Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
2.
Virology ; 426(2): 100-5, 2012 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-22341308

RESUMO

Currently there are no licensed antiviral treatments for the Alphaviruses Venezuelan equine encephalitis virus (VEEV), Everglades virus and Mucambo virus. We previously developed a humanised version of the mouse monoclonal antibody 1A3B-7 (Hu1A3B-7) which exhibited a wide range of reactivity in vitro and was able to protect mice from infection with VEEV. Continued work with the humanised antibody has now demonstrated that it has the potential to be a new human therapeutic. Hu1A3B-7 successfully protected mice from infection with multiple Alphaviruses. The effectiveness of the humanisation process was determined by assessing proliferation responses in human T-cells to peptides derived from the murine and humanised versions of the V(H) and V(L) domains. This analysis showed that the number of human T-cell epitopes within the humanised antibody had been substantially reduced, indicating that Hu1A3B-7 may have reduced immunogenicity in vivo.


Assuntos
Infecções por Alphavirus/prevenção & controle , Alphavirus/imunologia , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Antivirais/imunologia , Vírus da Encefalite Equina Venezuelana/imunologia , Microbiologia do Ar , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/virologia , Sequência de Aminoácidos , Animais , Encefalomielite Equina Venezuelana/imunologia , Encefalomielite Equina Venezuelana/prevenção & controle , Encefalomielite Equina Venezuelana/virologia , Humanos , Imunização Passiva , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular
3.
J Med Microbiol ; 61(Pt 1): 8-15, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21852521

RESUMO

Normal immunocompetent mice are not susceptible to non-adapted filoviruses. There are therefore two strategies available to establish a murine model of filovirus infection: adaptation of the virus to the host or the use of genetically modified mice that are susceptible to the virus. A number of knockout (KO) strains of mice with defects in either their adaptive or innate immunity are susceptible to non-adapted filoviruses. In this study, A129 α/ß -/- interferon receptor-deficient KO mice, strain A129 IFN-α/ß -/-, were used to determine the lethality of a range of filoviruses, including Lake Victoria marburgvirus (MARV), Zaire ebolavirus (ZEBOV), Sudan ebolavirus (SEBOV), Reston ebolavirus (REBOV) and Côte d'Ivoire ebolavirus (CIEBOV), administered by using intraperitoneal (IP) or aerosol routes of infection. One hundred percent mortality was observed in all groups of KO mice that were administered with a range of challenge doses of MARV and ZEBOV by either IP or aerosol routes. Mean time to death for both routes was dose-dependent and ranged from 5.4 to 7.4 days in the IP injection challenge, and from 10.2 to 13 days in the aerosol challenge. The lethal dose (50 % tissue culture infective dose, TCID(50)) of ZEBOV for KO mice was <1 TCID(50) ml(-1) when administered by either the IP or aerosol route of infection; for MARV the lethal dose was <1 TCID(50) ml(-1) by the IP route of infection and <10 TCID(50) ml(-1) by the aerosol route. In contrast, there was no mortality after infection with SEBOV or REBOV by either IP or aerosol routes of infection; all the mice lost weight (~15 % loss of group mean body weight with SEBOV and ~7 % with REBOV) but recovered to their original weights by day 14 post-challenge. There was no mortality in mice administered with CIEBOV via the IP route of infection and no clinical signs of infection were observed. The progression of disease was faster following infection with ZEBOV than with MARV but ultimately both viruses caused widespread infection with high titres of the infectious viruses in multiple organs. Histopathological observations were consistent with other animal models and showed widespread organ damage. This study suggests that MARV and ZEBOV are more virulent when administered via the IP route rather than by aerosol infection, although both are highly virulent by either route. The KO mouse may provide a useful model to test potential antiviral therapeutics against wild-type filoviruses.


Assuntos
Aerossóis , Modelos Animais de Doenças , Infecções por Filoviridae/mortalidade , Infecções por Filoviridae/fisiopatologia , Filoviridae/patogenicidade , Receptor de Interferon alfa e beta/genética , Animais , Ebolavirus/patogenicidade , Feminino , Filoviridae/classificação , Infecções por Filoviridae/virologia , Doença pelo Vírus Ebola/mortalidade , Doença pelo Vírus Ebola/fisiopatologia , Doença pelo Vírus Ebola/virologia , Humanos , Injeções Intraperitoneais , Masculino , Doença do Vírus de Marburg/mortalidade , Doença do Vírus de Marburg/fisiopatologia , Doença do Vírus de Marburg/virologia , Marburgvirus/patogenicidade , Camundongos , Camundongos Knockout , Receptor de Interferon alfa e beta/deficiência , Virulência
4.
Antiviral Res ; 90(1): 1-8, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21310183

