1
|
Chai YY, Zhou L, Yang Y, Tan L, Ma K, Huang ZJ. Development and validation of an LC-MS/MS method for pharmacokinetic assessment of a bispecific antibody (YBSW015) targeting SARS-CoV-2. J Pharm Biomed Anal 2025; 260:116770. [PMID: 40054108 DOI: 10.1016/j.jpba.2025.116770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/20/2025] [Accepted: 02/23/2025] [Indexed: 04/06/2025]
Abstract
YBSW015 is a bispecific antibody that targets severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). A pair of high-affinity parental monoclonal antibodies (B38 and H4) were discovered from the sera of convalescent patients, and YBSW015 was developed based on these two parental antibodies. To support pharmacokinetic research, a simple, specific and reproducible liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was developed and validated for the quantifying YBSW015 in human serum. Two signature peptides (SPs) were selected as surrogate analytes for quantification and qualitative detection, respectively. Two stable isotope-labeled synthetic peptides with the same sequence of two SPs were set as internal standards (ISs). Total antibody was precipitated with 1.0 % (w/v) trichloroacetic acid (TCA) in isopropanol (IPA) following by trypsin enzymatic and further purified by solid phase extraction (SPE). The performance of this bioanalytical assay was well evaluated with respect to linearity, accuracy and precision, selectivity, specificity, matrix effect, recovery, and stability. YBSW015 concentrations in human serum were determined over the concentration range of 2.00-1000 μg/mL, and the quantification was not interfered by the matrix components and pre-existing antibodies. This precise and sensitive method was successfully employed in a phase I clinical study.
Collapse
Affiliation(s)
- Yuan-Yuan Chai
- Suzhou Yabao Pharmaceutical R&D Co., LTD., Building B7-801, 218 Xinghu Street, Suzhou, Jiangsu, China
| | - Ling Zhou
- United-Power Pharma Tech (Shanghai) Co., LTD., Building 30, Lane 908, Ziping Road, Pudong New Area, Shanghai, China
| | - Yan Yang
- United-Power Pharma Tech (Shanghai) Co., LTD., Building 30, Lane 908, Ziping Road, Pudong New Area, Shanghai, China
| | - Lin Tan
- Suzhou Yabao Pharmaceutical R&D Co., LTD., Building B7-801, 218 Xinghu Street, Suzhou, Jiangsu, China
| | - Ke Ma
- United-Power Pharma Tech (Shanghai) Co., LTD., Building 30, Lane 908, Ziping Road, Pudong New Area, Shanghai, China.
| | - Zhi-Jiang Huang
- Suzhou Yabao Pharmaceutical R&D Co., LTD., Building B7-801, 218 Xinghu Street, Suzhou, Jiangsu, China.
| |
Collapse
|
2
|
Qiao R, Liu Y, Mao Q, Li J, Lu Y, Shi J, Li C, Yu J, Gong J, Wang X, Shao Y, Sun L, Zhang W, Yu H, Chu H, Wang P, Zhao X. Novel Trispecific Neutralizing Antibodies With Enhanced Potency and Breadth Against Pan-Sarbecoviruses. MedComm (Beijing) 2025; 6:e70191. [PMID: 40260012 PMCID: PMC12010136 DOI: 10.1002/mco2.70191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 04/23/2025] Open
Abstract
The ongoing emergence of new variants of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) underscores the urgent need for developing antivirals targeting both SARS-CoV-2 variants and related sarbecoviruses. To this end, we designed novel trispecific antibodies, Tri-1 and Tri-2, engineered by fusing the single-chain variable fragments (scFvs) of a potent antibody (PW5-570) to the Fc region of "Knob-into-Hole" bispecific antibodies (bsAbs) composed of two distinct broad antibodies (PW5-5 and PW5-535). Compared with the parental antibodies, Tri-1 and Tri-2 displayed enhanced binding affinities to the receptor-binding domains of SARS-CoV, SARS-CoV-2 wild type, and Omicron XBB.1.16, with each arm contributed to the overall enhancement. Furthermore, pseudovirus neutralization assays revealed that Tri-1 and Tri-2 effectively neutralized all tested SARS-CoV, SARS-CoV-2 variants, and related sarbecoviruses (Pangolin-GD, RaTG13, WIV1, and SHC014), demonstrating potency and breadth superior to any single parental antibody. Consistently, Tri-1 and Tri-2 were found to effectively neutralize authentic SARS-CoV and SARS-CoV-2 variants with IC50 values comparable to or better than those of parental antibodies. Taken together, this study highlights the potential effectiveness of Tri-1 and Tri-2 as novel formats for harnessing cross-neutralizing antibodies in the development of multivalent agents to combat both current and future SARS-like coronaviruses.
Collapse
Affiliation(s)
- Rui Qiao
- Shanghai Sci‐Tech Inno Center for Infection & ImmunityNational Medical Center for Infectious DiseasesHuashan HospitalInstitute of Infection and HealthShanghai Key Laboratory of Oncology Target Discovery and Antibody Drug DevelopmentFudan UniversityShanghaiChina
- Shanghai Pudong HospitalState Key Laboratory of Genetic EngineeringMOE Engineering Research Center of Gene TechnologySchool of Life SciencesShanghai Institute of Infectious Disease and BiosecurityFudan University Pudong Medical CenterFudan UniversityShanghaiChina
| | - Yuanchen Liu
- Department of MicrobiologySchool of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongPokfulamHong Kong Special Administrative RegionHong KongChina
| | - Qiyu Mao
- Shanghai Fifth People's HospitalShanghai Institute of Infectious Disease and BiosecurityInstitutes of Biomedical SciencesFudan UniversityShanghaiChina
| | - Jiayan Li
- Shanghai Pudong HospitalState Key Laboratory of Genetic EngineeringMOE Engineering Research Center of Gene TechnologySchool of Life SciencesShanghai Institute of Infectious Disease and BiosecurityFudan University Pudong Medical CenterFudan UniversityShanghaiChina
| | - Yinying Lu
- Shanghai Sci‐Tech Inno Center for Infection & ImmunityNational Medical Center for Infectious DiseasesHuashan HospitalInstitute of Infection and HealthShanghai Key Laboratory of Oncology Target Discovery and Antibody Drug DevelopmentFudan UniversityShanghaiChina
| | - Jialu Shi
- Department of MicrobiologySchool of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongPokfulamHong Kong Special Administrative RegionHong KongChina
| | - Chen Li
- Shanghai Pudong HospitalState Key Laboratory of Genetic EngineeringMOE Engineering Research Center of Gene TechnologySchool of Life SciencesShanghai Institute of Infectious Disease and BiosecurityFudan University Pudong Medical CenterFudan UniversityShanghaiChina
| | - Jizhen Yu
- Shanghai Pudong HospitalState Key Laboratory of Genetic EngineeringMOE Engineering Research Center of Gene TechnologySchool of Life SciencesShanghai Institute of Infectious Disease and BiosecurityFudan University Pudong Medical CenterFudan UniversityShanghaiChina
| | - Jiami Gong
- Shanghai Pudong HospitalState Key Laboratory of Genetic EngineeringMOE Engineering Research Center of Gene TechnologySchool of Life SciencesShanghai Institute of Infectious Disease and BiosecurityFudan University Pudong Medical CenterFudan UniversityShanghaiChina
| | - Xun Wang
- Shanghai Pudong HospitalState Key Laboratory of Genetic EngineeringMOE Engineering Research Center of Gene TechnologySchool of Life SciencesShanghai Institute of Infectious Disease and BiosecurityFudan University Pudong Medical CenterFudan UniversityShanghaiChina
| | - Yuchen Shao
- Shanghai Pudong HospitalState Key Laboratory of Genetic EngineeringMOE Engineering Research Center of Gene TechnologySchool of Life SciencesShanghai Institute of Infectious Disease and BiosecurityFudan University Pudong Medical CenterFudan UniversityShanghaiChina
| | - Lei Sun
- Shanghai Fifth People's HospitalShanghai Institute of Infectious Disease and BiosecurityInstitutes of Biomedical SciencesFudan UniversityShanghaiChina
| | - Wenhong Zhang
- Department of Infectious DiseasesShanghai Key Laboratory of Infectious Diseases and Biosafety Emergency ResponseNational Medical Center for Infectious DiseasesHuashan HospitalFudan UniversityShanghaiChina
| | - Hongjie Yu
- School of Public HealthKey Laboratory of Public Health SafetyFudan UniversityMinistry of EducationShanghaiChina
| | - Hin Chu
- Department of MicrobiologySchool of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongPokfulamHong Kong Special Administrative RegionHong KongChina
| | - Pengfei Wang
- Shanghai Sci‐Tech Inno Center for Infection & ImmunityNational Medical Center for Infectious DiseasesHuashan HospitalInstitute of Infection and HealthShanghai Key Laboratory of Oncology Target Discovery and Antibody Drug DevelopmentFudan UniversityShanghaiChina
- Shanghai Pudong HospitalState Key Laboratory of Genetic EngineeringMOE Engineering Research Center of Gene TechnologySchool of Life SciencesShanghai Institute of Infectious Disease and BiosecurityFudan University Pudong Medical CenterFudan UniversityShanghaiChina
| | - Xiaoyu Zhao
- Shanghai Sci‐Tech Inno Center for Infection & ImmunityNational Medical Center for Infectious DiseasesHuashan HospitalInstitute of Infection and HealthShanghai Key Laboratory of Oncology Target Discovery and Antibody Drug DevelopmentFudan UniversityShanghaiChina
| |
Collapse
|
3
|
Zhao C, Hao M, Bian T, Zhao X, Chi X, Chen Z, Fu G, Zhu Z, Fang T, Yu C, Li J, Chen W. Screening of Neutralizing Antibodies Targeting Gc Protein of RVFV. Viruses 2025; 17:559. [PMID: 40285002 PMCID: PMC12031069 DOI: 10.3390/v17040559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
Rift Valley fever virus (RVFV) is a mosquito-transmitted bunyavirus that can cause substantial morbidity and mortality in livestock and humans, for which there are no currently available licensed human therapeutics or vaccines. Therefore, the development of safe and effective antivirals is both necessary and urgent. The Gc protein is the primary target of the neutralizing antibody response related to Rift Valley fever virus. Here, we report one Gc-specific neutralizing antibody (NA137) isolated from an alpaca and one bispecific antibody (E2-NA137), the protective efficacies of which we evaluated in A129 mice. In this prophylactic study, the survival rates of the NA137 and E2-NA137 groups were both 80%, and in the treatment study, the survival rates were 20% and 60%, respectively. Altogether, our results emphasize that NA137 and E2-NA137 provide a potential approach for treating RVFV either prophylactically or therapeutically.
Collapse
Affiliation(s)
- Chuanyi Zhao
- School of Medicine, Zhejiang University, Hangzhou 310058, China;
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (M.H.); (T.B.); (X.Z.); (X.C.); (Z.C.); (G.F.); (Z.Z.); (T.F.); (C.Y.)
| | - Meng Hao
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (M.H.); (T.B.); (X.Z.); (X.C.); (Z.C.); (G.F.); (Z.Z.); (T.F.); (C.Y.)
| | - Ting Bian
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (M.H.); (T.B.); (X.Z.); (X.C.); (Z.C.); (G.F.); (Z.Z.); (T.F.); (C.Y.)
| | - Xiaofan Zhao
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (M.H.); (T.B.); (X.Z.); (X.C.); (Z.C.); (G.F.); (Z.Z.); (T.F.); (C.Y.)
| | - Xiangyang Chi
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (M.H.); (T.B.); (X.Z.); (X.C.); (Z.C.); (G.F.); (Z.Z.); (T.F.); (C.Y.)
| | - Zhengshan Chen
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (M.H.); (T.B.); (X.Z.); (X.C.); (Z.C.); (G.F.); (Z.Z.); (T.F.); (C.Y.)
| | - Guangcheng Fu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (M.H.); (T.B.); (X.Z.); (X.C.); (Z.C.); (G.F.); (Z.Z.); (T.F.); (C.Y.)
| | - Zheng Zhu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (M.H.); (T.B.); (X.Z.); (X.C.); (Z.C.); (G.F.); (Z.Z.); (T.F.); (C.Y.)
| | - Ting Fang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (M.H.); (T.B.); (X.Z.); (X.C.); (Z.C.); (G.F.); (Z.Z.); (T.F.); (C.Y.)
| | - Changming Yu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (M.H.); (T.B.); (X.Z.); (X.C.); (Z.C.); (G.F.); (Z.Z.); (T.F.); (C.Y.)
| | - Jianmin Li
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (M.H.); (T.B.); (X.Z.); (X.C.); (Z.C.); (G.F.); (Z.Z.); (T.F.); (C.Y.)
| | - Wei Chen
- School of Medicine, Zhejiang University, Hangzhou 310058, China;
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (M.H.); (T.B.); (X.Z.); (X.C.); (Z.C.); (G.F.); (Z.Z.); (T.F.); (C.Y.)
| |
Collapse
|
4
|
Shcherbakov DN, Isaeva AA, Mustaev EA. Treatment of Ebola Virus Disease: From Serotherapy to the Use of Monoclonal Antibodies. Antibodies (Basel) 2025; 14:22. [PMID: 40136471 PMCID: PMC11939263 DOI: 10.3390/antib14010022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/27/2025] Open
Abstract
Ebola virus disease (EVD) is an acute illness with a high-case fatality rate (CFR) caused by an RNA virus belonging to the Filoviridae family. Over the past 50 years, regular EVD outbreaks have been reported. The West African EVD outbreak of 2013-2016 proved to be significantly more widespread and complex than previous ones, resulting in approximately 11,000 deaths. A coordinated international effort was required to bring the outbreak under control. One of the main challenges faced by clinicians and researchers combating EVD was the absence of vaccines and preventive treatments. Only recently have efforts led to the development of effective therapeutic options. Among these, monoclonal antibody-based drugs have emerged as the most promising agents for the urgent treatment of EVD. This article aims to review the key milestones in the development of antibody-based therapies for EVD, tracing the journey from the use of convalescent serum to the creation of effective monoclonal antibody-based drugs and their combinations.
Collapse
Affiliation(s)
- Dmitriy N. Shcherbakov
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia;
| | - Anastasiya A. Isaeva
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia;
| | - Egor A. Mustaev
- Department of Natural Sciences, Novosibirsk State University, Pirogova st., 2, Novosibirsk 630090, Russia;
- Synchrotron Radiation Facility—Siberian Circular Photon Source “SKlF” Boreskov Institute of Catalysis of Siberian Branch of the Russian Academy of Sciences, Nikolskiy pr-t, 1, Koltsovo 630559, Russia
| |
Collapse
|
5
|
Ren Z, Li M, Chen J, Gong X, Song S, Li D, Yang M, Yu J, Asghar S, Cui Y, Niu S, Liao Z, Jiang Y, Liu J, Li Y, Zhang B, Zhao W, Peng J, Yang Y, Shen C. Identification of mpox M1R and B6R monoclonal and bispecific antibodies that efficiently neutralize authentic mpox virus. Emerg Microbes Infect 2024; 13:2401931. [PMID: 39233480 DOI: 10.1080/22221751.2024.2401931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/29/2024] [Accepted: 09/04/2024] [Indexed: 09/06/2024]
Abstract
In 2022, the monkeypox virus (mpox virus, MPXV) exhibited global dissemination across six continents, representing a notable challenge owing to the scarcity of targeted antiviral interventions. Passive immunotherapy, such as the use of monoclonal antibodies (mAbs) and bispecific antibodies (bsAbs), has emerged as a promising option for antiviral regimens. Here, we generated several mAbs against M1R and B6R of MPXV, and subsequently characterized the antiviral activity of these antibodies both in vitro and in vivo. Two neutralizing mAbs, M1H11 and M3B2, targeting M1R, and one B6R-specific mAb, B7C9, were identified. They exhibited varying antiviral efficacy against vaccinia virus (VACV) in vitro and in vivo. A cocktail comprising M1H11 and M3B2 demonstrated a superior protective effect in vivo. A bsAb, Bis-M1M3, was engineered by conjugating the fragment crystallizable (Fc) region of the human-mouse chimeric engineered M1H11 with the single-chain fragment variable (scFv) of M3B2. In mice challenged with MPXV, Bis-M1M3 showed a notable protective effects. Analysis of neutralization mechanism showed that these mAbs and Bis-M1M3 exerted virus-neutralizing effects before the virus infects cells. In vivo pharmacokinetic experiments showed that Bis-M1M3 has a long half-life in rhesus macaques. This study provides crucial insights for further research on broad-spectrum antiviral drugs against MPXV and other orthopoxviruses.
Collapse
Affiliation(s)
- Zuning Ren
- State Key Laboratory of Organ Failure Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Mengjun Li
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Jiayin Chen
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Xiaohua Gong
- Shenzhen Key Laboratory of Pathogen and Immunity, State Key Discipline of Infectious Disease, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, People's Republic of China
| | - Shuo Song
- Shenzhen Key Laboratory of Pathogen and Immunity, State Key Discipline of Infectious Disease, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, People's Republic of China
| | - Delin Li
- Laboratory of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, People's Republic of China
| | - Minghui Yang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, People's Republic of China
| | - Jianhai Yu
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Sadia Asghar
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Yanxin Cui
- Shenzhen Key Laboratory of Pathogen and Immunity, State Key Discipline of Infectious Disease, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, People's Republic of China
| | - Shiyu Niu
- Shenzhen Key Laboratory of Pathogen and Immunity, State Key Discipline of Infectious Disease, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, People's Republic of China
| | - Zhonghui Liao
- Shenzhen Key Laboratory of Pathogen and Immunity, State Key Discipline of Infectious Disease, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, People's Republic of China
| | - Yushan Jiang
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Jiahui Liu
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Yuqing Li
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Bao Zhang
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Wei Zhao
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Jie Peng
- State Key Laboratory of Organ Failure Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Yang Yang
- Shenzhen Key Laboratory of Pathogen and Immunity, State Key Discipline of Infectious Disease, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, People's Republic of China
| | - Chenguang Shen
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
- Key Laboratory of Infectious Diseases Research in South China, Southern Medical University, Ministry of Education, People's Republic of China
| |
Collapse
|
6
|
Ramos KE, Okba NMA, Tan J, Bandawane P, Meade PS, Loganathan M, Francis B, Shulenin S, Holtsberg FW, Aman MJ, McMahon M, Krammer F, Lai JR. Broadly protective bispecific antibodies that simultaneously target influenza virus hemagglutinin and neuraminidase. mBio 2024; 15:e0108524. [PMID: 38899870 PMCID: PMC11253627 DOI: 10.1128/mbio.01085-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/06/2024] [Indexed: 06/21/2024] Open
Abstract
Monoclonal antibodies (mAbs) are an attractive therapeutic platform for the prevention and treatment of influenza virus infection. There are two major glycoproteins on the influenza virion surface: hemagglutinin (HA), which is responsible for viral attachment and entry, and neuraminidase (NA), which mediates viral egress by enzymatically cleaving sialic acid to release budding particles from the host cell surface. Broadly neutralizing antibodies (bNAbs) that target the conserved HA central stalk region, such as CR9114, can inhibit both viral entry and egress. More recently, broadly binding mAbs that engage and inhibit the NA active site, such as 1G01, have been described to prevent viral egress. Here, we engineered bispecific antibodies (bsAbs) that combine the variable domains of CR9114 and 1G01 into a single molecule and evaluated if simultaneous targeting of two different glycoproteins improved antiviral properties in vitro and in vivo. Several CR9114/1G01 bsAbs were generated with various configurations of the two sets of the variable domains ("bsAb formats"). We found that combinations employing the addition of a single-chain variable fragment in the hinge region of an IgG scaffold had the best properties in terms of expression, stability, and binding. Further characterization of selected bsAbs showed potent neutralizing and egress-inhibiting activity. One such bsAb ("hSC_CR9114_1G01") provided higher levels of prophylactic protection from mortality and morbidity upon challenge with H1N1 than either of the parental mAbs at low dosing (1 mg/kg). These results highlight the potential use of bsAbs that simultaneously target HA and NA as new influenza immunotherapeutics. IMPORTANCE Infection by the influenza virus remains a global health burden. The approaches utilized here to augment the activity of broadly protective influenza virus antibodies may lead to a new class of immunotherapies with enhanced activity.
Collapse
MESH Headings
- Neuraminidase/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/pharmacology
- Antibodies, Viral/immunology
- Animals
- Humans
- Mice
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/immunology
- Antibodies, Neutralizing/immunology
- Antibodies, Monoclonal/immunology
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Mice, Inbred BALB C
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/drug effects
Collapse
Affiliation(s)
- Kevin E. Ramos
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Nisreen M. A. Okba
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jessica Tan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Pooja Bandawane
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Philip S. Meade
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Madhumathi Loganathan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Benjamin Francis
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | | - M. Javad Aman
- Integrated BioTherapeutics, Inc., Rockville, Maryland, USA
| | - Meagan McMahon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
| | - Jonathan R. Lai
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
7
|
Li Z, Zhang Z, Rosen ST, Feng M. Function and mechanism of bispecific antibodies targeting SARS-CoV-2. CELL INSIGHT 2024; 3:100150. [PMID: 38374826 PMCID: PMC10875118 DOI: 10.1016/j.cellin.2024.100150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 02/21/2024]
Abstract
As the dynamic evolution of SARS-CoV-2 led to reduced efficacy in monoclonal neutralizing antibodies and emergence of immune escape, the role of bispecific antibodies becomes crucial in bolstering antiviral activity and suppressing immune evasion. This review extensively assesses a spectrum of representative bispecific antibodies targeting SARS-CoV-2, delving into their characteristics, design formats, mechanisms of action, and associated advantages and limitations. The analysis encompasses factors influencing the selection of parental antibodies and strategies for incorporating added benefits in bispecific antibody design. Furthermore, how different classes of parental antibodies contribute to augmenting the broad-spectrum neutralization capability within bispecific antibodies is discussed. In summary, this review presents analyses and discussions aimed at offering valuable insights for shaping future strategies in bispecific antibody design to effectively confront the challenges posed by SARS-CoV-2 and propel advancements in antiviral therapeutic development.
Collapse
Affiliation(s)
- Zhaohui Li
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Zengyuan Zhang
- Department of Molecular Microbiology & Immunology, University of Southern California, CA, USA
| | - Steven T. Rosen
- Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Mingye Feng
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| |
Collapse
|
8
|
Wirchnianski AS, Nyakatura EK, Herbert AS, Kuehne AI, Abbasi SA, Florez C, Storm N, McKay LGA, Dailey L, Kuang E, Abelson DM, Wec AZ, Chakraborti S, Holtsberg FW, Shulenin S, Bornholdt ZA, Aman MJ, Honko AN, Griffiths A, Dye JM, Chandran K, Lai JR. Design and characterization of protective pan-ebolavirus and pan-filovirus bispecific antibodies. PLoS Pathog 2024; 20:e1012134. [PMID: 38603762 PMCID: PMC11037526 DOI: 10.1371/journal.ppat.1012134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 04/23/2024] [Accepted: 03/18/2024] [Indexed: 04/13/2024] Open
Abstract
Monoclonal antibodies (mAbs) are an important class of antiviral therapeutics. MAbs are highly selective, well tolerated, and have long in vivo half-life as well as the capacity to induce immune-mediated virus clearance. Their activities can be further enhanced by integration of their variable fragments (Fvs) into bispecific antibodies (bsAbs), affording simultaneous targeting of multiple epitopes to improve potency and breadth and/or to mitigate against viral escape by a single mutation. Here, we explore a bsAb strategy for generation of pan-ebolavirus and pan-filovirus immunotherapeutics. Filoviruses, including Ebola virus (EBOV), Sudan virus (SUDV), and Marburg virus (MARV), cause severe hemorrhagic fever. Although there are two FDA-approved mAb therapies for EBOV infection, these do not extend to other filoviruses. Here, we combine Fvs from broad ebolavirus mAbs to generate novel pan-ebolavirus bsAbs that are potently neutralizing, confer protection in mice, and are resistant to viral escape. Moreover, we combine Fvs from pan-ebolavirus mAbs with those of protective MARV mAbs to generate pan-filovirus protective bsAbs. These results provide guidelines for broad antiviral bsAb design and generate new immunotherapeutic candidates.
Collapse
MESH Headings
- Animals
- Mice
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/pharmacology
- Antibodies, Bispecific/therapeutic use
- Ebolavirus/immunology
- Hemorrhagic Fever, Ebola/immunology
- Hemorrhagic Fever, Ebola/prevention & control
- Hemorrhagic Fever, Ebola/virology
- Antibodies, Viral/immunology
- Humans
- Filoviridae/immunology
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/therapeutic use
- Antibodies, Monoclonal/immunology
- Female
- Mice, Inbred BALB C
- Filoviridae Infections/immunology
- Filoviridae Infections/therapy
- Filoviridae Infections/prevention & control
Collapse
Affiliation(s)
- Ariel S. Wirchnianski
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Elisabeth K. Nyakatura
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Andrew S. Herbert
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
- The Geneva Foundation, Tacoma, Washington, United States of America
| | - Ana I. Kuehne
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Shawn A. Abbasi
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
- The Geneva Foundation, Tacoma, Washington, United States of America
| | - Catalina Florez
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
- The Geneva Foundation, Tacoma, Washington, United States of America
| | - Nadia Storm
- Department of Virology, Immunology, and Microbiology; and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Lindsay G. A. McKay
- Department of Virology, Immunology, and Microbiology; and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Leandrew Dailey
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Erin Kuang
- Mapp Biopharmaceutical Inc., San Diego, California, United States of America
| | - Dafna M. Abelson
- Mapp Biopharmaceutical Inc., San Diego, California, United States of America
| | - Anna Z. Wec
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Srinjoy Chakraborti
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | | | - Sergey Shulenin
- Integrated BioTherapeutics, Inc., Rockville, Maryland, United States of America
| | | | - M. Javad Aman
- Integrated BioTherapeutics, Inc., Rockville, Maryland, United States of America
| | - Anna N. Honko
- Department of Virology, Immunology, and Microbiology; and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Anthony Griffiths
- Department of Virology, Immunology, and Microbiology; and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - John M. Dye
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Jonathan R. Lai
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| |
Collapse
|
9
|
Xu S, Li W, Jiao C, Cao Z, Wu F, Yan F, Wang H, Feng N, Zhao Y, Yang S, Wang J, Xia X. A Bivalent Bacterium-like Particles-Based Vaccine Induced Potent Immune Responses against the Sudan Virus and Ebola Virus in Mice. Transbound Emerg Dis 2023; 2023:9248581. [PMID: 40303775 PMCID: PMC12017122 DOI: 10.1155/2023/9248581] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/04/2023] [Accepted: 03/06/2023] [Indexed: 05/02/2025]
Abstract
Ebola virus disease (EVD) is an acute viral hemorrhagic fever disease causing thousands of deaths. The large Ebola outbreak in 2014-2016 posed significant threats to global public health, requiring the development of multiple medical measures for disease control. Sudan virus (SUDV) and Zaire virus (EBOV) are responsible for severe disease and occasional deadly outbreaks in West Africa and Middle Africa. This study shows that bivalent bacterium-like particles (BLPs)-based vaccine, SUDV-EBOV BLPs (S/ZBLP + 2 + P), generated by mixing SUDV-BLPs and EBOV-BLPs at a 1 : 1 ratio, is immunogenic in mice. The SUDV-EBOV BLPs induced potent immune responses against SUDV and EBOV and elicited both T-helper 1 (Th1) and T-helper 2 (Th2) immune responses. The results indicated that SUDV-EBOV BLPs-based vaccine has the potential to be a promising candidate against SUDV and EBOV infections and provide a strategy to develop universal vaccines for EVD.
Collapse
Affiliation(s)
- Shengnan Xu
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- Shandong Agricultural University, Taian, China
| | - Wujian Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Cuicui Jiao
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zengguo Cao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Fangfang Wu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Feihu Yan
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Hualei Wang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Na Feng
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yongkun Zhao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Songtao Yang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Jianzhong Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
| | - Xianzhu Xia
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- Collaborative Innovation Center for Healthy Sheep Breeding and Zoonoses Prevention and Control, Shihezi University, Shihezi, Xinjiang, China
| |
Collapse
|
10
|
Ren P, Hu Y, Peng L, Yang L, Suzuki K, Fang Z, Bai M, Zhou L, Feng Y, Zou Y, Xiong Y, Chen S. Function and Cryo-EM structures of broadly potent bispecific antibodies against multiple SARS-CoV-2 Omicron sublineages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2022.08.09.503414. [PMID: 35982661 PMCID: PMC9387138 DOI: 10.1101/2022.08.09.503414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The SARS-CoV-2 variant, Omicron (B.1.1.529), rapidly swept the world since its emergence. Compared with previous variants, Omicron has a high number of mutations, especially those in its spike glycoprotein that drastically dampen or abolish the efficacy of currently available vaccines and therapeutic antibodies. Several major sublineages of Omicron evolved, including BA.1, BA.1.1, BA.2, BA.2.12.1, BA.3, BA.4/5, and BA.2.75, which rapidly changing the global and regional landscape of the pandemic. Although vaccines are available, therapeutic antibodies remain critical for infected and especially hospitalized patients. To address this, we have designed and generated a panel of human/humanized therapeutic bispecific antibodies against Omicron and its sub-lineage variants, with activity spectrum against other lineages. Among these, the top clone CoV2-0213 has broadly potent activities against multiple SARS-CoV-2 ancestral and Omicron lineages, including BA.1, BA.1.1, BA.2, BA.2.12.1, BA.3, BA.4/5, and BA.2.75. We have solved the cryo-EM structure of the lead bi-specific antibody CoV-0213 and its major Fab arm MB.02. Three-dimensional structural analysis shows distinct epitope of antibody - spike receptor binding domain (RBD) interactions and reveals that both Fab fragments of CoV2-0213 can simultaneously target one single spike RBD or two adjacent ones in the same spike trimer, further corroborating its mechanism of action. CoV2-0213 represents a unique and potent broad-spectrum SARS-CoV-2 neutralizing bispecific antibody (nbsAb) against the currently circulating major Omicron variants (BA.1, BA.1.1, BA.2, BA.2.12.1, BA.2.75, BA.3, and BA.4/5). CoV2-0213 is primarily human and ready for translational testing as a countermeasure against the ever-evolving pathogen.
Collapse
Affiliation(s)
- Ping Ren
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
| | - Yingxia Hu
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Lei Peng
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
| | - Luojia Yang
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Kazushi Suzuki
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
| | - Zhenhao Fang
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
| | - Meizhu Bai
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
| | - Liqun Zhou
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
- Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, CT, USA
| | - Yanzhi Feng
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
- Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, CT, USA
| | - Yongji Zou
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
| | - Yong Xiong
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Correspondence: SC (), +1-203-737-3825 (office), +1-203-737-4952 (lab), YX (), +1 (203) 436-2609 (lab)
| | - Sidi Chen
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, CT, USA
- Immunobiology Program, Yale University, New Haven, CT, USA
- Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT, USA
- Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA
- Center for Biomedical Data Science, Yale University School of Medicine, New Haven, CT, USA
- Correspondence: SC (), +1-203-737-3825 (office), +1-203-737-4952 (lab), YX (), +1 (203) 436-2609 (lab)
| |
Collapse
|
11
|
Chang MR, Tomasovic L, Kuzmina NA, Ronk AJ, Byrne PO, Johnson R, Storm N, Olmedillas E, Hou YJ, Schäfer A, Leist SR, Tse LV, Ke H, Coherd C, Nguyen K, Kamkaew M, Honko A, Zhu Q, Alter G, Saphire EO, McLellan JS, Griffiths A, Baric RS, Bukreyev A, Marasco WA. IgG-like bispecific antibodies with potent and synergistic neutralization against circulating SARS-CoV-2 variants of concern. Nat Commun 2022; 13:5814. [PMID: 36192374 PMCID: PMC9528872 DOI: 10.1038/s41467-022-33030-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 08/26/2022] [Indexed: 11/25/2022] Open
Abstract
Monoclonal antibodies are a promising approach to treat COVID-19, however the emergence of SARS-CoV-2 variants has challenged the efficacy and future of these therapies. Antibody cocktails are being employed to mitigate these challenges, but neutralization escape remains a major challenge and alternative strategies are needed. Here we present two anti-SARS-CoV-2 spike binding antibodies, one Class 1 and one Class 4, selected from our non-immune human single-chain variable fragment (scFv) phage library, that are engineered into four, fully-human IgG-like bispecific antibodies (BsAb). Prophylaxis of hACE2 mice and post-infection treatment of golden hamsters demonstrates the efficacy of the monospecific antibodies against the original Wuhan strain, while promising in vitro results with the BsAbs demonstrate enhanced binding and distinct synergistic effects on neutralizing activity against circulating variants of concern. In particular, one BsAb engineered in a tandem scFv-Fc configuration shows synergistic neutralization activity against several variants of concern including B.1.617.2. This work provides evidence that synergistic neutralization can be achieved using a BsAb scaffold, and serves as a foundation for the future development of broadly reactive BsAbs against emerging variants of concern.
Collapse
Affiliation(s)
- Matthew R Chang
- Department of Cancer Immunology & Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Luke Tomasovic
- Department of Cancer Immunology & Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Natalia A Kuzmina
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Galveston National Laboratory, Galveston, TX, 77555, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Adam J Ronk
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Galveston National Laboratory, Galveston, TX, 77555, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Patrick O Byrne
- Department of Molecular Biosciences, University of Texas, Austin, TX, 78712, USA
| | - Rebecca Johnson
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University, School of Medicine, Boston, MA, 02118, USA
| | - Nadia Storm
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University, School of Medicine, Boston, MA, 02118, USA
| | | | - Yixuan J Hou
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Sarah R Leist
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Longping V Tse
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Hanzhong Ke
- Department of Cancer Immunology & Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Christian Coherd
- Department of Cancer Immunology & Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Katrina Nguyen
- Department of Cancer Immunology & Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Maliwan Kamkaew
- Department of Cancer Immunology & Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Anna Honko
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University, School of Medicine, Boston, MA, 02118, USA
| | - Quan Zhu
- Department of Cancer Immunology & Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | | | - Jason S McLellan
- Department of Molecular Biosciences, University of Texas, Austin, TX, 78712, USA
| | - Anthony Griffiths
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University, School of Medicine, Boston, MA, 02118, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Galveston National Laboratory, Galveston, TX, 77555, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Wayne A Marasco
- Department of Cancer Immunology & Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
12
|
Liu CH, Hu YT, Wong SH, Lin LT. Therapeutic Strategies against Ebola Virus Infection. Viruses 2022; 14:v14030579. [PMID: 35336986 PMCID: PMC8954160 DOI: 10.3390/v14030579] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/03/2022] [Accepted: 03/08/2022] [Indexed: 12/10/2022] Open
Abstract
Since the 2014–2016 epidemic, Ebola virus (EBOV) has spread to several countries and has become a major threat to global health. EBOV is a risk group 4 pathogen, which imposes significant obstacles for the development of countermeasures against the virus. Efforts have been made to develop anti-EBOV immunization and therapeutics, with three vaccines and two antibody-based therapeutics approved in recent years. Nonetheless, the high fatality of Ebola virus disease highlights the need to continuously develop antiviral strategies for the future management of EBOV outbreaks in conjunction with vaccination programs. This review aims to highlight potential EBOV therapeutics and their target(s) of inhibition, serving as a summary of the literature to inform readers of the novel candidates available in the continued search for EBOV antivirals.
Collapse
Affiliation(s)
- Ching-Hsuan Liu
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Yee-Tung Hu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Shu Hui Wong
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Liang-Tzung Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Correspondence:
| |
Collapse
|
13
|
An engineered bispecific human monoclonal antibody against SARS-CoV-2. Nat Immunol 2022; 23:423-430. [PMID: 35228696 DOI: 10.1038/s41590-022-01138-w] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 01/13/2022] [Indexed: 12/23/2022]
Abstract
The global severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic requires effective therapies against coronavirus disease 2019 (COVID-19), and neutralizing antibodies are a promising therapy. A noncompeting pair of human neutralizing antibodies (B38 and H4) blocking SARS-CoV-2 binding to its receptor, ACE2, have been described previously. Here, we develop bsAb15, a bispecific monoclonal antibody (bsAb) based on B38 and H4. bsAb15 has greater neutralizing efficiency than these parental antibodies, results in less selective pressure and retains neutralizing ability to most SARS-CoV-2 variants of concern (with more potent neutralizing activity against the Delta variant). We also selected for escape mutants of the two parental mAbs, a mAb cocktail and bsAb15, demonstrating that bsAb15 can efficiently neutralize all single-mAb escape mutants. Furthermore, prophylactic and therapeutic application of bsAb15 reduced the viral titer in infected nonhuman primates and human ACE2 transgenic mice. Therefore, this bsAb is a feasible and effective strategy to treat and prevent severe COVID-19.
Collapse
|
14
|
Ricolinostat promotes the generation of megakaryocyte progenitors from human hematopoietic stem and progenitor cells. Stem Cell Res Ther 2022; 13:54. [PMID: 35123563 PMCID: PMC8817546 DOI: 10.1186/s13287-022-02722-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background Ex vivo production of induced megakaryocytes (MKs) and platelets from stem cells is an alternative approach for supplying transfusible platelets. However, it is difficult to generate large numbers of MKs and platelets from hematopoietic stem cells and progenitor cells (HSPCs).
Methods To optimize the differentiation efficiency of megakaryocytic cells from HSPCs, we first employed a platelet factor 4 (PF4)-promoter reporter and high-throughput screening strategy to screen for small molecules. We also investigated the effects and possible mechanisms of candidate small molecules on megakaryocytic differentiation of human HSPCs. Results The small molecule Ricolinostat remarkably promoted the expression of PF4-promoter reporter in the megakaryocytic cell line. Notably, Ricolinostat significantly enhanced the cell fate commitment of MK progenitors (MkPs) from cord blood HSPCs and promoted the proliferation of MkPs based on cell surface marker detection, colony-forming unit-MK assay, and quantitative real-time PCR analyses. MkPs generated from Ricolinostat-induced HSPCs differentiated into mature MKs and platelets. Mechanistically, we found that Ricolinostat enhanced MkP fate mainly by inhibiting the secretion of IL-8 and decreasing the expression of the IL-8 receptor CXCR2. Conclusion The addition of Ricolinostat to the culture medium promoted MkP differentiation from HSPCs and enhanced the proliferation of MkPs mainly by suppressing the IL-8/CXCR2 pathway. Our results can help the development of manufacturing protocols for the efficient generation of MKs and platelets from stem cells in vitro. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02722-5.
Collapse
|
15
|
Wirchnianski AS, Wec AZ, Nyakatura EK, Herbert AS, Slough MM, Kuehne AI, Mittler E, Jangra RK, Teruya J, Dye JM, Lai JR, Chandran K. Two Distinct Lysosomal Targeting Strategies Afford Trojan Horse Antibodies With Pan-Filovirus Activity. Front Immunol 2021; 12:729851. [PMID: 34721393 PMCID: PMC8551868 DOI: 10.3389/fimmu.2021.729851] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
Multiple agents in the family Filoviridae (filoviruses) are associated with sporadic human outbreaks of highly lethal disease, while others, including several recently identified agents, possess strong zoonotic potential. Although viral glycoprotein (GP)-specific monoclonal antibodies have demonstrated therapeutic utility against filovirus disease, currently FDA-approved molecules lack antiviral breadth. The development of broadly neutralizing antibodies has been challenged by the high sequence divergence among filovirus GPs and the complex GP proteolytic cleavage cascade that accompanies filovirus entry. Despite this variability in the antigenic surface of GP, all filoviruses share a site of vulnerability-the binding site for the universal filovirus entry receptor, Niemann-Pick C1 (NPC1). Unfortunately, this site is shielded in extracellular GP and only uncovered by proteolytic cleavage by host proteases in late endosomes and lysosomes, which are generally inaccessible to antibodies. To overcome this obstacle, we previously developed a 'Trojan horse' therapeutic approach in which engineered bispecific antibodies (bsAbs) coopt viral particles to deliver GP:NPC1 interaction-blocking antibodies to their endo/lysosomal sites of action. This approach afforded broad protection against members of the genus Ebolavirus but could not neutralize more divergent filoviruses. Here, we describe next-generation Trojan horse bsAbs that target the endo/lysosomal GP:NPC1 interface with pan-filovirus breadth by exploiting the conserved and widely expressed host cation-independent mannose-6-phosphate receptor for intracellular delivery. Our work highlights a new avenue for the development of single therapeutics protecting against all known and newly emerging filoviruses.
Collapse
Affiliation(s)
- Ariel S. Wirchnianski
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Anna Z. Wec
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Elisabeth K. Nyakatura
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Andrew S. Herbert
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
- The Geneva Foundation, Tacoma, WA, United States
| | - Megan M. Slough
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Ana I. Kuehne
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Eva Mittler
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Rohit K. Jangra
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Jonathan Teruya
- Antibody Discovery and Research group, Mapp Biopharmaceutical, San Diego, CA, United States
| | - John M. Dye
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Jonathan R. Lai
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
16
|
Tshiani Mbaya O, Mukumbayi P, Mulangu S. Review: Insights on Current FDA-Approved Monoclonal Antibodies Against Ebola Virus Infection. Front Immunol 2021; 12:721328. [PMID: 34526994 PMCID: PMC8435780 DOI: 10.3389/fimmu.2021.721328] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 08/12/2021] [Indexed: 11/29/2022] Open
Abstract
The unprecedented 2013-2016 West Africa Ebola outbreak accelerated several medical countermeasures (MCMs) against Ebola virus disease (EVD). Several investigational products (IPs) were used throughout the outbreak but were not conclusive for efficacy results. Only the Randomized Controlled Trial (RCT) on ZMapp was promising but inconclusive. More recently, during the second-largest Ebola outbreak in North Kivu and Ituri provinces, Democratic Republic of the Congo (DRC), four IPs, including one small molecule (Remdesivir), two monoclonal antibody (mAb) cocktails (ZMapp and REGN-EB3) and a single mAb (mAb114), were evaluated in an RCT, the Pamoja Tulinde Maisha (PALM) study. Two products (REGN-EB3 and mAb114) demonstrated efficacy as compared to the control arm, ZMapp. There were remarkably few side effects recorded in the trial. The FDA approved both medications in this scientifically sound study, marking a watershed moment in the field of EVD therapy. These products can be produced relatively inexpensively and can be stockpiled. The administration of mAbs in EVD patients appears to be safe and effective, while several critical knowledge gaps remain; the impact of early administration of Ebola-specific mAbs on developing a robust immune response for future Ebola virus exposure is unknown. The viral mutation escape, leading to resistance, presents a potential limitation for single mAb therapy; further improvements need to be explored. Understanding the contribution of Fc-mediated antibody functions such as antibody-dependent cellular cytotoxicity (ADCC) of those approved mAbs is still critical. The potential merit of combination therapy and post-exposure prophylaxis (PEP) need to be demonstrated. Furthermore, the PALM trial has accounted for 30% of mortality despite the administration of specific treatments. The putative role of EBOV soluble Glycoprotein (sGP) as a decoy to the immune system, the virus persistence, and relapses might be investigated for treatment failure. The development of pan-filovirus or pan-species mAbs remains essential for protection. The interaction between FDA-approved mAbs and vaccines remains unclear and needs to be investigated. In this review, we summarize the efficacy and safety results of the PALM study and review current research questions for the further development of mAbs in pre-exposure or emergency post-exposure use.
Collapse
Affiliation(s)
- Olivier Tshiani Mbaya
- Clinical Monitoring Research Program Directorate, Leidos Biomedical Research, Frederick, MD, United States
| | - Philippe Mukumbayi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Sabue Mulangu
- Global Medical Affairs, Ridgeback Biotherapeutics, Miami, FL, United States
| |
Collapse
|
17
|
Escaffre O, Juelich TL, Neef N, Massey S, Smith J, Brasel T, Smith JK, Kalveram B, Zhang L, Perez D, Ikegami T, Freiberg AN, Comer JE. STAT-1 Knockout Mice as a Model for Wild-Type Sudan Virus (SUDV). Viruses 2021; 13:v13071388. [PMID: 34372594 PMCID: PMC8310124 DOI: 10.3390/v13071388] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 12/03/2022] Open
Abstract
Currently there is no FDA-licensed vaccine or therapeutic against Sudan ebolavirus (SUDV) infections. The largest ever reported 2014–2016 West Africa outbreak, as well as the 2021 outbreak in the Democratic Republic of Congo, highlight the critical need for countermeasures against filovirus infections. A well-characterized small animal model that is susceptible to wild-type filoviruses would greatly add to the screening of antivirals and vaccines. Here, we infected signal transducer and activator of transcription-1 knock out (STAT-1 KO) mice with five different wildtype filoviruses to determine susceptibility. SUDV and Marburg virus (MARV) were the most virulent, and caused 100% or 80% lethality, respectively. Zaire ebolavirus (EBOV), Bundibugyo ebolavirus (BDBV), and Taï Forest ebolavirus (TAFV) caused 40%, 20%, and no mortality, respectively. Further characterization of SUDV in STAT-1 KO mice demonstrated lethality down to 3.1 × 101 pfu. Viral genomic material was detectable in serum as early as 1 to 2 days post-challenge. The onset of viremia was closely followed by significant changes in total white blood cells and proportion of neutrophils and lymphocytes, as well as by an influx of neutrophils in the liver and spleen. Concomitant significant fluctuations in blood glucose, albumin, globulin, and alanine aminotransferase were also noted, altogether consistent with other models of filovirus infection. Finally, favipiravir treatment fully protected STAT-1 KO mice from lethal SUDV challenge, suggesting that this may be an appropriate small animal model to screen anti-SUDV countermeasures.
Collapse
Affiliation(s)
- Olivier Escaffre
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; (O.E.); (T.L.J.); (J.K.S.); (B.K.); (L.Z.); (T.I.)
| | - Terry L. Juelich
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; (O.E.); (T.L.J.); (J.K.S.); (B.K.); (L.Z.); (T.I.)
| | - Natasha Neef
- XTR Toxicologic Pathology Services LLC, Sterling, VA 20165, USA;
| | - Shane Massey
- Office of Regulated Nonclinical Studies, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; (S.M.); (J.S.); (T.B.)
| | - Jeanon Smith
- Office of Regulated Nonclinical Studies, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; (S.M.); (J.S.); (T.B.)
| | - Trevor Brasel
- Office of Regulated Nonclinical Studies, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; (S.M.); (J.S.); (T.B.)
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
- The Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Jennifer K. Smith
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; (O.E.); (T.L.J.); (J.K.S.); (B.K.); (L.Z.); (T.I.)
| | - Birte Kalveram
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; (O.E.); (T.L.J.); (J.K.S.); (B.K.); (L.Z.); (T.I.)
| | - Lihong Zhang
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; (O.E.); (T.L.J.); (J.K.S.); (B.K.); (L.Z.); (T.I.)
| | - David Perez
- Texas A&M University Division of Research, Texas A&M University, College Station, TX 77843, USA;
| | - Tetsuro Ikegami
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; (O.E.); (T.L.J.); (J.K.S.); (B.K.); (L.Z.); (T.I.)
- The Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Alexander N. Freiberg
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; (O.E.); (T.L.J.); (J.K.S.); (B.K.); (L.Z.); (T.I.)
- The Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
- Correspondence: (A.N.F.); (J.E.C.)
| | - Jason E. Comer
- Office of Regulated Nonclinical Studies, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; (S.M.); (J.S.); (T.B.)
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
- The Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
- Institute of Translational Sciences, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
- Correspondence: (A.N.F.); (J.E.C.)
| |
Collapse
|
18
|
Zivcec M, Spiropoulou CF, Spengler JR. The use of mice lacking type I or both type I and type II interferon responses in research on hemorrhagic fever viruses. Part 2: Vaccine efficacy studies. Antiviral Res 2020; 174:104702. [PMID: 31982149 DOI: 10.1016/j.antiviral.2019.104702] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/13/2019] [Accepted: 12/20/2019] [Indexed: 12/24/2022]
Abstract
For more than 20 years, researchers have used laboratory mice lacking type I or both type I and II interferon (IFN) responses to study high-containment viruses that cause hemorrhagic fevers (HF) in humans. With the exception of Rift Valley fever virus, agents that cause viral HF in humans, such as Ebola and Lassa virus, do not cause disease in mature immunocompetent mice. In contrast, IFN-deficient mice typically develop severe or fatal disease when inoculated with these agents. The sensitivity of IFN-deficient mice to disease has led to their widespread use in biocontainment laboratories to assess the efficacy of novel vaccines against HF viruses, often without considering whether adaptive immune responses in IFN-deficient mice accurately mirror those in immunocompetent humans. Failure to recognize these questions may lead to inappropriate expectations of the predictive value of mouse experiments. In two invited articles, we investigate these questions. The present article reviews the use of IFN-deficient mice for assessing novel vaccines against HF viruses, including Ebola, Lassa, Crimean-Congo hemorrhagic fever and Rift Valley fever viruses. A companion paper examines the general question of how the lack of IFN signaling may affect adaptive immune responses and the outcome of vaccine studies in mice.
Collapse
Affiliation(s)
- Marko Zivcec
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Christina F Spiropoulou
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jessica R Spengler
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| |
Collapse
|
19
|
Cholesterol-conjugated stapled peptides inhibit Ebola and Marburg viruses in vitro and in vivo. Antiviral Res 2019; 171:104592. [PMID: 31473342 DOI: 10.1016/j.antiviral.2019.104592] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 08/22/2019] [Accepted: 08/25/2019] [Indexed: 02/07/2023]
Abstract
Filoviridae currently includes five official and one proposed genera. Genus Ebolavirus includes five established and one proposed ebolavirus species for Bombali virus (BOMV), Bundibugyo virus (BDBV), Ebola virus (EBOV), Reston virus (RESTV), Sudan virus (SUDV) and Taï Forest virus (TAFV), and genus Marburgvirus includes a single species for Marburg virus (MARV) and Ravn virus (RAVV). Ebola virus (EBOV) has emerged as a significant public health concern since the 2013-2016 Ebola Virus Disease outbreak in Western Africa. Currently, there are no therapeutics approved and the need for Ebola-specific therapeutics remains a gap. In search for anti-Ebola therapies we tested the idea of using inhibitory properties of peptides corresponding to the C-terminal heptad-repeat (HR2) domains of class I fusion proteins against EBOV infection. The fusion protein GP2 of EBOV belongs to class I, suggesting that a similar strategy to HIV may be applied to inhibit EBOV infection. The serum half-life of peptides was expanded by cholesterol conjugation to allow daily dosing. The peptides were further constrained to stabilize a helical structure to increase the potency of inhibition. The EC50s of lead peptides were in low micromolar range, as determined by a high-content imaging test of EBOV-infected cells. Lead peptides were tested in an EBOV lethal mouse model and efficacy of the peptides were determined following twice-daily administration of peptides for 9 days. The most potent peptide was able to protect mice from lethal challenge of mouse-adapted Ebola virus. These data show that engineered peptides coupled with cholesterol can inhibit viral production, protect mice against lethal EBOV infection, and may be used to build novel therapeutics against EBOV.
Collapse
|
20
|
Banadyga L, Schiffman Z, He S, Qiu X. Virus inoculation and treatment regimens for evaluating anti-filovirus monoclonal antibody efficacy in vivo. BIOSAFETY AND HEALTH 2019; 1:6-13. [PMID: 32835206 PMCID: PMC7347303 DOI: 10.1016/j.bsheal.2019.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/07/2019] [Accepted: 02/21/2019] [Indexed: 01/05/2023] Open
Abstract
The development of monoclonal antibodies to treat disease caused by filoviruses, particularly Ebola virus, has risen steeply in recent years thanks to several key studies demonstrating their remarkable therapeutic potential. The increased drive to develop new and better monoclonal antibodies has necessarily seen an increase in animal model efficacy testing, which is critical to the pre-clinical development of any novel countermeasure. Primary and secondary efficacy testing against filoviruses typically makes use of one or more rodent models (mice, guinea pigs, and occasionally hamsters) or the more recently described ferret model, although the exact choice of model depends on the specific filovirus being evaluated. Indeed, no single small animal model exists for all filoviruses, and the use of any given model must consider the nature of that model as well as the nature of the therapeutic and the experimental objectives. Confirmatory evaluation, on the other hand, is performed in nonhuman primates (rhesus or cynomolgus macaques) regardless of the filovirus. In light of the number of different animal models that are currently used in monoclonal antibody efficacy testing, we sought to better understand how these efficacy tests are being performed by numerous different laboratories around the world. To this end, we review the animal models that are being used for antibody efficacy testing against filoviruses, and we highlight the challenge doses and routes of infection that are used. We also describe the various antibody treatment regimens, including antibody dose, route, and schedule of administration, that are used in these model systems. We do not identify any single best model or treatment regimen, and we do not advocate for field-wide protocol standardization. Instead, we hope to provide a comprehensive resource that will facilitate and enhance the continued pre-clinical development of novel monoclonal antibody therapeutics.
Collapse
Affiliation(s)
- Logan Banadyga
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada
| | - Zachary Schiffman
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada.,Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Shihua He
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada
| | - Xiangguo Qiu
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada.,Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
21
|
Meyer M, Malherbe DC, Bukreyev A. Can Ebola Virus Vaccines Have Universal Immune Correlates of protection? Trends Microbiol 2019; 27:8-16. [PMID: 30201511 PMCID: PMC6309495 DOI: 10.1016/j.tim.2018.08.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 07/30/2018] [Accepted: 08/15/2018] [Indexed: 12/22/2022]
Abstract
Testing vaccine efficacy against the highly lethal Ebola virus (EBOV) in humans is almost impossible due to obvious ethical reasons and the sporadic nature of outbreaks. For such situations, the 'animal rule' was established, requiring the product be tested in animal models, expected to predict the response observed in humans. For vaccines, this testing aims to identify immune correlates of protection, such as antibody or cell-mediated responses. In the wake of the 2013-2016 EBOV epidemic, and despite advancement of promising candidates into clinical trials, protective correlates remain ambiguous. In the hope of identifying a reliable correlate by comparing preclinical and clinical trial data on immune responses to vaccination, we conclude that correlates are not universal for all EBOV vaccines.
Collapse
Affiliation(s)
- Michelle Meyer
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77555, USA; These authors contributed equally to this work
| | - Delphine C Malherbe
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77555, USA; These authors contributed equally to this work
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Department Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77555, USA.
| |
Collapse
|
22
|
Nyakatura EK, Zak SE, Wec AZ, Hofmann D, Shulenin S, Bakken RR, Aman MJ, Chandran K, Dye JM, Lai JR. Design and evaluation of bi- and trispecific antibodies targeting multiple filovirus glycoproteins. J Biol Chem 2018; 293:6201-6211. [PMID: 29500195 DOI: 10.1074/jbc.ra117.001627] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/27/2018] [Indexed: 12/21/2022] Open
Abstract
Filoviruses (family Filoviridae) include five ebolaviruses and Marburg virus. These pathogens cause a rapidly progressing and severe viral disease with high mortality rates (generally 30-90%). Outbreaks of filovirus disease are sporadic and, until recently, were limited to less than 500 cases. However, the 2013-2016 epidemic in western Africa, caused by Ebola virus (EBOV), illustrated the potential of filovirus outbreaks to escalate to a much larger scale (over 28,000 suspected cases). mAbs against the envelope glycoprotein represent a promising therapeutic platform for managing filovirus infections. However, mAbs that exhibit neutralization or protective properties against multiple filoviruses are rare. Here we examined a panel of engineered bi- and trispecific antibodies, in which variable domains of mAbs that target epitopes from multiple filoviruses were combined, for their capacity to neutralize viral infection across filovirus species. We found that bispecific combinations targeting EBOV and Sudan virus (another ebolavirus), provide potent cross-neutralization and protection in mice. Furthermore, trispecific combinations, targeting EBOV, Sudan virus, and Marburg virus, exhibited strong neutralization potential against all three viruses. These results provide important insights into multispecific antibody engineering against filoviruses and will inform future immunotherapeutic discoveries.
Collapse
Affiliation(s)
| | - Samantha E Zak
- the Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland 21702, and
| | - Anna Z Wec
- Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York 10461
| | | | - Sergey Shulenin
- Integrated Biotherapeutics Inc., Gaithersburg, Maryland 20878
| | - Russell R Bakken
- the Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland 21702, and
| | - M Javad Aman
- Integrated Biotherapeutics Inc., Gaithersburg, Maryland 20878
| | - Kartik Chandran
- Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - John M Dye
- the Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland 21702, and
| | | |
Collapse
|
23
|
Liu JL, Shriver-Lake LC, Anderson GP, Zabetakis D, Goldman ER. Selection, characterization, and thermal stabilization of llama single domain antibodies towards Ebola virus glycoprotein. Microb Cell Fact 2017; 16:223. [PMID: 29233140 PMCID: PMC5726015 DOI: 10.1186/s12934-017-0837-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 11/28/2017] [Indexed: 12/26/2022] Open
Abstract
Background A key advantage of recombinant antibody technology is the ability to optimize and tailor reagents. Single domain antibodies (sdAbs), the recombinantly produced variable domains derived from camelid and shark heavy chain antibodies, provide advantages of stability and solubility and can be further engineered to enhance their properties. In this study, we generated sdAbs specific for Ebola virus envelope glycoprotein (GP) and increased their stability to expand their utility for use in austere locals. Ebola virus is extremely virulent and causes fatal hemorrhagic fever in ~ 50 percent of the cases. The viral GP binds to host cell receptors to facilitate viral entry and thus plays a critical role in pathogenicity. Results An immune phage display library containing more than 107 unique clones was developed from a llama immunized with a combination of killed Ebola virus and recombinantly produced GP. We panned the library to obtain GP binding sdAbs and isolated sdAbs from 5 distinct sequence families. Three GP binders with dissociation constants ranging from ~ 2 to 20 nM, and melting temperatures from ~ 57 to 72 °C were selected for protein engineering in order to increase their stability through a combination of consensus sequence mutagenesis and the addition of a non-canonical disulfide bond. These changes served to increase the melting temperatures of the sdAbs by 15–17 °C. In addition, fusion of a short positively charged tail to the C-terminus which provided ideal sites for the chemical modification of these sdAbs resulted in improved limits of detection of GP and Ebola virus like particles while serving as tracer antibodies. Conclusions SdAbs specific for Ebola GP were selected and their stability and functionality were improved utilizing protein engineering. Thermal stability of antibody reagents may be of particular importance when operating in austere locations that lack reliable refrigeration. Future efforts can evaluate the potential of these isolated sdAbs as candidates for diagnostic or therapeutic applications for Ebola. Electronic supplementary material The online version of this article (10.1186/s12934-017-0837-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jinny L Liu
- US Naval Research Laboratory, Center for Bio/Molecular Science and Engineering, 4555 Overlook Ave SW, Washington, DC, 20375, USA
| | - Lisa C Shriver-Lake
- US Naval Research Laboratory, Center for Bio/Molecular Science and Engineering, 4555 Overlook Ave SW, Washington, DC, 20375, USA
| | - George P Anderson
- US Naval Research Laboratory, Center for Bio/Molecular Science and Engineering, 4555 Overlook Ave SW, Washington, DC, 20375, USA
| | - Dan Zabetakis
- US Naval Research Laboratory, Center for Bio/Molecular Science and Engineering, 4555 Overlook Ave SW, Washington, DC, 20375, USA
| | - Ellen R Goldman
- US Naval Research Laboratory, Center for Bio/Molecular Science and Engineering, 4555 Overlook Ave SW, Washington, DC, 20375, USA.
| |
Collapse
|
24
|
Hofmann D, Zak SE, Nyakatura EK, Mittler E, Bakken RR, Chandran K, Dye JM, Lai JR. Mechanistic and Fc requirements for inhibition of Sudan virus entry and in vivo protection by a synthetic antibody. Immunol Lett 2017; 190:289-295. [PMID: 28890093 DOI: 10.1016/j.imlet.2017.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 08/28/2017] [Accepted: 09/05/2017] [Indexed: 11/18/2022]
Abstract
The Sudan virus (SUDV), an ebolavirus, causes severe hemorrhagic fever with human case fatality rates of ∼50%. Previous work from our lab demonstrated the synthetic antibody F4 potently inhibits viral entry and protects against lethal virus challenge in mice [Chen et al., ACS Chem. Biol., 2014, 9, 2263-2273]. Here, we explore mechanistic requirements as well as contribution of the Fc region and function on neutralization and in vivo protection. Live cell imaging demonstrates that the antibody colocalizes with vesicular stomatitis virus particles containing the Sudan virus glycoprotein (VSV-GPSUDV) and that the antibody is rapidly degraded within cellular endosomes. A viral escape mutant contained substitutions on the N-heptad repeat (NHR) segment of GP2, the fusion subunit. Truncation studies indicated that the size of the Fc impacts virus neutralization potential. Finally, we examined the protective efficacy of Fc-null mutants in mice, and found that Fc function was not required for high levels of protection. Altogether, these results indicate that neutralization of SUDV GP-mediated cell entry likely involves blockade of viral membrane fusion within endosomes, and that inhibition of viral entry is the likely mechanism of in vivo protection.
Collapse
Affiliation(s)
- Daniel Hofmann
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA
| | - Samantha E Zak
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD 21702, USA
| | - Elisabeth K Nyakatura
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA
| | - Eva Mittler
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA
| | - Russell R Bakken
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD 21702, USA
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA
| | - John M Dye
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD 21702, USA
| | - Jonathan R Lai
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA.
| |
Collapse
|
25
|
Duehr J, Wohlbold TJ, Oestereich L, Chromikova V, Amanat F, Rajendran M, Gomez-Medina S, Mena I, tenOever BR, García-Sastre A, Basler CF, Munoz-Fontela C, Krammer F. Novel Cross-Reactive Monoclonal Antibodies against Ebolavirus Glycoproteins Show Protection in a Murine Challenge Model. J Virol 2017; 91:e00652-17. [PMID: 28592526 PMCID: PMC5533894 DOI: 10.1128/jvi.00652-17] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 05/28/2017] [Indexed: 11/20/2022] Open
Abstract
Out of an estimated 31,100 cases since their discovery in 1976, ebolaviruses have caused approximately 13,000 deaths. The vast majority (∼11,000) of these occurred during the 2013-2016 West African epidemic. Three out of five species in the genus are known to cause Ebola Virus Disease in humans. Several monoclonal antibodies against the ebolavirus glycoprotein are currently in development as therapeutics. However, there is still a paucity of monoclonal antibodies that can cross-react between the glycoproteins of different ebolavirus species, and the mechanism of these monoclonal antibody therapeutics is still not understood in detail. Here, we generated a panel of eight murine monoclonal antibodies (MAbs) utilizing a prime-boost vaccination regimen with a Zaire ebolavirus glycoprotein expression plasmid followed by infection with a vesicular stomatitis virus expressing the Zaire ebolavirus glycoprotein. We tested the binding breadth of the resulting monoclonal antibodies using a set of recombinant surface glycoproteins from Reston, Taï Forest, Bundibugyo, Zaire, Sudan, and Marburg viruses and found two antibodies that showed pan-ebolavirus binding. An in vivo Stat2-/- mouse model was utilized to test the ability of these MAbs to protect from infection with a vesicular stomatitis virus expressing the Zaire ebolavirus glycoprotein. Several of our antibodies, including the broadly binding ones, protected mice from mortality despite lacking neutralization capability in vitro, suggesting their protection may be mediated by Fc-FcR interactions. Indeed, three antibodies displayed cellular phagocytosis and/or antibody-dependent cell-mediated cytotoxicity in vitro Our antibodies, specifically the two identified cross-reactive monoclonal antibodies (KL-2E5 and KL-2H7), might add to the understanding of anti-ebolavirus humoral immunity.IMPORTANCE This study describes the generation of a panel of novel anti-ebolavirus glycoprotein monoclonal antibodies, including two antibodies with broad cross-reactivity to all known ebolavirus species. The antibodies were raised using a heterologous DNA-viral vector prime-boost regimen, resulting in a high proportion of cross-reactive antibodies (25%). Similar vaccination regimens have been used successfully to induce broad protection against influenza viruses in humans, and our limited data indicate that this might be a useful strategy for filovirus vaccines as well. Several of our antibodies showed protective efficacy when tested in a novel murine challenge model and may be developed into future therapeutics.
Collapse
Affiliation(s)
- James Duehr
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Teddy John Wohlbold
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lisa Oestereich
- Department of Virology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Veronika Chromikova
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fatima Amanat
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Madhusudan Rajendran
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sergio Gomez-Medina
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Ignacio Mena
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benjamin R tenOever
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christopher F Basler
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Cesar Munoz-Fontela
- Department of Virology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
26
|
Antibodies from a Human Survivor Define Sites of Vulnerability for Broad Protection against Ebolaviruses. Cell 2017; 169:878-890.e15. [PMID: 28525755 DOI: 10.1016/j.cell.2017.04.037] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/16/2017] [Accepted: 04/26/2017] [Indexed: 12/19/2022]
Abstract
Experimental monoclonal antibody (mAb) therapies have shown promise for treatment of lethal Ebola virus (EBOV) infections, but their species-specific recognition of the viral glycoprotein (GP) has limited their use against other divergent ebolaviruses associated with human disease. Here, we mined the human immune response to natural EBOV infection and identified mAbs with exceptionally potent pan-ebolavirus neutralizing activity and protective efficacy against three virulent ebolaviruses. These mAbs recognize an inter-protomer epitope in the GP fusion loop, a critical and conserved element of the viral membrane fusion machinery, and neutralize viral entry by targeting a proteolytically primed, fusion-competent GP intermediate (GPCL) generated in host cell endosomes. Only a few somatic hypermutations are required for broad antiviral activity, and germline-approximating variants display enhanced GPCL recognition, suggesting that such antibodies could be elicited more efficiently with suitably optimized GP immunogens. Our findings inform the development of both broadly effective immunotherapeutics and vaccines against filoviruses.
Collapse
|
27
|
Nelson EA, Dyall J, Hoenen T, Barnes AB, Zhou H, Liang JY, Michelotti J, Dewey WH, DeWald LE, Bennett RS, Morris PJ, Guha R, Klumpp-Thomas C, McKnight C, Chen YC, Xu X, Wang A, Hughes E, Martin S, Thomas C, Jahrling PB, Hensley LE, Olinger GG, White JM. The phosphatidylinositol-3-phosphate 5-kinase inhibitor apilimod blocks filoviral entry and infection. PLoS Negl Trop Dis 2017; 11:e0005540. [PMID: 28403145 PMCID: PMC5402990 DOI: 10.1371/journal.pntd.0005540] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 04/24/2017] [Accepted: 03/30/2017] [Indexed: 12/12/2022] Open
Abstract
Phosphatidylinositol-3-phosphate 5-kinase (PIKfyve) is a lipid kinase involved in endosome maturation that emerged from a haploid genetic screen as being required for Ebola virus (EBOV) infection. Here we analyzed the effects of apilimod, a PIKfyve inhibitor that was reported to be well tolerated in humans in phase 2 clinical trials, for its effects on entry and infection of EBOV and Marburg virus (MARV). We first found that apilimod blocks infections by EBOV and MARV in Huh 7, Vero E6 and primary human macrophage cells, with notable potency in the macrophages (IC50, 10 nM). We next observed that similar doses of apilimod block EBOV-glycoprotein-virus like particle (VLP) entry and transcription-replication competent VLP infection, suggesting that the primary mode of action of apilimod is as an entry inhibitor, preventing release of the viral genome into the cytoplasm to initiate replication. After providing evidence that the anti-EBOV action of apilimod is via PIKfyve, we showed that it blocks trafficking of EBOV VLPs to endolysosomes containing Niemann-Pick C1 (NPC1), the intracellular receptor for EBOV. Concurrently apilimod caused VLPs to accumulate in early endosome antigen 1-positive endosomes. We did not detect any effects of apilimod on bulk endosome acidification, on the activity of cathepsins B and L, or on cholesterol export from endolysosomes. Hence by antagonizing PIKfyve, apilimod appears to block EBOV trafficking to its site of fusion and entry into the cytoplasm. Given the drug's observed anti-filoviral activity, relatively unexplored mechanism of entry inhibition, and reported tolerability in humans, we propose that apilimod be further explored as part of a therapeutic regimen to treat filoviral infections.
Collapse
Affiliation(s)
- Elizabeth A. Nelson
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Julie Dyall
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Thomas Hoenen
- Laboratory of Virology, Division of Intramural Research, National Institutes of Health, Hamilton, Montana, United States of America
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald–Insel Riems, Germany
| | - Alyson B. Barnes
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Huanying Zhou
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Janie Y. Liang
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Julia Michelotti
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - William H. Dewey
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Lisa Evans DeWald
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Richard S. Bennett
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Patrick J. Morris
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Rajarshi Guha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Carleen Klumpp-Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Crystal McKnight
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yu-Chi Chen
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Xin Xu
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Amy Wang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Emma Hughes
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Scott Martin
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Craig Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Peter B. Jahrling
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Lisa E. Hensley
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Gene G. Olinger
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Judith M. White
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
28
|
Liang J, Jangra RK, Bollinger L, Wada J, Radoshitzky SR, Chandran K, Jahrling PB, Kuhn JH, Jensen KS. Candidate medical countermeasures targeting Ebola virus cell entry. Future Virol 2017. [DOI: 10.2217/fvl-2016-0113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Medical countermeasures (MCMs) against virus infections ideally prevent the adsorption or entry of virions into target cells, thereby circumventing infection. Recent significant advances in elucidating the mechanism of Ebola virus (EBOV) host-cell penetration include the involvement of two-pore channels at the early stage of entry, and identification of cellular proteases for EBOV spike glycoprotein maturation and the intracellular EBOV receptor, Niemann–Pick type C1. This improved understanding of the initial steps of EBOV infection is now increasingly applied to rapid development of candidate MCMs, some of which have already entered the clinic. Candidate MCMs discussed include antibodies, small molecules and peptides that target various stages of the described EBOV cell-entry pathway. In this review, we summarize the currently known spectrum of EBOV cell-entry inhibitors, describe their mechanism of action and evaluate their potential for future development.
Collapse
Affiliation(s)
- Janie Liang
- Integrated Research Facility at Fort Detrick, National Institute of Allergy & Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Rohit K Jangra
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Laura Bollinger
- Integrated Research Facility at Fort Detrick, National Institute of Allergy & Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Jiro Wada
- Integrated Research Facility at Fort Detrick, National Institute of Allergy & Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Sheli R Radoshitzky
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Kartik Chandran
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Peter B Jahrling
- Integrated Research Facility at Fort Detrick, National Institute of Allergy & Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Jens H Kuhn
- Integrated Research Facility at Fort Detrick, National Institute of Allergy & Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Kenneth S Jensen
- Integrated Research Facility at Fort Detrick, National Institute of Allergy & Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| |
Collapse
|
29
|
Abstract
Mouse models of Ebola virus (EBOV) have demonstrated their utility as important tools for screening the efficacy of candidate therapeutics and vaccines. In this chapter we explain the various mouse models that utilize either wild-type or mouse-adapted EBOV variants.
Collapse
|
30
|
Abstract
Bispecific antibody engineering, in which binding specificities toward 2 distinct epitopes are combined into a single molecule, can greatly enhance immunotherapeutic properties of monoclonal antibodies. While the bispecific antibody approach has been applied widely to targets for indications such as cancer and inflammation, the development of such agents for viral immunotherapy is only now emerging. Here, we review recent advances in the development of bispecific antibodies for viral immunotherapy, highlighting promising in vitro and in vivo results.
Collapse
Affiliation(s)
- Elisabeth K Nyakatura
- a Department of Biochemistry , Albert Einstein College of Medicine , Bronx , NY , USA
| | - Alexandra Y Soare
- a Department of Biochemistry , Albert Einstein College of Medicine , Bronx , NY , USA
| | - Jonathan R Lai
- a Department of Biochemistry , Albert Einstein College of Medicine , Bronx , NY , USA
| |
Collapse
|
31
|
Wec AZ, Nyakatura EK, Herbert AS, Howell KA, Holtsberg FW, Bakken RR, Mittler E, Christin JR, Shulenin S, Jangra RK, Bharrhan S, Kuehne AI, Bornholdt ZA, Flyak AI, Saphire EO, Crowe JE, Aman MJ, Dye JM, Lai JR, Chandran K. A "Trojan horse" bispecific-antibody strategy for broad protection against ebolaviruses. Science 2016; 354:350-354. [PMID: 27608667 PMCID: PMC5647781 DOI: 10.1126/science.aag3267] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/25/2016] [Indexed: 12/26/2022]
Abstract
There is an urgent need for monoclonal antibody (mAb) therapies that broadly protect against Ebola virus and other filoviruses. The conserved, essential interaction between the filovirus glycoprotein, GP, and its entry receptor Niemann-Pick C1 (NPC1) provides an attractive target for such mAbs but is shielded by multiple mechanisms, including physical sequestration in late endosomes. Here, we describe a bispecific-antibody strategy to target this interaction, in which mAbs specific for NPC1 or the GP receptor-binding site are coupled to a mAb against a conserved, surface-exposed GP epitope. Bispecific antibodies, but not parent mAbs, neutralized all known ebolaviruses by coopting viral particles themselves for endosomal delivery and conferred postexposure protection against multiple ebolaviruses in mice. Such "Trojan horse" bispecific antibodies have potential as broad antifilovirus immunotherapeutics.
Collapse
Affiliation(s)
- Anna Z Wec
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Elisabeth K Nyakatura
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Andrew S Herbert
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Katie A Howell
- Integrated Biotherapeutics Inc., Gaithersburg, MD 20878, USA
| | | | - Russell R Bakken
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Eva Mittler
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - John R Christin
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Sergey Shulenin
- Integrated Biotherapeutics Inc., Gaithersburg, MD 20878, USA
| | - Rohit K Jangra
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Sushma Bharrhan
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ana I Kuehne
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Zachary A Bornholdt
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 10550, USA
| | - Andrew I Flyak
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN 37235, USA
| | - Erica Ollmann Saphire
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 10550, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 10550, USA
| | - James E Crowe
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN 37235, USA.
- Department of Pediatrics, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Vaccine Center, Vanderbilt University, Nashville, TN 37232, USA
| | - M Javad Aman
- Integrated Biotherapeutics Inc., Gaithersburg, MD 20878, USA.
| | - John M Dye
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA.
| | - Jonathan R Lai
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
32
|
Thi EP, Lee ACH, Geisbert JB, Ursic-Bedoya R, Agans KN, Robbins M, Deer DJ, Fenton KA, Kondratowicz AS, MacLachlan I, Geisbert TW, Mire CE. Rescue of non-human primates from advanced Sudan ebolavirus infection with lipid encapsulated siRNA. Nat Microbiol 2016; 1:16142. [PMID: 27670117 DOI: 10.1038/nmicrobiol.2016.142] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 07/14/2016] [Indexed: 02/03/2023]
Abstract
Although significant progress has been made in developing therapeutics against Zaire ebolavirus, these therapies do not protect against other Ebola species such as Sudan ebolavirus (SUDV). Here, we describe an RNA interference therapeutic comprising siRNA targeting the SUDV VP35 gene encapsulated in lipid nanoparticle (LNP) technology with increased potency beyond formulations used in TKM-Ebola clinical trials. Twenty-five rhesus monkeys were challenged with a lethal dose of SUDV. Twenty animals received siRNA-LNP beginning at 1, 2, 3, 4 or 5 days post-challenge. VP35-targeting siRNA-LNP treatment resulted in up to 100% survival, even when initiated when fever, viraemia and disease signs were evident. Treatment effectively controlled viral replication, mediating up to 4 log10 reductions after dosing. Mirroring clinical findings, a correlation between high viral loads and fatal outcome was observed, emphasizing the importance of stratifying efficacy according to viral load. In summary, strong survival benefit and rapid control of SUDV replication by VP35-targeting LNP confirm its therapeutic potential in combatting this lethal disease.
Collapse
Affiliation(s)
- Emily P Thi
- Arbutus Biopharma Corporation, Burnaby, British Columbia V5J 5J8, Canada
| | - Amy C H Lee
- Arbutus Biopharma Corporation, Burnaby, British Columbia V5J 5J8, Canada
| | - Joan B Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas 77550, USA.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Raul Ursic-Bedoya
- Arbutus Biopharma Corporation, Burnaby, British Columbia V5J 5J8, Canada
| | - Krystle N Agans
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas 77550, USA.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Marjorie Robbins
- Arbutus Biopharma Corporation, Burnaby, British Columbia V5J 5J8, Canada
| | - Daniel J Deer
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas 77550, USA.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Karla A Fenton
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas 77550, USA.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | | | - Ian MacLachlan
- Arbutus Biopharma Corporation, Burnaby, British Columbia V5J 5J8, Canada
| | - Thomas W Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas 77550, USA.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Chad E Mire
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas 77550, USA.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555, USA
| |
Collapse
|
33
|
Howell KA, Qiu X, Brannan JM, Bryan C, Davidson E, Holtsberg FW, Wec AZ, Shulenin S, Biggins JE, Douglas R, Enterlein SG, Turner HL, Pallesen J, Murin CD, He S, Kroeker A, Vu H, Herbert AS, Fusco ML, Nyakatura EK, Lai JR, Keck ZY, Foung SKH, Saphire EO, Zeitlin L, Ward AB, Chandran K, Doranz BJ, Kobinger GP, Dye JM, Aman MJ. Antibody Treatment of Ebola and Sudan Virus Infection via a Uniquely Exposed Epitope within the Glycoprotein Receptor-Binding Site. Cell Rep 2016; 15:1514-1526. [PMID: 27160900 PMCID: PMC4871745 DOI: 10.1016/j.celrep.2016.04.026] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/07/2016] [Accepted: 04/03/2016] [Indexed: 12/02/2022] Open
Abstract
Previous efforts to identify cross-neutralizing antibodies to the receptor-binding site (RBS) of ebolavirus glycoproteins have been unsuccessful, largely because the RBS is occluded on the viral surface. We report a monoclonal antibody (FVM04) that targets a uniquely exposed epitope within the RBS; cross-neutralizes Ebola (EBOV), Sudan (SUDV), and, to a lesser extent, Bundibugyo viruses; and shows protection against EBOV and SUDV in mice and guinea pigs. The antibody cocktail ZMapp™ is remarkably effective against EBOV (Zaire) but does not cross-neutralize other ebolaviruses. By replacing one of the ZMapp™ components with FVM04, we retained the anti-EBOV efficacy while extending the breadth of protection to SUDV, thereby generating a cross-protective antibody cocktail. In addition, we report several mutations at the base of the ebolavirus glycoprotein that enhance the binding of FVM04 and other cross-reactive antibodies. These findings have important implications for pan-ebolavirus vaccine development and defining broadly protective antibody cocktails.
Collapse
Affiliation(s)
- Katie A Howell
- Integrated BioTherapeutics, Inc., Gaithersburg, MD 20878, USA
| | - Xiangguo Qiu
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; Deparment of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Jennifer M Brannan
- U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA
| | | | | | | | - Anna Z Wec
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Sergey Shulenin
- Integrated BioTherapeutics, Inc., Gaithersburg, MD 20878, USA
| | - Julia E Biggins
- Integrated BioTherapeutics, Inc., Gaithersburg, MD 20878, USA
| | - Robin Douglas
- Integrated BioTherapeutics, Inc., Gaithersburg, MD 20878, USA
| | | | - Hannah L Turner
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jesper Pallesen
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Charles D Murin
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Shihua He
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; Deparment of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Andrea Kroeker
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; Deparment of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Hong Vu
- Integrated BioTherapeutics, Inc., Gaithersburg, MD 20878, USA
| | - Andrew S Herbert
- U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA
| | - Marnie L Fusco
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Elisabeth K Nyakatura
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jonathan R Lai
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Zhen-Yong Keck
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Steven K H Foung
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Erica Ollmann Saphire
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA; Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | - Gary P Kobinger
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; Deparment of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - John M Dye
- U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA
| | - M Javad Aman
- Integrated BioTherapeutics, Inc., Gaithersburg, MD 20878, USA.
| |
Collapse
|
34
|
Human Vaccines & Immunotherapeutics: News. Hum Vaccin Immunother 2016; 12:558-9. [DOI: 10.1080/21645515.2016.1160701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|