1
|
Jitender J, Hong T, Sherman A, Wong P, Aniogo E, Kujawski M, Shively JE, Yazaki PJ. Development of Anti-CEA C H2 Domain-Deleted Antibody (M5A∆C H2) for the PET Imaging of Colorectal Cancer. Mol Imaging Biol 2025; 27:192-200. [PMID: 40085171 PMCID: PMC12062029 DOI: 10.1007/s11307-025-01997-3] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
PURPOSE Recombinant antibody fragments represent a novel class of in vivo biological immunoPET imaging agents. This study developed a series of anti-carcinoembryonic antigen (CEA) CH2 domain-deleted antibodies to evaluate their rapid, high-level tumor targeting combined with fast blood clearance for immunoPET imaging in two colorectal cancer mouse models. PROCEDURE A series of humanized anti-CEA M5A∆CH2 recombinant antibody fragments were synthesized via transient mammalian expression and purified using a two-step process. The M5A∆CH2 antibody series was characterized by HPLC-SEC, SDS-PAGE and binding affinities. The M5A∆CH2-C5 antibody, which has five disulfide bridges in the modified IgG1/IgG3 hinge domain, was selected for positron emission tomography (PET) imaging. Site-specific thiol conjugation of the reduced hinge disulfides with the 1,4,7,10 tetraazacyclododecane-1,4,7-triacetic acid trisodium salt-vinyl sulfone (DO3A-VS) chelate was performed, followed by labeling with [64Cu-CuCl2]. The [64Cu]Cu-DO3A-M5A∆CH2-C5 was evaluated for CEA-positive tumor PET imaging at serial time points, pharmacokinetics and a terminal biodistribution study conducted in two CEA-positive colorectal cancer mouse models. RESULTS The anti-CEA M5A∆CH2 antibodies had high expression, were purified using a new CH3 domain affinity resin and were stable up to one year. ImmunoPET imaging and biodistribution studies were performed in athymic mice bearing human colorectal cancer LS174T tumors and immunocompetent transgenic-CEA (Tg-CEA) mice bearing MC-38 tumors transfected with the human CEA gene. The [64Cu]Cu-DO3A-M5A∆CH2-C5 showed rapid, high tumor localization and the expected fast blood clearance. CONCLUSIONS A series of humanized anti-CEA M5A∆CH2 antibodies were designed for immunoPET imaging of colorectal cancer, and the [64Cu]Cu-DO3A-M5A∆CH2-C5 showed high tumor targeting and fast blood clearance supporting its potential for clinical trials.
Collapse
Affiliation(s)
- Jitender Jitender
- Department of Immunology and Theranostics, Beckman Research Institute, City of Hope, USA
| | - Teresa Hong
- Department of Immunology and Theranostics, Beckman Research Institute, City of Hope, USA
| | - Anakim Sherman
- Department of Immunology and Theranostics, Beckman Research Institute, City of Hope, USA
| | - Patty Wong
- Department of Immunology and Theranostics, Beckman Research Institute, City of Hope, USA
| | - Eric Aniogo
- Department of Immunology and Theranostics, Beckman Research Institute, City of Hope, USA
| | - Maciej Kujawski
- Department of Immunology and Theranostics, Beckman Research Institute, City of Hope, USA
| | - John E Shively
- Department of Immunology and Theranostics, Beckman Research Institute, City of Hope, USA
| | - Paul J Yazaki
- Department of Immunology and Theranostics, Beckman Research Institute, City of Hope, USA.
| |
Collapse
|
2
|
Pang G, Wang R, Yang H, Chai M, Gao Y, Chen S, Mao T, Du L, Lan Y, Li S, Xu J, Cui P, Cheng R, Huang Y, Wang X, Yang Y. A synthetic heavy chain variable domain antibody library (VHL) provides highly functional antibodies with favorable developability. Protein Sci 2025; 34:e70090. [PMID: 40100169 PMCID: PMC11917115 DOI: 10.1002/pro.70090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/20/2025]
Abstract
Synthetic antibody libraries have been developed as an efficient source for the discovery of the heavy chain variable (VH) domain, which exhibits low immunogenicity, high tissue penetration, and diverse binding epitopes in therapeutic biopharmaceuticals. In this study, the human IGHV3-23*04 germline gene was chosen as the scaffold with a high expression level and favorable thermal stability. Amino acid diversity was introduced into the complementarity determining region 3 (CDR3) to exclude potential sequence liabilities. A library containing 2.6 × 1011 independent clones was successfully constructed. The receptor-binding domain (RBD) of the SARS-CoV-2 spike protein, interleukin-17A (IL17A), B-cell maturation antigen (BCMA), and G-protein coupled receptor family C group 5 member D (GPRC5D) were used as target antigens to screen and identify VHs. In each case, Thirty-one to fifty-five VHs were screened out. The VH-Fc antibodies showed superior affinities (as high as 4.6 nM) to the corresponding antigens but did not bind to antigen-irrelevant cell CHO-S. Furthermore, the anti-RBD and anti-IL17A VH-Fc antibodies showed strong functional activity in the receptor-blocking assays. The VH-Fc antibodies from the synthetic library exhibited favorable developability (thermal stability, colloidal stability, hydrophilicity, anti-aggregation ability, and no interaction with human IgGs). We demonstrated that high-affinity and highly functional VH domain antibodies were generated from the rationally designed library with desired physicochemical properties. This approach is generally universal to target any antigen and has significant potential to accelerate candidate selection.
Collapse
Affiliation(s)
- Guiying Pang
- College of PharmacyAnhui University of Traditional Chinese MedicineHefeiPeople's Republic of China
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd.BeijingPeople's Republic of China
- Joint Graduate SchoolYangtze Delta Drug Advanced Research InstituteNantongPeople's Republic of China
| | - Ruixue Wang
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd.BeijingPeople's Republic of China
| | - Hongxu Yang
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd.BeijingPeople's Republic of China
| | - Mengya Chai
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd.BeijingPeople's Republic of China
| | - Yanzhe Gao
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd.BeijingPeople's Republic of China
| | - Sisi Chen
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd.BeijingPeople's Republic of China
| | - Ting Mao
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd.BeijingPeople's Republic of China
| | - Luheng Du
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd.BeijingPeople's Republic of China
| | - Yujia Lan
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd.BeijingPeople's Republic of China
| | - Shu Li
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd.BeijingPeople's Republic of China
| | - Jiale Xu
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd.BeijingPeople's Republic of China
| | - Panpan Cui
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd.BeijingPeople's Republic of China
| | - Ruqing Cheng
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd.BeijingPeople's Republic of China
| | - Yuxin Huang
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd.BeijingPeople's Republic of China
| | - Xuncui Wang
- College of PharmacyAnhui University of Traditional Chinese MedicineHefeiPeople's Republic of China
| | - Yi Yang
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd.BeijingPeople's Republic of China
- Joint Graduate SchoolYangtze Delta Drug Advanced Research InstituteNantongPeople's Republic of China
| |
Collapse
|
3
|
Lee NJ, Jung M, Yang HY, Shim H. A single-domain antibody library based on a stability-engineered human VH3 scaffold. Sci Rep 2024; 14:17747. [PMID: 39085444 PMCID: PMC11291719 DOI: 10.1038/s41598-024-68680-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024] Open
Abstract
Using conventional immunoglobulin G (IgG) molecules as therapeutic agents presents several well-known disadvantages owing to their large size and structural complexity, negatively impacting development and production efficiency. Single-domain antibodies (sdAbs) are the smallest functional antibody format (~ 15 kDa) and represent a viable alternative to IgG in many applications. However, unlike natural single-domain antibodies, such as camelid VHH, the variable domains of conventional antibodies show poor physicochemical properties when expressed as sdAbs. This report identified stable sdAb variants of human VH3-23 from a framework region 2-randomized human VH library by phage display selection under thermal challenge. Synthetic complementarity determining region diversity was introduced to one of the selected variants with high thermal stability, expression level, and monomeric content to construct a human VH sdAb library. The library was validated by panning against a panel of antigens, and target-specific binders were identified and characterized for their affinity and biophysical properties. The results of this study suggest that a synthetic sdAb library based on a stability-engineered human VH scaffold could be a facile source of high-quality sdAb for many practical applications.
Collapse
Affiliation(s)
- Nam Ju Lee
- Department of Bioinspired Sciences, Ewha Womans University, Seoul, Korea
| | - Mooyoung Jung
- Department of Bioinspired Sciences, Ewha Womans University, Seoul, Korea
| | - Hye Young Yang
- Department of Life Sciences, Ewha Womans University, Seoul, Korea
| | - Hyunbo Shim
- Department of Bioinspired Sciences, Ewha Womans University, Seoul, Korea.
- Department of Life Sciences, Ewha Womans University, Seoul, Korea.
| |
Collapse
|
4
|
Tripathy RK, Pande AH. Molecular and functional insight into anti-EGFR nanobody: Theranostic implications for malignancies. Life Sci 2024; 345:122593. [PMID: 38554946 DOI: 10.1016/j.lfs.2024.122593] [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: 01/09/2024] [Revised: 02/27/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
Targeted therapy and imaging are the most popular techniques for the intervention and diagnosis of cancer. A potential therapeutic target for the treatment of cancer is the epidermal growth factor receptor (EGFR), primarily for glioblastoma, lung, and breast cancer. Over-production of ligand, transcriptional up-regulation due to autocrine/paracrine signalling, or point mutations at the genomic locus may contribute to the malfunction of EGFR in malignancies. This exploit makes use of EGFR, an established biomarker for cancer diagnostics and treatment. Despite considerable development in the last several decades in making EGFR inhibitors, they are still not free from limitations like toxicity and a short serum half-life. Nanobodies and antibodies share similar binding properties, but nanobodies have the additional advantage that they can bind to antigenic epitopes deep inside the target that conventional antibodies are unable to access. For targeted therapy, anti-EGFR nanobodies can be conjugated to various molecules such as drugs, peptides, toxins and photosensitizers. These nanobodies can be designed as novel immunoconjugates using the universal modular antibody-based platform technology (UniCAR). Furthermore, Anti-EGFR nanobodies can be expressed in neural stem cells and visualised by effective fluorescent and radioisotope labelling.
Collapse
Affiliation(s)
- Rajan K Tripathy
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, (Mohali) 160062, Punjab, India
| | - Abhay H Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, (Mohali) 160062, Punjab, India.
| |
Collapse
|
5
|
Chen L, Sun M, Zhang H, Zhang X, Yao Y, Li M, Li K, Fan P, Zhang H, Qin Y, Zhang Z, Li E, Chen Z, Guan W, Li S, Yu C, Zhang K, Gong R, Chiu S. Potent human neutralizing antibodies against Nipah virus derived from two ancestral antibody heavy chains. Nat Commun 2024; 15:2987. [PMID: 38582870 PMCID: PMC10998907 DOI: 10.1038/s41467-024-47213-8] [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: 08/14/2023] [Accepted: 03/18/2024] [Indexed: 04/08/2024] Open
Abstract
Nipah virus (NiV) is a World Health Organization priority pathogen and there are currently no approved drugs for clinical immunotherapy. Through the use of a naïve human phage-displayed Fab library, two neutralizing antibodies (NiV41 and NiV42) targeting the NiV receptor binding protein (RBP) were identified. Following affinity maturation, antibodies derived from NiV41 display cross-reactivity against both NiV and Hendra virus (HeV), whereas the antibody based on NiV42 is only specific to NiV. Results of immunogenetic analysis reveal a correlation between the maturation of antibodies and their antiviral activity. In vivo testing of NiV41 and its mature form (41-6) show protective efficacy against a lethal NiV challenge in hamsters. Furthermore, a 2.88 Å Cryo-EM structure of the tetrameric RBP and antibody complex demonstrates that 41-6 blocks the receptor binding interface. These findings can be beneficial for the development of antiviral drugs and the design of vaccines with broad spectrum against henipaviruses.
Collapse
Affiliation(s)
- Li Chen
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mengmeng Sun
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Huajun Zhang
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Xinghai Zhang
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Yanfeng Yao
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Ming Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Kangyin Li
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Pengfei Fan
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Haiwei Zhang
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Ye Qin
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhe Zhang
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Entao Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China
| | - Zhen Chen
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Wuxiang Guan
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Shanshan Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Changming Yu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China.
| | - Kaiming Zhang
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China.
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, MOE Key Laboratory for Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
- Department of Urology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| | - Rui Gong
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Hubei Jiangxia Laboratory, Wuhan, Hubei, China.
| | - Sandra Chiu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China.
| |
Collapse
|
6
|
Ma X, Wang M, Ying T, Wu Y. Reforming solid tumor treatment: the emerging potential of smaller format antibody-drug conjugate. Antib Ther 2024; 7:114-122. [PMID: 38566971 PMCID: PMC10983081 DOI: 10.1093/abt/tbae005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/23/2024] [Accepted: 02/07/2024] [Indexed: 04/04/2024] Open
Abstract
In recent years, substantial therapeutic efficacy of antibody-drug conjugates (ADCs) has been validated through approvals of 16 ADCs for the treatment of malignant tumors. However, realization of the maximum clinical use of ADCs requires surmounting extant challenges, mainly the limitations in tumor penetration capabilities when targeting solid tumors. To resolve the hurdle of suboptimal tumor penetration, miniaturized antibody fragments with engineered formats have been harnessed for ADC assembly. By virtue of their reduced molecular sizes, antibody fragment-drug conjugates hold considerable promise for efficacious delivery of cytotoxic agents, thus conferring superior therapeutic outcomes. This review will focus on current advancements in novel ADC development utilizing smaller antibody formats from ~6 to 80 kDa, with particular emphasis on single-domain antibodies, which have been widely applied in novel ADC design. Additionally, strategies to optimize clinical translation are discussed, including half-life extension, acceleration of internalization, and reduction of immunogenic potential.
Collapse
Affiliation(s)
- Xiaojie Ma
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Mingkai Wang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Tianlei Ying
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center for Synthetic Immunology, Fudan University, Shanghai 200032, China
| | - Yanling Wu
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center for Synthetic Immunology, Fudan University, Shanghai 200032, China
| |
Collapse
|
7
|
Yamamoto M, Funada R, Taki R, Shiroishi M. Production and characterization of an Fv-clasp of rheumatoid factor, a low-affinity human autoantibody. J Biochem 2024; 175:387-394. [PMID: 38102734 DOI: 10.1093/jb/mvad112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/23/2023] [Indexed: 12/17/2023] Open
Abstract
Rheumatoid factor (RF) is an autoantibody against IgG that affects autoimmune diseases and inhibits the effectiveness of pharmaceuticals and diagnostic agents. Although RFs derived from various germline genes have been identified, little is known about their molecular recognition mechanisms. In this study, the Fv-clasp format was used to prepare YES8c, an RF. We developed an Escherichia coli secretion expression system capable of producing milligram-scale of YES8c Fv-clasp per 1 L of culture. Although YES8c is an autoantibody with very low affinity, the produced Fv-clasp maintained specific binding to IgG. Interestingly, the molecules prepared by E. coli secretion had a higher affinity than those prepared by refolding. In the structure of the YES8c-Fc complex, the N-terminus of the light chain is close to Fc; therefore, it is suggested that the addition of the N-terminal methionine may cause collisions with Fc, resulting in reduced affinity. Our findings suggest that the Fv-clasp, which provides sufficient stability and a high bacterial yield, is a useful format for studying RFs with very low affinity. Furthermore, the Fv-clasp produced from a secretion expression system, which can properly process the N-terminus, would be suitable for analysis of RFs in which the N-terminus may be involved in interactions.
Collapse
Affiliation(s)
- Maruto Yamamoto
- Department of Biological Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, 125-8585, Japan
| | - Ryoma Funada
- Department of Biological Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, 125-8585, Japan
| | - Ryota Taki
- Department of Biological Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, 125-8585, Japan
| | - Mitsunori Shiroishi
- Department of Biological Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, 125-8585, Japan
| |
Collapse
|
8
|
Zhu XY, Li QX, Kong Y, Huang KK, Wang G, Wang YJ, Lu J, Hua GQ, Wu YL, Ying TL. A novel human single-domain antibody-drug conjugate targeting CEACAM5 exhibits potent in vitro and in vivo antitumor activity. Acta Pharmacol Sin 2024; 45:609-618. [PMID: 38030799 PMCID: PMC10834580 DOI: 10.1038/s41401-023-01200-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 11/13/2023] [Indexed: 12/01/2023]
Abstract
Leveraging the specificity of antibody to deliver cytotoxic agent into tumor, antibody-drug conjugates (ADCs) have become one of the hotspots in the development of anticancer therapies. Although significant progress has been achieved, there remain challenges to overcome, including limited penetration into solid tumors and potential immunogenicity. Fully human single-domain antibodies (UdAbs), with their small size and human nature, represent a promising approach for addressing these challenges. Carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) is a glycosylated cell surface protein that rarely expressed in normal adult tissues but overexpressed in diverse cancers, taking part in tumorigenesis, progression, and metastasis. In this study, we investigated the therapeutic potential of UdADC targeting CEACAM5. We performed biopanning in our library and obtained an antibody candidate B9, which bound potently and specifically to CEACAM5 protein (KD = 4.84 nM) and possessed excellent biophysical properties (low aggregation tendency, high homogeneity, and thermal stability). The conjugation of B9 with a potent cytotoxic agent, monomethyl auristatin E (MMAE), exhibited superior antitumor efficacy against CEACAM5-expressing human gastric cancer cell line MKN-45, human pancreatic carcinoma cell line BxPC-3 and human colorectal cancer cell line LS174T with IC50 values of 38.14, 25.60, and 101.4 nM, respectively. In BxPC-3 and MKN-45 xenograft mice, administration of UdADC B9-MMAE (5 mg/kg, i.v.) every 2 days for 4 times markedly inhibited the tumor growth without significant change in body weight. This study may have significant implications for the design of next-generation ADCs.
Collapse
Affiliation(s)
- Xiao-Yi Zhu
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, China
| | - Quan-Xiao Li
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, China
| | - Yu Kong
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, China
| | - Ke-Ke Huang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, China
| | - Gang Wang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yun-Ji Wang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jun Lu
- Auckland Bioengineering Institute, University of Auckland, Auckland, 1010, New Zealand
| | - Guo-Qiang Hua
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Yan-Ling Wu
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, China.
| | - Tian-Lei Ying
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, China.
| |
Collapse
|
9
|
Hao Y, Yan J, Fraser C, Jiang A, Anuganti M, Zhang R, Lloyd K, Jardine J, Coppola J, Meijers R, Li J, Springer TA. Synthetic integrin antibodies discovered by yeast display reveal αV subunit pairing preferences with β subunits. MAbs 2024; 16:2365891. [PMID: 38889315 PMCID: PMC11188837 DOI: 10.1080/19420862.2024.2365891] [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: 02/16/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024] Open
Abstract
Integrins are cell surface receptors that mediate the interactions of cells with their surroundings and play essential roles in cell adhesion, migration, and homeostasis. Eight of the 24 integrins bind to the tripeptide Arg-Gly-Asp (RGD) motif in their extracellular ligands, comprising the RGD-binding integrin subfamily. Despite similarity in recognizing the RGD motif and some redundancy, these integrins can selectively recognize RGD-containing ligands to fulfill specific functions in cellular processes. Antibodies against individual RGD-binding integrins are desirable for investigating their specific functions, and were selected here from a synthetic yeast-displayed Fab library. We discovered 11 antibodies that exhibit high specificity and affinity toward their target integrins, i.e. αVβ3, αVβ5, αVβ6, αVβ8, and α5β1. Of these, six are function-blocking antibodies and contain a ligand-mimetic R(G/L/T)D motif in their CDR3 sequences. We report antibody-binding specificity, kinetics, and binding affinity for purified integrin ectodomains, as well as intact integrins on the cell surface. We further used these antibodies to reveal binding preferences of the αV subunit for its 5 β-subunit partners: β6 = β8 > β3 > β1 = β5.
Collapse
Affiliation(s)
- Yuxin Hao
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Jiabin Yan
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Courtney Fraser
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
| | - Aiping Jiang
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | | | - Roushu Zhang
- Institute for Protein Innovation, Boston, MA, USA
| | | | | | | | - Rob Meijers
- Institute for Protein Innovation, Boston, MA, USA
| | - Jing Li
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Timothy A. Springer
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Aertker KM, Pilvankar MR, Prass TM, Blech M, Higel F, Kasturirangan S. Exploring molecular determinants and pharmacokinetic properties of IgG1-scFv bispecific antibodies. MAbs 2024; 16:2318817. [PMID: 38444390 PMCID: PMC10936634 DOI: 10.1080/19420862.2024.2318817] [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: 11/29/2023] [Accepted: 02/09/2024] [Indexed: 03/07/2024] Open
Abstract
Bispecific antibodies (BsAbs) capable of recognizing two distinct epitopes or antigens offer promising therapeutic options for various diseases by targeting multiple pathways. The favorable pharmacokinetic (PK) properties of monoclonal antibodies (mAbs) are crucial, as they directly influence patient safety and therapeutic efficacy. For numerous mAb therapeutics, optimization of neonatal Fc receptor (FcRn) interactions and elimination of unfavorable molecular properties have led to improved PK properties. However, many BsAbs exhibit unfavorable PK, which has precluded their development as drugs. In this report, we present studies on the molecular determinants underlying the distinct PK profiles of three IgG1-scFv BsAbs. Our study indicated that high levels of nonspecific interactions, elevated isoelectric point (pI), and increased number of positively charged patches contributed to the fast clearance of IgG1-scFv. FcRn chromatography results revealed specific scFv-FcRn interactions that are unique to the IgG1-scFv, which was further supported by molecular dynamics (MD) simulation. These interactions likely stabilize the BsAb FcRn interaction at physiological pH, which in turn could disrupt FcRn-mediated BsAb recycling. In addition to the empirical observations, we also evaluated the impact of in silico properties, including pI differential between the Fab and scFv and the ratio of dipole moment to hydrophobic moment (RM) and their correlation with the observed clearance. These findings highlight that the PK properties of BsAbs may be governed by novel determinants, owing to their increased structural complexity compared to immunoglobulin G (IgG) 1 antibodies.
Collapse
Affiliation(s)
- Kristina M.J. Aertker
- Analytical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | | | - Tobias M. Prass
- Center for Theoretical Chemistry, Ruhr University Bochum, Bochum, Germany
| | - Michaela Blech
- Analytical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Fabian Higel
- Global CMC Experts NBE, Global Quality Development, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Srinath Kasturirangan
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, CT, USA
| |
Collapse
|
11
|
Amash A, Volkers G, Farber P, Griffin D, Davison KS, Goodman A, Tonikian R, Yamniuk A, Barnhart B, Jacobs T. Developability considerations for bispecific and multispecific antibodies. MAbs 2024; 16:2394229. [PMID: 39189686 DOI: 10.1080/19420862.2024.2394229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/08/2024] [Accepted: 08/15/2024] [Indexed: 08/28/2024] Open
Abstract
Bispecific antibodies (bsAb) and multispecific antibodies (msAb) encompass a diverse variety of formats that can concurrently bind multiple epitopes, unlocking mechanisms to address previously difficult-to-treat or incurable diseases. Early assessment of candidate developability enables demotion of antibodies with low potential and promotion of the most promising candidates for further development. Protein-based therapies have a stringent set of developability requirements in order to be competitive (e.g. high-concentration formulation, and long half-life) and their assessment requires a robust toolkit of methods, few of which are validated for interrogating bsAbs/msAbs. Important considerations when assessing the developability of bsAbs/msAbs include their molecular format, likelihood for immunogenicity, specificity, stability, and potential for high-volume production. Here, we summarize the critical aspects of developability assessment, and provide guidance on how to develop a comprehensive plan tailored to a given bsAb/msAb.
Collapse
Affiliation(s)
- Alaa Amash
- AbCellera Biologics Inc, Vancouver, BC, Canada
| | | | | | | | | | | | | | | | | | - Tim Jacobs
- AbCellera Biologics Inc, Vancouver, BC, Canada
| |
Collapse
|
12
|
Lutz S, Klausz K, Albici AM, Ebinger L, Sellmer L, Teipel H, Frenzel A, Langner A, Winterberg D, Krohn S, Hust M, Schirrmann T, Dübel S, Scherließ R, Humpe A, Gramatzki M, Kellner C, Peipp M. Novel NKG2D-directed bispecific antibodies enhance antibody-mediated killing of malignant B cells by NK cells and T cells. Front Immunol 2023; 14:1227572. [PMID: 37965326 PMCID: PMC10641740 DOI: 10.3389/fimmu.2023.1227572] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/05/2023] [Indexed: 11/16/2023] Open
Abstract
The activating receptor natural killer group 2, member D (NKG2D) represents an attractive target for immunotherapy as it exerts a crucial role in cancer immunosurveillance by regulating the activity of cytotoxic lymphocytes. In this study, a panel of novel NKG2D-specific single-chain fragments variable (scFv) were isolated from naïve human antibody gene libraries and fused to the fragment antigen binding (Fab) of rituximab to obtain [CD20×NKG2D] bibodies with the aim to recruit cytotoxic lymphocytes to lymphoma cells. All bispecific antibodies bound both antigens simultaneously. Two bibody constructs, [CD20×NKG2D#3] and [CD20×NKG2D#32], efficiently activated natural killer (NK) cells in co-cultures with CD20+ lymphoma cells. Both bibodies triggered NK cell-mediated lysis of lymphoma cells and especially enhanced antibody-dependent cell-mediated cytotoxicity (ADCC) by CD38 or CD19 specific monoclonal antibodies suggesting a synergistic effect between NKG2D and FcγRIIIA signaling pathways in NK cell activation. The [CD20×NKG2D] bibodies were not effective in redirecting CD8+ T cells as single agents, but enhanced cytotoxicity when combined with a bispecific [CD19×CD3] T cell engager, indicating that NKG2D signaling also supports CD3-mediated T cell activation. In conclusion, engagement of NKG2D with bispecific antibodies is attractive to directly activate cytotoxic lymphocytes or to support their activation by monoclonal antibodies or bispecific T cell engagers. As a perspective, co-targeting of two tumor antigens may allow fine-tuning of antibody cancer therapies. Our proposed combinatorial approach is potentially applicable for many existing immunotherapies but further testing in different preclinical models is necessary to explore the full potential.
Collapse
Affiliation(s)
- Sebastian Lutz
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, Ludwig Maximilians University (LMU) Munich, Munich, Germany
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Katja Klausz
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Anca-Maria Albici
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Lea Ebinger
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Lea Sellmer
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Hannah Teipel
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | | | - Anna Langner
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Dorothee Winterberg
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Steffen Krohn
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Michael Hust
- YUMAB GmbH, Braunschweig, Germany
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | | | - Stefan Dübel
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Regina Scherließ
- Department of Pharmaceutics and Biopharmaceutics, Kiel University, Kiel, Germany
| | - Andreas Humpe
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, Ludwig Maximilians University (LMU) Munich, Munich, Germany
| | - Martin Gramatzki
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Christian Kellner
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, Ludwig Maximilians University (LMU) Munich, Munich, Germany
| | - Matthias Peipp
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| |
Collapse
|
13
|
Zhang Y, Li Q, Luo L, Duan C, Shen J, Wang Z. Application of germline antibody features to vaccine development, antibody discovery, antibody optimization and disease diagnosis. Biotechnol Adv 2023; 65:108143. [PMID: 37023966 DOI: 10.1016/j.biotechadv.2023.108143] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/26/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023]
Abstract
Although the efficacy and commercial success of vaccines and therapeutic antibodies have been tremendous, designing and discovering new drug candidates remains a labor-, time- and cost-intensive endeavor with high risks. The main challenges of vaccine development are inducing a strong immune response in broad populations and providing effective prevention against a group of highly variable pathogens. Meanwhile, antibody discovery faces several great obstacles, especially the blindness in antibody screening and the unpredictability of the developability and druggability of antibody drugs. These challenges are largely due to poorly understanding of germline antibodies and the antibody responses to pathogen invasions. Thanks to the recent developments in high-throughput sequencing and structural biology, we have gained insight into the germline immunoglobulin (Ig) genes and germline antibodies and then the germline antibody features associated with antigens and disease manifestation. In this review, we firstly outline the broad associations between germline antibodies and antigens. Moreover, we comprehensively review the recent applications of antigen-specific germline antibody features, physicochemical properties-associated germline antibody features, and disease manifestation-associated germline antibody features on vaccine development, antibody discovery, antibody optimization, and disease diagnosis. Lastly, we discuss the bottlenecks and perspectives of current and potential applications of germline antibody features in the biotechnology field.
Collapse
Affiliation(s)
- Yingjie Zhang
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China
| | - Qing Li
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China
| | - Liang Luo
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China
| | - Changfei Duan
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China
| | - Zhanhui Wang
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China.
| |
Collapse
|
14
|
Pomarici ND, Waibl F, Quoika PK, Bujotzek A, Georges G, Fernández-Quintero ML, Liedl KR. Structural mechanism of Fab domain dissociation as a measure of interface stability. J Comput Aided Mol Des 2023; 37:201-215. [PMID: 36918473 PMCID: PMC10049950 DOI: 10.1007/s10822-023-00501-9] [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: 01/02/2023] [Accepted: 02/23/2023] [Indexed: 03/16/2023]
Abstract
Therapeutic antibodies should not only recognize antigens specifically, but also need to be free from developability issues, such as poor stability. Thus, the mechanistic understanding and characterization of stability are critical determinants for rational antibody design. In this study, we use molecular dynamics simulations to investigate the melting process of 16 antigen binding fragments (Fabs). We describe the Fab dissociation mechanisms, showing a separation in the VH-VL and in the CH1-CL domains. We found that the depths of the minima in the free energy curve, corresponding to the bound states, correlate with the experimentally determined melting temperatures. Additionally, we provide a detailed structural description of the dissociation mechanism and identify key interactions in the CDR loops and in the CH1-CL interface that contribute to stabilization. The dissociation of the VH-VL or CH1-CL domains can be represented by conformational changes in the bend angles between the domains. Our findings elucidate the melting process of antigen binding fragments and highlight critical residues in both the variable and constant domains, which are also strongly germline dependent. Thus, our proposed mechanisms have broad implications in the development and design of new and more stable antigen binding fragments.
Collapse
Affiliation(s)
- Nancy D Pomarici
- Institute of General, Inorganic and Theoretical Chemistry, and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Franz Waibl
- Institute of General, Inorganic and Theoretical Chemistry, and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Patrick K Quoika
- Institute of General, Inorganic and Theoretical Chemistry, and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
- Center for Protein Assemblies (CPA), Physics Department, Chair of Theoretical Biophysics, Technical University of Munich, Ernst-Otto-Fischer-Str. 8, 85748, Garching, Germany
| | - Alexander Bujotzek
- Roche Pharma Research and Early Development, Large Molecule Research, Roche Innovation Center Munich, Nonnenwald 2, 82377, Penzberg, Germany
| | - Guy Georges
- Roche Pharma Research and Early Development, Large Molecule Research, Roche Innovation Center Munich, Nonnenwald 2, 82377, Penzberg, Germany
| | - Monica L Fernández-Quintero
- Institute of General, Inorganic and Theoretical Chemistry, and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria.
| | - Klaus R Liedl
- Institute of General, Inorganic and Theoretical Chemistry, and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria.
| |
Collapse
|
15
|
Gómez-Ramírez IV, Corrales-García LL, Possani LD, Riaño-Umbarila L, Becerril B. Expression in Pichia pastoris of human antibody fragments that neutralize venoms of Mexican scorpions. Toxicon 2023; 223:107012. [PMID: 36592762 DOI: 10.1016/j.toxicon.2022.107012] [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: 11/09/2022] [Revised: 12/16/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022]
Abstract
The methylotrophic yeast Pichia pastoris has been one of the most widely used organisms in recent years as an expression system for a wide variety of recombinant proteins with therapeutic potential. Its popularity as an alternative system to Escherichia coli is mainly due to the easy genetic manipulation and the ability to produce high levels of heterologous proteins, either intracellularly or extracellularly. Being a eukaryotic organism, P. pastoris carries out post-translational modifications that allow it to produce soluble and correctly folded recombinant proteins. This work, evaluated the expression capacity in P. pastoris of two single-chain variable fragments (scFvs) of human origin, 10FG2 and LR. These scFvs were previously obtained by directed evolution against scorpion venom toxins and are able to neutralize different toxins and venoms of Mexican species. The yield obtained in P. pastoris was higher than that obtained in bacterial periplasm (E. coli), and most importantly, biochemical and functional properties were not modified. These results confirm that P. pastoris yeast can be a good expression system for the production of antibody fragments of a new recombinant antivenom.
Collapse
Affiliation(s)
- Ilse V Gómez-Ramírez
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, UNAM, Apartado Postal 510-3, Cuernavaca, Morelos, 62250, Mexico
| | - Ligia Luz Corrales-García
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, UNAM, Apartado Postal 510-3, Cuernavaca, Morelos, 62250, Mexico; Departamento de Alimentos, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia, AA 1226, Medellín, 050010, Colombia
| | - Lourival D Possani
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, UNAM, Apartado Postal 510-3, Cuernavaca, Morelos, 62250, Mexico
| | - Lidia Riaño-Umbarila
- Investigadora por México, CONACyT, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Apartado Postal 510-3, Cuernavaca, Morelos, 62250, Mexico.
| | - Baltazar Becerril
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, UNAM, Apartado Postal 510-3, Cuernavaca, Morelos, 62250, Mexico.
| |
Collapse
|
16
|
Kolmar H, Grzeschik J, Könning D, Krah S, Zielonka S. Construction of Semisynthetic Shark vNAR Yeast Surface Display Antibody Libraries. Methods Mol Biol 2023; 2702:227-243. [PMID: 37679622 DOI: 10.1007/978-1-0716-3381-6_11] [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: 09/09/2023]
Abstract
The adaptive immune system of sharks comprises a unique heavy chain-only antibody isotype, termed immunoglobulin new antigen receptor (IgNAR), in which antigen binding is mediated by a single variable domain, referred to as vNAR. In recent years, efforts were made to harness these domains for biomedical and biotechnological applications particularly due to their high affinity and specificity combined with a small size and high stability. Herein, we describe protocols for the construction of semisynthetic, CDR3-randomized vNAR libraries for the isolation of target-specific paratopes by yeast surface display. Additionally, we provide guidance for affinity maturation of a panel of antigen-enriched vNAR domains through CDR1 diversification of the FACS-selected, antigen-enriched population and sublibrary establishment.
Collapse
Affiliation(s)
- Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany.
| | - Julius Grzeschik
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Doreen Könning
- Antibody-Drug Conjugates and Targeted NBE Therapeutics, Merck KGaA, Darmstadt, Germany
| | - Simon Krah
- Antibody Discovery & Protein Engineering, Merck KGaA, Darmstadt, Germany
| | - Stefan Zielonka
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany.
- Antibody Discovery & Protein Engineering, Merck KGaA, Darmstadt, Germany.
| |
Collapse
|
17
|
Ausserwöger H, Schneider MM, Herling TW, Arosio P, Invernizzi G, Knowles TPJ, Lorenzen N. Non-specificity as the sticky problem in therapeutic antibody development. Nat Rev Chem 2022; 6:844-861. [PMID: 37117703 DOI: 10.1038/s41570-022-00438-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2022] [Indexed: 11/16/2022]
Abstract
Antibodies are highly potent therapeutic scaffolds with more than a hundred different products approved on the market. Successful development of antibody-based drugs requires a trade-off between high target specificity and target binding affinity. In order to better understand this problem, we here review non-specific interactions and explore their fundamental physicochemical origins. We discuss the role of surface patches - clusters of surface-exposed amino acid residues with similar physicochemical properties - as inducers of non-specific interactions. These patches collectively drive interactions including dipole-dipole, π-stacking and hydrophobic interactions to complementary moieties. We elucidate links between these supramolecular assembly processes and macroscopic development issues, such as decreased physical stability and poor in vivo half-life. Finally, we highlight challenges and opportunities for optimizing protein binding specificity and minimizing non-specificity for future generations of therapeutics.
Collapse
|
18
|
Fernández-Quintero ML, Fischer ALM, Kokot J, Waibl F, Seidler CA, Liedl KR. The influence of antibody humanization on shark variable domain (VNAR) binding site ensembles. Front Immunol 2022; 13:953917. [PMID: 36177031 PMCID: PMC9514858 DOI: 10.3389/fimmu.2022.953917] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Sharks and other cartilaginous fish produce new antigen receptor (IgNAR) antibodies, as key part of their humoral immune response and are the phylogenetically oldest living organisms that possess an immunoglobulin (Ig)-based adaptive immune system. IgNAR antibodies are naturally occurring heavy-chain-only antibodies, that recognize antigens with their single domain variable regions (VNARs). In this study, we structurally and biophysically elucidate the effect of antibody humanization of a previously published spiny dogfish VNAR (parent E06), which binds with high affinity to the human serum albumin (HSA). We analyze different humanization variants together with the parental E06 VNAR and the human Vκ1 light chain germline DPK9 antibody to characterize the influence of point mutations in the framework and the antigen binding site on the specificity of VNARs as reported by Kovalenko et al. We find substantially higher flexibility in the humanized variants, reflected in a broader conformational space and a higher conformational entropy, as well as population shifts of the dominant binding site ensembles in solution. A further variant, in which some mutations are reverted, largely restores the conformational stability and the dominant binding minimum of the parent E06. We also identify differences in surface hydrophobicity between the human Vκ1 light chain germline DPK9 antibody, the parent VNAR E06 and the humanized variants. Additional simulations of VNAR-HSA complexes of the parent E06 VNAR and a humanized variant reveal that the parent VNAR features a substantially stronger network of stabilizing interactions. Thus, we conclude that a structural and dynamic understanding of the VNAR binding site upon humanization is a key aspect in antibody humanization.
Collapse
Affiliation(s)
| | | | | | | | | | - Klaus R. Liedl
- Department of General, Inorganic and Theoretical Chemistry, and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
19
|
Jia L, Jain M, Sun Y. Improving antibody thermostability based on statistical analysis of sequence and structural consensus data. Antib Ther 2022; 5:202-210. [PMID: 35967906 PMCID: PMC9372885 DOI: 10.1093/abt/tbac017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/21/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
Background
The use of Monoclonal Antibodies (MAbs) as therapeutics has been increasing over the past 30 years due to their high specificity and strong affinity towards the target. One of the major challenges towarding their use as drugs is their low thermostability, which impacts both efficacy as well as manufacturing and delivery.
Methods
To aid the design of thermally more stable mutants, consensus sequence-based method has been widely used. These methods typically have a success rate of about 50% with maximum melting temperature increment ranging from 10 to 32 °C. In order to improve the prediction performance, we have developed a new and fast MAbs specific method by adding a 3D structural layer to the consensus sequence method. This is done by analyzing the close-by residue pairs which are conserved in more than eight hundred MAbs’ 3D structures.
Results
Combining consensus sequence and structural residue pair covariance methods, we developed an in-house application for predicting human MAb thermostability to guide protein engineers to design stable molecules. Major advantage of this structural level assessment is in significantly reducing the false positives by almost half from the consensus sequence method alone. This application has shown success in designing MAb engineering panels in multiple biologics programs.
Conclusions
Our data science-based method shows impacts in Mab engineering.
Collapse
Affiliation(s)
- Lei Jia
- Discovery Research , Amgen, Thousand Oaks, CA, USA
| | - Mani Jain
- Discovery Research , Amgen, Thousand Oaks, CA, USA
| | - Yaxiong Sun
- Discovery Research , Amgen, Thousand Oaks, CA, USA
| |
Collapse
|
20
|
Darcy M, Crocker K, Wang Y, Le JV, Mohammadiroozbahani G, Abdelhamid MAS, Craggs TD, Castro CE, Bundschuh R, Poirier MG. High-Force Application by a Nanoscale DNA Force Spectrometer. ACS NANO 2022; 16:5682-5695. [PMID: 35385658 PMCID: PMC9048690 DOI: 10.1021/acsnano.1c10698] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/28/2022] [Indexed: 05/06/2023]
Abstract
The ability to apply and measure high forces (>10 pN) on the nanometer scale is critical to the development of nanomedicine, molecular robotics, and the understanding of biological processes such as chromatin condensation, membrane deformation, and viral packaging. Established force spectroscopy techniques including optical traps, magnetic tweezers, and atomic force microscopy rely on micron-sized or larger handles to apply forces, limiting their applications within constrained geometries including cellular environments and nanofluidic devices. A promising alternative to these approaches is DNA-based molecular calipers. However, this approach is currently limited to forces on the scale of a few piconewtons. To study the force application capabilities of DNA devices, we implemented DNA origami nanocalipers with tunable mechanical properties in a geometry that allows application of force to rupture a DNA duplex. We integrated static and dynamic single-molecule characterization methods and statistical mechanical modeling to quantify the device properties including force output and dynamic range. We found that the thermally driven dynamics of the device are capable of applying forces of at least 20 piconewtons with a nanometer-scale dynamic range. These characteristics could eventually be used to study other biomolecular processes such as protein unfolding or to control high-affinity interactions in nanomechanical devices or molecular robots.
Collapse
Affiliation(s)
- Michael Darcy
- Department
of Physics, Department of Mechanical and Aerospace Engineering, Biophysics Graduate
Program, Department of Chemistry and Biochemistry, and Division of Hematology, Department
of Internal Medicine, The Ohio State University, Columbus, Ohio 43210, United States
| | - Kyle Crocker
- Department
of Physics, Department of Mechanical and Aerospace Engineering, Biophysics Graduate
Program, Department of Chemistry and Biochemistry, and Division of Hematology, Department
of Internal Medicine, The Ohio State University, Columbus, Ohio 43210, United States
| | - Yuchen Wang
- Department
of Physics, Department of Mechanical and Aerospace Engineering, Biophysics Graduate
Program, Department of Chemistry and Biochemistry, and Division of Hematology, Department
of Internal Medicine, The Ohio State University, Columbus, Ohio 43210, United States
| | - Jenny V. Le
- Department
of Physics, Department of Mechanical and Aerospace Engineering, Biophysics Graduate
Program, Department of Chemistry and Biochemistry, and Division of Hematology, Department
of Internal Medicine, The Ohio State University, Columbus, Ohio 43210, United States
| | - Golbarg Mohammadiroozbahani
- Department
of Physics, Department of Mechanical and Aerospace Engineering, Biophysics Graduate
Program, Department of Chemistry and Biochemistry, and Division of Hematology, Department
of Internal Medicine, The Ohio State University, Columbus, Ohio 43210, United States
| | | | - Timothy D. Craggs
- Department
of Chemistry, University of Sheffield, Sheffield S3 7HF, U.K.
| | - Carlos E. Castro
- Department
of Physics, Department of Mechanical and Aerospace Engineering, Biophysics Graduate
Program, Department of Chemistry and Biochemistry, and Division of Hematology, Department
of Internal Medicine, The Ohio State University, Columbus, Ohio 43210, United States
| | - Ralf Bundschuh
- Department
of Physics, Department of Mechanical and Aerospace Engineering, Biophysics Graduate
Program, Department of Chemistry and Biochemistry, and Division of Hematology, Department
of Internal Medicine, The Ohio State University, Columbus, Ohio 43210, United States
| | - Michael G. Poirier
- Department
of Physics, Department of Mechanical and Aerospace Engineering, Biophysics Graduate
Program, Department of Chemistry and Biochemistry, and Division of Hematology, Department
of Internal Medicine, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
21
|
Wu Y, Li Q, Kong Y, Wang Z, Lei C, Li J, Ding L, Wang C, Cheng Y, Wei Y, Song Y, Yang Z, Tu C, Ding Y, Ying T. A highly stable human single-domain antibody-drug conjugate exhibits superior penetration and treatment of solid tumors. Mol Ther 2022; 30:2785-2799. [PMID: 35462042 PMCID: PMC9372316 DOI: 10.1016/j.ymthe.2022.04.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/04/2022] [Accepted: 04/18/2022] [Indexed: 10/18/2022] Open
Abstract
The inefficient tumor penetration of therapeutic antibodies has hampered their effective use in treating solid tumors. Here, we report the identification of a fully human single-domain antibody (UdAb), designated as n501, targeting the oncofetal antigen 5T4. The high-resolution crystal structure indicates that n501 adopts a compact structure very similar to that of camelid nanobodies, and binds tightly to all eight leucine-rich repeats of 5T4. Furthermore, the UdAb n501 exhibits exceptionally high stability, with no apparent activity changes over 4 weeks of storage at various temperatures. Importantly, the UdAb-based antibody-drug conjugate (n501-SN38) showed much deeper tumor penetration, significantly higher tumor uptake, and faster accumulation at tumor sites than conventional IgG1-based antibody-drug conjugate (m603-SN38), resulting in improved tumor inhibition. These results highlight the potential of UdAb-based antibody-drug conjugates as a potential class of antitumor therapeutics with characteristics of high stability and strong tumor penetration for the effective treatment of solid tumors.
Collapse
|
22
|
Barakat S, Berksöz M, Zahedimaram P, Piepoli S, Erman B. Nanobodies as molecular imaging probes. Free Radic Biol Med 2022; 182:260-275. [PMID: 35240292 DOI: 10.1016/j.freeradbiomed.2022.02.031] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/08/2022] [Accepted: 02/14/2022] [Indexed: 12/12/2022]
Abstract
Camelidae derived single-domain antibodies (sdAbs), commonly known as nanobodies (Nbs), are the smallest antibody fragments with full antigen-binding capacity. Owing to their desirable properties such as small size, high specificity, strong affinity, excellent stability, and modularity, nanobodies are on their way to overtake conventional antibodies in terms of popularity. To date, a broad range of nanobodies have been generated against different molecular targets with applications spanning basic research, diagnostics, and therapeutics. In the field of molecular imaging, nanobody-based probes have emerged as a powerful tool. Radioactive or fluorescently labeled nanobodies are now used to detect and track many targets in different biological systems using imaging techniques. In this review, we provide an overview of the use of nanobodies as molecular probes. Additionally, we discuss current techniques for the generation, conjugation, and intracellular delivery of nanobodies.
Collapse
Affiliation(s)
- Sarah Barakat
- Faculty of Engineering and Natural Sciences, Sabanci University, 34956, Tuzla, Istanbul, Turkey.
| | - Melike Berksöz
- Faculty of Engineering and Natural Sciences, Sabanci University, 34956, Tuzla, Istanbul, Turkey.
| | - Pegah Zahedimaram
- Faculty of Engineering and Natural Sciences, Sabanci University, 34956, Tuzla, Istanbul, Turkey.
| | - Sofia Piepoli
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bogazici University, 34342, Bebek, Istanbul, Turkey.
| | - Batu Erman
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bogazici University, 34342, Bebek, Istanbul, Turkey.
| |
Collapse
|
23
|
Obeng EM, Dzuvor CKO, Danquah MK. Anti-SARS-CoV-1 and -2 nanobody engineering towards avidity-inspired therapeutics. NANO TODAY 2022; 42:101350. [PMID: 34840592 PMCID: PMC8608585 DOI: 10.1016/j.nantod.2021.101350] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/22/2021] [Accepted: 11/18/2021] [Indexed: 05/15/2023]
Abstract
In the past two decades, the emergence of coronavirus diseases has been dire distress on both continental and global fronts and has resulted in the search for potent treatment strategies. One crucial challenge in this search is the recurrent mutations in the causative virus spike protein, which lead to viral escape issues. Among the current promising therapeutic discoveries is the use of nanobodies and nanobody-like molecules. While these nanobodies have demonstrated high-affinity interaction with the virus, the unpredictable spike mutations have warranted the need for avidity-inspired therapeutics of potent inhibitors such as nanobodies. This article discusses novel approaches for the design of anti-SARS-CoV-1 and -2 nanobodies to facilitate advanced innovations in treatment technologies. It further discusses molecular interactions and suggests multivalent protein nanotechnology and chemistry approaches to translate mere molecular affinity into avidity.
Collapse
Affiliation(s)
- Eugene M Obeng
- Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Christian K O Dzuvor
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Michael K Danquah
- Department of Chemical Engineering, University of Tennessee, Chattanooga 615 McCallie Ave, Chattanooga, TN 37403, United States
| |
Collapse
|
24
|
Engineering an Enhanced EGFR Engager: Humanization of Cetuximab for Improved Developability. Antibodies (Basel) 2022; 11:antib11010006. [PMID: 35076451 PMCID: PMC8788544 DOI: 10.3390/antib11010006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/12/2021] [Accepted: 01/05/2022] [Indexed: 01/05/2023] Open
Abstract
The epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase whose proliferative effects can contribute to the development of many types of solid tumors when overexpressed. For this reason, EGFR inhibitors such as cetuximab can play an important role in treating cancers such as colorectal cancer and head and neck cancer. Cetuximab is a chimeric monoclonal antibody containing mouse variable regions that bind to EGFR and prevent it from signaling. Although cetuximab has been used clinically since 2004 to successfully control solid tumors, advances in protein engineering have created the opportunity to address some of its shortcomings. In particular, the presence of mouse sequences could contribute to immunogenicity in the form of anti-cetuximab antibodies, and an occupied glycosylation site in FR3 can contribute to hypersensitivity reactions and product heterogeneity. Using simple framework graft or sequence-/structure-guided approaches, cetuximab was humanized onto 11 new frameworks. In addition to increasing humanness and removing the VH glycosylation site, dynamic light scattering revealed increases in stability, and bio-layer interferometry confirmed minimal changes in binding affinity, with patterns emerging across the humanization method. This work demonstrates the potential to improve the biophysical and clinical properties of first-generation protein therapeutics and highlights the advantages of computationally guided engineering.
Collapse
|
25
|
Dingus JG, Tang JCY, Amamoto R, Wallick GK, Cepko CL. A general approach for stabilizing nanobodies for intracellular expression. eLife 2022; 11:68253. [PMID: 36416528 PMCID: PMC9683787 DOI: 10.7554/elife.68253] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 11/08/2022] [Indexed: 11/24/2022] Open
Abstract
Conventional antibodies and their derived fragments are difficult to deploy against intracellular targets in live cells, due to their bulk and structural complexity. Nanobodies provide an alternative modality, with well-documented examples of intracellular expression. Despite their promise as intracellular reagents, there has not been a systematic study of nanobody intracellular expression. Here, we examined intracellular expression of 75 nanobodies from the Protein Data Bank. Surprisingly, a majority of these nanobodies were unstable in cells, illustrated by aggregation and clearance. Using comparative analysis and framework mutagenesis, we developed a general approach that stabilized a great majority of nanobodies that were originally unstable intracellularly, without significantly compromising target binding. This approach led to the identification of distinct sequence features that impacted the intracellular stability of tested nanobodies. Mutationally stabilized nanobody expression was found to extend to in vivo contexts, in the murine retina and in E. coli. These data provide for improvements in nanobody engineering for intracellular applications, potentiating a growing field of intracellular interrogation and intervention.
Collapse
Affiliation(s)
- John G Dingus
- Howard Hughes Medical Institute, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
| | - Jonathan CY Tang
- Howard Hughes Medical Institute, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
| | - Ryoji Amamoto
- Howard Hughes Medical Institute, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
| | - Grace K Wallick
- Howard Hughes Medical Institute, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
| | - Constance L Cepko
- Howard Hughes Medical Institute, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
26
|
Lee S, Jang S, Kang J, Park SB, Han YW, Nam H, Kim M, Lee J, Cho KJ, Kim J, Oh M, Ryu J, Seok JH, Kim Y, Lee JB, Park MS, Kim YS, Park H, Kim DS. MG1141A as a Highly Potent Monoclonal Neutralizing Antibody Against SARS-CoV-2 Variants. Front Immunol 2021; 12:778829. [PMID: 34868052 PMCID: PMC8637776 DOI: 10.3389/fimmu.2021.778829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022] Open
Abstract
Since the coronavirus disease outbreak in 2019, several antibody therapeutics have been developed to treat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections. Antibody therapeutics are effective in neutralizing the virus and reducing hospitalization in patients with mild and moderate infections. These therapeutics target the spike protein of SARS-CoV-2; however, emerging mutations in this protein reduce their efficiency. In this study, we developed a universal SARS-CoV-2 neutralizing antibody. We generated a humanized monoclonal antibody, MG1141A, against the receptor-binding domain of the spike protein through traditional mouse immunization. We confirmed that MG1141A could effectively neutralize live viruses, with an EC50 of 92 pM, and that it exhibited effective Fc-mediated functions. Additionally, it retained its neutralizing activity against the alpha (UK), beta (South Africa), and gamma (Brazil) variants of SARS-CoV-2. Taken together, our study contributes to the development of a novel antibody therapeutic approach, which can effectively combat emerging SARS-CoV-2 mutations.
Collapse
Affiliation(s)
- Sua Lee
- Department of Protein Engineering, Mogam Institute for Biomedical Research, Yongin, South Korea
| | - Shina Jang
- Department of Protein Engineering, Mogam Institute for Biomedical Research, Yongin, South Korea
| | - Jihoon Kang
- Department of Protein Engineering, Mogam Institute for Biomedical Research, Yongin, South Korea
| | - Soo Bin Park
- Department of Protein Engineering, Mogam Institute for Biomedical Research, Yongin, South Korea
| | - Young Woo Han
- Department of Infectious Disease Research, Mogam Institute for Biomedical Research, Yongin, South Korea
| | - Hyemi Nam
- Department of Target ID & Assay Development, Mogam Institute for Biomedical Research, Yongin, South Korea
| | - Munkyung Kim
- Department of Target ID & Assay Development, Mogam Institute for Biomedical Research, Yongin, South Korea
| | - Jeewon Lee
- Department of Target ID & Assay Development, Mogam Institute for Biomedical Research, Yongin, South Korea
| | - Ki Joon Cho
- Department of Protein Engineering, Mogam Institute for Biomedical Research, Yongin, South Korea
| | - Jeonghun Kim
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul, South Korea
| | - Miyoung Oh
- Department of Protein Engineering, Mogam Institute for Biomedical Research, Yongin, South Korea
| | - Jihye Ryu
- Department of Translational Research, Mogam Institute for Biomedical Research, Yongin, South Korea
| | - Jong Hyeon Seok
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul, South Korea
| | - Yunhwa Kim
- Department of Microbiology, College of Medicine, Yeungnam University, Daegu, South Korea
| | - Jee-Boong Lee
- Department of Target ID & Assay Development, Mogam Institute for Biomedical Research, Yongin, South Korea
| | - Man-Seong Park
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul, South Korea
| | - Yong-Sung Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Hosun Park
- Department of Microbiology, College of Medicine, Yeungnam University, Daegu, South Korea
| | - Dong-Sik Kim
- Department of Protein Engineering, Mogam Institute for Biomedical Research, Yongin, South Korea.,Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| |
Collapse
|
27
|
Wagner TR, Rothbauer U. Nanobodies - Little helpers unravelling intracellular signaling. Free Radic Biol Med 2021; 176:46-61. [PMID: 34536541 DOI: 10.1016/j.freeradbiomed.2021.09.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/26/2021] [Accepted: 09/08/2021] [Indexed: 11/21/2022]
Abstract
The identification of diagnostic and therapeutic targets requires a comprehensive understanding of cellular processes, for which advanced technologies in biomedical research are needed. The emergence of nanobodies (Nbs) derived from antibody fragments of camelid heavy chain-only antibodies as intracellular research tools offers new possibilities to study and modulate target antigens in living cells. Here we summarize this rapidly changing field, beginning with a brief introduction of Nbs, followed by an overview of how target-specific Nbs can be generated, and introduce the selection of intrabodies as research tools. Intrabodies, by definition, are intracellular functional Nbs that target ectopic or endogenous intracellular antigens within living cells. Such binders can be applied in various formats, e.g. as chromobodies for live cell microscopy or as biosensors to decipher complex intracellular signaling pathways. In addition, protein knockouts can be achieved by target-specific Nbs, while modulating Nbs have the potential as future therapeutics. The development of fine-tunable and switchable Nb-based systems that simultaneously provide spatial and temporal control has recently taken the application of these binders to the next level.
Collapse
Affiliation(s)
- Teresa R Wagner
- Pharmaceutical Biotechnology, Eberhard Karls University, Tübingen, Germany; NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Ulrich Rothbauer
- Pharmaceutical Biotechnology, Eberhard Karls University, Tübingen, Germany; NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany; Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany.
| |
Collapse
|
28
|
Hrynchak I, Santos L, Falcão A, Gomes CM, Abrunhosa AJ. Nanobody-Based Theranostic Agents for HER2-Positive Breast Cancer: Radiolabeling Strategies. Int J Mol Sci 2021; 22:ijms221910745. [PMID: 34639086 PMCID: PMC8509594 DOI: 10.3390/ijms221910745] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 02/07/2023] Open
Abstract
The overexpression of human epidermal growth factor 2 (HER2) in breast cancer (BC) has been associated with a more aggressive tumor subtype, poorer prognosis and shorter overall survival. In this context, the development of HER2-targeted radiotracers is crucial to provide a non-invasive assessment of HER2 expression to select patients for HER2-targeted therapies, monitor response and identify those who become resistant. Antibodies represent ideal candidates for this purpose, as they provide high contrast images for diagnosis and low toxicity in the therapeutic setting. Of those, nanobodies (Nb) are of particular interest considering their favorable kinetics, crossing of relevant biological membranes and intratumoral distribution. The purpose of this review is to highlight the unique characteristics and advantages of Nb-based radiotracers in BC imaging and therapy. Additionally, radiolabeling methods for Nb including direct labeling, indirect labeling via prosthetic group and indirect labeling via complexation will be discussed, reporting advantages and drawbacks. Furthermore, the preclinical to clinical translation of radiolabeled Nbs as promising theranostic agents will be reported.
Collapse
Affiliation(s)
- Ivanna Hrynchak
- ICNAS-Produção Unipessoal, Lda.—University of Coimbra, 3000-548 Coimbra, Portugal; (I.H.); (L.S.)
- CIBIT/ICNAS—Institute for Nuclear Sciences Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal;
| | - Liliana Santos
- ICNAS-Produção Unipessoal, Lda.—University of Coimbra, 3000-548 Coimbra, Portugal; (I.H.); (L.S.)
- CIBIT/ICNAS—Institute for Nuclear Sciences Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal;
| | - Amílcar Falcão
- CIBIT/ICNAS—Institute for Nuclear Sciences Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal;
| | - Célia M. Gomes
- iCBR—Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal;
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-548 Coimbra, Portugal
- CACC—Clinical Academic Center of Coimbra, 3000-075 Coimbra, Portugal
| | - Antero J. Abrunhosa
- ICNAS-Produção Unipessoal, Lda.—University of Coimbra, 3000-548 Coimbra, Portugal; (I.H.); (L.S.)
- CIBIT/ICNAS—Institute for Nuclear Sciences Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal;
- Correspondence:
| |
Collapse
|
29
|
Lan W, Valente JJ, Ilott A, Chennamsetty N, Liu Z, Rizzo JM, Yamniuk AP, Qiu D, Shackman HM, Bolgar MS. Investigation of anomalous charge variant profile reveals discrete pH-dependent conformations and conformation-dependent charge states within the CDR3 loop of a therapeutic mAb. MAbs 2021; 12:1763138. [PMID: 32432964 PMCID: PMC7299213 DOI: 10.1080/19420862.2020.1763138] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
During the development of a therapeutic monoclonal antibody (mAb-1), the charge variant profile obtained by pH-gradient cation exchange chromatography (CEX) contained two main peaks, each of which exhibited a unique intrinsic fluorescence profile and demonstrated inter-convertibility upon reinjection of isolated peak fractions. Domain analysis of mAb-1 by CEX and liquid chromatography-mass spectrometry indicated that the antigen-binding fragment chromatographed as two separate peaks that had identical mass. Surface plasmon resonance binding analysis to antigen demonstrated comparable kinetics/affinity between these fractionated peaks and unfractionated starting material. Subsequent molecular modeling studies revealed that the relatively long and flexible complementarity-determining region 3 (CDR3) loop on the heavy chain could adopt two discrete pH-dependent conformations: an “open” conformation at neutral pH where the HC-CDR3 is largely solvent exposed, and a “closed” conformation at lower pH where the solvent exposure of a neighboring tryptophan in the light chain is reduced and two aspartic acid residues near the ends of the HC-CDR3 loop have atypical pKa values. The pH-dependent equilibrium between “open” and “closed” conformations of the HC-CDR3, and its proposed role in the anomalous charge variant profile of mAb-1, were supported by further CEX and hydrophobic interaction chromatography studies. This work is an example of how pH-dependent conformational changes and conformation-dependent changes to net charge can unexpectedly contribute to perceived instability and require thorough analytical, biophysical, and functional characterization during biopharmaceutical drug product development.
Collapse
Affiliation(s)
- Wenkui Lan
- Drug Product Development, Bristol Myers Squibb, New Brunswick, United States
| | - Joseph J Valente
- Drug Product Development, Bristol Myers Squibb, New Brunswick, United States
| | - Andrew Ilott
- Drug Product Development, Bristol Myers Squibb, New Brunswick, United States
| | - Naresh Chennamsetty
- Biophysics Center of Excellence, Global Product Development and Supply, Bristol Myers Squibb, New Brunswick, United States
| | - Zhihua Liu
- Drug Product Development, Bristol Myers Squibb, New Brunswick, United States
| | - Joseph M Rizzo
- Discovery Biotherapeutics, Bristol Myers Squibb, Pennington, United States
| | - Aaron P Yamniuk
- Discovery Biotherapeutics, Bristol Myers Squibb, Pennington, United States
| | - Difei Qiu
- Chemical Process Department, Bristol Myers Squibb, New Brunswick, United States
| | - Holly M Shackman
- Chemical Process Department, Bristol Myers Squibb, New Brunswick, United States
| | - Mark S Bolgar
- Drug Product Development, Bristol Myers Squibb, New Brunswick, United States
| |
Collapse
|
30
|
Altunay B, Morgenroth A, Beheshti M, Vogg A, Wong NCL, Ting HH, Biersack HJ, Stickeler E, Mottaghy FM. HER2-directed antibodies, affibodies and nanobodies as drug-delivery vehicles in breast cancer with a specific focus on radioimmunotherapy and radioimmunoimaging. Eur J Nucl Med Mol Imaging 2021; 48:1371-1389. [PMID: 33179151 PMCID: PMC8113197 DOI: 10.1007/s00259-020-05094-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE The aim of the present paper is to review the role of HER2 antibodies, affibodies and nanobodies as vehicles for imaging and therapy approaches in breast cancer, including a detailed look at recent clinical data from antibody drug conjugates and nanobodies as well as affibodies that are currently under development. RESULTS Clinical and preclinical studies have shown that the use of monoclonal antibodies in molecular imaging is impaired by slow blood clearance, associated with slow and low tumor uptake and with limited tumor penetration potential. Antibody fragments, such as nanobodies, on the other hand, can be radiolabelled with short-lived radioisotopes and provide high-contrast images within a few hours after injection, allowing early diagnosis and reduced radiation exposure of patients. Even in therapy, the small radioactively labeled nanobodies prove to be superior to radioactively labeled monoclonal antibodies due to their higher specificity and their ability to penetrate the tumor. CONCLUSION While monoclonal antibodies are well established drug delivery vehicles, the current literature on molecular imaging supports the notion that antibody fragments, such as affibodies or nanobodies, might be superior in this approach.
Collapse
Affiliation(s)
- Betül Altunay
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074, Aachen, Germany
| | - Agnieszka Morgenroth
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074, Aachen, Germany
| | - Mohsen Beheshti
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074, Aachen, Germany
- Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne and Düsseldorf, Kerpener Str. 62, 50937, Cologne, Germany
- Division of Molecular PET-Imaging and Theranostics , Paracelsus Medical University , Salzburg, 5020, Austria
| | - Andreas Vogg
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074, Aachen, Germany
| | | | - Hong Hoi Ting
- Nanomab Technology Limited, Shanghai, People's Republic of China
| | | | - Elmar Stickeler
- Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne and Düsseldorf, Kerpener Str. 62, 50937, Cologne, Germany
- Department of Gynecology and Obstetrics, RWTH Aachen, Aachen, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074, Aachen, Germany.
- Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne and Düsseldorf, Kerpener Str. 62, 50937, Cologne, Germany.
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), 6202, Maastricht, The Netherlands.
| |
Collapse
|
31
|
Kim YJ, Lee MH, Lee SR, Chung HY, Kim K, Lee TG, Kim DY. Neutralizing Human Antibodies against Severe Acute Respiratory Syndrome Coronavirus 2 Isolated from a Human Synthetic Fab Phage Display Library. Int J Mol Sci 2021; 22:1913. [PMID: 33671877 PMCID: PMC7918989 DOI: 10.3390/ijms22041913] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 12/20/2022] Open
Abstract
Since it was first reported in Wuhan, China, in 2019, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a pandemic outbreak resulting in a tremendous global threat due to its unprecedented rapid spread and an absence of a prophylactic vaccine or therapeutic drugs treating the virus. The receptor-binding domain (RBD) of the SARS-CoV-2 spike protein is a key player in the viral entry into cells through its interaction with the angiotensin-converting enzyme 2 (ACE2) receptor protein, and the RBD has therefore been crucial as a drug target. In this study, we used phage display to develop human monoclonal antibodies (mAbs) that neutralize SARS-CoV-2. A human synthetic Fab phage display library was panned against the RBD of the SARS-CoV-2 spike protein (SARS-2 RBD), yielding ten unique Fabs with moderate apparent affinities (EC50 = 19-663 nM) for the SARS-2 RBD. All of the Fabs showed no cross-reactivity to the MERS-CoV spike protein, while three Fabs cross-reacted with the SARS-CoV spike protein. Five Fabs showed neutralizing activities in in vitro assays based on the Fabs' activities antagonizing the interaction between the SARS-2 RBD and ACE2. Reformatting the five Fabs into immunoglobulin Gs (IgGs) greatly increased their apparent affinities (KD = 0.08-1.0 nM), presumably due to the effects of avidity, without compromising their non-aggregating properties and thermal stability. Furthermore, two of the mAbs (D12 and C2) significantly showed neutralizing activities on pseudo-typed and authentic SARS-CoV-2. Given their desirable properties and neutralizing activities, we anticipate that these human anti-SARS-CoV-2 mAbs would be suitable reagents to be further developed as antibody therapeutics to treat COVID-19, as well as for diagnostics and research tools.
Collapse
|
32
|
DutaFabs are engineered therapeutic Fab fragments that can bind two targets simultaneously. Nat Commun 2021; 12:708. [PMID: 33514724 PMCID: PMC7846786 DOI: 10.1038/s41467-021-20949-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 12/18/2020] [Indexed: 01/07/2023] Open
Abstract
We report the development of a platform of dual targeting Fab (DutaFab) molecules, which comprise two spatially separated and independent binding sites within the human antibody CDR loops: the so-called H-side paratope encompassing HCDR1, HCDR3 and LCDR2, and the L-side paratope encompassing LCDR1, LCDR3 and HCDR2. Both paratopes can be independently selected and combined into the desired bispecific DutaFabs in a modular manner. X-ray crystal structures illustrate that DutaFabs are able to bind two target molecules simultaneously at the same Fv region comprising a VH-VL heterodimer. In the present study, this platform is applied to generate DutaFabs specific for VEGFA and PDGF-BB, which show high affinities, physico-chemical stability and solubility, as well as superior efficacy over anti-VEGF monotherapy in vivo. These molecules exemplify the usefulness of DutaFabs as a distinct class of antibody therapeutics, which is currently being evaluated in patients. Bispecific antibodies can bind to two distinct targets though the fusion of two different Fv regions. In this study, the authors develop DutaFabs that present two separated and independent antigen binding sites within the same Fv region, giving rise to bispecific Fab fragments.
Collapse
|
33
|
Mimura Y, Saldova R, Mimura-Kimura Y, Rudd PM, Jefferis R. Micro-Heterogeneity of Antibody Molecules. EXPERIENTIA SUPPLEMENTUM (2012) 2021; 112:1-26. [PMID: 34687006 DOI: 10.1007/978-3-030-76912-3_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Therapeutic monoclonal antibodies (mAbs) are mostly of the IgG class and constitute highly efficacious biopharmaceuticals for a wide range of clinical indications. Full-length IgG mAbs are large proteins that are subject to multiple posttranslational modifications (PTMs) during biosynthesis, purification, or storage, resulting in micro-heterogeneity. The production of recombinant mAbs in nonhuman cell lines may result in loss of structural fidelity and the generation of variants having altered stability, biological activities, and/or immunogenic potential. Additionally, even fully human therapeutic mAbs are of unique specificity, by design, and, consequently, of unique structure; therefore, structural elements may be recognized as non-self by individuals within an outbred human population to provoke an anti-therapeutic/anti-drug antibody (ATA/ADA) response. Consequently, regulatory authorities require that the structure of a potential mAb drug product is comprehensively characterized employing state-of-the-art orthogonal analytical technologies; the PTM profile may define a set of critical quality attributes (CQAs) for the drug product that must be maintained, employing quality by design parameters, throughout the lifetime of the drug. Glycosylation of IgG-Fc, at Asn297 on each heavy chain, is an established CQA since its presence and fine structure can have a profound impact on efficacy and safety. The glycoform profile of serum-derived IgG is highly heterogeneous while mAbs produced in mammalian cells in vitro is less heterogeneous and can be "orchestrated" depending on the cell line employed and the culture conditions adopted. Thus, the gross structure and PTM profile of a given mAb, established for the drug substance gaining regulatory approval, have to be maintained for the lifespan of the drug. This review outlines our current understanding of common PTMs detected in mAbs and endogenous IgG and the relationship between a variant's structural attribute and its impact on clinical performance.
Collapse
Affiliation(s)
- Yusuke Mimura
- Department of Clinical Research, National Hospital Organization Yamaguchi Ube Medical Center, Ube, Japan.
| | - Radka Saldova
- NIBRT GlycoScience Group, National Institute for Bioprocessing Research and Training, Mount Merrion, Blackrock, Co Dublin, Ireland
- UCD School of Medicine, College of Health and Agricultural Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Yuka Mimura-Kimura
- Department of Clinical Research, National Hospital Organization Yamaguchi Ube Medical Center, Ube, Japan
| | - Pauline M Rudd
- NIBRT GlycoScience Group, National Institute for Bioprocessing Research and Training, Mount Merrion, Blackrock, Co Dublin, Ireland
- Bioprocessing Technology Institute, Singapore, Singapore
| | - Roy Jefferis
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| |
Collapse
|
34
|
Transgenic Animals for the Generation of Human Antibodies. LEARNING MATERIALS IN BIOSCIENCES 2021. [DOI: 10.1007/978-3-030-54630-4_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
35
|
Root AR, Guntas G, Katragadda M, Apgar JR, Narula J, Chang CS, Hanscom S, McKenna M, Wade J, Meade C, Ma W, Guo Y, Liu Y, Duan W, Hendershot C, King AC, Zhang Y, Sousa E, Tam A, Benard S, Yang H, Kelleher K, Jin F, Piche-Nicholas N, Keating SE, Narciandi F, Lawrence-Henderson R, Arai M, Stochaj WR, Svenson K, Mosyak L, Lam K, Francis C, Marquette K, Wroblewska L, Zhu HL, Sheehan AD, LaVallie ER, D’Antona AM, Betts A, King L, Rosfjord E, Cunningham O, Lin L, Sapra P, Tchistiakova L, Mathur D, Bloom L. Discovery and optimization of a novel anti-GUCY2c x CD3 bispecific antibody for the treatment of solid tumors. MAbs 2021; 13:1850395. [PMID: 33459147 PMCID: PMC7833764 DOI: 10.1080/19420862.2020.1850395] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/26/2020] [Accepted: 11/10/2020] [Indexed: 12/29/2022] Open
Abstract
We report here the discovery and optimization of a novel T cell retargeting anti-GUCY2C x anti-CD3ε bispecific antibody for the treatment of solid tumors. Using a combination of hybridoma, phage display and rational design protein engineering, we have developed a fully humanized and manufacturable CD3 bispecific antibody that demonstrates favorable pharmacokinetic properties and potent in vivo efficacy. Anti-GUCY2C and anti-CD3ε antibodies derived from mouse hybridomas were first humanized into well-behaved human variable region frameworks with full retention of binding and T-cell mediated cytotoxic activity. To address potential manufacturability concerns, multiple approaches were taken in parallel to optimize and de-risk the two antibody variable regions. These approaches included structure-guided rational mutagenesis and phage display-based optimization, focusing on improving stability, reducing polyreactivity and self-association potential, removing chemical liabilities and proteolytic cleavage sites, and de-risking immunogenicity. Employing rapid library construction methods as well as automated phage display and high-throughput protein production workflows enabled efficient generation of an optimized bispecific antibody with desirable manufacturability properties, high stability, and low nonspecific binding. Proteolytic cleavage and deamidation in complementarity-determining regions were also successfully addressed. Collectively, these improvements translated to a molecule with potent single-agent in vivo efficacy in a tumor cell line adoptive transfer model and a cynomolgus monkey pharmacokinetic profile (half-life>4.5 days) suitable for clinical development. Clinical evaluation of PF-07062119 is ongoing.
Collapse
Affiliation(s)
- Adam R. Root
- BioMedicine Design, Pfizer Inc., Cambridge, MA, USA
| | | | | | | | - Jatin Narula
- BioMedicine Design, Pfizer Inc., Cambridge, MA, USA
| | | | - Sara Hanscom
- BioMedicine Design, Pfizer Inc., Cambridge, MA, USA
| | | | - Jason Wade
- BioMedicine Design, Pfizer Inc., Cambridge, MA, USA
| | - Caryl Meade
- BioMedicine Design, Pfizer Inc., Cambridge, MA, USA
| | - Weijun Ma
- BioMedicine Design, Pfizer Inc., Cambridge, MA, USA
| | - Yongjing Guo
- BioMedicine Design, Pfizer Inc., Cambridge, MA, USA
| | - Yan Liu
- BioMedicine Design, Pfizer Inc., Cambridge, MA, USA
| | - Weili Duan
- BioMedicine Design, Pfizer Inc., Cambridge, MA, USA
| | | | - Amy C. King
- BioMedicine Design, Pfizer Inc., Cambridge, MA, USA
| | - Yan Zhang
- BioMedicine Design, Pfizer Inc., Cambridge, MA, USA
| | - Eric Sousa
- BioMedicine Design, Pfizer Inc., Cambridge, MA, USA
| | - Amy Tam
- BioMedicine Design, Pfizer Inc., Cambridge, MA, USA
| | - Susan Benard
- BioMedicine Design, Pfizer Inc., Cambridge, MA, USA
| | - Han Yang
- BioMedicine Design, Pfizer Inc., Cambridge, MA, USA
| | | | - Fang Jin
- BioMedicine Design, Pfizer Inc., Cambridge, MA, USA
| | | | | | | | | | - Maya Arai
- BioMedicine Design, Pfizer Inc., Cambridge, MA, USA
| | | | | | - Lidia Mosyak
- BioMedicine Design, Pfizer Inc., Cambridge, MA, USA
| | | | | | | | | | - H. Lily Zhu
- BioMedicine Design, Pfizer Inc., Andover, MA, USA
| | | | | | | | - Alison Betts
- BioMedicine Design, Pfizer Inc., Cambridge, MA, USA
| | - Lindsay King
- BioMedicine Design, Pfizer Inc., Andover, MA, USA
| | - Edward Rosfjord
- Oncology Research & Development, Pfizer Inc., Pearl River, NY, USA
| | | | - Laura Lin
- BioMedicine Design, Pfizer Inc., Cambridge, MA, USA
| | - Puja Sapra
- Oncology Research & Development, Pfizer Inc., Pearl River, NY, USA
| | | | - Divya Mathur
- Oncology Research & Development, Pfizer Inc., Pearl River, NY, USA
| | - Laird Bloom
- BioMedicine Design, Pfizer Inc., Cambridge, MA, USA
| |
Collapse
|
36
|
Nanobodies as Versatile Tool for Multiscale Imaging Modalities. Biomolecules 2020; 10:biom10121695. [PMID: 33353213 PMCID: PMC7767244 DOI: 10.3390/biom10121695] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
Molecular imaging is constantly growing in different areas of preclinical biomedical research. Several imaging methods have been developed and are continuously updated for both in vivo and in vitro applications, in order to increase the information about the structure, localization and function of molecules involved in physiology and disease. Along with these progresses, there is a continuous need for improving labeling strategies. In the last decades, the single domain antigen-binding fragments nanobodies (Nbs) emerged as important molecular imaging probes. Indeed, their small size (~15 kDa), high stability, affinity and modularity represent desirable features for imaging applications, providing higher tissue penetration, rapid targeting, increased spatial resolution and fast clearance. Accordingly, several Nb-based probes have been generated and applied to a variety of imaging modalities, ranging from in vivo and in vitro preclinical imaging to super-resolution microscopy. In this review, we will provide an overview of the state-of-the-art regarding the use of Nbs in several imaging modalities, underlining their extreme versatility and their enormous potential in targeting molecules and cells of interest in both preclinical and clinical studies.
Collapse
|
37
|
Patasic L, Seifried J, Bezler V, Kaljanac M, Schneider IC, Schmitz H, Tondera C, Hartmann J, Hombach A, Buchholz CJ, Abken H, König R, Cichutek K. Designed Ankyrin Repeat Protein (DARPin) to target chimeric antigen receptor (CAR)-redirected T cells towards CD4 + T cells to reduce the latent HIV + cell reservoir. Med Microbiol Immunol 2020; 209:681-691. [PMID: 32918599 PMCID: PMC7568711 DOI: 10.1007/s00430-020-00692-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 08/19/2020] [Indexed: 10/25/2022]
Abstract
Chimeric Antigen Receptor (CAR)-redirected T cells show great efficacy in the patient-specific therapy of hematologic malignancies. Here, we demonstrate that a DARPin with specificity for CD4 specifically redirects and triggers the activation of CAR engineered T cells resulting in the depletion of CD4+ target cells aiming for elimination of the human immunodeficiency virus (HIV) reservoir.
Collapse
Affiliation(s)
- Lea Patasic
- Host-Pathogen Interactions, Paul-Ehrlich-Institut, Langen, Germany
| | - Janna Seifried
- Host-Pathogen Interactions, Paul-Ehrlich-Institut, Langen, Germany.
- Department for Infectious Disease Epidemiology, Robert Koch-Institute, Berlin, Germany.
| | - Valerie Bezler
- Regensburg Center for Interventional Immunology (RCI), Department of Genetic Immunotherapy, University Hospital Regensburg, Regensburg, Germany
| | - Marcell Kaljanac
- Regensburg Center for Interventional Immunology (RCI), Department of Genetic Immunotherapy, University Hospital Regensburg, Regensburg, Germany
| | - Irene C Schneider
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Langen, Germany
| | - Heike Schmitz
- Host-Pathogen Interactions, Paul-Ehrlich-Institut, Langen, Germany
| | | | - Jessica Hartmann
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Langen, Germany
| | - Andreas Hombach
- Center for Molecular Medicine Cologne, University of Cologne, and Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Christian J Buchholz
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Langen, Germany
| | - Hinrich Abken
- Center for Molecular Medicine Cologne, University of Cologne, and Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany
- Regensburg Center for Interventional Immunology (RCI), Department of Genetic Immunotherapy, University Hospital Regensburg, Regensburg, Germany
| | - Renate König
- Host-Pathogen Interactions, Paul-Ehrlich-Institut, Langen, Germany
- German Center for Infection Research (DZIF), Langen, Germany
- Immunity and Pathogenesis Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Klaus Cichutek
- Host-Pathogen Interactions, Paul-Ehrlich-Institut, Langen, Germany.
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Langen, Germany.
- German Center for Infection Research (DZIF), Langen, Germany.
| |
Collapse
|
38
|
Antibody Fragments as Tools for Elucidating Structure-Toxicity Relationships and for Diagnostic/Therapeutic Targeting of Neurotoxic Amyloid Oligomers. Int J Mol Sci 2020; 21:ijms21238920. [PMID: 33255488 PMCID: PMC7727795 DOI: 10.3390/ijms21238920] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/01/2020] [Accepted: 10/01/2020] [Indexed: 02/06/2023] Open
Abstract
The accumulation of amyloid protein aggregates in tissues is the basis for the onset of diseases known as amyloidoses. Intriguingly, many amyloidoses impact the central nervous system (CNS) and usually are devastating diseases. It is increasingly apparent that neurotoxic soluble oligomers formed by amyloidogenic proteins are the primary molecular drivers of these diseases, making them lucrative diagnostic and therapeutic targets. One promising diagnostic/therapeutic strategy has been the development of antibody fragments against amyloid oligomers. Antibody fragments, such as fragment antigen-binding (Fab), scFv (single chain variable fragments), and VHH (heavy chain variable domain or single-domain antibodies) are an alternative to full-length IgGs as diagnostics and therapeutics for a variety of diseases, mainly because of their increased tissue penetration (lower MW compared to IgG), decreased inflammatory potential (lack of Fc domain), and facile production (low structural complexity). Furthermore, through the use of in vitro-based ligand selection, it has been possible to identify antibody fragments presenting marked conformational selectivity. In this review, we summarize significant reports on antibody fragments selective for oligomers associated with prevalent CNS amyloidoses. We discuss promising results obtained using antibody fragments as both diagnostic and therapeutic agents against these diseases. In addition, the use of antibody fragments, particularly scFv and VHH, in the isolation of unique oligomeric assemblies is discussed as a strategy to unravel conformational moieties responsible for neurotoxicity. We envision that advances in this field may lead to the development of novel oligomer-selective antibody fragments with superior selectivity and, hopefully, good clinical outcomes.
Collapse
|
39
|
Selection and Characterization of YKL-40-Targeting Monoclonal Antibodies from Human Synthetic Fab Phage Display Libraries. Int J Mol Sci 2020; 21:ijms21176354. [PMID: 32883029 PMCID: PMC7504393 DOI: 10.3390/ijms21176354] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 12/14/2022] Open
Abstract
YKL-40, also known as chitinase-3-like 1 (CHI3L1), is a glycoprotein that is expressed and secreted by various cell types, including cancers and macrophages. Due to its implications for and upregulation in a variety of diseases, including inflammatory conditions, fibrotic disorders, and tumor growth, YKL-40 has been considered as a significant therapeutic biomarker. Here, we used a phage display to develop novel monoclonal antibodies (mAbs) targeting human YKL-40 (hYKL-40). Human synthetic antibody phage display libraries were panned against a recombinant hYKL-40 protein, yielding seven unique Fabs (Antigen-binding fragment), of which two Fabs (H1 and H2) were non-aggregating and thermally stable (75.5 °C and 76.5 °C, respectively) and had high apparent affinities (KD = 2.3 nM and 4.0 nM, respectively). Reformatting the Fabs into IgGs (Immunoglobulin Gs) increased their apparent affinities (notably, for H1 and H2, KD = 0.5 nM and 0.3 nM, respectively), presumably due to the effects of avidity, with little change to their non-aggregation property. The six anti-hYKL-40 IgGs were analyzed using a trans-well migration assay in vitro, revealing that three clones (H1, H2, and H4) were notably effective in reducing cell migration from both A549 and H460 lung cancer cell lines. The three clones were further analyzed in an in vivo animal test that assessed their anti-cancer activities, demonstrating that the tumor area and the number of tumor nodules were significantly reduced in the lung tissues treated with H1 (IgG). Given its high affinity and desirable properties, we expect that the H1 anti-hYKL-40 mAb will be a suitable candidate for developing anti-cancer therapeutics.
Collapse
|
40
|
Yang EY, Shah K. Nanobodies: Next Generation of Cancer Diagnostics and Therapeutics. Front Oncol 2020; 10:1182. [PMID: 32793488 PMCID: PMC7390931 DOI: 10.3389/fonc.2020.01182] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022] Open
Abstract
The development of targeted medicine has greatly expanded treatment options and spurred new research avenues in cancer therapeutics, with monoclonal antibodies (mAbs) emerging as a prevalent treatment in recent years. With mixed clinical success, mAbs still hold significant shortcomings, as they possess limited tumor penetration, high manufacturing costs, and the potential to develop therapeutic resistance. However, the recent discovery of “nanobodies,” the smallest-known functional antibody fragment, has demonstrated significant translational potential in preclinical and clinical studies. This review highlights their various applications in cancer and analyzes their trajectory toward their translation into the clinic.
Collapse
Affiliation(s)
- Emily Y Yang
- Center for Stem Cell Therapeutics and Imaging, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Departments of Neurosurgery, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States
| | - Khalid Shah
- Center for Stem Cell Therapeutics and Imaging, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Departments of Neurosurgery, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, United States
| |
Collapse
|
41
|
van der Kant R, Bauer J, Karow-Zwick AR, Kube S, Garidel P, Blech M, Rousseau F, Schymkowitz J. Adaption of human antibody λ and κ light chain architectures to CDR repertoires. Protein Eng Des Sel 2020; 32:109-127. [PMID: 31535139 PMCID: PMC6908821 DOI: 10.1093/protein/gzz012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 06/11/2019] [Indexed: 12/16/2022] Open
Abstract
Monoclonal antibodies bind with high specificity to a wide range of diverse antigens, primarily mediated by their hypervariable complementarity determining regions (CDRs). The defined antigen binding loops are supported by the structurally conserved β-sandwich framework of the light chain (LC) and heavy chain (HC) variable regions. The LC genes are encoded by two separate loci, subdividing the entity of antibodies into kappa (LCκ) and lambda (LCλ) isotypes that exhibit distinct sequence and conformational preferences. In this work, a diverse set of techniques were employed including machine learning, force field analysis, statistical coupling analysis and mutual information analysis of a non-redundant antibody structure collection. Thereby, it was revealed how subtle changes between the structures of LCκ and LCλ isotypes increase the diversity of antibodies, extending the predetermined restrictions of the general antibody fold and expanding the diversity of antigen binding. Interestingly, it was found that the characteristic framework scaffolds of κ and λ are stabilized by diverse amino acid clusters that determine the interplay between the respective fold and the embedded CDR loops. In conclusion, this work reveals how antibodies use the remarkable plasticity of the beta-sandwich Ig fold to incorporate a large diversity of CDR loops.
Collapse
Affiliation(s)
- Rob van der Kant
- Switch Laboratory, VIB Center for Brain and Disease Research, Herestraat 49, Leuven, Belgium.,Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49 Box, B-3000 Leuven, Belgium
| | - Joschka Bauer
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach/Riss, Germany
| | | | - Sebastian Kube
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach/Riss, Germany
| | - Patrick Garidel
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach/Riss, Germany
| | - Michaela Blech
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach/Riss, Germany
| | - Frederic Rousseau
- Switch Laboratory, VIB Center for Brain and Disease Research, Herestraat 49, Leuven, Belgium.,Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49 Box, B-3000 Leuven, Belgium
| | - Joost Schymkowitz
- Switch Laboratory, VIB Center for Brain and Disease Research, Herestraat 49, Leuven, Belgium.,Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49 Box, B-3000 Leuven, Belgium
| |
Collapse
|
42
|
Riaño-Umbarila L, Rojas-Trejo VM, Romero-Moreno JA, Costas M, Utrera-Espíndola I, Olamendi-Portugal T, Possani LD, Becerril B. Comparative assessment of the VH-VL and VL-VH orientations of single-chain variable fragments of scorpion toxin-neutralizing antibodies. Mol Immunol 2020; 122:141-147. [PMID: 32361416 DOI: 10.1016/j.molimm.2020.04.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/03/2020] [Accepted: 04/14/2020] [Indexed: 01/31/2023]
Abstract
The present study evaluated the effect of the change in the orientation of the VH-VL variable domains to VL-VH on the physicochemical and functional properties of two scorpion toxin-neutralizing scFvs. The results showed that the level of expression of proteins obtained from the periplasm of E. coli is the factor mainly affected, either with an increase or decrease in the amount of protein recovered. Likewise, the functional recognition activity in the presence of a denaturing agent showed slight variations in the two orientations. In contrast, recognition and biological activity (neutralizing capacity) are maintained. At the interaction level, the change marginally modified the kinetic association and dissociation constants without significantly modifying the value of the affinity constants. Similarly, it was observed that the thermodynamic stability of the proteins did not show significant variations either. These results contrast with some reports of the effect of changing the orientation of domains, suggesting that it is not possible to predict which orientation of the variable domains of an scFv is more favorable or if they are equivalent, as in the case of scFvs previously matured by directed evolution techniques.
Collapse
Affiliation(s)
- Lidia Riaño-Umbarila
- Cátedra CONACYT, Instituto de Biotecnología-Universidad Nacional Autónoma de México, Apartado Postal 510-3, Cuernavaca, Morelos, 62250, Mexico.
| | - Vianey Margarita Rojas-Trejo
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, UNAM, Apartado Postal 510-3, Cuernavaca, Morelos, 62250, Mexico
| | - José Alberto Romero-Moreno
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, UNAM, Apartado Postal 510-3, Cuernavaca, Morelos, 62250, Mexico
| | - Miguel Costas
- Laboratorio de Bio-Fisicoquímica, Departamento de Fisicoquímica, Facultad de Química, Cd. Universitaria, UNAM, Ciudad de México, 04510, Mexico
| | - Irving Utrera-Espíndola
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, UNAM, Apartado Postal 510-3, Cuernavaca, Morelos, 62250, Mexico
| | - Timoteo Olamendi-Portugal
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, UNAM, Apartado Postal 510-3, Cuernavaca, Morelos, 62250, Mexico
| | - Lourival D Possani
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, UNAM, Apartado Postal 510-3, Cuernavaca, Morelos, 62250, Mexico
| | - Baltazar Becerril
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, UNAM, Apartado Postal 510-3, Cuernavaca, Morelos, 62250, Mexico.
| |
Collapse
|
43
|
Najafi M, Safdari Y. Obtaining active recombinant proteins from bacterial inclusion bodies using salt solutions under neutral pH conditions. Protein Expr Purif 2020; 169:105586. [PMID: 32001358 DOI: 10.1016/j.pep.2020.105586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 10/25/2022]
Abstract
Eukaryotic recombinant proteins expressed in bacterial cells usually aggregate within the cells as inclusion bodies. Despite the widely-accepted theory considering inclusion bodies as inactive materials, inclusion bodies may contain large amounts of correctly-folded active recombinant proteins. Proteins trapped in inclusion bodies can be released using a high pH solution (pH ≥ 11); however, they may undergo structural changes in such pH conditions that may lead to their inactivation. Shifting in pH alongside the use of metal ions can help recover protein activity. The model protein we used in this study, 9R-Nimo.scFv, is highly active when extracted from bacterial inclusion bodies at high pH condition (pH 12) but loses its activity when pH is reduced to pH 7. We evaluated the capacity of nine salt solutions in terms of recovering protein activity in neutral pH conditions and found that ZnSO4 solution was the best one for this purpose. KNO3 and MnSO4 were also found to have a good capacity for recovering protein activity, as well.
Collapse
Affiliation(s)
- Marzieh Najafi
- Student Research Committee, Department of Medical Biotechnology, Faculty of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Yaghoub Safdari
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran; Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
44
|
Mittl PR, Ernst P, Plückthun A. Chaperone-assisted structure elucidation with DARPins. Curr Opin Struct Biol 2020; 60:93-100. [PMID: 31918361 DOI: 10.1016/j.sbi.2019.12.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/16/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022]
Abstract
Designed ankyrin repeat proteins (DARPins) are artificial binding proteins that have found many uses in therapy, diagnostics and biochemical research. They substantially extend the scope of antibody-derived binders. Their high affinity and specificity, rigidity, extended paratope, and facile bacterial production make them attractive for structural biology. Complexes with simple DARPins have been crystallized for a long time, but particularly the rigid helix fusion strategy has opened new opportunities. Rigid DARPin fusions expand crystallization space, enable recruitment of targets in a host lattice and reduce the size limit for cryo-EM. Besides applications in structural biology, rigid DARPin fusions also serve as molecular probes in cells to investigate spatial restraints in targets.
Collapse
Affiliation(s)
- Peer Re Mittl
- Department of Biochemistry, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Patrick Ernst
- Department of Biochemistry, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Andreas Plückthun
- Department of Biochemistry, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland.
| |
Collapse
|
45
|
Goulet DR, Atkins WM. Considerations for the Design of Antibody-Based Therapeutics. J Pharm Sci 2020; 109:74-103. [PMID: 31173761 PMCID: PMC6891151 DOI: 10.1016/j.xphs.2019.05.031] [Citation(s) in RCA: 189] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/02/2019] [Accepted: 05/29/2019] [Indexed: 02/06/2023]
Abstract
Antibody-based proteins have become an important class of biologic therapeutics, due in large part to the stability, specificity, and adaptability of the antibody framework. Indeed, antibodies not only have the inherent ability to bind both antigens and endogenous immune receptors but also have proven extremely amenable to protein engineering. Thus, several derivatives of the monoclonal antibody format, including bispecific antibodies, antibody-drug conjugates, and antibody fragments, have demonstrated efficacy for treating human disease, particularly in the fields of immunology and oncology. Reviewed here are considerations for the design of antibody-based therapeutics, including immunological context, therapeutic mechanisms, and engineering strategies. First, characteristics of antibodies are introduced, with emphasis on structural domains, functionally important receptors, isotypic and allotypic differences, and modifications such as glycosylation. Then, aspects of therapeutic antibody design are discussed, including identification of antigen-specific variable regions, choice of expression system, use of multispecific formats, and design of antibody derivatives based on fragmentation, oligomerization, or conjugation to other functional moieties. Finally, strategies to enhance antibody function through protein engineering are reviewed while highlighting the impact of fundamental biophysical properties on protein developability.
Collapse
Affiliation(s)
- Dennis R Goulet
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195.
| | - William M Atkins
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195
| |
Collapse
|
46
|
Banaszek A, Bumm TGP, Nowotny B, Geis M, Jacob K, Wölfl M, Trebing J, Kucka K, Kouhestani D, Gogishvili T, Krenz B, Lutz J, Rasche L, Hönemann D, Neuweiler H, Heiby JC, Bargou RC, Wajant H, Einsele H, Riethmüller G, Stuhler G. On-target restoration of a split T cell-engaging antibody for precision immunotherapy. Nat Commun 2019; 10:5387. [PMID: 31772172 PMCID: PMC6879491 DOI: 10.1038/s41467-019-13196-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 10/02/2019] [Indexed: 12/19/2022] Open
Abstract
T cell-engaging immunotherapies are changing the landscape of current cancer care. However, suitable target antigens are scarce, restricting these strategies to very few tumor types. Here, we report on a T cell-engaging antibody derivative that comes in two complementary halves and addresses antigen combinations instead of single molecules. Each half, now coined hemibody, contains an antigen-specific single-chain variable fragment (scFv) fused to either the variable light (VL) or variable heavy (VH) chain domain of an anti-CD3 antibody. When the two hemibodies simultaneously bind their respective antigens on a single cell, they align and reconstitute the original CD3-binding site to engage T cells. Employing preclinical models for aggressive leukemia and breast cancer, we show that by the combinatorial nature of this approach, T lymphocytes exclusively eliminate dual antigen-positive cells while sparing single positive bystanders. This allows for precision targeting of cancers not amenable to current immunotherapies. The restriction of appropriate tumour-specific antigens is a current limitation for T cell-engaging immunotherapy. Here, the authors have designed a new system constituted by two halve antibodies, which engage T cells once binding to two different antigens, to specifically eliminate double positive cells in preclinical leukemia and breast cancer mouse models.
Collapse
Affiliation(s)
- Agnes Banaszek
- University Clinic Würzburg, Department of Internal Medicine II, Hematology and Oncology, Würzburg, Germany
| | - Thomas G P Bumm
- University Clinic Würzburg, Department of Internal Medicine II, Hematology and Oncology, Würzburg, Germany
| | - Boris Nowotny
- University Clinic Würzburg, Department of Internal Medicine II, Hematology and Oncology, Würzburg, Germany
| | - Maria Geis
- University Clinic Würzburg, Department of Internal Medicine II, Hematology and Oncology, Würzburg, Germany
| | - Kim Jacob
- University Clinic Würzburg, Department of Internal Medicine II, Hematology and Oncology, Würzburg, Germany
| | - Matthias Wölfl
- University Clinic Würzburg, Children's Hospital, Pediatric Oncology, Hematology and Stem Cell Transplantation, Würzburg, Germany
| | - Johannes Trebing
- University Clinic Würzburg, Department of Internal Medicine II, Division of Molecular Internal Medicine, Würzburg, Germany
| | - Kirstin Kucka
- University Clinic Würzburg, Department of Internal Medicine II, Division of Molecular Internal Medicine, Würzburg, Germany
| | - Dina Kouhestani
- University Clinic Würzburg, Department of Internal Medicine II, Hematology and Oncology, Würzburg, Germany
| | - Tea Gogishvili
- University Clinic Würzburg, Department of Internal Medicine II, Hematology and Oncology, Würzburg, Germany
| | - Bastian Krenz
- University Clinic Würzburg, Department of Internal Medicine II, Hematology and Oncology, Würzburg, Germany
| | - Justina Lutz
- University Clinic Würzburg, Department of Internal Medicine II, Hematology and Oncology, Würzburg, Germany
| | - Leo Rasche
- University Clinic Würzburg, Department of Internal Medicine II, Hematology and Oncology, Würzburg, Germany
| | - Dirk Hönemann
- University Clinic Würzburg, Department of Internal Medicine II, Hematology and Oncology, Würzburg, Germany
| | - Hannes Neuweiler
- Department of Biotechnology and Biophysics, University Würzburg, Würzburg, Germany
| | - Julia C Heiby
- Department of Biotechnology and Biophysics, University Würzburg, Würzburg, Germany
| | - Ralf C Bargou
- University Clinic Würzburg, Department of Internal Medicine II, Hematology and Oncology, Würzburg, Germany.,Comprehensive Cancer Center Mainfranken, Universitätsklinikum, Würzburg, Germany
| | - Harald Wajant
- University Clinic Würzburg, Department of Internal Medicine II, Division of Molecular Internal Medicine, Würzburg, Germany
| | - Hermann Einsele
- University Clinic Würzburg, Department of Internal Medicine II, Hematology and Oncology, Würzburg, Germany
| | - Gert Riethmüller
- Ludwig-Maximilians-University, Institute for Immunology, Munich, Germany
| | - Gernot Stuhler
- University Clinic Würzburg, Department of Internal Medicine II, Hematology and Oncology, Würzburg, Germany.
| |
Collapse
|
47
|
Generation of a Nebulizable CDR-Modified MERS-CoV Neutralizing Human Antibody. Int J Mol Sci 2019; 20:ijms20205073. [PMID: 31614869 PMCID: PMC6829326 DOI: 10.3390/ijms20205073] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/10/2019] [Accepted: 10/10/2019] [Indexed: 12/26/2022] Open
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) induces severe aggravating respiratory failure in infected patients, frequently resulting in mechanical ventilation. As limited therapeutic antibody is accumulated in lung tissue following systemic administration, inhalation is newly recognized as an alternative, possibly better, route of therapeutic antibody for pulmonary diseases. The nebulization process, however, generates diverse physiological stresses, and thus, the therapeutic antibody must be resistant to these stresses, remain stable, and form minimal aggregates. We first isolated a MERS-CoV neutralizing antibody that is reactive to the receptor-binding domain (RBD) of spike (S) glycoprotein. To increase stability, we introduced mutations into the complementarity-determining regions (CDRs) of the antibody. In the HCDRs (excluding HCDR3) in this clone, two hydrophobic residues were replaced with Glu, two residues were replaced with Asp, and four residues were replaced with positively charged amino acids. In LCDRs, only two Leu residues were replaced with Val. These modifications successfully generated a clone with significantly greater stability and equivalent reactivity and neutralizing activity following nebulization compared to the original clone. In summary, we generated a MERS-CoV neutralizing human antibody that is reactive to recombinant MERS-CoV S RBD protein for delivery via a pulmonary route by introducing stabilizing mutations into five CDRs.
Collapse
|
48
|
Teng Y, Young JL, Edwards B, Hayes P, Thompson L, Johnston C, Edwards C, Sanders Y, Writer M, Pinto D, Zhang Y, Roode M, Chovanec P, Matheson L, Corcoran AE, Fernandez A, Montoliu L, Rossi B, Tosato V, Gjuracic K, Nikitin D, Bruschi C, McGuinness B, Sandal T, Romanos M. Diverse human V H antibody fragments with bio-therapeutic properties from the Crescendo Mouse. N Biotechnol 2019; 55:65-76. [PMID: 31600579 DOI: 10.1016/j.nbt.2019.10.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 09/04/2019] [Accepted: 10/04/2019] [Indexed: 01/26/2023]
Abstract
We describe the 'Crescendo Mouse', a human VH transgenic platform combining an engineered heavy chain locus with diverse human heavy chain V, D and J genes, a modified mouse Cγ1 gene and complete 3' regulatory region, in a triple knock-out (TKO) mouse background devoid of endogenous immunoglobulin expression. The addition of the engineered heavy chain locus to the TKO mouse restored B cell development, giving rise to functional B cells that responded to immunization with a diverse response that comprised entirely 'heavy chain only' antibodies. Heavy chain variable (VH) domain libraries were rapidly mined using phage display technology, yielding diverse high-affinity human VH that had undergone somatic hypermutation, lacked aggregation and showed enhanced expression in E. coli. The Crescendo Mouse produces human VH fragments, or Humabody® VH, with excellent bio-therapeutic potential, as exemplified here by the generation of antagonistic Humabody® VH specific for human IL17A and IL17RA.
Collapse
Affiliation(s)
- Yumin Teng
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK.
| | - Joyce L Young
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Bryan Edwards
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Philip Hayes
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Lorraine Thompson
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Colette Johnston
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Carolyn Edwards
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Yun Sanders
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Michele Writer
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Debora Pinto
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Yanjing Zhang
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Mila Roode
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Peter Chovanec
- Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Louise Matheson
- Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Anne E Corcoran
- Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Almudena Fernandez
- Centro Nacional de Biotecnologia (CNB-CSIC) & CIBER de Enfermedades Raras (CIBERER-ISCIII), Darwin 3, 28049, Madrid, Spain
| | - Lluis Montoliu
- Centro Nacional de Biotecnologia (CNB-CSIC) & CIBER de Enfermedades Raras (CIBERER-ISCIII), Darwin 3, 28049, Madrid, Spain
| | - Beatrice Rossi
- International Centre for Genetic Engineering and Biotechnology, Yeast Molecular Genetics Laboratory, Padriciano 99, 34149, Trieste, Italy
| | - Valentina Tosato
- International Centre for Genetic Engineering and Biotechnology, Yeast Molecular Genetics Laboratory, Padriciano 99, 34149, Trieste, Italy
| | - Kresimir Gjuracic
- International Centre for Genetic Engineering and Biotechnology, Yeast Molecular Genetics Laboratory, Padriciano 99, 34149, Trieste, Italy
| | - Dmitri Nikitin
- International Centre for Genetic Engineering and Biotechnology, Yeast Molecular Genetics Laboratory, Padriciano 99, 34149, Trieste, Italy
| | - Carlo Bruschi
- International Centre for Genetic Engineering and Biotechnology, Yeast Molecular Genetics Laboratory, Padriciano 99, 34149, Trieste, Italy
| | - Brian McGuinness
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Thomas Sandal
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Mike Romanos
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| |
Collapse
|
49
|
Ollier R, Wassmann P, Monney T, Ries Fecourt C, Gn S, C A V, Ayoub D, Stutz C, Gudi GS, Blein S. Single-step Protein A and Protein G avidity purification methods to support bispecific antibody discovery and development. MAbs 2019; 11:1464-1478. [PMID: 31462177 PMCID: PMC6816383 DOI: 10.1080/19420862.2019.1660564] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Heavy chain (Hc) heterodimers represent a majority of bispecific antibodies (bsAbs) under clinical development. Although recent technologies achieve high levels of Hc heterodimerization (HD), traces of homodimer contaminants are often present, and as a consequence robust purification techniques for generating highly pure heterodimers in a single step are needed. Here, we describe two different purification methods that exploit differences in Protein A (PA) or Protein G (PG) avidity between homo- and heterodimers. Differential elution between species was enabled by removing PA or PG binding in one of the Hcs of the bsAb. The PA method allowed the avidity purification of heterodimers based on the VH3 subclass, which naturally binds PA and interferes with separation, by using a combination of IgG3 Fc and a single amino acid change in VH3, N82aS. The PG method relied on a combination of three mutations that completely disrupts PG binding, M428G/N434A in IgG1 Fc and K213V in IgG1 CH1. Both methods achieved a high level of heterodimer purity as single-step techniques without Hc HD (93–98%). Since PA and PG have overlapping binding sites with the neonatal Fc receptor (FcRn), we investigated the effects of our engineering both in vitro and in vivo. Mild to moderate differences in FcRn binding and Fc thermal stability were observed, but these did not significantly change the serum half-lives of engineered control antibodies and heterodimers. The methods are conceptually compatible with various Hc HD platforms such as BEAT® (Bispecific Engagement by Antibodies based on the T cell receptor), in which the PA method has already been successfully implemented.
Collapse
Affiliation(s)
- Romain Ollier
- Department of Antibody Engineering, Glenmark Biotherapeutics SA, Biopôle Lausanne - Epalinges, Bâtiment SE-B , Epalinges , Switzerland
| | - Paul Wassmann
- Department of Antibody Engineering, Glenmark Biotherapeutics SA, Biopôle Lausanne - Epalinges, Bâtiment SE-B , Epalinges , Switzerland
| | - Thierry Monney
- Department of Antibody Engineering, Glenmark Biotherapeutics SA, Biopôle Lausanne - Epalinges, Bâtiment SE-B , Epalinges , Switzerland
| | - Christelle Ries Fecourt
- Department of Antibody Engineering, Glenmark Biotherapeutics SA, Biopôle Lausanne - Epalinges, Bâtiment SE-B , Epalinges , Switzerland
| | - Sunitha Gn
- Department of Drug Metabolism and Pharmacokinetics, Glenmark Pharmaceuticals Limited, Glenmark Research Centre , Navi Mumbai , India
| | - Vinu C A
- Department of Drug Metabolism and Pharmacokinetics, Glenmark Pharmaceuticals Limited, Glenmark Research Centre , Navi Mumbai , India
| | - Daniel Ayoub
- Department of Formulation and Analytical Development, Glenmark Pharmaceuticals SA , La Chaux-de-Fonds , Switzerland
| | - Cian Stutz
- Department of Antibody Engineering, Glenmark Biotherapeutics SA, Biopôle Lausanne - Epalinges, Bâtiment SE-B , Epalinges , Switzerland
| | - Girish S Gudi
- Department of Drug Metabolism and Pharmacokinetics, Glenmark Pharmaceuticals Inc ., Paramus , NJ , USA
| | - Stanislas Blein
- Department of Antibody Engineering, Glenmark Biotherapeutics SA, Biopôle Lausanne - Epalinges, Bâtiment SE-B , Epalinges , Switzerland
| |
Collapse
|
50
|
Optimization of an Antibody Light Chain Framework Enhances Expression, Biophysical Properties and Pharmacokinetics. Antibodies (Basel) 2019; 8:antib8030046. [PMID: 31544852 PMCID: PMC6784111 DOI: 10.3390/antib8030046] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/26/2019] [Accepted: 08/30/2019] [Indexed: 11/17/2022] Open
Abstract
Efficacy, safety, and manufacturability of therapeutic antibodies are influenced by their biopharmaceutical and biophysical properties. These properties can be optimized by library approaches or rationale protein design. Here, we employed a protein engineering approach to modify the variable domain of the light chain (VL) framework of an oxidized macrophage migration inhibitory factor (oxMIF)-specific antibody. The amendment of the antibody sequence was based on homology to human germline VL genes. Three regions or positions were identified in the VL domain—L1-4, L66, L79—and mutated independently or in combination to match the closest germline V gene. None of the mutations altered oxMIF specificity or affinity, but some variants improved thermal stability, aggregation propensity, and resulted in up to five-fold higher expression. Importantly, the improved biopharmaceutical properties translated into a superior pharmacokinetic profile of the antibody. Thus, optimization of the V domain framework can ameliorate the biophysical qualities of a therapeutic antibody candidate, and as result its manufacturability, and also has the potential to improve pharmacokinetics.
Collapse
|