RESUMO

In murine models of Venezuelan equine encephalitis virus (VEEV) infection, the neutralising monoclonal antibody 1A3B-7 has been shown to be effective in passive protection from challenge by the aerosol route with serogroups I, II and Mucambo virus (formally VEE complex subtype IIIA). This antibody is able to bind to all serogroups of the VEEV complex when used in ELISA and therefore is an excellent candidate for protein engineering in order to derive a humanised molecule suitable for therapeutic use in humans. A Complementarity Determining Region (CDR) grafting approach using human germline IgG frameworks was used to produce a panel of humanised variants of 1A3B-7, from which a single candidate molecule with retained binding specificity was identified. Evaluation of humanised 1A3B-7 (Hu1A3B-7) in in vitro studies indicated that Hu1A3B-7 retained both broad specificity and neutralising activity. Furthermore, in vivo experiments showed that Hu1A3B-7 successfully protected mice against lethal subcutaneous and aerosol challenges with VEEV strain TrD (serogroup I). Hu1A3B-7 is therefore a promising candidate for the future development of a broad-spectrum antiviral therapy to treat VEEV disease in humans.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Anticorpos Neutralizantes/administração & dosagem , Produtos Biológicos/administração & dosagem , Vírus da Encefalite Equina Venezuelana/imunologia , Encefalomielite Equina Venezuelana/prevenção & controle , Imunoterapia/métodos , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Produtos Biológicos/imunologia , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos BALB C
5.
Vaccine ; 28(34): 5558-64, 2010 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-20600509

RESUMO

A recombinant humanized antibody to Venezuelan equine encephalitis virus (VEEV) was constructed in a monocistronic adenoviral expression vector with a foot-and-mouth-disease virus-derived 2A self-cleavage oligopeptide inserted between the antibody heavy and light chains. After expression in mammalian cells, the heavy and light chains of the humanized antibody (hu1A4A1IgG1-2A) were completely cleaved and properly dimerized. The purified hu1A4A1IgG1-2A retained VEEV binding affinity and neutralizing activity similar to its parental murine antibody. The half-life of hu1A4A1IgG1-2A in mice was approximately 2 days. Passive immunization of hu1A4A1IgG1-2A in mice (50 microg/mouse) 24 h before or after virulent VEEV challenge provided complete protection, indicating that hu1A4A1IgG1-2A has potent prophylactic and therapeutic effects against VEEV infection.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Especificidade de Anticorpos , Encefalomielite Equina Venezuelana/prevenção & controle , Imunização Passiva , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/imunologia , Linhagem Celular , Vírus da Encefalite Equina Venezuelana/imunologia , Meia-Vida , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Proteínas Virais/imunologia
6.
Virol J ; 6: 206, 2009 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-19925641

RESUMO

BACKGROUND: There is currently a requirement for antiviral therapies capable of protecting against infection with Venezuelan equine encephalitis virus (VEEV), as a licensed vaccine is not available for general human use. Monoclonal antibodies are increasingly being developed as therapeutics and are potential treatments for VEEV as they have been shown to be protective in the mouse model of disease. However, to be truly effective, the antibody should recognise multiple strains of VEEV and broadly reactive monoclonal antibodies are rarely and only coincidentally isolated using classical hybridoma technology. RESULTS: In this work, methods were developed to reliably derive broadly reactive murine antibodies. A phage library was created that expressed single chain variable fragments (scFv) isolated from mice immunised with multiple strains of VEEV. A broadly reactive scFv was identified and incorporated into a murine IgG2a framework. This novel antibody retained the broad reactivity exhibited by the scFv but did not possess virus neutralising activity. However, the antibody was still able to protect mice against VEEV disease induced by strain TrD when administered 24 h prior to challenge. CONCLUSION: A monoclonal antibody possessing reactivity to a wide range of VEEV strains may be of benefit as a generic antiviral therapy. However, humanisation of the murine antibody will be required before it can be tested in humans.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Vírus da Encefalite Equina Venezuelana/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Antivirais/uso terapêutico , Linhagem Celular , Chlorocebus aethiops , Encefalomielite Equina Venezuelana/prevenção & controle , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Testes de Neutralização , Biblioteca de Peptídeos
7.
Virol J ; 6: 118, 2009 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-19646224

RESUMO

BACKGROUND: Optimisation of genes has been shown to be beneficial for expression of proteins in a range of applications. Optimisation has increased protein expression levels through improved codon usage of the genes and an increase in levels of messenger RNA. We have applied this to an adenovirus (ad)-based vaccine encoding structural proteins (E3-E2-6K) of Venezuelan equine encephalitis virus (VEEV). RESULTS: Following administration of this vaccine to Balb/c mice, an approximately ten-fold increase in antibody response was elicited and increased protective efficacy compared to an ad-based vaccine containing non-optimised genes was observed after challenge. CONCLUSION: This study, in which the utility of optimising genes encoding the structural proteins of VEEV is demonstrated for the first time, informs us that including optimised genes in gene-based vaccines for VEEV is essential to obtain maximum immunogenicity and protective efficacy.


Assuntos
Adenoviridae/genética , Códon , Vírus da Encefalite Equina Venezuelana/imunologia , Vetores Genéticos , Proteínas Estruturais Virais/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Sequência de Bases , Vírus da Encefalite Equina Venezuelana/genética , Ensaio de Imunoadsorção Enzimática/métodos , Imunoglobulina G/sangue , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Análise de Sobrevida , Proteínas Estruturais Virais/biossíntese , Proteínas Estruturais Virais/genética , Vacinas Virais/genética
8.
Viral Immunol ; 21(4): 451-7, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19115934

RESUMO

An adenovirus-based (ad-based) vaccine delivering antigens from the Alphavirus Venezuelan equine encephalitis virus (VEEV) is a strategy that offers clinical potential. A vaccine against VEEV is desirable because of the re-emerging nature of this virus, and also the potential that it may be used as a biological weapon. This study was designed to investigate whether the co-administration of CpG oligodeoxynucleotides (ODNs) with an ad-based VEEV vaccine could enhance the protective efficacy of the vaccine. We report that the co-administration of CpG ODN was unable to increase VEEV-specific antibody responses in mice, and was unable to increase the protective efficacy of the vaccine against aerosol challenge with virulent VEEV. However, it was noted that antibody responses directed against the adenovirus vaccine vector were increased, which may be detrimental, particularly in the context of homologous boosting.


Assuntos
Adenoviridae/imunologia , Adjuvantes Imunológicos/administração & dosagem , Vírus da Encefalite Equina Venezuelana/imunologia , Encefalomielite Equina Venezuelana/imunologia , Vetores Genéticos/imunologia , Oligodesoxirribonucleotídeos/imunologia , Vacinas Virais/imunologia , Adenoviridae/genética , Animais , Anticorpos Antivirais/sangue , Vírus da Encefalite Equina Venezuelana/genética , Encefalomielite Equina Venezuelana/prevenção & controle , Encefalomielite Equina Venezuelana/virologia , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Imunoglobulina G/sangue , Camundongos , Camundongos Endogâmicos BALB C , Oligodesoxirribonucleotídeos/administração & dosagem , Transgenes/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
9.
Vaccine ; 24(17): 3440-5, 2006 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-16527377

RESUMO

There is a requirement for a vaccine that protects against the alphavirus, Venezuelan equine encephalitis virus (VEEV). Previous work has shown that DNA vaccines encoding structural proteins of VEEV can elicit immune responses and protection against VEEV though this protection is incomplete against airborne VEEV. In this study, we demonstrate that particle-mediated epidermal delivery of a DNA vaccine encoding the E2 glycoprotein of VEEV can be boosted with a mucosally-delivered Ad-based vaccine encoding the same E2 glycoprotein. This results in an improved Th2-type IgG response, an increase in neutralising antibody and a significant increase in protection against airborne VEEV. This indicates that prime-boost may be a suitable immunisation regimen for providing protection against airborne VEEV.


Assuntos
Vírus da Encefalite Equina Venezuelana/imunologia , Vacinas de DNA/administração & dosagem , Vacinas Sintéticas/administração & dosagem , Proteínas do Envelope Viral/genética , Vacinas Virais/administração & dosagem , Adenoviridae/genética , Administração Cutânea , Animais , Anticorpos Antivirais/sangue , Feminino , Imunização Secundária , Imunoglobulina G/sangue , Imunoglobulina G/classificação , Camundongos , Camundongos Endogâmicos BALB C , Vacinas de DNA/imunologia , Vacinas Sintéticas/imunologia , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia
10.
Adv Drug Deliv Rev ; 57(9): 1343-61, 2005 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-15935877

RESUMO

The advantages associated with DNA vaccines include the speed with which they may be constructed and produced at large-scale, the ability to produce a broad spectrum of immune responses, and the ability for delivery using non-invasive means. In addition, DNA vaccines may be manipulated to express multiple antigens and may be tailored for the induction of appropriate immune responses. These advantages make DNA vaccination a promising approach for the development of vaccines for biodefence. In this review, the potential of DNA vaccines for biodefence is discussed.


Assuntos
Infecções Bacterianas/prevenção & controle , Guerra Biológica , Sistemas de Liberação de Medicamentos/métodos , Tecnologia Farmacêutica/tendências , Vacinas de DNA , Viroses/prevenção & controle , Animais , Infecções Bacterianas/imunologia , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/genética , Vacinas Bacterianas/imunologia , Portadores de Fármacos , Humanos , Lipossomos , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Vacinas Virais/imunologia , Viroses/imunologia
11.
Genet Vaccines Ther ; 3(1): 3, 2005 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-15842732

RESUMO

BACKGROUND: Previously, antigens expressed from DNA vaccines have been fused to the VP22 protein from Herpes Simplex Virus type I in order to improve efficacy. However, the immune enhancing mechanism of VP22 is poorly understood and initial suggestions that VP22 can mediate intercellular spread have been questioned. Despite this, fusion of VP22 to antigens expressed from DNA vaccines has improved immune responses, particularly to non-secreted antigens. METHODS: In this study, we fused the gene for the VP22 protein to the gene for Protective Antigen (PA) from Bacillus anthracis, the causative agent of anthrax. Protective immunity against infection with B. anthracis is almost entirely based on a response to PA and we have generated two constructs, where VP22 is fused to either the N- or the C-terminus of the 63 kDa protease-cleaved fragment of PA (PA63). RESULTS: Following gene gun immunisation of A/J mice with these constructs, we observed no improvement in the anti-PA antibody response generated. Following an intraperitoneal challenge with 70 50% lethal doses of B. anthracis strain STI spores, no difference in protection was evident in groups immunised with the DNA vaccine expressing PA63 and the DNA vaccines expressing fusion proteins of PA63 with VP22. CONCLUSION: VP22 fusion does not improve the protection of A/J mice against live spore challenge following immunisation of DNA vaccines expressing PA63.

12.
Vaccine ; 23(16): 1931-40, 2005 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-15734065

RESUMO

In some species DNA vaccines elicit potent humoral and cellular immune responses. However, their performance in humans and non-human primates is less impressive. There are suggestions in the literature that an increase in the intercellular distribution of protein expressed from a DNA vaccine may enhance immunogenicity. We incorporated the Herpes Simplex Virus type 1 (HSV) VP22 gene, which encodes a protein that has been described as promoting intercellular spread, into a DNA vector in which it was fused to enhanced green fluorescent protein (EGFP). Following transfection of the plasmid DNA into mammalian cells, distribution of the fusion protein VP22-EGFP was not increased compared to EGFP alone. Furthermore, we found no evidence to suggest that VP22 was capable of mediating intercellular spread. However, when these constructs were used as DNA vaccines to immunise mice, antibody levels specific to EGFP were significantly enhanced when EGFP was fused to VP22. These data suggest that amplification of the immune response may occur via mechanisms other than VP22-mediated intercellular spread of antigen.


Assuntos
Adjuvantes Imunológicos , Vacinas de DNA/imunologia , Proteínas Estruturais Virais/farmacologia , Animais , Formação de Anticorpos/efeitos dos fármacos , Western Blotting , Células COS , Chlorocebus aethiops , Proteínas de Fluorescência Verde/imunologia , Metanol , Camundongos , Camundongos Endogâmicos BALB C , Plasmídeos/imunologia , Fixação de Tecidos , Transfecção , Proteínas Virais de Fusão/farmacologia
13.
J Med Virol ; 67(3): 384-93, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12116032

RESUMO

Venezuelan equine encephalitis virus (VEEV) replicates in lymphoid tissues following peripheral inoculation and a high titre viraemia develops. Encephalitis develops after the virus enters the central nervous system from the blood, with the earliest neuronal involvement being via the olfactory nerve. Following aerosol challenge with virulent VEEV, the virus is thought to replicate in the nasal mucosa and there could be direct entry into the olfactory nerve via infected neuroepithelial cells. Protection from VEEV infection is believed to be primarily mediated by virus specific antibody. The correlation between protection and neutralising serum antibody titres is, however, inconsistent when the virulent virus is administered by the airborne route. This study demonstrates a link between antibody in serum and the nasal mucosa and protection by means of passive immunisation studies. Intra-nasal administration of antibody increased protection against airborne virus in Balb/c mice. Vaccination of mu MT strain mice that do not have functional B cells and cannot produce antibody revealed normal proliferation of spleen cells in vitro and robust cytokine production. Aerosol challenge of mu MT mice demonstrated that complete protection was only achieved when passive immunisation with antibody was supplemented with active immunisation with the TC-83 vaccine strain of the virus. This implies that cell-mediated immune functions are required for protection against airborne challenge with virulent VEEV.


Assuntos
Anticorpos Antivirais/sangue , Vírus da Encefalite Equina Venezuelana/patogenicidade , Encefalomielite Equina Venezuelana/imunologia , Encefalomielite Equina Venezuelana/prevenção & controle , Mucosa Nasal/imunologia , Administração Intranasal , Animais , Anticorpos Antivirais/imunologia , Especificidade de Anticorpos , Linhagem Celular , Vírus da Encefalite Equina Venezuelana/imunologia , Encefalomielite Equina Venezuelana/virologia , Imunidade Celular , Imunidade nas Mucosas , Imunização Passiva , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Baço/citologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA