1
|
Lu H, Sun Y, Chen J, Li J, Chen Y, Long T, She T, Zhu H, Jin Y, Yang H, Lu X, Tao Z. Non-immunoglobin scaffold binders as efficient affinity ligands for purification of broad-spectrum serum albumins. Talanta 2025; 285:127262. [PMID: 39615087 DOI: 10.1016/j.talanta.2024.127262] [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: 09/16/2024] [Revised: 11/16/2024] [Accepted: 11/20/2024] [Indexed: 01/23/2025]
Abstract
Although non-immunoglobin scaffold binders with high affinity and broad spectrum for albumin are attractive for lab-scale albumin purification, affinity chromatography based on these binders has not been developed. Here, the albumin-binding capabilities of representative binders, including protein G-derived albumin binding domain (ABD), albumin binding nanofitins (ABNF), and human serum albumin affimer 31 (HSA31) were predicted by interaction structure analysis and verified by experimental assays. Interaction structure prediction suggested that ABD possessed great potential to bind human (HSA), rhesus monkey (RhSA), mouse (MSA), and rat serum albumin (RSA), whereas ABNF might only bind HSA and bovine serum albumin (BSA), and HSA31 might not bind any of the tested albumins. Subsequent experimental assays demonstrated that ABD was similar to ABNF in HSA and RhSA binding but was superior to ABNF in MSA and RSA binding. ABNF exhibited greater BSA-binding capability than that exhibited by ABD. Both ABD and ABNF showed little binding to rabbit serum albumin (RbSA) and HSA31 showed no binding to all tested albumins. ABD and ABNF could be conjugated to prepackaged NHS-activated HP columns at similar ligand densities. For purification of albumin from plasma, the yields of ABD-HP column for HSA, RhSA, MSA, and RSA were approximately 20 mg, which was 3-4 times higher than that of ABNF-HP column using either Gly-HCl or sodium caprylate as elution buffer. Notably, BSA could only be recovered from plasma using ABNF-HP column with a yield of 5-8 mg. However, little albumin could be purified using HSA31-HP column. These results indicate that both ABD- and ABNF-HP columns could be used as novel tools for purification of broad-spectrum albumins.
Collapse
Affiliation(s)
- Hongyu Lu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yunqing Sun
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jie Chen
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Li
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuzhe Chen
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tingting Long
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Tianshan She
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hong Zhu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Youmei Jin
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hao Yang
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China; Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University, Chengdu, 610041, China; Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaofeng Lu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China; Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University, Chengdu, 610041, China; Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Ze Tao
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China; Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University, Chengdu, 610041, China; Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
2
|
Chen Z, Wang M, Duan W, Xia Y, Liu H, Qian F. Modulating the complement system through epitope-specific inhibition by complement C3 inhibitors. J Biol Chem 2025; 301:108250. [PMID: 39894217 PMCID: PMC11910092 DOI: 10.1016/j.jbc.2025.108250] [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: 07/16/2024] [Revised: 01/01/2025] [Accepted: 01/24/2025] [Indexed: 02/04/2025] Open
Abstract
As an integral part of the innate immune system, the complement system is a tightly regulated proteolytic cascade, playing a critical role in microbial defense, inflammation activation, and dying host cell clearance. Complement proteins are now emerging as subjects of intense research and drug development, since dysregulation of the complement system plays a critical role in several diseases and disorders, such as paroxysmal nocturnal hemoglobinuria (PNH) and geographic atrophy (GA). Within the complement cascade, complement C3 is the central component, situated at the convergence of all complement activation pathways, rendering it an attractive target for complement-related diseases. However, due to the complicated structure-activity relationship (SAR) of C3, elucidating the mechanisms of C3 inhibition on diverse epitopes is the basis for the rational design of C3-targeted therapeutics. Here, we have developed a set of comprehensive biochemical assays that are tailored to the specific steps within the complement cascade, allowing for a thorough understanding of the pharmacological consequences of different C3 inhibitors at each stage. Utilizing three model inhibitors (MIs) with different epitopes, we found that inhibition of MG4/MG5 domains has potent inhibition efficacy across all the complement activation pathways by interrupting C3-C3 convertase interaction, while inhibition of C345C domain displays a bias over the Alternative pathway (AP) inhibition by impairing AP C3 proconvertase formation. This study elucidates the intricate impact of C3 inhibition by targeting different epitopes, offering valuable insights into understanding the mechanism and facilitating the rational design of C3-targeted therapeutics.
Collapse
Affiliation(s)
- Zhidong Chen
- School of Pharmaceutical Sciences, Beijing Frontier Research Center for Biological Structure, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing, PR China
| | - Mingshuang Wang
- School of Pharmaceutical Sciences, Beijing Frontier Research Center for Biological Structure, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing, PR China; Quaerite Biopharm Research Co., Ltd., Beijing, PR China
| | - Wenqian Duan
- School of Pharmaceutical Sciences, Beijing Frontier Research Center for Biological Structure, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing, PR China
| | - Yi Xia
- School of Pharmaceutical Sciences, Beijing Frontier Research Center for Biological Structure, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing, PR China
| | - Huiqin Liu
- Quaerite Biopharm Research Co., Ltd., Beijing, PR China.
| | - Feng Qian
- School of Pharmaceutical Sciences, Beijing Frontier Research Center for Biological Structure, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing, PR China.
| |
Collapse
|
3
|
Kang Q, Li G, Wu Y, Wang S, Chen Z, Zai X, Pan X, Wang R, Lu J, Du P, Yang Z, Chi X, Xiao G, Xu J. A hTfR1 Receptor-Specific VHH Antibody Neutralizes Pseudoviruses Expressing Glycoproteins from Junín and Machupo Viruses. Viruses 2024; 16:1951. [PMID: 39772257 PMCID: PMC11680233 DOI: 10.3390/v16121951] [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: 10/23/2024] [Revised: 12/14/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
The Junín virus (JUNV) is one of the New World arenaviruses that cause severe hemorrhagic fever. Human transferrin receptor 1 (hTfR1) has been identified as the main receptor for JUNV for virus entry into host cells. To date, no treatment has been approved for JUNV. Herein, we investigated 12 anti-hTfR1 VHH (variable domain of the heavy chain of heavy-chain antibody) antibodies and confirmed their interaction with hTfR1. Most of them could bind to the hTfR1 apical domain, which is the glycoprotein 1 (GP1) binding domain of JUNV. Among them, 18N18 exhibited neutralizing activity against both the human immunodeficiency virus (HIV)-vectored lentiviral Junín pseudoviruses and the recombinant vesicular stomatitis virus (VSV)-vectored Junín pseudoviruses. We also verified that 18N18 blocked the interaction between hTfR1 and JUNV GP1. In addition, 18N18 could neutralize another New World arenavirus, the Machupo virus. Using AlphaFold 3-based simulation of 18N18-hTfR1 docking, we determined that 18N18's binding epitope was located at the JUNV GP1 binding epitope. 18N18 represents a candidate for JUNV treatment and provides a potential approach that could be applied to New World arenaviruses.
Collapse
MESH Headings
- Humans
- Junin virus/immunology
- Junin virus/genetics
- Antibodies, Neutralizing/immunology
- Glycoproteins/immunology
- Glycoproteins/genetics
- Glycoproteins/metabolism
- Receptors, Transferrin/immunology
- Receptors, Transferrin/metabolism
- Arenaviruses, New World/immunology
- Arenaviruses, New World/genetics
- Antibodies, Viral/immunology
- Epitopes/immunology
- Animals
- HEK293 Cells
- Antigens, CD/metabolism
- Antigens, CD/genetics
- Antigens, CD/immunology
- Virus Internalization
- Receptors, Virus/metabolism
- Receptors, Virus/immunology
- Viral Envelope Proteins/immunology
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/metabolism
- Single-Domain Antibodies/immunology
- Single-Domain Antibodies/genetics
- Single-Domain Antibodies/metabolism
Collapse
Affiliation(s)
- Qinglin Kang
- School of Medicine, Zhejiang University, Hangzhou 310063, China;
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (G.L.); (S.W.); (Z.C.); (X.Z.); (R.W.); (J.L.); (P.D.); (Z.Y.); (X.C.)
| | - Gege Li
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (G.L.); (S.W.); (Z.C.); (X.Z.); (R.W.); (J.L.); (P.D.); (Z.Y.); (X.C.)
| | - Yan Wu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China; (Y.W.); (X.P.); (G.X.)
| | - Shaoyan Wang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (G.L.); (S.W.); (Z.C.); (X.Z.); (R.W.); (J.L.); (P.D.); (Z.Y.); (X.C.)
| | - Zhengshan Chen
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (G.L.); (S.W.); (Z.C.); (X.Z.); (R.W.); (J.L.); (P.D.); (Z.Y.); (X.C.)
| | - Xiaodong Zai
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (G.L.); (S.W.); (Z.C.); (X.Z.); (R.W.); (J.L.); (P.D.); (Z.Y.); (X.C.)
| | - Xiaoyan Pan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China; (Y.W.); (X.P.); (G.X.)
| | - Rong Wang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (G.L.); (S.W.); (Z.C.); (X.Z.); (R.W.); (J.L.); (P.D.); (Z.Y.); (X.C.)
| | - Jiansheng Lu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (G.L.); (S.W.); (Z.C.); (X.Z.); (R.W.); (J.L.); (P.D.); (Z.Y.); (X.C.)
| | - Peng Du
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (G.L.); (S.W.); (Z.C.); (X.Z.); (R.W.); (J.L.); (P.D.); (Z.Y.); (X.C.)
| | - Zhixin Yang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (G.L.); (S.W.); (Z.C.); (X.Z.); (R.W.); (J.L.); (P.D.); (Z.Y.); (X.C.)
| | - Xiangyang Chi
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (G.L.); (S.W.); (Z.C.); (X.Z.); (R.W.); (J.L.); (P.D.); (Z.Y.); (X.C.)
| | - Gengfu Xiao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China; (Y.W.); (X.P.); (G.X.)
| | - Junjie Xu
- School of Medicine, Zhejiang University, Hangzhou 310063, China;
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (G.L.); (S.W.); (Z.C.); (X.Z.); (R.W.); (J.L.); (P.D.); (Z.Y.); (X.C.)
| |
Collapse
|
4
|
Linciano S, Vigolo E, Rosato A, Kumada Y, Angelini A. Albumin-based strategies to effectively prolong the circulation half-life of small immunomodulatory payloads in cancer therapy. Curr Opin Biotechnol 2024; 90:103218. [PMID: 39481162 DOI: 10.1016/j.copbio.2024.103218] [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: 04/01/2023] [Revised: 08/14/2024] [Accepted: 09/26/2024] [Indexed: 11/02/2024]
Abstract
Small immunomodulatory payloads (IMMs) such as peptide vaccines and cytokines have the capability to activate and boost the immune response against cancer. However, their clinical use has often been hindered by their poor stability and short circulating half-lives. To enhance the pharmacokinetic properties of small IMMs and promote their trafficking and accumulation in lymphatic and tumor tissues, a large variety of strategies have been developed. One of the most successful relies on the use of serum albumin (SA), the most abundant protein in the circulatory and lymphatic system. Here, we report a comparative analysis of the different covalent and noncovalent SA-based strategies applied so far to improve the efficacy of small IMMs in cancer therapy.
Collapse
Affiliation(s)
- Sara Linciano
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, Via Torino 155, 30172 Venice, Italy; Department of Functional Chemistry and Engineering, Kyoto Institute of Technology, 1 Matsugasaki-Hashikami-Cho, Sakyo-ku, Kyoto 606-0951, Japan
| | - Emilia Vigolo
- Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata, 64, 35128 Padua, Italy
| | - Antonio Rosato
- Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata, 64, 35128 Padua, Italy; Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padua, Via Giustiniani 2, 35124 Padua, Italy
| | - Yoichi Kumada
- Department of Functional Chemistry and Engineering, Kyoto Institute of Technology, 1 Matsugasaki-Hashikami-Cho, Sakyo-ku, Kyoto 606-0951, Japan
| | - Alessandro Angelini
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, Via Torino 155, 30172 Venice, Italy; European Centre for Living Technology (ECLT), Ca' Bottacin, Dorsoduro 3911, Calle Crosera, 30123 Venice, Italy.
| |
Collapse
|
5
|
Fridy PC, Rout MP, Ketaren NE. Nanobodies: From High-Throughput Identification to Therapeutic Development. Mol Cell Proteomics 2024; 23:100865. [PMID: 39433212 PMCID: PMC11609455 DOI: 10.1016/j.mcpro.2024.100865] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 10/08/2024] [Accepted: 10/13/2024] [Indexed: 10/23/2024] Open
Abstract
The camelid single-domain antibody fragment, commonly referred to as a nanobody, achieves the targeting power of conventional monoclonal antibodies (mAbs) at only a fraction of their size. Isolated from camelid species (including llamas, alpacas, and camels), their small size at ∼15 kDa, low structural complexity, and high stability compared with conventional antibodies have propelled nanobody technology into the limelight of biologic development. Nanobodies are proving themselves to be a potent complement to traditional mAb therapies, showing success in the treatment of, for example, autoimmune diseases and cancer, and more recently as therapeutic options to treat infectious diseases caused by rapidly evolving biological targets such as the SARS-CoV-2 virus. This review highlights the benefits of applying a proteomic approach to identify diverse nanobody sequences against a single antigen. This proteomic approach coupled with conventional yeast/phage display methods enables the production of highly diverse repertoires of nanobodies able to bind the vast epitope landscape of an antigen, with epitope sampling surpassing that of mAbs. Additionally, we aim to highlight recent findings illuminating the structural attributes of nanobodies that make them particularly amenable to comprehensive antigen sampling and to synergistic activity-underscoring the powerful advantage of acquiring a large, diverse nanobody repertoire against a single antigen. Lastly, we highlight the efforts being made in the clinical development of nanobodies, which have great potential as powerful diagnostic reagents and treatment options, especially when targeting infectious disease agents.
Collapse
Affiliation(s)
- Peter C Fridy
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA
| | - Michael P Rout
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA
| | - Natalia E Ketaren
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA.
| |
Collapse
|
6
|
Xiang Y, Xu J, McGovern BL, Ranzenigo A, Huang W, Sang Z, Shen J, Diaz-Tapia R, Pham ND, Teunissen AJP, Rodriguez ML, Benjamin J, Taylor DJ, van Leent MMT, White KM, García-Sastre A, Zhang P, Shi Y. Adaptive multi-epitope targeting and avidity-enhanced nanobody platform for ultrapotent, durable antiviral therapy. Cell 2024; 187:6966-6980.e23. [PMID: 39447570 PMCID: PMC11748749 DOI: 10.1016/j.cell.2024.09.043] [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: 02/26/2024] [Revised: 07/30/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024]
Abstract
Pathogens constantly evolve and can develop mutations that evade host immunity and treatment. Addressing these escape mechanisms requires targeting evolutionarily conserved vulnerabilities, as mutations in these regions often impose fitness costs. We introduce adaptive multi-epitope targeting with enhanced avidity (AMETA), a modular and multivalent nanobody platform that conjugates potent bispecific nanobodies to a human immunoglobulin M (IgM) scaffold. AMETA can display 20+ nanobodies, enabling superior avidity binding to multiple conserved and neutralizing epitopes. By leveraging multi-epitope SARS-CoV-2 nanobodies and structure-guided design, AMETA constructs exponentially enhance antiviral potency, surpassing monomeric nanobodies by over a million-fold. These constructs demonstrate ultrapotent, broad, and durable efficacy against pathogenic sarbecoviruses, including Omicron sublineages, with robust preclinical results. Structural analysis through cryoelectron microscopy and modeling has uncovered multiple antiviral mechanisms within a single construct. At picomolar to nanomolar concentrations, AMETA efficiently induces inter-spike and inter-virus cross-linking, promoting spike post-fusion and striking viral disarmament. AMETA's modularity enables rapid, cost-effective production and adaptation to evolving pathogens.
Collapse
Affiliation(s)
- Yufei Xiang
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jialu Xu
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Briana L McGovern
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Anna Ranzenigo
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Wei Huang
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Zhe Sang
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Juan Shen
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Randy Diaz-Tapia
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ngoc Dung Pham
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Abraham J P Teunissen
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - M Luis Rodriguez
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jared Benjamin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Derek J Taylor
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Mandy M T van Leent
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kris M White
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Peijun Zhang
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK; Diamond Light Source, Harwell Science and Innovation Campus, Didcot, UK; Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK.
| | - Yi Shi
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
7
|
Fridy PC, Farrell RJ, Molloy KR, Keegan S, Wang J, Jacobs EY, Li Y, Trivedi J, Sehgal V, Fenyö D, Wu Z, Chait BT, Rout MP. A new generation of nanobody research tools using improved mass spectrometry-based discovery methods. J Biol Chem 2024; 300:107623. [PMID: 39098531 PMCID: PMC11401214 DOI: 10.1016/j.jbc.2024.107623] [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: 03/13/2024] [Revised: 07/01/2024] [Accepted: 07/22/2024] [Indexed: 08/06/2024] Open
Abstract
Single-domain antibodies ("nanobodies") derived from the variable region of camelid heavy-chain only antibody variants have proven to be widely useful tools for research, therapeutic, and diagnostic applications. In addition to traditional display techniques, methods to generate nanobodies using direct detection by mass spectrometry and DNA sequencing have been highly effective. However, certain technical challenges have limited widespread application. We have optimized a new pipeline for this approach that greatly improves screening sensitivity, depth of antibody coverage, antigen compatibility, and overall hit rate and affinity. We have applied this improved methodology to generate significantly higher affinity nanobody repertoires against widely used targets in biological research-i.e., GFP, tdTomato, GST, and mouse, rabbit, and goat immunoglobulin G. We have characterized these reagents in affinity isolations and tissue immunofluorescence microscopy, identifying those that are optimal for these particularly demanding applications, and engineering dimeric constructs for ultra-high affinity. This study thus provides new nanobody tools directly applicable to a wide variety of research problems, and improved techniques enabling future nanobody development against diverse targets.
Collapse
Affiliation(s)
- Peter C Fridy
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA
| | - Ryan J Farrell
- Laboratory of Brain Development and Repair, The Rockefeller University, New York, New York, USA; Department of Biochemistry, Weill Cornell Medicine, New York, New York, USA
| | - Kelly R Molloy
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, New York, USA
| | - Sarah Keegan
- Department of Biochemistry and Molecular Pharmacology, Institute for Systems Genetics, NYU Grossman School of Medicine, New York, New York, USA
| | - Junjie Wang
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, New York, USA
| | - Erica Y Jacobs
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, New York, USA; Chemistry Department, St John's University, Queens, New York, USA
| | - Yinyin Li
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, New York, USA
| | - Jill Trivedi
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA
| | - Viren Sehgal
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA
| | - David Fenyö
- Department of Biochemistry and Molecular Pharmacology, Institute for Systems Genetics, NYU Grossman School of Medicine, New York, New York, USA
| | - Zhuhao Wu
- Laboratory of Brain Development and Repair, The Rockefeller University, New York, New York, USA
| | - Brian T Chait
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, New York, USA.
| | - Michael P Rout
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA.
| |
Collapse
|
8
|
Zettl I, Bauernfeind C, Kollárová J, Flicker S. Single-Domain Antibodies-Novel Tools to Study and Treat Allergies. Int J Mol Sci 2024; 25:7602. [PMID: 39062843 PMCID: PMC11277559 DOI: 10.3390/ijms25147602] [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: 06/07/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
IgE-mediated allergies represent a major health problem in the modern world. Apart from allergen-specific immunotherapy (AIT), the only disease-modifying treatment, researchers focus on biologics that target different key molecules such as allergens, IgE, or type 2 cytokines to ameliorate allergic symptoms. Single-domain antibodies, or nanobodies, are the newcomers in biotherapeutics, and their huge potential is being investigated in various research fields since their discovery 30 years ago. While they are dominantly applied for theranostics of cancer and treatment of infectious diseases, nanobodies have become increasingly substantial in allergology over the last decade. In this review, we discuss the prerequisites that we consider to be important for generating useful nanobody-based drug candidates for treating allergies. We further summarize the available research data on nanobodies used as allergen monitoring and detection probes and for therapeutic approaches. We reflect on the limitations that have to be addressed during the development process, such as in vivo half-life and immunogenicity. Finally, we speculate about novel application formats for allergy treatment that might be available in the future.
Collapse
Affiliation(s)
- Ines Zettl
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Clarissa Bauernfeind
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Jessica Kollárová
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Sabine Flicker
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
9
|
Li W, Chen M, Wang T, Feng X, Jiang X, Dong X, Zhang H, Tang X, Tian R, Zhang Y, Li Z. Characterization and humanization of VNARs targeting human serum albumin from the whitespotted bamboo shark (Chiloscyllium plagiosum). Int J Biol Macromol 2024; 273:133082. [PMID: 38878923 DOI: 10.1016/j.ijbiomac.2024.133082] [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: 03/12/2024] [Revised: 05/31/2024] [Accepted: 06/09/2024] [Indexed: 06/18/2024]
Abstract
The Shark-derived immunoglobulin new antigen receptors (IgNARs) have gained increasing attention for their high solubility, exceptional thermal stability, and intricate sequence variation. In this study, we immunized whitespotted bamboo shark (Chiloscyllium plagiosum) to create phage display library of variable domains of IgNAR (VNARs) for screening against Human Serum Albumin (HSA), a versatile vehicle in circulation due to its long in vivo half-life. We identified two HSA-binding VNAR clones, 2G5 and 2G6, and enhanced their expression in E. coli with the FKPA chaperone. 2G6 exhibited a strong binding affinity of 13 nM with HSA and an EC50 of 1 nM. In vivo study with a murine model further provided initial validation of 2G6's ability to prolong circulation time by binding to HSA. Additionally, we employed computational molecular docking to predict the binding affinities of both 2G6 and its humanized derivative, H2G6, to HSA. Our analysis unveiled that the complementarity-determining regions (CDR1 and CDR3) are pivotal in the antigen recognition process. Therefore, our study has advanced the understanding of the potential applications of VNARs in biomedical research aimed at extending drug half-life, holding promise for future therapeutic and diagnostic progressions.
Collapse
Affiliation(s)
- Weijie Li
- State Key Laboratory Breeding Base of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, Fujian 361005, China
| | - Mingliang Chen
- State Key Laboratory Breeding Base of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, Fujian 361005, China; School of Marine Biology, Xiamen Ocean Vocational College, Xiamen, Fujian 361100, China.
| | - Tao Wang
- The Key Laboratory of Urinary Tract Tumors and Calculi, Department of Urology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361003, China
| | - Xin Feng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, Fujian 361102, China
| | - Xierui Jiang
- State Key Laboratory Breeding Base of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, Fujian 361005, China
| | - Xiaoning Dong
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, National Institute for Data Science in Health and Medicine Engineering, Faculty of Medicine and Life Sciences, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Huan Zhang
- State Key Laboratory Breeding Base of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, Fujian 361005, China
| | - Xixiang Tang
- State Key Laboratory Breeding Base of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, Fujian 361005, China.
| | - Rui Tian
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, Fujian 361102, China.
| | - Yongyou Zhang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, National Institute for Data Science in Health and Medicine Engineering, Faculty of Medicine and Life Sciences, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China.
| | - Zengpeng Li
- State Key Laboratory Breeding Base of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, Fujian 361005, China.
| |
Collapse
|
10
|
Wang Y, Chen J, Zhang S, Jiang H, Zhu J, Jiang G, Liu Y, Zhu Y, Li J. Bispecific Nanobody-Aptamer Conjugates for Enhanced Cancer Therapy in Solid Tumors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308265. [PMID: 38225704 DOI: 10.1002/smll.202308265] [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: 09/20/2023] [Revised: 12/30/2023] [Indexed: 01/17/2024]
Abstract
Bispecific antibodies possess exceptional potential as therapeutic agents due to their capacity to bind to two different antigens simultaneously. However, challenges pertain to unsatisfactory stability, manufacturing complexity, and limited tumor penetration hinder their broad applicability. In this study, a versatile technology is presented for the rapid generation of bispecific nanobody-aptamer conjugates with efficient tumor penetration. The approach utilizes microbial transglutaminase (MTGase) and click chemistry to achieve site-specific conjugation of nanobodies and aptamers, which are termed nanotamers. The nanotamers recognize and bind to two types of molecular targets expressed on cancer cells. As a prototype, a bispecific nanotamer is developed that binds both clusters of differentiation 47 (CD47) and mesenchymal epithelial transition receptor (Met) expressed on the tumor cell membrane. This CD47-Met nanotamer demonstrates high affinity and specificity toward tumor cells expressing both targets, exhibits improved receptor functional inhibition through a strong steric hindrance effect. Moreover, its capacity for deep tumor penetration greatly enhances the impact of conventional chemotherapy on antitumor efficacy. The as-developed nanotamer synthesis approach shows promise to customize bispecific molecular probes targeting different cancer types and different therapeutic goals.
Collapse
Affiliation(s)
- Ya Wang
- School of Chemistry and Materials, University of Science and Technology of China, Hefei, 230026, China
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Jie Chen
- School of Chemistry and Materials, University of Science and Technology of China, Hefei, 230026, China
| | - Sen Zhang
- School of Chemistry and Materials, University of Science and Technology of China, Hefei, 230026, China
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Hang Jiang
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Jianqing Zhu
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Guangyi Jiang
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Yichang Liu
- School of Pharmacy, Nantong University, Nantong, 226019, China
| | - Yingdi Zhu
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Juan Li
- School of Chemistry and Materials, University of Science and Technology of China, Hefei, 230026, China
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| |
Collapse
|
11
|
Cavalieri G, Marson D, Giurgevich N, Valeri R, Felluga F, Laurini E, Pricl S. Molecular Ballet: Investigating the Complex Interaction between Self-Assembling Dendrimers and Human Serum Albumin via Computational and Experimental Methods. Pharmaceutics 2024; 16:533. [PMID: 38675194 PMCID: PMC11054399 DOI: 10.3390/pharmaceutics16040533] [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: 03/21/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Dendrimers, intricate macromolecules with highly branched nanostructures, offer unique attributes including precise control over size, shape, and functionality, making them promising candidates for a wide range of biomedical applications. The exploration of their interaction with biological environments, particularly human serum albumin (HSA), holds significant importance for biomedical utilization. In this study, the interaction between HSA and a recently developed self-assembling amphiphilic dendrimer (AD) was investigated using various experimental techniques. Fluorescence spectroscopy and isothermal titration calorimetry revealed moderate interactions between the protein and the AD nanomicelles (NMs), primarily attributed to favorable enthalpic contributions arising from electrostatic interactions and hydrogen bonding. Structural analysis indicated minimal changes in HSA upon complexation with the AD NMs, which was further supported by computational simulations demonstrating stable interactions at the atomistic level. These findings provide valuable insights into the binding mechanisms and thermodynamic parameters governing HSA/AD NM interactions, thereby contributing to the understanding of their potential biomedical applications.
Collapse
Affiliation(s)
- Gabriele Cavalieri
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy; (G.C.); (D.M.); (N.G.); (R.V.); (S.P.)
| | - Domenico Marson
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy; (G.C.); (D.M.); (N.G.); (R.V.); (S.P.)
| | - Nicoletta Giurgevich
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy; (G.C.); (D.M.); (N.G.); (R.V.); (S.P.)
| | - Rachele Valeri
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy; (G.C.); (D.M.); (N.G.); (R.V.); (S.P.)
| | - Fulvia Felluga
- Department of Chemical and Pharmaceutical Sciences, DSCF, University of Trieste, Via Giorgieri 1, 34127 Trieste, Italy;
| | - Erik Laurini
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy; (G.C.); (D.M.); (N.G.); (R.V.); (S.P.)
| | - Sabrina Pricl
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy; (G.C.); (D.M.); (N.G.); (R.V.); (S.P.)
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, ul. Pomorska 141/143, 90-236 Łódź, Poland
| |
Collapse
|
12
|
De Felice S, Romanyuk Z, Chinellato M, Zoia G, Linciano S, Kumada Y, Pardon E, Steyaert J, Angelini A, Cendron L. Crystal structure of human serum albumin in complex with megabody reveals unique human and murine cross-reactive binding site. Protein Sci 2024; 33:e4887. [PMID: 38152025 PMCID: PMC10804666 DOI: 10.1002/pro.4887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/22/2023] [Accepted: 12/22/2023] [Indexed: 12/29/2023]
Abstract
The pharmacokinetic properties of small biotherapeutics can be enhanced via conjugation to cross-reactive albumin-binding ligands in a process that improves their safety and accelerates testing through multiple pre-clinical animal models. In this context, the small and stable heavy-chain-only nanobody NbAlb1, capable of binding both human and murine albumin, has recently been successfully applied to improve the stability and prolong the in vivo plasma residence time of multiple small therapeutic candidates. Despite its clinical efficacy, the mechanism of cross-reactivity of NbAlb1 between human and murine serum albumins has not yet been investigated. To unveil the molecular basis of such an interaction, we solved the crystal structure of human serum albumin (hSA) in complex with NbAlb1. The structure was obtained by harnessing the unique features of a megabody chimeric protein, comprising NbAlb1 grafted onto a modified version of the circularly permutated and bacterial-derived protein HopQ. This structure showed that NbAlb1 contacts a yet unexplored binding site located in the peripheral region of domain II that is conserved in both human and mouse serum albumin proteins. Furthermore, we show that the binding of NbAlb1 to both serum albumin proteins is retained even at acidic pH levels, thus explaining its extended in vivo half-life. The elucidation of the molecular basis of NbAlb1 cross-reactivity to human and murine albumins might guide the design of novel nanobodies with broader reactivity toward a larger panel of serum albumins, thus facilitating the pre-clinical and clinical phases in humans.
Collapse
Affiliation(s)
| | - Zhanna Romanyuk
- Department of Molecular Sciences and NanosystemsCa’ Foscari University of VeniceVeniceItaly
| | | | - Giulia Zoia
- Department of Molecular Sciences and NanosystemsCa’ Foscari University of VeniceVeniceItaly
| | - Sara Linciano
- Department of Molecular Sciences and NanosystemsCa’ Foscari University of VeniceVeniceItaly
| | - Yoichi Kumada
- Department of Functional Chemistry and EngineeringKyoto Institute of TechnologyKyotoJapan
| | - Els Pardon
- VIB‐VUB Center for Structural Biology, VIBBrusselsBelgium
- Structural Biology Brussels, Vrije Universiteit Brussel (VUB)BrusselsBelgium
| | - Jan Steyaert
- VIB‐VUB Center for Structural Biology, VIBBrusselsBelgium
- Structural Biology Brussels, Vrije Universiteit Brussel (VUB)BrusselsBelgium
| | - Alessandro Angelini
- Department of Molecular Sciences and NanosystemsCa’ Foscari University of VeniceVeniceItaly
- European Centre for Living Technology (ECLT), Ca’ BottacinVeniceItaly
| | | |
Collapse
|
13
|
Schriek AI, Aldon YLT, van Gils MJ, de Taeye SW. Next-generation bNAbs for HIV-1 cure strategies. Antiviral Res 2024; 222:105788. [PMID: 38158130 DOI: 10.1016/j.antiviral.2023.105788] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/03/2024]
Abstract
Despite the ability to suppress viral replication using anti-retroviral therapy (ART), HIV-1 remains a global public health problem. Curative strategies for HIV-1 have to target and eradicate latently infected cells across the body, i.e. the viral reservoir. Broadly neutralizing antibodies (bNAbs) targeting the HIV-1 envelope glycoprotein (Env) have the capacity to neutralize virions and bind to infected cells to initiate elimination of these cells. To improve the efficacy of bNAbs in terms of viral suppression and viral reservoir eradication, next generation antibodies (Abs) are being developed that address the current limitations of Ab treatment efficacy; (1) low antigen (Env) density on (reactivated) HIV-1 infected cells, (2) high viral genetic diversity, (3) exhaustion of immune cells and (4) short half-life of Abs. In this review we summarize and discuss preclinical and clinical studies in which anti-HIV-1 Abs demonstrated potent viral control, and describe the development of engineered Abs that could address the limitations described above. Next generation Abs with optimized effector function, avidity, effector cell recruitment and immune cell activation have the potential to contribute to an HIV-1 cure or durable control.
Collapse
Affiliation(s)
- A I Schriek
- Amsterdam UMC Location University of Amsterdam, Department of Medical Microbiology, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands.
| | - Y L T Aldon
- Amsterdam UMC Location University of Amsterdam, Department of Medical Microbiology, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
| | - M J van Gils
- Amsterdam UMC Location University of Amsterdam, Department of Medical Microbiology, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
| | - S W de Taeye
- Amsterdam UMC Location University of Amsterdam, Department of Medical Microbiology, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands.
| |
Collapse
|
14
|
Harmsen MM, Ackerschott B, de Smit H. Serum immunoglobulin or albumin binding single-domain antibodies that enable tailored half-life extension of biologics in multiple animal species. Front Immunol 2024; 15:1346328. [PMID: 38352869 PMCID: PMC10862077 DOI: 10.3389/fimmu.2024.1346328] [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: 11/29/2023] [Accepted: 01/12/2024] [Indexed: 02/16/2024] Open
Abstract
Single-domain antibody fragments (sdAbs) can be isolated from heavy-chain-only antibodies that occur in camelids or the heavy chain of conventional antibodies, that also occur in camelids. Therapeutic application of sdAbs is often complicated by their low serum half-life. Fusion to sdAb that bind to long-lived serum proteins albumin or IgG can prolong serum half-life of fusion partners. Such studies mostly focused on human application. For half-life prolongation in multiple animal species novel species cross-reacting sdAb are needed. We here describe the isolation from immunized llamas of sdAbs G6 and G13 that bound IgG of 9-10 species analysed, including horse, dog, cat, and swine, as well as sdAb A12 that bound horse, dog, swine and cat albumin. A12 bound albumin with 13 to 271 nM affinity dependent on the species. G13 affinity was difficult to determine by biolayer interferometry due to low and heterogeneous signals. G13 and G6 compete for the same binding domain on Fab fragments. Furthermore, they both lack the hallmark residues typical of camelid sdAbs derived from heavy-chain antibodies and had sequence characteristics typical of human sdAbs with high solubility and stability. This suggests they are derived from conventional llama antibodies. They most likely bind IgG through pairing with VL domains at the VH-VL interface rather than a paratope involving complementarity determining regions. None of the isolated sdAb interfered with FcRn binding to albumin or IgG, and thus do not prevent endosomal albumin/IgG-sdAb complex recycling. Fusions of albumin-binding sdAb A12 to several tetanus neurotoxin (TeNT) binding sdAbs prolonged the terminal serum half-life in piglets to about 4 days, comparable to authentic swine albumin. However, G13 conferred a much lower half-life of 0.84 days. Similarly, in horse, G13 prolonged half-life to only 1.2 days whereas A12 fused to two TeNT binding domains (T6T16A12) had a half-life of 21 days. The high half-life of T6T16A12, which earlier proved to be a highly potent TeNT antitoxin, further supports its therapeutic value. Furthermore, we have identified several additional sdAbs that enable tailored half-life extension of biologicals in multiple animal species.
Collapse
Affiliation(s)
- Michiel M. Harmsen
- Wageningen Bioveterinary Research, Wageningen University & Research, Lelystad, Netherlands
| | | | - Hans de Smit
- Research and Development, Smivet B.V., Wijchen, Netherlands
| |
Collapse
|
15
|
Tamburini P, Pedersen DV, Devore D, Cone J, Patel R, Hunter T, Sun F, Andersen GR, Hunter J. Characterization of the bispecific VHH antibody tarperprumig (ALXN1820) specific for properdin and designed for low-volume administration. MAbs 2024; 16:2415060. [PMID: 39397258 PMCID: PMC11485714 DOI: 10.1080/19420862.2024.2415060] [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: 08/12/2024] [Revised: 09/23/2024] [Accepted: 10/06/2024] [Indexed: 10/15/2024] Open
Abstract
The bispecific antibody tarperprumig (ALXN1820) was developed as a treatment option for diseases involving dysregulated complement alternative pathway (AP) activity that could be administered in small volumes, either subcutaneously or intravenously. Tarperprumig incorporates a C-terminal variable domain of a heavy chain only antibody (VHH) that binds properdin (FP) connected via a flexible linker to an N-terminal VHH that binds human serum albumin (HSA). The purified bispecific VHH antibody exhibits an experimental molecular weight average of 27.4 kDa and can be formulated at > 100 mg/mL. Tarperprumig binds tightly to FP and HSA with sub-nanomolar affinity at pH 7.4 and can associate simultaneously with FP and HSA to form a ternary complex. Tarperprumig potently and dose-dependently inhibits to completion in vitro AP-dependent complement C5b-9 formation, AP-dependent hemolysis, and the AP deposition of C3, FP and C9. X-ray crystallography revealed that the isolated FP-binding VHH recognizes the thrombospondin repeat 5 domain of FP, thereby preventing FP from binding to the AP convertase owing to severe steric hindrance. Tarperprumig cross-reacts with cynomolgus monkey FP and serum albumin. In summary, tarperprumig exhibits properties tailored for subcutaneous administration and is currently in clinical development for the treatment of complement AP-related disorders.
Collapse
Affiliation(s)
- Paul Tamburini
- Research and Product Development, Alexion, AstraZeneca Rare Disease, New Haven, CT, USA
| | | | - Denise Devore
- Research and Product Development, Alexion, AstraZeneca Rare Disease, New Haven, CT, USA
| | - Josh Cone
- Research and Product Development, Alexion, AstraZeneca Rare Disease, New Haven, CT, USA
| | - Rekha Patel
- Research and Product Development, Alexion, AstraZeneca Rare Disease, New Haven, CT, USA
| | - Todd Hunter
- Research and Product Development, Alexion, AstraZeneca Rare Disease, New Haven, CT, USA
| | - Fang Sun
- Research and Product Development, Alexion, AstraZeneca Rare Disease, New Haven, CT, USA
| | - Gregers Rom Andersen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Jeffrey Hunter
- Research and Product Development, Alexion, AstraZeneca Rare Disease, New Haven, CT, USA
| |
Collapse
|
16
|
Ullah A, Shin G, Lim SI. Human serum albumin binders: A piggyback ride for long-acting therapeutics. Drug Discov Today 2023; 28:103738. [PMID: 37591409 DOI: 10.1016/j.drudis.2023.103738] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 07/29/2023] [Accepted: 08/10/2023] [Indexed: 08/19/2023]
Abstract
Human serum albumin (HSA) is the most abundant protein in the blood and has desirable properties as a drug carrier. One of the most promising ways to exploit HSA as a carrier is to append an albumin-binding moiety (ABM) to a drug for in situ HSA binding upon administration. Nature- and library-derived ABMs vary in size, affinity, and epitope, differentially improving the pharmacokinetics of an appended drug. In this review, we evaluate the current state of knowledge regarding various aspects of ABMs and the unique advantages of ABM-mediated drug delivery. Furthermore, we discuss how ABMs can be specifically modulated to maximize potential benefits in clinical development.
Collapse
Affiliation(s)
- Aziz Ullah
- Department of Chemical Engineering, Pukyong National University, Busan 48513, Republic of Korea; Gomal Centre of Pharmaceutical Sciences, Faculty of Pharmacy, Gomal University, Dera Ismail Khan 29050, Khyber Pakhtunkhwa, Pakistan
| | - Goeun Shin
- Department of Chemical Engineering, Pukyong National University, Busan 48513, Republic of Korea; Nbios Inc, 7, Jukheon-gil, Gangneung-si, Gangwon-do, Republic of Korea
| | - Sung In Lim
- Department of Chemical Engineering, Pukyong National University, Busan 48513, Republic of Korea; Marine BioResource Co., Ltd., 365, Sinseon-ro, Nam-gu, Busan 48548, Republic of Korea.
| |
Collapse
|
17
|
Esparza TJ, Su S, Francescutti CM, Rodionova E, Kim JH, Brody DL. Enhanced in vivo blood brain barrier transcytosis of macromolecular cargo using an engineered pH-sensitive mouse transferrin receptor binding nanobody. Fluids Barriers CNS 2023; 20:64. [PMID: 37620930 PMCID: PMC10463325 DOI: 10.1186/s12987-023-00462-z] [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: 04/26/2023] [Accepted: 08/10/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND The blood brain barrier limits entry of macromolecular diagnostic and therapeutic cargos. Blood brain barrier transcytosis via receptor mediated transport systems, such as the transferrin receptor, can be used to carry macromolecular cargos with variable efficiency. Transcytosis involves trafficking through acidified intracellular vesicles, but it is not known whether pH-dependent unbinding of transport shuttles can be used to improve blood brain barrier transport efficiency. METHODS A mouse transferrin receptor binding nanobody, NIH-mTfR-M1, was engineered to confer greater unbinding at pH 5.5 vs 7.4 by introducing multiple histidine mutations. The histidine mutant nanobodies were coupled to neurotensin for in vivo functional blood brain barrier transcytosis testing via central neurotensin-mediated hypothermia in wild-type mice. Multi-nanobody constructs including the mutant M1R56H, P96H, Y102H and two copies of the P2X7 receptor-binding 13A7 nanobody were produced to test proof-of-concept macromolecular cargo transport in vivo using quantitatively verified capillary depleted brain lysates and in situ histology. RESULTS The most effective histidine mutant, M1R56H, P96H, Y102H-neurotensin, caused > 8 °C hypothermia after 25 nmol/kg intravenous injection. Levels of the heterotrimeric construct M1R56H, P96H, Y102H-13A7-13A7 in capillary depleted brain lysates peaked at 1 h and were 60% retained at 8 h. A control construct with no brain targets was only 15% retained at 8 h. Addition of the albumin-binding Nb80 nanobody to make M1R56H, P96H, Y102H-13A7-13A7-Nb80 extended blood half-life from 21 min to 2.6 h. At 30-60 min, biotinylated M1R56H, P96H, Y102H-13A7-13A7-Nb80 was visualized in capillaries using in situ histochemistry, whereas at 2-16 h it was detected in diffuse hippocampal and cortical cellular structures. Levels of M1R56H, P96H, Y102H-13A7-13A7-Nb80 reached more than 3.5 percent injected dose/gram of brain tissue after 30 nmol/kg intravenous injection. However, higher injected concentrations did not result in higher brain levels, compatible with saturation and an apparent substrate inhibitory effect. CONCLUSION The pH-sensitive mouse transferrin receptor binding nanobody M1R56H, P96H, Y102H may be a useful tool for rapid and efficient modular transport of diagnostic and therapeutic macromolecular cargos across the blood brain barrier in mouse models. Additional development will be required to determine whether this nanobody-based shuttle system will be useful for imaging and fast-acting therapeutic applications.
Collapse
Affiliation(s)
- Thomas J Esparza
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
| | - Shiran Su
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Elvira Rodionova
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Joong Hee Kim
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
| | - David L Brody
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA.
- Center for Neuroscience and Regenerative Medicine, Bethesda, MD, USA.
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
18
|
Esparza TJ, Su S, Francescutti CM, Rodionova E, Kim JH, Brody DL. Enhanced in Vivo Blood Brain Barrier Transcytosis of Macromolecular Cargo Using an Engineered pH-sensitive Mouse Transferrin Receptor Binding Nanobody. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.26.538462. [PMID: 37333358 PMCID: PMC10274906 DOI: 10.1101/2023.04.26.538462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Background The blood brain barrier limits entry of macromolecular diagnostic and therapeutic cargos. Blood brain barrier transcytosis via receptor mediated transport systems, such as the transferrin receptor, can be used to carry macromolecular cargos with variable efficiency. Transcytosis involves trafficking through acidified intracellular vesicles, but it is not known whether pH-dependent unbinding of transport shuttles can be used to improve blood brain barrier transport efficiency. Methods A mouse transferrin receptor binding nanobody, NIH-mTfR-M1, was engineered to confer greater unbinding at pH 5.5 vs 7.4 by introducing multiple histidine mutations. The histidine mutant nanobodies were coupled to neurotensin for in vivo functional blood brain barrier transcytosis testing via central neurotensin-mediated hypothermia in wild-type mice. Multi-nanobody constructs including the mutant M1 R56H, P96H, Y102H and two copies of the P2X7 receptor-binding 13A7 nanobody were produced to test proof-of-concept macromolecular cargo transport in vivo using quantitatively verified capillary depleted brain lysates and in situ histology. Results The most effective histidine mutant, M1 R56H, P96H, Y102H -neurotensin, caused >8°C hypothermia after 25 nmol/kg intravenous injection. Levels of the heterotrimeric construct M1 56,96,102His -13A7-13A7 in capillary depleted brain lysates peaked at 1 hour and were 60% retained at 8 hours. A control construct with no brain targets was only 15% retained at 8 hours. Addition of the albumin-binding Nb80 nanobody to make M1 R56H, P96H, Y102H -13A7-13A7-Nb80 extended blood half-life from 21 minutes to 2.6 hours. At 30-60 minutes, biotinylated M1 R56H, P96H, Y102H -13A7-13A7-Nb80 was visualized in capillaries using in situ histochemistry, whereas at 2-16 hours it was detected in diffuse hippocampal and cortical cellular structures. Levels of M1 R56H, P96H, Y102H -13A7-13A7-Nb80 reached more than 3.5 percent injected dose/gram of brain tissue after 30 nmol/kg intravenous injection. However, higher injected concentrations did not result in higher brain levels, compatible with saturation and an apparent substrate inhibitory effect. Conclusion The pH-sensitive mouse transferrin receptor binding nanobody M1 R56H, P96H, Y102H may be a useful tool for rapid and efficient modular transport of diagnostic and therapeutic macromolecular cargos across the blood brain barrier in mouse models. Additional development will be required to determine whether this nanobody-based shuttle system will be useful for imaging and fast-acting therapeutic applications.
Collapse
Affiliation(s)
- Thomas J. Esparza
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, United States of America
| | - Shiran Su
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States of America
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, United States of America
| | | | - Elvira Rodionova
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States of America
| | - Joong Hee Kim
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, United States of America
| | - David L. Brody
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States of America
- Center for Neuroscience and Regenerative Medicine, Bethesda, MD, United States of America
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States of America
| |
Collapse
|
19
|
Siepe DH, Picton LK, Garcia KC. Receptor Elimination by E3 Ubiquitin Ligase Recruitment (REULR): A Targeted Protein Degradation Toolbox. ACS Synth Biol 2023; 12:1081-1093. [PMID: 37011906 PMCID: PMC10127277 DOI: 10.1021/acssynbio.2c00587] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Indexed: 04/05/2023]
Abstract
In recent years, targeted protein degradation (TPD) of plasma membrane proteins by hijacking the ubiquitin proteasome system (UPS) or the lysosomal pathway has emerged as a novel therapeutic avenue in drug development to address and inhibit canonically difficult targets. While TPD strategies have been successful in targeting cell surface receptors, these approaches are limited by the availability of suitable binders to generate heterobifunctional molecules. Here, we present the development of a nanobody (VHH)-based degradation toolbox termed REULR (Receptor Elimination by E3 Ubiquitin Ligase Recruitment). We generated human and mouse cross-reactive nanobodies against five transmembrane PA-TM-RING-type E3 ubiquitin ligases (RNF128, RNF130, RNF167, RNF43, and ZNRF3), covering a broad range and selectivity of tissue expression, with which we characterized the expression in human and mouse cell lines and immune cells (PBMCs). We demonstrate that heterobifunctional REULR molecules can enforce transmembrane E3 ligase interactions with a variety of disease-relevant target receptors (EGFR, EPOR, and PD-1) by induced proximity, resulting in effective membrane clearance of the target receptor at varying levels. In addition, we designed E3 ligase self-degrading molecules, "fratricide" REULRs (RNF128, RNF130, RENF167, RNF43, and ZNRF3), that allow downregulation of one or several E3 ligases from the cell surface and consequently modulate receptor signaling strength. REULR molecules represent a VHH-based modular and versatile "mix and match" targeting strategy for the facile modulation of cell surface proteins by induced proximity to transmembrane PA-TM-RING E3 ligases.
Collapse
Affiliation(s)
- Dirk H. Siepe
- Department
of Molecular and Cellular Physiology, Stanford
University School of Medicine, Stanford, California 94305, United States
| | - Lora K. Picton
- Department
of Molecular and Cellular Physiology, Stanford
University School of Medicine, Stanford, California 94305, United States
| | - K. Christopher Garcia
- Department
of Molecular and Cellular Physiology, Stanford
University School of Medicine, Stanford, California 94305, United States
- Department
of Structural Biology, Stanford University
School of Medicine, Stanford, California 94305, United States
- Howard
Hughes Medical Institute, Stanford University
School of Medicine, Stanford, California 94305, United States
| |
Collapse
|
20
|
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
|
21
|
Screening and characterization of inhibitory vNAR targeting nanodisc-assembled influenza M2 proteins. iScience 2022; 26:105736. [PMID: 36570769 PMCID: PMC9771723 DOI: 10.1016/j.isci.2022.105736] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 10/16/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Influenza A virus poses a constant challenge to human health. The highly conserved influenza matrix-2 (M2) protein is an attractive target for the development of a universal antibody-based drug. However, screening using antigens with subphysiological conformation in a nonmembrane environment significantly reduces the generation of efficient antibodies. Here, M2(1-46) was incorporated into nanodiscs (M2-nanodiscs) with M2 in a membrane-embedded tetrameric conformation, closely resembling its natural physiological state in the influenza viral envelope. M2-nanodisc generation, an antigen, was followed by Chiloscyllium plagiosum immunization. The functional vNARs were selected by phage display panning strategy from the shark immune library. One of the isolated vNARs, AM2H10, could specifically bind to tetrameric M2 instead of monomeric M2e (the ectodomain of M2 protein). Furthermore, AM2H10 blocked ion influx through amantadine-sensitive and resistant M2 channels. Our findings indicated the possibility of developing functional shark nanobodies against various influenza viruses by targeting the M2 protein.
Collapse
|
22
|
Shen Z, Sang Z, Shi Y. Nanobodies as a powerful platform for biomedicine. Trends Mol Med 2022; 28:1006-1007. [PMID: 36075813 PMCID: PMC11721352 DOI: 10.1016/j.molmed.2022.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/13/2022] [Accepted: 08/15/2022] [Indexed: 10/31/2022]
Affiliation(s)
- Zhuolun Shen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Zhe Sang
- Center for Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, NY 10029, USA
| | - Yi Shi
- Center for Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, NY 10029, USA.
| |
Collapse
|
23
|
Lou H, Cao X. Antibody variable region engineering for improving cancer immunotherapy. Cancer Commun (Lond) 2022; 42:804-827. [PMID: 35822503 PMCID: PMC9456695 DOI: 10.1002/cac2.12330] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/25/2022] [Accepted: 06/22/2022] [Indexed: 04/09/2023] Open
Abstract
The efficacy and specificity of conventional monoclonal antibody (mAb) drugs in the clinic require further improvement. Currently, the development and application of novel antibody formats for improving cancer immunotherapy have attracted much attention. Variable region-retaining antibody fragments, such as antigen-binding fragment (Fab), single-chain variable fragment (scFv), bispecific antibody, and bi/trispecific cell engagers, are engineered with humanization, multivalent antibody construction, affinity optimization and antibody masking for targeting tumor cells and killer cells to improve antibody-based therapy potency, efficacy and specificity. In this review, we summarize the application of antibody variable region engineering and discuss the future direction of antibody engineering for improving cancer therapies.
Collapse
Affiliation(s)
- Hantao Lou
- Ludwig Institute of Cancer ResearchUniversity of OxfordOxfordOX3 7DRUK
- Chinese Academy for Medical Sciences Oxford InstituteNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
| | - Xuetao Cao
- Chinese Academy for Medical Sciences Oxford InstituteNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
- Department of ImmunologyCentre for Immunotherapy, Institute of Basic Medical SciencesChinese Academy of Medical SciencesBeijing100005P. R. China
| |
Collapse
|
24
|
Lee YCJ, Shirkey JD, Park J, Bisht K, Cowan AJ. An Overview of Antiviral Peptides and Rational Biodesign Considerations. BIODESIGN RESEARCH 2022; 2022:9898241. [PMID: 37850133 PMCID: PMC10521750 DOI: 10.34133/2022/9898241] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/04/2022] [Indexed: 10/19/2023] Open
Abstract
Viral diseases have contributed significantly to worldwide morbidity and mortality throughout history. Despite the existence of therapeutic treatments for many viral infections, antiviral resistance and the threat posed by novel viruses highlight the need for an increased number of effective therapeutics. In addition to small molecule drugs and biologics, antimicrobial peptides (AMPs) represent an emerging class of potential antiviral therapeutics. While AMPs have traditionally been regarded in the context of their antibacterial activities, many AMPs are now known to be antiviral. These antiviral peptides (AVPs) have been shown to target and perturb viral membrane envelopes and inhibit various stages of the viral life cycle, from preattachment inhibition through viral release from infected host cells. Rational design of AMPs has also proven effective in identifying highly active and specific peptides and can aid in the discovery of lead peptides with high therapeutic selectivity. In this review, we highlight AVPs with strong antiviral activity largely curated from a publicly available AMP database. We then compile the sequences present in our AVP database to generate structural predictions of generic AVP motifs. Finally, we cover the rational design approaches available for AVPs taking into account approaches currently used for the rational design of AMPs.
Collapse
Affiliation(s)
- Ying-Chiang J. Lee
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Jaden D. Shirkey
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Jongbeom Park
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Karishma Bisht
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Alexis J. Cowan
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| |
Collapse
|
25
|
Mast FD, Fridy PC, Ketaren NE, Wang J, Jacobs EY, Olivier JP, Sanyal T, Molloy KR, Schmidt F, Rutkowska M, Weisblum Y, Rich LM, Vanderwall ER, Dambrauskas N, Vigdorovich V, Keegan S, Jiler JB, Stein ME, Olinares PDB, Herlands L, Hatziioannou T, Sather DN, Debley JS, Fenyö D, Sali A, Bieniasz PD, Aitchison JD, Chait BT, Rout MP. Highly synergistic combinations of nanobodies that target SARS-CoV-2 and are resistant to escape. eLife 2021; 10:e73027. [PMID: 34874007 PMCID: PMC8651292 DOI: 10.7554/elife.73027] [Citation(s) in RCA: 42] [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: 08/13/2021] [Accepted: 11/07/2021] [Indexed: 02/06/2023] Open
Abstract
The emergence of SARS-CoV-2 variants threatens current vaccines and therapeutic antibodies and urgently demands powerful new therapeutics that can resist viral escape. We therefore generated a large nanobody repertoire to saturate the distinct and highly conserved available epitope space of SARS-CoV-2 spike, including the S1 receptor binding domain, N-terminal domain, and the S2 subunit, to identify new nanobody binding sites that may reflect novel mechanisms of viral neutralization. Structural mapping and functional assays show that indeed these highly stable monovalent nanobodies potently inhibit SARS-CoV-2 infection, display numerous neutralization mechanisms, are effective against emerging variants of concern, and are resistant to mutational escape. Rational combinations of these nanobodies that bind to distinct sites within and between spike subunits exhibit extraordinary synergy and suggest multiple tailored therapeutic and prophylactic strategies.
Collapse
Affiliation(s)
- Fred D Mast
- Center for Global Infectious Disease Research, Seattle Children's Research InstituteSeattleUnited States
| | - Peter C Fridy
- Laboratory of Cellular and Structural Biology, The Rockefeller UniversityNew YorkUnited States
| | - Natalia E Ketaren
- Laboratory of Cellular and Structural Biology, The Rockefeller UniversityNew YorkUnited States
| | - Junjie Wang
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller UniversityNew YorkUnited States
| | - Erica Y Jacobs
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller UniversityNew YorkUnited States
- Department of Chemistry, St. John’s UniversityQueensUnited States
| | - Jean Paul Olivier
- Center for Global Infectious Disease Research, Seattle Children's Research InstituteSeattleUnited States
| | - Tanmoy Sanyal
- Department of Bioengineering and Therapeutic Sciences, Department of Pharmaceutical Chemistry, California Institute for Quantitative Biosciences, University of California, San FranciscoSan FranciscoUnited States
| | - Kelly R Molloy
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller UniversityNew YorkUnited States
| | - Fabian Schmidt
- Laboratory of Retrovirology, The Rockefeller UniversityNew YorkUnited States
| | - Magdalena Rutkowska
- Laboratory of Retrovirology, The Rockefeller UniversityNew YorkUnited States
| | - Yiska Weisblum
- Laboratory of Retrovirology, The Rockefeller UniversityNew YorkUnited States
| | - Lucille M Rich
- Center for Immunity and Immunotherapies, Seattle Children’s Research InstituteSeattleUnited States
| | - Elizabeth R Vanderwall
- Center for Immunity and Immunotherapies, Seattle Children’s Research InstituteSeattleUnited States
| | - Nicholas Dambrauskas
- Center for Global Infectious Disease Research, Seattle Children's Research InstituteSeattleUnited States
| | - Vladimir Vigdorovich
- Center for Global Infectious Disease Research, Seattle Children's Research InstituteSeattleUnited States
| | - Sarah Keegan
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of MedicineNew YorkUnited States
| | - Jacob B Jiler
- Laboratory of Cellular and Structural Biology, The Rockefeller UniversityNew YorkUnited States
| | - Milana E Stein
- Laboratory of Cellular and Structural Biology, The Rockefeller UniversityNew YorkUnited States
| | - Paul Dominic B Olinares
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller UniversityNew YorkUnited States
| | | | | | - D Noah Sather
- Center for Global Infectious Disease Research, Seattle Children's Research InstituteSeattleUnited States
- Department of Pediatrics, University of WashingtonSeattleUnited States
| | - Jason S Debley
- Center for Immunity and Immunotherapies, Seattle Children’s Research InstituteSeattleUnited States
- Department of Pediatrics, University of WashingtonSeattleUnited States
- Division of Pulmonary and Sleep Medicine, Seattle Children’s HospitalSeattleUnited States
| | - David Fenyö
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of MedicineNew YorkUnited States
| | - Andrej Sali
- Department of Bioengineering and Therapeutic Sciences, Department of Pharmaceutical Chemistry, California Institute for Quantitative Biosciences, University of California, San FranciscoSan FranciscoUnited States
| | - Paul D Bieniasz
- Laboratory of Retrovirology, The Rockefeller UniversityNew YorkUnited States
- Howard Hughes Medical Institute, The Rockefeller UniversityNew YorkUnited States
| | - John D Aitchison
- Center for Global Infectious Disease Research, Seattle Children's Research InstituteSeattleUnited States
- Department of Pediatrics, University of WashingtonSeattleUnited States
- Department of Biochemistry, University of WashingtonSeattleUnited States
| | - Brian T Chait
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller UniversityNew YorkUnited States
| | - Michael P Rout
- Laboratory of Cellular and Structural Biology, The Rockefeller UniversityNew YorkUnited States
| |
Collapse
|