1
|
Ballesteros ME, Tolbert T, Schöneich C. Near UV and visible light-induced site-specific fragmentation of IgG1-based modalities mediated by histidine and Fe(III): a role for intra-domain interactions? J Pharm Sci 2025:103810. [PMID: 40324686 DOI: 10.1016/j.xphs.2025.103810] [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: 01/24/2025] [Revised: 04/28/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
Photo-stability represents a critical quality attribute for the development of therapeutic proteins where the exposure to near UV or visible light can lead to protein fragmentation. Here, we compare the photo-stability of three IgG1 based modalities, formulated in histidine (His) buffer containing various levels of Fe(III). We report a significant difference in the extent of photo-degradation between a high mannose Fc fragment (HM-Fc), NISTmAb and a fusion protein, Flt-3L-Ig. Our results indicate that despite preserving the Fc domain sequence, the NISTmAb and Flt-3L-Ig are more susceptible to site specific Thr259 photo-fragmentation in the CH2 domain compared to the HM-Fc (amino acid numbering based on the NISTmAb sequence). Enzymatic deglycoslyation enhanced the susceptibility of both NISTmAb and HM-Fc to photo-fragmentation, while enzymatic cleavage of the Fab domain from NISTmAb decreased the extent of photo-fragmentation. Our findings suggest that differences in photo-stability may be attributed to differences in domain-domain interactions, glycan structure, and the thermal stability of these modalities. Therefore, careful consideration should be given to photostability studies during the development of such proteins into therapeutic drug products.
Collapse
Affiliation(s)
| | - Thomas Tolbert
- Department of Pharmaceutical Chemistry, University of Kansas, 2093 Constant Avenue, Lawrence, KS 66047, USA
| | - Christian Schöneich
- Department of Pharmaceutical Chemistry, University of Kansas, 2093 Constant Avenue, Lawrence, KS 66047, USA.
| |
Collapse
|
2
|
Chen C, Zhang H, Lin Y, Lu M, Liao Q, Zhang S, Chen W, Zheng X, Li Y, Ding R, Wan Z. Identification of potential therapeutic drugs targeting core genes for systemic lupus erythematosus (SLE) and coexisting COVID-19: Insights from bioinformatic analyses. Immun Inflamm Dis 2023; 11:e1087. [PMID: 38018597 PMCID: PMC10659756 DOI: 10.1002/iid3.1087] [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: 06/23/2023] [Revised: 10/24/2023] [Accepted: 11/02/2023] [Indexed: 11/30/2023] Open
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) patients are at risk during the COVID-19 pandemic, yet the underlying molecular mechanisms remain incompletely understood. This study sought to analyze the potential molecular connections between COVID-19 and SLE, employing a bioinformatics approach to identify effective drugs for both conditions. METHODS The data sets GSE100163 and GSE183071 were utilized to determine share differentially expressed genes (DEGs). These DEGs were later analyzed by various bioinformatic methods, including functional enrichment, protein-protein interaction (PPI) network analysis, regulatory network construction, and gene-drug interaction construction. RESULTS A total of 50 common DEGs were found between COVID-19 and SLE. Gene ontology (GO) functional annotation revealed that "immune response," "innate immune response," "plasma membrane," and "protein binding" were most enriched in. Additionally, the pathways that were enriched include "Th1 and Th2 cell differentiation." The study identified 48 genes/nodes enriched with 292 edges in the PPI network, of which the top 10 hub genes were CD4, IL7R, CD3E, CD5, CD247, KLRB1, CD40LG, CD7, CR2, and GZMK. Furthermore, the study found 48 transcription factors and 8 microRNAs regulating these hub genes. Finally, four drugs namely ibalizumab (targeted to CD4), blinatumomab (targeted to CD3E), muromonab-CD3 (targeted to CD3E), and catumaxomab (targeted to CD3E) were found in gene-drug interaction. CONCLUSION Four possible drugs that targeted two specific genes, which may be beneficial for COVID-19 patients with SLE.
Collapse
Affiliation(s)
- Chao Chen
- School of Medicine, Institute of GenomicsHuaqiao UniversityXiamenChina
| | - Hongjian Zhang
- Department of Oncology and Vascular Interventional RadiologyZhongshan Hospital Xiamen University, School of Medicine, Xiamen UniversityXiamenFujianChina
| | - Yanbin Lin
- Department of Oncology and Vascular Interventional RadiologyZhongshan Hospital Xiamen University, School of Medicine, Xiamen UniversityXiamenFujianChina
| | - Meiqi Lu
- Department of Oncology and Vascular Interventional RadiologyZhongshan Hospital Xiamen University, School of Medicine, Xiamen UniversityXiamenFujianChina
| | - Quan Liao
- Department of Oncology and Vascular Interventional RadiologyZhongshan Hospital Xiamen University, School of Medicine, Xiamen UniversityXiamenFujianChina
| | - Shichao Zhang
- Department of Oncology and Vascular Interventional RadiologyZhongshan Hospital Xiamen University, School of Medicine, Xiamen UniversityXiamenFujianChina
| | - Weibin Chen
- Department of Oncology and Vascular Interventional RadiologyZhongshan Hospital Xiamen University, School of Medicine, Xiamen UniversityXiamenFujianChina
| | - Xiongwei Zheng
- Department of Oncology and Vascular Interventional RadiologyZhongshan Hospital Xiamen University, School of Medicine, Xiamen UniversityXiamenFujianChina
| | - Yunpeng Li
- Department of Oncology and Vascular Interventional RadiologyZhongshan Hospital Xiamen University, School of Medicine, Xiamen UniversityXiamenFujianChina
| | - Rui Ding
- Department of Oncology and Vascular Interventional RadiologyZhongshan Hospital Xiamen University, School of Medicine, Xiamen UniversityXiamenFujianChina
| | - Zheng Wan
- Department of Oncology and Vascular Interventional RadiologyZhongshan Hospital Xiamen University, School of Medicine, Xiamen UniversityXiamenFujianChina
| |
Collapse
|
3
|
Yu K, Liu B, Yu H, Sun C, Wang X, Li G, Dong M, Wang Y, Zhang J, Xu N, Liu W. A neutralizing bispecific single-chain antibody against SARS-CoV-2 Omicron variant produced based on CR3022. Front Cell Infect Microbiol 2023; 13:1155293. [PMID: 37207187 PMCID: PMC10189128 DOI: 10.3389/fcimb.2023.1155293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/21/2023] [Indexed: 05/21/2023] Open
Abstract
Introduction The constantly mutating SARS-CoV-2 has been infected an increasing number of people, hence the safe and efficacious treatment are urgently needed to combat the COVID-19 pandemic. Currently, neutralizing antibodies (Nabs), targeting the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein are potentially effective therapeutics against COVID-19. As a new form of antibody, bispecific single chain antibodies (BscAbs) can be easily expressed in E. coli and exhibits broad-spectrum antiviral activity. Methods In this study, we constructed two BscAbs 16-29, 16-3022 and three single chain variable fragments (scFv) S1-16, S2-29 and S3022 as a comparison to explore their antiviral activity against SARS-CoV-2. The affinity of the five antibodies was characterized by ELISA and SPR and the neutralizing activity of them was analyzed using pseudovirus or authentic virus neutralization assay. Bioinformatics and competitive ELISA methods were used to identify different epitopes on RBD. Results Our results revealed the potent neutralizing activity of two BscAbs 16-29 and 16-3022 against SARS-CoV-2 original strain and Omicron variant infection. In addition, we also found that SARS-CoV RBD-targeted scFv S3022 could play a synergistic role with other SARS-CoV-2 RBD-targeted antibodies to enhance neutralizing activity in the form of a BscAb or in cocktail therapies. Discussion This innovative approach offers a promising avenue for the development of subsequent antibody therapies against SARSCoV-2. Combining the advantages of cocktails and single-molecule strategies, BscAb therapy has the potential to be developed as an effective immunotherapeutic for clinical use to mitigate the ongoing pandemic.
Collapse
Affiliation(s)
- Kaikai Yu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Science, Changchun, Jilin, China
| | - Bin Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Haotian Yu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Science, Changchun, Jilin, China
| | - Chengbiao Sun
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Science, Changchun, Jilin, China
| | - Xuefeng Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Science, Changchun, Jilin, China
| | - Guorui Li
- College of Life Sciences and Food Engineering, Inner Mongolia Minzu University, Tongliao, China
| | - Mingxin Dong
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Science, Changchun, Jilin, China
| | - Yan Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Science, Changchun, Jilin, China
| | - Jianxu Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Science, Changchun, Jilin, China
| | - Na Xu
- Academic Affairs Office, Jilin Medical University, Jilin, Jilin, China
| | - Wensen Liu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Science, Changchun, Jilin, China
| |
Collapse
|
4
|
Identification of potent human neutralizing antibodies against SARS-CoV-2 implications for development of therapeutics and prophylactics. Nat Commun 2021; 12:4887. [PMID: 34373446 PMCID: PMC8352940 DOI: 10.1038/s41467-021-25153-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 07/20/2021] [Indexed: 12/12/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a novel coronavirus that is spreading rapidly, which seriously impacts global public health and economy. Thus, developing effective drugs remains urgent. We identify two potent antibodies, nCoVmab1 and nCoVmab2, targeting the SARS-CoV-2 spike protein receptor-binding domain (RBD) with high affinities from a naïve human phage-displayed Fab library. nCoVmab1 and nCoVmab2 neutralize authentic SARS-CoV-2 with picomolar and nanomolar IC50 values, respectively. No detectable defects of nCoVmab1 and nCoVmab2 are found during the preliminary druggability evaluation. nCoVmab1 could reduce viral titer and lung injury when administered prophylactically and therapeutically in human angiotensin-converting enzyme II (hACE2)-transgenic mice. Therefore, phage display platform could be efficiently used for rapid development of neutralizing monoclonal antibodies (nmabs) with clinical potential against emerging infectious diseases. In addition, we determinate epitopes in RBD of these antibodies to elucidate the neutralizing mechanism. We also convert nCoVmab1 and nCoVmab2 to their germline formats for further analysis, which reveals the contribution of somatic hypermutation (SHM) during nCoVmab1 and nCoVmab2 maturation. Our findings not only provide two highly potent nmabs against SARS-CoV-2 as prophylactic and therapeutic candidates, but also give some clues for development of anti-SARS-CoV-2 agents (e.g., drugs and vaccines) targeting the RBD.
Collapse
|
5
|
Zhou G, Zhao Q. Perspectives on therapeutic neutralizing antibodies against the Novel Coronavirus SARS-CoV-2. Int J Biol Sci 2020; 16:1718-1723. [PMID: 32226289 PMCID: PMC7098029 DOI: 10.7150/ijbs.45123] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 03/01/2020] [Indexed: 12/28/2022] Open
Abstract
A newly identified novel coronavirus (SARS-CoV-2) is causing pneumonia-associated respiratory syndrome across the world. Epidemiology, genomics, and pathogenesis of the SARS-CoV-2 show high homology with that of SARS-CoV. Current efforts are focusing on development of specific antiviral drugs. Therapeutic neutralizing antibodies (NAbs) against SARS-CoV-2 will be greatly important therapeutic agents for the treatment of coronavirus disease 2019 (COVID-19). Herein, the host immune responses against SARS-CoV discussed in this review provide implications for developing NAbs and understanding clinical interventions against SARS-CoV-2. Further, we describe the benefits, challenges and considerations of NAbs against SARS-CoV-2. Although many challenges exist, NAbs still offer a therapeutic option to control the current pandemic and the possible re-emergence of the virus in the future, and their development therefore remains a high priority.
Collapse
Affiliation(s)
- Guangyu Zhou
- Faculty of Health Sciences, University of Macau, Taipa, Macau SPR, China
- Institute of Translational Medicine, University of Macau, Taipa, Macau SPR, China
| | - Qi Zhao
- Faculty of Health Sciences, University of Macau, Taipa, Macau SPR, China
- Institute of Translational Medicine, University of Macau, Taipa, Macau SPR, China
| |
Collapse
|
6
|
Li M, Chen L, Wang Q, Hao M, Zhang X, Liu L, Yu X, Yang C, Xu J, Chen J, Gong R. A cross-reactive human monoclonal antibody targets the conserved H7 antigenic site A from fifth wave H7N9-infected humans. Antiviral Res 2019; 170:104556. [PMID: 31299269 DOI: 10.1016/j.antiviral.2019.104556] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/06/2019] [Accepted: 07/08/2019] [Indexed: 10/26/2022]
Abstract
Subtype H7 avian influenza viruses have been found to be associated with human infection and represent a risk for global public health. In 2013, the emergence of H7N9 virus in human beings and persistent human infection in China raised the most serious pandemic threat. Here we identified a human monoclonal antibody, P52E03, targeting the hemagglutinin (HA) of subtype H7 influenza viruses (H7 antigen), from a convalescent patient infected with H7N9 in 2017. P52E03 showed in vitro hemagglutination inhibiting (HI) and neutralizing activity against subtype H7 viruses belonging to both North American and Eurasian lineages. Moreover, it could prophylactically protect mice against weight loss and death caused by challenge with lethal H7N9 viruses in vivo and, therefore, is a candidate for development of antiviral agent against H7N9 infection. By generating escape mutant variants, we found that a single G151E substitution in the viral H7 antigenic site A could abort the neutralizing activity. Computational structural prediction of the P52E03/H7 complex revealed that residues including G151 in and around the conserved antigenic site A region are important for antigen recognition by the H7 cross-reactive antibody. Finally, we found that the P52E03 germline precursor (gHgL) antibody recognizes HA with measurable affinity, suggesting that its epitope is vulnerable to the human immune system and might elicit neutralizing antibodies (nAbs) in vivo after vaccination.
Collapse
Affiliation(s)
- Mingxin Li
- 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
| | - Li Chen
- 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
| | - Qingguang Wang
- 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
| | - Mengchan Hao
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China; National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xiaoqing Zhang
- 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
| | - Linlin Liu
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, 430079, China
| | - Xiao Yu
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, 430079, China
| | - 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
| | - Junqiang Xu
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, 430079, China.
| | - Jianjun Chen
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China; National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Rui Gong
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China.
| |
Collapse
|
7
|
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
|
8
|
Tatsumi K, Sakashita G, Nariai Y, Okazaki K, Kato H, Obayashi E, Yoshida H, Sugiyama K, Park SY, Sekine J, Urano T. G196 epitope tag system: a novel monoclonal antibody, G196, recognizes the small, soluble peptide DLVPR with high affinity. Sci Rep 2017; 7:43480. [PMID: 28266535 PMCID: PMC5339894 DOI: 10.1038/srep43480] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/24/2017] [Indexed: 11/09/2022] Open
Abstract
The recognition specificity of monoclonal antibodies (mAbs) has made mAbs among the most frequently used tools in both basic science research and in clinical diagnosis and therapies. Precise determination of the epitope allows the development of epitope tag systems to be used with recombinant proteins for various purposes. Here we describe a new family of tag derived from the epitope recognized by a highly specific mAb G196. The minimal epitope was identified as the five amino acid sequence Asp-Leu-Val-Pro-Arg. Permutation analysis was used to characterize the binding requirements of mAb G196, and the variable regions of the mAb G196 were identified and structurally analyzed by X-ray crystallography. Isothermal titration calorimetry revealed the high affinity (Kd = 1.25 nM) of the mAb G196/G196-epitope peptide interaction, and G196-tag was used to detect several recombinant cytosolic and nuclear proteins in human and yeast cells. mAb G196 is valuable for developing a new peptide tagging system for cell biology and biochemistry research.
Collapse
Affiliation(s)
- Kasumi Tatsumi
- Department of Biochemistry, Shimane University School of Medicine, Izumo 693-8501, Japan.,Department of Oral and Maxillofacial Surgery Shimane University School of Medicine, Izumo 693-8501, Japan
| | - Gyosuke Sakashita
- Department of Biochemistry, Shimane University School of Medicine, Izumo 693-8501, Japan
| | - Yuko Nariai
- Department of Biochemistry, Shimane University School of Medicine, Izumo 693-8501, Japan
| | - Kosuke Okazaki
- Department of Biochemistry, Shimane University School of Medicine, Izumo 693-8501, Japan
| | - Hiroaki Kato
- Department of Biochemistry, Shimane University School of Medicine, Izumo 693-8501, Japan
| | - Eiji Obayashi
- Department of Biochemistry, Shimane University School of Medicine, Izumo 693-8501, Japan
| | - Hisashi Yoshida
- Drug Design Group, Kanagawa Academy of Science and Technology, Kawasaki, Kanagawa, 213-0012, Japan
| | - Kanako Sugiyama
- Drug Design Group, Kanagawa Academy of Science and Technology, Kawasaki, Kanagawa, 213-0012, Japan
| | - Sam-Yong Park
- Drug Design Group, Kanagawa Academy of Science and Technology, Kawasaki, Kanagawa, 213-0012, Japan.,Protein Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Tsurumi, Yokohama 230-0045, Japan
| | - Joji Sekine
- Department of Oral and Maxillofacial Surgery Shimane University School of Medicine, Izumo 693-8501, Japan
| | - Takeshi Urano
- Department of Biochemistry, Shimane University School of Medicine, Izumo 693-8501, Japan
| |
Collapse
|
9
|
Robinson JE, Hastie KM, Cross RW, Yenni RE, Elliott DH, Rouelle JA, Kannadka CB, Smira AA, Garry CE, Bradley BT, Yu H, Shaffer JG, Boisen ML, Hartnett JN, Zandonatti MA, Rowland MM, Heinrich ML, Martínez-Sobrido L, Cheng B, de la Torre JC, Andersen KG, Goba A, Momoh M, Fullah M, Gbakie M, Kanneh L, Koroma VJ, Fonnie R, Jalloh SC, Kargbo B, Vandi MA, Gbetuwa M, Ikponmwosa O, Asogun DA, Okokhere PO, Follarin OA, Schieffelin JS, Pitts KR, Geisbert JB, Kulakoski PC, Wilson RB, Happi CT, Sabeti PC, Gevao SM, Khan SH, Grant DS, Geisbert TW, Saphire EO, Branco LM, Garry RF. Most neutralizing human monoclonal antibodies target novel epitopes requiring both Lassa virus glycoprotein subunits. Nat Commun 2016; 7:11544. [PMID: 27161536 PMCID: PMC4866400 DOI: 10.1038/ncomms11544] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 04/07/2016] [Indexed: 01/19/2023] Open
Abstract
Lassa fever is a severe multisystem disease that often has haemorrhagic manifestations. The epitopes of the Lassa virus (LASV) surface glycoproteins recognized by naturally infected human hosts have not been identified or characterized. Here we have cloned 113 human monoclonal antibodies (mAbs) specific for LASV glycoproteins from memory B cells of Lassa fever survivors from West Africa. One-half bind the GP2 fusion subunit, one-fourth recognize the GP1 receptor-binding subunit and the remaining fourth are specific for the assembled glycoprotein complex, requiring both GP1 and GP2 subunits for recognition. Notably, of the 16 mAbs that neutralize LASV, 13 require the assembled glycoprotein complex for binding, while the remaining 3 require GP1 only. Compared with non-neutralizing mAbs, neutralizing mAbs have higher binding affinities and greater divergence from germline progenitors. Some mAbs potently neutralize all four LASV lineages. These insights from LASV human mAb characterization will guide strategies for immunotherapeutic development and vaccine design.
Collapse
Affiliation(s)
- James E Robinson
- Section of Infectious Disease, Department of Pediatrics, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, Louisiana 70112, USA
| | - Kathryn M Hastie
- Department of Immunology and Microbial Science, Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Robert W Cross
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, 301 University Boulevard, Galveston, Texas 77555, USA
| | - Rachael E Yenni
- Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, Louisiana 70112, USA
| | - Deborah H Elliott
- Section of Infectious Disease, Department of Pediatrics, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, Louisiana 70112, USA
| | - Julie A Rouelle
- Section of Infectious Disease, Department of Pediatrics, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, Louisiana 70112, USA
| | - Chandrika B Kannadka
- Section of Infectious Disease, Department of Pediatrics, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, Louisiana 70112, USA
| | - Ashley A Smira
- Section of Infectious Disease, Department of Pediatrics, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, Louisiana 70112, USA
| | - Courtney E Garry
- Section of Infectious Disease, Department of Pediatrics, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, Louisiana 70112, USA.,Autoimmune Technologies, LLC, 1010 Common St #1705, New Orleans, Louisiana 70112, USA
| | - Benjamin T Bradley
- Section of Infectious Disease, Department of Pediatrics, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, Louisiana 70112, USA
| | - Haini Yu
- Section of Infectious Disease, Department of Pediatrics, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, Louisiana 70112, USA
| | - Jeffrey G Shaffer
- Department of Biostatistics and Bioinformatics, Tulane School of Public Health and Tropical Medicine, New Orleans, Louisiana 70112, USA
| | - Matt L Boisen
- Corgenix, Inc., 11575 Main Street #400, Broomfield, Colorado 80020, USA
| | - Jessica N Hartnett
- Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, Louisiana 70112, USA
| | - Michelle A Zandonatti
- Department of Immunology and Microbial Science, Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Megan M Rowland
- Zalgen Labs, LLC, 20271 Goldenrod Lane, Suite 2083, Germantown, Maryland 20876, USA
| | - Megan L Heinrich
- Zalgen Labs, LLC, 20271 Goldenrod Lane, Suite 2083, Germantown, Maryland 20876, USA
| | - Luis Martínez-Sobrido
- Department of Microbiology and Immunology, University of Rochester, 601 Elmwood Avenue, Rochester, New York 14642, USA
| | - Benson Cheng
- Department of Microbiology and Immunology, University of Rochester, 601 Elmwood Avenue, Rochester, New York 14642, USA
| | - Juan C de la Torre
- Department of Immunology and Microbial Science, Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Kristian G Andersen
- Department of Immunology and Microbial Science, Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Augustine Goba
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, 1 Combema Road, Kenema, Sierra Leone
| | - Mambu Momoh
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, 1 Combema Road, Kenema, Sierra Leone.,Department of Laboratory Sciences Polytechnic College, 2 Combema Road, Kenema, Sierra Leone
| | - Mohamed Fullah
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, 1 Combema Road, Kenema, Sierra Leone.,Department of Laboratory Sciences Polytechnic College, 2 Combema Road, Kenema, Sierra Leone
| | - Michael Gbakie
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, 1 Combema Road, Kenema, Sierra Leone
| | - Lansana Kanneh
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, 1 Combema Road, Kenema, Sierra Leone
| | - Veronica J Koroma
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, 1 Combema Road, Kenema, Sierra Leone
| | - Richard Fonnie
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, 1 Combema Road, Kenema, Sierra Leone
| | - Simbirie C Jalloh
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, 1 Combema Road, Kenema, Sierra Leone
| | - Brima Kargbo
- Ministry of Health and Sanitation, 4th Floor Youyi Building, Freetown, Sierra Leone
| | - Mohamed A Vandi
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, 1 Combema Road, Kenema, Sierra Leone.,Ministry of Health and Sanitation, 4th Floor Youyi Building, Freetown, Sierra Leone
| | - Momoh Gbetuwa
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, 1 Combema Road, Kenema, Sierra Leone.,Ministry of Health and Sanitation, 4th Floor Youyi Building, Freetown, Sierra Leone
| | - Odia Ikponmwosa
- Department of Medicine, Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Km. 87, Benin/Auchi Road, Irrua, Nigeria
| | - Danny A Asogun
- Department of Medicine, Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Km. 87, Benin/Auchi Road, Irrua, Nigeria
| | - Peter O Okokhere
- Department of Medicine, Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Km. 87, Benin/Auchi Road, Irrua, Nigeria
| | - Onikepe A Follarin
- Department of Medicine, Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Km. 87, Benin/Auchi Road, Irrua, Nigeria.,Department of Biological Sciences, College of Natural Sciences, Redeemer's University, Off Gbongan-Oshogbo Road, Ede, Nigeria.,African Center of Excellence for Genomics of Infectious Disease (ACEGID), Redeemer's University, Off Gbongan-Oshogbo Road, Ede, Nigeria
| | - John S Schieffelin
- Section of Infectious Disease, Department of Pediatrics, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, Louisiana 70112, USA.,Section of Infectious Disease, Department of Internal Medicine, Tulane University School of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, Louisiana 70112, USA
| | - Kelly R Pitts
- Corgenix, Inc., 11575 Main Street #400, Broomfield, Colorado 80020, USA
| | - Joan B Geisbert
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, 301 University Boulevard, Galveston, Texas 77555, USA
| | - Peter C Kulakoski
- Autoimmune Technologies, LLC, 1010 Common St #1705, New Orleans, Louisiana 70112, USA
| | - Russell B Wilson
- Autoimmune Technologies, LLC, 1010 Common St #1705, New Orleans, Louisiana 70112, USA
| | - Christian T Happi
- Department of Medicine, Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Km. 87, Benin/Auchi Road, Irrua, Nigeria.,Department of Biological Sciences, College of Natural Sciences, Redeemer's University, Off Gbongan-Oshogbo Road, Ede, Nigeria.,African Center of Excellence for Genomics of Infectious Disease (ACEGID), Redeemer's University, Off Gbongan-Oshogbo Road, Ede, Nigeria
| | - Pardis C Sabeti
- Department of Organismic and Evolutionary Biology, Center for Systems Biology, Harvard University, 1350 Massachusetts Avenue, Cambridge, Massachusetts 02138, USA.,Center for Systems Biology, Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, 415 Main Street, Cambridge, Massachusetts 02142, USA.,Department of Immunology and Infectious Disease, Harvard School of Public Health, 677 Huntington Avenue, Boston, Massachusetts 02115, USA
| | - Sahr M Gevao
- Department of Medicine, University of Sierra Leone, Freetown, Sierra Leone
| | - S Humarr Khan
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, 1 Combema Road, Kenema, Sierra Leone.,Ministry of Health and Sanitation, 4th Floor Youyi Building, Freetown, Sierra Leone
| | - Donald S Grant
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, 1 Combema Road, Kenema, Sierra Leone.,Ministry of Health and Sanitation, 4th Floor Youyi Building, Freetown, Sierra Leone
| | - Thomas W Geisbert
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, 301 University Boulevard, Galveston, Texas 77555, USA
| | - Erica Ollmann Saphire
- Department of Immunology and Microbial Science, Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA.,The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Luis M Branco
- Zalgen Labs, LLC, 20271 Goldenrod Lane, Suite 2083, Germantown, Maryland 20876, USA
| | - Robert F Garry
- Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, Louisiana 70112, USA.,Zalgen Labs, LLC, 20271 Goldenrod Lane, Suite 2083, Germantown, Maryland 20876, USA
| |
Collapse
|
10
|
Ying T, Du L, Ju TW, Prabakaran P, Lau CCY, Lu L, Liu Q, Wang L, Feng Y, Wang Y, Zheng BJ, Yuen KY, Jiang S, Dimitrov DS. Exceptionally potent neutralization of Middle East respiratory syndrome coronavirus by human monoclonal antibodies. J Virol 2014; 88:7796-805. [PMID: 24789777 PMCID: PMC4097770 DOI: 10.1128/jvi.00912-14] [Citation(s) in RCA: 197] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 04/21/2014] [Indexed: 11/20/2022] Open
Abstract
The recently discovered Middle East respiratory syndrome coronavirus (MERS-CoV) continues to infect humans, with high mortality. Specific, highly effective therapeutics and vaccines against the MERS-CoV are urgently needed to save human lives and address the pandemic concerns. We identified three human monoclonal antibodies (MAbs), m336, m337, and m338, targeting the receptor (CD26/DPP4) binding domain (RBD) of the MERS-CoV spike glycoprotein from a very large naïve-antibody library (containing ∼10(11) antibodies). They bound with high affinity: equilibrium dissociation constants for the three MAbs were equal to 4.2, 9.3, and 15 nM, respectively, as measured by Biacore for Fabs binding to RBD. The avidity for IgG1 m336, m337, and m338 was even higher: 99, 820, and 560 pM, respectively. The antibodies bound to overlapping epitopes that overlap the receptor binding site on the RBD as suggested by competition experiments and further supported by site-directed mutagenesis of the RBD and a docking model of the m336-RBD complex. The highest-affinity MAb, m336, neutralized both pseudotyped and live MERS-CoV with exceptional potency, 50% neutralization at 0.005 and 0.07 μg/ml, respectively, likely by competing with DPP4 for binding to the S glycoprotein. The exceptionally high neutralization activity of these antibodies and especially m336 suggests that they have great potential for prophylaxis and therapy of MERS-CoV infection in humans and as a tool for development of vaccine immunogens. The rapid identification (within several weeks) of potent MAbs suggests a possibility to use the new large antibody library and related methodology for a quick response to the public threat resulting from emerging coronaviruses. Importance: A novel human coronavirus, the Middle East respiratory syndrome coronavirus (MERS-CoV), was found to infect humans with a high mortality rate in 2012, just 1 decade after the appearance of the first highly pathogenic coronavirus, severe acute respiratory syndrome coronavirus (SARS-CoV). There are no effective therapeutics available. It is highly desirable to find an approach for rapidly developing potent therapeutics against MERS-CoV, which not only can be implemented for MERS treatment but also can help to develop a platform strategy to combat future emerging coronaviruses. We report here the identification of human monoclonal antibodies (MAbs) from a large nonimmune antibody library that target MERS-CoV. One of the antibodies, m336, neutralized the virus with exceptional potency. It therefore may have great potential as a candidate therapeutic and as a reagent to facilitate the development of vaccines against MERS-CoV.
Collapse
Affiliation(s)
- Tianlei Ying
- Protein Interactions Group, Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland, USA
| | - Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, USA
| | - Tina W Ju
- Protein Interactions Group, Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland, USA
| | - Ponraj Prabakaran
- Protein Interactions Group, Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland, USA Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland, USA
| | - Candy C Y Lau
- Department of Microbiology, University of Hong Kong, Pokfulam, Hong Kong
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University, Shanghai, China
| | - Qi Liu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University, Shanghai, China
| | - Lili Wang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, USA
| | - Yang Feng
- Protein Interactions Group, Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland, USA
| | - Yanping Wang
- Protein Interactions Group, Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland, USA
| | - Bo-Jian Zheng
- Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland, USA
| | - Kwok-Yung Yuen
- Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland, USA
| | - Shibo Jiang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, USA Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University, Shanghai, China
| | - Dimiter S Dimitrov
- Protein Interactions Group, Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland, USA
| |
Collapse
|
11
|
Broder CC, Xu K, Nikolov DB, Zhu Z, Dimitrov DS, Middleton D, Pallister J, Geisbert TW, Bossart KN, Wang LF. A treatment for and vaccine against the deadly Hendra and Nipah viruses. Antiviral Res 2013; 100:8-13. [PMID: 23838047 DOI: 10.1016/j.antiviral.2013.06.012] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 06/19/2013] [Accepted: 06/20/2013] [Indexed: 12/29/2022]
Abstract
Hendra virus and Nipah virus are bat-borne paramyxoviruses that are the prototypic members of the genus Henipavirus. The henipaviruses emerged in the 1990s, spilling over from their natural bat hosts and causing serious disease outbreaks in humans and livestock. Hendra virus emerged in Australia and since 1994 there have been 7 human infections with 4 case fatalities. Nipah virus first appeared in Malaysia and subsequent outbreaks have occurred in Bangladesh and India. In total, there have been an estimated 582 human cases of Nipah virus and of these, 54% were fatal. Their broad species tropism and ability to cause fatal respiratory and/or neurologic disease in humans and animals make them important transboundary biological threats. Recent experimental findings in animals have demonstrated that a human monoclonal antibody targeting the viral G glycoprotein is an effective post-exposure treatment against Hendra and Nipah virus infection. In addition, a subunit vaccine based on the G glycoprotein of Hendra virus affords protection against Hendra and Nipah virus challenge. The vaccine has been developed for use in horses in Australia and is the first vaccine against a Biosafety Level-4 (BSL-4) agent to be licensed and commercially deployed. Together, these advances offer viable approaches to address Hendra and Nipah virus infection of livestock and people.
Collapse
Affiliation(s)
- Christopher C Broder
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814, United States.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Gong R, Wang Y, Ying T, Feng Y, Streaker E, Prabakaran P, Dimitrov DS. N-terminal truncation of an isolated human IgG1 CH2 domain significantly increases its stability and aggregation resistance. Mol Pharm 2013; 10:2642-52. [PMID: 23641816 PMCID: PMC3795862 DOI: 10.1021/mp400075f] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Isolated human immunoglobulin G (IgG) CH2 domains are promising scaffolds for novel candidate therapeutics. Unlike other human IgG domains, CH2 is not involved in strong interchain interactions, and isolated CH2 is relatively stable. However, isolated single CH2 is prone to aggregation. In native IgG and Fc molecules, the N-terminal residues of CH2 from the two heavy chains interact with each other and form hinge regions. By contrast, the N-terminal residues are highly disordered in isolated CH2. We have hypothesized that the removal of the CH2 N-terminal residues may not only increase its stability but also its aggregation resistance. To test this hypothesis we constructed a shortened variant of IgG1 CH2 (CH2s) where the first seven residues of the N-terminus were deleted. We found that the thermal stability of CH2s was increased by 5 °C compared to CH2. Importantly, we demonstrated that CH2s is significantly less prone to aggregation than CH2 as measured by Thioflavin T (ThT) fluorescence, turbidity, and light scattering. We also found that the CH2s exhibited pH-dependent binding to a soluble single-chain human neonatal Fc receptor (shFcRn) which was significantly stronger than the very weak binding of CH2 to shFcRn as measured by flow cytometry. Computer modeling suggested a possible mode of CH2 aggregation involving its N-terminal residues. Therefore, deletion of the N-terminal residues could increase drugability of CH2-based therapeutic candidates. This strategy to increase stability and aggregation resistance could also be applicable to other Ig-related proteins.
Collapse
Affiliation(s)
- Rui Gong
- Antibody Engineering Group, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- Protein Interactions Group, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Yanping Wang
- Protein Interactions Group, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
- Basic Research Program, Science Applications International Corporation-Frederick, Inc., Frederick, MD 21702, USA
| | - Tianlei Ying
- Protein Interactions Group, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Yang Feng
- Protein Interactions Group, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Emily Streaker
- Protein Interactions Group, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
- Basic Research Program, Science Applications International Corporation-Frederick, Inc., Frederick, MD 21702, USA
| | - Ponraj Prabakaran
- Protein Interactions Group, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
- Basic Research Program, Science Applications International Corporation-Frederick, Inc., Frederick, MD 21702, USA
| | - Dimiter S. Dimitrov
- Protein Interactions Group, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| |
Collapse
|