Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2682: 33-58, 2023.
Article in English | MEDLINE | ID: mdl-37610572

ABSTRACT

Henipaviruses possess two envelope glycoproteins, the attachment (G) and the fusion (F) proteins that mediate cellular entry and are the major targets of virus-neutralizing antibody responses. Recombinant expression technologies have been used to produce soluble G and F proteins (sG and sF) that retain native-like oligomeric conformations and epitopes, which are advantageous for the development and characterization of vaccines and antiviral antibody therapeutics. In addition to Hendra virus and Nipah virus tetrameric sG and trimeric sF production, we also describe the expression and purification of Cedar virus tetrameric sG and Ghana virus trimeric sF glycoproteins. These henipavirus glycoproteins were also used as immunizing antigens to generate monoclonal antibodies, and binding was demonstrated with a pan-henipavirus multiplex microsphere immunoassay.


Subject(s)
Henipavirus , Henipavirus/genetics , Antibodies, Blocking , Antibodies, Monoclonal
2.
Nat Struct Mol Biol ; 28(5): 426-434, 2021 05.
Article in English | MEDLINE | ID: mdl-33927387

ABSTRACT

Hendra virus (HeV) and Nipah virus (NiV) are henipaviruses (HNVs) causing respiratory illness and severe encephalitis in humans, with fatality rates of 50-100%. There are no licensed therapeutics or vaccines to protect humans. HeV and NiV use a receptor-binding glycoprotein (G) and a fusion glycoprotein (F) to enter host cells. HNV F and G are the main targets of the humoral immune response, and the presence of neutralizing antibodies is a correlate of protection against NiV and HeV in experimentally infected animals. We describe here two cross-reactive F-specific antibodies, 1F5 and 12B2, that neutralize NiV and HeV through inhibition of membrane fusion. Cryo-electron microscopy structures reveal that 1F5 and 12B2 recognize distinct prefusion-specific, conserved quaternary epitopes and lock F in its prefusion conformation. We provide proof-of-concept for using antibody cocktails for neutralizing NiV and HeV and define a roadmap for developing effective countermeasures against these highly pathogenic viruses.


Subject(s)
Antibodies, Viral/immunology , Broadly Neutralizing Antibodies/immunology , Hendra Virus/immunology , Nipah Virus/immunology , Viral Fusion Proteins/immunology , Animals , Antibodies, Monoclonal, Humanized/immunology , CHO Cells , Cricetulus , Cross Reactions , HEK293 Cells , Henipavirus Infections/immunology , Henipavirus Infections/prevention & control , Humans , Mice , Virus Internalization
3.
J Infect Dis ; 221(Suppl 4): S471-S479, 2020 05 11.
Article in English | MEDLINE | ID: mdl-31686101

ABSTRACT

BACKGROUND: Nipah virus (NiV) and Hendra virus (HeV) are zoonotic paramyxoviruses that cause severe disease in both animals and humans. There are no approved vaccines or treatments for use in humans; however, therapeutic treatment of both NiV and HeV infection in ferrets and non-human primates with a cross-reactive, neutralizing human monoclonal antibody (mAb), m102.4, targeting the G glycoprotein has been demonstrated. In a previous study, we isolated, characterized, and humanized a cross-reactive, neutralizing anti-F mAb (h5B3.1). The mAb h5B3.1 blocks the required F conformational change needed to facilitate membrane fusion and virus infection, and the epitope recognized by h5B3.1 has been structurally defined; however, the efficacy of h5B3.1 in vivo is unknown. METHODS: The post-infection antiviral activity of h5B3.1 was evaluated in vivo by administration in ferrets after NiV and HeV virus challenge. RESULTS: All subjects that received h5B3.1 from 1 to several days after infection with a high-dose, oral-nasal virus challenge were protected from disease, whereas all controls died. CONCLUSIONS: This is the first successful post-exposure antibody therapy for NiV and HeV using a humanized cross-reactive mAb targeting the F glycoprotein, and the findings suggest that a combination therapy targeting both F and G should be evaluated as a therapy for NiV/HeV infection.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Hendra Virus , Henipavirus Infections/therapy , Nipah Virus , Viral Fusion Proteins/immunology , Animals , Cross Reactions , Ferrets , Henipavirus Infections/prevention & control , Henipavirus Infections/virology , Humans
4.
Nat Struct Mol Biol ; 26(10): 980-987, 2019 10.
Article in English | MEDLINE | ID: mdl-31570878

ABSTRACT

Nipah virus (NiV) and Hendra virus (HeV) are zoonotic henipaviruses (HNVs) responsible for outbreaks of encephalitis and respiratory illness with fatality rates of 50-100%. No vaccines or licensed therapeutics currently exist to protect humans against NiV or HeV. HNVs enter host cells by fusing the viral and cellular membranes via the concerted action of the attachment (G) and fusion (F) glycoproteins, the main targets of the humoral immune response. Here, we describe the isolation and humanization of a potent monoclonal antibody cross-neutralizing NiV and HeV. Cryo-electron microscopy, triggering and fusion studies show the antibody binds to a prefusion-specific quaternary epitope, conserved in NiV F and HeV F glycoproteins, and prevents membrane fusion and viral entry. This work supports the importance of the HNV prefusion F conformation for eliciting a robust immune response and paves the way for using this antibody for prophylaxis and post-exposure therapy with NiV- and HeV-infected individuals.


Subject(s)
Antibodies, Neutralizing/pharmacology , Antiviral Agents/pharmacology , Hendra Virus/drug effects , Henipavirus Infections/drug therapy , Nipah Virus/drug effects , Viral Fusion Proteins/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized/pharmacology , HEK293 Cells , Hendra Virus/metabolism , Henipavirus Infections/metabolism , Henipavirus Infections/virology , Humans , Models, Molecular , Nipah Virus/metabolism , Viral Fusion Proteins/metabolism , Virus Internalization/drug effects
5.
Sci Rep ; 6: 30916, 2016 08 03.
Article in English | MEDLINE | ID: mdl-27484128

ABSTRACT

Nipah virus (NiV) is a paramyxovirus that causes severe disease in humans and animals. There are two distinct strains of NiV, Malaysia (NiVM) and Bangladesh (NiVB). Differences in transmission patterns and mortality rates suggest that NiVB may be more pathogenic than NiVM. To investigate pathogenic differences between strains, 4 African green monkeys (AGM) were exposed to NiVM and 4 AGMs were exposed to NiVB. While NiVB was uniformly lethal, only 50% of NiVM-infected animals succumbed to infection. Histopathology of lungs and spleens from NiVB-infected AGMs was significantly more severe than NiVM-infected animals. Importantly, a second study utilizing 11 AGMs showed that the therapeutic window for human monoclonal antibody m102.4, previously shown to rescue AGMs from NiVM infection, was much shorter in NiVB-infected AGMs. Together, these data show that NiVB is more pathogenic in AGMs under identical experimental conditions and suggests that postexposure treatments may need to be NiV strain specific for optimal efficacy.


Subject(s)
Henipavirus Infections/veterinary , Nipah Virus/classification , Nipah Virus/pathogenicity , Animals , Antibodies, Monoclonal/therapeutic use , Bangladesh , Chlorocebus aethiops , Female , Henipavirus Infections/prevention & control , Henipavirus Infections/transmission , Henipavirus Infections/virology , Malaysia , Male
6.
PLoS Pathog ; 11(12): e1005322, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26646856

ABSTRACT

Nipah virus (NiV) is a paramyxovirus that infects host cells through the coordinated efforts of two envelope glycoproteins. The G glycoprotein attaches to cell receptors, triggering the fusion (F) glycoprotein to execute membrane fusion. Here we report the first crystal structure of the pre-fusion form of the NiV-F glycoprotein ectodomain. Interestingly this structure also revealed a hexamer-of-trimers encircling a central axis. Electron tomography of Nipah virus-like particles supported the hexameric pre-fusion model, and biochemical analyses supported the hexamer-of-trimers F assembly in solution. Importantly, structure-assisted site-directed mutagenesis of the interfaces between F trimers highlighted the functional relevance of the hexameric assembly. Shown here, in both cell-cell fusion and virus-cell fusion systems, our results suggested that this hexamer-of-trimers assembly was important during fusion pore formation. We propose that this assembly would stabilize the pre-fusion F conformation prior to cell attachment and facilitate the coordinated transition to a post-fusion conformation of all six F trimers upon triggering of a single trimer. Together, our data reveal a novel and functional pre-fusion architecture of a paramyxoviral fusion glycoprotein.


