1
|
Li L, Lan Z, Qiao H, Meng X, Shi Z, Zhang W, Wang Y, Sun Z, Cui Q, Wang L, Zhou S, Hu F, Zhang D, Dai Y, Chen H, Geng Y. Design of NanoBiT-Nanobody-based FGL1 biosensors for early assisted diagnosis of esophageal cancer. Biomaterials 2025; 320:123286. [PMID: 40138964 DOI: 10.1016/j.biomaterials.2025.123286] [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: 12/09/2024] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
Esophageal cancer (EC) is one of the most common causes of cancer-related mortality due in part to challenges in early diagnosis. Biomarker identification is crucial for improved early screening and treatment strategies for patients. Firstly, we employed serum proteomics techniques to screen for potential biomarkers in 15 early-stage EC patients and 5 healthy individuals. Among the differentially expressed proteins, FGL1 emerged as a promising candidate (AUC = 0.974) for early detection of EC. Subsequently, we developed NanoBiT-conjugated dual nanobodies (NBNB) sensors for robust and quantitative signal detection in fetal bovine serum (FBS) in 30 min or less, with a limit of detection (LoD) of 11.38 pM. In a case-control study recruiting 96 EC patients and 99 control samples, testing serum samples with the developed NBNB sensors revealed significantly elevated serum level of FGL1 in all-stage EC patients (AUC = 0.7880) and early-stage EC patients (AUC = 0.8286). Additionally, the combined diagnostic performance of FGL1 and CEA in EC samples is notably enhanced (AUC = 0.8847). These findings propose FGL1 as a novel and promising target for the early-stage EC diagnosis and treatment selection. Furthermore, we applied the assay to patients across six types of cancer, suggesting FGL1 as a potential pan-cancer marker. This study introduces a rapid, easy-to-use, cost-effective, reliable, universal, and high-throughput alternative to meet the growing demand for cancer biomarker testing in both academic and clinical settings.
Collapse
Affiliation(s)
- Lingyun Li
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhongyun Lan
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Huarui Qiao
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiangjing Meng
- Shandong Provincial Key Laboratory of Biopharmaceuticals, Shandong Academy of Pharmaceutical Sciences, Jinan, 250101, China
| | - Ziyang Shi
- Queen Mary University of London Engineering School, Northwestern Polytechnical University, Xi'an, 710129, China
| | - Wanting Zhang
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yi'ang Wang
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zengchao Sun
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Qianqian Cui
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Lu Wang
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Siyu Zhou
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fangzheng Hu
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Daizhou Zhang
- Shandong Provincial Key Laboratory of Biopharmaceuticals, Shandong Academy of Pharmaceutical Sciences, Jinan, 250101, China.
| | - Yuanyuan Dai
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital of Chinese Academy of Medical Sciences Langfang Campus, Langfang, 065001, China.
| | - Hao Chen
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Yong Geng
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
2
|
Sun M, Cao D, Zhao M, Zhu X, Sun Y, Wang Q, Zhang H, Yang Y, Tian X, Wang H, Cai X, An T. Nanobody-based competitive enzyme-linked immunosorbent assay for detecting antibodies of porcine reproductive and respiratory syndrome virus. Int J Biol Macromol 2025; 312:144248. [PMID: 40379183 DOI: 10.1016/j.ijbiomac.2025.144248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 05/12/2025] [Accepted: 05/13/2025] [Indexed: 05/19/2025]
Abstract
The pandemic of porcine reproductive and respiratory syndrome (PRRS) continues a global threat to the swine industry and causes huge economic losses. Large-scale serological surveillance, particularly through enzyme-linked immunosorbent assay (ELISA), is an effective measure for PRRS control. Nanobodies have been used in diagnostic immunoassays due to their high affinity and specificity. In this study, two novel nanobodies against N protein of PRRSV were generated that exhibited ability to block the recognition of serum antibodies to N protein. A high-affinity nanobody was conjugated with horseradish peroxidase (HRP), then nanobody-based competitive ELISA (cELISA) was subsequently developed to detect antibodies against both genotypes 1 and 2 PRRSV. Notably, the developed cELISA exhibited enhanced sensitivity, specificity and reproducibility. The comparative analysis demonstrated a high agreement of 96.69 % between the cELISA and a commercial ELISA kit. Collectively, the developed nanobody-based cELISA can be used for epidemiological study of PRRSV infection and immune evaluations with advantages of simple operation and low costing.
Collapse
Affiliation(s)
- Mingxia Sun
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Dan Cao
- Soybean Research Institute, Heilongjiang Academy of Agricultural Science, Harbin 150086, China
| | - Man Zhao
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Xulong Zhu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Yue Sun
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Qian Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Hongliang Zhang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Yongbo Yang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Xiaoxiao Tian
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Haiwei Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Xuehui Cai
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| | - Tongqing An
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin 150069, China.
| |
Collapse
|
3
|
Hou J, Du K, Li J, Li Z, Cao S, Zhang S, Huang W, Liu H, Yang X, Sun S, Mo S, Qin T, Zhang X, Yin S, Nie X, Lu X. Research trends in the use of nanobodies for cancer therapy. J Control Release 2025; 381:113454. [PMID: 39922288 DOI: 10.1016/j.jconrel.2025.01.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 02/10/2025]
Abstract
Although there are many challenges in using nanobodies for treating various complex tumor diseases, including rapid renal clearance and the complex blood-brain barrier environment, nanobodies have shown great potential due to their high antigen affinity, excellent tumor penetration ability, and favorable safety profile. Since the discovery of the variable domain (VHH) of camelid heavy-chain antibodies in 1993, nanobodies have been progressively applied to various cancer therapy platforms, such as antagonistic drugs and targeting agents for effector domains. In recent years, several nanobody-based drugs, including Caplacizumab, KN-035, and Ozoralizumab, have been approved for clinical use. Among them, KN-035 is used for treating advanced solid tumors, and these advancements have propelled nanobody development to new heights. Currently, nanobodies are being rapidly applied to the treatment of a wide range of diseases, from viral infections to cancer, demonstrating strong advantages in areas such as targeted protein degradation, bioimaging, nanobody-drug conjugation, bispecific T-cell engagers, and vaccine applications. Bibliometric tools, including CiteSpace, HisCite Pro, and Alluvial Generator, were employed to trace the historical development of nanobodies in cancer research. The contributions of authors, countries, and institutions in this field were analyzed, and research hotspots and emerging trends were identified through keyword analysis and influential articles. Future trends were also predicted. This study provides a unique, comprehensive, and objective perspective on the use of nanobodies in tumor research, laying a foundation for future research directions and offering valuable insights for researchers in the field.
Collapse
Affiliation(s)
- Jun Hou
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Kejiang Du
- Department of Otorhinolaryngology-Head and Neck Surgery, Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China; Liuzhou People's Hospital affiliated to Guangxi Medical University, Liuzhou 545006, China
| | - Jinling Li
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Zhenghui Li
- Department of Neurosurgery, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450000, China
| | - Shaorui Cao
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Shilin Zhang
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Wenxing Huang
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Heng Liu
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Xiaomei Yang
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Shuyang Sun
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Shanzhao Mo
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Tianyu Qin
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Xilei Zhang
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Shihua Yin
- Department of Otorhinolaryngology-Head and Neck Surgery, Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China.
| | - Xinyu Nie
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230002, China.
| | - Xiaoling Lu
- College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China.
| |
Collapse
|
4
|
Flynn CD, Wu Z, Bantle A, Isaacson SE, Chang D, Mahmud A, Yousefi H, Das J, Kelley SO. Nanobody Receptors Enable High-Sensitivity Monitoring of IL-6 Using Molecular Pendulum Bioanalysis. Anal Chem 2025; 97:7825-7832. [PMID: 40173279 DOI: 10.1021/acs.analchem.4c06305] [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: 04/04/2025]
Abstract
The development of biomolecular sensing technologies with high sensitivity and specificity remains an important goal in modern analytical science. Molecular pendulum sensing has emerged as a new reagentless method capable of detecting a wide array of biomolecules directly in biological fluids. This sensing approach relies heavily on the modulation of hydrodynamic drag of molecular probes through solution, such that alterations in hydrodynamic diameter can transduce biomolecular interactions. Here, we explore the use of nanobodies as an alternative receptor in pendulum-based systems due to their small size and robust affinities. We compare the performance of nanobodies with that of aptamers and antibodies integrated into the molecular pendulum system by targeting the inflammatory indicator interleukin-6 (IL-6). Nanobody molecular pendulums demonstrate enhanced sensor response and sensitivity compared to those of the other receptors, enabling fine control over detection in the low physiological range of IL-6. In addition, we demonstrate the ability of nanobody sensors to function in complex biological matrices and at physiological temperature.
Collapse
Affiliation(s)
- Connor D Flynn
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada
- Department of Chemistry, Weinberg College of Arts & Sciences, Northwestern University, Evanston, Illinois 60208, United States
| | - Zhenwei Wu
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Amy Bantle
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3H6, Canada
| | - Scott E Isaacson
- Department of Chemistry, Weinberg College of Arts & Sciences, Northwestern University, Evanston, Illinois 60208, United States
| | - Dingran Chang
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Alam Mahmud
- Department of Chemistry, Weinberg College of Arts & Sciences, Northwestern University, Evanston, Illinois 60208, United States
| | - Hanie Yousefi
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Jagotamoy Das
- Department of Chemistry, Weinberg College of Arts & Sciences, Northwestern University, Evanston, Illinois 60208, United States
| | - Shana O Kelley
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada
- Department of Chemistry, Weinberg College of Arts & Sciences, Northwestern University, Evanston, Illinois 60208, United States
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3H6, Canada
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
- Department of Biochemistry, Feinberg School of Medicine, Northwestern University, Evanston, Illinois 60611, United States
- Simpson Querrey Institute, Northwestern University, Chicago, Illinois 60611, United States
- Chan Zuckerberg Biohub Chicago, Chicago, Illinois 60642, United States
| |
Collapse
|
5
|
Zeven K, Dierick H, Sevenois M, Baudhuin H, Navarro L, Van den Block S, Saliën J, Breckpot K, Bridoux J, De Groof TWM, Devoogdt N. Optimizing antibody PET imaging: a comparative preclinical analysis of nanobody and minibody-like PET tracers. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07205-2. [PMID: 40159541 DOI: 10.1007/s00259-025-07205-2] [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: 01/16/2025] [Accepted: 03/11/2025] [Indexed: 04/02/2025]
Abstract
PURPOSE Positron emission tomography remains the most important medical imaging technique for noninvasive imaging. Advances in radiotracer design, particularly those based on antibody(-formats), have broadened diagnostic applications. While minibodies and nanobodies stand out among antibody-type radiotracers due to their unique characteristics and pharmacokinetics, a direct comparison between these formats remains unexplored. In this study, we generated a so-called minabody, a minibody-like protein consisting of a nanobody fused to a CH3 domain, and compared it with its nanobody counterpart in vitro and in vivo upon labeling with various PET radionuclides. METHODS A previously developed human TIGIT-targeting nanobody was reformatted to a minabody format to allow head-to-head comparison of both formats. The minabody and nanobody formats were compared in vitro for binding, specificity and stability. The minabody and/or nanobody formats were labeled with copper-64, gallium-68 or fluor-18 for non-invasive imaging of human TIGIT-expressing tumors by PET/CT over time. RESULTS The minabody format showed higher binding affinities compared to the nanobody format. Despite lower binding affinity, the nanobody format outperformed the minabody format as PET imaging tracer showing higher specificity and allowing imaging of human TIGIT expression from 1 to 48 h post-injection upon labeling with copper-64. In addition, copper-64 showed superior tumor uptake and contrast for nanobody imaging compared to gallium-68 and fluor-18. CONCLUSION This study describes the first head-to-head comparison between the minibody and nanobody format for PET imaging. Our results underscore the importance of considering both targeting vector format and the radionuclide to achieve accurate and high-quality PET imaging.
Collapse
Affiliation(s)
- Katty Zeven
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel, Brussels, Belgium
| | - Herlinde Dierick
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Matthijs Sevenois
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | - Sonja Van den Block
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Jelena Saliën
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel, Brussels, Belgium
- Institut de Chimie Moléculaire de l'Université de Bourgogne (ICMUB), UMR CNRS 6302, Université de Bourgogne, Dijon, France
| | - Karine Breckpot
- Translational Oncology Research Center (TORC), Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jessica Bridoux
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel, Brussels, Belgium
| | - Timo W M De Groof
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel, Brussels, Belgium.
| | - Nick Devoogdt
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
6
|
Li W, Xu Z, He Q, Pan J, Zhang Y, El-Sheikh ESA, Hammock BD, Li D. Nanobody-Based Immunoassays for the Detection of Food Hazards-A Review. BIOSENSORS 2025; 15:183. [PMID: 40136980 PMCID: PMC11939871 DOI: 10.3390/bios15030183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025]
Abstract
Food safety remains a significant global challenge that affects human health. Various hazards, including microbiological and chemical threats, can compromise food safety throughout the supply chain. To address food safety issues and ensure public health, it is necessary to adopt rapid, accurate, and highly specific detection methods. Immunoassays are considered to be an effective method for the detection of highly sensitive biochemical indicators and provide an efficient platform for the identification of food hazards. In immunoassays, antibodies function as the primary recognition elements. Nanobodies have significant potential as valuable biomolecules in diagnostic applications. Their distinctive physicochemical and structural characteristics make them excellent candidates for the development of reliable diagnostic assays, and as promising alternatives to monoclonal and polyclonal antibodies. Herein, we summarize a comprehensive overview of the status and prospects of nanobody-based immunoassays in ensuring food safety. First, we begin with a historical perspective on the development of nanobodies and their unique characteristics. Subsequently, we explore the definitions and boundaries of immunoassays and immunosensors, before discussing the potential applications of nanobody-based immunoassays in food safety testing that have emerged over the past five years, and follow the different immunoassays, highlighting their advantages over traditional detection methods. Finally, the directions and challenges of nanobody-based immunoassays in food safety are discussed. Due to their remarkable sensitivity, specificity and versatility, nanobody-based immunoassays hold great promise in revolutionizing food safety testing and ensuring public health and well-being.
Collapse
Affiliation(s)
- Wenkai Li
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; (W.L.); (Z.X.); (Q.H.); (J.P.); (Y.Z.)
- Key Laboratory of Intelligent Equipment and Robotics for Agriculture of Zhejiang Province, Hangzhou 310058, China
| | - Zhihao Xu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; (W.L.); (Z.X.); (Q.H.); (J.P.); (Y.Z.)
- Key Laboratory of Intelligent Equipment and Robotics for Agriculture of Zhejiang Province, Hangzhou 310058, China
| | - Qiyi He
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; (W.L.); (Z.X.); (Q.H.); (J.P.); (Y.Z.)
- Key Laboratory of Intelligent Equipment and Robotics for Agriculture of Zhejiang Province, Hangzhou 310058, China
| | - Junkang Pan
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; (W.L.); (Z.X.); (Q.H.); (J.P.); (Y.Z.)
- Key Laboratory of Intelligent Equipment and Robotics for Agriculture of Zhejiang Province, Hangzhou 310058, China
| | - Yijia Zhang
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; (W.L.); (Z.X.); (Q.H.); (J.P.); (Y.Z.)
- Key Laboratory of Intelligent Equipment and Robotics for Agriculture of Zhejiang Province, Hangzhou 310058, China
| | | | - Bruce D. Hammock
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California Davis, Davis, CA 95616, USA
| | - Dongyang Li
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; (W.L.); (Z.X.); (Q.H.); (J.P.); (Y.Z.)
- Key Laboratory of Intelligent Equipment and Robotics for Agriculture of Zhejiang Province, Hangzhou 310058, China
| |
Collapse
|
7
|
El Salamouni NS, Cater JH, Spenkelink LM, Yu H. Nanobody engineering: computational modelling and design for biomedical and therapeutic applications. FEBS Open Bio 2025; 15:236-253. [PMID: 38898362 PMCID: PMC11788755 DOI: 10.1002/2211-5463.13850] [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: 04/05/2024] [Revised: 05/25/2024] [Accepted: 06/10/2024] [Indexed: 06/21/2024] Open
Abstract
Nanobodies, the smallest functional antibody fragment derived from camelid heavy-chain-only antibodies, have emerged as powerful tools for diverse biomedical applications. In this comprehensive review, we discuss the structural characteristics, functional properties, and computational approaches driving the design and optimisation of synthetic nanobodies. We explore their unique antigen-binding domains, highlighting the critical role of complementarity-determining regions in target recognition and specificity. This review further underscores the advantages of nanobodies over conventional antibodies from a biosynthesis perspective, including their small size, stability, and solubility, which make them ideal candidates for economical antigen capture in diagnostics, therapeutics, and biosensing. We discuss the recent advancements in computational methods for nanobody modelling, epitope prediction, and affinity maturation, shedding light on their intricate antigen-binding mechanisms and conformational dynamics. Finally, we examine a direct example of how computational design strategies were implemented for improving a nanobody-based immunosensor, known as a Quenchbody. Through combining experimental findings and computational insights, this review elucidates the transformative impact of nanobodies in biotechnology and biomedical research, offering a roadmap for future advancements and applications in healthcare and diagnostics.
Collapse
Affiliation(s)
- Nehad S. El Salamouni
- Molecular Horizons and School of Chemistry and Molecular BioscienceUniversity of WollongongAustralia
| | - Jordan H. Cater
- Molecular Horizons and School of Chemistry and Molecular BioscienceUniversity of WollongongAustralia
| | - Lisanne M. Spenkelink
- Molecular Horizons and School of Chemistry and Molecular BioscienceUniversity of WollongongAustralia
| | - Haibo Yu
- Molecular Horizons and School of Chemistry and Molecular BioscienceUniversity of WollongongAustralia
- ARC Centre of Excellence in Quantum BiotechnologyUniversity of WollongongAustralia
| |
Collapse
|
8
|
Krapež G, Šamec N, Zottel A, Katrašnik M, Kump A, Šribar J, Križaj I, Stojan J, Romih R, Bajc G, Butala M, Muyldermans S, Jovčevska I. In Vitro Functional Validation of an Anti-FREM2 Nanobody for Glioblastoma Cell Targeting. Antibodies (Basel) 2025; 14:8. [PMID: 39982223 PMCID: PMC11843905 DOI: 10.3390/antib14010008] [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: 12/11/2024] [Revised: 01/17/2025] [Accepted: 01/22/2025] [Indexed: 02/22/2025] Open
Abstract
Background/Objectives: Glioblastomas are the most common brain malignancies. Despite the implementation of multimodal therapy, patient life expectancy after diagnosis is barely 12 to 18 months. Glioblastomas are highly heterogeneous at the genetic and epigenetic level and comprise multiple different cell subpopulations. Therefore, small molecules such as nanobodies, able to target membrane proteins specific to glioblastoma cells or specific cell types within the tumor are being investigated as novel tools to treat glioblastomas. Methods: Here, we describe the identification of such a nanobody and its in silico and in vitro validation. NB3F18, as we named it, is directed against the membrane-associated protein FREM2, overexpressed in glioblastoma stem cells. Results: Three dimensional in silico modeling indicated that NB3F18 and FREM2 form a stable complex. Surface plasmon resonance confirmed their interaction with moderate affinity. As we demonstrated by flow cytometry, NB3F18 binds to glioblastoma stem cells to a greater extent than to differentiated glioblastoma cells and astrocytes. Immunocytochemistry revealed surface localization of NB3F18 on glioblastoma stem cells, whereas cytoplasmic localization of NB3F18 was observed in other cell lines. NB3F18 was detected by transmission electron microscopy on the plasma membrane and in various compartments of the endocytic pathway, from endocytic vesicles to multivesicular bodies (endosomes) and lysosomes. Interestingly, NB3F18 was cytotoxic to glioblastoma stem cells. Conclusions: Collectively, NB3F18 has been qualified as an interesting tool to target glioblastoma cells and as a potential vehicle to deliver biological or pharmaceutical agents to these cells.
Collapse
Affiliation(s)
- Gloria Krapež
- Center for Functional Genomics and Biochips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia; (G.K.); (N.Š.); (A.Z.); (M.K.)
| | - Neja Šamec
- Center for Functional Genomics and Biochips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia; (G.K.); (N.Š.); (A.Z.); (M.K.)
| | - Alja Zottel
- Center for Functional Genomics and Biochips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia; (G.K.); (N.Š.); (A.Z.); (M.K.)
| | - Mojca Katrašnik
- Center for Functional Genomics and Biochips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia; (G.K.); (N.Š.); (A.Z.); (M.K.)
| | - Ana Kump
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia; (A.K.); (I.K.)
- Jožef Stefan International Postgraduate School, Jamova 39, 1000 Ljubljana, Slovenia
| | - Jernej Šribar
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia; (A.K.); (I.K.)
| | - Igor Križaj
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia; (A.K.); (I.K.)
| | - Jurij Stojan
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia;
| | - Rok Romih
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia;
| | - Gregor Bajc
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000 Ljubljana, Slovenia; (G.B.); (M.B.)
| | - Matej Butala
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000 Ljubljana, Slovenia; (G.B.); (M.B.)
| | - Serge Muyldermans
- Cellular and Molecular Immunology, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Ivana Jovčevska
- Center for Functional Genomics and Biochips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia; (G.K.); (N.Š.); (A.Z.); (M.K.)
| |
Collapse
|
9
|
Markusson S, Raasakka A, Schröder M, Sograte‐Idrissi S, Rahimi AM, Asadpour O, Körner H, Lodygin D, Eichel‐Vogel MA, Chowdhury R, Sutinen A, Muruganandam G, Iyer M, Cooper MH, Weigel MK, Ambiel N, Werner HB, Zuchero JB, Opazo F, Kursula P. Nanobodies against the myelin enzyme CNPase as tools for structural and functional studies. J Neurochem 2025; 169:e16274. [PMID: 39655780 PMCID: PMC11629607 DOI: 10.1111/jnc.16274] [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: 05/27/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 12/13/2024]
Abstract
2',3'-Cyclic nucleotide 3'-phosphodiesterase (CNPase) is an abundant constituent of central nervous system non-compact myelin, and its loss in mice and humans causes neurodegeneration. Additionally, CNPase is frequently used as a marker antigen for myelinating cells. The catalytic activity of CNPase, the 3'-hydrolysis of 2',3'-cyclic nucleotides, is well characterised in vitro, but the in vivo function of CNPase remains unclear. CNPase interacts with the actin cytoskeleton to counteract the developmental closure of cytoplasmic channels that travel through compact myelin; its enzymatic activity may be involved in adenosine metabolism and RNA degradation. We developed a set of high-affinity nanobodies recognising the phosphodiesterase domain of CNPase, and the crystal structures of each complex show that the five nanobodies have distinct epitopes. One of the nanobodies bound deep into the CNPase active site and acted as an inhibitor. Moreover, the nanobodies were characterised in imaging applications and as intrabodies, expressed in mammalian cells, such as primary oligodendrocytes. Fluorescently labelled nanobodies functioned in imaging of teased nerve fibres and whole brain tissue sections, as well as super-resolution microscopy. These anti-CNPase nanobodies provide new tools for structural and functional studies on myelin formation, dynamics, and disease, including high-resolution imaging of nerve tissue.
Collapse
Affiliation(s)
| | - Arne Raasakka
- Department of BiomedicineUniversity of BergenBergenNorway
- Neurosurgery DepartmentStanford University School of MedicineStanfordCaliforniaUSA
| | | | - Shama Sograte‐Idrissi
- Center for Biostructural Imaging of Neurodegeneration (BIN)University of Göttingen Medical CenterGöttingenGermany
| | - Amir Mohammad Rahimi
- Center for Biostructural Imaging of Neurodegeneration (BIN)University of Göttingen Medical CenterGöttingenGermany
| | - Ommolbanin Asadpour
- Center for Biostructural Imaging of Neurodegeneration (BIN)University of Göttingen Medical CenterGöttingenGermany
| | - Henrike Körner
- Department for Neuroimmunology and Multiple Sclerosis ResearchUniversity of Göttingen Medical CenterGöttingenGermany
| | - Dmitri Lodygin
- Department for Neuroimmunology and Multiple Sclerosis ResearchUniversity of Göttingen Medical CenterGöttingenGermany
| | - Maria A. Eichel‐Vogel
- Department of NeurogeneticsMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | | | - Aleksi Sutinen
- Faculty of Biochemistry and Molecular Medicine & Biocenter OuluUniversity of OuluOuluFinland
| | - Gopinath Muruganandam
- VIB‐VUB Center for Structural BiologyVlaams Instituut voor BiotechnologieBrusselsBelgium
- Department of Bioengineering Sciences, Structural Biology BrusselsVrije Universiteit BrusselBrusselsBelgium
| | - Manasi Iyer
- Neurosurgery DepartmentStanford University School of MedicineStanfordCaliforniaUSA
| | - Madeline H. Cooper
- Neurosurgery DepartmentStanford University School of MedicineStanfordCaliforniaUSA
| | - Maya K. Weigel
- Neurosurgery DepartmentStanford University School of MedicineStanfordCaliforniaUSA
| | - Nicholas Ambiel
- Neurosurgery DepartmentStanford University School of MedicineStanfordCaliforniaUSA
| | - Hauke B. Werner
- Department of NeurogeneticsMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - J. Bradley Zuchero
- Neurosurgery DepartmentStanford University School of MedicineStanfordCaliforniaUSA
| | - Felipe Opazo
- Center for Biostructural Imaging of Neurodegeneration (BIN)University of Göttingen Medical CenterGöttingenGermany
- Institute of Neuro‐ and Sensory PhysiologyUniversity Medical Center GöttingenGöttingenGermany
- NanoTag Biotechnologies GmbHGöttingenGermany
| | - Petri Kursula
- Department of BiomedicineUniversity of BergenBergenNorway
- Faculty of Biochemistry and Molecular Medicine & Biocenter OuluUniversity of OuluOuluFinland
| |
Collapse
|
10
|
Lauwers Y, De Groof TWM, Vincke C, Van Craenenbroeck J, Jumapili NA, Barthelmess RM, Courtoy G, Waelput W, De Pauw T, Raes G, Devoogdt N, Van Ginderachter JA. Imaging of tumor-associated macrophage dynamics during immunotherapy using a CD163-specific nanobody-based immunotracer. Proc Natl Acad Sci U S A 2024; 121:e2409668121. [PMID: 39693339 DOI: 10.1073/pnas.2409668121] [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: 05/26/2024] [Accepted: 11/13/2024] [Indexed: 12/20/2024] Open
Abstract
Immunotherapies have emerged as an effective treatment option for immune-related diseases, such as cancer and inflammatory diseases. However, variations in patient responsiveness limit the broad applicability and success of these immunotherapies. Noninvasive whole-body imaging of the immune status of individual patients during immunotherapy could enable the prediction and monitoring of the patient's response, resulting in more personalized treatments. In this study, we developed a nanobody-based immunotracer targeting CD163, a receptor specifically expressed on macrophages. This anti-CD163 immunotracer bound to human and mouse CD163 with high affinity and specificity without competing for ligand binding. Furthermore, the tracer showed no unwanted immune cell activation and was nonimmunogenic. Upon radiolabeling of the anti-CD163 immunotracer, specific imaging of CD163+ macrophages using micro-single-photon emission computerized tomography/computed tomography or micro-positron emission tomography/CT was performed. The anti-CD163 immunotracer was able to stratify immunotherapy responders from nonresponders (NR) by visualizing differences in the intratumoral CD163+ TAM distribution in Lewis lung carcinoma-ovalbumin tumor-bearing mice receiving an anti-programmed cell death protein-1 (PD-1)/CSF1R combination treatment. Immunotherapy-responding mice showed a more homogeneous distribution of the PET signal in the middle of the tumor, while CD163+ TAMs were located at the tumor periphery in NR. As such, visualization of CD163+ TAM distribution in the tumor microenvironment could allow a prediction or follow-up of therapy response. Altogether, this study describes an immunotracer, specific for CD163+ macrophages, that allows same-day imaging and follow-up of these immune cells in the tumor microenvironment, providing a good basis for the prediction and follow-up of immunotherapy responses in cancer patients.
Collapse
Affiliation(s)
- Yoline Lauwers
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Timo W M De Groof
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Cécile Vincke
- Laboratory of Myeloid Cell Immunology, Vlaams Instituut voor Biotechnologie Center for Inflammation Research, Brussels 1050, Belgium
- Laboratory of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels 1050, Belgium
| | - Jolien Van Craenenbroeck
- Laboratory of Myeloid Cell Immunology, Vlaams Instituut voor Biotechnologie Center for Inflammation Research, Brussels 1050, Belgium
- Laboratory of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels 1050, Belgium
| | - Neema Ahishakiye Jumapili
- Laboratory of Myeloid Cell Immunology, Vlaams Instituut voor Biotechnologie Center for Inflammation Research, Brussels 1050, Belgium
- Laboratory of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels 1050, Belgium
| | - Romina Mora Barthelmess
- Laboratory of Myeloid Cell Immunology, Vlaams Instituut voor Biotechnologie Center for Inflammation Research, Brussels 1050, Belgium
- Laboratory of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels 1050, Belgium
| | - Guillaume Courtoy
- Department of Pathology, Universitair Ziekenhuis Brussel, Brussels B-1090, Belgium
- Laboratory of Experimental Pathology, Supporting Clinical Sciences, Vrije Universiteit Brussel, Brussels B-1090, Belgium
| | - Wim Waelput
- Department of Pathology, Universitair Ziekenhuis Brussel, Brussels B-1090, Belgium
- Laboratory of Experimental Pathology, Supporting Clinical Sciences, Vrije Universiteit Brussel, Brussels B-1090, Belgium
| | - Tessa De Pauw
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Geert Raes
- Laboratory of Myeloid Cell Immunology, Vlaams Instituut voor Biotechnologie Center for Inflammation Research, Brussels 1050, Belgium
- Laboratory of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels 1050, Belgium
| | - Nick Devoogdt
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Jo A Van Ginderachter
- Laboratory of Myeloid Cell Immunology, Vlaams Instituut voor Biotechnologie Center for Inflammation Research, Brussels 1050, Belgium
- Laboratory of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels 1050, Belgium
| |
Collapse
|
11
|
Meeus F, Funeh CN, Awad RM, Zeven K, Autaers D, De Becker A, Van Riet I, Goyvaerts C, Tuyaerts S, Neyns B, Devoogdt N, De Vlaeminck Y, Breckpot K. Preclinical evaluation of antigen-sensitive B7-H3-targeting nanobody-based CAR-T cells in glioblastoma cautions for on-target, off-tumor toxicity. J Immunother Cancer 2024; 12:e009110. [PMID: 39562005 PMCID: PMC11575280 DOI: 10.1136/jitc-2024-009110] [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: 02/21/2024] [Accepted: 10/27/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Glioblastoma is the most common lethal primary brain tumor, urging evaluation of new treatment options. Chimeric antigen receptor (CAR)-T cells targeting B7 homolog 3 (B7-H3) are promising because of the overexpression of B7-H3 on glioblastoma cells but not on healthy brain tissue. Nanobody-based (nano)CARs are gaining increasing attention as promising alternatives to classical single-chain variable fragment-based (scFv)CARs, because of their single-domain nature and low immunogenicity. Still, B7-H3 nanoCAR-T cells have not been extensively studied in glioblastoma. METHODS B7-H3 nanoCAR- and scFvCAR-T cells were developed and evaluated in human glioblastoma models. NanoCAR-T cells targeting an irrelevant antigen served as control. T cell activation, cytokine secretion and killing capacity were evaluated in vitro using ELISA, live cell imaging and flow cytometry. Antigen-specific killing was assessed by generating B7-H3 knock-out cells using Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9-genome editing. The tumor tracing capacity of the B7-H3 nanobody was first evaluated in vivo using nuclear imaging. Then, the therapeutic potential of the nanoCAR-T cells was evaluated in a xenograft glioblastoma model. RESULTS We showed that B7-H3 nanoCAR-T cells were most efficient in lysing B7-H3pos glioblastoma cells in vitro. Lack of glioblastoma killing by control nanoCAR-T cells and lack of B7-H3neg glioblastoma killing by B7-H3 nanoCAR-T cells showed antigen-specificity. We showed in vivo tumor targeting capacity of the B7-H3 nanobody-used for the nanoCAR design-in nuclear imaging experiments. Evaluation of the nanoCAR-T cells in vivo showed tumor control in mice treated with B7-H3 nanoCAR-T cells in contrast to progressive disease in mice treated with control nanoCAR-T cells. However, we observed limiting toxicity in mice treated with B7-H3 nanoCAR-T cells and showed that the B7-H3 nanoCAR-T cells are activated even by low levels of mouse B7-H3 expression. CONCLUSIONS B7-H3 nanoCAR-T cells showed promise for glioblastoma therapy following in vitro characterization, but limiting in vivo toxicity was observed. Off-tumor recognition of healthy mouse tissue by the cross-reactive B7-H3 nanoCAR-T cells was identified as a potential cause for this toxicity, warranting caution when using highly sensitive nanoCAR-T cells, recognizing the low-level expression of B7-H3 on healthy tissue.
Collapse
Affiliation(s)
- Fien Meeus
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Laboratory for Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel, Brussels, Belgium
- Department of Medical Imaging (MIMA), Molecular Imaging and Therapy (MITH) research group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Cyprine Neba Funeh
- Department of Medical Imaging (MIMA), Molecular Imaging and Therapy (MITH) research group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Robin Maximilian Awad
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Laboratory for Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel, Brussels, Belgium
| | - Katty Zeven
- Department of Medical Imaging (MIMA), Molecular Imaging and Therapy (MITH) research group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Dorien Autaers
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Laboratory for Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel, Brussels, Belgium
| | - Ann De Becker
- Department of Hematology, Cellular Therapy Laboratory, University Hospital Brussels, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Ivan Van Riet
- Department of Hematology, Cellular Therapy Laboratory, University Hospital Brussels, Universitair Ziekenhuis Brussel, Brussels, Belgium
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Hematology and Immunology Research Team (HEIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Cleo Goyvaerts
- Department of Medical Imaging (MIMA), Molecular Imaging and Therapy (MITH) research group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sandra Tuyaerts
- Department of Medical Oncology, University Hospital Brussels, Universitair Ziekenhuis Brussel, Brussels, Belgium
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Laboratory for Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel, Brussels, Belgium
| | - Bart Neyns
- Department of Medical Oncology, University Hospital Brussels, Universitair Ziekenhuis Brussel, Brussels, Belgium
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Laboratory for Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel, Brussels, Belgium
| | - Nick Devoogdt
- Department of Medical Imaging (MIMA), Molecular Imaging and Therapy (MITH) research group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yannick De Vlaeminck
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Laboratory for Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine Breckpot
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Laboratory for Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
12
|
Robinson SR, Dayao DA, Medina JA, Martone CJ, Yauch AK, Hinkley T, Erasmus JH, Shoemaker CB, Tzipori S. An anti-Shiga toxin VHH nanobody multimer protects mice against fatal toxicosis when administered intramuscularly as repRNA. Infect Immun 2024; 92:e0023924. [PMID: 39392311 PMCID: PMC11556087 DOI: 10.1128/iai.00239-24] [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: 06/06/2024] [Accepted: 09/21/2024] [Indexed: 10/12/2024] Open
Abstract
Hemolytic uremic syndrome (HUS) is a systemic sequelae from gastrointestinal infection with Shiga toxin (Stx) producing Escherichia coli (STEC) that can result in acute kidney injury, lasting renal disease, and death. Despite a window for intervention between hemorrhagic diarrhea and onset of HUS, no specific therapies exist to prevent or treat HUS following STEC infection. Furthermore, there is no way to predict which patients with STEC will develop HUS or any rapid way to determine which Stx variant is present. To address this, we have broadened the therpay to neutralize additional toxin variants. It contains a multimer of nanobodies derived from camelid heavy chain antibody fragments (VHHs). An improved VHH-based neutralizing agent (VNA2) is delivered intramuscularly as RNA combined with LION nanoparticles rather than mRNA, that replicates on administration (repRNA), resulting in a rapidly circulating VNA that can bind systemic toxin. The RNA/VNA2-Stx administered intramuscularly prevents toxicity and death in a mouse model of acute Stx toxicity.
Collapse
Affiliation(s)
- Sally R. Robinson
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
| | - Denise Ann Dayao
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
| | - Jhon A. Medina
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
| | - Cara J. Martone
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
| | - Anne K. Yauch
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
| | | | | | - Charles B. Shoemaker
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
| | - Saul Tzipori
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
| |
Collapse
|
13
|
Zhang L, Li X, He Q, Chen M, Zhou M, Guo J, Li Y, Tu Z. Elastin-like polypeptide-functionalized nanobody for column-free immunoaffinity purification of aflatoxin B 1. Anal Bioanal Chem 2024; 416:6199-6208. [PMID: 39264463 DOI: 10.1007/s00216-024-05498-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/29/2024] [Accepted: 08/15/2024] [Indexed: 09/13/2024]
Abstract
A column-free immunoaffinity purification (CFIP) technique for sample preparation of aflatoxin B1 (AFB1) was developed using an AFB1-specific nanobody (named G8) and an elastin-like polypeptide (ELP). The reversible phase transition between liquid and solid in response to temperature changes was exhibited by the ELP which was derived from human elastin. The G8 was tagged with ELPs of various lengths (20, 40, 60, and 80 repeat units) at the C-terminus using recursive directional ligation (RDL). Coding sequences were then subcloned into pET30a at the multiple cloning sites. Bioactive recombinant proteins were produced by expressing them as inclusion bodies in Escherichia coli BL21 (DE3), then dissolved and refolded. Analysis by indirect competitive enzyme-linked immunosorbent assay (icELISA) and transition temperature (Tt) measurement confirmed that the refolded G8-ELPs preserved the ability to recognize AFB1 as well as phase transition when the temperature rose above Tt. To establish the optimal conditions for cleaning AFB1, the effects of various parameters on recovery were investigated. The recovery in ELISA tests was 95 ± 3.67% under the optimized CFIP workflow. Furthermore, the CFIP-prepared samples were applied for high-performance liquid chromatography (HPLC) detection. The recovery in the CFIP-HPLC test ranged from 54 ± 1.86% to 98 ± 3.58% for maize, rice, soy sauce, and vegetable oil samples. To the best of our knowledge, this is the first report combining the function of both nanobody and ELP to develop a cleanup technique for small molecules in a complex matrix. The CFIP for the sample pretreatment was easy to use and inexpensive. In contrast to conventional immunosensitivity materials, the reagent utilized in the CFIP was entirely biosynthesized without any chemical coupling reactions. This suggests that the nanobody-ELP may serve as a useful dual-functional reagent for the development of sample cleaning or purification methods.
Collapse
Affiliation(s)
- Leping Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
- College of Food Science, Nanchang University, Nanchang, 330031, China
- Jiangxi Province Key Laboratory of Modern Analytical Sciences, Nanchang University, Nanchang, 330031, China
| | - Xiaojiang Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
- College of Food Science, Nanchang University, Nanchang, 330031, China
- Jiangxi Province Key Laboratory of Modern Analytical Sciences, Nanchang University, Nanchang, 330031, China
| | - Qinghua He
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
- Jiangxi-OAI Joint Research Institution, Nanchang University, Nanchang, 330047, China
- Jiangxi Province Key Laboratory of Modern Analytical Sciences, Nanchang University, Nanchang, 330031, China
| | - Mengna Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
- College of Food Science, Nanchang University, Nanchang, 330031, China
- Jiangxi Province Key Laboratory of Modern Analytical Sciences, Nanchang University, Nanchang, 330031, China
| | - Mengmeng Zhou
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
- College of Food Science, Nanchang University, Nanchang, 330031, China
- Jiangxi Province Key Laboratory of Modern Analytical Sciences, Nanchang University, Nanchang, 330031, China
| | - Jiebiao Guo
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Shaoguan University, Shaoguan, 512005, China
| | - Yanping Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China.
- Jiangxi-OAI Joint Research Institution, Nanchang University, Nanchang, 330047, China.
| | - Zhui Tu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China.
- Jiangxi Province Key Laboratory of Modern Analytical Sciences, Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
14
|
Guo Y, Wang R, Wang Y, Zheng F, Chen J, Lyu Z, Yuan C, Liu L, Jiang X. Screening and anti-angiogenesis activity of Chiloscyllium plagiosum anti-human VEGFR2 single-domain antibody. Clin Exp Immunol 2024; 218:177-187. [PMID: 39028612 PMCID: PMC11482501 DOI: 10.1093/cei/uxae060] [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: 01/26/2024] [Revised: 05/25/2024] [Accepted: 07/18/2024] [Indexed: 07/21/2024] Open
Abstract
Recently, the incidence of malignant tumors is on the rise and searching for new treatments on it has become the research priority. Blocking the vascular endothelial growth factor (VEGF) and its receptor (VEGFR) is one of the treatment strategies that used in the development of specific anti-angiogenic drugs. The deficiencies in tissue penetration and affinity maturation become the weakness of these drugs in anti-tumors applications. The single heavy chain antibody found in Chiloscyllium plagiosum, which has a low molecular weight and superior tissue penetration of variable region (variable new antigen receptor, VNARs), was considered to have the high antigen-binding activity and stability. This type of antibody has a simple structure that can be prokaryoticaly expressed, which makes it easily to produce new antiangiogenic target drugs. Specific anti-IgNAR rabbit multiple antibodies have been used to assess the level of VNARs in sharks and have shown a significant enrichment of IgNAR after triple immunization. An anti-VEGFR2 phage library was used for the targeted VNARs screening, and five candidate VNARs sequences were subsequently obtained by phage screening, followed by combined screening with the transcriptome library, and analysis of conserved regions along with 3D modelling matched the VNAR profile. ELISA and cell-based assays showed that two of the VNARs, VNAR-A6, and VNAR-E3, had a superior antigen affinity and anti-angiogenic activity thereby being able to inhibit human Umbilical Vein Endothelial Cells proliferation and migration. The anti-VEGFR2 VNARs derived from the immunized C. plagiosum and screened by phage library, which provide the new research ideas and specific approaches for the development of new drugs. The anti-VEGFR2 VNARs are capable for blocking the VEGF-VEGFR pathway, which of these may contribute to expanding the use of anti-angiogenic drugs.
Collapse
Affiliation(s)
- Yanwen Guo
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Ruiqi Wang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yun Wang
- Tianxiang East Hospital, Yiwu, China
| | | | - Jianqing Chen
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Zhengbing Lyu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Chen Yuan
- The First People’s Hospital of Huzhou, Huzhou, China
| | - Lili Liu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiaofeng Jiang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
- Jiangsu Baiying Biotech C., Ltd, Taizhou, China
| |
Collapse
|
15
|
Hao Z, Dong X, Zhang Z, Qin Z. A Nanobody of PEDV S1 Protein: Screening and Expression in Escherichia coli. Biomolecules 2024; 14:1116. [PMID: 39334881 PMCID: PMC11430113 DOI: 10.3390/biom14091116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) has caused significant economic losses to the pig farming industry in various countries for a long time. Currently, there are no highly effective preventive or control measures available. Research into the pathogenic mechanism of PEDV has shown that it primarily causes infection by binding the S protein to the CD13 (APN) receptor on the membrane of porcine intestinal epithelial cells. The S1 region contains three neutralization epitopes and multiple receptor-binding domains, which are closely related to viral antigenicity and ad-sorption invasion. Nanobodies are a type of single-domain antibody that have been discovered in recent years. They can be expressed on a large scale through prokaryotic expression systems, which makes them cost-effective, stable, and less immunogenic. This study used a phage display library of nanobodies against the PEDV S1 protein. After three rounds of selection and enrichment, the DNA sequence of the highly specific nanobody S1Nb1 was successfully obtained. To obtain soluble nanobody S1Nb1, its DNA sequence was inserted into the vector Pcold and a solubility-enhancing SUMO tag was added. The resulting recombinant vector, Pcold-SUMO-S1Nb1, was then transformed into E. coli BL21(DE3) to determine the optimal expression conditions for the nanobody. Following purification using Ni-column affinity chromatography, Western blot analysis confirmed the successful purification of S1Nb1 carrying the solubility-enhancing tag. ELISA results demonstrated a strong affinity between the S1Nb1 nanobody and PEDV S1 protein.
Collapse
Affiliation(s)
| | | | | | - Zhihua Qin
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, China; (Z.H.); (X.D.); (Z.Z.)
| |
Collapse
|
16
|
Zeven K, Lauwers Y, De Mey L, Debacker JM, De Pauw T, De Groof TWM, Devoogdt N. Advancements in nuclear imaging using radiolabeled nanobody tracers to support cancer immunotherapy. IMMUNOTHERAPY ADVANCES 2024; 4:ltae006. [PMID: 39281708 PMCID: PMC11402390 DOI: 10.1093/immadv/ltae006] [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: 06/27/2024] [Accepted: 08/23/2024] [Indexed: 09/18/2024] Open
Abstract
The evolving landscape of cancer immunotherapy has revolutionized cancer treatment. However, the dynamic tumor microenvironment has led to variable clinical outcomes, indicating a need for predictive biomarkers. Noninvasive nuclear imaging, using radiolabeled modalities, has aided in patient selection and monitoring of their treatment response. This approach holds promise for improving diagnostic accuracy, providing a more personalized treatment regimen, and enhancing the clinical response. Nanobodies or single-domain antibodies, derived from camelid heavy-chain antibodies, allow early timepoint detection of targets with high target-to-background ratios. To date, a plethora of nanobodies have been developed for nuclear imaging of tumor-specific antigens, immune checkpoints, and immune cells, both at a preclinical and clinical level. This review comprehensively outlines the recent advancements in nanobody-based nuclear imaging, both on preclinical and clinical levels. Additionally, the impact and expected future advancements on the use of nanobody-based radiopharmaceuticals in supporting cancer diagnosis and treatment follow-up are discussed.
Collapse
Affiliation(s)
- Katty Zeven
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Yoline Lauwers
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Lynn De Mey
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Jens M Debacker
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Tessa De Pauw
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Timo W M De Groof
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Nick Devoogdt
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|
17
|
Wang X, Sheng Y, Ji P, Deng Y, Sun Y, Chen Y, Nan Y, Hiscox JA, Zhou EM, Liu B, Zhao Q. A Broad-specificity Neutralizing Nanobody against Hepatitis E Virus Capsid Protein. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:442-455. [PMID: 38905108 PMCID: PMC11299488 DOI: 10.4049/jimmunol.2300706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 06/03/2024] [Indexed: 06/23/2024]
Abstract
Hepatitis E virus (HEV) is a worldwide zoonotic and public health concern. The study of HEV biology is helpful for designing viral vaccines and drugs. Nanobodies have recently been considered appealing materials for viral biological research. In this study, a Bactrian camel was immunized with capsid proteins from different genotypes (1, 3, 4, and avian) of HEV. Then, a phage library (6.3 × 108 individual clones) was constructed using peripheral blood lymphocytes from the immunized camel, and 12 nanobodies against the truncated capsid protein of genotype 3 HEV (g3-p239) were screened. g3-p239-Nb55 can cross-react with different genotypes of HEV and block Kernow-C1/P6 HEV from infecting HepG2/C3A cells. To our knowledge, the epitope recognized by g3-p239-Nb55 was determined to be a novel conformational epitope located on the surface of viral particles and highly conserved among different mammalian HEV isolates. Next, to increase the affinity and half-life of the nanobody, it was displayed on the surface of ferritin, which can self-assemble into a 24-subunit nanocage, namely, fenobody-55. The affinities of fenobody-55 to g3-p239 were ∼20 times greater than those of g3-p239-Nb55. In addition, the half-life of fenobody-55 was nine times greater than that of g3-p239-Nb55. G3-p239-Nb55 and fenobody-55 can block p239 attachment and Kernow-C1/P6 infection of HepG2/C3A cells. Fenobody-55 can completely neutralize HEV infection in rabbits when it is preincubated with nonenveloped HEV particles. Our study reported a case in which a nanobody neutralized HEV infection by preincubation, identified a (to our knowledge) novel and conserved conformational epitope of HEV, and provided new material for researching HEV biology.
Collapse
Affiliation(s)
- Xueting Wang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Department of Veterinary Medicine, Shandong Vocational Animal Science and Veterinary College, Weifang, Shandong, China
| | - Yamin Sheng
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Pinpin Ji
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yingying Deng
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yani Sun
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yiyang Chen
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yuchen Nan
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Julian A. Hiscox
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - En-Min Zhou
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Baoyuan Liu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Qin Zhao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
18
|
Zhang T, Yang D, Tang L, Hu Y. Current development of severe acute respiratory syndrome coronavirus 2 neutralizing antibodies (Review). Mol Med Rep 2024; 30:148. [PMID: 38940338 PMCID: PMC11228696 DOI: 10.3892/mmr.2024.13272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/21/2024] [Indexed: 06/29/2024] Open
Abstract
The coronavirus disease 2019 pandemic due to severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2) seriously affected global public health security. Studies on vaccines, neutralizing antibodies (NAbs) and small molecule antiviral drugs are currently ongoing. In particular, NAbs have emerged as promising therapeutic agents due to their well‑defined mechanism, high specificity, superior safety profile, ease of large‑scale production and simultaneous application for both prevention and treatment of viral infection. Numerous NAb therapeutics have entered the clinical research stages, demonstrating promising therapeutic and preventive effects. These agents have been used for outbreak prevention and control under urgent authorization processes. The present review summarizes the molecular targets of SARS‑CoV‑2‑associated NAbs and screening and identification techniques for NAb development. Moreover, the current shortcomings and challenges that persist with the use of NAbs are discussed. The aim of the present review is to offer a reference for the development of NAbs for any future emergent infectious diseases, including SARS‑CoV‑2.
Collapse
Affiliation(s)
- Tong Zhang
- Department of Hematology, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Di Yang
- Department of Hematology, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Liang Tang
- Department of Hematology, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yu Hu
- Department of Hematology, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
19
|
Barbieri ES, Sosa-Holt C, Ibañez LI, Baztarrica J, Garaicoechea L, Gay CL, Caceres CJ, Aduriz M, Baumeister E, Escribano JA, Perez D, Wigdorovitz A, Parreño GV, Puntel M. Anti-hemagglutinin monomeric nanobody provides prophylactic immunity against H1 subtype influenza A viruses. PLoS One 2024; 19:e0301664. [PMID: 38985719 PMCID: PMC11236207 DOI: 10.1371/journal.pone.0301664] [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: 08/12/2022] [Accepted: 03/20/2024] [Indexed: 07/12/2024] Open
Abstract
Influenza viruses constitute a major threat to human health globally. The viral surface glycoprotein hemagglutinin (HA) is the immunodominant antigen, contains the site for binding to the cellular receptor (RBS), and it is the major target of neutralizing antibody responses post-infection. We developed llama-derived single chain antibody fragments (VHHs) specific for type A influenza virus. Four VHHs were identified and further characterized. VHH D81 bound residues in the proximity of the C-terminal region of HA1 of H1 and H5 subtypes, and showed weak neutralizing activity, whereas VHH B33 bound residues in the proximity of the N-terminal region of the HA's stem domain (HA2) of H1, H5, and H9 subtypes, and showed no neutralizing activity. Of most relevance, VHHs E13 and G41 recognized highly conserved conformational epitopes on the H1 HA's globular domain (HA1) and showed high virus neutralizing activity (ranging between 0.94 to 0.01μM), when tested against several human H1N1 isolates. Additionally, E13 displayed abrogated virus replication of a panel of H1N1 strains spanning over 80 years of antigenic drift and isolated from human, avian, and swine origin. Interestingly, E13 conferred protection in vivo at a dose as low as 0.05 mg/kg. Mice treated with E13 intranasally resulted in undetectable virus challenge loads in the lungs at day 4 post-challenge. The transfer of sterilizing pan-H1 immunity, by a dose in the range of micrograms given intranasally, is of major significance for a monomeric VHH and supports the further development of E13 as an immunotherapeutic agent for the mitigation of influenza infections.
Collapse
Affiliation(s)
- Elena Susana Barbieri
- Virology Institute IncuINTA (IVIT-CONICET), National Institute of Agricultural Technology, Hurlingham, Buenos Aires, Argentina
- National Council for Scientific and Technical Research (CONICET), Buenos Aires City, Buenos Aires, Argentina
| | - Carla Sosa-Holt
- Virology Institute IncuINTA (IVIT-CONICET), National Institute of Agricultural Technology, Hurlingham, Buenos Aires, Argentina
- National Council for Scientific and Technical Research (CONICET), Buenos Aires City, Buenos Aires, Argentina
| | - Lorena Itati Ibañez
- National Council for Scientific and Technical Research (CONICET), Buenos Aires City, Buenos Aires, Argentina
- Institute of Science and Technology, Buenos Aires City, Buenos Aires, Argentina
| | - Josefina Baztarrica
- Virology Institute IncuINTA (IVIT-CONICET), National Institute of Agricultural Technology, Hurlingham, Buenos Aires, Argentina
| | - Lorena Garaicoechea
- Virology Institute IncuINTA (IVIT-CONICET), National Institute of Agricultural Technology, Hurlingham, Buenos Aires, Argentina
- National Council for Scientific and Technical Research (CONICET), Buenos Aires City, Buenos Aires, Argentina
| | - Claire Lindsey Gay
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| | - Carlos Joaquin Caceres
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| | - Matias Aduriz
- Virology Institute IncuINTA (IVIT-CONICET), National Institute of Agricultural Technology, Hurlingham, Buenos Aires, Argentina
- National Council for Scientific and Technical Research (CONICET), Buenos Aires City, Buenos Aires, Argentina
| | - Elsa Baumeister
- National Institute of Infectious Diseases, Malbran Institute, Buenos Aires City, Buenos Aires, Argentina
| | | | - Daniel Perez
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| | - Andrés Wigdorovitz
- Virology Institute IncuINTA (IVIT-CONICET), National Institute of Agricultural Technology, Hurlingham, Buenos Aires, Argentina
- National Council for Scientific and Technical Research (CONICET), Buenos Aires City, Buenos Aires, Argentina
| | - Gladys Viviana Parreño
- Virology Institute IncuINTA (IVIT-CONICET), National Institute of Agricultural Technology, Hurlingham, Buenos Aires, Argentina
- National Council for Scientific and Technical Research (CONICET), Buenos Aires City, Buenos Aires, Argentina
| | - Mariana Puntel
- Virology Institute IncuINTA (IVIT-CONICET), National Institute of Agricultural Technology, Hurlingham, Buenos Aires, Argentina
- National Council for Scientific and Technical Research (CONICET), Buenos Aires City, Buenos Aires, Argentina
| |
Collapse
|
20
|
Tyritidis I, Tsioupros E, Christou P, Koutras N, Morfos V, Nika K. A non-invasive nanobody probe for high precision mapping of Lck spatial distribution. Front Immunol 2024; 15:1440499. [PMID: 39021567 PMCID: PMC11251914 DOI: 10.3389/fimmu.2024.1440499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 06/18/2024] [Indexed: 07/20/2024] Open
Abstract
The tyrosine kinase Lck is mandatory for initiating signaling responses downstream the antigenic T cell receptor (TCR). Numerous studies have shown that a prerequisite for efficient and well-balanced Lck regulation and function is its finely orchestrated spatial distribution pattern, especially at the plane of the plasma membrane. There is a wealth of knowledge on Lck localization sites, preference for specialized lipid microenvironments and colocalization partners. However, several questions concerning the spatial organization of its differentially phosphorylated conformers and the dynamics of their juxtaposition in relation to ligated and non-ligated TCRs remain elusive. In this brief report we introduce a non-invasive nanobody-based approach for mapping Lck subcellular allocation with high precision. Our initial data using this methodology, provide insight into the topology of Lck in resting T cells and its confined localization in a strictly delimited environment within the plane of the plasma membrane.
Collapse
Affiliation(s)
- Ioannis Tyritidis
- Department of Biochemistry, School of Medicine, University of Patras, Patras, Greece
| | - Evangelos Tsioupros
- Department of Biochemistry, School of Medicine, University of Patras, Patras, Greece
| | - Pantelis Christou
- Department of Biochemistry, School of Medicine, University of Patras, Patras, Greece
| | - Nikolaos Koutras
- Department of Biochemistry, School of Medicine, University of Patras, Patras, Greece
| | - Vasileios Morfos
- Department of Biochemistry, School of Medicine, University of Patras, Patras, Greece
- Department of Molecular Immunology, Faculty of Biology, Institute of Biology III, University of Freiburg, Freiburg, Germany
| | - Konstantina Nika
- Department of Biochemistry, School of Medicine, University of Patras, Patras, Greece
| |
Collapse
|
21
|
Nemecz D, Nowak WA, Nemecz Á. VHH Nanobody Versatility against Pentameric Ligand-Gated Ion Channels. J Med Chem 2024; 67:8502-8518. [PMID: 38829690 PMCID: PMC11181324 DOI: 10.1021/acs.jmedchem.4c00231] [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: 01/26/2024] [Revised: 04/17/2024] [Accepted: 05/20/2024] [Indexed: 06/05/2024]
Abstract
Pentameric ligand-gated ion channels provide rapid chemical-electrical signal transmission between cells in the central and peripheral nervous system. Their dysfunction is associated with many nervous system disorders. They are composed of five identical (homomeric receptors) or homologous (heteromeric receptors) subunits. VHH nanobodies, or single-chain antibodies, are the variable domain, VHH, of antibodies that are composed of the heavy chain only from camelids. Their unique structure results in many specific biochemical and biophysical properties that make them an excellent alternative to conventional antibodies. This Perspective explores the published VHH nanobodies which have been isolated against pentameric ligand-gated ion channel subfamilies. It outlines the genetic and chemical modifications available to alter nanobody function. An assessment of the available functional and structural data indicate that it is feasible to create therapeutic agents and impart, through their modification, a given desired modulatory effect of its target receptor for current stoichiometric-specific VHH nanobodies.
Collapse
Affiliation(s)
- Dorota Nemecz
- Biochemistry
Department, Nicolaus Copernicus University
in Torun, 87-100 Torun, Poland
| | - Weronika A. Nowak
- Biochemistry
Department, Nicolaus Copernicus University
in Torun, 87-100 Torun, Poland
| | - Ákos Nemecz
- Biochemistry
Department, Nicolaus Copernicus University
in Torun, 87-100 Torun, Poland
| |
Collapse
|
22
|
De Greve H, Fioravanti A. Single domain antibodies from camelids in the treatment of microbial infections. Front Immunol 2024; 15:1334829. [PMID: 38827746 PMCID: PMC11140111 DOI: 10.3389/fimmu.2024.1334829] [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: 11/07/2023] [Accepted: 04/29/2024] [Indexed: 06/04/2024] Open
Abstract
Infectious diseases continue to pose significant global health challenges. In addition to the enduring burdens of ailments like malaria and HIV, the emergence of nosocomial outbreaks driven by antibiotic-resistant pathogens underscores the ongoing threats. Furthermore, recent infectious disease crises, exemplified by the Ebola and SARS-CoV-2 outbreaks, have intensified the pursuit of more effective and efficient diagnostic and therapeutic solutions. Among the promising options, antibodies have garnered significant attention due to their favorable structural characteristics and versatile applications. Notably, nanobodies (Nbs), the smallest functional single-domain antibodies of heavy-chain only antibodies produced by camelids, exhibit remarkable capabilities in stable antigen binding. They offer unique advantages such as ease of expression and modification and enhanced stability, as well as improved hydrophilicity compared to conventional antibody fragments (antigen-binding fragments (Fab) or single-chain variable fragments (scFv)) that can aggregate due to their low solubility. Nanobodies directly target antigen epitopes or can be engineered into multivalent Nbs and Nb-fusion proteins, expanding their therapeutic potential. This review is dedicated to charting the progress in Nb research, particularly those derived from camelids, and highlighting their diverse applications in treating infectious diseases, spanning both human and animal contexts.
Collapse
Affiliation(s)
- Henri De Greve
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Antonella Fioravanti
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
- VIB-VUB Center for Structural Biology, Vrije Universiteit Brussel, Brussels, Belgium
- Fondazione ParSeC – Parco delle Scienze e della Cultura, Prato, Italy
| |
Collapse
|
23
|
Zhang R, Huang L, Zhang X, Yu Y, Liang T, Wang H, Zhang X, Hu D, Wang B, Wang Y, Jiang J, Yu X. Proteomics Platform Reveals Broad-Spectrum Nanobodies for SARS-CoV-2 Variant Neutralization. J Proteome Res 2024; 23:1559-1570. [PMID: 38603467 DOI: 10.1021/acs.jproteome.3c00569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
The ongoing evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to the emergence of different variants of concerns with immune evasion that have been prevalent over the past three years. Nanobodies, the functional variable regions of camelid heavy-chain-only antibodies, have garnered interest in developing neutralizing antibodies due to their smaller size, structural stability, ease of production, high affinity, and low immunogenicity, among other characteristics. In this work, we describe an integrated proteomics platform for the high-throughput screening of nanobodies against different SARS-CoV-2 spike variants. To demonstrate this platform, we immunized a camel with subunit 1 (S1) of the wild-type spike protein and constructed a nanobody phage library. The binding and neutralizing activities of the nanobodies against 72 spike variants were then measured, resulting in the identification of two nanobodies (C-282 and C-39) with broad neutralizing activity against six non-Omicron variants (D614G, Alpha, Beta, Gamma, Delta, Kappa) and five Omicron variants (BA.1-5). Their neutralizing capability was validated using in vitro pseudovirus-based neutralization assays. All these results demonstrate the utility of our proteomics platform to identify new nanobodies with broad neutralizing capability and to develop a treatment for patients with SARS-CoV-2 variant infection in the future.
Collapse
Affiliation(s)
- Ran Zhang
- School of Basic Medicine Sciences, Anhui Medical University, Hefei, Anhui 230031, PR China
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Lan Huang
- Changping Laboratory, Beijing 102206, China
| | - Xiaohan Zhang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | | | - Te Liang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Hongye Wang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xiaomei Zhang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Di Hu
- ProteomicsEra Medical Co., Ltd., Beijing 102206, China
| | - Bingwei Wang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | | | - Junyi Jiang
- Translational Medicine Technology Platform, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xiaobo Yu
- School of Basic Medicine Sciences, Anhui Medical University, Hefei, Anhui 230031, PR China
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| |
Collapse
|
24
|
Liu M, Jin D, Yu W, Yu J, Cao K, Cheng J, Zheng X, Wang A, Liu Y. Enhancing Tumor Immunotherapy by Multivalent Anti-PD-L1 Nanobody Assembled via Ferritin Nanocage. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308248. [PMID: 38491904 PMCID: PMC11132087 DOI: 10.1002/advs.202308248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/12/2024] [Indexed: 03/18/2024]
Abstract
Increasing immunotherapy response rate and durability can lead to significant improvements in cancer care. To address this challenge, a novel multivalent immune checkpoint therapeutic platform is constructed through site-specific ligation of anti-PD-L1 nanobody (Nb) on ferritin (Ftn) nanocage. Nb-Ftn blocks PD-1/PD-L1 interaction and downregulates PD-L1 levels via endocytosis-induced degradation. In addition, the cage structure of Ftn allows encapsulation of indocyanine green (ICG), an FDA-approved dye. Photothermal treatment with Nb-Ftn@ICG induces immunogenic death of tumor cells, which improves systemic immune response via maturation of dendritic cells and enhanced infiltration of T cells. Moreover, Nb-Ftn encapsulation significantly enhances cellular uptake, tumor accumulation and retention of ICG. In vivo assays showed that this nanoplatform ablates the primary tumor, suppresses abscopal tumors and inhibits tumor metastasis, leading to a prolonged survival rate. This work presents a novel strategy for improving cancer immunotherapy using multivalent nanobody-ferritin conjugates as immunological targeting and enhancing carriers.
Collapse
Affiliation(s)
- Manman Liu
- Department of Pharmacythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineDepartment of ChemistryUniversity of Science and Technology of ChinaHefei230001China
| | - Duo Jin
- Department of Pharmacythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineDepartment of ChemistryUniversity of Science and Technology of ChinaHefei230001China
| | - Wenxin Yu
- Department of Pharmacythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineDepartment of ChemistryUniversity of Science and Technology of ChinaHefei230001China
| | - Jiaji Yu
- Department of Pharmacythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineDepartment of ChemistryUniversity of Science and Technology of ChinaHefei230001China
| | - Kaiming Cao
- Department of Pharmacythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineDepartment of ChemistryUniversity of Science and Technology of ChinaHefei230001China
| | - Junjie Cheng
- Department of Pharmacythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineDepartment of ChemistryUniversity of Science and Technology of ChinaHefei230001China
| | - Xiaohu Zheng
- The CAS Key Laboratory of Innate Immunity and Chronic DiseaseSchool of Basic Medical SciencesCenter for Advanced Interdisciplinary Science and Biomedicine of IHMDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230027China
| | - Andrew Wang
- Department of Radiation OncologyUniversity of Texas Southwestern Medical CenterDallas75230USA
| | - Yangzhong Liu
- Department of Pharmacythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineDepartment of ChemistryUniversity of Science and Technology of ChinaHefei230001China
| |
Collapse
|
25
|
Sencanski M, Glisic S, Kubale V, Cotman M, Mavri J, Vrecl M. Computational Modeling and Characterization of Peptides Derived from Nanobody Complementary-Determining Region 2 (CDR2) Targeting Active-State Conformation of the β 2-Adrenergic Receptor (β 2AR). Biomolecules 2024; 14:423. [PMID: 38672440 PMCID: PMC11048008 DOI: 10.3390/biom14040423] [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: 02/27/2024] [Revised: 03/20/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
This study assessed the suitability of the complementarity-determining region 2 (CDR2) of the nanobody (Nb) as a template for the derivation of nanobody-derived peptides (NDPs) targeting active-state β2-adrenergic receptor (β2AR) conformation. Sequences of conformationally selective Nbs favoring the agonist-occupied β2AR were initially analyzed by the informational spectrum method (ISM). The derived NDPs in complex with β2AR were subjected to protein-peptide docking, molecular dynamics (MD) simulations, and metadynamics-based free-energy binding calculations. Computational analyses identified a 25-amino-acid-long CDR2-NDP of Nb71, designated P4, which exhibited the following binding free-energy for the formation of the β2AR:P4 complex (ΔG = -6.8 ± 0.8 kcal/mol or a Ki = 16.5 μM at 310 K) and mapped the β2AR:P4 amino acid interaction network. In vitro characterization showed that P4 (i) can cross the plasma membrane, (ii) reduces the maximum isoproterenol-induced cAMP level by approximately 40% and the isoproterenol potency by up to 20-fold at micromolar concentration, (iii) has a very low affinity to interact with unstimulated β2AR in the cAMP assay, and (iv) cannot reduce the efficacy and potency of the isoproterenol-mediated β2AR/β-arrestin-2 interaction in the BRET2-based recruitment assay. In summary, the CDR2-NDP, P4, binds preferentially to agonist-activated β2AR and disrupts Gαs-mediated signaling.
Collapse
Affiliation(s)
- Milan Sencanski
- Laboratory for Plant Molecular Biology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11000 Belgrade, Serbia
- Laboratory for Bioinformatics and Computational Chemistry, Institute of Nuclear Sciences VINCA, National Institute of Serbia, University of Belgrade, 11000 Belgrade, Serbia;
| | - Sanja Glisic
- Laboratory for Bioinformatics and Computational Chemistry, Institute of Nuclear Sciences VINCA, National Institute of Serbia, University of Belgrade, 11000 Belgrade, Serbia;
| | - Valentina Kubale
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (V.K.); (M.C.)
| | - Marko Cotman
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (V.K.); (M.C.)
| | - Janez Mavri
- Department of Computational Biochemistry and Drug Design, National Institute of Chemistry, 1000 Ljubljana, Slovenia;
| | - Milka Vrecl
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (V.K.); (M.C.)
| |
Collapse
|
26
|
Li S, Meng X, Li R, Huang B, Wang X. NanoBERTa-ASP: predicting nanobody paratope based on a pretrained RoBERTa model. BMC Bioinformatics 2024; 25:122. [PMID: 38515052 PMCID: PMC10956323 DOI: 10.1186/s12859-024-05750-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 03/18/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Nanobodies, also known as VHH or single-domain antibodies, are unique antibody fragments derived solely from heavy chains. They offer advantages of small molecules and conventional antibodies, making them promising therapeutics. The paratope is the specific region on an antibody that binds to an antigen. Paratope prediction involves the identification and characterization of the antigen-binding site on an antibody. This process is crucial for understanding the specificity and affinity of antibody-antigen interactions. Various computational methods and experimental approaches have been developed to predict and analyze paratopes, contributing to advancements in antibody engineering, drug development, and immunotherapy. However, existing predictive models trained on traditional antibodies may not be suitable for nanobodies. Additionally, the limited availability of nanobody datasets poses challenges in constructing accurate models. METHODS To address these challenges, we have developed a novel nanobody prediction model, named NanoBERTa-ASP (Antibody Specificity Prediction), which is specifically designed for predicting nanobody-antigen binding sites. The model adopts a training strategy more suitable for nanobodies, based on an advanced natural language processing (NLP) model called BERT (Bidirectional Encoder Representations from Transformers). To be more specific, the model utilizes a masked language modeling approach named RoBERTa (Robustly Optimized BERT Pretraining Approach) to learn the contextual information of the nanobody sequence and predict its binding site. RESULTS NanoBERTa-ASP achieved exceptional performance in predicting nanobody binding sites, outperforming existing methods, indicating its proficiency in capturing sequence information specific to nanobodies and accurately identifying their binding sites. Furthermore, NanoBERTa-ASP provides insights into the interaction mechanisms between nanobodies and antigens, contributing to a better understanding of nanobodies and facilitating the design and development of nanobodies with therapeutic potential. CONCLUSION NanoBERTa-ASP represents a significant advancement in nanobody paratope prediction. Its superior performance highlights the potential of deep learning approaches in nanobody research. By leveraging the increasing volume of nanobody data, NanoBERTa-ASP can further refine its predictions, enhance its performance, and contribute to the development of novel nanobody-based therapeutics. Github repository: https://github.com/WangLabforComputationalBiology/NanoBERTa-ASP.
Collapse
Affiliation(s)
- Shangru Li
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen, China
| | - Xiangpeng Meng
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen, China
| | - Rui Li
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen, China
| | - Bingding Huang
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen, China.
| | - Xin Wang
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen, China.
| |
Collapse
|
27
|
Zhang W, Wang H, Wu T, Gao X, Shang Y, Zhang Z, Liu X, Li Y. A SARS-CoV-2 Nanobody Displayed on the Surface of Human Ferritin with High Neutralization Activity. Int J Nanomedicine 2024; 19:2429-2440. [PMID: 38476285 PMCID: PMC10929646 DOI: 10.2147/ijn.s450829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Purpose COVID-19 is rampant throughout the world, which has caused great damage to human lives and seriously hindered the development of the global economy. Aiming at the treatment of SARS-CoV-2, in this study, we proposed a novel fenobody strategy based on ferritin (Fe) self-assembly technology. Methods The neutralizing nanobody H11-D4 of SARS-CoV-2 fused to the C-terminus of end-modified human ferritin was expressed in E. coli and silkworm baculovirus expression systems. A large number of nanoparticles were successfully self-assembled in silkworms, while relatively few nanoparticles can be observed in the treated products from E. coli by electron microscopy. Subsequently, the fenobody's expression level and neutralizing activity were then evaluated. Results The results showed that the IC50 of H11-D4 and fenobody Fe-H11-D4 expressed in E. coli were 171.1 nmol L-1 and 20.87 nmol L-1, respectively. However, the IC50 of Fe-HD11-D4 expressed in silkworms was 1.46 nmol L-1 showing better neutralization activity. Conclusion Therefore, fenobodies can be well self-assembled in silkworm baculovirus expression system, and ferritin self-assembly technology can effectively improve nanobody neutralization activity.
Collapse
Affiliation(s)
- Wenrong Zhang
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
- College of Life Sciences, Capital Normal University, Beijing, People’s Republic of China
| | - Haining Wang
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Tong Wu
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Xintao Gao
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Yuting Shang
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Zhifang Zhang
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Xingjian Liu
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Yinü Li
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| |
Collapse
|
28
|
Li M, Zhao X, Yu C, Wang L. Antibody-Drug Conjugate Overview: a State-of-the-art Manufacturing Process and Control Strategy. Pharm Res 2024; 41:419-440. [PMID: 38366236 DOI: 10.1007/s11095-023-03649-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 12/16/2023] [Indexed: 02/18/2024]
Abstract
Antibody-drug conjugates (ADCs) comprise an antibody, linker, and drug, which direct their highly potent small molecule drugs to target tumor cells via specific binding between the antibody and surface antigens. The antibody, linker, and drug should be properly designed or selected to achieve the desired efficacy while minimizing off-target toxicity. With a unique and complex structure, there is inherent heterogeneity introduced by product-related variations and the manufacturing process. Here this review primarily covers recent key advances in ADC history, clinical development status, molecule design, manufacturing processes, and quality control. The manufacturing process, especially the conjugation process, should be carefully developed, characterized, validated, and controlled throughout its lifecycle. Quality control is another key element to ensure product quality and patient safety. A patient-centric strategy has been well recognized and adopted by the pharmaceutical industry for therapeutic proteins, and has been successfully implemented for ADCs as well, to ensure that ADC products maintain their quality until the end of their shelf life. Deep product understanding and process knowledge defines attribute testing strategies (ATS). Quality by design (QbD) is a powerful approach for process and product development, and for defining an overall control strategy. Finally, we summarize the current challenges on ADC development and provide some perspectives that may help to give related directions and trigger more cross-functional research to surmount those challenges.
Collapse
Affiliation(s)
- Meng Li
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Xueyu Zhao
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Chuanfei Yu
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Lan Wang
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, National Institutes for Food and Drug Control, Beijing, People's Republic of China.
| |
Collapse
|
29
|
Heremans J, Maximilian Awad R, Bridoux J, Ertveldt T, Caveliers V, Madder A, Hoogenboom R, Devoogdt N, Ballet S, Hernot S, Breckpot K, Martin C. Sustained release of a human PD-L1 single-domain antibody using peptide-based hydrogels. Eur J Pharm Biopharm 2024; 196:114183. [PMID: 38246566 DOI: 10.1016/j.ejpb.2024.114183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 01/14/2024] [Indexed: 01/23/2024]
Abstract
Monoclonal antibodies (mAbs) targeting the immune checkpoint axis, which contains the programmed cell death protein-1 (PD-1) and its ligand PD-L1, revolutionized the field of oncology. Unfortunately, the large size of mAbs and the presence of an Fc fraction limit their tumor penetrative capacities and support off-target effects, potentially resulting in unresponsive patients and immune-related adverse events (irAEs) respectively. Single-domain antibodies (sdAbs) are ten times smaller than conventional mAbs and represent an emerging antibody subclass that has been proposed as next generation immune checkpoint inhibitor (ICI) therapeutics. They demonstrate favorable characteristics, such as an excellent stability, high antigen-binding affinity and an enhanced tumor penetration. Because sdAbs have a short half-life, methods to prolong their presence in the circulation and at the target site might be necessary in some cases to unfold their full therapeutic potential. In this study, we investigated a peptide-based hydrogel as an injectable biomaterial depot formulation for the sustained release of the human PD-L1 sdAb K2. We showed that a hydrogel composed of the amphipathic hexapeptide hydrogelator H-FQFQFK-NH2 prolonged the in vivo release of K2 after subcutaneous (s.c.) injection, up to at least 72 h, as monitored by SPECT/CT and fluorescence imaging. Additionally, after encapsulation in the hydrogel and s.c. administration, a significantly extended systemic presence and tumor uptake of K2 was observed in mice bearing a melanoma tumor expressing human PD-L1. Altogether, this study describes how peptide hydrogels can be exploited to provide the sustained release of sdAbs, thereby potentially enhancing its clinical and therapeutic effects.
Collapse
Affiliation(s)
- Julie Heremans
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, B-1050 Brussels, Belgium
| | - Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Jessica Bridoux
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Thomas Ertveldt
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Vicky Caveliers
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Annemieke Madder
- Organic and Biomimetic Chemistry Research Group, Ghent University, 9000 Ghent, Belgium
| | - Richard Hoogenboom
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, 9000 Ghent, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Steven Ballet
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, B-1050 Brussels, Belgium
| | - Sophie Hernot
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Charlotte Martin
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, B-1050 Brussels, Belgium
| |
Collapse
|
30
|
De Leiris N, Perret P, Lombardi C, Gözel B, Chierici S, Millet P, Debiossat M, Bacot S, Tournier BB, Chames P, Lenormand JL, Ghezzi C, Fagret D, Moulin M. A single-domain antibody for the detection of pathological Tau protein in the early stages of oligomerization. J Transl Med 2024; 22:163. [PMID: 38365700 PMCID: PMC10870657 DOI: 10.1186/s12967-024-04987-1] [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: 10/23/2023] [Accepted: 02/12/2024] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND Soluble oligomeric forms of Tau protein have emerged as crucial players in the propagation of Tau pathology in Alzheimer's disease (AD). Our objective is to introduce a single-domain antibody (sdAb) named 2C5 as a novel radiotracer for the efficient detection and longitudinal monitoring of oligomeric Tau species in the human brain. METHODS The development and production of 2C5 involved llama immunization with the largest human Tau isoform oligomers of different maturation states. Subsequently, 2C5 underwent comprehensive in vitro characterization for affinity and specificity via Enzyme-Linked Immunosorbent Assay and immunohistochemistry on human brain slices. Technetium-99m was employed to radiolabel 2C5, followed by its administration to healthy mice for biodistribution analysis. RESULTS 2C5 exhibited robust binding affinity towards Tau oligomers (Kd = 6.280 nM ± 0.557) and to Tau fibers (Kd = 5.024 nM ± 0.453), with relatively weaker binding observed for native Tau protein (Kd = 1791 nM ± 8.714) and amyloid peptide (Kd > 10,000 nM). Remarkably, this SdAb facilitated immuno-histological labeling of pathological forms of Tau in neurons and neuritic plaques, yielding a high-contrast outcome in AD patients, closely mirroring the performance of reference antibodies AT8 and T22. Furthermore, 2C5 SdAb was successfully radiolabeled with 99mTc, preserving stability for up to 6 h post-radiolabeling (radiochemical purity > 93%). However, following intravenous injection into healthy mice, the predominant uptake occurred in kidneys, amounting to 115.32 ± 3.67, 97.70 ± 43.14 and 168.20 ± 34.52% of injected dose per gram (% ID/g) at 5, 10 and 45 min respectively. Conversely, brain uptake remained minimal at all measured time points, registering at 0.17 ± 0.03, 0.12 ± 0.07 and 0.02 ± 0.01% ID/g at 5, 10 and 45 min post-injection respectively. CONCLUSION 2C5 demonstrates excellent affinity and specificity for pathological Tau oligomers, particularly in their early stages of oligomerization. However, the current limitation of insufficient blood-brain barrier penetration necessitates further modifications before considering its application in nuclear medicine imaging for humans.
Collapse
Affiliation(s)
- Nicolas De Leiris
- University Grenoble Alpes, Clinique Universitaire de Médecine Nucléaire, INSERM, Centre Hospitalier Universitaire Grenoble Alpes, LRB, CS 10217, 38043, Grenoble CEDEX 9, France.
| | - Pascale Perret
- University Grenoble Alpes, INSERM, LRB, 38000, Grenoble, France
| | | | - Bülent Gözel
- University Grenoble Alpes, INSERM, LRB, 38000, Grenoble, France
| | - Sabine Chierici
- University Grenoble Alpes, CNRS, DCM, 38000, Grenoble, France
| | - Philippe Millet
- Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | | | - Sandrine Bacot
- University Grenoble Alpes, INSERM, LRB, 38000, Grenoble, France
| | - Benjamin B Tournier
- Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | - Patrick Chames
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | | | | | - Daniel Fagret
- University Grenoble Alpes, INSERM, LRB, 38000, Grenoble, France
| | - Marcelle Moulin
- University Grenoble Alpes, INSERM, LRB, 38000, Grenoble, France
| |
Collapse
|
31
|
Sun M, Sun Y, Yang Y, Zhao M, Cao D, Zhang M, Xia D, Wang T, Gao Y, Wang S, Wang H, Cai X, An T. Multivalent nanobody-based sandwich enzyme-linked immunosorbent assay for sensitive detection of porcine reproductive and respiratory syndrome virus. Int J Biol Macromol 2024; 258:128896. [PMID: 38143067 DOI: 10.1016/j.ijbiomac.2023.128896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/13/2023] [Accepted: 12/17/2023] [Indexed: 12/26/2023]
Abstract
The pandemic of the porcine reproductive and respiratory syndrome virus (PRRSV) has caused huge economic losses and continues to threaten the swine industry worldwide. Nucleocapsid protein (N protein) is the primary antigen of PRRSV for development of sensitive diagnostic assays. Two high affinity nanobodies against N protein, Nb12 and Nb35, were selected and employed to develop a sandwich ELISA. Further we improved the ELISA method to obtain greater sensitivity, a trivalent nanobody (3 × Nb35) and a bivalent nanobody-HRP fusion protein (2 × Nb12-HRP) were expressed and used. This modified ELISA was found to have high sensitivity for detecting PRRSV, with a detection limit of 10 TCID50/ml (median tissue culture infectious dose), which was approximately 200-fold greater than the single-copy nanobody-based sandwich ELISA. The developed assay shows high specificity and can detect almost all circulating lineages of PRRSV-2 in China. This study provides suggestions for reforming nanobodies and for the further development of multivalent nanobody-based ELISAs for other various viruses.
Collapse
Affiliation(s)
- Mingxia Sun
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Yue Sun
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Yongbo Yang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Man Zhao
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Dan Cao
- Soybean Research Institute, Heilongjiang Academy of Agricultural Science, Harbin 150086, China
| | - Minmin Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Dasong Xia
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Tao Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Yanfei Gao
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Shanghui Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Haiwei Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Xuehui Cai
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; Heilongjiang Research Center of Veterinary Biopharmaceutical Technology, Harbin 150069, China.
| | - Tongqing An
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin 150069, China.
| |
Collapse
|
32
|
Xiong S, Liu Z, Yi X, Liu K, Huang B, Wang X. NanoLAS: a comprehensive nanobody database with data integration, consolidation and application. Database (Oxford) 2024; 2024:baae003. [PMID: 38300518 PMCID: PMC10833066 DOI: 10.1093/database/baae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/15/2023] [Accepted: 01/06/2024] [Indexed: 02/02/2024]
Abstract
Nanobodies, a unique subclass of antibodies first discovered in camelid animals, are composed solely of a single heavy chain's variable region. Their significantly reduced molecular weight, in comparison to conventional antibodies, confers numerous advantages in the treatment of various diseases. As research and applications involving nanobodies expand, the quantity of identified nanobodies is also rapidly growing. However, the existing antibody databases are deficient in type and coverage, failing to satisfy the comprehensive needs of researchers and thus impeding progress in nanobody research. In response to this, we have amalgamated data from multiple sources to successfully assemble a new and comprehensive nanobody database. This database has currently included the latest nanobody data and provides researchers with an excellent search and data display interface, thus facilitating the progression of nanobody research and their application in disease treatment. In summary, the newly constructed Nanobody Library and Archive System may significantly enhance the retrieval efficiency and application potential of nanobodies. We envision that Nanobody Library and Archive System will serve as an accessible, robust and efficient tool for nanobody research and development, propelling advancements in the field of biomedicine. Database URL: https://www.nanolas.cloud.
Collapse
Affiliation(s)
| | - Zhengwen Liu
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen 518118, China
| | - Xin Yi
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen 518118, China
| | - Kai Liu
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen 518118, China
| | - Bingding Huang
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen 518118, China
| | - Xin Wang
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen 518118, China
| |
Collapse
|
33
|
Babamohamadi M, Mohammadi N, Faryadi E, Haddadi M, Merati A, Ghobadinezhad F, Amirian R, Izadi Z, Hadjati J. Anti-CTLA-4 nanobody as a promising approach in cancer immunotherapy. Cell Death Dis 2024; 15:17. [PMID: 38191571 PMCID: PMC10774412 DOI: 10.1038/s41419-023-06391-x] [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: 06/21/2023] [Revised: 11/25/2023] [Accepted: 12/13/2023] [Indexed: 01/10/2024]
Abstract
Cancer is one of the most common diseases and causes of death worldwide. Since common treatment approaches do not yield acceptable results in many patients, developing innovative strategies for effective treatment is necessary. Immunotherapy is one of the promising approaches that has been highly regarded for preventing tumor recurrence and new metastases. Meanwhile, inhibiting immune checkpoints is one of the most attractive methods of cancer immunotherapy. Cytotoxic T lymphocyte-associated protein-4 (CTLA-4) is an essential immune molecule that plays a vital role in cell cycle modulation, regulation of T cell proliferation, and cytokine production. This molecule is classically expressed by stimulated T cells. Inhibition of overexpression of immune checkpoints such as CTLA-4 receptors has been confirmed as an effective strategy. In cancer immunotherapy, immune checkpoint-blocking drugs can be enhanced with nanobodies that target immune checkpoint molecules. Nanobodies are derived from the variable domain of heavy antibody chains. These small protein fragments have evolved entirely without a light chain and can be used as a powerful tool in imaging and treating diseases with their unique structure. They have a low molecular weight, which makes them smaller than conventional antibodies while still being able to bind to specific antigens. In addition to low molecular weight, specific binding to targets, resistance to temperature, pH, and enzymes, high ability to penetrate tumor tissues, and low toxicity make nanobodies an ideal approach to overcome the disadvantages of monoclonal antibody-based immunotherapy. In this article, while reviewing the cellular and molecular functions of CTLA-4, the structure and mechanisms of nanobodies' activity, and their delivery methods, we will explain the advantages and challenges of using nanobodies, emphasizing immunotherapy treatments based on anti-CTLA-4 nanobodies.
Collapse
Affiliation(s)
- Mehregan Babamohamadi
- Department of Biology, School of Natural Sciences, University of Tabriz, Tabriz, Iran
- Stem Cell and Regenerative Medicine Innovation Center, Tehran University of Medical Sciences, Tehran, Iran
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Nastaran Mohammadi
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Elham Faryadi
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Maryam Haddadi
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amirhossein Merati
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Medical Laboratory Sciences, School of Paramedical, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farbod Ghobadinezhad
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Roshanak Amirian
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zhila Izadi
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Jamshid Hadjati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
34
|
Farka Z, Brandmeier JC, Mickert MJ, Pastucha M, Lacina K, Skládal P, Soukka T, Gorris HH. Nanoparticle-Based Bioaffinity Assays: From the Research Laboratory to the Market. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307653. [PMID: 38039956 DOI: 10.1002/adma.202307653] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/16/2023] [Indexed: 12/03/2023]
Abstract
Advances in the development of new biorecognition elements, nanoparticle-based labels as well as instrumentation have inspired the design of new bioaffinity assays. This review critically discusses the potential of nanoparticles to replace current enzymatic or molecular labels in immunoassays and other bioaffinity assays. Successful implementations of nanoparticles in commercial assays and the need for rapid tests incorporating nanoparticles in different roles such as capture support, signal generation elements, and signal amplification systems are highlighted. The limited number of nanoparticles applied in current commercial assays can be explained by challenges associated with the analysis of real samples (e.g., blood, urine, or nasal swabs) that are difficult to resolve, particularly if the same performance can be achieved more easily by conventional labels. Lateral flow assays that are based on the visual detection of the red-colored line formed by colloidal gold are a notable exception, exemplified by SARS-CoV-2 rapid antigen tests that have moved from initial laboratory testing to widespread market adaption in less than two years.
Collapse
Affiliation(s)
- Zdeněk Farka
- Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Julian C Brandmeier
- Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Universitätsstr. 31, 93053, Regensburg, Germany
| | | | - Matěj Pastucha
- Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
- TestLine Clinical Diagnostics, Křižíkova 188, Brno, 612 00, Czech Republic
| | - Karel Lacina
- CEITEC-Central European Institute of Technology, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Petr Skládal
- Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Tero Soukka
- Department of Life Technologies/Biotechnology, University of Turku, Kiinamyllynkatu 10, Turku, 20520, Finland
| | - Hans H Gorris
- Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| |
Collapse
|
35
|
Yang X, Sun Z, He Z, Xie X, Liu X. Combination of nanobody and peptidomimetic to develop novel immunoassay platforms for detecting ochratoxin A in cereals. Food Chem 2023; 429:137018. [PMID: 37517225 DOI: 10.1016/j.foodchem.2023.137018] [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: 05/10/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 08/01/2023]
Abstract
Mimotope-based immunoassays for mycotoxins eliminate the requirement for large amounts of mycotoxin standards for the chemosynthesis of artificial antigens. Herein, the nanobody-based magnetic beads were used to screen the mimotope (peptidomimetic) of ochratoxin A (OTA) from the phage-displayed peptide library. The interactions between nanobody and the most sensitive Y4 peptidomimetic were investigated by computer-assisted simulation and compared with those between nanobody and OTA. By combining the nanobody, the phage-displayed Y4 and alkaline phosphatase-tagged Y4 fusion protein as the competing antigens, were used to develop two novel immunoassay platforms (PN-ELISA and APN-ELISA). The two methods are advantageous in the use of nontoxic substitutes of OTA and avoiding the use of monoclonal antibodies. Moreover, good analytical performances of both methods were obtained and confirmed by liquid chromatography tandem mass spectrometry. Therefore, the proposed novel methods based on nanobody and peptidomimetic were demonstrated to be highly reliable for detecting OTA in food.
Collapse
Affiliation(s)
- Xun Yang
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Zhichang Sun
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Zhenyun He
- School of Food Science and Engineering, Hainan University, Haikou 570228, China; Hainan College of Economics and Business, Haikou 571127, China
| | - Xiaoxia Xie
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Xing Liu
- School of Food Science and Engineering, Hainan University, Haikou 570228, China.
| |
Collapse
|
36
|
Zha J, Liu D, Ren J, Liu Z, Wu X. Advances in Metabolic Engineering of Pichia pastoris Strains as Powerful Cell Factories. J Fungi (Basel) 2023; 9:1027. [PMID: 37888283 PMCID: PMC10608127 DOI: 10.3390/jof9101027] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 10/28/2023] Open
Abstract
Pichia pastoris is the most widely used microorganism for the production of secreted industrial proteins and therapeutic proteins. Recently, this yeast has been repurposed as a cell factory for the production of chemicals and natural products. In this review, the general physiological properties of P. pastoris are summarized and the readily available genetic tools and elements are described, including strains, expression vectors, promoters, gene editing technology mediated by clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9, and adaptive laboratory evolution. Moreover, the recent achievements in P. pastoris-based biosynthesis of proteins, natural products, and other compounds are highlighted. The existing issues and possible solutions are also discussed for the construction of efficient P. pastoris cell factories.
Collapse
Affiliation(s)
- Jian Zha
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (D.L.); (J.R.); (Z.L.)
| | | | | | | | - Xia Wu
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (D.L.); (J.R.); (Z.L.)
| |
Collapse
|
37
|
Zhou S, Fang X, Lv J, Yang Y, Zeng Y, Liu Y, Wei W, Huang G, Zhang B, Wu C. Site-Specific Modification of Single Domain Antibodies by Enzyme-Immobilized Magnetic Beads. Bioconjug Chem 2023; 34:1914-1922. [PMID: 37804224 DOI: 10.1021/acs.bioconjchem.3c00423] [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: 10/09/2023]
Abstract
Nanobodies as imaging agents and drug conjugates have shown great potential for cancer diagnostics and therapeutics. However, site-specific modification of a nanobody with microbial transglutaminase (mTGase) encounters problems in protein separation and purification. Here, we describe a facile yet reliable strategy of immobilizing mTGase onto magnetic beads for site-specific nanobody modification. The mTGase immobilized on magnetic beads (MB-mTGase) exhibits catalytic activity nearly equivalent to that of the free mTGase, with good reusability and universality. Magnetic separation simplifies the protein purification step and reduces the loss of nanobody bioconjugates more effectively than size exclusion chromatography. Using MB-mTGase, we demonstrate site-specific conjugation of nanobodies with fluorescent dyes and polyethylene glycol molecules, enabling targeted immunofluorescence imaging and improved circulation dynamics and tumor accumulation in vivo. The combined advantages of MB-mTGase method, including high conjugation efficiency, quick purification, less protein loss, and recycling use, are promising for site-specific nanobody functionalization and biomedical applications.
Collapse
Affiliation(s)
- Siyu Zhou
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| | - Xiaofeng Fang
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| | - Jiahui Lv
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| | - Yicheng Yang
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| | - Yiqi Zeng
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| | - Ying Liu
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Bo Zhang
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| | - Changfeng Wu
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| |
Collapse
|
38
|
Monti G, Vincke C, Lunding M, Jensen AMG, Madsen P, Muyldermans S, Kjolby M, Andersen OM. Epitope mapping of nanobodies binding the Alzheimer's disease receptor SORLA. J Biotechnol 2023; 375:17-27. [PMID: 37634829 DOI: 10.1016/j.jbiotec.2023.08.005] [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: 05/09/2023] [Revised: 08/20/2023] [Accepted: 08/24/2023] [Indexed: 08/29/2023]
Abstract
Reduced levels of the Sortilin-related receptor with A-type repeats (SORLA) in different brain regions as well as in the cerebrospinal fluid have been associated with Alzheimer's disease. Methods and reagents to develop reliable detection assays to quantify SORLA and its specific isoforms are therefore much needed. Nanobodies (Nbs) are unique biomolecules derived from the blood of camelids that display advantageous physicochemical and antigen affinity properties, making them attractive tools with great relevance to both diagnostic and therapeutic applications. Here, we purified and characterized eight Nbs that were isolated from the blood of an alpaca immunized with the recombinant extracellular domain of SORLA. The selected Nbs showed high affinity to SORLA in the low nanomolar range as observed by surface plasmon resonance. For mapping of the Nbs' epitopes within the antigen, we transiently transfected HEK293 cells with a panel of SORLA deletion constructs, and developed a protocol of immunostaining by applying fluorescent dye conjugated Nbs. With this method, we showed that the selected Nbs specifically recognize a part of SORLA containing Fibronectin-type III domains, representing promising tools not only for disease clarifying research, but also for translational medicine as candidates for clinical diagnostic purposes.
Collapse
Affiliation(s)
- Giulia Monti
- Department of Biomedicine, Aarhus University, Høegh‑Guldbergs Gade 10, 8000 Aarhus C, Denmark
| | - Cécile Vincke
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Melanie Lunding
- Department of Biomedicine, Aarhus University, Høegh‑Guldbergs Gade 10, 8000 Aarhus C, Denmark
| | - Anne Mette G Jensen
- Department of Biomedicine, Aarhus University, Høegh‑Guldbergs Gade 10, 8000 Aarhus C, Denmark
| | - Peder Madsen
- Department of Biomedicine, Aarhus University, Høegh‑Guldbergs Gade 10, 8000 Aarhus C, Denmark
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Mads Kjolby
- Department of Biomedicine, Aarhus University, Høegh‑Guldbergs Gade 10, 8000 Aarhus C, Denmark; Department of Clinical Pharmacology and Steno Diabetes Center Aarhus, Aarhus University Hospital, Denmark
| | - Olav M Andersen
- Department of Biomedicine, Aarhus University, Høegh‑Guldbergs Gade 10, 8000 Aarhus C, Denmark.
| |
Collapse
|
39
|
Thran M, Pönisch M, Danz H, Horscroft N, Ichtchenko K, Tzipori S, Shoemaker CB. Co-administration of an effector antibody enhances the half-life and therapeutic potential of RNA-encoded nanobodies. Sci Rep 2023; 13:14632. [PMID: 37670025 PMCID: PMC10480410 DOI: 10.1038/s41598-023-41092-7] [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: 05/30/2023] [Accepted: 08/22/2023] [Indexed: 09/07/2023] Open
Abstract
The incidence of Clostridioides difficile infection (CDI) and associated mortality have increased rapidly worldwide in recent years. Therefore, it is critical to develop new therapies for CDI. Here we report on the development of mRNA-LNPs encoding camelid-derived VHH-based neutralizing agents (VNAs) targeting toxins A and/or B of C. difficile. In preclinical models, intravenous administration of the mRNA-LNPs provided serum VNA levels sufficient to confer protection of mice against severe disease progression following toxin challenge. Furthermore, we employed an mRNA-LNP encoded effector antibody, a molecular tool designed to specifically bind an epitopic tag linked to the VNAs, to prolong VNA serum half-life. Co-administration of VNA-encoding mRNA-LNPs and an effector antibody, either provided as recombinant protein or encoded by mRNA-LNP, increased serum VNA half-life in mice and in gnotobiotic piglets. Prolonged serum half-life was associated with higher concentrations of serum VNA and enhanced prophylactic protection of mice in challenge models.
Collapse
Affiliation(s)
| | | | - Hillary Danz
- Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary Medicine, North Grafton, MA, 01536, USA
| | | | - Konstantin Ichtchenko
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, 10016, USA
| | - Saul Tzipori
- Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary Medicine, North Grafton, MA, 01536, USA
| | - Charles B Shoemaker
- Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary Medicine, North Grafton, MA, 01536, USA.
| |
Collapse
|
40
|
Li Z, Ma S, Zhang S, Ma Z, Du L, Li M. Degradation of extracellular and membrane proteins in targeted therapy: Status quo and quo vadis. Drug Discov Today 2023; 28:103716. [PMID: 37467880 DOI: 10.1016/j.drudis.2023.103716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 06/29/2023] [Accepted: 07/12/2023] [Indexed: 07/21/2023]
Abstract
Targeted protein degradation (TPD) strategies, such as proteolysis-targeting chimeras (PROTACs) only work for intracellular protein degradation because they involve the intracellular protein degradation machinery. Several new technologies have emerged in recent years for TPD of extracellular and membrane proteins. Even though some progress has been demonstrated in the extracellular and membrane protein degradation field, the application of these technologies is still in its infancy. In this review, we survey the therapeutic potential of existing technologies by summarizing and reviewing discoveries and hurdles in extracellular and membrane protein-of-interest (POI) degradation.
Collapse
Affiliation(s)
- Zhenzhen Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Siyue Ma
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Shuxin Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Zhao Ma
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Lupei Du
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| | - Minyong Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China.
| |
Collapse
|
41
|
De Pauw T, De Mey L, Debacker JM, Raes G, Van Ginderachter JA, De Groof TWM, Devoogdt N. Current status and future expectations of nanobodies in oncology trials. Expert Opin Investig Drugs 2023; 32:705-721. [PMID: 37638538 DOI: 10.1080/13543784.2023.2249814] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/16/2023] [Indexed: 08/29/2023]
Abstract
INTRODUCTION Monoclonal antibodies have revolutionized personalized medicine for cancer in recent decades. Despite their broad application in oncology, their large size and complexity may interfere with successful tumor targeting for certain applications of cancer diagnosis and therapy. Nanobodies have unique structural and pharmacological features compared to monoclonal antibodies and have successfully been used as complementary anti-cancer diagnostic and/or therapeutic tools. AREAS COVERED Here, an overview is given of the nanobody-based diagnostics and therapeutics that have been or are currently being tested in oncological clinical trials. Furthermore, preclinical developments, which are likely to be translated into the clinic in the near future, are highlighted. EXPERT OPINION Overall, the presented studies show the application potential of nanobodies in the field of oncology, making it likely that more nanobodies will be clinically approved in the upcoming future.
Collapse
Affiliation(s)
- Tessa De Pauw
- In vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lynn De Mey
- In vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Jens M Debacker
- In vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Geert Raes
- Cellular and Molecular Immunology Lab, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Jo A Van Ginderachter
- Cellular and Molecular Immunology Lab, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Timo W M De Groof
- In vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Nick Devoogdt
- In vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
42
|
Zhao H, Wang G, Dong H, Wu S, Du Y, Wan B, Ji P, Wu Y, Jiang D, Zhuang G, Duan H, Zhang G, Zhang A. Identification of a Linear B Cell Epitope on p54 of African Swine Fever Virus Using Nanobodies as a Novel Tool. Microbiol Spectr 2023; 11:e0336222. [PMID: 37191526 PMCID: PMC10269858 DOI: 10.1128/spectrum.03362-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 04/14/2023] [Indexed: 05/17/2023] Open
Abstract
African swine fever (ASF) has received great attention from the swine industry due to the pandemic and the lack of vaccines or effective treatments. In the present study, 13 African swine fever virus (ASFV) p54-specific nanobodies (Nbs) were successfully screened based on Bactrian camel immunization of p54 protein and phage display technology, and their reactivity with the p54 C-terminal domain (p54-CTD) was determined; however, only Nb8-horseradish peroxidase (Nb8-HRP) exhibited the best reactivity. Immunoperoxidase monolayer assay (IPMA) and immunofluorescence assay (IFA) results indicated that Nb8-HRP specifically reacted with ASFV-infected cells. Then, the possible epitopes of p54 were identified using Nb8-HRP. The results showed that Nb8-HRP could recognize p54-CTD truncated mutant p54-T1. Then, 6 overlapping peptides covering p54-T1 were synthesized to determine the possible epitopes. Dot blot and peptide-based enzyme-linked immunosorbent assay (ELISA) results suggested that one novel minimal linear B cell epitope, 76QQWVEV81, which had never been reported before, was identified. Alanine-scanning mutagenesis revealed that 76QQWV79 was the core binding site for Nb8. Epitope 76QQWVEV81 was highly conserved among genotype II ASFV strains and could react with inactivated ASFV antibody-positive serum from naturally infected pigs, indicating that it was a natural linear B cell epitope. These findings provide valuable insights for vaccine design and p54 as an effective diagnostic tool. IMPORTANCE The ASFV p54 protein plays an important role in inducing neutralization antibodies in vivo after viral infection and is often used as a candidate protein for subunit vaccine development. The full understanding of the p54 protein epitope provides a sufficient theoretical basis for p54 as a vaccine candidate protein. The present study uses a p54-specific nanobody as a probe to identify a highly conserved antigenic epitope, 76QQWVEV81, among different ASFV strains, and it can induce humoral immune responses in pigs. This is the first report using virus-specific nanobodies as a tool to identify some special epitopes that cannot be recognized by conventional monoclonal antibodies. This study opens up nanobodies as a new tool for identifying epitopes and also provides a theoretical basis for understanding p54-induced neutralizing antibodies.
Collapse
Affiliation(s)
- Huijun Zhao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Henan Engineering Laboratory of Animal Biological Products, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Longhu Modern Immunology Laboratory, Zhengzhou, Henan, China
| | - Gaijie Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Henan Engineering Laboratory of Animal Biological Products, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Longhu Modern Immunology Laboratory, Zhengzhou, Henan, China
| | - Haoxin Dong
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Henan Engineering Laboratory of Animal Biological Products, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Longhu Modern Immunology Laboratory, Zhengzhou, Henan, China
| | - Shuya Wu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Henan Engineering Laboratory of Animal Biological Products, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Longhu Modern Immunology Laboratory, Zhengzhou, Henan, China
| | - Yongkun Du
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Henan Engineering Laboratory of Animal Biological Products, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Longhu Modern Immunology Laboratory, Zhengzhou, Henan, China
| | - Bo Wan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Henan Engineering Laboratory of Animal Biological Products, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Longhu Modern Immunology Laboratory, Zhengzhou, Henan, China
| | - Pengchao Ji
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Henan Engineering Laboratory of Animal Biological Products, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Longhu Modern Immunology Laboratory, Zhengzhou, Henan, China
| | - Yanan Wu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Henan Engineering Laboratory of Animal Biological Products, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Longhu Modern Immunology Laboratory, Zhengzhou, Henan, China
| | - Dawei Jiang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Henan Engineering Laboratory of Animal Biological Products, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Longhu Modern Immunology Laboratory, Zhengzhou, Henan, China
| | - Guoqing Zhuang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Henan Engineering Laboratory of Animal Biological Products, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Longhu Modern Immunology Laboratory, Zhengzhou, Henan, China
| | - Hong Duan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Gaiping Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Henan Engineering Laboratory of Animal Biological Products, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Longhu Modern Immunology Laboratory, Zhengzhou, Henan, China
| | - Angke Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Henan Engineering Laboratory of Animal Biological Products, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Longhu Modern Immunology Laboratory, Zhengzhou, Henan, China
| |
Collapse
|
43
|
Stuart T, Hao S, Zhang B, Mekerishvili L, Landau DA, Maniatis S, Satija R, Raimondi I. Nanobody-tethered transposition enables multifactorial chromatin profiling at single-cell resolution. Nat Biotechnol 2023; 41:806-812. [PMID: 36536150 PMCID: PMC10272075 DOI: 10.1038/s41587-022-01588-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 10/24/2022] [Indexed: 12/24/2022]
Abstract
Chromatin states are functionally defined by a complex combination of histone modifications, transcription factor binding, DNA accessibility and other factors. Current methods for defining chromatin states cannot measure more than one aspect in a single experiment at single-cell resolution. Here we introduce nanobody-tethered transposition followed by sequencing (NTT-seq), an assay capable of measuring the genome-wide presence of up to three histone modifications and protein-DNA binding sites at single-cell resolution. NTT-seq uses recombinant Tn5 transposase fused to a set of secondary nanobodies (nb). Each nb-Tn5 fusion protein specifically binds to different immunoglobulin-G antibodies, enabling a mixture of primary antibodies binding different epitopes to be used in a single experiment. We apply bulk-cell and single-cell NTT-seq to generate high-resolution multimodal maps of chromatin states in cell culture and in human immune cells. We also extend NTT-seq to enable simultaneous profiling of cell surface protein expression and multimodal chromatin states to study cells of the immune system.
Collapse
Affiliation(s)
- Tim Stuart
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Stephanie Hao
- Technology Innovation Lab, New York Genome Center, New York, NY, USA
| | - Bingjie Zhang
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Levan Mekerishvili
- New York Genome Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Dan A Landau
- New York Genome Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Silas Maniatis
- Technology Innovation Lab, New York Genome Center, New York, NY, USA
| | - Rahul Satija
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Ivan Raimondi
- Technology Innovation Lab, New York Genome Center, New York, NY, USA.
- Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
44
|
Alonso Villela SM, Kraïem-Ghezal H, Bouhaouala-Zahar B, Bideaux C, Aceves Lara CA, Fillaudeau L. Production of recombinant scorpion antivenoms in E. coli: current state and perspectives. Appl Microbiol Biotechnol 2023:10.1007/s00253-023-12578-1. [PMID: 37199752 DOI: 10.1007/s00253-023-12578-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/19/2023]
Abstract
Scorpion envenomation is a serious health problem in tropical and subtropical zones. The access to scorpion antivenom is sometimes limited in availability and specificity. The classical production process is cumbersome, from the hyper-immunization of the horses to the IgG digestion and purification of the F(ab)'2 antibody fragments. The production of recombinant antibody fragments in Escherichia coli is a popular trend due to the ability of this microbial host to produce correctly folded proteins. Small recombinant antibody fragments, such as single-chain variable fragments (scFv) and nanobodies (VHH), have been constructed to recognize and neutralize the neurotoxins responsible for the envenomation symptoms in humans. They are the focus of interest of the most recent studies and are proposed as potentially new generation of pharmaceuticals for their use in immunotherapy against scorpion stings of the Buthidae family. This literature review comprises the current status on the scorpion antivenom market and the analyses of cross-reactivity of commercial scorpion anti-serum against non-specific scorpion venoms. Recent studies on the production of new recombinant scFv and nanobodies will be presented, with a focus on the Androctonus and Centruroides scorpion species. Protein engineering-based technology could be the key to obtaining the next generation of therapeutics capable of neutralizing and cross-reacting against several types of scorpion venoms. KEY POINTS: • Commercial antivenoms consist of predominantly purified equine F(ab)'2fragments. • Nanobody-based antivenom can neutralize Androctonus venoms and have a low immunogenicity. • Affinity maturation and directed evolution are used to obtain potent scFv families against Centruroides scorpions.
Collapse
Affiliation(s)
| | - Hazar Kraïem-Ghezal
- Laboratoire Des Venins Et Molécules Thérapeutiques, Institut Pasteur de Tunis, Université de Tunis El Manar, 13 Place Pasteur BP74, 1002, Tunis, Tunisia
| | - Balkiss Bouhaouala-Zahar
- Laboratoire Des Venins Et Molécules Thérapeutiques, Institut Pasteur de Tunis, Université de Tunis El Manar, 13 Place Pasteur BP74, 1002, Tunis, Tunisia.
- Faculté de Médecine de Tunis, Université de Tunis El Manar, Tunis, Tunisia.
| | - Carine Bideaux
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France
| | | | - Luc Fillaudeau
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France
| |
Collapse
|
45
|
Dutt Y, Pandey RP, Dutt M, Gupta A, Vibhuti A, Vidic J, Raj VS, Chang CM, Priyadarshini A. Therapeutic applications of nanobiotechnology. J Nanobiotechnology 2023; 21:148. [PMID: 37149615 PMCID: PMC10163736 DOI: 10.1186/s12951-023-01909-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/24/2023] [Indexed: 05/08/2023] Open
Abstract
Nanobiotechnology, as a novel and more specialized branch of science, has provided a number of nanostructures such as nanoparticles, by utilizing the methods, techniques, and protocols of other branches of science. Due to the unique features and physiobiological characteristics, these nanostructures or nanocarriers have provided vast methods and therapeutic techniques, against microbial infections and cancers and for tissue regeneration, tissue engineering, and immunotherapies, and for gene therapies, through drug delivery systems. However, reduced carrying capacity, abrupt and non-targeted delivery, and solubility of therapeutic agents, can affect the therapeutic applications of these biotechnological products. In this article, we explored and discussed the prominent nanobiotechnological methods and products such as nanocarriers, highlighted the features and challenges associated with these products, and attempted to conclude if available nanostructures offer any scope of improvement or enhancement. We aimed to identify and emphasize the nanobiotechnological methods and products, with greater prospect and capacity for therapeutic improvements and enhancements. We found that novel nanocarriers and nanostructures, such as nanocomposites, micelles, hydrogels, microneedles, and artificial cells, can address the associated challenges and inherited drawbacks, with help of conjugations, sustained and stimuli-responsive release, ligand binding, and targeted delivery. We recommend that nanobiotechnology, despite having few challenges and drawbacks, offers immense opportunities that can be harnessed in delivering quality therapeutics with precision and prediction. We also recommend that, by exploring the branched domains more rigorously, bottlenecks and obstacles can also be addressed and resolved in return.
Collapse
Affiliation(s)
- Yogesh Dutt
- Department of Microbiology, SRM University, 39, Rajiv Gandhi Education City, Post Office P.S. Rai, Sonepat, Haryana 131029 India
| | - Ramendra Pati Pandey
- Department of Microbiology, SRM University, 39, Rajiv Gandhi Education City, Post Office P.S. Rai, Sonepat, Haryana 131029 India
- Department of Biotechnology, SRM University, 39, Rajiv Gandhi Education City, Post Office P.S. Rai, Sonepat, Haryana 131029 India
| | - Mamta Dutt
- Mamta Dental Clinic, Opposite Sector 29, Main Badkhal Road, Faridabad, Haryana 121002 India
| | - Archana Gupta
- Department of Biotechnology, SRM University, 39, Rajiv Gandhi Education City, Post Office P.S. Rai, Sonepat, Haryana 131029 India
| | - Arpana Vibhuti
- Department of Biotechnology, SRM University, 39, Rajiv Gandhi Education City, Post Office P.S. Rai, Sonepat, Haryana 131029 India
| | - Jasmina Vidic
- Université Paris-Saclay, Micalis Institute, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France
| | - V. Samuel Raj
- Department of Microbiology, SRM University, 39, Rajiv Gandhi Education City, Post Office P.S. Rai, Sonepat, Haryana 131029 India
| | - Chung-Ming Chang
- Master & Ph.D Program in Biotechnology Industry, Chang Gung University, No.259, Wenhua 1st Rd., Guishan Dist., Taoyuan City, 33302 Taiwan (ROC)
| | - Anjali Priyadarshini
- Department of Microbiology, SRM University, 39, Rajiv Gandhi Education City, Post Office P.S. Rai, Sonepat, Haryana 131029 India
- Department of Biotechnology, SRM University, 39, Rajiv Gandhi Education City, Post Office P.S. Rai, Sonepat, Haryana 131029 India
| |
Collapse
|
46
|
Hu Y, Zhang C, Lin J, Wang Y, Wu S, Sun Y, Zhang B, Lv H, Ji X, Lu Y, Wang S. Selection of specific nanobodies against peanut allergen through unbiased immunization strategy and the developed immuno-assay. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2022.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
47
|
Contreras MA, Serrano-Rivero Y, González-Pose A, Salazar-Uribe J, Rubio-Carrasquilla M, Soares-Alves M, Parra NC, Camacho-Casanova F, Sánchez-Ramos O, Moreno E. Design and Construction of a Synthetic Nanobody Library: Testing Its Potential with a Single Selection Round Strategy. Molecules 2023; 28:molecules28093708. [PMID: 37175117 PMCID: PMC10180287 DOI: 10.3390/molecules28093708] [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: 03/20/2023] [Revised: 04/18/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023] Open
Abstract
Nanobodies (Nbs) are single domain antibody fragments derived from heavy-chain antibodies found in members of the Camelidae family. They have become a relevant class of biomolecules for many different applications because of several important advantages such as their small size, high solubility and stability, and low production costs. On the other hand, synthetic Nb libraries are emerging as an attractive alternative to animal immunization for the selection of antigen-specific Nbs. Here, we present the design and construction of a new synthetic nanobody library using the phage display technology, following a structure-based approach in which the three hypervariable loops were subjected to position-specific randomization schemes. The constructed library has a clonal diversity of 108 and an amino acid variability that matches the codon distribution set by design at each randomized position. We have explored the capabilities of the new library by selecting nanobodies specific for three antigens: vascular endothelial growth factor (VEGF), tumor necrosis factor (TNF) and the glycoprotein complex (GnGc) of Andes virus. To test the potential of the library to yield a variety of antigen-specific Nbs, we introduced a biopanning strategy consisting of a single selection round using stringent conditions. Using this approach, we obtained several binders for each of the target antigens. The constructed library represents a promising nanobody source for different applications.
Collapse
Affiliation(s)
- María Angélica Contreras
- Pharmacology Department, School of Biological Sciences, University of Concepcion, Concepcion 4070386, Chile
| | | | - Alaín González-Pose
- Faculty of Basic Sciences, University of Medellin, Medellin 050026, Colombia
| | | | | | - Matheus Soares-Alves
- Pharmacology Department, School of Biological Sciences, University of Concepcion, Concepcion 4070386, Chile
| | - Natalie C Parra
- Pharmacology Department, School of Biological Sciences, University of Concepcion, Concepcion 4070386, Chile
| | - Frank Camacho-Casanova
- Pharmacology Department, School of Biological Sciences, University of Concepcion, Concepcion 4070386, Chile
| | - Oliberto Sánchez-Ramos
- Pharmacology Department, School of Biological Sciences, University of Concepcion, Concepcion 4070386, Chile
| | - Ernesto Moreno
- Faculty of Basic Sciences, University of Medellin, Medellin 050026, Colombia
| |
Collapse
|
48
|
Yong Joon Kim J, Sang Z, Xiang Y, Shen Z, Shi Y. Nanobodies: Robust miniprotein binders in biomedicine. Adv Drug Deliv Rev 2023; 195:114726. [PMID: 36754285 PMCID: PMC11725230 DOI: 10.1016/j.addr.2023.114726] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 12/30/2022] [Accepted: 02/02/2023] [Indexed: 02/10/2023]
Abstract
Variable domains of heavy chain-only antibodies (VHH), also known as nanobodies (Nbs), are monomeric antigen-binding domains derived from the camelid heavy chain-only antibodies. Nbs are characterized by small size, high target selectivity, and marked solubility and stability, which collectively facilitate high-quality drug development. In addition, Nbs are readily expressed from various expression systems, including E. coli and yeast cells. For these reasons, Nbs have emerged as preferred antibody fragments for protein engineering, disease diagnosis, and treatment. To date, two Nb-based therapies have been approved by the U.S. Food and Drug Administration (FDA). Numerous candidates spanning a wide spectrum of diseases such as cancer, immune disorders, infectious diseases, and neurodegenerative disorders are under preclinical and clinical investigation. Here, we discuss the structural features of Nbs that allow for specific, versatile, and strong target binding. We also summarize emerging technologies for identification, structural analysis, and humanization of Nbs. Our main focus is to review recent advances in using Nbs as a modular scaffold to facilitate the engineering of multivalent polymers for cutting-edge applications. Finally, we discuss remaining challenges for Nb development and envision new opportunities in Nb-based research.
Collapse
Affiliation(s)
- Jeffrey Yong Joon Kim
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, 1, Gustave L. Levy Pl, New York, NY 10029, USA; Medical Scientist Training Program, University of Pittsburgh School of Medicine and Carnegie Mellon University, Pittsburgh, PA, USA
| | - Zhe Sang
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, 1, Gustave L. Levy Pl, New York, NY 10029, USA
| | - Yufei Xiang
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, 1, Gustave L. Levy Pl, New York, NY 10029, USA
| | - Zhuolun Shen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yi Shi
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, 1, Gustave L. Levy Pl, New York, NY 10029, USA.
| |
Collapse
|
49
|
Lan Z, Li L, Sun Y, Meng X, Shi Z, Du M, Wang H, Sun Z, Cui Q, Wang L, Geng T, Zhou S, Wang Y, Hu F, Duan C, Geng Y, Zhu Y, Dai Y. A procedure for producing an anti-AXL nanobody in E. coli. Protein Expr Purif 2023; 207:106268. [PMID: 37023993 DOI: 10.1016/j.pep.2023.106268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/28/2023] [Accepted: 03/29/2023] [Indexed: 04/07/2023]
Abstract
As one of the receptors of the TAM family, AXL plays a vital role in stem cell maintenance, angiogenesis, immune escape of viruses and drug resistance against tumors. In this study, the truncated extracellular segment containing two immunoglobulin-like domains of human AXL (AXL-IG), which has been confirmed to bind growth arrest specific 6 (GAS6) by structural studies [1], was expressed in a prokaryotic expression system and then purified. Immunizing camelid with the purified AXL-IG as antigen could lead to the production of unique nanobodies composed of only variable domain of heavy chain of heavy-chain antibody (VHH), which are around 15 kD and stable. We screened out a nanobody A-LY01 specific binding to AXL-IG. We further determined the affinity of A-LY01 to AXL-IG and revealed that A-LY01 could specifically recognize full-length AXL on the surface of HEK 293T/17 cells. Our study provides appropriate support for the development of diagnostic reagents and antibody therapeutics targeting AXL.
Collapse
Affiliation(s)
- Zhongyun Lan
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; Laboratory of Quality & Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture, Shanghai, 201306, China; State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Lingyun Li
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yili Sun
- Drug Discovery Shandong Laboratory, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, China
| | - Xiangjing Meng
- Shandong Provincial Key Laboratory of Biopharmaceuticals, Shandong Academy of Pharmaceutical Sciences, Jinan, 250101, China
| | - Zhenzhong Shi
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Mengyang Du
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Hui Wang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zengchao Sun
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Qianqian Cui
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Lu Wang
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Tengjie Geng
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Siyu Zhou
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yi'ang Wang
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fangzheng Hu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Chonggang Duan
- Shandong Provincial Key Laboratory of Biopharmaceuticals, Shandong Academy of Pharmaceutical Sciences, Jinan, 250101, China
| | - Yong Geng
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Yongheng Zhu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; Laboratory of Quality & Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture, Shanghai, 201306, China.
| | - Yuanyuan Dai
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China; National Cancer Center/National Clinical Research Center for Cancer/Hebei Cancer Hospital, Chinese Academy of Medical Sciences, Langfang, 065001, China.
| |
Collapse
|
50
|
Maali A, Gholizadeh M, Feghhi-Najafabadi S, Noei A, Seyed-Motahari SS, Mansoori S, Sharifzadeh Z. Nanobodies in cell-mediated immunotherapy: On the road to fight cancer. Front Immunol 2023; 14:1012841. [PMID: 36761751 PMCID: PMC9905824 DOI: 10.3389/fimmu.2023.1012841] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
The immune system is essential in recognizing and eliminating tumor cells. The unique characteristics of the tumor microenvironment (TME), such as heterogeneity, reduced blood flow, hypoxia, and acidity, can reduce the efficacy of cell-mediated immunity. The primary goal of cancer immunotherapy is to modify the immune cells or the TME to enable the immune system to eliminate malignancies successfully. Nanobodies, known as single-domain antibodies, are light chain-free antibody fragments produced from Camelidae antibodies. The unique properties of nanobodies, including high stability, reduced immunogenicity, enhanced infiltration into the TME of solid tumors and facile genetic engineering have led to their promising application in cell-mediated immunotherapy. They can promote the cancer therapy either directly by bridging between tumor cells and immune cells and by targeting cancer cells using immune cell-bound nanobodies or indirectly by blocking the inhibitory ligands/receptors. The T-cell activation can be engaged through anti-CD3 and anti-4-1BB nanobodies in the bispecific (bispecific T-cell engagers (BiTEs)) and trispecific (trispecific T-cell engager (TriTEs)) manners. Also, nanobodies can be used as natural killer (NK) cell engagers (BiKEs, TriKEs, and TetraKEs) to create an immune synapse between the tumor and NK cells. Nanobodies can redirect immune cells to attack tumor cells through a chimeric antigen receptor (CAR) incorporating a nanobody against the target antigen. Various cancer antigens have been targeted by nanobody-based CAR-T and CAR-NK cells for treating both hematological and solid malignancies. They can also cause the continuation of immune surveillance against tumor cells by stopping inappropriate inhibition of immune checkpoints. Other roles of nanobodies in cell-mediated cancer immunotherapy include reprogramming macrophages to reduce metastasis and angiogenesis, as well as preventing the severe side effects occurring in cell-mediated immunotherapy. Here, we highlight the critical functions of various immune cells, including T cells, NK cells, and macrophages in the TME, and discuss newly developed immunotherapy methods based on the targeted manipulation of immune cells and TME with nanobodies.
Collapse
Affiliation(s)
- Amirhosein Maali
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran,Department of Medical Biotechnology, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Monireh Gholizadeh
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Ahmad Noei
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Seyedeh Sheila Seyed-Motahari
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran,Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Zahra Sharifzadeh
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran,*Correspondence: Zahra Sharifzadeh,
| |
Collapse
|