Skip to main content
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems
  • Log in
  • My alerts
  • My Cart

Main menu

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • COVID-19 Special Collection
    • Minireviews
    • JVI Classic Spotlights
    • Archive
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About JVI
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems

User menu

  • Log in
  • My alerts
  • My Cart

Search

  • Advanced search
Journal of Virology
publisher-logosite-logo

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • COVID-19 Special Collection
    • Minireviews
    • JVI Classic Spotlights
    • Archive
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About JVI
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
Vaccines and Antiviral Agents

Novel Cross-Reactive Monoclonal Antibodies against Ebolavirus Glycoproteins Show Protection in a Murine Challenge Model

James Duehr, Teddy John Wohlbold, Lisa Oestereich, Veronika Chromikova, Fatima Amanat, Madhusudan Rajendran, Sergio Gomez-Medina, Ignacio Mena, Benjamin R. tenOever, Adolfo García-Sastre, Christopher F. Basler, Cesar Munoz-Fontela, Florian Krammer
Douglas S. Lyles, Editor
James Duehr
aDepartment of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
bGraduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Teddy John Wohlbold
aDepartment of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
bGraduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lisa Oestereich
cDepartment of Virology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Veronika Chromikova
aDepartment of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Fatima Amanat
aDepartment of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Madhusudan Rajendran
aDepartment of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Madhusudan Rajendran
Sergio Gomez-Medina
dHeinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ignacio Mena
aDepartment of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
eGlobal Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Benjamin R. tenOever
aDepartment of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Adolfo García-Sastre
aDepartment of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
eGlobal Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
fDepartment of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Christopher F. Basler
gCenter for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Cesar Munoz-Fontela
cDepartment of Virology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
dHeinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Cesar Munoz-Fontela
Florian Krammer
aDepartment of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Florian Krammer
Douglas S. Lyles
Wake Forest University
Roles: Editor
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1128/JVI.00652-17
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

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.

INTRODUCTION

The 2013-2016 outbreak of ebolavirus (species Zaire ebolavirus) (EBOV) in Guinea, Liberia, and Sierra Leone had an undeniable worldwide effect. Its impact is measured not only in terms of human lives lost, but also in food scarcity and economic instability (1–3). The virus responsible emerged out of the Congolese forest and into the public eye in 1976 and has since caused ∼13,000 confirmed deaths (4, 5). The vast majority of these deaths (∼11,000) occurred during the 2013-2016 outbreak (6). Prior to this outbreak, only one other case of filoviral disease in West Africa had ever been recorded: a single nonfatal case of Taï Forest virus viremia in 1994 (7). This relative scarcity followed by an outright flood of cases in 2014 emphasizes the erratic nature of filovirus outbreaks and underscores the need for an effective pan-Filoviridae therapeutic.

Many potential prophylactics and therapeutics have been described in the literature (8–10). Of these, only the vesicular stomatitis virus expressing the Zaire ebolavirus glycoprotein (VSV-EBOV) vaccine has entered phase III clinical trials (11), and most other approaches are also focused on immune responses against the Zaire ebolavirus membrane glycoprotein (GP). The three monoclonal antibodies (MAbs) present in the ZMapp MAb cocktail (c13C6, c2G4, and c4G7) have been described as protective against EBOV challenge in macaques, but not all three are neutralizing (12). The recent EBOV outbreak in West Arica has spurred the development of additional anti-GP MAbs isolated from humans (Ebola Virus Disease [EVD] survivors) and animals, some of which have been developed further into novel therapeutics (13–17). They include novel approaches that combine different MAbs into bispecific molecules using antibody engineering (18, 19).

While EBOV has the highest case fatality rate of any member of the ebolavirus genus, it is not the only member of the genus with epidemic potential (20). Four out of five species in the genus are known to cause EVD in humans, with Sudan and Bundibugyo viruses having case fatality rates of approximately 53% and 32%, respectively (5). It is known that effective humoral immune responses during acute EVD may be important for patient survival in humans (21). The precise mechanism of this protective effect remains to be elucidated, but both virus neutralization and effector functions seem to be involved. To shed more light on these interactions and to investigate pan-filovirus therapeutic targets, we generated a panel of MAbs against the EBOV GP and characterized their binding breadth, neutralizing activity, effector functions, and protective efficacy in a novel lethal challenge model of VSV-EBOV infection in Stat2−/− mice. Two of the generated MAbs that displayed pan-ebolavirus binding activity—but were nonneutralizing—showed protection in the mouse model (KL-2E5 and KL-2H7). Another MAb, KL-2G12, which demonstrated specific binding to and neutralization of VSV-EBOV, also provided prophylactic and therapeutic protection in the mouse model.

RESULTS

Hybdridoma generation and screening.To generate hybridomas, a single female BALB/c mouse was administered two DNA vaccinations with EBOV GP expression plasmids followed by one immunization with VSV-EBOV. The rationale behind this vaccination regimen was based on one used for influenza virus hemagglutinin, where a DNA prime followed by a virus-vectored boost leads to the induction of broadly protective antibody responses (22). The mice were then boosted for the hybridoma fusion with virus-like particles (VLPs) derived from 293T cells transfected with plasmids expressing EBOV GP and viral protein 40 (VP40) (23). To screen the resulting hybridoma clones, Vero.E6 cells infected with a Newcastle disease virus (NDV) expressing the EBOV GP derived from a 2014 EBOV strain and 293T cells transfected with an EBOV GP mammalian expression plasmid were immunostained with individual hybridoma supernatants. This method ensures that only clones capable of binding the GP as it is expressed on the surfaces of mammalian cell membranes in the context of a viral infection are selected. Of 29 positive clones, 8 were IgG and 11 were IgM. The eight positive IgG hybridomas were expanded, and antibodies were purified from culture supernatants for further characterization.

Characterization of the MAb binding profile.To further characterize binding properties and elucidate the interaction of GP and MAbs, we expressed recombinant GPs from all member species of the genus Ebolavirus in insect cells. They included GPs from Zaire (EBOV), Reston (RESTV), Sudan (SUDV), Bundibugyo (BDBV), and Taï Forest (TAFV) ebolaviruses. We also generated the Marburg virus (MARV) glycoprotein as a suitable phylogenetic outgroup and the glycoprotein from the specific Guinea variant (Makona) of EBOV (EBOVG) that was the etiological agent responsible for the most recent outbreak in West Africa (24). To determine the breadth of reactivity of our antibodies, we assessed binding to recombinant GPs using enzyme-linked immunosorbent assays (ELISAs) (Fig. 1). All the MAbs bound to EBOV GP (Fig. 1A), albeit at different levels. When we analyzed cross-reactivity to other filovirus GPs, two MAbs (KL-2E5 and KL-2H7) displayed remarkable cross-reactivity against all five member species of the genus Ebolavirus (Fig. 1B). As was expected (due to the large genetic distance between EBOV and MARV), no cross-reactive clones bound MARV GP. Interestingly, the MAb that later presented the greatest neutralizing activity in vitro (KL-2G12) (Fig. 1B and 2A) had narrow breadth, binding only to the EBOV and EBOVG GPs. All the MAbs bound to cells transfected with a plasmid expressing EBOV GP, again verifying that they recognize GP expressed on the cell surface (Fig. 1C). Since the version of the GP present on VSV-EBOV might have a slightly different conformation than the GP found on the wild-type virus, we also evaluated binding to cells infected with wild-type EBOV in a biosafety level 4 (BSL4) laboratory. Most of our MAbs bound in both contexts (Fig. 1D). Finally, to determine if the MAbs could bind linear epitopes, we performed a reducing sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) under denaturing conditions, followed by Western blotting using our generated MAbs as primary antibodies. Only MAb KL-2G12 did not recognize EBOV GP in this assay, suggesting the antibody binds a more complex conformational epitope (Fig. 1E).

FIG 1
  • Open in new tab
  • Download powerpoint
FIG 1

EBOV MAbs display cross-reactivity in vitro. (A) ELISAs representative of assays performed to create the heat map shown in panel B. The ELISA shown was performed against recombinant Zaire ebolavirus glycoprotein. All assays were performed in technical duplicates with a glycoprotein coating concentration of 2 μg/ml. Error bars (depicting standard errors of the mean) smaller than the symbol were omitted for visualization. (B) Heat map of MAb reactivity against insect cell-generated recombinant glycoproteins. The scale bar represents 1% difference in amino acid sequence. (C) Immunofluorescence staining of 293T cells transfected with EBOV-GP pCAGGS plasmids and stained with eight EBOV-reactive MAbs at a concentration of 30 μg/ml. The positive-control antibody was a murine monoclonal previously raised against recombinant EBOV GP. (D) Confocal microscopy of Vero.E6 cells infected with wild-type EBOV (Mayinga variant) and fixed at 72 h postinfection. The cells were then stained with 10 μg/ml of the indicated MAbs, followed by secondary staining with the indicated fluorescent MAbs. Blue staining indicates the cell nucleus (DAPI), and red staining indicates the cell membrane. Anti-GP MAbs show slightly different staining patterns that might suggest that different forms of GP are recognized. Serum reactive to the nucleoprotein was used as a positive control, resulting in a punctate staining pattern. (E) Western blotting was performed with 300 ng of EBOV GP (lane 1) and H7 influenza virus HA (lane 2) (negative control; both proteins were hexahistidine tagged). Each membrane was stained separately with one of eight MAbs, followed by secondary staining with anti-mouse IgG coupled to HRP. A control membrane was stained with anti-His murine IgG.

Neutralization of VSV-EBOV in vitro.To further characterize these MAbs, we performed neutralization assays against VSV-EBOV. As others have shown, neutralization of VSV-EBOV in vitro is one of the most predictive factors of protective efficacy with MAb therapeutics against live Ebolavirus challenge in primate models (12, 25). Reactivity against the GP alone is necessary but might not be sufficient (26). We also wanted to assess if the presence or absence of the mucin-like domain (MLD) impacted MAb binding. Therefore, we assessed the neutralizing capacity of our MAbs using plaque reduction neutralization assays (PRNAs) against both the canonical VSV-EBOV and a companion replication-competent pseudotyped virus bearing a GP lacking the MLD but still capable of viral entry and multicycle growth in Vero cells (VSV-EBOVΔmuc) (Fig. 2). Interestingly, the MAbs that had the most restricted breadth (KL-1F11 and KL-2G12) were also those that possessed the most neutralizing activity (Fig. 2A and B). Consistent results were obtained using a neutralization assay based on a more sensitive fluorescent readout (Fig. 2C). As with binding, we also wanted to verify that neutralizing capacity extended to live ebolavirus (Mayinga variant) infection in an in vitro BSL4 environment. Surprisingly, this was not the case (data not shown). Antibodies (KL-2G12 and KL-1F11) that neutralize and protect against VSV-EBOV—but are nonneutralizing against wild-type EBOV—have yet to be described in the literature.

FIG 2
  • Open in new tab
  • Download powerpoint
FIG 2

Neutralization assays and escape mutants. (A) PRNAs performed against VSV-EBOV with an intact mucin-like domain. Percent inhibition was calculated as a decrease in plaque number in comparison to the average of the isotype control and blank wells. All assays were performed in duplicate. (B) PRNAs performed as for panel A, except with a VSV-EBOV lacking the mucin-like domain. (C) Fluorescence-based neutralization assay, with eGFP mean intensity as a readout. (D) Escape mutations visualized on a crystal structure of the GP trimer (45) (3INU). A side view is shown; the membrane is below. The arrow indicates the location of the KL-2G12 escape mutation. (E) Top view. (F) Bottom view. (G) Schematic of the mature GP with potential escape mutations from KL-2G12 and KL-1F11 noted. SP, signal peptide; RBD, receptor binding domain. (H) KL-2G12. (I) KL-1F11. PRNAs were performed on the escape mutant viruses using their respective MAbs and an isotype control. The error bars indicate standard deviations.

To characterize the binding epitope of our most neutralizing antibodies, we generated escape mutants of VSV-EBOV. First, we passaged VSV-EBOV in the presence of double the 50% inhibitory concentration (2× IC50) of either KL-2G12 or KL-1F11, the most potent neutralizing MAbs in our panel of eight. Then, after 72 h of incubation, we passaged the viral population in the presence of double the antibody concentration. After six such passages, we plaque purified isolates of each escape mutant and evaluated for mutations using deep sequencing (Fig. 2D to G). To verify that our escape mutants had truly “escaped” MAb binding, PRNAs were performed on the two escape viruses, showing no neutralization from either MAb (Fig. 2H and I). The escape mutant generated from passaging in the presence of KL-1F11 had nonsynonymous coding variants located on the MLD, GP2, and glycan cap. Since KL-1F11 appears to neutralize VSV-EBOVΔmuc as well as VSV-EBOV containing an intact MLD (Fig. 2B), there is no reason to believe KL-1F11's neutralization capacity requires an intact mucin-like domain. The VSV-EBOV escape mutant raised in the presence of KL-2G12 displayed a single variant located on the glycan cap near the receptor binding pocket.

Fc receptor engagement and cellular cytotoxicity/phagocytosis activity.Since effector functions are becoming an important topic for antiviral immunity in the influenza virus and HIV fields, we were also interested in investigating the ability of our antibodies to engage Fc receptors in vitro. Our MAb panel represents a diversity of IgG isotypes—IgG1, IgG2a, and IgG2b—that have noted differences in their abilities to activate effector functions (27). To investigate each MAb's ability to utilize these Fc-FcγR interactions, an antibody-dependent cellular phagocytosis (ADCP) assay and an antibody-dependent cell-mediated cytotoxicity (ADCC) reporter assay were established in the laboratory (Fig. 3). Our most neutralizing antibodies (KL-2G12 and KL-1F11) scored highest in the ADCC reporter assay, while KL-2G12 and KL-1F8 scored highest in the ADCP assay. The cross-reactive, nonneutralizing MAbs KL-2E5 and KL-2H7 showed little effect in either assay. Of note, KL-2E5 and KL-2H7 are murine IgG1 antibodies, which are known to have poor FcγR engagement (28).

FIG 3
  • Open in new tab
  • Download powerpoint
FIG 3

Several anti-GP MAbs exhibit effector functions in in vitro assays. (A) ADCC reporter assay. Threefold serial dilutions of anti-EBOV MAbs starting at 30 μg/ml. Values are depicted as fold induction over the negative-control MAb. Antibody isotypes are indicated. (B) ADCP assay. Fourfold serial dilutions of anti-EBOV MAbs starting at 10 μg/ml incubated with EBOV GP bound to neutravidin beads. The phagocytic score was calculated as the mean fluorescence intensity multiplied by the percentage of positive THP-1 cells as measured by flow cytometry. The error bars indicate standard deviations.

Neutralizing and nonneutralizing antibodies confer protection in a lethal model of VSV-EBOV infection in Stat2−/− mice.Since the goal was to determine which of our MAbs have the most potential as therapeutic agents, we evaluated their protective efficacies in an in vivo challenge model. In order to do so in the most efficient manner, we were interested in establishing an animal model of infection that could be used outside a BSL4 laboratory. We first examined the possibility of using immunodeficient mice infected with VSV-EBOV and found specifically that Stat2−/− mice readily succumb to infection when inoculated with this replication-competent pseudotyped virus. As is shown in Fig. 4, Stat2−/− mice can be infected with VSV-EBOV, resulting in a 50% lethal dose (LD50) of approximately 56 PFU. Notably, weight loss proved to be an ineffective predictor of mortality, with many mice succumbing to infection well above the humane endpoint of 25% weight loss. In pilot experiments, infectious VSV-EBOV was found in several organs of these mice, including the brain (Fig. 4C).

FIG 4
  • Open in new tab
  • Download powerpoint
FIG 4

VSV-EBOV causes lethal infections in Stat2−/− mice. (A) Groups of Stat2−/− mice (6 to 12 mice per group) were administered 104, 103, 102, 101, or 100 PFU of VSV-EBOV in 100 μl of PBS i.p. in the right flank. Weight was monitored daily, and mice that lost 25% or more of their initial weight were euthanized and scored as dead. The LD50 was calculated to be 56 PFU. (B) Survival data from the experiment in panel A. The error bars indicate standard deviations. (C) Titers of infectious virus found in different organs 48 and 96 h postinfection.

With this in mind, we next tested the protective efficacy of our MAbs in this model by performing preexposure prophylaxis experiments. Mice were first intraperitoneally (i.p.) given one of our 8 MAbs or an isotype control antibody at a dose of 10 mg/kg of body weight and then infected i.p. with VSV-EBOV 3 h later and evaluated for mortality daily (Fig. 5A). As might be expected, the most potent neutralizer (KL-2G12) was also the antibody with the most protective capacity. Unexpectedly, we also observed protective efficacy with the two nonneutralizing cross-reactive antibodies (KL-2E5 and KL-2H7; 80% and 40% survival, respectively) and observed limited protection from a nonneutralizing, highly specific MAb (KL-1F8; 40% survival).

FIG 5
  • Open in new tab
  • Download powerpoint
FIG 5

Several MAbs mediate protection in a lethal VSV-EBOV challenge model in Stat2−/− mice. (A) Survival data from MAb challenge experiments. Groups of five mice were administered 10 mg/kg (100 μl) of one of the eight MAbs i.p. into the left flank. Three hours later, each mouse was inoculated with 250 PFU (∼5 LD50) of VSV-EBOV in 100 μl of PBS i.p. The data shown are cumulative data from several experiments. The group size for the MAb-treated groups in each separate experiment was five mice per MAb, but in each experiment, negative controls (n = 3) were included. The cumulative data for the negative control therefore represent 18 mice. (B) KL-2G12 postexposure (p.e.) therapeutic treatment. Groups of four mice were infected with 250 PFU (∼5 LD50) of VSV-EBOV in 100 μl of PBS i.p. Either 24 or 48 h later, the mice were given an injection of 10 mg/kg KL-2G12 or a control IgG (CTRL IgG). For both experiments, weight was monitored daily, and mice that lost 25% or more of their initial weight were euthanized. **, P < 0.01.

We were also interested in evaluating the therapeutic efficacy of our most protective prophylactic MAb (KL-2G12). To this end, 6- to 8-week-old mice were infected i.p. with 250 PFU VSV-EBOV and then split into three groups (Fig. 5B). Group 1 was administered 10 mg/kg of KL-2G12 24 h postinfection. Group 2 received the same 48 h postinfection. As a control, the third group received 10 mg/kg of an isotype control antibody 24 h postinfection. As was expected, the mortality of the mice was dependent on the timing of MAb administration: all the mice given the therapeutic, highly neutralizing MAb 24 h postinfection survived, while those given the same MAb 48 h postinfection survived at a reduced rate (50%) and those receiving the control MAb survived infection at a much lower rate (25%).

DISCUSSION

Despite the threat that ebolaviruses pose to public health, the economy, and global stability, no vaccines or therapeutics have been approved for their treatment or prevention. Moreover, the approaches that are farthest advanced in the pipeline exclusively target the Zaire ebolavirus. Despite this focus, EBOV accounted for only 58% of EVD cases prior to the 2014 outbreak (20). Thus, the benefit of broader therapeutic approaches is clear. Here, we report the development and characterization of monoclonal antibodies raised against the Zaire ebolavirus glycoprotein that display cross-reactivity, VSV-EBOV neutralization, and protection in a mouse model. These eight MAbs encompass a broad swath of anti-EBOV antibody characteristics previously reported, and some unreported. Two of the isolated MAbs display a wide breadth of binding (KL-2E5 and KL-2H7) and cross-react against all five species in the ebolavirus genus. Another MAb, KL-2G12, binds and potently neutralizes VSV-EBOV but lacks neutralization against the wild-type virus (Mayinga variant), suggesting conformational differences between the glycoproteins expressed on the two viruses. As shown here, MAbs that bind but do not neutralize wild-type virus may still be protective in animal models. In fact, a nonneutralizing antibody was included in the ZMapp cocktail because it improved protective efficacy (12, 26). Of note, KL-2E5 and KL-2H7, which are partially protective against VSV-EBOV in Stat2−/− mice and cross-reactive in vitro, are nonneutralizing and exhibited low Fc receptor activation in the assays tested. The mechanisms of protection of these MAbs remain to be elucidated. Overall, two out of eight IgG MAbs purified were cross-reactive, an unusually high number with homologous vaccine regimens. It is possible that the prime-boost regimen used is the causative factor, especially since recent findings in the influenza virus field have indicated that heterologous prime-boost regimens (e.g., DNA/live attenuated virus prime followed by an inactivated virus vaccine boost) induce broader reactivity than prime alone or homologous prime-boost regimens (29–32). Despite unanswered questions, the findings reported here may have an important impact on the design of vaccines and therapeutics against filoviral disease.

MATERIALS AND METHODS

Cells and viruses.293T and Vero.E6 cells were obtained from the American Type Culture Collection (ATCC) and were maintained in Dulbecco's modified Eagle's medium (DMEM) (Gibco) supplemented with 10% fetal bovine serum (FBS) (Clontech), 100 U/ml of penicillin, and 100 μg/ml of streptomycin (antibiotics mixture). THP-1 cells were obtained from the ATCC and maintained in Roswell Park Memorial Institute (RPMI) medium supplemented with 10% FBS (Clontech). Sf9 insect cells (ATCC CRL-1711) were propagated in TNM-FH medium (Gemini Bio-Products) supplemented with 10% FBS and antibiotics mixture. High Five cells (BTI-TN-5B1-4 subclone; Vienna Institute of Biotechnology) were grown in serum-free SFX medium (HyClone) supplemented with antibiotics mixture (33).

VSV-EBOV was grown in Vero.E6 cells, and titers were determined by plaquing virus on Vero.E6 cell monolayers and staining with polyclonal anti-VSV mouse serum. VSV-EBOV and VSV-EBOVΔmuc are both replication-competent pseudotyped viruses derived from the VSV-EBOV-XN2 rescue system and featuring enhanced green fluorescent protein (eGFP) open reading frames (ORFs) in the first cloning site directly preceding NP (34, 35).

A recombinant Newcastle disease virus (rNDV-EBOV-GP) expressing the ectodomain of the EBOV glycoprotein (strain wt/GIN/2014/Makona-Gueckedou-C07; GenBank accession number KJ660347 ) was prepared as previously described (36). Briefly, a synthetic, codon-optimized sequence encoding the ectodomain of the EBOV-GP was fused in frame to the sequence coding the transmembrane and cytoplasmic tail of the NDV F protein and cloned as an additional transcript unit in a plasmid containing the full-length cDNA of the NDV vaccine strain LaSota. The recombinant virus was rescued by transfection following a previously described protocol (37) and amplified in specific-pathogen-free (SPF) embryonated chicken eggs (Charles River Laboratories).

All experiments with infectious EBOV were carried out in the biosafety level 4 laboratory of the Bernhard Nocht Institute for Tropical Medicine in Hamburg, Germany, according to institutional biosafety regulations. All personnel performing infections wore positive-pressure biosafety suits.

Production of ebolavirus VLPs.To produce ebolavirus VLPs, 293T cells in multiple 175-cm2 flasks were transfected with EBOV GP in a pCAGGS vector and VP40 in a pCAGGS vector using a ratio of 2:3. After incubating at 37°C for 48 h, the cell supernatant was harvested and clarified via low-speed centrifugation (3,000 × g for 30 min at 4°C). VLPs were pelleted through a 30% sucrose cushion (30% sucrose in NTE buffer [100 mM NaCl, 10 mM Tris-HCl, 1 mM EDTA], pH 7.4) by ultracentrifugation (Beckman L7-65 ultracentrifuge with an SW-28 rotor at 25,000 rpm for 2 h). Once all of the supernatant was aspirated, the VLP pellets were resuspended in phosphate-buffered saline (PBS) (pH 7.4). Presence of the ebolavirus GP in the final VLP preparation was confirmed via Western blotting by probing with murine positive-control MAb (supplied by the Basler Laboratory).

Recombinant proteins.Coding sequences of glycoprotein ectodomains (ending at D632 in EBOV, upstream of the ADAM17/TACE cleavage site at D637) from EBOV, an isolate of EBOVG, BDBV, SUDV, TAFV, RESTV, and MARV were amplified via PCR (primer sequences are available upon request) and inserted into a modified pFastBacDual vector in frame with a carboxy-terminal trimerization domain and a hexahistidine tag using an In-Fusion cloning kit (Clontech). The constructs obtained were Sanger sequenced and transformed into DH10Bac competent cells (ThermoFisher Scientific) to produce recombinant bacmids, which were then transfected into Sf9 cells as described previously (38, 39). Recombinant baculoviruses were propagated in Sf9 cells. Proteins were expressed in High Five cells and purified from culture supernatants following a detailed published protocol (38).

Hybridoma generation.Female BALB/c mice (6 to 8 weeks old; Jackson Laboratory) were initially vaccinated by intramuscular administration of plasmid DNA, followed immediately by the application of electrical stimulation (TriGrid delivery system; Ichor Medical Systems) (40, 41). The spacing of the TriGrid electrode array is 2.5 mm, and the electrical field is applied at an amplitude of 250 V/cm of electrode spacing for six pulses for a total duration of 40 ms applied over a 400-ms interval. DNA vaccination comprised 40 μg of pCAGGS EBOV GP. Three weeks later, a DNA boost was performed following the same procedure. Three weeks after that, the mice were given 104 PFU VSV-EBOV i.p. Approximately 3 weeks after this infection, one mouse was boosted with a unilateral intraperitoneal injection of EBOV VLPs containing EBOV GP and VP40 (100 μg total protein content) adjuvanted with 10 μg poly(I·C). Three days postboost, the mouse was sacrificed, and its spleen was sterilely removed. The spleen was flushed forcefully with serum-free DMEM (with antibiotic mixture) using a 10-ml syringe with a 20-gauge needle, followed by repeated pulverization with flat-ended forceps. Splenocytes and SP2/0 myeloma cells (in exponential growth phase) were combined in a 5:1 ratio, and cell fusion was mediated via slow, dropwise addition of 1 ml of polyethylene glycol (mass, ∼4,000 Da). The splenocyte-SP2 mixture was then resuspended in 25 ml of complete DMEM (supplemented with antibiotic mixture, FBS, and HEPES) and left to incubate for 24 h at 37°C. After this incubation, the cells were spun down, resuspended in 10 ml of complete DMEM, mixed with 90 ml of proprietary semisolid Clonacell-HY Medium D (Stemcell Technologies), and dispensed onto tissue culture dishes (10 ml each) using a 10-ml syringe with a 15-gauge Luer Stub adapter (Becton Dickinson). Individual colonies were picked 10 days later and transferred into 96-well plates containing Clonacell-HY Medium E. Five days after transfer to 96-well plates, hybridoma supernatants were individually screened by immunostaining Vero.E6 cells infected with NDV-EBOV-GP at a multiplicity of infection (MOI) of 1 and transfected 293T cells expressing EBOV GP. Positive clones were isotyped using a Pierce rapid antibody isotyping kit (Life Technologies); only the MAbs isotyped to the IgG heavy-chain subclasses were selected for further expansion and purification.

Antibody purification.Hybridoma cultures were initially expanded in Clonacell-HY Medium E but gradually switched to serum-free hybridoma medium until a final volume of 300 to 500 ml was achieved. When the cells appeared no longer viable (10 days after the final expansion step), the cultures were harvested by low-speed centrifugation (30 min; 5,500 relative centrifugal force [RCF]), and the supernatants were passed through 0.22-μm-pore-size sterile filtration units (Millipore). The filtered supernatants were passed through a gravity flow column packed with protein G-Sepharose 4 Fast Flow beads (GE Healthcare). After washing with 3 column volumes (450 ml) of sterile PBS (pH 7.4), antibodies were eluted with 45 ml of 0.1 M glycine-HCl buffer (pH 2.7), and the eluate was immediately neutralized with 5 ml of 2 M Tris-HCl buffer (pH 10). The MAbs were further concentrated and buffer exchanged against PBS (pH 7.4) using Amicon Ultra centrifugal filter units (30-kDa cutoff; Millipore). The final protein concentration was determined using a NanoDrop device (Thermo Scientific) and measuring absorbance at 280 nm.

Challenge studies in Stat2−/− mice. Stat2 −/− C57BL/6 mice were kindly provided by Christian Schindler and were bred at the Icahn School of Medicine at Mount Sinai. The LD50 of the VSV-EBOV pseudotyped virus was determined by injecting 6- to 8-week-old Stat2−/− mice with 1,000, 100, 10, or 1 PFU of PBS-diluted virus i.p. (left flank) and monitoring daily for survival and weight loss until 14 days postinfection (p.i.). Mice that lost 25% or more of their initial body weight were euthanized and scored dead. The LD50 was calculated using the Reed and Muench method.

To study in which organs the virus would replicate, two groups of two mice each were inoculated with 250 PFU of VSV-EBOV (i.p.; left flank) and sacrificed either 48 or 96 h postinfection for organ titer analysis. At these time points, the brain, kidneys, intestine, spleen, and liver were harvested and homogenized using a BeadBlaster 24 (Benchmark) homogenizer. Viral titers were then measured by incubating the homogenate in serial 10-fold dilutions (50-μl volume) on confluent Vero cells in triplicate. Live virus stock from cell culture was used as a positive control; 72 h after serial dilutions of homogenate were added, the cells were assessed for fluorescence to determine the 50% tissue culture infective dose (TCID50).

In prophylactic antibody protection studies, groups of two male and three female Stat2−/− mice aged 6 to 8 weeks were given a 10-mg/kg dose of purified monoclonal antibody i.p. (right flank). Negative-control mice received a 10-mg/kg dose of MAb 8H9, which is specific for influenza virus H6 hemagglutinin. At 3 h posttreatment, 5 times the LD50 of VSV-EBOV-EGFP (250 PFU) diluted in PBS (pH 7.4) was injected i.p. (left flank). All the mice were then monitored daily for survival and weight loss until 14 days postinfection. MAbs were tested in batches due to limited availability of the Stat2−/− mice. The group size for the MAb-treated groups in each separate experiment was five mice per MAb, but in every experiment, negative controls (n = 3) were included. The cumulative data for the negative controls therefore represent 18 mice. The data shown are cumulative, i.e., data from all experiments combined.

For therapeutic studies, groups of two male and two female Stat2−/− mice aged 6 to 8 weeks were given 5 times the LD50 of VSV-EBOV (250 PFU) diluted in PBS (pH 7.4) injected i.p. (left flank). At 24 and 48 h p.i., a 10-mg/kg dose of purified mouse monoclonal antibody, either 2G12 or control MAb (8H9), was injected in 200 μl i.p. (right flank). All mice were then monitored daily for survival and weight loss until 14 days postinfection. All procedures were performed in accordance with the Icahn School of Medicine at Mount Sinai Institutional Animal Care and Use Committee (IACUC) guidelines.

Generation of MAb escape mutant viruses.MAb escape mutant variants of VSV-EBOV were generated via serial passaging in Vero cells in the presence of increasing amounts of MAb, with a starting concentration of 2× IC50 (as calculated from the neutralization assay against VSV-EBOV). Initially, Vero cells in 12-well tissue culture plates (Sigma) were infected with VSV-EBOV at a multiplicity of infection (MOI) of 1 in the presence of 2× IC50 of MAb (performed in duplicate for each MAb) in MEM. After incubating for 72 h at 37°C, 100 μl of supernatant was collected and used to directly inoculate a fresh monolayer of Vero cells in the presence of a 2-fold increase in the MAb concentration. This process was repeated for six passages. Throughout serial passaging, successful infection was confirmed by the expression of eGFP. In the cases of both MAbs, eGFP was present after the last passage of virus. Virus was additionally passaged in the presence of an irrelevant mouse MAb against the influenza virus hemagglutinin (8H9; IgG) to control for mutational variants obtained from passaging alone. Escape mutant viruses were plaque purified once serial passaging was completed to create monoclonal stocks. These monoclonal stocks were then tested in plaque reduction neutralization assays, as described previously, against their corresponding escape MAbs, and no residual neutralization activity was detected.

Plaque reduction neutralization assays.PRNAs were performed according to a protocol established by Tan et al. (42) with modifications. Briefly, MAbs at 1:5 dilutions starting from 100 μg/ml were incubated with 70 PFU of either VSV-EBOV, VSV-EBOVΔmuc, or one of two escape mutant viruses derived from VSV-EBOV for 1 h at room temperature (RT). These dilutions were then plaqued on 90 to 100% confluent Vero.E6 cell monolayers in 12-well plates. Cells were overlaid with minimum essential medium (MEM) containing 0.64% agarose (Oxoid) and the corresponding MAb dilutions. After 4 days of incubation at 37°C, the cells were fixed with 3.7% paraformaldehyde (PFA), and the plaques were stained using a polyclonal anti-EBOV mouse serum (1:1,000), an anti-mouse secondary antibody conjugated to horseradish peroxidase (HRP) (Sigma), and Trueblue reagent (KPL). The plaques were counted in each MAb dilution, and the percent inhibition for each MAb and dilution was individually calculated based on a no-antibody control and an isotype control. All assays were performed in duplicate. The data were analyzed by using Prism software (GraphPad), and the concentration at which a MAb inhibited 50% of plaques (IC50) was calculated using a nonlinear 4-parameter regression.

Fluorescence-based neutralization assays.Vero.E6 cells were plated overnight at 37°C to achieve 100% confluence in sterile, flat-bottom, 96-well tissue culture plates (Sigma). Individual MAbs were serially diluted 1:2 in MEM, in duplicate, from a starting concentration of 600 μg/ml in separate 96-well, sterile, flat-bottom tissue culture plates (final volume, 75 μl/well). Extra dilutions were performed for 2G12 in a separate assay once it became clear neutralization activity did not reach 0%. VSV-EBOV was diluted to 20 times the half-maximal effective concentration (EC50) in MEM and added to the MAb dilution plates (75 μl/well). The plates were briefly tapped (for mixing) and incubated at RT for 1 h. Next, medium from the Vero cells was aspirated and replaced with 100 μl of the antibody-virus mixture. The plates were incubated at 37°C for 48 h and then read by an acumen Cellista Laser Scanning Imaging Cytometer (TPP Labtech). Readouts of the eGFP mean intensity were plotted and analyzed using Prism software (GraphPad), and the IC50 was defined as the concentration at which 50% of the mean intensity of eGFP was inhibited.

ELISAs.Immulon 4 HBX plates were coated overnight with recombinant purified glycoprotein from each ebolavirus species at 4°C at a concentration of 2 μg/ml (50 μl/well) in coating buffer (0.1 M Na2CO3-NaHCO3, pH 9.4). After removal of the coating buffer, the plates were washed three times with PBS (pH 7.4) containing 0.1% Tween 20 (TPBS). The plates were then blocked for 1 h with blocking buffer (TPBS plus 3% dry milk powder). After washing the plates three times with TPBS, purified IgG was diluted 1:3 in the plates, starting with 100 μg/ml in blocking buffer. Positive controls (serum from the vaccinated animal postboost) and negative controls (isotype control antibody; 8H9 [anti-influenza]) were used to evaluate standardization. Control serum was diluted 1:3 in the plates, with a 1:100 starting concentration in blocking buffer. After a 2-h incubation at RT, the plates were washed three times with TPBS, and secondary horseradish peroxidase-conjugated anti-mouse IgG antibody (Rockland; 610-603-002; 1:3,000) diluted in blocking buffer was added. After a 1-h incubation, the plates were washed four times with TPBS and developed with SigmaFast o-phenylenediamine dihydrochloride (OPD) (Sigma; 100 μl per well). After 10 min of development, the reaction was stopped with 3 M HCl. The optical density at 490 nm (OD490) was read on a plate reader, and background levels were analyzed in Microsoft Excel. The minimal binding concentration was calculated as the lowest dilution of antibody that rose above background (the average of blank wells on each plate plus 3 standard deviations). Minimal binding concentrations were imaged as a heat map using Interactive Tree of Life (iTOL) v3 (European Molecular Biology Laboratory) (43).

Phagocytosis assay.The phagocytosis assay was performed as previously described by Ackerman et al. (44). Briefly, duplicates of serial 4-fold dilutions of tested mouse IgG antibodies starting with 10 μg/ml were incubated for 2 h at 37°C with 1-μm fluorescent neutravidin beads (Invitrogen) conjugated with biotinylated EBOV-GP antigen (biotinylated using EZ-Link-NHS-PEG4 biotin in 1:1 molar ratio; ThermoFisher Scientific) in 96-well plate format. THP-1 cells (2 × 104 per well) were added and incubated for 12 h in an incubator at 37°C and 5% CO2. At the end of the incubation time, the plates were fixed by adding 100 μl of 3.7% PFA/well, and samples were measured using the yellow 2 laser channel of an LSRIII flow cytometer (BD Biosciences). Analysis was performed using FCS Express software (De Novo Software).

ADCC reporter assay.ADCC was measured using the ADCC Reporter Bioassay kit (Promega) according to the manufacturer's instructions. Briefly, 3 × 104 Vero cells per well were seeded in solid white 96-well plates (Corning Costar) and then 24 h later infected with VSV-EBOV at an MOI of 5. Serial 3-fold antibody dilutions in triplicates starting at 90 μg/ml were prepared in a separate 96-well U-bottom plate and transferred to the infected cells (1:3 of the total volume/well; therefore, the final starting concentration of antibodies was 30 μg/ml). After the addition of effector cells (part of the Reporter Bioassay kit; Promega), the plates were incubated at 37°C and 5% CO2 for 6 h, Bio-Glo Luciferase reagent (Promega) was added, and luminescence was measured immediately using a Synergy Hybrid Reader (BioTek).

Immunofluorescence.To test for antibody binding via immunofluorescence, 293T cells were plated overnight in sterile, flat-bottom, 96-well tissue culture plates (Sigma) and transfected with Zaire ebolavirus GP in the pCAGGS vector. After incubating for 24 h at 37°C, the cells were fixed with 3.7% formaldehyde for at least 1 h at 4°C. Next, the formaldehyde was discarded and the cell monolayer was blocked with 3% milk in PBS for at least 1 h. For the primary antibody step, the plates were incubated with either experimental anti-EBOV MAb (30 μg/ml), positive-control anti-EBOV MAb 8G9 (30 μg/ml), or no antibody as a negative control in 1% milk (100 μl/well) for 1 h at room temperature. The plates were washed 3 times with PBS and incubated with Alexa Fluor 488 goat anti-mouse secondary antibody in PBS, 1% milk (100 μl/well) for 1 h at room temperature in the dark. Finally, after washing 3 additional times, the cells were visualized via fluorescence microscopy.

To test for antibody binding to wild-type Zaire ebolavirus (Mayinga variant), Vero.E6 cells were grown on glass coverslips and infected at an MOI of 0.1 in a BSL4 laboratory. After incubating for 72 h at 37°C, the cells were washed with PBS and stained with tetramethyl rhodamine isocyanate (TRITC)-labeled cell mask (1:1,000) for 10 min. The cells were washed three times with PBS and fixed with 4% formaldehyde for 30 to 45 min at room temperature. Next, the formaldehyde was discarded, and 0.1% Triton X-100 in PBS plus 10% fetal calf serum (FCS) was added to permeabilize for 30 min. For the primary antibody step, the cells were incubated with either experimental anti-EBOV MAb (10 μg/ml) or positive-control anti-EBOV polyclonal mouse serum (which detects EBOV nucleoprotein) for 1 h at room temperature. The antibodies were diluted in PBS. The cells were then washed 3 times with PBS and incubated with FITC anti-mouse secondary and DAPI (4′,6-diamidino-2-phenylindole) (1:10.000) in PBS for 1 h at room temperature in the dark. Finally, after washing 3 additional times, the cells were mounted on glass slides with Prolong Antifade and visualized via confocal fluorescence microscopy.

Western blotting.Binding of MAbs to EBOV glycoprotein was analyzed on SDS-PAGE (10% polyacrylamide; Mini Protean TGX gels; Bio-Rad) under reducing conditions. Three hundred nanograms of either EBOV GP (lane 1) or influenza virus H7 hemagglutinin (HA) (lane 2; control) was added to the gel and electrophoresed at 200 V for 40 min (Fig. 2). After electrophoresis, each gel was transferred to a polyvinylidene difluoride (PVDF) membrane via an Owl HEP series semidry electroblotting system (Thermo Scientific), according to the manufacturer's instructions. The membranes were washed in 1× PBS and blocked with 10% nonfat milk-PBS for 2 h at room temperature. The membranes were then washed three times with 1× TPBS. One of eight anti-EBOV MAbs (10 μg/ml) or a control anti-polyhistidine antibody (Sigma-Aldrich; 1:1,000 in blocking buffer) was then incubated with the membrane for 1 h at RT and washed three times in TPBS. An anti-mouse IgG-HRP conjugate (1:3,000 in blocking buffer) was then incubated with the membrane for 1 h at RT. Subsequently, ECL substrate (Pierce) was added and chemiluminescence was detected on a Proteinsimple FluorChem E imager.

Visualization of escape mutants on three-dimensional (3D) structures and statistical analysis.Escape mutations were visualized based on a Protein Data Bank (PDB) file derived from a structure of the human MAb KZ52 bound to the Zaire ebolavirus GP trimer (3INU ) crystallized by Lee et al. (45) using the PyMOL Molecular Graphics System (version 1.8) from Schrödinger, LLC.

Data were evaluated for statistical significance using Prism v7 software (Graphpad). Survival curves were assessed using the log-rank (Mantel-Cox) test, with adjustments made for multiple comparisons using the Bonferroni correction.

ACKNOWLEDGMENTS

We thank Ariana Hirsh for excellent technical assistance with the expression of filovirus glycoproteins and Viviana Simon for letting us use her FluorChem imager.

This study was funded by Seed Funding from the Icahn School of Medicine at Mount Sinai (to F.K.) and by NIH grants U19AI109945 and U19AI109664 (to C.F.B.).

FOOTNOTES

    • Received 17 April 2017.
    • Accepted 28 May 2017.
    • Accepted manuscript posted online 7 June 2017.
  • Copyright © 2017 American Society for Microbiology.

All Rights Reserved .

REFERENCES

  1. 1.↵
    1. Thomas A-C,
    2. Nkunzimana T,
    3. Perez Hoyos A,
    4. Kayitakire F
    . 2014. Impact of the West African Ebola virus disease outbreak on food security. Joint Research Centre, European Commission, Ispra, Italy. http://reliefweb.int/sites/reliefweb.int/files/resources/JRC94257_ebola_impact_on_food_security_jrc_h04_final_report.pdf.pdf.
  2. 2.↵
    1. Evans D,
    2. Ferreira F,
    3. Panzer J
    . 2014. The economic impact of the 2014 Ebola epidemic: short and medium term estimates for West Africa. The World Bank Group. http://documents.worldbank.org/curated/en/524521468141287875/pdf/912190WP0see0a00070385314B00PUBLIC0.pdf.
  3. 3.↵
    1. Pavot V
    . 2016. Ebola virus vaccines: where do we stand?Clin Immunol173:44–49. doi:10.1016/j.clim.2016.10.016.
    OpenUrlCrossRefPubMed
  4. 4.↵
    1. Liu Bin W,
    2. Li ZX,
    3. Du Y,
    4. Cao GW
    . 2015. Ebola virus disease: from epidemiology to prophylaxis. Mil Med Res2:7. doi:10.1186/s40779-015-0035-4.
    OpenUrlCrossRef
  5. 5.↵
    1. Gebretadik FA,
    2. Seifu MF,
    3. Gelaw BK
    . 2015. Review on Ebola virus disease: its outbreak and current status. Epidemiol (Sunnyvale)5:204. doi:10.4172/2161-1165.1000204.
    OpenUrlCrossRef
  6. 6.↵
    1. Kaner J,
    2. Schaack S,
    3. Olival K,
    4. Hayman D,
    5. Paessler S,
    6. Walker D,
    7. Kuhn J,
    8. Suzuki Y,
    9. Gojobori T,
    10. Walsh P,
    11. Biek R,
    12. Real L,
    13. Vogel G,
    14. Gire S,
    15. Goba A,
    16. Andersen K,
    17. Sealfon R,
    18. Park D,
    19. Kanneh L,
    20. Jalloh S,
    21. Carroll M,
    22. Matthews D,
    23. Hiscox J,
    24. Elmore M,
    25. Pollakis G,
    26. Rambaut A,
    27. Hewson R,
    28. Simon-Loriere E,
    29. Faye O,
    30. Faye O,
    31. Koivogui L,
    32. Magassouba N,
    33. Keita S,
    34. Thiberge J-M,
    35. Lamunu M,
    36. Lutwama J,
    37. Kamugisha J,
    38. Opio A,
    39. Nambooze J,
    40. Ndayimirije N,
    41. Okware S,
    42. Borchert M,
    43. Mutyaba I,
    44. Kerkhove M,
    45. Lutwama J,
    46. Luwaga H,
    47. Bisoborwa G,
    48. Turyagaruka J,
    49. Leroy E,
    50. Kumulungui B,
    51. Pourrut X,
    52. Rouquet P,
    53. Hassanin A,
    54. Yaba P,
    55. Délicat A,
    56. Paweska J,
    57. Gonzalez J-P,
    58. Swanepoel R,
    59. Leroy E,
    60. Rouquet P,
    61. Formenty P,
    62. Souquière S,
    63. Kilbourne A,
    64. Froment J-M,
    65. Bermejo M,
    66. Raabe V,
    67. Borchert M,
    68. Rewar S,
    69. Mirdha D,
    70. Heymann D,
    71. Dowell S,
    72. Mukunu R,
    73. Ksaizek T,
    74. Khan A,
    75. Rollin P,
    76. Peters C,
    77. Bausch D,
    78. Towner J,
    79. Dowell S,
    80. Kaducu F,
    81. Lukwiya M,
    82. Sanchez A,
    83. Nichol S,
    84. Ksiazek T,
    85. Rollin P,
    86. Thorson A,
    87. Formenty P,
    88. Lofthouse C,
    89. Broutet N,
    90. Mate S,
    91. Kugelman J,
    92. Nyenswah T,
    93. Ladner J,
    94. Wiley M,
    95. Cordier-Lassalle T,
    96. Christie A,
    97. Perner A,
    98. Fowler R,
    99. Bellomo R,
    100. Roberts I,
    101. Kugelman J,
    102. Sanchez-Lockhart M,
    103. Andersen K,
    104. Gire S,
    105. Park D,
    106. Sealfon R,
    107. Lin A,
    108. Buttner S,
    109. Benjamin K,
    110. Olga D,
    111. Markus E,
    112. Tilo F,
    113. Sarah R,
    114. Jurgen E,
    115. Stephan B,
    116. Claudio R,
    117. Helmut G,
    118. Heesterbeek H,
    119. Anderson R,
    120. Andreasen V,
    121. Bansal S,
    122. Angelis D,
    123. Dye C,
    124. Eames K,
    125. Chowell G,
    126. Nishiura H,
    127. Woolhouse M,
    128. Haydon D,
    129. Antia R,
    130. Althaus C,
    131. Scarpino S,
    132. Iamarino A,
    133. Wells C,
    134. Yamin D,
    135. Ndeffo-Mbah M,
    136. Wenzel N,
    137. Fox S,
    138. Ki-moon B,
    139. Nabarro D,
    140. Baize S,
    141. Pannetier D,
    142. Oestereich L,
    143. Rieger T,
    144. Koivogui L,
    145. Magassouba N,
    146. Soropogui B,
    147. Saéz AM,
    148. Weiss S,
    149. Nowak K,
    150. Lapeyre V,
    151. Zimmermann F,
    152. Düx A,
    153. Kühl H,
    154. Takahashi S,
    155. Jessica C,
    156. Metcalf E,
    157. Ferrari M,
    158. Moss W,
    159. Truelove S,
    160. Tatem A,
    161. Grenfell B,
    162. Lessler J,
    163. Hayden E,
    164. Black B,
    165. Caluwaerts S,
    166. Achar J,
    167. Reardon S,
    168. Shultz J,
    169. Baingana F,
    170. Neria Y,
    171. Currie J,
    172. Grenfell B,
    173. Farrar J,
    174. Chang C,
    175. Ortiz K,
    176. Ansari A,
    177. Gershwin M,
    178. Chowell G,
    179. Miller M,
    180. Viboud C,
    181. Trotter C,
    182. Gay N,
    183. Edmunds WJ
    . 2014. Ebola virus disease in West Africa: the first 9 months of the epidemic and forward projections. N Engl J Med371:1481–1495. doi:10.1056/NEJMoa1411100.
    OpenUrlCrossRefPubMed
  7. 7.↵
    1. Le Guenno B,
    2. Formenty P,
    3. Formentry P,
    4. Wyers M,
    5. Gounon P,
    6. Walker F,
    7. Boesch C
    . 1995. Isolation and partial characterisation of a new strain of Ebola virus. Lancet345:1271–1274. doi:10.1016/S0140-6736(95)90925-7.
    OpenUrlCrossRefPubMedWeb of Science
  8. 8.↵
    1. Medaglini D,
    2. Harandi AM,
    3. Ottenhoff THM,
    4. Siegrist C-A
    , VSV-EbovacConsortium. 2015. Ebola vaccine R&D: filling the knowledge gaps. Sci Transl Med7:317ps24-317ps24. doi:10.1126/scitranslmed.aad3106.
    OpenUrlFREE Full Text
  9. 9.↵
    1. Schmaljohn A,
    2. Lewis GK
    . 2016. Cell-targeting antibodies in immunity to Ebola. Pathog Dis74:ftw021. doi:10.1093/femspd/ftw021.
    OpenUrlCrossRefPubMed
  10. 10.↵
    1. Nissen MD
    . 2016. Current status of Ebolavirus vaccine development. Pathology48:S52–S53.
    OpenUrl
  11. 11.↵
    1. Henao-Restrepo AM,
    2. Camacho A,
    3. Longini IM,
    4. Watson CH,
    5. Edmunds WJ,
    6. Egger M,
    7. Carroll MW,
    8. Dean NE,
    9. Diatta I,
    10. Doumbia M,
    11. Draguez B,
    12. Duraffour S,
    13. Enwere G,
    14. Grais R,
    15. Gunther S,
    16. Gsell P-S,
    17. Hossmann S,
    18. Watle SV,
    19. Kondé MK,
    20. Kéïta S,
    21. Kone S,
    22. Kuisma E,
    23. Levine MM,
    24. Mandal S,
    25. Mauget T,
    26. Norheim G,
    27. Riveros X,
    28. Soumah A,
    29. Trelle S,
    30. Vicari AS,
    31. Røttingen J-A,
    32. Kieny M-P
    . 2017. Efficacy and effectiveness of an rVSV-vectored vaccine in preventing Ebola virus disease: final results from the Guinea ring vaccination, open-label, cluster-randomised trial (Ebola Ça Suffit!). Lancet389:505–518. doi:10.1016/S0140-6736(16)32621-6.
    OpenUrlCrossRefPubMed
  12. 12.↵
    1. Qiu X,
    2. Audet J,
    3. Lv M,
    4. He S,
    5. Wong G,
    6. Wei H,
    7. Luo L,
    8. Fernando L,
    9. Kroeker A,
    10. Bovendo HF,
    11. Bello A,
    12. Li F,
    13. Ye P,
    14. Jacobs M,
    15. Ippolito G,
    16. Saphire EO,
    17. Bi S,
    18. Shen B,
    19. Gao GF,
    20. Zeitlin L,
    21. Feng J,
    22. Zhang B,
    23. Kobinger GP
    . 2016. Two-mAb cocktail protects macaques against the Makona variant of Ebola virus. Sci Transl Med8:329ra33. doi:10.1126/scitranslmed.aad9875.
    OpenUrlAbstract/FREE Full Text
  13. 13.↵
    1. Keck Z-Y,
    2. Enterlein SG,
    3. Howell KA,
    4. Vu H,
    5. Shulenin S,
    6. Warfield KL,
    7. Froude JW,
    8. Araghi N,
    9. Douglas R,
    10. Biggins J,
    11. Lear-Rooney CM,
    12. Wirchnianski AS,
    13. Lau P,
    14. Wang Y,
    15. Herbert AS,
    16. Dye JM,
    17. Glass PJ,
    18. Holtsberg FW,
    19. Foung SKH,
    20. Aman MJ
    . 2015. Macaque monoclonal antibodies targeting novel conserved epitopes within filovirus glycoprotein. J Virol90:279–291. doi:10.1128/JVI.02172-15.
    OpenUrlCrossRefPubMed
  14. 14.↵
    1. Flyak AI,
    2. Shen X,
    3. Murin CD,
    4. Turner HL,
    5. David JA,
    6. Fusco ML,
    7. Lampley R,
    8. Kose N,
    9. Ilinykh PA,
    10. Kuzmina N,
    11. Branchizio A,
    12. King H,
    13. Brown L,
    14. Bryan C,
    15. Davidson E,
    16. Doranz BJ,
    17. Slaughter JC,
    18. Sapparapu G,
    19. Klages C,
    20. Ksiazek TG,
    21. Saphire EO,
    22. Ward AB,
    23. Bukreyev A,
    24. Crowe JE
    . 2016. Cross-reactive and potent neutralizing antibody responses in human survivors of natural ebolavirus infection. Cell164:392–405. doi:10.1016/j.cell.2015.12.022.
    OpenUrlCrossRefPubMed
  15. 15.↵
    1. Bornholdt ZA,
    2. Turner HL,
    3. Murin CD,
    4. Li W,
    5. Sok D,
    6. Souders CA,
    7. Piper AE,
    8. Goff A,
    9. Shamblin JD,
    10. Wollen SE,
    11. Sprague TR,
    12. Fusco ML,
    13. Pommert KBJ,
    14. Cavacini LA,
    15. Smith HL,
    16. Klempner M,
    17. Reimann KA,
    18. Krauland E,
    19. Gerngross TU,
    20. Wittrup KD,
    21. Saphire EO,
    22. Burton DR,
    23. Glass PJ,
    24. Ward AB,
    25. Walker LM
    . 2016. Isolation of potent neutralizing antibodies from a survivor of the 2014 Ebola virus outbreak. Science351:1078–1083. doi:10.1126/science.aad5788.
    OpenUrlAbstract/FREE Full Text
  16. 16.↵
    1. Howell KA,
    2. Qiu X,
    3. Brannan JM,
    4. Bryan C,
    5. Davidson E,
    6. Holtsberg FW,
    7. Wec AZ,
    8. Shulenin S,
    9. Biggins JE,
    10. Douglas R,
    11. Enterlein SG,
    12. Turner HL,
    13. Pallesen J,
    14. Murin CD,
    15. He S,
    16. Kroeker A,
    17. Vu H,
    18. Herbert AS,
    19. Fusco ML,
    20. Nyakatura EK,
    21. Lai JR,
    22. Keck Z-Y,
    23. Foung SKH,
    24. Saphire EO,
    25. Zeitlin L,
    26. Ward AB,
    27. Chandran K,
    28. Doranz BJ,
    29. Kobinger GP,
    30. Dye JM,
    31. Aman MJ
    . 2016. Antibody treatment of Ebola and Sudan virus infection via a uniquely exposed epitope within the glycoprotein receptor-binding site. Cell Rep15:1514–1526. doi:10.1016/j.celrep.2016.04.026.
    OpenUrlCrossRefPubMed
  17. 17.↵
    1. Holtsberg FW,
    2. Shulenin S,
    3. Vu H,
    4. Howell KA,
    5. Patel SJ,
    6. Gunn B,
    7. Karim M,
    8. Lai JR,
    9. Frei JC,
    10. Nyakatura EK,
    11. Zeitlin L,
    12. Douglas R,
    13. Fusco ML,
    14. Froude JW,
    15. Saphire EO,
    16. Herbert AS,
    17. Wirchnianski AS,
    18. Lear-Rooney CM,
    19. Alter G,
    20. Dye JM,
    21. Glass PJ,
    22. Warfield KL,
    23. Aman MJ
    . 2016. Pan-ebolavirus and pan-filovirus mouse monoclonal antibodies: protection against Ebola and Sudan viruses. J Virol90:266–278. doi:10.1128/JVI.02171-15.
    OpenUrlAbstract/FREE Full Text
  18. 18.↵
    1. Frei JC,
    2. Nyakatura EK,
    3. Zak SE,
    4. Bakken RR,
    5. Chandran K,
    6. Dye JM,
    7. Lai JR
    . 2016. Bispecific antibody affords complete post-exposure protection of mice from both ebola (Zaire) and Sudan viruses. Sci Rep6:19193. doi:10.1038/srep19193.
    OpenUrlCrossRefPubMed
  19. 19.↵
    1. Wec AZ,
    2. Nyakatura EK,
    3. Herbert AS,
    4. Howell KA,
    5. Holtsberg FW,
    6. Bakken RR,
    7. Mittler E,
    8. Christin JR,
    9. Shulenin S,
    10. Jangra RK,
    11. Bharrhan S,
    12. Kuehne AI,
    13. Bornholdt ZA,
    14. Flyak AI,
    15. Saphire EO,
    16. Crowe JE,
    17. Aman MJ,
    18. Dye JM,
    19. Lai JR,
    20. Chandran K
    . 2016. A “Trojan horse” bispecific-antibody strategy for broad protection against ebolaviruses. Science354:350–354. doi:10.1126/science.aag3267.
    OpenUrlAbstract/FREE Full Text
  20. 20.↵
    1. Holmes EC,
    2. Dudas G,
    3. Rambaut A,
    4. Andersen KG
    . 2016. The evolution of Ebola virus: insights from the 2013-2016 epidemic. Nature538:193–200. doi:10.1038/nature19790.
    OpenUrlCrossRefPubMed
  21. 21.↵
    1. Moekotte AL,
    2. Huson MAM,
    3. van der Ende AJ,
    4. Agnandji ST,
    5. Huizenga E,
    6. Goorhuis A,
    7. Grobusch MP
    . 2016. Monoclonal antibodies for the treatment of Ebola virus disease. Expert Opin Investig Drugs25:1325–1335. doi:10.1080/13543784.2016.1240785.
    OpenUrlCrossRef
  22. 22.↵
    1. Wei C-J,
    2. Boyington JC,
    3. McTamney PM,
    4. Kong W-P,
    5. Pearce MB,
    6. Xu L,
    7. Andersen H,
    8. Rao S,
    9. Tumpey TM,
    10. Yang Z-Y,
    11. Nabel GJ
    . 2010. Induction of broadly neutralizing H1N1 influenza antibodies by vaccination. Science329:1060–1064. doi:10.1126/science.1192517.
    OpenUrlAbstract/FREE Full Text
  23. 23.↵
    1. Leung LW,
    2. Martinez O,
    3. Reynard O,
    4. Volchkov VE,
    5. Basler CF
    . 2011. Ebola virus failure to stimulate plasmacytoid dendritic cell interferon responses correlates with impaired cellular entry. J Infect Dis204:S973–S977. doi:10.1093/infdis/jir331.
    OpenUrlCrossRefPubMed
  24. 24.↵
    1. Baize S,
    2. Pannetier D,
    3. Oestereich L,
    4. Rieger T,
    5. Koivogui L,
    6. Magassouba N,
    7. Soropogui B,
    8. Sow MS,
    9. Keïta S,
    10. De Clerck H,
    11. Tiffany A,
    12. Dominguez G,
    13. Loua M,
    14. Traoré A,
    15. Kolié M,
    16. Malano ER,
    17. Heleze E,
    18. Bocquin A,
    19. Mély S,
    20. Raoul H,
    21. Caro V,
    22. Cadar D,
    23. Gabriel M,
    24. Pahlmann M,
    25. Tappe D,
    26. Schmidt-Chanasit J,
    27. Impouma B,
    28. Diallo AK,
    29. Formenty P,
    30. Van Herp M,
    31. Günther S
    . 2014. Emergence of Zaire Ebola virus disease in Guinea. N Engl J Med371:1418–1425. doi:10.1056/NEJMoa1404505.
    OpenUrlCrossRefPubMed
  25. 25.↵
    1. Furuyama W,
    2. Marzi A,
    3. Nanbo A,
    4. Haddock E,
    5. Maruyama J,
    6. Miyamoto H,
    7. Igarashi M,
    8. Yoshida R,
    9. Noyori O,
    10. Feldmann H,
    11. Takada A
    . 2016. Discovery of an antibody for pan-ebolavirus therapy. Sci Rep6:20514. doi:10.1038/srep20514.
    OpenUrlCrossRefPubMed
  26. 26.↵
    1. Tran EEH,
    2. Nelson EA,
    3. Bonagiri P,
    4. Simmons JA,
    5. Shoemaker CJ,
    6. Schmaljohn CS,
    7. Kobinger GP,
    8. Zeitlin L,
    9. Subramaniam S,
    10. White JM
    . 2016. Mapping of Ebolavirus neutralization by monoclonal antibodies in the ZMapp Cocktail using cryo-electron tomography and studies of cellular entry. J Virol90:7618–7627. doi:10.1128/JVI.00406-16.
    OpenUrlAbstract/FREE Full Text
  27. 27.↵
    1. Chan KR,
    2. Ong EZ,
    3. Mok DZL,
    4. Ooi EE
    . 2015. Fc receptors and their influence on efficacy of therapeutic antibodies for treatment of viral diseases. Expert Rev Anti Infect Ther13:1351–1360. doi:10.1586/14787210.2015.1079127.
    OpenUrlCrossRef
  28. 28.↵
    1. Nimmerjahn F,
    2. Gordan S,
    3. Lux A
    . 2015. FcγR dependent mechanisms of cytotoxic, agonistic, and neutralizing antibody activities. Trends Immunol36:325–336. doi:10.1016/j.it.2015.04.005.
    OpenUrlCrossRefPubMed
  29. 29.↵
    1. Ledgerwood JE,
    2. Wei C-J,
    3. Hu Z,
    4. Gordon IJ,
    5. Enama ME,
    6. Hendel CS,
    7. McTamney PM,
    8. Pearce MB,
    9. Yassine HM,
    10. Boyington JC,
    11. Bailer R,
    12. Tumpey TM,
    13. Koup RA,
    14. Mascola JR,
    15. Nabel GJ,
    16. Graham BS
    , VRC 306 Study Team. 2011. DNA priming and influenza vaccine immunogenicity: two phase 1 open label randomised clinical trials. Lancet Infect Dis11:916–924. doi:10.1016/S1473-3099(11)70240-7.
    OpenUrlCrossRefPubMedWeb of Science
  30. 30.↵
    1. Ledgerwood JE,
    2. Zephir K,
    3. Hu Z,
    4. Wei C-J,
    5. Chang L,
    6. Enama ME,
    7. Hendel CS,
    8. Sitar S,
    9. Bailer RT,
    10. Koup RA,
    11. Mascola JR,
    12. Nabel GJ,
    13. Graham BS
    , VRC 310 Study Team. 2013. Prime-boost interval matters: a randomized phase 1 study to identify the minimum interval necessary to observe the H5 DNA influenza vaccine priming effect. J Infect Dis208:418–422. doi:10.1093/infdis/jit180.
    OpenUrlCrossRefPubMed
  31. 31.↵
    1. Babu TM,
    2. Levine M,
    3. Fitzgerald T,
    4. Luke C,
    5. Sangster MY,
    6. Jin H,
    7. Topham D,
    8. Katz J,
    9. Treanor J,
    10. Subbarao K
    . 2014. Live attenuated H7N7 influenza vaccine primes for a vigorous antibody response to inactivated H7N7 influenza vaccine. Vaccine32:6798–6804. doi:10.1016/j.vaccine.2014.09.070.
    OpenUrlCrossRefPubMed
  32. 32.↵
    1. Halliley JL,
    2. Khurana S,
    3. Krammer F,
    4. Fitzgerald T,
    5. Coyle EM,
    6. Chung KY,
    7. Baker SF,
    8. Yang H,
    9. Martínez-Sobrido L,
    10. Treanor JJ,
    11. Subbarao K,
    12. Golding H,
    13. Topham DJ,
    14. Sangster MY
    . 2015. High-affinity H7 head and stalk domain-specific antibody responses to an inactivated influenza H7N7 vaccine after priming with live attenuated influenza vaccine. J Infect Dis212:1270–1278. doi:10.1093/infdis/jiv210.
    OpenUrlCrossRefPubMed
  33. 33.↵
    1. Krammer F,
    2. Schinko T,
    3. Palmberger D,
    4. Tauer C,
    5. Messner P,
    6. Grabherr R
    . 2010. Trichoplusia ni cells (High FiveTM) are highly efficient for the production of influenza A virus-like particles: a comparison of two insect cell lines as production platforms for influenza vaccines. Mol Biotechnol45:226–234. doi:10.1007/s12033-010-9268-3.
    OpenUrlCrossRefPubMed
  34. 34.↵
    1. Wong AC,
    2. Sandesara RG,
    3. Mulherkar N,
    4. Whelan SP,
    5. Chandran K
    . 2010. A forward genetic strategy reveals destabilizing mutations in the Ebolavirus glycoprotein that alter its protease dependence during cell entry. J Virol84:163–175. doi:10.1128/JVI.01832-09.
    OpenUrlAbstract/FREE Full Text
  35. 35.↵
    1. Carette JE,
    2. Raaben M,
    3. Wong AC,
    4. Herbert AS,
    5. Obernosterer G,
    6. Mulherkar N,
    7. Kuehne AI,
    8. Kranzusch PJ,
    9. Griffin AM,
    10. Ruthel G,
    11. Dal Cin P,
    12. Dye JM,
    13. Whelan SP,
    14. Chandran K,
    15. Brummelkamp TR
    . 2011. Ebola virus entry requires the cholesterol transporter Niemann-Pick C1. Nature477:340–343. doi:10.1038/nature10348.
    OpenUrlCrossRefPubMedWeb of Science
  36. 36.↵
    1. Liu Q,
    2. Mena I,
    3. Ma J,
    4. Bawa B,
    5. Krammer F,
    6. Lyoo YS,
    7. Lang Y,
    8. Morozov I,
    9. Mahardika GN,
    10. Ma W,
    11. García-Sastre A,
    12. Richt JA
    . 2015. Newcastle disease virus-vectored H7 and H5 live vaccines protect chickens from challenge with H7N9 or H5N1 avian influenza viruses. J Virol89:7401–7408. doi:10.1128/JVI.00031-15.
    OpenUrlAbstract/FREE Full Text
  37. 37.↵
    1. Ayllon J,
    2. García-Sastre A,
    3. Martínez-Sobrido L
    . 11October2013. Rescue of recombinant Newcastle disease virus from cDNA. J Vis Exp. doi:10.3791/50830.
    OpenUrlCrossRef
  38. 38.↵
    1. Margine I,
    2. Palese P,
    3. Krammer F
    . 2013. Expression of functional recombinant hemagglutinin and neuraminidase proteins from the novel H7N9 influenza virus using the baculovirus expression system. J Vis Exp81:e51112. doi:10.3791/51112.
    OpenUrlCrossRefPubMed
  39. 39.↵
    1. Krammer F,
    2. Margine I,
    3. Tan GS,
    4. Pica N,
    5. Krause JC,
    6. Palese P
    . 2012. A carboxy-terminal trimerization domain stabilizes conformational epitopes on the stalk domain of soluble recombinant hemagglutinin substrates. PLoS One7:e43603. doi:10.1371/journal.pone.0043603.
    OpenUrlCrossRefPubMed
  40. 40.↵
    1. Luxembourg A,
    2. Evans CF,
    3. Hannaman D
    . 2007. Electroporation-based DNA immunisation: translation to the clinic. Expert Opin Biol Ther7:1647–1664. doi:10.1517/14712598.7.11.1647.
    OpenUrlCrossRefPubMedWeb of Science
  41. 41.↵
    1. Luxembourg A,
    2. Hannaman D,
    3. Nolan E,
    4. Ellefsen B,
    5. Nakamura G,
    6. Chau L,
    7. Tellez O,
    8. Little S,
    9. Bernard R
    . 2008. Potentiation of an anthrax DNA vaccine with electroporation. Vaccine26:5216–5222. doi:10.1016/j.vaccine.2008.03.064.
    OpenUrlCrossRefPubMed
  42. 42.↵
    1. Tan GS,
    2. Krammer F,
    3. Eggink D,
    4. Kongchanagul A,
    5. Moran TM,
    6. Palese P
    . 2012. A pan-H1 anti-hemagglutinin monoclonal antibody with potent broad-spectrum efficacy in vivo. J Virol86:6179–6188. doi:10.1128/JVI.00469-12.
    OpenUrlAbstract/FREE Full Text
  43. 43.↵
    1. Letunic I,
    2. Bork P
    . 2016. Interactive tree of life (iTOL) v3: an online tool for the display and annotation of phylogenetic and other trees. Nucleic Acids Res44:W242. doi:10.1093/nar/gkw290.
    OpenUrlCrossRefPubMed
  44. 44.↵
    1. Ackerman ME,
    2. Moldt B,
    3. Wyatt RT,
    4. Dugast A-S,
    5. McAndrew E,
    6. Tsoukas S,
    7. Jost S,
    8. Berger CT,
    9. Sciaranghella G,
    10. Liu Q,
    11. Irvine DJ,
    12. Burton DR,
    13. Alter G
    . 2011. A robust, high-throughput assay to determine the phagocytic activity of clinical antibody samples. J Immunol Methods366:8–19. doi:10.1016/j.jim.2010.12.016.
    OpenUrlCrossRefPubMedWeb of Science
  45. 45.↵
    1. Lee JE,
    2. Fusco ML,
    3. Abelson DM,
    4. Hessell AJ,
    5. Burton DR,
    6. Saphire EO
    . 2009. Techniques and tactics used in determining the structure of the trimeric ebolavirus glycoprotein. Acta Crystallogr D Biol Crystallogr65:1162–1180. doi:10.1107/S0907444909032314.
    OpenUrlCrossRefPubMedWeb of Science
PreviousNext
Back to top
Download PDF
Citation Tools
Novel Cross-Reactive Monoclonal Antibodies against Ebolavirus Glycoproteins Show Protection in a Murine Challenge Model
James Duehr, Teddy John Wohlbold, Lisa Oestereich, Veronika Chromikova, Fatima Amanat, Madhusudan Rajendran, Sergio Gomez-Medina, Ignacio Mena, Benjamin R. tenOever, Adolfo García-Sastre, Christopher F. Basler, Cesar Munoz-Fontela, Florian Krammer
Journal of Virology Jul 2017, 91 (16) e00652-17; DOI: 10.1128/JVI.00652-17

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Print

Alerts
Sign In to Email Alerts with your Email Address
Email

Thank you for sharing this Journal of Virology article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Novel Cross-Reactive Monoclonal Antibodies against Ebolavirus Glycoproteins Show Protection in a Murine Challenge Model
(Your Name) has forwarded a page to you from Journal of Virology
(Your Name) thought you would be interested in this article in Journal of Virology.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
Novel Cross-Reactive Monoclonal Antibodies against Ebolavirus Glycoproteins Show Protection in a Murine Challenge Model
James Duehr, Teddy John Wohlbold, Lisa Oestereich, Veronika Chromikova, Fatima Amanat, Madhusudan Rajendran, Sergio Gomez-Medina, Ignacio Mena, Benjamin R. tenOever, Adolfo García-Sastre, Christopher F. Basler, Cesar Munoz-Fontela, Florian Krammer
Journal of Virology Jul 2017, 91 (16) e00652-17; DOI: 10.1128/JVI.00652-17
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Top
  • Article
    • ABSTRACT
    • INTRODUCTION
    • RESULTS
    • DISCUSSION
    • MATERIALS AND METHODS
    • ACKNOWLEDGMENTS
    • FOOTNOTES
    • REFERENCES
  • Figures & Data
  • Info & Metrics
  • PDF

KEYWORDS

Antibodies, Monoclonal
Antibodies, Viral
cross protection
ebolavirus
Hemorrhagic Fever, Ebola
Immunologic Factors
Viral Envelope Proteins
EVD
ebolavirus
filovirus
glycoprotein
MAb

Related Articles

Cited By...

About

  • About JVI
  • Editor in Chief
  • Editorial Board
  • Policies
  • For Reviewers
  • For the Media
  • For Librarians
  • For Advertisers
  • Alerts
  • RSS
  • FAQ
  • Permissions
  • Journal Announcements

Authors

  • ASM Author Center
  • Submit a Manuscript
  • Article Types
  • Ethics
  • Contact Us

Follow #Jvirology

@ASMicrobiology

       

 

JVI in collaboration with

American Society for Virology

ASM Journals

ASM journals are the most prominent publications in the field, delivering up-to-date and authoritative coverage of both basic and clinical microbiology.

About ASM | Contact Us | Press Room

 

ASM is a member of

Scientific Society Publisher Alliance

 

American Society for Microbiology
1752 N St. NW
Washington, DC 20036
Phone: (202) 737-3600

Copyright © 2021 American Society for Microbiology | Privacy Policy | Website feedback

Print ISSN: 0022-538X; Online ISSN: 1098-5514