Subject(s)
Henipavirus Infections/metabolism , Nipah Virus/chemistry , Viral Envelope Proteins/chemistry , Virus Internalization , Crystallography, X-Ray , Electrophoresis, Polyacrylamide Gel , HEK293 Cells , Humans , Mutagenesis, Site-Directed , Nipah Virus/metabolism , Protein Conformation , Viral Envelope Proteins/metabolism
7.
Sci Transl Med ; 6(242): 242ra82, 2014 Jun 25.
Article in English | MEDLINE | ID: mdl-24964990

ABSTRACT

Nipah virus (NiV) is an emerging zoonotic paramyxovirus that causes severe and often fatal disease in pigs and humans. There are currently no vaccines or treatments approved for human use. Studies in small-animal models of NiV infection suggest that antibody therapy may be a promising treatment. However, most studies have assessed treatment at times shortly after virus exposure before animals show signs of disease. We assessed the efficacy of a fully human monoclonal antibody, m102.4, at several time points after virus exposure including at the onset of clinical illness in a uniformly lethal nonhuman primate model of NiV disease. Sixteen African green monkeys (AGMs) were challenged intratracheally with a lethal dose of NiV, and 12 animals were infused twice with m102.4 (15 mg/kg) beginning at either 1, 3, or 5 days after virus challenge and again about 2 days later. The presence of viral RNA, infectious virus, and/or NiV-specific immune responses demonstrated that all subjects were infected after challenge. All 12 AGMs that received m102.4 survived infection, whereas the untreated control subjects succumbed to disease between days 8 and 10 after infection. AGMs in the day 5 treatment group exhibited clinical signs of disease, but all animals recovered by day 16. These results represent the successful therapeutic in vivo efficacy by an investigational drug against NiV in a nonhuman primate and highlight the potential impact that a monoclonal antibody can have on a highly pathogenic zoonotic human disease.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Chlorocebus aethiops/immunology , Henipavirus Infections/prevention & control , Henipavirus Infections/therapy , Nipah Virus/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/immunology , Antigens, Viral/immunology , Henipavirus Infections/immunology , Henipavirus Infections/virology , Humans , Immunohistochemistry , Neutralization Tests , Treatment Outcome , Viral Load
8.
J Virol ; 88(9): 4624-31, 2014 May.
Article in English | MEDLINE | ID: mdl-24522928

ABSTRACT

UNLABELLED: Hendra virus (HeV) is a zoonotic emerging virus belonging to the family Paramyxoviridae. HeV causes severe and often fatal respiratory and/or neurologic disease in both animals and humans. Currently, there are no licensed vaccines or antiviral drugs approved for human use. A number of animal models have been developed for studying HeV infection, with the African green monkey (AGM) appearing to most faithfully reproduce the human disease. Here, we assessed the utility of a newly developed recombinant subunit vaccine based on the HeV attachment (G) glycoprotein in the AGM model. Four AGMs were vaccinated with two doses of the HeV vaccine (sGHeV) containing Alhydrogel, four AGMs received the sGHeV with Alhydrogel and CpG, and four control animals did not receive the sGHeV vaccine. Animals were challenged with a high dose of infectious HeV 21 days after the boost vaccination. None of the eight specifically vaccinated animals showed any evidence of clinical illness and survived the challenge. All four controls became severely ill with symptoms consistent with HeV infection, and three of the four animals succumbed 8 days after exposure. Success of the recombinant subunit vaccine in AGMs provides pivotal data in supporting its further preclinical development for potential human use. IMPORTANCE: A Hendra virus attachment (G) glycoprotein subunit vaccine was tested in nonhuman primates to assess its ability to protect them from a lethal infection with Hendra virus. It was found that all vaccinated African green monkeys were completely protected against subsequent Hendra virus infection and disease. The success of this new subunit vaccine in nonhuman primates provides critical data in support of its further development for future human use.


Subject(s)
Hendra Virus/immunology , Henipavirus Infections/prevention & control , Viral Envelope Proteins/immunology , Viral Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Aluminum Hydroxide/administration & dosage , Animals , Chlorocebus aethiops , Disease Models, Animal , Hendra Virus/genetics , Henipavirus Infections/pathology , Oligodeoxyribonucleotides/administration & dosage , Survival Analysis , Vaccination/methods , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Envelope Proteins/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
9.
PLoS Pathog ; 9(10): e1003684, 2013.
Article in English | MEDLINE | ID: mdl-24130486

ABSTRACT

The henipaviruses, represented by Hendra (HeV) and Nipah (NiV) viruses are highly pathogenic zoonotic paramyxoviruses with uniquely broad host tropisms responsible for repeated outbreaks in Australia, Southeast Asia, India and Bangladesh. The high morbidity and mortality rates associated with infection and lack of licensed antiviral therapies make the henipaviruses a potential biological threat to humans and livestock. Henipavirus entry is initiated by the attachment of the G envelope glycoprotein to host cell membrane receptors. Previously, henipavirus-neutralizing human monoclonal antibodies (hmAb) have been isolated using the HeV-G glycoprotein and a human naïve antibody library. One cross-reactive and receptor-blocking hmAb (m102.4) was recently demonstrated to be an effective post-exposure therapy in two animal models of NiV and HeV infection, has been used in several people on a compassionate use basis, and is currently in development for use in humans. Here, we report the crystal structure of the complex of HeV-G with m102.3, an m102.4 derivative, and describe NiV and HeV escape mutants. This structure provides detailed insight into the mechanism of HeV and NiV neutralization by m102.4, and serves as a blueprint for further optimization of m102.4 as a therapeutic agent and for the development of entry inhibitors and vaccines.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Neutralizing/chemistry , Antibodies, Viral/chemistry , Hendra Virus/chemistry , Viral Fusion Proteins/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cell Line , Cross Reactions , Crystallography, X-Ray , Hendra Virus/genetics , Hendra Virus/immunology , Henipavirus Infections/genetics , Henipavirus Infections/immunology , Humans , Protein Structure, Quaternary , Protein Structure, Tertiary , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology
10.
Proc Natl Acad Sci U S A ; 110(36): 14634-9, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-23959867

ABSTRACT

Eph receptor tyrosine kinases and their ephrin ligands mediate cell signaling during normal and oncogenic development. Eph signaling is initiated in a multistep process leading to the assembly of higher-order Eph/ephrin clusters that set off bidirectional signaling in interacting cells. Eph and ephrins are divided in two subclasses based on their abilities to bind and activate each other and on sequence conservation. EphA4 is an exception to the general rule because it can be activated by both A- and B-class ephrin ligands. Here we present high-resolution structures of the complete EphA4 ectodomain and its complexes with ephrin-A5. The structures reveal how ligand binding promotes conformational changes in the EphA4 ligand-binding domain allowing the formation of signaling clusters at the sites of cell-cell contact. In addition, the structural data, combined with structure-based mutagenesis, reveal a previously undescribed receptor-receptor interaction between the EphA4 ligand-binding and membrane-proximal fibronectin domains, which is functionally important for efficient receptor activation.


Subject(s)
Ephrin-A5/chemistry , Protein Structure, Tertiary , Receptor, EphA4/chemistry , Signal Transduction , Binding Sites/genetics , Blotting, Western , Crystallography, X-Ray , Enzyme Activation , Ephrin-A5/genetics , Ephrin-A5/metabolism , HEK293 Cells , Humans , Models, Molecular , Mutation , Protein Binding , Protein Conformation , Protein Multimerization , Receptor, EphA4/genetics , Receptor, EphA4/metabolism
11.
PLoS One ; 7(11): e48742, 2012.
Article in English | MEDLINE | ID: mdl-23144952

ABSTRACT

Hendra virus and Nipah virus, comprising the genus Henipavirus, are recently emerged, highly pathogenic and often lethal zoonotic agents against which there are no approved therapeutics. Two surface glycoproteins, the attachment (G) and fusion (F), mediate host cell entry. The crystal structures of the Hendra G glycoprotein alone and in complex with the ephrin-B2 receptor reveal that henipavirus uses Tryptophan 122 on ephrin-B2/B3 as a "latch" to facilitate the G-receptor association. Structural-based mutagenesis of residues in the Hendra G glycoprotein at the receptor binding interface document their importance for viral attachments and entry, and suggest that the stability of the Hendra-G-ephrin attachment complex does not strongly correlate with the efficiency of viral entry. In addition, our data indicates that conformational rearrangements of the G glycoprotein head domain upon receptor binding may be the trigger leading to the activation of the viral F fusion glycoprotein during virus infection.


