1
|
Chen K, Wei F, Zhang X, Jin H, Zhou R, Zuo Y, Fan K. Dynamics of an SVEIR transmission model with protection awareness and two strains. Infect Dis Model 2025; 10:207-228. [PMID: 39469221 PMCID: PMC11513685 DOI: 10.1016/j.idm.2024.10.001] [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: 07/19/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/30/2024] Open
Abstract
As of May 2024, the main strains of COVID-19 caused hundreds of millions of infection cases and millions of deaths worldwide. In this study, we consider the COVID-19 epidemics with the main strains in the Chinese mainland. We study complex interactions among hosts, non-pharmaceutical interventions, and vaccinations for the main strains by a differential equation model called SVEIR. The disease transmission model incorporates two strains and protection awareness of the susceptible population. Results of this study show that the protection awareness plays a crucial role against infection of the population, and that the vaccines are effective against the circulation of the earlier strains, but ineffective for emerging strains. By using the next generation matrix method, the basic reproduction number of the SVEIR model is firstly obtained. Our analysis by Hurwitz criterion and LaSalle's invariance principle shows that the disease free-equilibrium point is locally and globally asymptotically stable when the threshold value is below one. The existences of endemic equilibrium points are also established, and the global asymptotic stabilities are analyzed using the Lyapunov function method. Further, the SVEIR model is confirmed to satisfy the principle of competitive exclusion, of which the strain with the larger value of the basic reproduction number is dominant. Numerically, the surveillance data with the Omicron strain and the XBB strain are split by the cubic spline interpolation method. The fitting curves against the surveillance data are plotted using the least-squares method from MATLAB. The results indicate that the XBB strain dominates in this study. Moreover, a global sensitivity analysis of the key parameters is performed by using of PRCC. The numerical simulations imply that combination control strategy positively impacts on the infection scale than what separate control strategy does, and that the earlier time producing protection awareness for the public creates less infection scale, further that the increment of protection awareness also reduces the infection scale. Therefore, the policymakers of the local government are suggested to concern the changes of protection awareness of the public.
Collapse
Affiliation(s)
- Kaijing Chen
- School of Mathematics and Statistics, Fuzhou University, Fuzhou, 350116, Fujian, China
| | - Fengying Wei
- School of Mathematics and Statistics, Fuzhou University, Fuzhou, 350116, Fujian, China
- Key Laboratory of Operations Research and Control of Universities in Fujian, Fuzhou University, Fuzhou, 350116, Fujian, China
- Center for Applied Mathematics of Fujian Province, Fuzhou University, Fuzhou, 350116, Fujian, China
| | - Xinyan Zhang
- Jinzhou Center for Disease Control and Prevention, Jinzhou, 121000, Liaoning, China
| | - Hao Jin
- Jinzhou Center for Disease Control and Prevention, Jinzhou, 121000, Liaoning, China
| | - Ruiyang Zhou
- School of Mathematics and Statistics, Fuzhou University, Fuzhou, 350116, Fujian, China
| | - Yue Zuo
- Jinzhou Center for Disease Control and Prevention, Jinzhou, 121000, Liaoning, China
| | - Kai Fan
- Jinzhou Center for Disease Control and Prevention, Jinzhou, 121000, Liaoning, China
| |
Collapse
|
2
|
Singh R, Im C, Qiu Y, Mackness B, Gupta A, Joren T, Sledzieski S, Erlach L, Wendt M, Fomekong Nanfack Y, Bryson B, Berger B. Learning the language of antibody hypervariability. Proc Natl Acad Sci U S A 2025; 122:e2418918121. [PMID: 39793083 PMCID: PMC11725859 DOI: 10.1073/pnas.2418918121] [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: 09/15/2024] [Accepted: 11/19/2024] [Indexed: 01/12/2025] Open
Abstract
Protein language models (PLMs) have demonstrated impressive success in modeling proteins. However, general-purpose "foundational" PLMs have limited performance in modeling antibodies due to the latter's hypervariable regions, which do not conform to the evolutionary conservation principles that such models rely on. In this study, we propose a transfer learning framework called Antibody Mutagenesis-Augmented Processing (AbMAP), which fine-tunes foundational models for antibody-sequence inputs by supervising on antibody structure and binding specificity examples. Our learned feature representations accurately predict mutational effects on antigen binding, paratope identification, and other key antibody properties. We experimentally validate AbMAP for antibody optimization by applying it to refine a set of antibodies that bind to a SARS-CoV-2 peptide, and obtain an 82% hit-rate and up to 22-fold increase in binding affinity. AbMAP also unlocks large-scale analyses of immune repertoires, revealing that B-cell receptor repertoires of individuals, while remarkably different in sequence, converge toward similar structural and functional coverage. Importantly, AbMAP's transfer learning approach can be readily adapted to advances in foundational PLMs. We anticipate AbMAP will accelerate the efficient design and modeling of antibodies, expedite the discovery of antibody-based therapeutics, and deepen our understanding of humoral immunity.
Collapse
Affiliation(s)
- Rohit Singh
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Chiho Im
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Yu Qiu
- Sanofi R&D Large Molecule Research, Cambridge, MA02141
| | | | - Abhinav Gupta
- Sanofi R&D Large Molecule Research, Cambridge, MA02141
| | - Taylor Joren
- Sanofi R&D Data and Data Science, Artificial Intelligence and Deep Analytics, Cambridge, MA02141
| | - Samuel Sledzieski
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Lena Erlach
- Department of Biosystems Science and Engineering, ETH Zürich, 8092, Switzerland
| | - Maria Wendt
- Sanofi R&D Large Molecule Research, Cambridge, MA02141
| | | | - Bryan Bryson
- Department of Biological Engineering, Massachusetts Institute of Technology, Technology, Cambridge, MA02139
| | - Bonnie Berger
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Mathematics, Massachusetts Institute of Technology, Cambridge, MA02139
| |
Collapse
|
3
|
Liu H, Chen P, Zhai X, Huo KG, Zhou S, Han L, Fan G. PPB-Affinity: Protein-Protein Binding Affinity dataset for AI-based protein drug discovery. Sci Data 2024; 11:1316. [PMID: 39627219 PMCID: PMC11615212 DOI: 10.1038/s41597-024-03997-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/11/2024] [Indexed: 12/06/2024] Open
Abstract
Prediction of protein-protein binding (PPB) affinity plays an important role in large-molecular drug discovery. Deep learning (DL) has been adopted to predict the changes of PPB binding affinities upon mutations, but there was a scarcity of studies predicting the PPB affinity itself. The major reason is the paucity of open-source dataset with PPB affinity data. To address this gap, the current study introduced a large comprehensive PPB affinity (PPB-Affinity) dataset. The PPB-Affinity dataset contains key information such as crystal structures of protein-protein complexes (with or without protein mutation patterns), PPB affinity, receptor protein chain, ligand protein chain, etc. To the best of our knowledge, this is the largest publicly available PPB affinity dataset, and we believe it will significantly advance drug discovery by streamlining the screening of potential large-molecule drugs. We also developed a deep-learning benchmark model with this dataset to predict the PPB affinity, providing a foundational comparison for the research community.
Collapse
Affiliation(s)
- Huaqing Liu
- Artificial Intelligence Innovation Center, Research Institute of Tsinghua, Pearl River Delta, Guangzhou, 510700, China
| | - Peiyi Chen
- Artificial Intelligence Innovation Center, Research Institute of Tsinghua, Pearl River Delta, Guangzhou, 510700, China
| | - Xiaochen Zhai
- Cyagen Biosciences (Suzhou) Inc., Guangzhou, 215000, China
| | - Ku-Geng Huo
- Cyagen Biosciences (Guangzhou) Inc., Guangzhou, 510700, China
| | - Shuxian Zhou
- Artificial Intelligence Innovation Center, Research Institute of Tsinghua, Pearl River Delta, Guangzhou, 510700, China
| | - Lanqing Han
- Artificial Intelligence Innovation Center, Research Institute of Tsinghua, Pearl River Delta, Guangzhou, 510700, China.
- Cyagen Biomodels (Guangzhou) Co., Ltd, Guangzhou, 510700, China.
| | - Guoxin Fan
- Department of Pain Medicine, Shenzhen Nanshan People's Hospital, Shenzhen University Medical School, Shenzhen, 518056, China.
| |
Collapse
|
4
|
Matson RP, Comba IY, Silvert E, Niesen MJM, Murugadoss K, Patwardhan D, Suratekar R, Goel EG, Poelaert BJ, Wan KK, Brimacombe KR, Venkatakrishnan AJ, Soundararajan V. A deep learning approach predicting the activity of COVID-19 therapeutics and vaccines against emerging variants. NPJ Syst Biol Appl 2024; 10:138. [PMID: 39604453 PMCID: PMC11603192 DOI: 10.1038/s41540-024-00471-0] [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: 11/07/2023] [Accepted: 11/09/2024] [Indexed: 11/29/2024] Open
Abstract
Understanding which viral variants evade neutralization is crucial for improving antibody-based treatments, especially with rapidly evolving viruses like SARS-CoV-2. Yet, conventional assays are labor intensive and cannot capture the full spectrum of variants. We present a deep learning approach to predict changes in neutralizing antibody activity of COVID-19 therapeutics and vaccine-elicited sera/plasma against emerging viral variants. Our approach leverages data of 67,885 unique SARS-CoV-2 Spike sequences and 7,069 in vitro assays. The resulting model accurately predicted fold changes in neutralizing activity (R2 = 0.77) for a test set (N = 980) of data collected up to eight months after the training data. Next, the model was used to predict changes in activity of current therapeutic and vaccine-induced antibodies against emerging SARS-CoV-2 lineages. Consistent with other work, we found significantly reduced activity against newer XBB descendants, notably EG.5, FL.1.5.1, and XBB.1.16; primarily attributed to the F456L spike mutation.
Collapse
Affiliation(s)
| | - Isin Y Comba
- nference, Cambridge, MA, 02139, USA
- Division of Public Health, Infectious Diseases and Occupational Medicine, Mayo Clinic Rochester, Rochester, NY, 55905, USA
| | | | | | | | | | | | | | - Brittany J Poelaert
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA
| | - Kanny K Wan
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA
| | - Kyle R Brimacombe
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA
| | | | - Venky Soundararajan
- nference, Cambridge, MA, 02139, USA.
- nference Labs, Bengaluru, Karnataka, 560017, India.
| |
Collapse
|
5
|
Kim J, Kim S, Park S, Kim D, Kim M, Baek K, Kang BM, Shin HE, Lee MH, Lee Y, Kwon HJ. Production of a monoclonal antibody targeting the SARS-CoV-2 Omicron spike protein and analysis of SARS-CoV-2 Omicron mutations related to monoclonal antibody resistance. Microbes Infect 2024:105461. [PMID: 39580070 DOI: 10.1016/j.micinf.2024.105461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 11/25/2024]
Abstract
SARS-CoV-2 mutations have resulted in the emergence of multiple concerning variants, with Omicron being the dominant strain presently. Therefore, we developed a monoclonal antibody (mAb) against the spike (S) protein of SARS-CoV-2 Omicron for therapeutic applications. We established the 1E3H12 mAb, recognizing the receptor binding domain (RBD) of the Omicron S protein, and found that the 1E3H12 mAb can efficiently recognize the Omicron S protein with weak affinity to the Alpha, Beta, and Mu variants, but not to the parental strain and Delta variant. Based on in vitro assays, the mAb demonstrated neutralizing activity against Omicron BA.1, BA.4/5, BQ.1.1, and XBB. A humanized antibody was further produced and proved to have neutralizing activity. To verify the potential limitations of the 1E3H12 mAb due to viral escape of SARS-CoV-2 Omicron variants, we analyzed the emergence of variants by whole genome deep sequencing after serial passage in cell culture. The results showed a few unique S protein mutations in the genome associated with resistance to the mAb. These findings suggest that this antibody not only contributes to the therapeutic arsenal against COVID-19 but also addresses the ongoing challenge of antibody resistance among the evolving subvariants of SARS-CoV-2 Omicron.
Collapse
Affiliation(s)
- Jinsoo Kim
- Institute of Medical Science, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Suyeon Kim
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Sangkyu Park
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, Republic of Korea
| | - Dongbum Kim
- Institute of Medical Science, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Minyoung Kim
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Kyeongbin Baek
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Bo Min Kang
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Ha-Eun Shin
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, Republic of Korea
| | - Myeong-Heon Lee
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, Republic of Korea
| | - Younghee Lee
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, Republic of Korea.
| | - Hyung-Joo Kwon
- Institute of Medical Science, College of Medicine, Hallym University, Chuncheon, Republic of Korea; Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Republic of Korea.
| |
Collapse
|
6
|
Cong B, Dong X, Yang Z, Yu P, Chai Y, Liu J, Zhang M, Zang Y, Kang J, Feng Y, Liu Y, Feng W, Wang D, Deng W, Li F, Song Z, Wang Z, Chen X, Qin H, Yu Q, Li Z, Liu S, Xu X, Zhong N, Ren X, Qin C, Liu L, Wang J, Cao X. Single-cell spatiotemporal analysis reveals alveolar dendritic cell-T cell immunity hubs defending against pulmonary infection. Cell Discov 2024; 10:103. [PMID: 39414763 PMCID: PMC11484931 DOI: 10.1038/s41421-024-00733-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/08/2024] [Indexed: 10/18/2024] Open
Abstract
How immune cells are spatiotemporally coordinated in the lung to effectively monitor, respond to, and resolve infection and inflammation in primed form needs to be fully illustrated. Here we apply immunocartography, a high-resolution technique that integrates spatial and single-cell RNA sequencing (scRNA-seq) through deconvolution and co-localization analyses, to the SARS-CoV-2-infected Syrian hamster model. We generate a comprehensive transcriptome map of the whole process of pulmonary infection from physiological condition, infection initiation, severe pneumonia to natural recovery at organ scale and single-cell resolution, with 142,965 cells and 45 lung lobes from 25 hamsters at 5 time points. Integrative analysis identifies that alveolar dendritic cell-T cell immunity hubs, where Ccr7+Ido1+ dendritic cells, Cd160+Cd8+ T cells, and Tnfrsf4+Cd4+ T cells physiologically co-localize, rapidly expand during SARS-CoV-2 infection, eliminate SARS-CoV-2 with the aid of Slamf9+ macrophages, and then restore to physiological levels after viral clearance. We verify the presence of these cell subpopulations in the immunity hubs in normal and SARS-CoV-2-infected hACE2 mouse models, as well as in publicly available human scRNA-seq datasets, demonstrating the potential broad relevance of our findings in lung immunity.
Collapse
Affiliation(s)
- Boyi Cong
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuan Dong
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Zongheng Yang
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Pin Yu
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Yangyang Chai
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiaqi Liu
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Meihan Zhang
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China
| | | | | | - Yu Feng
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Yi Liu
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | | | - Dehe Wang
- Changping Laboratory, Beijing, China
| | - Wei Deng
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Fengdi Li
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhiqi Song
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Ziqiao Wang
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaosu Chen
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China
| | - Hua Qin
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China
| | - Qinyi Yu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhiqing Li
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Navy Medical University, Shanghai, China
- Guangzhou Laboratory, Guangzhou, Guangdong, China
| | - Shuxun Liu
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Navy Medical University, Shanghai, China
- Guangzhou Laboratory, Guangzhou, Guangdong, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | | | | | - Chuan Qin
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China.
| | - Longqi Liu
- BGI-Shenzhen, Shenzhen, Guangdong, China.
| | - Jian Wang
- BGI-Shenzhen, Shenzhen, Guangdong, China.
| | - Xuetao Cao
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China.
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
7
|
Bendix AF, Trentin AB, Vasconcelos MW, Pilonetto JC, Kuhn BC, Leite DCDA, De Barros FRO, Cardoso JMK, Gabiatti NC, Wendt SN, Ghisi NDC. From chaos to clarity: The scientometric breakthrough in COVID-19 research. Diagn Microbiol Infect Dis 2024; 110:116438. [PMID: 39047387 DOI: 10.1016/j.diagmicrobio.2024.116438] [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: 04/29/2024] [Revised: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND The COVID-19 pandemic paralyzed the world for over three years, generating unprecedented social changes in recent human history. AIMS We aimed to scientometrically summarize a global and temporal overview of publications on COVID-19 in the two worst years of the pandemic and its progression in early 2022, after the start of vaccination. METHODS Using the Web of Science database, this review covered the period from late 2019 to March 2022 and included all publications identified using the following terms: "SARS-CoV-2", "COVID-19", "Coronavirus Disease 19", and "2019-nCoV". We retrieved 268,904 publications, with evident global spreading, demonstrating that the pandemic triggered worldwide scientific research efforts. RESULTS Within the dataset, 195 countries have published about Covid-19. In initial publications, a solid trend in genotyping, sequencing, and detection of the virus was evident; however, in the development of the pandemic, new knowledge and research focus gained relevance, with continental solid trends, revealed by the keywords sustainability (eastern Europe); material sciences (Asia); public and mental health (Africa); information sciences (western Europe); education (Latin America). It identified high-impact research, mainly on diagnosis and vaccines, but also equally essential topics for returning life to the new normal, such as mental health, education, and remote work. The world experienced a highly transmissible infection that proved how fragile we are regarding organization and society. CONCLUSIONS It is necessary to learn from such an event and establish a protocol of actions and measures to be taken and avoided in a health emergency, aiming to act differently from the chaos experienced during the pandemic. Following the One Health approach, humanity must be aware of the need for more sustainable attitudes, given the inseparability of human beings from the environment.
Collapse
Affiliation(s)
- Andre Felipe Bendix
- Programa de Pós-Graduação em Biotecnologia - PPGBIOTEC, Universidade Tecnológica Federal do Paraná, Dois Vizinhos, Brasil; Dois Vizinhos/ Laboratório Multiusuário de Análises Biológicas e Biologia Molecular (BioMol) - UTFPR, Grupo de Pesquisa em Biologia Molecular - UTFPR, Brasil
| | - Alex Batista Trentin
- Programa de Pós-Graduação em Biotecnologia - PPGBIOTEC, Universidade Tecnológica Federal do Paraná, Dois Vizinhos, Brasil; Dois Vizinhos/ Laboratório Multiusuário de Análises Biológicas e Biologia Molecular (BioMol) - UTFPR, Grupo de Pesquisa em Biologia Molecular - UTFPR, Brasil
| | - Marina Wust Vasconcelos
- Dois Vizinhos/ Laboratório Multiusuário de Análises Biológicas e Biologia Molecular (BioMol) - UTFPR, Grupo de Pesquisa em Biologia Molecular - UTFPR, Brasil; Programa de Pós-Graduação em Genética (PPGGEN), Universidade Federal do Paraná, Curitiba, Brasil
| | - Jessica Cousseau Pilonetto
- Programa de Pós-Graduação em Biotecnologia - PPGBIOTEC, Universidade Tecnológica Federal do Paraná, Dois Vizinhos, Brasil; Dois Vizinhos/ Laboratório Multiusuário de Análises Biológicas e Biologia Molecular (BioMol) - UTFPR, Grupo de Pesquisa em Biologia Molecular - UTFPR, Brasil
| | - Betty Cristiane Kuhn
- Coordenação do Curso de Engenharia de Bioprocessos e Biotecnologia, Universidade Tecnológica Federal do Paraná, Brasil; Dois Vizinhos/ Laboratório Multiusuário de Análises Biológicas e Biologia Molecular (BioMol) - UTFPR, Grupo de Pesquisa em Biologia Molecular - UTFPR, Brasil
| | - Deborah Catharine De Assis Leite
- Programa de Pós-Graduação em Biotecnologia - PPGBIOTEC, Universidade Tecnológica Federal do Paraná, Dois Vizinhos, Brasil; Dois Vizinhos/ Laboratório Multiusuário de Análises Biológicas e Biologia Molecular (BioMol) - UTFPR, Grupo de Pesquisa em Biologia Molecular - UTFPR, Brasil; Programa de Pós-Graduação em Tecnologias Computacionais para o Agronegócio-PPGTCA, Universidade Tecnológica Federal do Paraná, Medianeira, Brasil
| | - Flavia Regina Oliveira De Barros
- Coordenação do Curso de Engenharia de Bioprocessos e Biotecnologia, Universidade Tecnológica Federal do Paraná, Brasil; Programa de Pós-Graduação em Zootecnia (PPZ), Universidade Tecnológica Federal do Paraná, Dois Vizinhos, Brasil; Dois Vizinhos/ Laboratório Multiusuário de Análises Biológicas e Biologia Molecular (BioMol) - UTFPR, Grupo de Pesquisa em Biologia Molecular - UTFPR, Brasil
| | - Juliana Morini Küpper Cardoso
- Dois Vizinhos/ Laboratório Multiusuário de Análises Biológicas e Biologia Molecular (BioMol) - UTFPR, Grupo de Pesquisa em Biologia Molecular - UTFPR, Brasil
| | - Naiana Cristine Gabiatti
- Programa de Pós-Graduação em Biotecnologia - PPGBIOTEC, Universidade Tecnológica Federal do Paraná, Dois Vizinhos, Brasil; Dois Vizinhos/ Laboratório Multiusuário de Análises Biológicas e Biologia Molecular (BioMol) - UTFPR, Grupo de Pesquisa em Biologia Molecular - UTFPR, Brasil
| | - Simone Neumann Wendt
- Coordenação do Curso de Engenharia Florestal, Universidade Tecnológica Federal do Paraná, Brasil; Dois Vizinhos/ Laboratório Multiusuário de Análises Biológicas e Biologia Molecular (BioMol) - UTFPR, Grupo de Pesquisa em Biologia Molecular - UTFPR, Brasil
| | - Nédia de Castilhos Ghisi
- Programa de Pós-Graduação em Biotecnologia - PPGBIOTEC, Universidade Tecnológica Federal do Paraná, Dois Vizinhos, Brasil; Dois Vizinhos/ Laboratório Multiusuário de Análises Biológicas e Biologia Molecular (BioMol) - UTFPR, Grupo de Pesquisa em Biologia Molecular - UTFPR, Brasil.
| |
Collapse
|
8
|
Cai H, Zhang Z, Wang M, Zhong B, Li Q, Zhong Y, Wu Y, Ying T, Tang J. Pretrainable geometric graph neural network for antibody affinity maturation. Nat Commun 2024; 15:7785. [PMID: 39242604 PMCID: PMC11379722 DOI: 10.1038/s41467-024-51563-8] [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: 09/13/2023] [Accepted: 08/13/2024] [Indexed: 09/09/2024] Open
Abstract
Increasing the binding affinity of an antibody to its target antigen is a crucial task in antibody therapeutics development. This paper presents a pretrainable geometric graph neural network, GearBind, and explores its potential in in silico affinity maturation. Leveraging multi-relational graph construction, multi-level geometric message passing and contrastive pretraining on mass-scale, unlabeled protein structural data, GearBind outperforms previous state-of-the-art approaches on SKEMPI and an independent test set. A powerful ensemble model based on GearBind is then derived and used to successfully enhance the binding of two antibodies with distinct formats and target antigens. ELISA EC50 values of the designed antibody mutants are decreased by up to 17 fold, and KD values by up to 6.1 fold. These promising results underscore the utility of geometric deep learning and effective pretraining in macromolecule interaction modeling tasks.
Collapse
Affiliation(s)
- Huiyu Cai
- BioGeometry, Beijing, China
- Mila-Québec AI Institute, Montréal, QC, Canada
- Department of Computer Science and Operations Research, Université de Montréal, Montréal, QC, Canada
| | - Zuobai Zhang
- Mila-Québec AI Institute, Montréal, QC, Canada
- Department of Computer Science and Operations Research, Université de Montréal, Montréal, QC, Canada
| | - Mingkai Wang
- Shanghai Engineering Research Center for Synthetic Immunology, Fudan University, Shanghai, China
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Bozitao Zhong
- Mila-Québec AI Institute, Montréal, QC, Canada
- Department of Computer Science and Operations Research, Université de Montréal, Montréal, QC, Canada
| | - Quanxiao Li
- Shanghai Engineering Research Center for Synthetic Immunology, Fudan University, Shanghai, China
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yuxuan Zhong
- Shanghai Engineering Research Center for Synthetic Immunology, Fudan University, Shanghai, China
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yanling Wu
- Shanghai Engineering Research Center for Synthetic Immunology, Fudan University, Shanghai, China.
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Tianlei Ying
- Shanghai Engineering Research Center for Synthetic Immunology, Fudan University, Shanghai, China.
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Jian Tang
- BioGeometry, Beijing, China.
- Mila-Québec AI Institute, Montréal, QC, Canada.
- Department of Decision Sciences, HEC Montréal, Montréal, QC, Canada.
| |
Collapse
|
9
|
Almubarak HF, Tan W, Hoffmann AD, Sun Y, Wei J, El-Shennawy L, Squires JR, Dashzeveg NK, Simonton B, Jia Y, Iyer R, Xu Y, Nicolaescu V, Elli D, Randall GC, Schipma MJ, Swaminathan S, Ison MG, Liu H, Fang D, Shen Y. Novel antibody language model accelerates IgG screening and design for broad-spectrum antiviral therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.01.582176. [PMID: 38496411 PMCID: PMC10942297 DOI: 10.1101/2024.03.01.582176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Therapeutic antibodies have become one of the most influential therapeutics in modern medicine to fight against infectious pathogens, cancer, and many other diseases. However, experimental screening for highly efficacious targeting antibodies is labor-intensive and of high cost, which is exacerbated by evolving antigen targets under selective pressure such as fast-mutating viral variants. As a proof-of-concept, we developed a machine learning-assisted antibody generation pipeline AbGen that greatly accelerates the screening and re-design of immunoglobulins G (IgGs) against a broad spectrum of SARS-CoV-2 coronavirus variant strains. Our AbGen centers around a novel antibody language model (AbLM) that is pretrained on 12 million generic protein domain sequences and fine-tuned on 4,000+ paired VH-VL sequences, with IgG-specific CDR-masking and VH-VL cross-attention. AbLM provides a latent space of IgG sequence embeddings for AbGen, including (a) landscapes of IgGs' activities in neutralizing the wild-type virus are analyzed through structure prediction for IgG and IgG-antigen (viral protein spike's receptor binding domain, RBD) interactions; and (b) landscapes of IgGs' susceptibility in neutralizing variant viruses are predicted through Gaussian process regression, despite that as few as 14 clinical antibodies' responses to variants of concern are available. The AbGen pipeline was applied to over 1300 IgG sequences we collected from RBD-binding B cells of convalescent patients. With experimental validations, AbGen efficiently prioritized IgG candidates against a broad spectrum of viral variants (wildtype, Delta, and Omicron), preventing the infection of host cells in vitro and hACE2 transgenic mice in vivo. Compared to other existing protein language models that require 10-100 times more model parameters, AbLM improved the precision from around 50% to 75% to predict IgGs with low variant susceptibility. Furthermore, AbGen enables structure-based computational protein redesign for selected IgG clones with single amino acid substitutions at the RBD-binding interface that doubled the IgG blockade efficacy for one of the severe, therapy-resistant strains - Delta (B.1.617). Our work expedites applications of artificial intelligence in antibody screen and re-design combining data-driven protein language models and Kriging for antibody sequence analysis and activity prediction, in synergy with physics-driven protein docking and design for antibody-antigen interface analyses and functional optimization.
Collapse
Affiliation(s)
- Hannah Faisal Almubarak
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611
- Driskill Graduate Program, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611
| | - Wuwei Tan
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843
| | - Andrew D. Hoffmann
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611
| | - Yuanfei Sun
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843
| | - Juncheng Wei
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611
| | - Lamiaa El-Shennawy
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611
| | - Joshua R. Squires
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611
| | - Nurmaa K. Dashzeveg
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611
| | - Brooke Simonton
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611
| | - Yuzhi Jia
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611
| | - Radhika Iyer
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611
| | - Yanan Xu
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611
| | - Vlad Nicolaescu
- Howard T. Ricketts Laboratory and Department of Microbiology, the University of Chicago, Chicago, IL 60637
| | - Derek Elli
- Howard T. Ricketts Laboratory and Department of Microbiology, the University of Chicago, Chicago, IL 60637
| | - Glenn C. Randall
- Howard T. Ricketts Laboratory and Department of Microbiology, the University of Chicago, Chicago, IL 60637
| | - Matthew J. Schipma
- NUseq Core Facility, Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611
| | - Suchitra Swaminathan
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611
- Division of Rheumatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611
| | | | - Huiping Liu
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611
| | - Yang Shen
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843
| |
Collapse
|
10
|
He H, He B, Guan L, Zhao Y, Jiang F, Chen G, Zhu Q, Chen CYC, Li T, Yao J. De novo generation of SARS-CoV-2 antibody CDRH3 with a pre-trained generative large language model. Nat Commun 2024; 15:6867. [PMID: 39127753 PMCID: PMC11316817 DOI: 10.1038/s41467-024-50903-y] [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: 11/14/2023] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Artificial Intelligence (AI) techniques have made great advances in assisting antibody design. However, antibody design still heavily relies on isolating antigen-specific antibodies from serum, which is a resource-intensive and time-consuming process. To address this issue, we propose a Pre-trained Antibody generative large Language Model (PALM-H3) for the de novo generation of artificial antibodies heavy chain complementarity-determining region 3 (CDRH3) with desired antigen-binding specificity, reducing the reliance on natural antibodies. We also build a high-precision model antigen-antibody binder (A2binder) that pairs antigen epitope sequences with antibody sequences to predict binding specificity and affinity. PALM-H3-generated antibodies exhibit binding ability to SARS-CoV-2 antigens, including the emerging XBB variant, as confirmed through in-silico analysis and in-vitro assays. The in-vitro assays validate that PALM-H3-generated antibodies achieve high binding affinity and potent neutralization capability against spike proteins of SARS-CoV-2 wild-type, Alpha, Delta, and the emerging XBB variant. Meanwhile, A2binder demonstrates exceptional predictive performance on binding specificity for various epitopes and variants. Furthermore, by incorporating the attention mechanism inherent in the Roformer architecture into the PALM-H3 model, we improve its interpretability, providing crucial insights into the fundamental principles of antibody design.
Collapse
Affiliation(s)
- Haohuai He
- AI Lab, Tencent, Shenzhen, 518052, China
- Artificial Intelligence Medical Research Center, School of Intelligent Systems Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Bing He
- AI Lab, Tencent, Shenzhen, 518052, China.
| | - Lei Guan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Xi'an, China
| | - Yu Zhao
- AI Lab, Tencent, Shenzhen, 518052, China
| | - Feng Jiang
- AI Lab, Tencent, Shenzhen, 518052, China
| | - Guanxing Chen
- Artificial Intelligence Medical Research Center, School of Intelligent Systems Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Qingge Zhu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Xi'an, China
| | - Calvin Yu-Chian Chen
- AI for Science (AI4S)-Preferred Program, School of Electronic and Computer Engineering, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
- Department of Medical Research, China Medical University Hospital, Taichung, 40447, Taiwan.
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, 41354, Taiwan.
- Guangdong L-Med Biotechnology Co. Ltd, Meizhou, 514699, Guangdong, China.
| | - Ting Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Xi'an, China.
| | | |
Collapse
|
11
|
Barletta GP, Tandiana R, Soler M, Fortuna S, Rocchia W. Locuaz: an in silico platform for protein binders optimization. Bioinformatics 2024; 40:btae492. [PMID: 39107888 PMCID: PMC11324344 DOI: 10.1093/bioinformatics/btae492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 07/28/2024] [Accepted: 08/03/2024] [Indexed: 08/16/2024] Open
Abstract
MOTIVATION Engineering high-affinity binders targeting specific antigenic determinants remains a challenging and often daunting task, requiring extensive experimental screening. Computational methods have the potential to accelerate this process, reducing costs and time, but only if they demonstrate broad applicability and efficiency in exploring mutations, evaluating affinity, and pruning unproductive mutation paths. RESULTS In response to these challenges, we introduce a new computational platform for optimizing protein binders towards their targets. The platform is organized as a series of modules, performing mutation selection and application, molecular dynamics simulations to sample conformations around interaction poses, and mutation prioritization using suitable scoring functions. Notably, the platform supports parallel exploration of different mutation streams, enabling in silico high-throughput screening on High Performance Computing (HPC) systems. Furthermore, the platform is highly customizable, allowing users to implement their own protocols. AVAILABILITY AND IMPLEMENTATION The source code is available at https://github.com/pgbarletta/locuaz and documentation is at https://locuaz.readthedocs.io/. The data underlying this article are available at https://github.com/pgbarletta/suppl_info_locuaz.
Collapse
Affiliation(s)
- German P Barletta
- CONCEPT, Istituto Italiano di Tecnologia, Via Enrico Melen, 83 Genova Liguria 16152, Italy
- The Abdus Salam International Centre for Theoretical Physics (ICTP), Str. Costiera, 11, Trieste, Friuli-Venezia Giulia, 34151, Italy
| | - Rika Tandiana
- CONCEPT, Istituto Italiano di Tecnologia, Via Enrico Melen, 83 Genova Liguria 16152, Italy
| | - Miguel Soler
- CONCEPT, Istituto Italiano di Tecnologia, Via Enrico Melen, 83 Genova Liguria 16152, Italy
- Dipartimento di Scienze Matematiche, Informatiche e Fisiche (DMIF), University of Udine, Via delle Scienze, 206, Udine, Friuli-Venezia Giulia, 33100, Italy
| | - Sara Fortuna
- CONCEPT, Istituto Italiano di Tecnologia, Via Enrico Melen, 83 Genova Liguria 16152, Italy
- Cresset, New Cambridge House, Litlington, Royston, SG8-0SS, United Kingdom
| | - Walter Rocchia
- CONCEPT, Istituto Italiano di Tecnologia, Via Enrico Melen, 83 Genova Liguria 16152, Italy
| |
Collapse
|
12
|
Natsrita P, Charoenkwan P, Shoombuatong W, Mahalapbutr P, Faksri K, Chareonsudjai S, Rungrotmongkol T, Pipattanaboon C. Machine-learning-assisted high-throughput identification of potent and stable neutralizing antibodies against all four dengue virus serotypes. Sci Rep 2024; 14:17165. [PMID: 39060292 PMCID: PMC11282219 DOI: 10.1038/s41598-024-67487-8] [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: 12/15/2023] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Several computational methods have been developed to identify neutralizing antibodies (NAbs) covering four dengue virus serotypes (DENV-1 to DENV-4); however, limitations of the dataset and the resulting performance remain. Here, we developed a new computational framework to predict potent and stable NAbs against DENV-1 to DENV-4 using only antibody (CDR-H3) and epitope sequences as input. Specifically, our proposed computational framework employed sequence-based ML and molecular dynamic simulation (MD) methods to achieve more accurate identification. First, we built a novel dataset (n = 1108) by compiling the interactions of CDR-H3 and epitope sequences with the half maximum inhibitory concentration (IC50) values, which represent neutralizing activities. Second, we achieved an accurately predictive ML model that showed high AUC values of 0.879 and 0.885 by tenfold cross-validation and independent tests, respectively. Finally, our computational framework could be applied to filter approximately 2.5 million unseen antibodies into two final candidates that showed strong and stable binding to all four serotypes. In addition, the most potent and stable candidate (1B3B9_V21) was evaluated for its development potential as a therapeutic agent by molecular docking and MD simulations. This study provides an antibody computational approach to facilitate the high-throughput identification of NAbs and accelerate the development of therapeutic antibodies.
Collapse
Affiliation(s)
- Piyatida Natsrita
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Phasit Charoenkwan
- Modern Management and Information Technology, College of Arts, Media and Technology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Watshara Shoombuatong
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Panupong Mahalapbutr
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Kiatichai Faksri
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Research and Diagnostic Center for Emerging Infectious Diseases, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Sorujsiri Chareonsudjai
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Thanyada Rungrotmongkol
- Center of Excellent in Biocatalyst and Sustainable Biotechnology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Chonlatip Pipattanaboon
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.
| |
Collapse
|
13
|
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
|
14
|
Yi Y, Wan X, Zhao K, Ou-Yang L, Zhao P. Equivariant Line Graph Neural Network for Protein-Ligand Binding Affinity Prediction. IEEE J Biomed Health Inform 2024; 28:4336-4347. [PMID: 38551822 DOI: 10.1109/jbhi.2024.3383245] [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: 07/03/2024]
Abstract
Binding affinity prediction of three-dimensional (3D) protein-ligand complexes is critical for drug repositioning and virtual drug screening. Existing approaches usually transform a 3D protein-ligand complex to a two-dimensional (2D) graph, and then use graph neural networks (GNNs) to predict its binding affinity. However, the node and edge features of the 2D graph are extracted based on invariant local coordinate systems of the 3D complex. As a result, these approaches can not fully learn the global information of the complex, such as the physical symmetry and the topological information of bonds. To address these issues, we propose a novel Equivariant Line Graph Network (ELGN) for binding affinity prediction of 3D protein-ligand complexes. The proposed ELGN firstly adds a super node to the 3D complex, and then builds a line graph based on the 3D complex. After that, ELGN uses a new E(3)-equivariant network layer to pass the messages between nodes and edges based on the global coordinate system of the 3D complex. Experimental results on two real datasets demonstrate the effectiveness of ELGN over several state-of-the-art baselines.
Collapse
|
15
|
Yu G, Zhao Q, Bi X, Wang J. DDAffinity: predicting the changes in binding affinity of multiple point mutations using protein 3D structure. Bioinformatics 2024; 40:i418-i427. [PMID: 38940145 PMCID: PMC11211828 DOI: 10.1093/bioinformatics/btae232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2024] Open
Abstract
MOTIVATION Mutations are the crucial driving force for biological evolution as they can disrupt protein stability and protein-protein interactions which have notable impacts on protein structure, function, and expression. However, existing computational methods for protein mutation effects prediction are generally limited to single point mutations with global dependencies, and do not systematically take into account the local and global synergistic epistasis inherent in multiple point mutations. RESULTS To this end, we propose a novel spatial and sequential message passing neural network, named DDAffinity, to predict the changes in binding affinity caused by multiple point mutations based on protein 3D structures. Specifically, instead of being on the whole protein, we perform message passing on the k-nearest neighbor residue graphs to extract pocket features of the protein 3D structures. Furthermore, to learn global topological features, a two-step additive Gaussian noising strategy during training is applied to blur out local details of protein geometry. We evaluate DDAffinity on benchmark datasets and external validation datasets. Overall, the predictive performance of DDAffinity is significantly improved compared with state-of-the-art baselines on multiple point mutations, including end-to-end and pre-training based methods. The ablation studies indicate the reasonable design of all components of DDAffinity. In addition, applications in nonredundant blind testing, predicting mutation effects of SARS-CoV-2 RBD variants, and optimizing human antibody against SARS-CoV-2 illustrate the effectiveness of DDAffinity. AVAILABILITY AND IMPLEMENTATION DDAffinity is available at https://github.com/ak422/DDAffinity.
Collapse
Affiliation(s)
- Guanglei Yu
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
- Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha 410083, China
- Medical Engineering and Technology College, Xinjiang Medical University, Urumqi 830017, China
| | - Qichang Zhao
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
- Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha 410083, China
| | - Xuehua Bi
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
- Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha 410083, China
- Medical Engineering and Technology College, Xinjiang Medical University, Urumqi 830017, China
| | - Jianxin Wang
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
- Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha 410083, China
| |
Collapse
|
16
|
Chen H, Fan X, Zhu S, Pei Y, Zhang X, Zhang X, Liu L, Qian F, Tian B. Accurate prediction of CDR-H3 loop structures of antibodies with deep learning. eLife 2024; 12:RP91512. [PMID: 38921957 PMCID: PMC11208048 DOI: 10.7554/elife.91512] [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] [Indexed: 06/27/2024] Open
Abstract
Accurate prediction of the structurally diverse complementarity determining region heavy chain 3 (CDR-H3) loop structure remains a primary and long-standing challenge for antibody modeling. Here, we present the H3-OPT toolkit for predicting the 3D structures of monoclonal antibodies and nanobodies. H3-OPT combines the strengths of AlphaFold2 with a pre-trained protein language model and provides a 2.24 Å average RMSDCα between predicted and experimentally determined CDR-H3 loops, thus outperforming other current computational methods in our non-redundant high-quality dataset. The model was validated by experimentally solving three structures of anti-VEGF nanobodies predicted by H3-OPT. We examined the potential applications of H3-OPT through analyzing antibody surface properties and antibody-antigen interactions. This structural prediction tool can be used to optimize antibody-antigen binding and engineer therapeutic antibodies with biophysical properties for specialized drug administration route.
Collapse
Affiliation(s)
- Hedi Chen
- MOE Key Laboratory of Bioinformatics, State Key Laboratory of Molecular Oncology, School of Pharmaceutical Sciences, Tsinghua UniversityBeijingChina
| | - Xiaoyu Fan
- MOE Key Laboratory of Bioinformatics, State Key Laboratory of Molecular Oncology, School of Pharmaceutical Sciences, Tsinghua UniversityBeijingChina
| | - Shuqian Zhu
- MOE Key Laboratory of Bioinformatics, State Key Laboratory of Molecular Oncology, School of Pharmaceutical Sciences, Tsinghua UniversityBeijingChina
| | - Yuchan Pei
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua UniversityBeijingChina
| | - Xiaochun Zhang
- MOE Key Laboratory of Bioinformatics, State Key Laboratory of Molecular Oncology, School of Pharmaceutical Sciences, Tsinghua UniversityBeijingChina
| | - Xiaonan Zhang
- Department of Natural Language Processing, Baidu International Technology (Shenzhen) Co LtdShenzhenChina
| | - Lihang Liu
- Department of Natural Language Processing, Baidu International Technology (Shenzhen) Co LtdShenzhenChina
| | - Feng Qian
- MOE Key Laboratory of Bioinformatics, State Key Laboratory of Molecular Oncology, School of Pharmaceutical Sciences, Tsinghua UniversityBeijingChina
| | - Boxue Tian
- MOE Key Laboratory of Bioinformatics, State Key Laboratory of Molecular Oncology, School of Pharmaceutical Sciences, Tsinghua UniversityBeijingChina
| |
Collapse
|
17
|
Joubbi S, Micheli A, Milazzo P, Maccari G, Ciano G, Cardamone D, Medini D. Antibody design using deep learning: from sequence and structure design to affinity maturation. Brief Bioinform 2024; 25:bbae307. [PMID: 38960409 PMCID: PMC11221890 DOI: 10.1093/bib/bbae307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 05/20/2024] [Accepted: 06/12/2024] [Indexed: 07/05/2024] Open
Abstract
Deep learning has achieved impressive results in various fields such as computer vision and natural language processing, making it a powerful tool in biology. Its applications now encompass cellular image classification, genomic studies and drug discovery. While drug development traditionally focused deep learning applications on small molecules, recent innovations have incorporated it in the discovery and development of biological molecules, particularly antibodies. Researchers have devised novel techniques to streamline antibody development, combining in vitro and in silico methods. In particular, computational power expedites lead candidate generation, scaling and potential antibody development against complex antigens. This survey highlights significant advancements in protein design and optimization, specifically focusing on antibodies. This includes various aspects such as design, folding, antibody-antigen interactions docking and affinity maturation.
Collapse
Affiliation(s)
- Sara Joubbi
- Department of Computer Science, University of Pisa, Largo B. Pontecorvo, 3, 56127, Pisa, Italy
- Data Science for Health (DaScH) Lab, Fondazione Toscana Life Sciences, Via Fiorentina, 1, 53100, Siena, Italy
| | - Alessio Micheli
- Department of Computer Science, University of Pisa, Largo B. Pontecorvo, 3, 56127, Pisa, Italy
| | - Paolo Milazzo
- Department of Computer Science, University of Pisa, Largo B. Pontecorvo, 3, 56127, Pisa, Italy
| | - Giuseppe Maccari
- Data Science for Health (DaScH) Lab, Fondazione Toscana Life Sciences, Via Fiorentina, 1, 53100, Siena, Italy
| | - Giorgio Ciano
- Data Science for Health (DaScH) Lab, Fondazione Toscana Life Sciences, Via Fiorentina, 1, 53100, Siena, Italy
| | - Dario Cardamone
- Data Science for Health (DaScH) Lab, Fondazione Toscana Life Sciences, Via Fiorentina, 1, 53100, Siena, Italy
| | - Duccio Medini
- Data Science for Health (DaScH) Lab, Fondazione Toscana Life Sciences, Via Fiorentina, 1, 53100, Siena, Italy
| |
Collapse
|
18
|
Zhou B, Zheng L, Wu B, Tan Y, Lv O, Yi K, Fan G, Hong L. Protein Engineering with Lightweight Graph Denoising Neural Networks. J Chem Inf Model 2024; 64:3650-3661. [PMID: 38630581 DOI: 10.1021/acs.jcim.4c00036] [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: 04/19/2024]
Abstract
Protein engineering faces challenges in finding optimal mutants from a massive pool of candidate mutants. In this study, we introduce a deep-learning-based data-efficient fitness prediction tool to steer protein engineering. Our methodology establishes a lightweight graph neural network scheme for protein structures, which efficiently analyzes the microenvironment of amino acids in wild-type proteins and reconstructs the distribution of the amino acid sequences that are more likely to pass natural selection. This distribution serves as a general guidance for scoring proteins toward arbitrary properties on any order of mutations. Our proposed solution undergoes extensive wet-lab experimental validation spanning diverse physicochemical properties of various proteins, including fluorescence intensity, antigen-antibody affinity, thermostability, and DNA cleavage activity. More than 40% of ProtLGN-designed single-site mutants outperform their wild-type counterparts across all studied proteins and targeted properties. More importantly, our model can bypass the negative epistatic effect to combine single mutation sites and form deep mutants with up to seven mutation sites in a single round, whose physicochemical properties are significantly improved. This observation provides compelling evidence of the structure-based model's potential to guide deep mutations in protein engineering. Overall, our approach emerges as a versatile tool for protein engineering, benefiting both the computational and bioengineering communities.
Collapse
Affiliation(s)
- Bingxin Zhou
- Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- Shanghai National Center for Applied Mathematics (SJTU Center), Shanghai 200240, China
| | - Lirong Zheng
- Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Banghao Wu
- Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yang Tan
- Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- School of Information Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
- Shanghai Artificial Intelligence Laboratory, Shanghai 200232, China
| | - Outongyi Lv
- Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Kai Yi
- School of Mathematics and Statistics, University of New South Wales, Sydney 2052, Australia
| | - Guisheng Fan
- School of Information Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Liang Hong
- Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- Shanghai National Center for Applied Mathematics (SJTU Center), Shanghai 200240, China
- Shanghai Artificial Intelligence Laboratory, Shanghai 200232, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 201203, China
| |
Collapse
|
19
|
Ding K, Luo J, Luo Y. Leveraging conformal prediction to annotate enzyme function space with limited false positives. PLoS Comput Biol 2024; 20:e1012135. [PMID: 38809942 PMCID: PMC11164347 DOI: 10.1371/journal.pcbi.1012135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 06/10/2024] [Accepted: 05/03/2024] [Indexed: 05/31/2024] Open
Abstract
Machine learning (ML) is increasingly being used to guide biological discovery in biomedicine such as prioritizing promising small molecules in drug discovery. In those applications, ML models are used to predict the properties of biological systems, and researchers use these predictions to prioritize candidates as new biological hypotheses for downstream experimental validations. However, when applied to unseen situations, these models can be overconfident and produce a large number of false positives. One solution to address this issue is to quantify the model's prediction uncertainty and provide a set of hypotheses with a controlled false discovery rate (FDR) pre-specified by researchers. We propose CPEC, an ML framework for FDR-controlled biological discovery. We demonstrate its effectiveness using enzyme function annotation as a case study, simulating the discovery process of identifying the functions of less-characterized enzymes. CPEC integrates a deep learning model with a statistical tool known as conformal prediction, providing accurate and FDR-controlled function predictions for a given protein enzyme. Conformal prediction provides rigorous statistical guarantees to the predictive model and ensures that the expected FDR will not exceed a user-specified level with high probability. Evaluation experiments show that CPEC achieves reliable FDR control, better or comparable prediction performance at a lower FDR than existing methods, and accurate predictions for enzymes under-represented in the training data. We expect CPEC to be a useful tool for biological discovery applications where a high yield rate in validation experiments is desired but the experimental budget is limited.
Collapse
Affiliation(s)
- Kerr Ding
- School of Computational Science and Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Jiaqi Luo
- School of Computational Science and Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Yunan Luo
- School of Computational Science and Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| |
Collapse
|
20
|
Chen Y, Zha J, Xu S, Shao J, Liu X, Li D, Zhang X. Structure-Based Optimization of One Neutralizing Antibody against SARS-CoV-2 Variants Bearing the L452R Mutation. Viruses 2024; 16:566. [PMID: 38675908 PMCID: PMC11053997 DOI: 10.3390/v16040566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Neutralizing antibodies (nAbs) play an important role against SARS-CoV-2 infections. Previously, we have reported one potent receptor binding domain (RBD)-binding nAb Ab08 against the SARS-CoV-2 prototype and a panel of variants, but Ab08 showed much less efficacy against the variants harboring the L452R mutation. To overcome the antibody escape caused by the L452R mutation, we generated several structure-based Ab08 derivatives. One derivative, Ab08-K99E, displayed the mostly enhanced neutralizing potency against the Delta pseudovirus bearing the L452R mutation compared to the Ab08 and other derivatives. Ab08-K99E also showed improved neutralizing effects against the prototype, Omicron BA.1, and Omicron BA.4/5 pseudoviruses. In addition, compared to the original Ab08, Ab08-K99E exhibited high binding properties and affinities to the RBDs of the prototype, Delta, and Omicron BA.4/5 variants. Altogether, our findings report an optimized nAb, Ab08-K99E, against SARS-CoV-2 variants and demonstrate structure-based optimization as an effective way for antibody development against pathogens.
Collapse
Affiliation(s)
- Yamin Chen
- Suzhou Medical College, Soochow University, Suzhou 215123, China; (Y.C.); (X.L.)
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; (S.X.); (J.S.)
| | - Jialu Zha
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China;
| | - Shiqi Xu
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; (S.X.); (J.S.)
- The CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China
| | - Jiang Shao
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; (S.X.); (J.S.)
| | - Xiaoshan Liu
- Suzhou Medical College, Soochow University, Suzhou 215123, China; (Y.C.); (X.L.)
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; (S.X.); (J.S.)
| | - Dianfan Li
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China;
| | - Xiaoming Zhang
- Suzhou Medical College, Soochow University, Suzhou 215123, China; (Y.C.); (X.L.)
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; (S.X.); (J.S.)
- Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai 200052, China
| |
Collapse
|
21
|
Ozden B, Şamiloğlu E, Özsan A, Erguven M, Yükrük C, Koşaca M, Oktayoğlu M, Menteş M, Arslan N, Karakülah G, Barlas AB, Savaş B, Karaca E. Benchmarking the accuracy of structure-based binding affinity predictors on Spike-ACE2 deep mutational interaction set. Proteins 2024; 92:529-539. [PMID: 37991066 DOI: 10.1002/prot.26645] [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: 02/23/2023] [Revised: 10/25/2023] [Accepted: 11/13/2023] [Indexed: 11/23/2023]
Abstract
Since the start of COVID-19 pandemic, a huge effort has been devoted to understanding the Spike (SARS-CoV-2)-ACE2 recognition mechanism. To this end, two deep mutational scanning studies traced the impact of all possible mutations across receptor binding domain (RBD) of Spike and catalytic domain of human ACE2. By concentrating on the interface mutations of these experimental data, we benchmarked six commonly used structure-based binding affinity predictors (FoldX, EvoEF1, MutaBind2, SSIPe, HADDOCK, and UEP). These predictors were selected based on their user-friendliness, accessibility, and speed. As a result of our benchmarking efforts, we observed that none of the methods could generate a meaningful correlation with the experimental binding data. The best correlation is achieved by FoldX (R = -0.51). When we simplified the prediction problem to a binary classification, that is, whether a mutation is enriching or depleting the binding, we showed that the highest accuracy is achieved by FoldX with a 64% success rate. Surprisingly, on this set, simple energetic scoring functions performed significantly better than the ones using extra evolutionary-based terms, as in Mutabind and SSIPe. Furthermore, we demonstrated that recent AI approaches, mmCSM-PPI and TopNetTree, yielded comparable performances to the force field-based techniques. These observations suggest plenty of room to improve the binding affinity predictors in guessing the variant-induced binding profile changes of a host-pathogen system, such as Spike-ACE2. To aid such improvements we provide our benchmarking data at https://github.com/CSB-KaracaLab/RBD-ACE2-MutBench with the option to visualize our mutant models at https://rbd-ace2-mutbench.github.io/.
Collapse
Affiliation(s)
- Burcu Ozden
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, Turkey
| | - Eda Şamiloğlu
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, Turkey
| | - Atakan Özsan
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Mehmet Erguven
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Can Yükrük
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Mehdi Koşaca
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, Turkey
| | - Melis Oktayoğlu
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Muratcan Menteş
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Nazmiye Arslan
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Gökhan Karakülah
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, Turkey
| | - Ayşe Berçin Barlas
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, Turkey
| | - Büşra Savaş
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, Turkey
| | - Ezgi Karaca
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, Turkey
| |
Collapse
|
22
|
Liu W, Wang Z, You R, Xie C, Wei H, Xiong Y, Yang J, Zhu S. PLMSearch: Protein language model powers accurate and fast sequence search for remote homology. Nat Commun 2024; 15:2775. [PMID: 38555371 PMCID: PMC10981738 DOI: 10.1038/s41467-024-46808-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 03/08/2024] [Indexed: 04/02/2024] Open
Abstract
Homologous protein search is one of the most commonly used methods for protein annotation and analysis. Compared to structure search, detecting distant evolutionary relationships from sequences alone remains challenging. Here we propose PLMSearch (Protein Language Model), a homologous protein search method with only sequences as input. PLMSearch uses deep representations from a pre-trained protein language model and trains the similarity prediction model with a large number of real structure similarity. This enables PLMSearch to capture the remote homology information concealed behind the sequences. Extensive experimental results show that PLMSearch can search millions of query-target protein pairs in seconds like MMseqs2 while increasing the sensitivity by more than threefold, and is comparable to state-of-the-art structure search methods. In particular, unlike traditional sequence search methods, PLMSearch can recall most remote homology pairs with dissimilar sequences but similar structures. PLMSearch is freely available at https://dmiip.sjtu.edu.cn/PLMSearch .
Collapse
Affiliation(s)
- Wei Liu
- Institute of Science and Technology for Brain-Inspired Intelligence and MOE Frontiers Center for Brain Science, Fudan University, 200433, Shanghai, China
| | - Ziye Wang
- Institute of Science and Technology for Brain-Inspired Intelligence and MOE Frontiers Center for Brain Science, Fudan University, 200433, Shanghai, China
| | - Ronghui You
- Institute of Science and Technology for Brain-Inspired Intelligence and MOE Frontiers Center for Brain Science, Fudan University, 200433, Shanghai, China
| | - Chenghan Xie
- School of Mathematical Sciences, Fudan University, 200433, Shanghai, China
| | - Hong Wei
- School of Mathematical Sciences, Nankai University, 300071, Tianjin, China
| | - Yi Xiong
- Department of Bioinformatics and Biostatistics, Shanghai Jiao Tong University, 200240, Shanghai, China
| | - Jianyi Yang
- Ministry of Education Frontiers Science Center for Nonlinear Expectations, Research Center for Mathematics and Interdisciplinary Science, Shandong University, 266237, Qingdao, China.
| | - Shanfeng Zhu
- Institute of Science and Technology for Brain-Inspired Intelligence and MOE Frontiers Center for Brain Science, Fudan University, 200433, Shanghai, China.
- Shanghai Qi Zhi Institute, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China.
- Shanghai Key Lab of Intelligent Information Processing and Shanghai Institute of Artificial Intelligence Algorithm, Fudan University, Shanghai, China.
- Zhangjiang Fudan International Innovation Center, Shanghai, China.
| |
Collapse
|
23
|
Orlando G, Serrano L, Schymkowitz J, Rousseau F. Integrating physics in deep learning algorithms: a force field as a PyTorch module. Bioinformatics 2024; 40:btae160. [PMID: 38514422 PMCID: PMC11007235 DOI: 10.1093/bioinformatics/btae160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 02/08/2024] [Accepted: 03/19/2024] [Indexed: 03/23/2024] Open
Abstract
MOTIVATION Deep learning algorithms applied to structural biology often struggle to converge to meaningful solutions when limited data is available, since they are required to learn complex physical rules from examples. State-of-the-art force-fields, however, cannot interface with deep learning algorithms due to their implementation. RESULTS We present MadraX, a forcefield implemented as a differentiable PyTorch module, able to interact with deep learning algorithms in an end-to-end fashion. AVAILABILITY AND IMPLEMENTATION MadraX documentation, together with tutorials and installation guide, is available at madrax.readthedocs.io.
Collapse
Affiliation(s)
- Gabriele Orlando
- Switch Laboratory, VIB Center for Brain and Disease Research, VIB, Leuven 3000, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven 3000, Belgium
- Switch Laboratory, VIB Center for AI & Computational Biology, VIB, Leuven 3000, Belgium
| | - Luis Serrano
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr Aiguader 88, Barcelona 08003, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- IC REA, Pg. Lluis Companys 23, Barcelona 08010, Spain
| | - Joost Schymkowitz
- Switch Laboratory, VIB Center for Brain and Disease Research, VIB, Leuven 3000, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven 3000, Belgium
- Switch Laboratory, VIB Center for AI & Computational Biology, VIB, Leuven 3000, Belgium
| | - Frederic Rousseau
- Switch Laboratory, VIB Center for Brain and Disease Research, VIB, Leuven 3000, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven 3000, Belgium
- Switch Laboratory, VIB Center for AI & Computational Biology, VIB, Leuven 3000, Belgium
| |
Collapse
|
24
|
Kim DN, McNaughton AD, Kumar N. Leveraging Artificial Intelligence to Expedite Antibody Design and Enhance Antibody-Antigen Interactions. Bioengineering (Basel) 2024; 11:185. [PMID: 38391671 PMCID: PMC10886287 DOI: 10.3390/bioengineering11020185] [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: 12/30/2023] [Revised: 01/30/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024] Open
Abstract
This perspective sheds light on the transformative impact of recent computational advancements in the field of protein therapeutics, with a particular focus on the design and development of antibodies. Cutting-edge computational methods have revolutionized our understanding of protein-protein interactions (PPIs), enhancing the efficacy of protein therapeutics in preclinical and clinical settings. Central to these advancements is the application of machine learning and deep learning, which offers unprecedented insights into the intricate mechanisms of PPIs and facilitates precise control over protein functions. Despite these advancements, the complex structural nuances of antibodies pose ongoing challenges in their design and optimization. Our review provides a comprehensive exploration of the latest deep learning approaches, including language models and diffusion techniques, and their role in surmounting these challenges. We also present a critical analysis of these methods, offering insights to drive further progress in this rapidly evolving field. The paper includes practical recommendations for the application of these computational techniques, supplemented with independent benchmark studies. These studies focus on key performance metrics such as accuracy and the ease of program execution, providing a valuable resource for researchers engaged in antibody design and development. Through this detailed perspective, we aim to contribute to the advancement of antibody design, equipping researchers with the tools and knowledge to navigate the complexities of this field.
Collapse
Affiliation(s)
| | | | - Neeraj Kumar
- Pacific Northwest National Laboratory, 902 Battelle Blvd., Richland, WA 99352, USA; (D.N.K.); (A.D.M.)
| |
Collapse
|
25
|
Hie BL, Shanker VR, Xu D, Bruun TUJ, Weidenbacher PA, Tang S, Wu W, Pak JE, Kim PS. Efficient evolution of human antibodies from general protein language models. Nat Biotechnol 2024; 42:275-283. [PMID: 37095349 PMCID: PMC10869273 DOI: 10.1038/s41587-023-01763-2] [Citation(s) in RCA: 104] [Impact Index Per Article: 104.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 03/28/2023] [Indexed: 04/26/2023]
Abstract
Natural evolution must explore a vast landscape of possible sequences for desirable yet rare mutations, suggesting that learning from natural evolutionary strategies could guide artificial evolution. Here we report that general protein language models can efficiently evolve human antibodies by suggesting mutations that are evolutionarily plausible, despite providing the model with no information about the target antigen, binding specificity or protein structure. We performed language-model-guided affinity maturation of seven antibodies, screening 20 or fewer variants of each antibody across only two rounds of laboratory evolution, and improved the binding affinities of four clinically relevant, highly mature antibodies up to sevenfold and three unmatured antibodies up to 160-fold, with many designs also demonstrating favorable thermostability and viral neutralization activity against Ebola and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pseudoviruses. The same models that improve antibody binding also guide efficient evolution across diverse protein families and selection pressures, including antibiotic resistance and enzyme activity, suggesting that these results generalize to many settings.
Collapse
Affiliation(s)
- Brian L Hie
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA.
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA.
| | - Varun R Shanker
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Duo Xu
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Theodora U J Bruun
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Payton A Weidenbacher
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Shaogeng Tang
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Wesley Wu
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - John E Pak
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Peter S Kim
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA.
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
26
|
Irvine EB, Reddy ST. Advancing Antibody Engineering through Synthetic Evolution and Machine Learning. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:235-243. [PMID: 38166249 DOI: 10.4049/jimmunol.2300492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/20/2023] [Indexed: 01/04/2024]
Abstract
Abs are versatile molecules with the potential to achieve exceptional binding to target Ags, while also possessing biophysical properties suitable for therapeutic drug development. Protein display and directed evolution systems have transformed synthetic Ab discovery, engineering, and optimization, vastly expanding the number of Ab clones able to be experimentally screened for binding. Moreover, the burgeoning integration of high-throughput screening, deep sequencing, and machine learning has further augmented in vitro Ab optimization, promising to accelerate the design process and massively expand the Ab sequence space interrogated. In this Brief Review, we discuss the experimental and computational tools employed in synthetic Ab engineering and optimization. We also explore the therapeutic challenges posed by developing Abs for infectious diseases, and the prospects for leveraging machine learning-guided protein engineering to prospectively design Abs resistant to viral escape.
Collapse
Affiliation(s)
- Edward B Irvine
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Sai T Reddy
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| |
Collapse
|
27
|
Luo M, Zhou B, Reddem ER, Tang B, Chen B, Zhou R, Liu H, Liu L, Katsamba PS, Au KK, Man HO, To KKW, Yuen KY, Shapiro L, Dang S, Ho DD, Chen Z. Structural insights into broadly neutralizing antibodies elicited by hybrid immunity against SARS-CoV-2. Emerg Microbes Infect 2023; 12:2146538. [PMID: 36354024 PMCID: PMC9817130 DOI: 10.1080/22221751.2022.2146538] [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: 09/30/2022] [Accepted: 11/08/2022] [Indexed: 11/11/2022]
Abstract
ABSTRACTIncreasing spread by SARS-CoV-2 Omicron variants challenges existing vaccines and broadly reactive neutralizing antibodies (bNAbs) against COVID-19. Here we determine the diversity, potency, breadth and structural insights of bNAbs derived from memory B cells of BNT162b2-vaccinee after homogeneous Omicron BA.1 breakthrough infection. The infection activates diverse memory B cell clonotypes for generating potent class I/II and III bNAbs with new epitopes mapped to the receptor-binding domain (RBD). The top eight bNAbs neutralize wildtype and BA.1 potently but display divergent IgH/IgL sequences and neuralization profiles against other variants of concern (VOCs). Two of them (P2D9 and P3E6) belonging to class III NAbs display comparable potency against BA.4/BA.5, although structural analysis reveals distinct modes of action. P3E6 neutralizes all variants tested through a unique bivalent interaction with two RBDs. Our findings provide new insights into hybrid immunity on BNT162b2-induced diverse memory B cells in response to Omicron breakthrough infection for generating diverse bNAbs with distinct structural basis.
Collapse
Affiliation(s)
- Mengxiao Luo
- AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Biao Zhou
- AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | | | - Bingjie Tang
- Division of Life Science, Center of Systems Biology and Human Health, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong Special Administrative Region, People’s Republic of China
| | - Bohao Chen
- AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Runhong Zhou
- AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Hang Liu
- Division of Life Science, Center of Systems Biology and Human Health, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong Special Administrative Region, People’s Republic of China
| | - Lihong Liu
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | | | - Ka-Kit Au
- AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Hiu-On Man
- AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Kelvin Kai-Wang To
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Centre for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, People’s Republic of China
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Kwok-Yung Yuen
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Centre for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, People’s Republic of China
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Lawrence Shapiro
- Zuckerman Mind Brain Behaviour Institute, New York, NY, USA
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Shangyu Dang
- Division of Life Science, Center of Systems Biology and Human Health, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong Special Administrative Region, People’s Republic of China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, People’s Republic of China
- HKUST-Shenzhen Research Institute, Nanshan, People’s Republic of China
| | - David D. Ho
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Zhiwei Chen
- AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Centre for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, People’s Republic of China
| |
Collapse
|
28
|
Yang X. Passive antibody therapy in emerging infectious diseases. Front Med 2023; 17:1117-1134. [PMID: 38040914 DOI: 10.1007/s11684-023-1021-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 07/20/2023] [Indexed: 12/03/2023]
Abstract
The epidemic of corona virus disease 2019 (COVID-19) caused by severe acute respiratory syndrome Coronavirus 2 and its variants of concern (VOCs) has been ongoing for over 3 years. Antibody therapies encompassing convalescent plasma, hyperimmunoglobulin, and neutralizing monoclonal antibodies (mAbs) applied in passive immunotherapy have yielded positive outcomes and played a crucial role in the early COVID-19 treatment. In this review, the development path, action mechanism, clinical research results, challenges, and safety profile associated with the use of COVID-19 convalescent plasma, hyperimmunoglobulin, and mAbs were summarized. In addition, the prospects of applying antibody therapy against VOCs was assessed, offering insights into the coping strategies for facing new infectious disease outbreaks.
Collapse
Affiliation(s)
- Xiaoming Yang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan, 430207, China.
- Wuhan Institute of Biological Products Co., Ltd., Wuhan, 430207, China.
- China National Biotec Group Company Limited, Beijing, 100029, China.
| |
Collapse
|
29
|
Sun H, Wang Y, Chen X, Jiang Y, Wang S, Huang Y, Liu L, Li Y, Lan M, Guo H, Yuan Q, Zhang Y, Li T, Yu H, Gu Y, Zhang J, Li S, Zheng Z, Zheng Q, Xia N. Structural basis for broad neutralization of human antibody against Omicron sublineages and evasion by XBB variant. J Virol 2023; 97:e0113723. [PMID: 37855619 PMCID: PMC10688377 DOI: 10.1128/jvi.01137-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/18/2023] [Indexed: 10/20/2023] Open
Abstract
IMPORTANCE The ongoing COVID-19 pandemic has been characterized by the emergence of new SARS-CoV-2 variants including the highly transmissible Omicron XBB sublineages, which have shown significant resistance to neutralizing antibodies (nAbs). This resistance has led to decreased vaccine effectiveness and therefore result in breakthrough infections and reinfections, which continuously threaten public health. To date, almost all available therapeutic nAbs, including those authorized under Emergency Use Authorization nAbs that were previously clinically useful against early strains, have recently been found to be ineffective against newly emerging variants. In this study, we provide a comprehensive structural basis about how the Class 3 nAbs, including 1G11 in this study and noted LY-CoV1404, are evaded by the newly emerged SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Hui Sun
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yizhen Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, China
| | - Xiuting Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yanan Jiang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, China
| | - Siling Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yang Huang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, China
| | - Liqin Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yu Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, China
| | - Miaolin Lan
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, China
| | - Huilin Guo
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, China
| | - Quan Yuan
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, China
- Xiang An Biomedicine Laboratory, Xiamen, China
| | - Yali Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, China
- Xiang An Biomedicine Laboratory, Xiamen, China
| | - Tingting Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, China
- Xiang An Biomedicine Laboratory, Xiamen, China
| | - Hai Yu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, China
- Xiang An Biomedicine Laboratory, Xiamen, China
| | - Ying Gu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, China
- Xiang An Biomedicine Laboratory, Xiamen, China
| | - Jun Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, China
- Xiang An Biomedicine Laboratory, Xiamen, China
| | - Shaowei Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, China
- Xiang An Biomedicine Laboratory, Xiamen, China
| | - Zizheng Zheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, China
- Xiang An Biomedicine Laboratory, Xiamen, China
| | - Qingbing Zheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, China
- Xiang An Biomedicine Laboratory, Xiamen, China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, China
- Xiang An Biomedicine Laboratory, Xiamen, China
- Research Unit of Frontier Technology of Structural Vaccinology, Chinese Academy of Medical Sciences, Xiamen, China
| |
Collapse
|
30
|
Clark T, Subramanian V, Jayaraman A, Fitzpatrick E, Gopal R, Pentakota N, Rurak T, Anand S, Viglione A, Raman R, Tharakaraman K, Sasisekharan R. Enhancing antibody affinity through experimental sampling of non-deleterious CDR mutations predicted by machine learning. Commun Chem 2023; 6:244. [PMID: 37945793 PMCID: PMC10636138 DOI: 10.1038/s42004-023-01037-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 10/20/2023] [Indexed: 11/12/2023] Open
Abstract
The application of machine learning (ML) models to optimize antibody affinity to an antigen is gaining prominence. Unfortunately, the small and biased nature of the publicly available antibody-antigen interaction datasets makes it challenging to build an ML model that can accurately predict binding affinity changes due to mutations (ΔΔG). Recognizing these inherent limitations, we reformulated the problem to ask whether an ML model capable of classifying deleterious vs non-deleterious mutations can guide antibody affinity maturation in a practical setting. To test this hypothesis, we developed a Random Forest classifier (Antibody Random Forest Classifier or AbRFC) with expert-guided features and integrated it into a computational-experimental workflow. AbRFC effectively predicted non-deleterious mutations on an in-house validation dataset that is free of biases seen in the publicly available training datasets. Furthermore, experimental screening of a limited number of predictions from the model (<10^2 designs) identified affinity-enhancing mutations in two unrelated SARS-CoV-2 antibodies, resulting in constructs with up to 1000-fold increased binding to the SARS-COV-2 RBD. Our findings indicate that accurate prediction and screening of non-deleterious mutations using machine learning offers a powerful approach to improving antibody affinity.
Collapse
Affiliation(s)
- Thomas Clark
- Altus Enterprises, 900 Middlesex Turnpike, Billerica, MA, USA
| | | | - Akila Jayaraman
- Altus Enterprises, 900 Middlesex Turnpike, Billerica, MA, USA
| | | | - Ranjani Gopal
- Altus Enterprises, 900 Middlesex Turnpike, Billerica, MA, USA
| | | | - Troy Rurak
- Altus Enterprises, 900 Middlesex Turnpike, Billerica, MA, USA
| | - Shweta Anand
- Altus Enterprises, 900 Middlesex Turnpike, Billerica, MA, USA
| | | | - Rahul Raman
- Department of Biological Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA.
| | | | - Ram Sasisekharan
- Department of Biological Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA.
| |
Collapse
|
31
|
Kouba P, Kohout P, Haddadi F, Bushuiev A, Samusevich R, Sedlar J, Damborsky J, Pluskal T, Sivic J, Mazurenko S. Machine Learning-Guided Protein Engineering. ACS Catal 2023; 13:13863-13895. [PMID: 37942269 PMCID: PMC10629210 DOI: 10.1021/acscatal.3c02743] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/20/2023] [Indexed: 11/10/2023]
Abstract
Recent progress in engineering highly promising biocatalysts has increasingly involved machine learning methods. These methods leverage existing experimental and simulation data to aid in the discovery and annotation of promising enzymes, as well as in suggesting beneficial mutations for improving known targets. The field of machine learning for protein engineering is gathering steam, driven by recent success stories and notable progress in other areas. It already encompasses ambitious tasks such as understanding and predicting protein structure and function, catalytic efficiency, enantioselectivity, protein dynamics, stability, solubility, aggregation, and more. Nonetheless, the field is still evolving, with many challenges to overcome and questions to address. In this Perspective, we provide an overview of ongoing trends in this domain, highlight recent case studies, and examine the current limitations of machine learning-based methods. We emphasize the crucial importance of thorough experimental validation of emerging models before their use for rational protein design. We present our opinions on the fundamental problems and outline the potential directions for future research.
Collapse
Affiliation(s)
- Petr Kouba
- Loschmidt
Laboratories, Department of Experimental Biology and RECETOX, Faculty
of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech
Republic
- Czech Institute
of Informatics, Robotics and Cybernetics, Czech Technical University in Prague, Jugoslavskych partyzanu 1580/3, 160 00 Prague 6, Czech Republic
- Faculty of
Electrical Engineering, Czech Technical
University in Prague, Technicka 2, 166 27 Prague 6, Czech Republic
| | - Pavel Kohout
- Loschmidt
Laboratories, Department of Experimental Biology and RECETOX, Faculty
of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech
Republic
- International
Clinical Research Center, St. Anne’s
University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| | - Faraneh Haddadi
- Loschmidt
Laboratories, Department of Experimental Biology and RECETOX, Faculty
of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech
Republic
- International
Clinical Research Center, St. Anne’s
University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| | - Anton Bushuiev
- Czech Institute
of Informatics, Robotics and Cybernetics, Czech Technical University in Prague, Jugoslavskych partyzanu 1580/3, 160 00 Prague 6, Czech Republic
| | - Raman Samusevich
- Czech Institute
of Informatics, Robotics and Cybernetics, Czech Technical University in Prague, Jugoslavskych partyzanu 1580/3, 160 00 Prague 6, Czech Republic
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 160 00 Prague 6, Czech Republic
| | - Jiri Sedlar
- Czech Institute
of Informatics, Robotics and Cybernetics, Czech Technical University in Prague, Jugoslavskych partyzanu 1580/3, 160 00 Prague 6, Czech Republic
| | - Jiri Damborsky
- Loschmidt
Laboratories, Department of Experimental Biology and RECETOX, Faculty
of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech
Republic
- International
Clinical Research Center, St. Anne’s
University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| | - Tomas Pluskal
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 160 00 Prague 6, Czech Republic
| | - Josef Sivic
- Czech Institute
of Informatics, Robotics and Cybernetics, Czech Technical University in Prague, Jugoslavskych partyzanu 1580/3, 160 00 Prague 6, Czech Republic
| | - Stanislav Mazurenko
- Loschmidt
Laboratories, Department of Experimental Biology and RECETOX, Faculty
of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech
Republic
- International
Clinical Research Center, St. Anne’s
University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| |
Collapse
|
32
|
Guo Y, Li T. Modeling the competitive transmission of the Omicron strain and Delta strain of COVID-19. JOURNAL OF MATHEMATICAL ANALYSIS AND APPLICATIONS 2023; 526:127283. [PMID: 37035507 PMCID: PMC10065814 DOI: 10.1016/j.jmaa.2023.127283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Indexed: 06/19/2023]
Abstract
Since November 2021, there have been cases of COVID-19's Omicron strain spreading in competition with Delta strains in many parts of the world. To explore how these two strains developed in this competitive spread, a new compartmentalized model was established. First, we analyzed the fundamental properties of the model, obtained the expression of the basic reproduction number, proved the local and global asymptotic stability of the disease-free equilibrium. Then by means of the cubic spline interpolation method, we obtained the data of new Omicron and Delta cases in the United States of new cases starting from December 8, 2021, to February 12, 2022. Using the weighted nonlinear least squares estimation method, we fitted six time series (cumulative confirmed cases, cumulative deaths, new cases, new deaths, new Omicron cases, and new Delta cases), got estimates of the unknown parameters, and obtained an approximation of the basic reproduction number in the United States during this time period as R 0 ≈ 1.5165 . Finally, each control strategy was evaluated by cost-effectiveness analysis to obtain the optimal control strategy under different perspectives. The results not only show the competitive transmission characteristics of the new strain and existing strain, but also provide scientific suggestions for effectively controlling the spread of these strains.
Collapse
Affiliation(s)
- Youming Guo
- College of Science, Guilin University of Technology, Guilin, Guangxi 541004, PR China
- Guangxi Colleges and Universities Key Laboratory of Applied Statistics, Guilin University of Technology, Guilin, Guangxi 541004, PR China
| | - Tingting Li
- College of Science, Guilin University of Technology, Guilin, Guangxi 541004, PR China
- Guangxi Colleges and Universities Key Laboratory of Applied Statistics, Guilin University of Technology, Guilin, Guangxi 541004, PR China
| |
Collapse
|
33
|
Zhou Y, Huang Z, Li W, Wei J, Jiang Q, Yang W, Huang J. Deep learning in preclinical antibody drug discovery and development. Methods 2023; 218:57-71. [PMID: 37454742 DOI: 10.1016/j.ymeth.2023.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/20/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023] Open
Abstract
Antibody drugs have become a key part of biotherapeutics. Patients suffering from various diseases have benefited from antibody therapies. However, its development process is rather long, expensive and risky. To speed up the process, reduce cost and improve success rate, artificial intelligence, especially deep learning methods, have been widely used in all aspects of preclinical antibody drug development, from library generation to hit identification, developability screening, lead selection and optimization. In this review, we systematically summarize antibody encodings, deep learning architectures and models used in preclinical antibody drug discovery and development. We also critically discuss challenges and opportunities, problems and possible solutions, current applications and future directions of deep learning in antibody drug development.
Collapse
Affiliation(s)
- Yuwei Zhou
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Ziru Huang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Wenzhen Li
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Jinyi Wei
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Qianhu Jiang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Wei Yang
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Jian Huang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China.
| |
Collapse
|
34
|
Shah M, Woo HG. Assessment of neutralization susceptibility of Omicron subvariants XBB.1.5 and BQ.1.1 against broad-spectrum neutralizing antibodies through epitopes mapping. Front Mol Biosci 2023; 10:1236617. [PMID: 37828918 PMCID: PMC10565033 DOI: 10.3389/fmolb.2023.1236617] [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: 06/08/2023] [Accepted: 08/31/2023] [Indexed: 10/14/2023] Open
Abstract
The emergence of new variants of the SARS-CoV-2 virus has posed a significant challenge in developing broadly neutralizing antibodies (nAbs) with guaranteed therapeutic potential. Some nAbs, such as Sotrovimab, have exhibited varying levels of efficacy against different variants, while others, such as Bebtelovimab and Bamlanivimab-etesevimab are ineffective against specific variants, including BQ.1.1 and XBB. This highlights the urgent need for developing broadly active monoclonal antibodies (mAbs) providing prophylactic and therapeutic benefits to high-risk patients, especially in the face of the risk of reinfection from new variants. Here, we aimed to investigate the feasibility of redirecting existing mAbs against new variants of SARS-CoV-2, as well as to understand how BQ.1.1 and XBB.1.5 can evade broadly neutralizing mAbs. By mapping epitopes and escape sites, we discovered that the new variants evade multiple mAbs, including FDA-approved Bebtelovimab, which showed resilience against other Omicron variants. Our approach, which included simulations, endpoint free energy calculation, and shape complementarity analysis, revealed the possibility of identifying mAbs that are effective against both BQ.1.1 and XBB.1.5. We identified two broad-spectrum mAbs, R200-1F9 and R207-2F11, as potential candidates with increased binding affinity to XBB.1.5 and BQ.1.1 compared to the reference (Wu01) strain. Additionally, we propose that these mAbs do not interfere with Angiotensin Converting Enzyme 2 (ACE2) and bind to conserved epitopes on the receptor binding domain of Spike that are not-overlapping, potentially providing a solution to neutralize these new variants either independently or as part of a combination (cocktail) treatment.
Collapse
Affiliation(s)
- Masaud Shah
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Hyun Goo Woo
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
- Department of Biomedical Science, Graduate School, Ajou University, Suwon, Republic of Korea
| |
Collapse
|
35
|
Qiu Y, Wei GW. Artificial intelligence-aided protein engineering: from topological data analysis to deep protein language models. Brief Bioinform 2023; 24:bbad289. [PMID: 37580175 PMCID: PMC10516362 DOI: 10.1093/bib/bbad289] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/14/2023] [Accepted: 07/26/2023] [Indexed: 08/16/2023] Open
Abstract
Protein engineering is an emerging field in biotechnology that has the potential to revolutionize various areas, such as antibody design, drug discovery, food security, ecology, and more. However, the mutational space involved is too vast to be handled through experimental means alone. Leveraging accumulative protein databases, machine learning (ML) models, particularly those based on natural language processing (NLP), have considerably expedited protein engineering. Moreover, advances in topological data analysis (TDA) and artificial intelligence-based protein structure prediction, such as AlphaFold2, have made more powerful structure-based ML-assisted protein engineering strategies possible. This review aims to offer a comprehensive, systematic, and indispensable set of methodological components, including TDA and NLP, for protein engineering and to facilitate their future development.
Collapse
Affiliation(s)
- Yuchi Qiu
- Department of Mathematics, Michigan State University, East Lansing, 48824 MI, USA
| | - Guo-Wei Wei
- Department of Mathematics, Michigan State University, East Lansing, 48824 MI, USA
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, 48824 MI, USA
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, 48824 MI, USA
| |
Collapse
|
36
|
Wang G, Liu X, Wang K, Gao Y, Li G, Baptista-Hon DT, Yang XH, Xue K, Tai WH, Jiang Z, Cheng L, Fok M, Lau JYN, Yang S, Lu L, Zhang P, Zhang K. Deep-learning-enabled protein-protein interaction analysis for prediction of SARS-CoV-2 infectivity and variant evolution. Nat Med 2023; 29:2007-2018. [PMID: 37524952 DOI: 10.1038/s41591-023-02483-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 06/28/2023] [Indexed: 08/02/2023]
Abstract
Host-pathogen interactions and pathogen evolution are underpinned by protein-protein interactions between viral and host proteins. An understanding of how viral variants affect protein-protein binding is important for predicting viral-host interactions, such as the emergence of new pathogenic SARS-CoV-2 variants. Here we propose an artificial intelligence-based framework called UniBind, in which proteins are represented as a graph at the residue and atom levels. UniBind integrates protein three-dimensional structure and binding affinity and is capable of multi-task learning for heterogeneous biological data integration. In systematic tests on benchmark datasets and further experimental validation, UniBind effectively and scalably predicted the effects of SARS-CoV-2 spike protein variants on their binding affinities to the human ACE2 receptor, as well as to SARS-CoV-2 neutralizing monoclonal antibodies. Furthermore, in a cross-species analysis, UniBind could be applied to predict host susceptibility to SARS-CoV-2 variants and to predict future viral variant evolutionary trends. This in silico approach has the potential to serve as an early warning system for problematic emerging SARS-CoV-2 variants, as well as to facilitate research on protein-protein interactions in general.
Collapse
Affiliation(s)
- Guangyu Wang
- State Key Laboratory of Networking and Switching Technology, Beijing University of Posts and Telecommunications, Beijing, China.
| | - Xiaohong Liu
- Instutite for Artificial Intelligence in Medicine and Faculty of Medicine, Macau University of Science and Technology, Macau, China
- UCL Cancer Institute, University College London, London, UK
| | - Kai Wang
- Department of Big Data and Biomedical Artificial Intelligence, National Biomedical Imaging Center, College of Future Technology, Peking University and Peking-Tsinghua Center for Life Sciences, Beijing, China
| | - Yuanxu Gao
- Guangzhou National Laboratory, Guangzhou, China
| | - Gen Li
- Guangzhou National Laboratory, Guangzhou, China
- Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Daniel T Baptista-Hon
- Instutite for Artificial Intelligence in Medicine and Faculty of Medicine, Macau University of Science and Technology, Macau, China
- Zhuhai International Eye Center and Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital and the First Affiliated Hospital of Faculty of Medicine, Macau University of Science and Technology, Guangdong, China
| | - Xiaohong Helena Yang
- Instutite for Artificial Intelligence in Medicine and Faculty of Medicine, Macau University of Science and Technology, Macau, China
| | - Kanmin Xue
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Wa Hou Tai
- Instutite for Artificial Intelligence in Medicine and Faculty of Medicine, Macau University of Science and Technology, Macau, China
| | - Zeyu Jiang
- State Key Laboratory of Networking and Switching Technology, Beijing University of Posts and Telecommunications, Beijing, China
| | - Linling Cheng
- Instutite for Artificial Intelligence in Medicine and Faculty of Medicine, Macau University of Science and Technology, Macau, China
- Zhuhai International Eye Center and Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital and the First Affiliated Hospital of Faculty of Medicine, Macau University of Science and Technology, Guangdong, China
| | - Manson Fok
- Instutite for Artificial Intelligence in Medicine and Faculty of Medicine, Macau University of Science and Technology, Macau, China
| | - Johnson Yiu-Nam Lau
- Departments of Biology and Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Shengyong Yang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ligong Lu
- Instutite for Artificial Intelligence in Medicine and Faculty of Medicine, Macau University of Science and Technology, Macau, China
- Zhuhai International Eye Center and Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital and the First Affiliated Hospital of Faculty of Medicine, Macau University of Science and Technology, Guangdong, China
| | - Ping Zhang
- State Key Laboratory of Networking and Switching Technology, Beijing University of Posts and Telecommunications, Beijing, China
| | - Kang Zhang
- Instutite for Artificial Intelligence in Medicine and Faculty of Medicine, Macau University of Science and Technology, Macau, China.
- Department of Big Data and Biomedical Artificial Intelligence, National Biomedical Imaging Center, College of Future Technology, Peking University and Peking-Tsinghua Center for Life Sciences, Beijing, China.
- Guangzhou National Laboratory, Guangzhou, China.
- Zhuhai International Eye Center and Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital and the First Affiliated Hospital of Faculty of Medicine, Macau University of Science and Technology, Guangdong, China.
| |
Collapse
|
37
|
Qiu Y, Wei GW. Artificial intelligence-aided protein engineering: from topological data analysis to deep protein language models. ARXIV 2023:arXiv:2307.14587v1. [PMID: 37547662 PMCID: PMC10402185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Protein engineering is an emerging field in biotechnology that has the potential to revolutionize various areas, such as antibody design, drug discovery, food security, ecology, and more. However, the mutational space involved is too vast to be handled through experimental means alone. Leveraging accumulative protein databases, machine learning (ML) models, particularly those based on natural language processing (NLP), have considerably expedited protein engineering. Moreover, advances in topological data analysis (TDA) and artificial intelligence-based protein structure prediction, such as AlphaFold2, have made more powerful structure-based ML-assisted protein engineering strategies possible. This review aims to offer a comprehensive, systematic, and indispensable set of methodological components, including TDA and NLP, for protein engineering and to facilitate their future development.
Collapse
Affiliation(s)
- Yuchi Qiu
- Department of Mathematics, Michigan State University, East Lansing, 48824, MI, USA
| | - Guo-Wei Wei
- Department of Mathematics, Michigan State University, East Lansing, 48824, MI, USA
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, 48824, MI, USA
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, 48824, MI, USA
| |
Collapse
|
38
|
Yang X, Duan H, Liu X, Zhang X, Pan S, Zhang F, Gao P, Liu B, Yang J, Chi X, Yang W. Broad Sarbecovirus Neutralizing Antibodies Obtained by Computational Design and Synthetic Library Screening. J Virol 2023:e0061023. [PMID: 37367229 PMCID: PMC10373554 DOI: 10.1128/jvi.00610-23] [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: 04/27/2023] [Accepted: 06/10/2023] [Indexed: 06/28/2023] Open
Abstract
Members of the Sarbecovirus subgenus of Coronaviridae have twice caused deadly threats to humans. There is increasing concern about the rapid mutation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which has evolved into multiple generations of epidemic variants in 3 years. Broad neutralizing antibodies are of great importance for pandemic preparedness against SARS-CoV-2 variants and divergent zoonotic sarbecoviruses. Here, we analyzed the structural conservation of the receptor-binding domain (RBD) from representative sarbecoviruses and chose S2H97, a previously reported RBD antibody with ideal breadth and resistance to escape, as a template for computational design to enhance the neutralization activity and spectrum. A total of 35 designs were purified for evaluation. The neutralizing activity of a large proportion of these designs against multiple variants was increased from several to hundreds of times. Molecular dynamics simulation suggested that extra interface contacts and enhanced intermolecular interactions between the RBD and the designed antibodies are established. After light and heavy chain reconstitution, AI-1028, with five complementarity determining regions optimized, showed the best neutralizing activity across all tested sarbecoviruses, including SARS-CoV, multiple SARS-CoV-2 variants, and bat-derived viruses. AI-1028 recognized the same cryptic RBD epitope as the parental prototype antibody. In addition to computational design, chemically synthesized nanobody libraries are also a precious resource for rapid antibody development. By applying distinct RBDs as baits for reciprocal screening, we identified two novel nanobodies with broad activities. These findings provide potential pan-sarbecovirus neutralizing drugs and highlight new pathways to rapidly optimize therapeutic candidates when novel SARS-CoV-2 escape variants or new zoonotic coronaviruses emerge. IMPORTANCE The subgenus Sarbecovirus includes human SARS-CoV, SARS-CoV-2, and hundreds of genetically related bat viruses. The continuous evolution of SARS-CoV-2 has led to the striking evasion of neutralizing antibody (NAb) drugs and convalescent plasma. Antibodies with broad activity across sarbecoviruses would be helpful to combat current SARS-CoV-2 mutations and longer term animal virus spillovers. The study of pan-sarbecovirus NAbs described here is significant for the following reasons. First, we established a structure-based computational pipeline to design and optimize NAbs to obtain more potent and broader neutralizing activity across multiple sarbecoviruses. Second, we screened and identified nanobodies from a highly diversified synthetic library with a broad neutralizing spectrum using an elaborate screening strategy. These methodologies provide guidance for the rapid development of antibody therapeutics against emerging pathogens with highly variable characteristics.
Collapse
Affiliation(s)
- Xuehua Yang
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Huarui Duan
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiuying Liu
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xinhui Zhang
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shengnan Pan
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fangyuan Zhang
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Peixiang Gao
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Bo Liu
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jian Yang
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaojing Chi
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wei Yang
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
39
|
Li L, Gupta E, Spaeth J, Shing L, Jaimes R, Engelhart E, Lopez R, Caceres RS, Bepler T, Walsh ME. Machine learning optimization of candidate antibody yields highly diverse sub-nanomolar affinity antibody libraries. Nat Commun 2023; 14:3454. [PMID: 37308471 DOI: 10.1038/s41467-023-39022-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 05/23/2023] [Indexed: 06/14/2023] Open
Abstract
Therapeutic antibodies are an important and rapidly growing drug modality. However, the design and discovery of early-stage antibody therapeutics remain a time and cost-intensive endeavor. Here we present an end-to-end Bayesian, language model-based method for designing large and diverse libraries of high-affinity single-chain variable fragments (scFvs) that are then empirically measured. In a head-to-head comparison with a directed evolution approach, we show that the best scFv generated from our method represents a 28.7-fold improvement in binding over the best scFv from the directed evolution. Additionally, 99% of designed scFvs in our most successful library are improvements over the initial candidate scFv. By comparing a library's predicted success to actual measurements, we demonstrate our method's ability to explore tradeoffs between library success and diversity. Results of our work highlight the significant impact machine learning models can have on scFv development. We expect our method to be broadly applicable and provide value to other protein engineering tasks.
Collapse
Affiliation(s)
- Lin Li
- Massachusetts Institute of Technology Lincoln Laboratory, Lexington, MA, USA.
| | - Esther Gupta
- Massachusetts Institute of Technology Lincoln Laboratory, Lexington, MA, USA
| | - John Spaeth
- Massachusetts Institute of Technology Lincoln Laboratory, Lexington, MA, USA
| | - Leslie Shing
- Massachusetts Institute of Technology Lincoln Laboratory, Lexington, MA, USA
| | - Rafael Jaimes
- Massachusetts Institute of Technology Lincoln Laboratory, Lexington, MA, USA
| | | | | | - Rajmonda S Caceres
- Massachusetts Institute of Technology Lincoln Laboratory, Lexington, MA, USA
| | - Tristan Bepler
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Simons Electron Microscopy Center, New York Structural Biology Center, New York, NY, USA
| | - Matthew E Walsh
- Massachusetts Institute of Technology Lincoln Laboratory, Lexington, MA, USA
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
40
|
Sheng Z, Bimela JS, Wang M, Li Z, Guo Y, Ho DD. An optimized thermodynamics integration protocol for identifying beneficial mutations in antibody design. Front Immunol 2023; 14:1190416. [PMID: 37275896 PMCID: PMC10235760 DOI: 10.3389/fimmu.2023.1190416] [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: 03/20/2023] [Accepted: 04/28/2023] [Indexed: 06/07/2023] Open
Abstract
Accurate identification of beneficial mutations is central to antibody design. Many knowledge-based (KB) computational approaches have been developed to predict beneficial mutations, but their accuracy leaves room for improvement. Thermodynamic integration (TI) is an alchemical free energy algorithm that offers an alternative technique for identifying beneficial mutations, but its performance has not been evaluated. In this study, we developed an efficient TI protocol with high accuracy for predicting binding free energy changes of antibody mutations. The improved TI method outperforms KB methods at identifying both beneficial and deleterious mutations. We observed that KB methods have higher accuracies in predicting deleterious mutations than beneficial mutations. A pipeline using KB methods to efficiently exclude deleterious mutations and TI to accurately identify beneficial mutations was developed for high-throughput mutation scanning. The pipeline was applied to optimize the binding affinity of a broadly sarbecovirus neutralizing antibody 10-40 against the circulating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) omicron variant. Three identified beneficial mutations show strong synergy and improve both binding affinity and neutralization potency of antibody 10-40. Molecular dynamics simulation revealed that the three mutations improve the binding affinity of antibody 10-40 through the stabilization of an altered binding mode with increased polar and hydrophobic interactions. Above all, this study presents an accurate and efficient TI-based approach for optimizing antibodies and other biomolecules.
Collapse
Affiliation(s)
- Zizhang Sheng
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Jude S. Bimela
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, United States
| | - Maple Wang
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Zhiteng Li
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Yicheng Guo
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - David D. Ho
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| |
Collapse
|
41
|
Durham J, Zhang J, Humphreys IR, Pei J, Cong Q. Recent advances in predicting and modeling protein-protein interactions. Trends Biochem Sci 2023; 48:527-538. [PMID: 37061423 DOI: 10.1016/j.tibs.2023.03.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 03/03/2023] [Accepted: 03/17/2023] [Indexed: 04/17/2023]
Abstract
Protein-protein interactions (PPIs) drive biological processes, and disruption of PPIs can cause disease. With recent breakthroughs in structure prediction and a deluge of genomic sequence data, computational methods to predict PPIs and model spatial structures of protein complexes are now approaching the accuracy of experimental approaches for permanent interactions and show promise for elucidating transient interactions. As we describe here, the key to this success is rich evolutionary information deciphered from thousands of homologous sequences that coevolve in interacting partners. This covariation signal, revealed by sophisticated statistical and machine learning (ML) algorithms, predicts physiological interactions. Accurate artificial intelligence (AI)-based modeling of protein structures promises to provide accurate 3D models of PPIs at a proteome-wide scale.
Collapse
Affiliation(s)
- Jesse Durham
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jing Zhang
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ian R Humphreys
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Jimin Pei
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Qian Cong
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
42
|
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: 53] [Impact Index Per Article: 26.5] [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
|
43
|
Luo X, Tong F, Zhao W, Zheng X, Li J, Li J, Zhao D. BERT2DAb: a pre-trained model for antibody representation based on amino acid sequences and 2D-structure. MAbs 2023; 15:2285904. [PMID: 38010801 DOI: 10.1080/19420862.2023.2285904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 11/16/2023] [Indexed: 11/29/2023] Open
Abstract
Prior research has generated a vast amount of antibody sequences, which has allowed the pre-training of language models on amino acid sequences to improve the efficiency of antibody screening and optimization. However, compared to those for proteins, there are fewer pre-trained language models available for antibody sequences. Additionally, existing pre-trained models solely rely on embedding representations using amino acids or k-mers, which do not explicitly take into account the role of secondary structure features. Here, we present a new pre-trained model called BERT2DAb. This model incorporates secondary structure information based on self-attention to learn representations of antibody sequences. Our model achieves state-of-the-art performance on three downstream tasks, including two antigen-antibody binding classification tasks (precision: 85.15%/94.86%; recall:87.41%/86.15%) and one antigen-antibody complex mutation binding free energy prediction task (Pearson correlation coefficient: 0.77). Moreover, we propose a novel method to analyze the relationship between attention weights and contact states of pairs of subsequences in tertiary structures. This enhances the interpretability of BERT2DAb. Overall, our model demonstrates strong potential for improving antibody screening and design through downstream applications.
Collapse
Affiliation(s)
- Xiaowei Luo
- Information Center, Academy of Military Medical Sciences, Beijing, China
| | - Fan Tong
- Information Center, Academy of Military Medical Sciences, Beijing, China
| | - Wenbin Zhao
- Information Center, Academy of Military Medical Sciences, Beijing, China
| | - Xiangwen Zheng
- Information Center, Academy of Military Medical Sciences, Beijing, China
| | - Jiangyu Li
- Information Center, Academy of Military Medical Sciences, Beijing, China
| | - Jing Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Dongsheng Zhao
- Information Center, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
44
|
Miller NL, Clark T, Raman R, Sasisekharan R. Learned features of antibody-antigen binding affinity. Front Mol Biosci 2023; 10:1112738. [PMID: 36895805 PMCID: PMC9989197 DOI: 10.3389/fmolb.2023.1112738] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/18/2023] [Indexed: 02/23/2023] Open
Abstract
Defining predictors of antigen-binding affinity of antibodies is valuable for engineering therapeutic antibodies with high binding affinity to their targets. However, this task is challenging owing to the huge diversity in the conformations of the complementarity determining regions of antibodies and the mode of engagement between antibody and antigen. In this study, we used the structural antibody database (SAbDab) to identify features that can discriminate high- and low-binding affinity across a 5-log scale. First, we abstracted features based on previously learned representations of protein-protein interactions to derive 'complex' feature sets, which include energetic, statistical, network-based, and machine-learned features. Second, we contrasted these complex feature sets with additional 'simple' feature sets based on counts of contacts between antibody and antigen. By investigating the predictive potential of 700 features contained in the eight complex and simple feature sets, we observed that simple feature sets perform comparably to complex feature sets in classification of binding affinity. Moreover, combining features from all eight feature-sets provided the best classification performance (median cross-validation AUROC and F1-score of 0.72). Of note, classification performance is substantially improved when several sources of data leakage (e.g., homologous antibodies) are not removed from the dataset, emphasizing a potential pitfall in this task. We additionally observe a classification performance plateau across diverse featurization approaches, highlighting the need for additional affinity-labeled antibody-antigen structural data. The findings from our present study set the stage for future studies aimed at multiple-log enhancement of antibody affinity through feature-guided engineering.
Collapse
Affiliation(s)
- Nathaniel L Miller
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States.,Koch Institute of Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Thomas Clark
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States.,Koch Institute of Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Rahul Raman
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States.,Koch Institute of Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Ram Sasisekharan
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States.,Koch Institute of Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
45
|
Lou H, Zheng J, Fang X(L, Liang Z, Zhang M, Chen Y, Wang C, Cao X. Deep learning-based rapid generation of broadly reactive antibodies against SARS-CoV-2 and its Omicron variant. Cell Res 2023; 33:80-82. [PMID: 36167982 PMCID: PMC9514701 DOI: 10.1038/s41422-022-00727-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 09/06/2022] [Indexed: 01/06/2023] Open
Affiliation(s)
- Hantao Lou
- Frontier Research Center for Cell Response, Nankai-Oxford International Advanced Research Institute, College of Life Sciences, Nankai University, Tianjin, China. .,Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK.
| | - Jianqing Zheng
- grid.4991.50000 0004 1936 8948The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK ,grid.4991.50000 0004 1936 8948Big Data Institute, University of Oxford, Oxford, UK
| | - Xiaohang (Leo) Fang
- grid.4991.50000 0004 1936 8948Department of Engineering Science, University of Oxford, Oxford, UK
| | - Zhu Liang
- grid.4991.50000 0004 1936 8948Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK ,grid.4991.50000 0004 1936 8948Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Meihan Zhang
- grid.216938.70000 0000 9878 7032Frontier Research Center for Cell Response, Nankai-Oxford International Advanced Research Institute, College of Life Sciences, Nankai University, Tianjin, China
| | - Yu Chen
- grid.216938.70000 0000 9878 7032Frontier Research Center for Cell Response, Nankai-Oxford International Advanced Research Institute, College of Life Sciences, Nankai University, Tianjin, China
| | - Chunmei Wang
- grid.4991.50000 0004 1936 8948Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK ,grid.506261.60000 0001 0706 7839Department of Immunology, Centre for Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuetao Cao
- Frontier Research Center for Cell Response, Nankai-Oxford International Advanced Research Institute, College of Life Sciences, Nankai University, Tianjin, China. .,Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK. .,Department of Immunology, Centre for Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
46
|
Wang Z, Wang G, Lu H, Li H, Tang M, Tong A. Development of therapeutic antibodies for the treatment of diseases. MOLECULAR BIOMEDICINE 2022; 3:35. [PMID: 36418786 PMCID: PMC9684400 DOI: 10.1186/s43556-022-00100-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/24/2022] [Indexed: 11/25/2022] Open
Abstract
Since the first monoclonal antibody drug, muromonab-CD3, was approved for marketing in 1986, 165 antibody drugs have been approved or are under regulatory review worldwide. With the approval of new drugs for treating a wide range of diseases, including cancer and autoimmune and metabolic disorders, the therapeutic antibody drug market has experienced explosive growth. Monoclonal antibodies have been sought after by many biopharmaceutical companies and scientific research institutes due to their high specificity, strong targeting abilities, low toxicity, side effects, and high development success rate. The related industries and markets are growing rapidly, and therapeutic antibodies are one of the most important research and development areas in the field of biology and medicine. In recent years, great progress has been made in the key technologies and theoretical innovations provided by therapeutic antibodies, including antibody-drug conjugates, antibody-conjugated nuclides, bispecific antibodies, nanobodies, and other antibody analogs. Additionally, therapeutic antibodies can be combined with technologies used in other fields to create new cross-fields, such as chimeric antigen receptor T cells (CAR-T), CAR-natural killer cells (CAR-NK), and other cell therapy. This review summarizes the latest approved or in regulatory review therapeutic antibodies that have been approved or that are under regulatory review worldwide, as well as clinical research on these approaches and their development, and outlines antibody discovery strategies that have emerged during the development of therapeutic antibodies, such as hybridoma technology, phage display, preparation of fully human antibody from transgenic mice, single B-cell antibody technology, and artificial intelligence-assisted antibody discovery.
Collapse
Affiliation(s)
- Zeng Wang
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Guoqing Wang
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, China
| | - Huaqing Lu
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hongjian Li
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Mei Tang
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Aiping Tong
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
47
|
Evaluating the ability of the NLHA2 and artificial neural network models to predict COVID-19 severity, and comparing them with the four existing scoring systems. Microb Pathog 2022; 171:105735. [PMID: 36007846 PMCID: PMC9395227 DOI: 10.1016/j.micpath.2022.105735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/04/2022] [Accepted: 08/18/2022] [Indexed: 01/08/2023]
Abstract
To improve the identification and subsequent intervention of COVID-19 patients at risk for ICU admission, we constructed COVID-19 severity prediction models using logistic regression and artificial neural network (ANN) analysis and compared them with the four existing scoring systems (PSI, CURB-65, SMARTCOP, and MuLBSTA). In this prospective multi-center study, 296 patients with COVID-19 pneumonia were enrolled and split into the General-Ward-Care group (N = 238) and the ICU-Admission group (N = 58). The PSI model (AUC = 0.861) had the best results among the existing four scoring systems, followed by SMARTCOP (AUC = 0.770), motified-MuLBSTA (AUC = 0.761), and CURB-65 (AUC = 0.712). Data from 197 patients (training set) were analyzed for modeling. The beta coefficients from logistic regression were used to develop a severity prediction model and risk score calculator. The final model (NLHA2) included five covariates (consumes alcohol, neutrophil count, lymphocyte count, hemoglobin, and AKP). The NLHA2 model (training: AUC = 0.959; testing: AUC = 0.857) had similar results to the PSI model, but with fewer variable items. ANN analysis was used to build another complex model, which had higher accuracy (training: AUC = 1.000; testing: AUC = 0.907). Discrimination and calibration were further verified through bootstrapping (2000 replicates), Hosmer-Lemeshow goodness of fit testing, and Brier score calculation. In conclusion, the PSI model is the best existing system for predicting ICU admission among COVID-19 patients, while two newly-designed models (NLHA2 and ANN) performed better than PSI, and will provide a new approach for the development of prognostic evaluation system in a novel respiratory viral epidemic.
Collapse
|
48
|
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
|
49
|
Ackermann K, Wort JL, Bode BE. Pulse dipolar EPR for determining nanomolar binding affinities. Chem Commun (Camb) 2022; 58:8790-8793. [PMID: 35837993 PMCID: PMC9350988 DOI: 10.1039/d2cc02360a] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Protein interaction studies often require very low concentrations and highly sensitive biophysical methods. Here, we demonstrate that pulse dipolar electron paramagnetic resonance spectroscopy allows measuring dissociation constants in the nanomolar range. This approach is appealing for concentration-limited biomolecular systems and medium-to-high-affinity binding studies, demonstrated here at 50 nanomolar protein concentration. CuII-nitroxide RIDME measurements at 100 nM protein concentration allow reliable extraction of dissociation constants and distances, while measurements at 50 nM protein concentration allow reliable extraction of dissociation constants only.![]()
Collapse
Affiliation(s)
- Katrin Ackermann
- EaStCHEM School of Chemistry, Biomedical Sciences Research Complex and Centre of Magnetic resonance, University of St Andrews, North Haugh, St Andrews, KY16 9ST, Scotland, UK.
| | - Joshua L Wort
- EaStCHEM School of Chemistry, Biomedical Sciences Research Complex and Centre of Magnetic resonance, University of St Andrews, North Haugh, St Andrews, KY16 9ST, Scotland, UK.
| | - Bela E Bode
- EaStCHEM School of Chemistry, Biomedical Sciences Research Complex and Centre of Magnetic resonance, University of St Andrews, North Haugh, St Andrews, KY16 9ST, Scotland, UK.
| |
Collapse
|
50
|
Zhang Y, Tan W, Lou Z, Huang B, Zhou W, Zhao Y, Zhang J, Liang H, Li N, Zhu X, Ding L, Guo Y, He Z, He Y, Wang Z, Ma B, Ma M, Zhao S, Chang Z, Zhao X, Zheng X, Wu G, Wang H, Yang X. Immunogenicity Evaluating of the Multivalent COVID-19 Inactivated Vaccine against the SARS-CoV-2 Variants. Vaccines (Basel) 2022; 10:vaccines10060956. [PMID: 35746564 PMCID: PMC9228943 DOI: 10.3390/vaccines10060956] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 12/15/2022] Open
Abstract
It has been reported that the novel coronavirus (COVID-19) has caused more than 286 million cases and 5.4 million deaths to date. Several strategies have been implemented globally, such as social distancing and the development of the vaccines. Several severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants have appeared, such as Alpha, Beta, Gamma, Delta, and Omicron. With the rapid spread of the novel coronavirus and the rapidly changing mutants, the development of a broad-spectrum multivalent vaccine is considered to be the most effective way to defend against the constantly mutating virus. Here, we evaluated the immunogenicity of the multivalent COVID-19 inactivated vaccine. Mice were immunized by multivalent COVID-19 inactivated vaccine, and the neutralizing antibodies in serum were analyzed. The results show that HB02 + Delta + Omicron trivalent vaccine could provide broad spectrum protection against HB02, Beta, Delta, and Omicron virus. Additionally, the different multivalent COVID-19 inactivated vaccines could enhance cellular immunity. Together, our findings suggest that the multivalent COVID-19 inactivated vaccine can provide broad spectrum protection against HB02 and other virus variants in humoral and cellular immunity, providing new ideas for the development of a broad-spectrum COVID-19 vaccine.
Collapse
Affiliation(s)
- Yuntao Zhang
- China National Biotec Group Company Limited, Beijing 100024, China;
| | - Wenjie Tan
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China; (W.T.); (B.H.); (W.Z.)
| | - Zhiyong Lou
- MOE Key Laboratory of Protein Science & Collaborative Innovation Center of Biotherapy, School of Medicine, Tsinghua University, Beijing 100084, China;
| | - Baoying Huang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China; (W.T.); (B.H.); (W.Z.)
| | - Weimin Zhou
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China; (W.T.); (B.H.); (W.Z.)
| | - Yuxiu Zhao
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Jin Zhang
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Hongyang Liang
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Na Li
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Xiujuan Zhu
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Ling Ding
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Yancen Guo
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Zhenyu He
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Yao He
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Zhanhui Wang
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Bo Ma
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Meng Ma
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Suhua Zhao
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Zhen Chang
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Xue Zhao
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Xiaotong Zheng
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
| | - Guizhen Wu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China; (W.T.); (B.H.); (W.Z.)
- Correspondence: (G.W.); (H.W.); (X.Y.)
| | - Hui Wang
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Y.Z.); (J.Z.); (H.L.); (N.L.); (X.Z.); (L.D.); (Y.G.); (Z.H.); (Y.H.); (Z.W.); (B.M.); (M.M.); (S.Z.); (Z.C.); (X.Z.); (X.Z.)
- Correspondence: (G.W.); (H.W.); (X.Y.)
| | - Xiaoming Yang
- China National Biotec Group Company Limited, Beijing 100024, China;
- Correspondence: (G.W.); (H.W.); (X.Y.)
| |
Collapse
|