Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Infect Dis ; 218(4): 572-580, 2018 07 13.
Article in English | MEDLINE | ID: mdl-29617879

ABSTRACT

Background: Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection among infants and young children. To date, no vaccine is approved for the broad population of healthy infants. MEDI8897, a potent anti-RSV fusion antibody with extended serum half-life, is currently under clinical investigation as a potential passive RSV vaccine for all infants. As a ribonucleic acid virus, RSV is prone to mutation, and the possibility of viral escape from MEDI8897 neutralization is a potential concern. Methods: We generated RSV monoclonal antibody (mAb)-resistant mutants (MARMs) in vitro and studied the effect of the amino acid substitutions identified on binding and viral neutralization susceptibility to MEDI8897. The impact of resistance-associated mutations on in vitro growth kinetics and the prevalence of these mutations in currently circulating strains of RSV in the United States was assessed. Results: Critical residues identified in MARMs for MEDI8897 neutralization were located in the MEDI8897 binding site defined by crystallographic analysis. Substitutions in these residues affected the binding of mAb to virus, without significant impact on viral replication in vitro. The frequency of natural resistance-associated polymorphisms was low. Conclusions: Results from this study provide insights into the mechanism of MEDI8897 escape and the complexity of monitoring for emergence of resistance.


Subject(s)
Amino Acid Substitution , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Immunologic Factors/pharmacology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/immunology , Viral Fusion Proteins/immunology , Binding Sites , Biological Products/pharmacology , Crystallography, X-Ray , Drug Resistance, Viral , Gene Frequency , Humans , Immune Evasion , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/immunology , Neutralization Tests , Prevalence , Protein Conformation , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/isolation & purification , United States/epidemiology , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics , Virus Attachment/drug effects , Virus Replication/drug effects
2.
PLoS Pathog ; 11(7): e1005035, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26161532

ABSTRACT

Prevention efforts for respiratory syncytial virus (RSV) have been advanced due to the recent isolation and characterization of antibodies that specifically recognize the prefusion conformation of the RSV fusion (F) glycoprotein. These potently neutralizing antibodies are in clinical development for passive prophylaxis and have also aided the design of vaccine antigens that display prefusion-specific epitopes. To date, prefusion-specific antibodies have been shown to target two antigenic sites on RSV F, but both of these sites are also present on monomeric forms of F. Here we present a structural and functional characterization of human antibody AM14, which potently neutralized laboratory strains and clinical isolates of RSV from both A and B subtypes. The crystal structure and location of escape mutations revealed that AM14 recognizes a quaternary epitope that spans two protomers and includes a region that undergoes extensive conformational changes in the pre- to postfusion F transition. Binding assays demonstrated that AM14 is unique in its specific recognition of trimeric furin-cleaved prefusion F, which is the mature form of F on infectious virions. These results demonstrate that the prefusion F trimer contains potent neutralizing epitopes not present on monomers and that AM14 should be particularly useful for characterizing the conformational state of RSV F-based vaccine antigens.


Subject(s)
Antibodies, Neutralizing/ultrastructure , Antibodies, Viral/ultrastructure , Epitopes, B-Lymphocyte/ultrastructure , Respiratory Syncytial Viruses/immunology , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , Antigens, Viral/immunology , Cell Line , Chromatography, Gel , Crystallography, X-Ray , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/immunology , Flow Cytometry , Glycoproteins/chemistry , Glycoproteins/immunology , Glycoproteins/ultrastructure , Humans , Protein Structure, Quaternary , Surface Plasmon Resonance
3.
J Virol ; 84(16): 8132-40, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20519399

ABSTRACT

Respiratory syncytial virus (RSV) is a major cause of virus-induced respiratory disease and hospitalization in infants. Palivizumab, an RSV-neutralizing monoclonal antibody, is used clinically to prevent serious RSV-related respiratory disease in high-risk infants. Motavizumab, an affinity-optimized version of palivizumab, was developed to improve protection against RSV. These antibodies bind RSV F protein, which plays a role in virus attachment and mediates fusion. Determining how these antibodies neutralize RSV is important to help guide development of new antibody drugs against RSV and, potentially, other viruses. This study aims to uncover the mechanism(s) by which palivizumab and motavizumab neutralize RSV. Assays were developed to test the effects of these antibodies at distinct steps during RSV replication. Pretreatment of virus with palivizumab or motavizumab did not inhibit virus attachment or the ability of F protein to interact with the target cell membrane. However, pretreatment of virus with either of these antibodies resulted in the absence of detectable viral transcription. These results show that palivizumab and motavizumab act at a point after F protein initiates interaction with the cell membrane and before virus transcription. Palivizumab and motavizumab also inhibited F protein-mediated cell-to-cell fusion. Therefore, these results strongly suggest that these antibodies block both cell-to-cell and virus-to-cell fusion, since these processes are likely similar. Finally, palivizumab and motavizumab did not reduce viral budding. Based on models developed from numerous studies of viral fusion proteins, our results indicate that these antibodies may prevent conformational changes in F protein required for the fusion process.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antiviral Agents/pharmacology , Respiratory Syncytial Virus, Human/drug effects , Virus Internalization/drug effects , Animals , Antibodies, Monoclonal, Humanized , Antibodies, Viral/pharmacology , Cell Fusion , Cell Line , Chlorocebus aethiops , Humans , Palivizumab , Protein Binding , Respiratory Syncytial Virus, Human/immunology , Viral Fusion Proteins/immunology , Viral Fusion Proteins/metabolism , Virus Attachment/drug effects , Virus Replication/drug effects
4.
Virus Genes ; 40(2): 212-21, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20111897

ABSTRACT

Studies of the fusion activity of respiratory syncytial virus (RSV) F protein are significantly hindered by low recombinant expression levels. While infection produces F protein levels detectable by western blot, recombinant expression produces undetectable to low levels of F protein. Identifying the obstacles that hinder recombinant F protein expression may lead to improved expression and facilitate the study of F protein function. We hypothesized that nuclear localization and/or inefficient RNA polymerase II-mediated transcription contribute to poor recombinant F protein expression. This study shows a combination of stalled nuclear export, premature polyadenylation, and low mRNA abundance all contribute to low recombinant F protein expression levels. In addition, this study provides an expression optimization strategy that results in greater F protein expression levels than observed by codon-optimization of the F protein gene, which will be useful for future studies of F protein function.


Subject(s)
Active Transport, Cell Nucleus , Gene Expression , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , RNA, Viral/metabolism , Viral Fusion Proteins/biosynthesis , Animals , Cell Line , Humans , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Viral Fusion Proteins/genetics
5.
Sci Transl Med ; 9(388)2017 05 03.
Article in English | MEDLINE | ID: mdl-28469033

ABSTRACT

Prevention of respiratory syncytial virus (RSV) illness in all infants is a major public health priority. However, no vaccine is currently available to protect this vulnerable population. Palivizumab, the only approved agent for RSV prophylaxis, is limited to high-risk infants, and the cost associated with the requirement for dosing throughout the RSV season makes its use impractical for all infants. We describe the development of a monoclonal antibody as potential RSV prophylaxis for all infants with a single intramuscular dose. MEDI8897*, a highly potent human antibody, was optimized from antibody D25, which targets the prefusion conformation of the RSV fusion (F) protein. Crystallographic analysis of Fab in complex with RSV F from subtypes A and B reveals that MEDI8897* binds a highly conserved epitope. MEDI8897* neutralizes a diverse panel of RSV A and B strains with >50-fold higher activity than palivizumab. At similar serum concentrations, prophylactic administration of MEDI8897* was ninefold more potent than palivizumab at reducing pulmonary viral loads by >3 logs in cotton rats infected with either RSV A or B subtypes. MEDI8897 was generated by the introduction of triple amino acid substitutions (YTE) into the Fc domain of MEDI8897*, which led to more than threefold increased half-life in cynomolgus monkeys compared to non-YTE antibody. Considering the pharmacokinetics of palivizumab in infants, which necessitates five monthly doses for protection during an RSV season, the high potency and extended half-life of MEDI8897 support its development as a cost-effective option to protect all infants from RSV disease with once-per-RSV-season dosing in the clinic.


Subject(s)
Palivizumab/therapeutic use , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/therapeutic use , Respiratory Syncytial Viruses/pathogenicity , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Antiviral Agents/pharmacokinetics , Antiviral Agents/therapeutic use , Female , Humans , Infant , Infant, Newborn , Male , Palivizumab/pharmacokinetics , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Viruses/drug effects
6.
Mol Immunol ; 41(10): 985-1000, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15302161

ABSTRACT

A panel of anti-human CD2 monoclonal antibodies (mAb) and soluble human CD58 (LFA-3) were tested for binding to human peripheral blood mononuclear cells (PBMCs), recombinant human CD2 and mononuclear cells from Cynomolgus, Rhesus and African green monkey, Stump-tail, Pig-tail and Assamese macaque, Chimpanzee and Baboon. This analysis revealed that whilst some antibodies recognized all species, there were differential binding profiles with others. Three antibodies, MEDI-507, 6F10.3 and 4B2, recognized CD2 from human and Chimpanzee but not that from the other primates. We have cloned eight of the previously unknown primate CD2 molecules and report here their sequences for the first time. This analysis revealed that 12 amino acids formed a common set of residues in the extra cellular domain of human and Chimpanzee CD2. Using a "knock-in" mutagenesis approach starting with Baboon CD2, which does not bind MEDI-507, 6F10.3 and 4B2, we have identified three residues in the adhesion domain of human CD2 which are critical for its binding to these mAbs. These residues, N18, K55 and T59 define a region located outside of the previously described binding regions on CD2. Affinity measurements of the mutants revealed a variety of degrees of binding restoration for MEDI-507, 6F10.3 and 4B2, indicating that there are fine differences within a given epitope. Furthermore, the analysis of the competition of several of the anti-human CD2 antibodies with each other and CD58 demonstrated the existence of a continuum of overlapping epitopes on human CD2, which is in contrast to the commonly held belief that epitopes on human CD2 are clearly segregated.


Subject(s)
Antibodies/immunology , CD2 Antigens/immunology , Animals , CD2 Antigens/chemistry , CD2 Antigens/genetics , CD58 Antigens/immunology , Epitope Mapping , Haplorhini/genetics , Haplorhini/immunology , Humans , Pan troglodytes/genetics , Pan troglodytes/immunology , Sequence Analysis, Protein , T-Lymphocytes/immunology
7.
J Gen Virol ; 89(Pt 12): 3113-3118, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19008400

ABSTRACT

Human metapneumovirus (hMPV) is genetically related to respiratory syncytial virus (RSV); both cause respiratory tract illnesses ranging from a mild cough to bronchiolitis and pneumonia. The F protein-directed monoclonal antibody (mAb) palivizumab has been shown to prevent severe lower respiratory tract RSV infection in animals and humans. We have previously reported on a panel of mAbs against the hMPV F protein that neutralize hMPV in vitro and, in two cases, in vivo. Here we describe the generation of hMPV mAb-resistant mutants (MARMs) to these neutralizing antibodies. Sequencing the F proteins of the hMPV MARMs identified several neutralizing epitopes. Interestingly, some of the epitopes mapped on the hMPV F protein coincide with homologous regions mapped previously on the RSV F protein, including the site against which the broadly protective mAb palivizumab is directed. This suggests that these homologous regions play important, conserved functions in both viruses.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Metapneumovirus/immunology , Viral Fusion Proteins/immunology , Antibodies, Monoclonal/biosynthesis , Antibodies, Viral/biosynthesis , Epitope Mapping , Epitopes/chemistry , Epitopes/immunology , Humans , Metapneumovirus/genetics , Mutation , Neutralization Tests , Structure-Activity Relationship , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics
8.
J Gen Virol ; 88(Pt 10): 2702-2709, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17872522

ABSTRACT

Human metapneumovirus (hMPV), a newly discovered paramyxovirus, is associated with acute respiratory-tract illness, primarily in young children, individuals with underlying disease and the elderly. Two genetic lineages of hMPV circulate around the world, and viruses from these two lineages demonstrate antigenic differences. The clinical impact of hMPV warrants the development of vaccines. Recombinant soluble fusion (F) proteins of prototype viruses of the two main lineages of hMPV that can be produced in high yields have been constructed. In this study, the antigenicity, immunogenicity and protective efficacy of these soluble F subunit vaccines were evaluated in Syrian golden hamsters (Mesocricetus auratus). Immunization of hamsters with the soluble F proteins, adjuvanted with Specol or iscom matrix, induced high virus-neutralization titres, with higher titres against the homologous than the heterologous virus. The neutralizing antibodies protected from subsequent infection of the lungs with both homologous and heterologous virus. Upon challenge, viral titres in the nasal turbinates of immunized animals were reduced significantly compared with those of PBS-immunized animals. In conclusion, a soluble F subunit vaccine for hMPV that induces cross-protective immunity for infection of the lower respiratory tract in Syrian golden hamsters has been generated.


Subject(s)
Metapneumovirus/immunology , Respiratory Tract Infections/immunology , Viral Vaccines/administration & dosage , Animals , Base Sequence , Child , Chlorocebus aethiops , Cricetinae , Haplorhini , Humans , Mesocricetus , Metapneumovirus/genetics , Molecular Sequence Data , Plasmids , Restriction Mapping , Species Specificity , Vero Cells , Viral Proteins/genetics , Viral Proteins/immunology
9.
J Virol ; 80(16): 7799-806, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16873237

ABSTRACT

Human metapneumovirus (hMPV) is a recently described member of the Paramyxoviridae family/Pneumovirinae subfamily and shares many common features with respiratory syncytial virus (RSV), another member of the same subfamily. hMPV causes respiratory tract illnesses that, similar to human RSV, occur predominantly during the winter months and have symptoms that range from mild to severe cough, bronchiolitis, and pneumonia. Like RSV, the hMPV virus can be subdivided into two genetic subgroups, A and B. With RSV, a single monoclonal antibody directed at the fusion (F) protein can prevent severe lower respiratory tract RSV infection. Because of the high level of sequence conservation of the F protein across all the hMPV subgroups, this protein is likely to be the preferred antigenic target for the generation of cross-subgroup neutralizing antibodies. Here we describe the generation of a panel of neutralizing monoclonal antibodies that bind to the hMPV F protein. A subset of these antibodies has the ability to neutralize prototypic strains of both the A and B hMPV subgroups in vitro. Two of these antibodies exhibited high-affinity binding to the F protein and were shown to protect hamsters against infection with hMPV. The data suggest that a monoclonal antibody could be used prophylactically to prevent lower respiratory tract disease caused by hMPV.


Subject(s)
Antibodies, Monoclonal/isolation & purification , Antibodies, Viral/isolation & purification , Metapneumovirus/immunology , Paramyxoviridae Infections/prevention & control , Respiratory Tract Infections/prevention & control , Viral Fusion Proteins/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Viral/pharmacology , Antibodies, Viral/therapeutic use , Cells, Cultured , Humans , Respiratory Tract Infections/virology , Viral Fusion Proteins/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL