Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
PLoS Pathog ; 11(7): e1005035, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26161532

RESUMO

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.


Assuntos
Anticorpos Neutralizantes/ultraestrutura , Anticorpos Antivirais/ultraestrutura , Epitopos de Linfócito B/ultraestrutura , Vírus Sinciciais Respiratórios/imunologia , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/química , Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Linhagem Celular , Cromatografia em Gel , Cristalografia por Raios X , Ensaio de Imunoadsorção Enzimática , Mapeamento de Epitopos , Epitopos de Linfócito B/química , Epitopos de Linfócito B/imunologia , Citometria de Fluxo , Glicoproteínas/química , Glicoproteínas/imunologia , Glicoproteínas/ultraestrutura , Humanos , Estrutura Quaternária de Proteína , Ressonância de Plasmônio de Superfície
2.
Antimicrob Agents Chemother ; 57(12): 6147-53, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24080653

RESUMO

The study objective was to evaluate the pharmacokinetics (PK), antidrug antibody (ADA), and safety of motavizumab-YTE (motavizumab with amino acid substitutions M252Y/S254T/T256E [YTE]), an Fc-modified anti-respiratory syncytial virus (RSV) monoclonal antibody. Healthy adults (n = 31) were randomized to receive a single intravenous (i.v.) dose of motavizumab-YTE or motavizumab (0.3, 3, 15, or 30 mg/kg) and followed for 240 days. Clearance of motavizumab-YTE was significantly lower (71% to 86%) and the half-life (t1/2) was 2- to 4-fold longer than with motavizumab. However, similar peak concentrations and volume-of-distribution values, indicative of similar distribution properties, were seen at all dose levels. The sustained serum concentrations of motavizumab-YTE were fully functional, as shown by RSV neutralizing activity that persisted for 240 days with motavizumab-YTE versus 90 days postdose for motavizumab. Safety and incidence of ADA were comparable between groups. In this first study of an Fc-modified monoclonal antibody in humans, motavizumab-YTE was well tolerated and exhibited an extended half-life of up to 100 days. (This study has been registered at ClinicalTrials.gov under registration no. NCT00578682.).


Assuntos
Anticorpos Monoclonais Humanizados/farmacocinética , Antivirais/farmacocinética , Drogas em Investigação/farmacocinética , Fragmentos Fc das Imunoglobulinas/química , Adulto , Anticorpos Monoclonais Humanizados/sangue , Antivirais/sangue , Método Duplo-Cego , Feminino , Meia-Vida , Humanos , Injeções Intravenosas , Masculino , Taxa de Depuração Metabólica
3.
J Infect Dis ; 205(4): 635-8, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22184728

RESUMO

Specific mutations in respiratory syncytial virus (RSV) fusion protein can cause palivizumab resistance. We assessed the incidence of sequence polymorphisms and palivizumab resistance in clinical RSV isolates collected from immunoprophylaxis-naive subjects. Polymorphisms were identified at low frequency, and only polymorphic mutations in antigenic site A (<1% of all polymorphisms) conferred palivizumab resistance.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Antivirais/farmacologia , Farmacorresistência Viral , Mutação de Sentido Incorreto , Vírus Sinciciais Respiratórios/genética , Proteínas Virais de Fusão/genética , Antígenos Virais/genética , Antígenos Virais/imunologia , Pré-Escolar , Epitopos/genética , Humanos , Lactente , Recém-Nascido , Palivizumab , Polimorfismo Genético , Proteínas Virais de Fusão/imunologia
4.
J Infect Dis ; 203(5): 674-82, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21208913

RESUMO

BACKGROUND: Palivizumab is a US Food and Drug Administration-approved monoclonal antibody for the prevention of respiratory syncytial virus (RSV) lower respiratory disease in high-risk infants. Motavizumab, derived from palivizumab with enhanced antiviral activity, has recently been tested in humans. Although palivizumab escape mutants have been generated in the laboratory, the development of resistant RSV in patients receiving palivizumab has not been reported previously. METHODS: We generated palivizumab and motavizumab escape mutants in vitro and examined the development of resistant mutants in RSV-breakthrough patients receiving immunoprophylaxis. The effect of these mutations on neutralization by palivizumab and motavizumab and in vitro fitness was studied. RESULTS: Antibody-resistant RSV variants selected in vitro had mutations at position 272 of the fusion protein, from lysine to asparagine, methionine, threonine, glutamine, or glutamate. Variants containing mutations at positions 272 and 275 were detected in breakthrough patients. All these variants were resistant to palivizumab, but only the glutamate variant at position 272 demonstrated resistance to motavizumab. Mixtures of wild-type and variant RSV soon lost the resistant phenotype in the absence of selection. CONCLUSIONS: Resistant RSV variants were detected in a small subset (∼ 5%) of RSV breakthrough cases. The fitness of these variants was impaired, compared to wild-type RSV.


Assuntos
Anticorpos Monoclonais/imunologia , Antivirais/imunologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vírus Sinciciais Respiratórios/imunologia , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Anticorpos Antivirais , Farmacorresistência Viral/genética , Farmacorresistência Viral/imunologia , Humanos , Lactente , Dados de Sequência Molecular , Mutação , Mucosa Nasal/virologia , Palivizumab , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Vírus Sinciciais Respiratórios/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
J Mol Biol ; 368(3): 652-65, 2007 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-17362988

RESUMO

Respiratory syncytial virus (RSV) is the leading cause of viral bronchiolitis and pneumonia in infants and children. Currently, palivizumab is the only approved monoclonal antibody (mAb) for prophylaxis of RSV. However, a small percentage of patients are not protected by palivizumab; in addition, palivizumab does not inhibit RSV replication effectively in the upper respiratory tract. We report here the development and characterization of motavizumab, an ultra-potent, affinity-matured, humanized mAb derived from palivizumab. Several palivizumab variants that enhanced the neutralization of RSV in vitro by up to 44-fold were generated; however, in vivo prophylaxis of cotton rats with these antibodies conferred only about a twofold improvement in potency over palivizumab. This unexpected small increase of in vivo potency was caused by poor serum pharmacokinetics and lung bio-availability that resulted from unexpectedly broad tissue binding. Subsequent analyses revealed that changes at three amino acids arising from the affinity maturation markedly increased the non-specific binding to various tissues. Our results suggested that k(on)-driven mutations are more likely to initiate non-specific binding events than k(off)-driven mutations. Reversion of these three residues to the original sequences greatly diminished the tissue binding. The resulting mAb, motavizumab, binds to RSV F protein 70-fold better than palivizumab, and exhibits about a 20-fold improvement in neutralization of RSV in vitro. In cotton rats, at equivalent concentrations, motavizumab reduced pulmonary RSV titers to up to 100-fold lower levels than did palivizumab and, unlike palivizumab, motavizumab very potently inhibited viral replication in the upper respiratory tract. This affinity-enhanced mAb is being investigated in pivotal clinical trials. Importantly, our engineering process offers precious insights into the improvement of other therapeutic mAbs.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antivirais/uso terapêutico , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Sistema Respiratório/virologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais Humanizados , Antivirais/farmacocinética , Linhagem Celular , Reações Cruzadas , Feminino , Humanos , Pulmão/metabolismo , Macaca fascicularis , Dados de Sequência Molecular , Mutação , Testes de Neutralização , Palivizumab , Pan troglodytes , Vírus Sinciciais Respiratórios/fisiologia , Sistema Respiratório/efeitos dos fármacos , Sistema Respiratório/imunologia , Sigmodontinae , Distribuição Tecidual , Replicação Viral/efeitos dos fármacos
6.
Sci Transl Med ; 9(388)2017 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-28469033

RESUMO

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.


Assuntos
Palivizumab/uso terapêutico , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/uso terapêutico , Vírus Sinciciais Respiratórios/patogenicidade , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Antivirais/farmacocinética , Antivirais/uso terapêutico , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Palivizumab/farmacocinética , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Vírus Sinciciais Respiratórios/efeitos dos fármacos
7.
J Mol Biol ; 350(1): 126-44, 2005 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15907931

RESUMO

We describe here the selection of ultra-potent anti-respiratory syncytial virus (RSV) antibodies for preventing RSV infection. A large number of antibody variants derived from Synagis (palivizumab), an anti-RSV monoclonal antibody that targets RSV F protein, were generated by a directed evolution approach that allowed convenient manipulation of the binding kinetics. Palivizumab variants with about 100-fold slower dissociation rates or with fivefold faster association rates were identified and tested for their ability to neutralize virus in a microneutralization assay. Our data reveal a major differential effect of the association and dissociation rates on the RSV neutralization, particularly for intact antibodies wherein the association rate plays the predominant role. Furthermore, we found that antibody binding valence also plays a critical role in mediating the viral neutralization through a mechanism that is likely unrelated to antibody size or binding avidity. We applied an iterative mutagenesis approach, and thereafter were able to identify palivizumab Fab variants with up to 1500-fold improvement and palivizumab IgG variants with up to 44-fold improvement in the ability to neutralize RSV. These anti-RSV antibodies likely will offer great clinical potential for RSV immunoprophylaxis. In addition, our findings provide insights into engineering potent antibody therapeutics for other disease targets.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/genética , Anticorpos Monoclonais Humanizados , Anticorpos Antivirais/química , Anticorpos Antivirais/genética , Linhagem Celular , Chlorocebus aethiops , Ensaio de Imunoadsorção Enzimática , Humanos , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Mutação/genética , Testes de Neutralização , Palivizumab , Estrutura Terciária de Proteína , Infecções por Vírus Respiratório Sincicial/genética , Vírus Sinciciais Respiratórios/genética
8.
J Gen Virol ; 89(Pt 12): 3113-3118, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19008400

RESUMO

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.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Metapneumovirus/imunologia , Proteínas Virais de Fusão/imunologia , Anticorpos Monoclonais/biossíntese , Anticorpos Antivirais/biossíntese , Mapeamento de Epitopos , Epitopos/química , Epitopos/imunologia , Humanos , Metapneumovirus/genética , Mutação , Testes de Neutralização , Relação Estrutura-Atividade , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/genética
9.
J Virol ; 80(16): 7799-806, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16873237

RESUMO

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.


Assuntos
Anticorpos Monoclonais/isolamento & purificação , Anticorpos Antivirais/isolamento & purificação , Metapneumovirus/imunologia , Infecções por Paramyxoviridae/prevenção & controle , Infecções Respiratórias/prevenção & controle , Proteínas Virais de Fusão/imunologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Antivirais/farmacologia , Anticorpos Antivirais/uso terapêutico , Células Cultivadas , Humanos , Infecções Respiratórias/virologia , Proteínas Virais de Fusão/antagonistas & inibidores
10.
J Immunol ; 169(9): 5171-80, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12391234

RESUMO

Many biological functions, including control of the homeostasis and maternofetal transfer of serum gamma-globulins, are mediated by the MHC class I-related neonatal FcR (FcRn). A correlation exists in mice between the binding affinity of IgG1/Fc fragments to FcRn at pH 6.0 and their serum t(1/2). To expand this observation, phage display of mutagenized Fc fragments derived from a human IgG1 was used to increase their affinity to both murine and human FcRn. Ten variants were identified that have a higher affinity toward murine and human FcRn at pH 6.0, with DeltaDeltaG (DeltaG(wild type) - DeltaG(mutant)) from 1.0 to 2.0 kcal/mol and from 0.6 to 2.4 kcal/mol, respectively. Those variants exhibit a parallel increase in binding at pH 7.4 to murine, but not human, FcRn. Although not degraded in blood in vitro, accumulated in tissues, nor excreted in urine, their serum concentration in mice is decreased. We propose that higher affinity to FcRn at pH 7.4 adversely affects release into the serum and offsets the benefit of the enhanced binding at pH 6.0.


Assuntos
Animais Recém-Nascidos/imunologia , Sítios de Ligação de Anticorpos , Receptores Fc/metabolismo , Animais , Animais Recém-Nascidos/genética , Bacteriófago M13/genética , Bacteriófago M13/imunologia , Sítios de Ligação de Anticorpos/genética , Antígenos de Histocompatibilidade Classe I , Humanos , Concentração de Íons de Hidrogênio , Imunoglobulina G/administração & dosagem , Imunoglobulina G/sangue , Imunoglobulina G/genética , Imunoglobulina G/metabolismo , Recém-Nascido , Injeções Intramusculares , Injeções Intravenosas , Camundongos , Camundongos Endogâmicos BALB C , Mutagênese Sítio-Dirigida , Especificidade de Órgãos/genética , Especificidade de Órgãos/imunologia , Biblioteca de Peptídeos , Testes de Precipitina , Receptores Fc/administração & dosagem , Receptores Fc/sangue , Receptores Fc/genética , Receptores de IgG/administração & dosagem , Receptores de IgG/sangue , Receptores de IgG/genética , Receptores de IgG/metabolismo , Ressonância de Plasmônio de Superfície
11.
J Infect Dis ; 190(5): 975-8, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15295704

RESUMO

Premature infants and those with chronic lung disease or congenital heart disease are at high risk of severe respiratory syncytial virus (RSV) disease. Palivizumab (Synagis), a humanized anti-RSV monoclonal antibody, has been used extensively since 1998 to prevent severe RSV disease in high-risk infants. To monitor for possible palivizumab-resistant mutants, an immunofluorescence binding assay that predicts palivizumab neutralization of RSV was developed. RSV isolates were collected at 8 US sites from 458 infants hospitalized for RSV disease (1998-2002). Palivizumab bound to all 371 RSV isolates able to be evaluated, including 25 from active-palivizumab recipients. The palivizumab epitope appears to be highly conserved, even in infants receiving prophylaxis with palivizumab.


Assuntos
Anticorpos Monoclonais/farmacologia , Antivirais/farmacologia , Farmacorresistência Viral , Hospitalização , Mutação , Vírus Sincicial Respiratório Humano/efeitos dos fármacos , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Antivirais/uso terapêutico , Linhagem Celular , Imunofluorescência , Humanos , Lactente , Testes de Sensibilidade Microbiana , Testes de Neutralização , Palivizumab , Infecções por Vírus Respiratório Sincicial/epidemiologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Infecções por Vírus Respiratório Sincicial/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA