1
|
Li M, Zhan Y, Wu B, Qin Y, Gao J, Liu L, Gong R. Comparison of C-type Nanoantibody Produced in Different Expression Systems Implying Potential Clinical Applications. Mol Pharm 2025; 22:387-396. [PMID: 39630426 DOI: 10.1021/acs.molpharmaceut.4c00980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
In the pharmaceutical industry, the Chinese hamster ovary cell, a type of mammalian cell, is extensively employed for the production of conventional full-length monoclonal antibodies. Nanobody is one of the most attractive directions for the development of next-generation antibody drugs. However, a suitable expression system for its manufacture has not yet been comprehensively evaluated. Previously, we proposed that the immunoglobulin constant CH2 domain could be a promising scaffold for developing C-type nanoantibodies (C-Nabs) as candidate therapeutics. Here, we used an antiviral C-Nab, which we identified previously (under review), as a model for investigation. We expressed C-Nabs without a tag in different systems, including a bacterium (C-Nabbac), yeast (C-Nabyeast), and mammalian cell (C-Nabmam). After purification, the binding and neutralizing activities of C-Nabs from different expression systems are similar. Their secondary structures are rich in β-strand. The melting temperatures of C-Nabbac (71.5 °C) and C-Nabmam (70.2 °C) are similar, which are slightly higher than that of C-Nabyeast (65.6 °C), while C-Nabyeast and C-Nabmam are more resistant to urea-induced unfolding than C-Nabbac. C-Nabyeast and C-Nabmam demonstrate higher resistance to aggregation compared to C-Nabbac. C-Nabyeast exhibits greater resistance to enzyme digestion compared to C-Nabbac and C-Nabmam. Notably, when administered via intraperitoneal injection in mice, C-Nabyeast shows superior pharmacokinetics. Overall, after comparing C-Nab proteins from various expression systems, we determined that yeast is the most suitable host for producing C-Nabs. This finding is beneficial for the production of nanobodies as potential drug candidates.
Collapse
Affiliation(s)
- Moxuan Li
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430207, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yancheng Zhan
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430207, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bihao Wu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430207, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ye Qin
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430207, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiazhao Gao
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430207, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lan Liu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430207, China
| | - Rui Gong
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430207, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Hubei Jiangxia Laboratory, Wuhan, Hubei 430200, China
| |
Collapse
|
2
|
Li C, Yao QQ, Li J. Druggability properties of a L309K mutation in the antibody CH2 domain. 3 Biotech 2024; 14:152. [PMID: 38742229 PMCID: PMC11088599 DOI: 10.1007/s13205-024-04000-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/28/2024] [Indexed: 05/16/2024] Open
Abstract
In the early stages of antibody drug development, it is imperative to conduct a comprehensive assessment and enhancement of the druggability attributes of potential molecules by considering their fundamental physicochemical properties. This study specifically concentrates on the surface-exposed hydrophobic region of the candidate antibody aPDL1-WT and explores the effectiveness of the L309K mutation strategy. The resulting aPDL1-LK variant demonstrates a notable enhancement over the original antibody in addressing the issue of aggregation and formation of large molecular impurities under accelerated high-temperature conditions. The mutated molecule, aPDL1-LK, exhibits excellent physicochemical properties such as hydrophilicity, conformational stability, charge variant stability, post-translational modifications, and serum stability. In terms of biological function, aPDL1-LK maintains the same glycosylation pattern as the original antibody and shows no significant difference in affinity for antigen hPDL1 protein, CD16a-F158, CD64, CD32a-H131, and complement C1q, compared to aPDL1-WT. The L309K mutation results in an approximately twofold reduction in its affinity for CD16a-V158 and CD32a-R131. In vitro biological assays, including antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), and complement-dependent cytotoxicity (CDC), reveal that the L309K mutation may decrease CD16a-V158-mediated ADCC activity due to the mutation-induced decrease in ligand affinity, while not affect CD32a-R131-mediated ADCP activity. In conclusion, the L309K mutation offers a promising strategy to enhance the druggability properties of candidate antibodies.
Collapse
Affiliation(s)
- Cui Li
- Department of Pharmacy, Zhejiang Provincial Hospital of Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310000 Zhejiang China
| | - Qing-qing Yao
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, 215000 Jiangsu China
| | - Jiang Li
- Department of Pharmacy, Zhejiang Provincial Hospital of Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310000 Zhejiang China
| |
Collapse
|
3
|
Ge S, Dias ACP, Zhang X. Chimerism of avian IgY-scFv and truncated IgG-Fc: A novel strategy in cross-species antibody generation and enhancement. Immunology 2024; 172:46-60. [PMID: 38247105 DOI: 10.1111/imm.13752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 12/15/2023] [Indexed: 01/23/2024] Open
Abstract
Chicken single-chain fragment variable (IgY-scFv) is a functional fragment and an emerging development in genetically engineered antibodies with a wide range of biomedical applications. However, scFvs have considerably shorter serum half-life due to the absence of antibody Fc region compared with the full-length antibody, and usually requires continuous intravenous administration for efficacy. A promising approach to overcome this limitation is to fuse scFv with immunoglobulin G (IgG) Fc region, for better recognition and mediation by the neonatal Fc receptor (FcRn) in the host. In this study, engineered mammalian ΔFc domains (CH2, CH3, and intact Fc region) were fused with anti-canine parvovirus-like particles avian IgY-scFv to produce chimeric antibodies and expressed in the HEK293 cell expression system. The obtained scFv-CH2, scFv-CH3, and scFv-Fc can bind with antigen specifically and dose-dependently. Surface plasmon resonance investigation confirmed that scFv-CH2, scFv-CH3, and scFv-Fc had different degrees of binding to FcRn, with scFv-Fc showing the highest affinity. scFv-Fc had a significantly longer half-life in mice compared with the unfused scFv. The identified ΔFcs are promising for the development of engineered Fc-based therapeutic antibodies and proteins with longer half-lives. The avian IgY-scFv-mammalian IgG Fc region opens up new avenues for antibody engineering, and it is a novel strategy to enhance the rapid development and screening of functional antibodies in veterinary and human medicine.
Collapse
Affiliation(s)
- Shikun Ge
- Department of Biology, Centre of Molecular and Environmental Biology, University of Minho, Braga, Portugal
- College of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, China
| | - Alberto Carlos Pires Dias
- Department of Biology, Centre of Molecular and Environmental Biology, University of Minho, Braga, Portugal
| | - Xiaoying Zhang
- Department of Biology, Centre of Molecular and Environmental Biology, University of Minho, Braga, Portugal
- College of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, China
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
4
|
Oyama K, Nakakido M, Ohkuri T, Nakamura H, Tsumoto K, Ueda T. Enhancing thermal stability in the CH 2 domain to suppress aggregation through the introduction of simultaneous disulfide bonds in Pichia pastoris. Protein Sci 2023; 32:e4831. [PMID: 37924310 PMCID: PMC10680342 DOI: 10.1002/pro.4831] [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: 09/05/2023] [Revised: 10/21/2023] [Accepted: 11/01/2023] [Indexed: 11/06/2023]
Abstract
Protein aggregations decrease production yields and impair the efficacy of therapeutics. The CH2 domain is a crucial part of the constant region of human IgG. But, it is also the least stable domain in IgG, which can result in antibody instability and aggregation problems. We created a novel mutant of the CH2 domain (T250C/L314C, mut10) by introducing a disulfide bond and expressed it using Pichia pastoris. The mut10 variant exhibited enhanced thermal stability, resistance to enzymatic degradation, and reduced aggregation in comparison to the original CH2 domain. However, it was less stable than mut20 (L242C/K334C), which is the variant prepared in a previous study (Gong et al., J. Biol. Chem., 2009). A more advanced mutant, mut25, was created by combining mut10 and mut20. Mut25 artificially contains two disulfide bonds. The new mutant, mut25, showed enhanced thermal stability, increased resistance to enzymatic digestion, and reduced aggregation in comparison to mut20. According to our knowledge, mut25 achieves an unprecedented level of stability among the humanized whole CH2 domains that have been reported so far. Mut25 has the potential to serve as a new platform for antibody therapeutics due to its ability to reduce immunogenicity by decreasing aggregation.
Collapse
Affiliation(s)
- Kosuke Oyama
- Graduate School of Pharmaceutical SciencesKyushu UniversityFukuokaJapan
| | - Makoto Nakakido
- Graduate School of EngineeringThe University of TokyoTokyoJapan
| | | | - Hitomi Nakamura
- Faculty of Pharmaceutical SciencesSojo UniversityKumamotoJapan
| | - Kouhei Tsumoto
- Graduate School of EngineeringThe University of TokyoTokyoJapan
| | - Tadashi Ueda
- Graduate School of Pharmaceutical SciencesKyushu UniversityFukuokaJapan
| |
Collapse
|
5
|
Cano-Garrido O, Serna N, Unzueta U, Parladé E, Mangues R, Villaverde A, Vázquez E. Protein scaffolds in human clinics. Biotechnol Adv 2022; 61:108032. [PMID: 36089254 DOI: 10.1016/j.biotechadv.2022.108032] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/30/2022] [Accepted: 09/03/2022] [Indexed: 11/02/2022]
Abstract
Fundamental clinical areas such as drug delivery and regenerative medicine require biocompatible materials as mechanically stable scaffolds or as nanoscale drug carriers. Among the wide set of emerging biomaterials, polypeptides offer enticing properties over alternative polymers, including full biocompatibility, biodegradability, precise interactivity, structural stability and conformational and functional versatility, all of them tunable by conventional protein engineering. However, proteins from non-human sources elicit immunotoxicities that might bottleneck further development and narrow their clinical applicability. In this context, selecting human proteins or developing humanized protein versions as building blocks is a strict demand to design non-immunogenic protein materials. We review here the expanding catalogue of human or humanized proteins tailored to execute different levels of scaffolding functions and how they can be engineered as self-assembling materials in form of oligomers, polymers or complex networks. In particular, we emphasize those that are under clinical development, revising their fields of applicability and how they have been adapted to offer, apart from mere mechanical support, highly refined functions and precise molecular interactions.
Collapse
Affiliation(s)
- Olivia Cano-Garrido
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès (Barcelona), Spain
| | - Naroa Serna
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès (Barcelona), Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 08193 Cerdanyola del Vallès (Barcelona), Spain
| | - Ugutz Unzueta
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 08193 Cerdanyola del Vallès (Barcelona), Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès (Barcelona), Spain; Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025 Barcelona, Spain; Josep Carreras Leukaemia Research Institute, 08916 Badalona (Barcelona), Spain
| | - Eloi Parladé
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès (Barcelona), Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 08193 Cerdanyola del Vallès (Barcelona), Spain
| | - Ramón Mangues
- Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025 Barcelona, Spain; Josep Carreras Leukaemia Research Institute, 08916 Badalona (Barcelona), Spain
| | - Antonio Villaverde
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès (Barcelona), Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 08193 Cerdanyola del Vallès (Barcelona), Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès (Barcelona), Spain.
| | - Esther Vázquez
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès (Barcelona), Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 08193 Cerdanyola del Vallès (Barcelona), Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès (Barcelona), Spain.
| |
Collapse
|
6
|
Wang Q, Gong R. Immunotherapy targeting mesothelin in acute myeloid leukemia. J Leukoc Biol 2022; 112:813-821. [PMID: 35946307 DOI: 10.1002/jlb.5mr0622-483r] [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: 03/01/2022] [Revised: 06/06/2022] [Indexed: 11/08/2022] Open
Abstract
Mesothelin (MSLN) is an emerging target that exists in soluble and membrane-associated forms. It is usually used for the diagnosis and treatment of MSLN-positive solid tumors. Interestingly, recent studies have shown that MSLN is highly expressed in 36% of acute myeloid leukemia (AML) patients and barely expressed in normal hematopoietic cells, which makes MSLN a promising target for the treatment of AML. It has been shown that MSLN is detectable as a diagnostic marker in its soluble form. Although the mechanism of action is unclear, MSLN remains a promising target for immunotherapy. Most MSLN research has been conducted in solid tumors, and less research has been conducted in hematopoietic tumors. Increasing research on MSLN is underway in AML, a hematopoietic neoplasm. For example, MSLN is related to extramedullary disease, minimal residual disease, and relapse in AML patients. Decreasing the expression of MSLN reduces the severity of the disease course. This information suggests that MSLN may be an ideal target for the treatment of many AML-related diseases to improve the prognosis and survival rate. At present, there are a few immunotherapies targeting MSLN in AML in preclinical and clinical trials, such as antibody-drug conjugates, bispecific T-cell engagers, and chimeric antigen receptor-T cells, which opens new room for the treatment of MSLN-related AML.
Collapse
Affiliation(s)
- Qingguang Wang
- 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
| | - 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
| |
Collapse
|
7
|
Ni D, Kırtel O, Yin D, Xu W, Chen Q, Öner ET, Mu W. Improving the catalytic behaviors of Lactobacillus-derived fructansucrases by truncation strategies. Enzyme Microb Technol 2021; 149:109857. [PMID: 34311894 DOI: 10.1016/j.enzmictec.2021.109857] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/04/2021] [Accepted: 06/21/2021] [Indexed: 01/17/2023]
Abstract
Fructansucrases (FSs), including inulosucrase (IS) and levansucrase (LS), are the members of the Glycoside Hydrolase family 68 (GH68) enzymes. IS and LS catalyze the polymerization of the fructosyl moiety from sucrose to inulin- and levan-type fructans, respectively. Lactobacillus-derived FSs have relatively extended N- and C-terminal sequences. However, the functional roles of these sequences in their enzymatic properties and fructan biosynthesis remain largely unknown. Limosilactobacillus reuteri (basionym: Lactobacillus reuteri) 121 could produce both IS and LS, abbreviated as Lare121-IS and Lare121-LS, respectively. In this study, it was found that the terminal truncation displayed an obvious effect on their activities and the N-terminal truncated variants, Lare121-ISΔ177-701 and Lare121-LSΔ154-686, displayed the highest activities. Melting temperature (Tm) and the thermostability at 50 °C were measured to evaluate the stability of various truncated versions, revealing the different effects of N-terminal on the stability. The average molecular weight and polymerization degree of the fructans produced by different truncated variants did not change considerably, indicating that N-terminal truncation had low influence on fructan biosynthesis. In addition, it was found that N-terminal truncation could also improve the activity of other reported FSs from Lactobacillus species.
Collapse
Affiliation(s)
- Dawei Ni
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Onur Kırtel
- IBSB-Industrial Biotechnology and Systems Biology Research Group, Department of Bioengineering, Marmara University, Göztepe Campus, Istanbul, Turkey
| | - Dejing Yin
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Wei Xu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qiuming Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ebru Toksoy Öner
- IBSB-Industrial Biotechnology and Systems Biology Research Group, Department of Bioengineering, Marmara University, Göztepe Campus, Istanbul, Turkey
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, 214122, China.
| |
Collapse
|
8
|
Pastrana B, Nieves S, Li W, Liu X, Dimitrov DS. Developability Assessment of an Isolated C H2 Immunoglobulin Domain. Anal Chem 2021; 93:1342-1351. [PMID: 33325681 DOI: 10.1021/acs.analchem.0c02663] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The IgG CH2 domain continues to hold promise for the development of new therapeutic entities because of its bifunctional role as a biomarker and effector protein. The need for further understanding of molecular stability and aggregation in therapeutic proteins has led to the development of a breakthrough quantum cascade laser microscope to allow for real-time comparability assessment of an array of related proteins in solution upon thermal perturbation. Our objective was to perform a comprehensive developability assessment of three similar monoclonal antibody (mAb) fragments: CH2, CH2s, and m01s. The CH2 construct consists of residues Pro238 to Lys340 of the IgG1 heavy chain sequence. CH2s has a 7-residue deletion at the N-terminus and a 16-residue C-terminal extension containing a histidine tag. The m01s construct is identical to CH2s, except for two cysteines introduced at positions 242 and 334. A series of hyperspectral images was acquired during thermal perturbation from 28 to 60 °C for all three proteins in an array. Co-distribution and two-dimensional infrared correlation spectroscopies yielded the mechanism of aggregation and stability for these three proteins. The level of detail is unprecedented, identifying the regions within CH2 and CH2s that are prone to self-association and establishing the differences in stability. Furthermore, CH2 helical segments, β-sheets, β-turns, and random coil regions were less stable than in CH2s and m01s because of the presence of the N-terminal 310-helix and β-turn type III. The engineered disulfide bridge in m01s eliminated the self-association process and rendered this mAb fragment the most stable.
Collapse
Affiliation(s)
- Belinda Pastrana
- Protein Dynamic Solutions, 9 Audubon Road, Wakefield, Massachusetts 01880-1256, United States
| | - Sherly Nieves
- Protein Dynamic Solutions, 9 Audubon Road, Wakefield, Massachusetts 01880-1256, United States
| | - Wei Li
- National Cancer Institute, Frederick, Maryland 21702-1201, United States.,Department of Medicine, Center for Antibody Therapeutics, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, Pennsylvania 15261, United States
| | - Xianglei Liu
- Department of Medicine, Center for Antibody Therapeutics, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, Pennsylvania 15261, United States
| | - Dimiter S Dimitrov
- National Cancer Institute, Frederick, Maryland 21702-1201, United States.,Department of Medicine, Center for Antibody Therapeutics, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, Pennsylvania 15261, United States
| |
Collapse
|
9
|
Cao G, Gao X, Zhan Y, Wang Q, Zhang Z, Dimitrov DS, Gong R. An engineered human IgG1 CH2 domain with decreased aggregation and nonspecific binding. MAbs 2021; 12:1689027. [PMID: 31795802 PMCID: PMC6927756 DOI: 10.1080/19420862.2019.1689027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The immunoglobulin (Ig) CH2 domain is a promising scaffold for the development of candidate therapeutics. We have previously shown that the stability of isolated CH2 could be increased by the introduction of an additional disulfide bond and removal of seven N-terminal residues (m01s). However, both isolated CH2 and m01s aggregate, likely due to the existence of aggregation-prone regions (APRs) that we identified by using computational methods. This knowledge was used to generate a phage display library of mutants. The library was incubated at high temperature to remove aggregating CH2 domains, and then panned against a mouse anti-human CH2 monoclonal antibody targeting a conformational epitope to remove misfolded CH2s. After two rounds of panning, one clone, m01s5, with smaller APRs, was identified. After additional mutagenesis one clone, m01s5.4, which aggregated much less than m01s as measured by a turbidity assay and dynamic light scattering, was identified. m01s5.4 also exhibited much lower nonspecific binding than m01s. Engineering of a previously identified m01s-based tumor antigen-specific binder led to a dramatic reduction of its aggregation without affecting its binding. In summary, we describe a new approach for reducing aggregation based on a combination of computational and phage display methodologies, and show that aggregation of CH2-based scaffolds can be significantly reduced by the newly identified mutants, which can improve the developability of potential CH2-based therapeutics.
Collapse
Affiliation(s)
- Guangcan Cao
- 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
| | - Xinyu Gao
- 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
| | - Yancheng Zhan
- 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
| | - Qingguang Wang
- 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
| | - Dimiter S Dimitrov
- Center for Antibody Therapeutics, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA
| | - 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
| |
Collapse
|
10
|
da Silva ES, Aglas L, Pinheiro CS, de Andrade Belitardo EMM, Silveira EF, Huber S, Torres RT, Wallner M, Briza P, Lackner P, Laimer J, Pacheco LGC, Cruz ÁA, Alcântara-Neves NM, Ferreira F. A hybrid of two major Blomia tropicalis allergens as an allergy vaccine candidate. Clin Exp Allergy 2020; 50:835-847. [PMID: 32314444 PMCID: PMC7384089 DOI: 10.1111/cea.13611] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/27/2020] [Accepted: 04/07/2020] [Indexed: 12/21/2022]
Abstract
Introduction Allergen‐specific immunotherapy (AIT) represents a curative approach for treating allergies. In the tropical and subtropical regions of the world, Blomia tropicalis (Blo t 5 and Blo t 21) is the likely dominant source of indoor allergens. Aim To generate a hypoallergenic Blo t 5/Blo t 21 hybrid molecule that can treat allergies caused by B tropicalis. Methods Using in silico design of B tropicalis hybrid proteins, we chose two hybrid proteins for heterologous expression. Wild‐type Blo t 5/Blo t 21 hybrid molecule and a hypoallergenic version, termed BTH1 and BTH2, respectively, were purified by ion exchange and size exclusion chromatography and characterized by physicochemical, as well as in vitro and in vivo immunological, experiments. Results BTH1, BTH2 and the parental allergens were purified to homogeneity and characterized in detail. BTH2 displayed the lowest IgE reactivity that induced basophil degranulation using sera from allergic rhinitis and asthmatic patients. BTH2 essentially presented the same endolysosomal degradation pattern as the shortened rBlo t 5 and showed a higher resistance towards degradation than the full‐length Blo t 5. In vivo immunization of mice with BTH2 led to the production of IgG antibodies that competed with human IgE for allergen binding. Stimulation of splenocytes from BTH2‐immunized mice produced higher levels of IL‐10 and decreased secretion of IL‐4 and IL‐5. In addition, BTH2 stimulated T‐cell proliferation in PBMCs isolated from allergic patients, with secretion of higher levels of IL‐10 and lower levels of IL‐5 and IL‐13, when compared to parental allergens. Conclusions and Clinical Relevance BTH2 is a promising hybrid vaccine candidate for immunotherapy of Blomia allergy. However, further pre‐clinical studies addressing its efficacy and safety are needed.
Collapse
Affiliation(s)
- Eduardo Santos da Silva
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil.,Programa de Pós-Graduação em Biotecnologia da Rede Nordeste de Biotecnologia (RENORBIO), Natal, Brazil.,Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Lorenz Aglas
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Carina Silva Pinheiro
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Emília M M de Andrade Belitardo
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil.,Programa de Pós-Graduação em Imunologia da Universidade Federal da Bahia, Salvador, Brazil
| | - Elisânia Fontes Silveira
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Sara Huber
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Rogério Tanan Torres
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Michael Wallner
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Peter Briza
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Peter Lackner
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Josef Laimer
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Luis Gustavo C Pacheco
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Álvaro A Cruz
- Núcleo de Excelência de Asma da, Universidade Federal da Bahia, Salvador, Brazil
| | - Neuza Maria Alcântara-Neves
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil.,Programa de Pós-Graduação em Biotecnologia da Rede Nordeste de Biotecnologia (RENORBIO), Natal, Brazil.,Programa de Pós-Graduação em Imunologia da Universidade Federal da Bahia, Salvador, Brazil
| | - Fatima Ferreira
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| |
Collapse
|
11
|
Gao X, Conard A, Yang C, Zhan Y, Zeng F, Shi J, Li W, Dimitrov DS, Gong R. Optimization of the C-Terminus of an Autonomous Human IgG1 CH2 Domain for Stability and Aggregation Resistance. Mol Pharm 2019; 16:3647-3656. [DOI: 10.1021/acs.molpharmaceut.9b00544] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Xinyu Gao
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Alex Conard
- Center for Antibody Therapeutics, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania 15261, United States
| | - Chunpeng Yang
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yancheng Zhan
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fang Zeng
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jian Shi
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
| | - Wei Li
- Center for Antibody Therapeutics, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania 15261, United States
| | - Dimiter S. Dimitrov
- Center for Antibody Therapeutics, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania 15261, United States
| | - Rui Gong
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
| |
Collapse
|
12
|
Zeng F, Yang C, Gao X, Li X, Zhang Z, Gong R. Comprehensive elucidation of the structural and functional roles of engineered disulfide bonds in antibody Fc fragment. J Biol Chem 2018; 293:19127-19135. [PMID: 30327432 DOI: 10.1074/jbc.ra118.005367] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/28/2018] [Indexed: 11/06/2022] Open
Abstract
Therapeutic monoclonal antibodies and Fc-fusion proteins containing antibody Fc fragment may tend to destabilize (e.g. unfold and aggregate), which leads to loss of functions and increase of adverse risks. Although engineering of an additional disulfide bond has been performed in Fc or Fc domains for optimization, the relationships between introduced disulfide bond and alteration of the stability, aggregation propensity and function were still unclear and should be addressed for achievement of better therapeutic outcome. Here, we constructed three human IgG1 Fc mutants including FcCH2-s-s- (one engineered disulfide bond in CH2 domain), FcCH3-s-s- (one engineered disulfide bond in CH3 domain), and FcCH3-s-s- CH2-s-s- (two engineered disulfide bonds in CH2 and CH3 domains, respectively) for evaluation. As expected, each mutated domain shows obviously increased stability during thermo-induced unfolding, and FcCH3-s-s- CH2-s-s- is most thermo-stable among wildtype Fc (wtFc) and three mutants. The order of overall stability against denaturant is FcCH3-s-s- CH2-s-s- > FcCH2-s-s- > FcCH3-s-s- > wtFc. Then the aggregation propensity was compared among these four proteins. Under conditions of incubation at 60 °C, their aggregation resistance is in the order of FcCH3-s-s- CH2-s-s- > FcCH2-s-s- > FcCH3-s-s- ≈ wtFc. In contrast, the order is FcCH3-s-s- CH2-s-s- > FcCH3-s-s- > FcCH2-s-s- ≈ wtFc under acidic conditions. In addition, the Fc-mediated functions are not obviously affected by engineered disulfide bond. Our results give a comprehensive elucidation of structural and functional effects caused by additional disulfide bonds in the Fc fragment, which is important for Fc engineering toward the desired clinical performance.
Collapse
Affiliation(s)
- Fang Zeng
- From the CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China and.,the University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunpeng Yang
- From the CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China and.,the University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinyu Gao
- From the CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China and.,the University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuan Li
- From the CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China and
| | - Zhe Zhang
- From the CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China and
| | - Rui Gong
- From the CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China and
| |
Collapse
|
13
|
Weber B, Brandl MJ, Pulido Cendales MD, Berner C, Pradhan T, Feind GM, Zacharias M, Reif B, Buchner J. A single residue switch reveals principles of antibody domain integrity. J Biol Chem 2018; 293:17107-17118. [PMID: 30228183 DOI: 10.1074/jbc.ra118.005475] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 09/14/2018] [Indexed: 11/06/2022] Open
Abstract
Despite their importance for antibody architecture and design, the principles governing antibody domain stability are still not understood in sufficient detail. Here, to address this question, we chose a domain from the invariant part of IgG, the CH2 domain. We found that compared with other Ig domains, the isolated CH2 domain is a surprisingly unstable monomer, exhibiting a melting temperature of ∼44 °C. We further show that the presence of an additional C-terminal lysine in a CH2 variant substantially increases the melting temperature by ∼14 °C relative to CH2 WT. To explore the molecular mechanism of this effect, we employed biophysical approaches to probe structural features of CH2. The results revealed that Lys101 is key for the formation of three secondary structure elements: the very C-terminal β-strand and two adjacent α-helices. We also noted that a dipole interaction between Lys101 and the nearby α-helix, is important for stabilizing the CH2 architecture by protecting the hydrophobic core. Interestingly, this interaction between the α-helix and C-terminal charged residues is highly conserved in antibody domains, suggesting that it represents a general mechanism for maintaining their integrity. We conclude that the observed interactions involving terminal residues have practical applications for defining domain boundaries in the development of antibody therapeutics and diagnostics.
Collapse
Affiliation(s)
- Benedikt Weber
- From the Center for Integrated Protein Science Munich, Departments of Chemie and
| | - Matthias J Brandl
- From the Center for Integrated Protein Science Munich, Departments of Chemie and
| | | | - Carolin Berner
- From the Center for Integrated Protein Science Munich, Departments of Chemie and
| | - Tejaswini Pradhan
- From the Center for Integrated Protein Science Munich, Departments of Chemie and
| | - Gina Maria Feind
- From the Center for Integrated Protein Science Munich, Departments of Chemie and
| | | | - Bernd Reif
- From the Center for Integrated Protein Science Munich, Departments of Chemie and
| | - Johannes Buchner
- From the Center for Integrated Protein Science Munich, Departments of Chemie and
| |
Collapse
|
14
|
Yang C, Gao X, Gong R. Engineering of Fc Fragments with Optimized Physicochemical Properties Implying Improvement of Clinical Potentials for Fc-Based Therapeutics. Front Immunol 2018; 8:1860. [PMID: 29375551 PMCID: PMC5766897 DOI: 10.3389/fimmu.2017.01860] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 12/07/2017] [Indexed: 01/09/2023] Open
Abstract
Therapeutic monoclonal antibodies and Fc-fusion proteins are successfully used in treatment of various diseases mainly including cancer, immune disease, and viral infection, which belong to the Fc-based therapeutics. In recent years, engineered Fc-derived antibody domains have also shown potential for Fc-based therapeutics. To increase the druggability of Fc-based therapeutic candidates, many efforts have been made in optimizing physicochemical properties and functions mediated by Fc fragment. The desired result is that we can simultaneously obtain Fc variants with increased physicochemical properties in vitro and capacity of mediating appropriate functions in vivo. However, changes of physicochemical properties of Fc may result in alternation of Fc-mediated functions and vice versa, which leads to undesired outcomes for further development of Fc-based therapeutics. Therefore, whether modified Fc fragments are suitable for achievement of expected clinical results or not needs to be seriously considered. Now, this question comes to be noticed and should be figured out to make better translation from the results of laboratory into clinical applications. In this review, we summarize different strategies on engineering physicochemical properties of Fc, and preliminarily elucidate the relationships between modified Fc in vitro and the subsequent therapeutic influence in vivo.
Collapse
Affiliation(s)
- Chunpeng Yang
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xinyu Gao
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Rui Gong
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
15
|
Li D, Gong R, Zheng J, Chen X, Dimitrov DS, Zhao Q. Engineered antibody CH2 domains binding to nucleolin: Isolation, characterization and improvement of aggregation. Biochem Biophys Res Commun 2017; 485:446-453. [PMID: 28202413 PMCID: PMC6957259 DOI: 10.1016/j.bbrc.2017.02.058] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 02/09/2017] [Indexed: 12/18/2022]
Abstract
Smaller recombinant antibody fragments are now emerging as alternatives of conventional antibodies. Especially, immunoglobulin (Ig) constant CH2 domain and engineered CH2 with improved stability are promising as scaffolds for selection of specific binders to various antigens. We constructed a yeast display library based on an engineered human IgG1 CH2 scaffold with diversified loop regions. A group of CH2 binders were isolated from this yeast display library by panning against nucleolin, which is a tumor-associated antigen involved in cell proliferation, tumor cell growth and angiogenesis. Out of 20 mutants, we selected 3 clones exhibiting relatively high affinities to nucleolin on yeasts. However, recombinant CH2 mutants aggregated when they were expressed. To find the mechanism of the aggregation, we employed computational prediction approaches through structural homology models of CH2 binders. The analysis of potential aggregation prone regions (APRs) and solvent accessible surface areas (ASAs) indicated two hydrophobic residues, Val264 and Leu309, in the β-sheet, in which replacement of both charged residues led to significant decrease of the protein aggregation. The newly identified CH2 binders could be improved to use as candidate therapeutics or research reagents in the future.
Collapse
Affiliation(s)
- Dezhi Li
- College of Life Science, Xiamen University, Xiamen, Fujian, China
| | - Rui Gong
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Jun Zheng
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Xihai Chen
- Department of General Surgery, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China.
| | - Dimiter S Dimitrov
- Protein Interaction Section, Cancer Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Qi Zhao
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| |
Collapse
|
16
|
Abstract
The crystallizable fragment (Fc) of the immunoglobulin class G (IgG) is a very attractive scaffold for the design of novel therapeutics due to its quality of uniting all essential antibody functions. This article reviews the functionalization of this homodimeric glycoprotein by diversification of structural loops of CH3 domains for the design of Fcabs, i.e. antigen-binding Fc proteins. It reports the design of libraries for the selection of nanomolar binders with wildtype-like in vivo half-life and correlation of Fc receptor binding and ADCC. The in vitro and preclinical biological activity of selected Fcabs is compared with that of clinically approved antibodies. Recently, the great potential of the scaffold for the development of therapeutics for clinical use has been shown when the HER2-binding Fcab FS102 entered clinical phase I. Furthermore, methods for the engineering of biophysical properties of Fcabs applicable to proteins in general are presented as well as the different approaches in the design of heterodimeric Fc-based scaffolds used in the generation of bispecific monoclonal antibodies. Finally, this work critically analyzes and compares the various efforts in the design of highly diverse and functional libraries that have been made in the engineering of IgG1-Fc and structurally similar scaffolds.
Collapse
Affiliation(s)
- Elisabeth Lobner
- Christian Doppler Laboratory for Antibody Engineering, Department of Chemistry, Vienna Institute of BioTechnology, BOKU - University of Natural Resources and Life Sciences, Vienna, Austria
| | - Michael W Traxlmayr
- Christian Doppler Laboratory for Antibody Engineering, Department of Chemistry, Vienna Institute of BioTechnology, BOKU - University of Natural Resources and Life Sciences, Vienna, Austria
| | - Christian Obinger
- Christian Doppler Laboratory for Antibody Engineering, Department of Chemistry, Vienna Institute of BioTechnology, BOKU - University of Natural Resources and Life Sciences, Vienna, Austria
| | - Christoph Hasenhindl
- Christian Doppler Laboratory for Antibody Engineering, Department of Chemistry, Vienna Institute of BioTechnology, BOKU - University of Natural Resources and Life Sciences, Vienna, Austria
| |
Collapse
|
17
|
Physicochemical characterization of a navy bean (Phaseolus vulgaris) protein fraction produced using a solvent-free method. Food Chem 2016; 208:35-41. [DOI: 10.1016/j.foodchem.2016.03.102] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 03/23/2016] [Accepted: 03/28/2016] [Indexed: 11/23/2022]
|
18
|
Dashivets T, Stracke J, Dengl S, Knaupp A, Pollmann J, Buchner J, Schlothauer T. Oxidation in the complementarity-determining regions differentially influences the properties of therapeutic antibodies. MAbs 2016; 8:1525-1535. [PMID: 27612038 PMCID: PMC5098445 DOI: 10.1080/19420862.2016.1231277] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Therapeutic antibodies can undergo a variety of chemical modification reactions in vitro. Depending on the site of modification, either antigen binding or Fc-mediated functions can be affected. Oxidation of tryptophan residues is one of the post-translational modifications leading to altered antibody functionality. In this study, we examined the structural and functional properties of a therapeutic antibody construct and 2 affinity matured variants thereof. Two of the 3 antibodies carry an oxidation-prone tryptophan residue in the complementarity-determining region of the VL domain. We demonstrate the differences in the stability and bioactivity of the 3 antibodies, and reveal differential degradation pathways for the antibodies susceptible to oxidation.
Collapse
Affiliation(s)
- Tetyana Dashivets
- a Roche Pharma Research and Early Development (pRED), Large Molecule Research, Roche Innovation Center Munich , Germany.,b Center for Integrated Protein Science Munich , Department Chemie, Technische Universität München , Garching , Germany
| | - Jan Stracke
- c Early-Stage Pharmaceutical Development & GLP Supplies, F. Hoffmann-La Roche Ltd Pharmaceutical Development & Supplies PTD Biologics Europe , Basel , Switzerland
| | - Stefan Dengl
- a Roche Pharma Research and Early Development (pRED), Large Molecule Research, Roche Innovation Center Munich , Germany
| | - Alexander Knaupp
- a Roche Pharma Research and Early Development (pRED), Large Molecule Research, Roche Innovation Center Munich , Germany
| | | | - Johannes Buchner
- b Center for Integrated Protein Science Munich , Department Chemie, Technische Universität München , Garching , Germany
| | - Tilman Schlothauer
- a Roche Pharma Research and Early Development (pRED), Large Molecule Research, Roche Innovation Center Munich , Germany
| |
Collapse
|
19
|
Zhou H, Yang W, Tian Y, Peng H, Wu Y. N-terminal truncation contributed to increasing thermal stability of mannanase Man1312 without activity loss. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2016; 96:1390-1395. [PMID: 25930671 DOI: 10.1002/jsfa.7240] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 04/20/2015] [Accepted: 04/25/2015] [Indexed: 06/04/2023]
Abstract
BACKGROUND The disordered residues on distal loops affect the molecular structural stability and on some occasions have regulatory roles in catalytic reaction. To increase understanding of the influence of distal residue mutation, this study explored the thermostability and enzymatic activity of mannanase Man1312 deletion mutants. The focus was on residues located on the N-terminal region because they are more disordered and changeable. The effects of N-terminal truncation on enzymatic activity and thermal dynamics were investigated by spectrophotometry, circular dichroism and differential scanning calorimetry assays. RESULTS The deletion mutants on V3, N7 and Q11 showed a marked increase in stability, while the enzymatic activity was significantly improved when triplet deletion was carried out. Triplet deletion MandVNQ showed around double the stability of its corresponding single-site and double-site deletion mutants. The Tm value of MandVNP was about 8 °C higher than that of Man1312. MandVNP had improved characteristics of Topt by 10 °C, t1/2 by 10 min and catalytic activity by 11% in comparison with Man1312. Analysis of spectra and modeling showed that MandVNQ had increased helix and strand contents. CONCLUSION N-terminal truncation had positive effects on the thermostability and activity of mannanase.
Collapse
Affiliation(s)
- Haiyan Zhou
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410128, China
| | - Wenjiao Yang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410128, China
| | - Yun Tian
- Key Lab of Agricultural Biochemistry and Biotransformation, Hunan Agricultural University, Changsha, 410128, China
| | - Hanhui Peng
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410128, China
| | - Yongyao Wu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410128, China
| |
Collapse
|
20
|
Fluorescence dye-based detection of mAb aggregates in CHO culture supernatants. Anal Bioanal Chem 2015; 407:4849-56. [DOI: 10.1007/s00216-015-8672-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 03/30/2015] [Accepted: 03/31/2015] [Indexed: 01/16/2023]
|
21
|
Brader ML, Estey T, Bai S, Alston RW, Lucas KK, Lantz S, Landsman P, Maloney KM. Examination of Thermal Unfolding and Aggregation Profiles of a Series of Developable Therapeutic Monoclonal Antibodies. Mol Pharm 2015; 12:1005-17. [DOI: 10.1021/mp400666b] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Mark L. Brader
- Protein Pharmaceutical Development, Biogen Idec, 14 Cambridge Center, Cambridge, Massachusetts 02142, United States
| | - Tia Estey
- Protein Pharmaceutical Development, Biogen Idec, 14 Cambridge Center, Cambridge, Massachusetts 02142, United States
| | - Shujun Bai
- Protein Pharmaceutical Development, Biogen Idec, 14 Cambridge Center, Cambridge, Massachusetts 02142, United States
| | - Roy W. Alston
- Protein Pharmaceutical Development, Biogen Idec, 14 Cambridge Center, Cambridge, Massachusetts 02142, United States
| | - Karin K. Lucas
- Protein Pharmaceutical Development, Biogen Idec, 14 Cambridge Center, Cambridge, Massachusetts 02142, United States
| | - Steven Lantz
- Protein Pharmaceutical Development, Biogen Idec, 14 Cambridge Center, Cambridge, Massachusetts 02142, United States
| | - Pavel Landsman
- Protein Pharmaceutical Development, Biogen Idec, 14 Cambridge Center, Cambridge, Massachusetts 02142, United States
| | - Kevin M. Maloney
- Protein Pharmaceutical Development, Biogen Idec, 14 Cambridge Center, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
22
|
Ying T, Gong R, Ju TW, Prabakaran P, Dimitrov DS. Engineered Fc based antibody domains and fragments as novel scaffolds. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1844:1977-1982. [PMID: 24792384 PMCID: PMC4185235 DOI: 10.1016/j.bbapap.2014.04.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 04/17/2014] [Accepted: 04/21/2014] [Indexed: 11/23/2022]
Abstract
Therapeutic monoclonal antibodies (mAbs) have been successful for the therapy of a number of diseases mostly cancer and immune disorders. However, the vast majority of mAbs approved for clinical use are full size, typically in IgG1 format. These mAbs may exhibit relatively poor tissue penetration and restricted epitope access due to their large size. A promising solution to this fundamental limitation is the engineering of smaller scaffolds based on the IgG1 Fc region. These scaffolds can be used for the generation of libraries of mutants from which high-affinity binders can be selected. Comprised of the CH2 and CH3 domains, the Fc region is important not only for the antibody effector function but also for its long half-life. This review focuses on engineered Fc based antibody fragments and domains including native (dimeric) Fc and monomeric Fc as well as CH2 and monomeric CH3, and their use as novel scaffolds and binders. The Fc based binders are promising candidate therapeutics with optimized half-life, enhanced tissue penetration and access to sterically restricted binding sites resulting in an increased therapeutic efficacy. This article is part of a Special Issue entitled: Recent advances in molecular engineering of antibody.
Collapse
Affiliation(s)
- Tianlei Ying
- Protein Interactions Group, Laboratory of Experimental Immunology, Cancer and Inflammation Program, National Cancer Institute, National Institutes of Health (NIH), Frederick, MD 21702, USA.
| | - Rui Gong
- Antibody Engineering Group, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
| | - Tina W Ju
- Protein Interactions Group, Laboratory of Experimental Immunology, Cancer and Inflammation Program, National Cancer Institute, National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Ponraj Prabakaran
- Protein Interactions Group, Laboratory of Experimental Immunology, Cancer and Inflammation Program, National Cancer Institute, National Institutes of Health (NIH), Frederick, MD 21702, USA; Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Dimiter S Dimitrov
- Protein Interactions Group, Laboratory of Experimental Immunology, Cancer and Inflammation Program, National Cancer Institute, National Institutes of Health (NIH), Frederick, MD 21702, USA
| |
Collapse
|
23
|
Abstract
Monoclonal antibodies have been successfully used for the therapy of various diseases. However, because of their large size (∼150 kD), many limitations have also been found during their development and manufacture. The use of antibody fragments with smaller sizes is one of the strategies to overcome these limitations. Antibody heavy chain variable domains (12∼15 kD) have already been widely used for the development of variable domain-based engineered antibody domains (termed V-based eAds) targeting different antigens. Recently, antibody second heavy chain constant domains (∼12 kD) were proposed as novel scaffolds for library construction and selection of specific binders termed constant domain-based eAds (C-based eAds) as novel candidate therapeutics, which might also confer additional crystallizable fragment functions. Both V- and C-based eAds are promising therapeutic candidates. This review summarizes progress in the development of eAds, and discusses the related patents and their potential applications.
Collapse
|