(s)
- Zahra Sepahdar
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Reza Saghiri
- Biochemistry Department, Pasteur Institute of Iran, Tehran, Iran
| | - Mehran Miroliaei
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Mona Salimi
- Physiology and Pharmacology Department, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
34
|
Structural and functional insights into a novel pre-clinical-stage antibody targeting IL-17A for treatment of autoimmune diseases. Int J Biol Macromol 2022; 202:529-538. [DOI: 10.1016/j.ijbiomac.2022.01.119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 02/04/2023]
|
35
|
Magalhães ICL, Souza PFN, Marques LEC, Girão NM, Araújo FMC, Guedes MIF. New insights into the recombinant proteins and monoclonal antibodies employed to immunodiagnosis and control of Zika virus infection: A review. Int J Biol Macromol 2022; 200:139-150. [PMID: 34998869 DOI: 10.1016/j.ijbiomac.2021.12.196] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 12/11/2022]
Abstract
An emergent positive-stranded RNA virus, transmitted by mosquitoes with its first case of vertical transmission confirmed in 2015 in Brazil. The Zika virus (ZIKV) fever has received particular attention, mainly related to neurological diseases such as microcephaly in newborns. However, the laboratory diagnosis for ZIKV still faces some challenges due to its cross-reactivity with other flaviviruses, requiring a correct and differential diagnosis, contributing to the good prognosis of patients, especially in pregnant women. Among these, for early diagnosis, the CDC considers the RT-PCR the gold standard, more sensitive and specific, but expensive. Serological tests for the diagnosis of ZIKV can also be found beyond the period when the viral components are detectable in the serum. Inputs to produce more sensitive and specific diagnostic kits and the possibility of viral detection in less invasive samples are among the objectives of recent research on ZIKV. This review outlines recent advances in developing recombinant antigen and antibody-based diagnostic tools for the main flaviviruses in Northeast Brazil, such as ZIKV and Dengue virus (DENV).
Collapse
Affiliation(s)
- Ilana C L Magalhães
- Biotechnology and Molecular Biology Laboratory, State University of Ceara, Fortaleza, Ceara, Brazil.
| | - Pedro F N Souza
- Department of Biochemistry and Molecular Biology, Laboratory of Plant Defense Proteins, Federal University of Ceara, Fortaleza, Brazil.
| | - Lívia E C Marques
- Biotechnology and Molecular Biology Laboratory, State University of Ceara, Fortaleza, Ceara, Brazil
| | - Nicolas M Girão
- Biotechnology and Molecular Biology Laboratory, State University of Ceara, Fortaleza, Ceara, Brazil
| | | | - Maria Izabel F Guedes
- Biotechnology and Molecular Biology Laboratory, State University of Ceara, Fortaleza, Ceara, Brazil
| |
Collapse
|
36
|
Liu B, Yang D. Easily Established and Multifunctional Synthetic Nanobody Libraries as Research Tools. Int J Mol Sci 2022; 23:ijms23031482. [PMID: 35163405 PMCID: PMC8835997 DOI: 10.3390/ijms23031482] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 02/06/2023] Open
Abstract
Nanobodies, or VHHs, refer to the antigen-binding domain of heavy-chain antibodies (HCAbs) from camelids. They have been widely used as research tools for protein purification and structure determination due to their small size, high specificity, and high stability, overcoming limitations with conventional antibody fragments. However, animal immunization and subsequent retrieval of antigen-specific nanobodies are expensive and complicated. Construction of synthetic nanobody libraries using DNA oligonucleotides is a cost-effective alternative for immunization libraries and shows great potential in identifying antigen-specific or even conformation-specific nanobodies. This review summarizes and analyses synthetic nanobody libraries in the current literature, including library design and biopanning methods, and further discusses applications of antigen-specific nanobodies obtained from synthetic libraries to research.
Collapse
|
37
|
Akbar R, Bashour H, Rawat P, Robert PA, Smorodina E, Cotet TS, Flem-Karlsen K, Frank R, Mehta BB, Vu MH, Zengin T, Gutierrez-Marcos J, Lund-Johansen F, Andersen JT, Greiff V. Progress and challenges for the machine learning-based design of fit-for-purpose monoclonal antibodies. MAbs 2022; 14:2008790. [PMID: 35293269 PMCID: PMC8928824 DOI: 10.1080/19420862.2021.2008790] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 11/04/2021] [Accepted: 11/17/2021] [Indexed: 12/15/2022] Open
Abstract
Although the therapeutic efficacy and commercial success of monoclonal antibodies (mAbs) are tremendous, the design and discovery of new candidates remain a time and cost-intensive endeavor. In this regard, progress in the generation of data describing antigen binding and developability, computational methodology, and artificial intelligence may pave the way for a new era of in silico on-demand immunotherapeutics design and discovery. Here, we argue that the main necessary machine learning (ML) components for an in silico mAb sequence generator are: understanding of the rules of mAb-antigen binding, capacity to modularly combine mAb design parameters, and algorithms for unconstrained parameter-driven in silico mAb sequence synthesis. We review the current progress toward the realization of these necessary components and discuss the challenges that must be overcome to allow the on-demand ML-based discovery and design of fit-for-purpose mAb therapeutic candidates.
Collapse
Affiliation(s)
- Rahmad Akbar
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Habib Bashour
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Puneet Rawat
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Philippe A. Robert
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Eva Smorodina
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Russia
| | | | - Karine Flem-Karlsen
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Department of Pharmacology, University of Oslo and Oslo University Hospital, Norway
| | - Robert Frank
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Brij Bhushan Mehta
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Mai Ha Vu
- Department of Linguistics and Scandinavian Studies, University of Oslo, Norway
| | - Talip Zengin
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Bioinformatics, Mugla Sitki Kocman University, Turkey
| | | | | | - Jan Terje Andersen
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Department of Pharmacology, University of Oslo and Oslo University Hospital, Norway
| | - Victor Greiff
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| |
Collapse
|
38
|
Lu S, Li Y, Wang F, Nan X, Zhang S. Leveraging Sequential and Spatial Neighbors Information by Using CNNs Linked With GCNs for Paratope Prediction. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:68-74. [PMID: 34029193 DOI: 10.1109/tcbb.2021.3083001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Antibodies consisting of variable and constant regions, are a special type of proteins playing a vital role in immune system of the vertebrate. They have the remarkable ability to bind a large range of diverse antigens with extraordinary affinity and specificity. This malleability of binding makes antibodies an important class of biological drugs and biomarkers. In this article, we propose a method to identify which amino acid residues of an antibody directly interact with its associated antigen based on the features from sequence and structure. Our algorithm uses convolution neural networks (CNNs) linked with graph convolution networks (GCNs) to make use of information from both sequential and spatial neighbors to understand more about the local environment of target amino acid residue. Furthermore, we process the antigen partner of an antibody by employing an attention layer. Our method improves on the state-of-the-art methodology.
Collapse
|
39
|
Kielczewska A, D'Angelo I, Amador MS, Wang T, Sudom A, Min X, Rathanaswami P, Pigott C, Foltz IN. Development of a potent high-affinity human therapeutic antibody via novel application of recombination signal sequence-based affinity maturation. J Biol Chem 2021; 298:101533. [PMID: 34973336 PMCID: PMC8808179 DOI: 10.1016/j.jbc.2021.101533] [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: 12/14/2021] [Accepted: 12/20/2021] [Indexed: 12/01/2022] Open
Abstract
Therapeutic antibody development requires discovery of an antibody molecule with desired specificities and drug-like properties. For toxicological studies, a therapeutic antibody must bind the ortholog antigen with a similar affinity to the human target to enable relevant dosing regimens, and antibodies falling short of this affinity design goal may not progress as therapeutic leads. Herein, we report the novel use of mammalian recombination signal sequence (RSS)–directed recombination for complementarity-determining region–targeted protein engineering combined with mammalian display to close the species affinity gap of human interleukin (IL)-13 antibody 731. This fully human antibody has not progressed as a therapeutic in part because of a 400-fold species affinity gap. Using this nonhypothesis-driven affinity maturation method, we generated multiple antibody variants with improved IL-13 affinity, including the highest affinity antibody reported to date (34 fM). Resolution of a cocrystal structure of the optimized antibody with the cynomolgus monkey (or nonhuman primate) IL-13 protein revealed that the RSS-derived mutations introduced multiple successive amino-acid substitutions resulting in a de novo formation of a π–π stacking–based protein–protein interaction between the affinity-matured antibody heavy chain and helix C on IL-13, as well as an introduction of an interface-distant residue, which enhanced the light chain–binding affinity to target. These mutations synergized binding of heavy and light chains to the target protein, resulting in a remarkably tight interaction, and providing a proof of concept for a new method of protein engineering, based on synergizing a mammalian display platform with novel RSS-mediated library generation.
Collapse
Affiliation(s)
| | - Igor D'Angelo
- Amgen Inc, Therapeutic Discovery, Thousand Oaks, California, USA
| | - Maria Sheena Amador
- Amgen British Columbia, Therapeutic Discovery, Burnaby, British Columbia, Canada
| | - Tina Wang
- Amgen British Columbia, Therapeutic Discovery, Burnaby, British Columbia, Canada
| | - Athena Sudom
- Amgen San Francisco, Therapeutic Discovery, San Francisco, California, USA
| | - Xiaoshan Min
- Amgen San Francisco, Therapeutic Discovery, San Francisco, California, USA
| | | | - Craig Pigott
- Innovative Targeting Solutions, Burnaby, British Columbia, Canada
| | - Ian N Foltz
- Amgen British Columbia, Therapeutic Discovery, Burnaby, British Columbia, Canada
| |
Collapse
|
40
|
Prabakaran P, Rao SP, Wendt M. Animal immunization merges with innovative technologies: A new paradigm shift in antibody discovery. MAbs 2021; 13:1924347. [PMID: 33947305 PMCID: PMC8118498 DOI: 10.1080/19420862.2021.1924347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Animal-derived antibody sources, particularly, transgenic mice that are engineered with human immunoglobulin loci, along with advanced antibody generation technology platforms have facilitated the discoveries of human antibody therapeutics. For example, isolation of antigen-specific B cells, microfluidics, and next-generation sequencing have emerged as powerful tools for identifying and developing monoclonal antibodies (mAbs). These technologies enable not only antibody drug discovery but also lead to the understanding of B cell biology, immune mechanisms and immunogenetics of antibodies. In this perspective article, we discuss the scientific merits of animal immunization combined with advanced methods for antibody generation as compared to animal-free alternatives through in-vitro-generated antibody libraries. The knowledge gained from animal-derived antibodies concerning the recombinational diversity, somatic hypermutation patterns, and physiochemical properties is found more valuable and prerequisite for developing in vitro libraries, as well as artificial intelligence/machine learning methods to discover safe and effective mAbs.
Collapse
Affiliation(s)
- Ponraj Prabakaran
- Biologics Research US, Global Large Molecules Research, Sanofi, Framingham, MA, USA
| | - Sambasiva P Rao
- Biologics Research US, Global Large Molecules Research, Sanofi, Framingham, MA, USA
| | - Maria Wendt
- Biologics Research US, Global Large Molecules Research, Sanofi, Framingham, MA, USA
| |
Collapse
|
41
|
Pertseva M, Gao B, Neumeier D, Yermanos A, Reddy ST. Applications of Machine and Deep Learning in Adaptive Immunity. Annu Rev Chem Biomol Eng 2021; 12:39-62. [PMID: 33852352 DOI: 10.1146/annurev-chembioeng-101420-125021] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Adaptive immunity is mediated by lymphocyte B and T cells, which respectively express a vast and diverse repertoire of B cell and T cell receptors and, in conjunction with peptide antigen presentation through major histocompatibility complexes (MHCs), can recognize and respond to pathogens and diseased cells. In recent years, advances in deep sequencing have led to a massive increase in the amount of adaptive immune receptor repertoire data; additionally, proteomics techniques have led to a wealth of data on peptide-MHC presentation. These large-scale data sets are now making it possible to train machine and deep learning models, which can be used to identify complex and high-dimensional patterns in immune repertoires. This article introduces adaptive immune repertoires and machine and deep learning related to biological sequence data and then summarizes the many applications in this field, which span from predicting the immunological status of a host to the antigen specificity of individual receptors and the engineering of immunotherapeutics.
Collapse
Affiliation(s)
- Margarita Pertseva
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland; .,Life Science Zurich Graduate School, ETH Zurich and University of Zurich, 8006 Zurich, Switzerland
| | - Beichen Gao
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland;
| | - Daniel Neumeier
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland;
| | - Alexander Yermanos
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland; .,Department of Pathology and Immunology, University of Geneva, 1205 Geneva, Switzerland.,Department of Biology, Institute of Microbiology and Immunology, ETH Zurich, 8093 Zurich, Switzerland
| | - Sai T Reddy
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland;
| |
Collapse
|
42
|
Kim DG, Choi Y, Kim HS. Epitopes of Protein Binders Are Related to the Structural Flexibility of a Target Protein Surface. J Chem Inf Model 2021; 61:2099-2107. [PMID: 33829791 DOI: 10.1021/acs.jcim.0c01397] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Protein binders including antibodies are known not to bind to random sites of target proteins, and their functional effectiveness mainly depends on the binding region, called the epitope. For the development of protein binders with desired functions, it is thus critical to understand which surface region protein binders prefer (or do not prefer) to bind. The current methods for epitope prediction focus on static indicators such as structural geometry or amino acid propensity, whereas protein binding events are in fact a consequence of dynamic interactions. Here, we demonstrate that the preference for a binding site by protein binders is strongly related to the structural flexibility of a target protein surface. Molecular dynamics simulations on unbound forms of antigen structures revealed that the antigen surface in direct contact with antibodies is less flexible than the rest of the surface. This tendency was shown to be similar in other non-antibody protein binders such as affibody, DARPin, monobody, and repebody. We also found that the relatedness of epitopes to the structural flexibility of a target protein surface is dependent on the secondary structure elements of paratopes. Monobody and repebody, whose binding sites are composed of β-strands, distinctively prefer to bind to a relatively more rigid region of a target protein. These observations enabled us to develop a simple epitope prediction method which shows a comparable performance to the commonly used ones.
Collapse
Affiliation(s)
- Dong-Gun Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Yoonjoo Choi
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun, Jeonnam 58128, Republic of Korea
| | - Hak-Sung Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| |
Collapse
|
43
|
Le HT, Do PC, Le L. Grafting Methionine on 1F1 Ab Increases the Broad-Activity on HA Structural-Conserved Residues of H1, H2, and H3 Influenza a Viruses. Evol Bioinform Online 2021; 17:11769343211003082. [PMID: 33795930 PMCID: PMC7975486 DOI: 10.1177/11769343211003082] [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: 11/05/2020] [Accepted: 02/24/2021] [Indexed: 11/27/2022] Open
Abstract
A high level of mutation enables the influenza A virus to resist antibiotics
previously effective against the influenza A virus. A portion of the structure
of hemagglutinin HA is assumed to be well-conserved to maintain its role in
cellular fusion, and the structure tends to be more conserved than sequence. We
designed peptide inhibitors to target the conserved residues on the HA surface,
which were identified based on structural alignment. Most of the conserved and
strongly similar residues are located in the receptor-binding and esterase
regions on the HA1 domain In a later step, fragments of anti-HA antibodies were
gathered and screened for the binding ability to the found conserved residues.
As a result, Methionine amino acid got the best docking score within the −2.8 Å
radius of Van der Waals when it is interacting with Tyrosine, Arginine, and
Glutamic acid. Then, the binding affinity and spectrum of the fragments were
enhanced by grafting hotspot amino acid into the fragments to form peptide
inhibitors. Our peptide inhibitor was able to form in silico contact with a
structurally conserved region across H1, H2, and H3 HA, with the binding site at
the boundary between HA1 and HA2 domains, spreading across different monomers,
suggesting a new target for designing broad-spectrum antibody and vaccine. This
research presents an affordable method to design broad-spectrum peptide
inhibitors using fragments of an antibody as a scaffold.
Collapse
Affiliation(s)
- Hoa Thanh Le
- School of Biotechnology, International University, Ho Chi Minh City, Vietnam.,Vietnam National University, Ho Chi Minh City, Vietnam
| | - Phuc-Chau Do
- School of Biotechnology, International University, Ho Chi Minh City, Vietnam.,Vietnam National University, Ho Chi Minh City, Vietnam
| | - Ly Le
- School of Biotechnology, International University, Ho Chi Minh City, Vietnam.,Vietnam National University, Ho Chi Minh City, Vietnam.,Vingroup Big Data Institute, Hanoi, Vietnam
| |
Collapse
|
44
|
Laustsen AH, Greiff V, Karatt-Vellatt A, Muyldermans S, Jenkins TP. Animal Immunization, in Vitro Display Technologies, and Machine Learning for Antibody Discovery. Trends Biotechnol 2021; 39:1263-1273. [PMID: 33775449 DOI: 10.1016/j.tibtech.2021.03.003] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023]
Abstract
For years, a discussion has persevered on the benefits and drawbacks of antibody discovery using animal immunization versus in vitro selection from non-animal-derived recombinant repertoires using display technologies. While it has been argued that using recombinant display libraries can reduce animal consumption, we hold that the number of animals used in immunization campaigns is dwarfed by the number sacrificed during preclinical studies. Thus, improving quality control of antibodies before entering in vivo studies will have a larger impact on animal consumption. Both animal immunization and recombinant repertoires present unique advantages for discovering antibodies that are fit for purpose. Furthermore, we anticipate that machine learning will play a significant role within discovery workflows, refining current antibody discovery practices.
Collapse
Affiliation(s)
- Andreas H Laustsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark.
| | - Victor Greiff
- Department of Immunology, University of Oslo, Oslo, Norway
| | | | - Serge Muyldermans
- Department of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Timothy P Jenkins
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
45
|
Akbar R, Robert PA, Pavlović M, Jeliazkov JR, Snapkov I, Slabodkin A, Weber CR, Scheffer L, Miho E, Haff IH, Haug DTT, Lund-Johansen F, Safonova Y, Sandve GK, Greiff V. A compact vocabulary of paratope-epitope interactions enables predictability of antibody-antigen binding. Cell Rep 2021; 34:108856. [PMID: 33730590 DOI: 10.1016/j.celrep.2021.108856] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 11/29/2020] [Accepted: 02/22/2021] [Indexed: 12/16/2022] Open
Abstract
Antibody-antigen binding relies on the specific interaction of amino acids at the paratope-epitope interface. The predictability of antibody-antigen binding is a prerequisite for de novo antibody and (neo-)epitope design. A fundamental premise for the predictability of antibody-antigen binding is the existence of paratope-epitope interaction motifs that are universally shared among antibody-antigen structures. In a dataset of non-redundant antibody-antigen structures, we identify structural interaction motifs, which together compose a commonly shared structure-based vocabulary of paratope-epitope interactions. We show that this vocabulary enables the machine learnability of antibody-antigen binding on the paratope-epitope level using generative machine learning. The vocabulary (1) is compact, less than 104 motifs; (2) distinct from non-immune protein-protein interactions; and (3) mediates specific oligo- and polyreactive interactions between paratope-epitope pairs. Our work leverages combined structure- and sequence-based learning to demonstrate that machine-learning-driven predictive paratope and epitope engineering is feasible.
Collapse
Affiliation(s)
- Rahmad Akbar
- Department of Immunology, University of Oslo, Oslo, Norway.
| | | | - Milena Pavlović
- Department of Informatics, University of Oslo, Oslo, Norway; Centre for Bioinformatics, University of Oslo, Norway; K.G. Jebsen Centre for Coeliac Disease Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Igor Snapkov
- Department of Immunology, University of Oslo, Oslo, Norway
| | | | - Cédric R Weber
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Lonneke Scheffer
- Department of Informatics, University of Oslo, Oslo, Norway; Centre for Bioinformatics, University of Oslo, Norway
| | - Enkelejda Miho
- Institute of Medical Engineering and Medical Informatics, School of Life Sciences, FHNW University of Applied Sciences and Arts Northwestern Switzerland, Muttenz, Switzerland
| | | | | | | | - Yana Safonova
- Computer Science and Engineering Department, University of California, San Diego, La Jolla, CA, USA
| | - Geir K Sandve
- Department of Informatics, University of Oslo, Oslo, Norway; Centre for Bioinformatics, University of Oslo, Norway; K.G. Jebsen Centre for Coeliac Disease Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Victor Greiff
- Department of Immunology, University of Oslo, Oslo, Norway.
| |
Collapse
|
46
|
Bonadio A, Shifman JM. Computational design and experimental optimization of protein binders with prospects for biomedical applications. Protein Eng Des Sel 2021; 34:gzab020. [PMID: 34436606 PMCID: PMC8388154 DOI: 10.1093/protein/gzab020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 07/11/2021] [Accepted: 07/11/2021] [Indexed: 11/12/2022] Open
Abstract
Protein-based binders have become increasingly more attractive candidates for drug and imaging agent development. Such binders could be evolved from a number of different scaffolds, including antibodies, natural protein effectors and unrelated small protein domains of different geometries. While both computational and experimental approaches could be utilized for protein binder engineering, in this review we focus on various computational approaches for protein binder design and demonstrate how experimental selection could be applied to subsequently optimize computationally-designed molecules. Recent studies report a number of designed protein binders with pM affinities and high specificities for their targets. These binders usually characterized with high stability, solubility, and low production cost. Such attractive molecules are bound to become more common in various biotechnological and biomedical applications in the near future.
Collapse
Affiliation(s)
- Alessandro Bonadio
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Julia M Shifman
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| |
Collapse
|
47
|
Norman RA, Ambrosetti F, Bonvin AMJJ, Colwell LJ, Kelm S, Kumar S, Krawczyk K. Computational approaches to therapeutic antibody design: established methods and emerging trends. Brief Bioinform 2020; 21:1549-1567. [PMID: 31626279 PMCID: PMC7947987 DOI: 10.1093/bib/bbz095] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/07/2019] [Accepted: 07/05/2019] [Indexed: 12/31/2022] Open
Abstract
Antibodies are proteins that recognize the molecular surfaces of potentially noxious molecules to mount an adaptive immune response or, in the case of autoimmune diseases, molecules that are part of healthy cells and tissues. Due to their binding versatility, antibodies are currently the largest class of biotherapeutics, with five monoclonal antibodies ranked in the top 10 blockbuster drugs. Computational advances in protein modelling and design can have a tangible impact on antibody-based therapeutic development. Antibody-specific computational protocols currently benefit from an increasing volume of data provided by next generation sequencing and application to related drug modalities based on traditional antibodies, such as nanobodies. Here we present a structured overview of available databases, methods and emerging trends in computational antibody analysis and contextualize them towards the engineering of candidate antibody therapeutics.
Collapse
|
48
|
Chaves B, Sartori GR, Vasconcelos DCA, Savino W, Caffarena ER, Cotta-de-Almeida V, da Silva JHM. Guidelines To Predict Binding Poses of Antibody-Integrin Complexes. ACS OMEGA 2020; 5:16379-16385. [PMID: 32685800 PMCID: PMC7364430 DOI: 10.1021/acsomega.0c00226] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/19/2020] [Indexed: 06/11/2023]
Abstract
Integrins are cell adhesion receptors that transmit bidirectional signals across the plasma membrane. They are noncovalently linked heterodimeric molecules consisting of two subunits and act as biomarkers in several pathologies. Thus, according to the increase of therapeutic antibody production, some efforts have been applied to produce anti-integrin antibodies. Here, we purposed to evaluate methods of generation and identification of the binding pose of integrin-antibody complexes, through protein-protein docking and molecular dynamics simulations, and propose a strategy to assure the confidence of the final model and avoid false-positive poses. The results show that ClusPro and GRAMM-X were the best programs to generate the native pose of integrin-antibody complexes. Furthermore, we were able to recover and to ensure that the selected pose is the native one by using a simple rule. All complexes from ClusPro in which the first model had the lowest energy, at least 5% more negative than the second one, were correctly predicted. Therefore, our methodology seems to be efficient to avoid misranking of wrong poses for integrin-antibody complexes. In cases where the rule is inconclusive, we proposed the use of heated molecular dynamics to identify the native pose characterized by RMSDi <0.5 nm. We believe that the set of methods presented here helps in the rational design of anti-integrin antibodies, giving some insights on the development of new biopharmaceuticals.
Collapse
Affiliation(s)
- Beatriz Chaves
- Computational
Modeling Group, Oswaldo Cruz Foundation, Ceara. Av Sao Jose, S/N, CEP, Eusebio, Ceará 61760-000, Brazil
| | - Geraldo R. Sartori
- Computational
Modeling Group, Oswaldo Cruz Foundation, Ceara. Av Sao Jose, S/N, CEP, Eusebio, Ceará 61760-000, Brazil
| | - Disraeli C. A. Vasconcelos
- Computational
Modeling Group, Oswaldo Cruz Foundation, Ceara. Av Sao Jose, S/N, CEP, Eusebio, Ceará 61760-000, Brazil
| | - Wilson Savino
- Laboratory
on Thymus Research, Oswaldo Cruz Institute/Oswaldo
Cruz Foundation, Rio de Janeiro 21040-360, Brazil
| | - Ernesto R. Caffarena
- Computational
Biophysics and Molecular Modeling Group, Scientific Computing Program
(PROCC), Oswaldo Cruz Foundation, Rio de Janeiro 21040-222, Brazil
| | - Vinícius Cotta-de-Almeida
- Laboratory
on Thymus Research, Oswaldo Cruz Institute/Oswaldo
Cruz Foundation, Rio de Janeiro 21040-360, Brazil
| | - João H. M. da Silva
- Computational
Modeling Group, Oswaldo Cruz Foundation, Ceara. Av Sao Jose, S/N, CEP, Eusebio, Ceará 61760-000, Brazil
| |
Collapse
|
49
|
Optimized methods for IL-17A refolding and anti-IL17A Fab production for co-crystallization with small molecules. Biotechniques 2020; 69:427-433. [DOI: 10.2144/btn-2019-0170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Refolding of human interleukin 17A (IL-17A) has been reported; however, the key refolding protocol was not robust enough to deliver consistent results and to be easily scaled up for crystallization. Here we report an optimized refolding method for IL-17A. Although co-crystal structures of IL-17A with ligands have been obtained with a high-affinity peptide and an anti-IL-17A Fab as stabilizers, neither the production yield nor the characterization of the IL-17A/Fab complex was reported. To facilitate co-crystallization of IL-17A with small-molecule compounds derived from our DNA encoded library, we also describe the method for yield enhancement of anti-IL-17A Fab production and characterize the IL-17A/Fab complex for the first time, providing an essential prerequisite for structure-based drug discovery targeting IL-17A.
Collapse
|
50
|
Tahir ul Qamar M, Shokat Z, Muneer I, Ashfaq UA, Javed H, Anwar F, Bari A, Zahid B, Saari N. Multiepitope-Based Subunit Vaccine Design and Evaluation against Respiratory Syncytial Virus Using Reverse Vaccinology Approach. Vaccines (Basel) 2020; 8:E288. [PMID: 32521680 PMCID: PMC7350008 DOI: 10.3390/vaccines8020288] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/12/2020] [Accepted: 05/15/2020] [Indexed: 02/07/2023] Open
Abstract
Respiratory syncytial virus (RSV) is primarily associated with respiratory disorders globally. Despite the availability of information, there is still no competitive vaccine available for RSV. Therefore, the present study has been designed to develop a multiepitope-based subunit vaccine (MEV) using a reverse vaccinology approach to curb RSV infections. Briefly, two highly antigenic and conserved proteins of RSV (glycoprotein and fusion protein) were selected and potential epitopes of different categories (B-cell and T-cell) were identified from them. Eminently antigenic and overlapping epitopes, which demonstrated strong associations with their respective human leukocyte antigen (HLA) alleles and depicted collective ~70% coverage of the world's populace, were shortlisted. Finally, 282 amino acids long MEV construct was established by connecting 13 major histocompatibility complex (MHC) class-I with two MHC class-II epitopes with appropriate adjuvant and linkers. Adjuvant and linkers were added to increase the immunogenic stimulation of the MEV. Developed MEV was stable, soluble, non-allergenic, non-toxic, flexible and highly antigenic. Furthermore, molecular docking and molecular dynamics (MD) simulations analyses were carried out. Results have shown a firm and robust binding affinity of MEV with human pathogenic toll-like receptor three (TLR3). The computationally mediated immune response of MEV demonstrated increased interferon-γ production, a significant abundance of immunoglobulin and activation of macrophages which are essential for immune-response against RSV. Moreover, MEV codons were optimized and in silico cloning was performed, to ensure its increased expression. These outcomes proposed that the MEV developed in this study will be a significant candidate against RSV to control and prevent RSV-related disorders if further investigated experimentally.
Collapse
Affiliation(s)
| | - Zeeshan Shokat
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad 38000, Pakistan; (Z.S.); (U.A.A.); (H.J.)
| | - Iqra Muneer
- School of Life Sciences, University of Science and Technology of China, Hefei 230052, China;
| | - Usman Ali Ashfaq
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad 38000, Pakistan; (Z.S.); (U.A.A.); (H.J.)
| | - Hamna Javed
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad 38000, Pakistan; (Z.S.); (U.A.A.); (H.J.)
| | - Farooq Anwar
- Department of Chemistry, University of Sargodha, Sargodha 40100, Pakistan;
| | - Amna Bari
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China;
| | - Barira Zahid
- Key Laboratory of Horticultural Plant Biology (Ministry of Education), Huazhong Agricultural University, Wuhan 430070, China;
| | - Nazamid Saari
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| |
Collapse
|