Subject(s)
Ephrin-B2/chemistry , Glycoproteins/chemistry , Hendra Virus/pathogenicity , Viral Proteins/chemistry , Crystallography, X-Ray , Ephrin-B2/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , HEK293 Cells , HeLa Cells , Hendra Virus/genetics , Humans , Models, Biological , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Receptors, Virus/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Attachment , Virus Internalization
12.
Sci Transl Med ; 4(146): 146ra107, 2012 Aug 08.
Article in English | MEDLINE | ID: mdl-22875827

ABSTRACT

In the 1990s, Hendra virus and Nipah virus (NiV), two closely related and previously unrecognized paramyxoviruses that cause severe disease and death in humans and a variety of animals, were discovered in Australia and Malaysia, respectively. Outbreaks of disease have occurred nearly every year since NiV was first discovered, with case fatality ranging from 10 to 100%. In the African green monkey (AGM), NiV causes a severe lethal respiratory and/or neurological disease that essentially mirrors fatal human disease. Thus, the AGM represents a reliable disease model for vaccine and therapeutic efficacy testing. We show that vaccination of AGMs with a recombinant subunit vaccine based on the henipavirus attachment G glycoprotein affords complete protection against subsequent NiV infection with no evidence of clinical disease, virus replication, or pathology observed in any challenged subjects. Success of the recombinant subunit vaccine in nonhuman primates provides crucial data in supporting its further preclinical development for potential human use.


Subject(s)
Antigens, Viral/immunology , Chlorocebus aethiops/immunology , Chlorocebus aethiops/virology , Glycoproteins/immunology , Hendra Virus/immunology , Nipah Virus/immunology , Animals , Henipavirus Infections/immunology , Henipavirus Infections/prevention & control , Nipah Virus/pathogenicity
13.
J Virol ; 86(21): 11457-71, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22915804

ABSTRACT

The henipaviruses, Hendra virus (HeV) and Nipah virus (NiV), are paramyxoviruses discovered in the mid- to late 1990s that possess a broad host tropism and are known to cause severe and often fatal disease in both humans and animals. HeV and NiV infect cells by a pH-independent membrane fusion mechanism facilitated by their attachment (G) and fusion (F) glycoproteins. Here, several soluble forms of henipavirus F (sF) were engineered and characterized. Recombinant sF was produced by deleting the transmembrane (TM) and cytoplasmic tail (CT) domains and appending a glycosylphosphatidylinositol (GPI) anchor signal sequence followed by GPI-phospholipase D digestion, appending a trimeric coiled-coil (GCNt) domain (sF(GCNt)), or deleting the TM, CT, and fusion peptide domain. These sF glycoproteins were produced as F(0) precursors, and all were apparent stable trimers recognized by NiV-specific antisera. Surprisingly, however, only the GCNt-appended constructs (sF(GCNt)) could elicit cross-reactive henipavirus-neutralizing antibody in mice. In addition, sF(GCNt) constructs could be triggered in vitro by protease cleavage and heat to transition from an apparent prefusion to postfusion conformation, transitioning through an intermediate that could be captured by a peptide corresponding to the C-terminal heptad repeat domain of F. The pre- and postfusion structures of sF(GCNt) and non-GCNt-appended sF could be revealed by electron microscopy and were distinguishable by F-specific monoclonal antibodies. These data suggest that only certain sF constructs could serve as potential subunit vaccine immunogens against henipaviruses and also establish important tools for further structural, functional, and diagnostic studies on these important emerging viruses.


Subject(s)
Henipavirus/immunology , Henipavirus/ultrastructure , Viral Fusion Proteins/immunology , Viral Fusion Proteins/ultrastructure , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cross Reactions , HeLa Cells , Henipavirus/genetics , Humans , Mice , Microscopy, Electron , Protein Conformation , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism
14.
Glycobiology ; 22(4): 572-84, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22171062

ABSTRACT

Hendra virus (HeV) continues to cause morbidity and mortality in both humans and horses with a number of sporadic outbreaks. HeV has two structural membrane glycoproteins that mediate the infection of host cells: the attachment (G) and the fusion (F) glycoproteins that are essential for receptor binding and virion-host cell membrane fusion, respectively. N-linked glycosylation of viral envelope proteins are critical post-translation modifications that have been implicated in roles of structural integrity, virus replication and evasion of the host immune response. Deciphering the glycan composition and structure on these glycoproteins may assist in the development of glycan-targeted therapeutic intervention strategies. We examined the site occupancy and glycan composition of recombinant soluble G (sG) glycoproteins expressed in two different mammalian cell systems, transient human embryonic kidney 293 (HEK293) cells and vaccinia virus (VV)-HeLa cells, using a suite of biochemical and biophysical tools: electrophoresis, lectin binding and tandem mass spectrometry. The N-linked glycans of both VV and HEK293-derived sG glycoproteins carried predominantly mono- and disialylated complex-type N-glycans and a smaller population of high mannose-type glycans. All seven consensus sequences for N-linked glycosylation were definitively found to be occupied in the VV-derived protein, whereas only four sites were found and characterized in the HEK293-derived protein. We also report, for the first time, the existence of O-linked glycosylation sites in both proteins. The striking characteristic of both proteins was glycan heterogeneity in both N- and O-linked sites. The structural features of G protein glycosylation were also determined by X-ray crystallography and interactions with the ephrin-B2 receptor are discussed.


Subject(s)
Hendra Virus , Polysaccharides/chemistry , Viral Envelope Proteins/chemistry , Amino Acid Motifs , Amino Acid Sequence , Carbohydrate Conformation , Carbohydrate Sequence , Crystallography, X-Ray , Electrophoretic Mobility Shift Assay , Glycosylation , HEK293 Cells , HeLa Cells , Humans , Lectins/chemistry , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemistry , Protein Binding , Protein Structure, Quaternary , Receptor, EphB2/chemistry , Recombinant Proteins/chemistry , Sequence Analysis, Protein
15.
Sci Transl Med ; 3(105): 105ra103, 2011 Oct 19.
Article in English | MEDLINE | ID: mdl-22013123

ABSTRACT

Hendra virus (HeV) is a recently emerged zoonotic paramyxovirus that can cause a severe and often fatal disease in horses and humans. HeV is categorized as a biosafety level 4 agent, which has made the development of animal models and testing of potential therapeutics and vaccines challenging. Infection of African green monkeys (AGMs) with HeV was recently demonstrated, and disease mirrored fatal HeV infection in humans, manifesting as a multisystemic vasculitis with widespread virus replication in vascular tissues and severe pathologic manifestations in the lung, spleen, and brain. Here, we demonstrate that m102.4, a potent HeV-neutralizing human monoclonal antibody (hmAb), can protect AGMs from disease after infection with HeV. Fourteen AGMs were challenged intratracheally with a lethal dose of HeV, and 12 subjects were infused twice with a 100-mg dose of m102.4 beginning at either 10, 24, or 72 hours after infection and again about 48 hours later. The presence of viral RNA, infectious virus, and HeV-specific immune responses demonstrated that all subjects were infected after challenge. All 12 AGMs that received m102.4 survived infection, whereas the untreated control subjects succumbed to disease on day 8 after infection. Animals in the 72-hour treatment group exhibited neurological signs of disease, but all animals started to recover by day 16 after infection. These results represent successful post-exposure in vivo efficacy by an investigational drug against HeV and highlight the potential impact a hmAb can have on human disease.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Hendra Virus/pathogenicity , Henipavirus Infections/drug therapy , Animals , Antibodies, Monoclonal/pharmacokinetics , Chlorocebus aethiops , Henipavirus Infections/prevention & control , Humans
16.
Vaccine ; 29(34): 5623-30, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21689706

ABSTRACT

The henipaviruses, Hendra virus (HeV) and Nipah virus (NiV), are two deadly zoonotic viruses for which no vaccines or therapeutics have yet been approved for human or livestock use. In 14 outbreaks since 1994 HeV has been responsible for multiple fatalities in horses and humans, with all known human infections resulting from close contact with infected horses. A vaccine that prevents virus shedding in infected horses could interrupt the chain of transmission to humans and therefore prevent HeV disease in both. Here we characterise HeV infection in a ferret model and show that it closely mirrors the disease seen in humans and horses with induction of systemic vasculitis, including involvement of the pulmonary and central nervous systems. This model of HeV infection in the ferret was used to assess the immunogenicity and protective efficacy of a subunit vaccine based on a recombinant soluble version of the HeV attachment glycoprotein G (HeVsG), adjuvanted with CpG. We report that ferrets vaccinated with a 100 µg, 20 µg or 4 µg dose of HeVsG remained free of clinical signs of HeV infection following a challenge with 5000 TCID50 of HeV. In addition, and of considerable importance, no evidence of virus or viral genome was detected in any tissues or body fluids in any ferret in the 100 and 20 µg groups, while genome was detected in the nasal washes only of one animal in the 4 µg group. Together, our findings indicate that 100 µg or 20 µg doses of HeVsG vaccine can completely prevent a productive HeV infection in the ferret, suggesting that vaccination to prevent the infection and shedding of HeV is possible.


Subject(s)
Hendra Virus/immunology , Vaccines, Subunit/immunology , Viral Envelope Proteins/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/immunology , Ferrets/immunology , Ferrets/virology , Glycoproteins/immunology , Henipavirus Infections/immunology , Henipavirus Infections/prevention & control , Recombinant Proteins/immunology , Vaccines, Synthetic/immunology , Virus Shedding/immunology
17.
PLoS One ; 5(5): e10690, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20502528

ABSTRACT

Nipah virus (NiV) is an enigmatic emerging pathogen that causes severe and often fatal neurologic and/or respiratory disease in both animals and humans. Amongst people, case fatality rates range between 40 and 75 percent and there are no vaccines or treatments approved for human use. Guinea pigs, hamsters, cats, ferrets, pigs and most recently squirrel monkeys (New World monkey) have been evaluated as animal models of human NiV infection, and with the exception of the ferret, no model recapitulates all aspects of NiV-mediated disease seen in humans. To identify a more viable nonhuman primate (NHP) model, we examined the pathogenesis of NiV in African green monkeys (AGM). Exposure of eight monkeys to NiV produced a severe systemic infection in all eight animals with seven of the animals succumbing to infection. Viral RNA was detected in the plasma of challenged animals and occurred in two of three subjects as a peak between days 7 and 21, providing the first clear demonstration of plasma-associated viremia in NiV experimentally infected animals and suggested a progressive infection that seeded multiple organs simultaneously from the initial site of virus replication. Unlike the cat, hamster and squirrel monkey models of NiV infection, severe respiratory pathology, neurological disease and generalized vasculitis all manifested in NiV-infected AGMs, providing an accurate reflection of what is observed in NiV-infected humans. Our findings demonstrate the first consistent and highly pathogenic NHP model of NiV infection, providing a new and critical platform in the evaluation and licensure of either passive and active immunization or therapeutic strategies for human use.


Subject(s)
Henipavirus Infections/immunology , Henipavirus Infections/virology , Nipah Virus/pathogenicity , Primates/immunology , Primates/virology , Animals , Autoradiography , Chlorocebus aethiops , Henipavirus Infections/blood , Humans , Models, Immunological , Nipah Virus/physiology , Organ Specificity/immunology , RNA, Viral/blood , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction , Viral Load/immunology , Viral Tropism/immunology
18.
PLoS Pathog ; 5(10): e1000642, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19888339

ABSTRACT

Nipah virus is a broadly tropic and highly pathogenic zoonotic paramyxovirus in the genus Henipavirus whose natural reservoirs are several species of Pteropus fruit bats. Nipah virus has repeatedly caused outbreaks over the past decade associated with a severe and often fatal disease in humans and animals. Here, a new ferret model of Nipah virus pathogenesis is described where both respiratory and neurological disease are present in infected animals. Severe disease occurs with viral doses as low as 500 TCID(50) within 6 to 10 days following infection. The underlying pathology seen in the ferret closely resembles that seen in Nipah virus infected humans, characterized as a widespread multisystemic vasculitis, with virus replicating in highly vascular tissues including lung, spleen and brain, with recoverable virus from a variety of tissues. Using this ferret model a cross-reactive neutralizing human monoclonal antibody, m102.4, targeting the henipavirus G glycoprotein was evaluated in vivo as a potential therapeutic agent. All ferrets that received m102.4 ten hours following a high dose oral-nasal Nipah virus challenge were protected from disease while all controls died. This study is the first successful post-exposure passive antibody therapy for Nipah virus using a human monoclonal antibody.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Henipavirus Infections/prevention & control , Nipah Virus/pathogenicity , Acute Disease , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/therapeutic use , Disease Models, Animal , Ferrets , Glycoproteins/immunology , Henipavirus Infections/immunology , Henipavirus Infections/pathology , Humans , Immunohistochemistry , Nipah Virus/immunology , RNA, Viral/metabolism , Tissue Distribution , Viral Envelope Proteins/immunology , Viral Load
19.
Methods Mol Biol ; 525: 31-58, xiii, 2009.
Article in English | MEDLINE | ID: mdl-19252850

ABSTRACT

Neutralizing antibodies are a critical component in the protection or recovery from viral infections. In the absence of available vaccines or antiviral drugs for many important human viral pathogens, the identification and characterization of new human monoclonal antibodies (hmAbs) that are able to neutralize viruses offers the possibility for effective pre- and/or post-exposure therapeutic modalities. Such hmAbs may also help in our understanding of the virus entry process, the mechanisms of virus neutralization, and in the eventual development of specific entry inhibitors, vaccines, and research tools. The majority of the more recently developed antiviral hmAbs have come from the use of antibody phage-display technologies using both naïve and immune libraries. Many of these agents are also enveloped viruses possessing important neutralizing determinants within their membrane-anchored envelope glycoproteins, and the use of recombinant, soluble versions of these viral glycoproteins is often critical in the isolation and development of antiviral hmAbs. This chapter will detail several methods that have been successfully employed to produce, purify, and characterize soluble and secreted versions of several viral envelope glycoproteins which have been successfully used as antigens to capture and isolate human phage-displayed monoclonal antibodies.


Subject(s)
Antibodies/therapeutic use , Glycoproteins/biosynthesis , Molecular Biology/methods , Recombinant Proteins/biosynthesis , Viral Proteins/biosynthesis , Animals , Cell Line , Chromatography, Gel , Electrophoresis, Polyacrylamide Gel , Humans , Molecular Weight , Solubility , Ultracentrifugation , Vaccinia virus
20.
J Infect Dis ; 198(6): 836-42, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18680407

ABSTRACT

BACKGROUND: Human metapneumovirus (hMPV) is a newly discovered paramyxovirus that causes acute respiratory illness. Despite apparent near-universal exposure during early childhood, immunity is transient. METHODS: An indirect screening enzyme-linked immunosorbent assay using a recombinant soluble fusion (F) glycoprotein derived from hMPV was used to test for anti-F IgG in 1,380 pairs of acute- and convalescent-stage serum samples collected from children in Kamphaeng Phet, Thailand. RESULTS: Of the 1,380 serum sample pairs tested, 1,376 (99.7%) showed evidence of prior infection with hMPV. Sixty-six paired specimens demonstrated a >or=4-fold rise in titer, for an overall reinfection rate of 4.9%. Two children demonstrated evidence of an initial infection. Forty-eight of the 68 new infections or reinfections occurred in 2000, accounting for 13.2% of all nonflaviviral febrile illnesses in the study population in that year. Of 68 positive cases, 85.3% complained of cough and 66.2% complained of rhinorrhea, compared with 61.4% and 49.0% of negative cases, respectively (P < .01). All positive samples were also tested for an increase in titer of antibodies to respiratory syncytial virus F, and 27% exhibited a >or=4-fold rise. CONCLUSION: These results demonstrate that hMPV reinfections cause illness at a rate equal to that seen for initial infections. hMPV may have a more significant impact in older children than previously realized and may be the cause of significant outbreaks in this population.


Subject(s)
Metapneumovirus , Paramyxoviridae Infections/epidemiology , Adolescent , Child , Demography , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin G/blood , Paramyxoviridae Infections/immunology , Recurrence , Thailand/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...