Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Vaccine ; 33(13): 1578-85, 2015 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-25698488

RESUMO

Therapeutic vaccinations have a potential application in infections where no curative treatment is available. In contrast to HIV, efficacious vaccines for a cat retrovirus, feline leukemia virus (FeLV), are commercially available. However, the infection is still prevalent, and no effective treatment of the infection is known. By vaccinating persistently FeLV-infected cats and presenting FeLV antigens to the immune system of the host, e.g., in the form of recombinant and/or adjuvanted antigens, we intended to shift the balance toward an advantage of the host so that persistent infection could be overcome by the infected cat. Two commercially available FeLV vaccines efficacious in protecting naïve cats from FeLV infection were tested in six experimentally and persistently FeLV-infected cats: first, a canarypox-vectored vaccine, and second, an adjuvanted, recombinant envelope vaccine was repeatedly administered with the aim to stimulate the immune system. No beneficial effects on p27 antigen and plasma viral RNA loads, anti-FeLV antibodies, or life expectancy of the cats were detected. The cats were unable to overcome or decrease viremia. Some cats developed antibodies to FeLV antigens although not protective. Thus, we cannot recommend vaccinating persistently FeLV-infected cats as a means of improving their FeLV status, quality of life or life expectancy. We suggest testing of all cats for FeLV infection prior to FeLV vaccination.


Assuntos
Doenças do Gato/terapia , Vírus da Leucemia Felina/imunologia , Infecções por Retroviridae/veterinária , Proteínas Oncogênicas de Retroviridae/uso terapêutico , Infecções Tumorais por Vírus/veterinária , Vacinas Sintéticas/uso terapêutico , Vacinas Virais/uso terapêutico , Animais , Anticorpos Antivirais/imunologia , Doenças do Gato/virologia , Gatos , Produtos do Gene gag/sangue , Vírus da Leucemia Felina/patogenicidade , Expectativa de Vida , Qualidade de Vida , Infecções por Retroviridae/terapia , Infecções por Retroviridae/virologia , Proteínas Oncogênicas de Retroviridae/administração & dosagem , Infecções Tumorais por Vírus/terapia , Infecções Tumorais por Vírus/virologia , Vacinação/veterinária , Vacinas Sintéticas/administração & dosagem , Carga Viral , Vacinas Virais/administração & dosagem , Viremia/terapia , Viremia/veterinária
4.
Vaccine ; 32(22): 2599-603, 2014 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-24662705

RESUMO

The aim of the study was to determine the efficacy of an inactivated feline leukemia virus (FeLV) vaccine (Versifel(®) FeLV, Zoetis.) compared to a recombinant FeLV vaccine (Purevax(®) FeLV, Merial Animal Health) in young cats, exposed under laboratory conditions to a highly virulent challenge model. The study was designed to be consistent with the general immunogenicity requirements of the European Pharmacopoeia 6.0 Monograph 01/2008:1321-Feline Leukaemia Vaccine (Inactivated) with the exception that commercial-strength vaccines were assessed. Fifty seronegative cats (8-9 weeks old) were vaccinated subcutaneously on two occasions, three weeks apart, with either placebo (treatment group T01), Versifel FeLV Vaccine (treatment group T02), or Purevax FeLV Vaccine (treatment group T03) according to the manufacturer's directions. Cats were challenged three weeks after the second vaccination with a virulent FeLV isolate (61E strain). Persistent FeLV antigenemia was determined from 3 to 15 weeks postchallenge. Bone marrow samples were tested for the presence of FeLV proviral DNA to determine FeLV latent infection. At week 15 after challenge with the virulent FeLV 61E strain, the Versifel FeLV Vaccine conferred 89.5% protection against FeLV persistent antigenemia and 94.7% protection against FeLV proviral DNA integration in bone marrow cells. In comparison, the Purevax FeLV Vaccine conferred 20% protection against FeLV persistent antigenemia and 35% protection against FeLV proviral DNA integration in bone marrow cells following challenge. The data from this study show that the Versifel FeLV Vaccine was efficacious in preventing both FeLV persistent p27 antigenemia and FeLV proviral DNA integration in bone marrow cells of cats challenged with this particular challenge model under laboratory conditions and provided better protection than Purevax FeLV in this experimental challenge model with highly virulent FeLV.


Assuntos
Antígenos Virais/sangue , Produtos do Gene gag/sangue , Leucemia Felina/prevenção & controle , Proteínas Oncogênicas de Retroviridae/administração & dosagem , Vacinação/veterinária , Vacinas Virais/administração & dosagem , Animais , Células da Medula Óssea/virologia , Gatos , DNA Viral/isolamento & purificação , Vírus da Leucemia Felina , Distribuição Aleatória , Vacinas de Produtos Inativados/administração & dosagem , Vacinas Sintéticas/administração & dosagem
5.
J Virol ; 88(2): 992-1001, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24198407

RESUMO

We previously delineated a highly conserved immunosuppressive (IS) domain within murine and primate retroviral envelope proteins that is critical for virus propagation in vivo. The envelope-mediated immunosuppression was assessed by the ability of the proteins, when expressed by allogeneic tumor cells normally rejected by engrafted mice, to allow these cells to escape, at least transiently, immune rejection. Using this approach, we identified key residues whose mutation (i) specifically abolishes immunosuppressive activity without affecting the "mechanical" function of the envelope protein and (ii) significantly enhances humoral and cellular immune responses elicited against the virus. The objective of this work was to study the immunosuppressive activity of the envelope protein (p15E) of feline leukemia virus (FeLV) and evaluate the effect of its abolition on the efficacy of a vaccine against FeLV. Here we demonstrate that the FeLV envelope protein is immunosuppressive in vivo and that this immunosuppressive activity can be "switched off" by targeted mutation of a specific amino acid. As a result of the introduction of the mutated envelope sequence into a previously well characterized canarypox virus-vectored vaccine (ALVAC-FeLV), the frequency of vaccine-induced FeLV-specific gamma interferon (IFN-γ)-producing cells was increased, whereas conversely, the frequency of vaccine-induced FeLV-specific interleukin-10 (IL-10)-producing cells was reduced. This shift in the IFN-γ/IL-10 response was associated with a higher efficacy of ALVAC-FeLV against FeLV infection. This study demonstrates that FeLV p15E is immunosuppressive in vivo, that the immunosuppressive domain of p15E can modulate the FeLV-specific immune response, and that the efficacy of FeLV vaccines can be enhanced by inhibiting the immunosuppressive activity of the IS domain through an appropriate mutation.


Assuntos
Vírus da Varíola dos Canários/genética , Produtos do Gene env/química , Produtos do Gene env/imunologia , Imunossupressores/química , Vírus da Leucemia Felina/genética , Leucemia Felina/imunologia , Mutação de Sentido Incorreto , Proteínas Oncogênicas de Retroviridae/genética , Vacinas Virais/genética , Animais , Vírus da Varíola dos Canários/metabolismo , Gatos , Feminino , Produtos do Gene env/administração & dosagem , Produtos do Gene env/genética , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Imunossupressores/administração & dosagem , Imunossupressores/imunologia , Interferons/genética , Interferons/imunologia , Interleucina-10/genética , Interleucina-10/imunologia , Vírus da Leucemia Felina/química , Vírus da Leucemia Felina/imunologia , Leucemia Felina/prevenção & controle , Leucemia Felina/virologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Estrutura Terciária de Proteína , Proteínas Oncogênicas de Retroviridae/administração & dosagem , Proteínas Oncogênicas de Retroviridae/química , Proteínas Oncogênicas de Retroviridae/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/química , Vacinas Virais/imunologia
6.
J Immunol ; 187(1): 361-71, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21646298

RESUMO

Infection of human cells by human T cell leukemia virus type 1 (HTLV-1) is mediated by the viral envelope glycoproteins. The gp46 surface glycoprotein binds to cell surface receptors, including heparan sulfate proteoglycans, neuropilin 1, and glucose transporter 1, allowing the transmembrane glycoprotein to initiate fusion of the viral and cellular membranes. The envelope glycoproteins are recognized by neutralizing Abs and CTL following a protective immune response, and therefore, represent attractive components for a HTLV-1 vaccine. To begin to explore the immunological properties of potential envelope-based subunit vaccine candidates, we have used a soluble recombinant surface glycoprotein (gp46, SU) fused to the Fc region of human IgG (sRgp46-Fc) as an immunogen to vaccinate mice. The recombinant SU protein is highly immunogenic and induces high titer Ab responses, facilitating selection of hybridomas that secrete mAbs targeting SU. Many of these mAbs recognize envelope displayed on the surface of HTLV-1-infected cells and virions and several of the mAbs robustly antagonize envelope-mediated membrane fusion and neutralize pseudovirus infectivity. The most potently neutralizing mAbs recognize the N-terminal receptor-binding domain of SU, though there is considerable variation in neutralizing proficiency of the receptor-binding domain-targeted mAbs. By contrast, Abs targeting the C-terminal domain of SU tend to lack robust neutralizing activity. Importantly, we find that both neutralizing and poorly neutralizing Abs strongly stimulate neutrophil-mediated cytotoxic responses to HTLV-1-infected cells. Our data demonstrate that recombinant forms of SU possess immunological features that are of significant utility to subunit vaccine design.


Assuntos
Anticorpos Neutralizantes/toxicidade , Anticorpos Antideltaretrovirus/toxicidade , Produtos do Gene env/imunologia , Vírus Linfotrópico T Tipo 1 Humano/imunologia , Proteínas Oncogênicas de Retroviridae/imunologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/virologia , Internalização do Vírus , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/toxicidade , Anticorpos Neutralizantes/biossíntese , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Anticorpos Antideltaretrovirus/biossíntese , Produtos do Gene env/administração & dosagem , Produtos do Gene env/genética , Infecções por HTLV-I/imunologia , Infecções por HTLV-I/prevenção & controle , Infecções por HTLV-I/virologia , Células HeLa , Vírus Linfotrópico T Tipo 1 Humano/patogenicidade , Humanos , Células Jurkat , Camundongos , Proteínas Oncogênicas de Retroviridae/administração & dosagem , Proteínas Oncogênicas de Retroviridae/genética , Vacinas de Subunidades Antigênicas/genética , Vacinas de Subunidades Antigênicas/imunologia , Vacinas de Subunidades Antigênicas/uso terapêutico
7.
Intervirology ; 54(2): 78-86, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20829603

RESUMO

To develop improved vaccination strategies against feline leukemia virus (FeLV), rats were immunized with the transmembrane envelope protein p15E of FeLV alone or in combination with the commercial vaccine Leucogen® comprising the nonglycosylated FeLV surface envelope protein. Binding and neutralizing antibodies were induced in both groups and in the group immunized with Leucogen alone. Higher titers of antibodies neutralizing FeLV were induced by simultaneous immunization with Leucogen and p15E compared to the responses using Leucogen or p15E alone, suggesting that combination vaccines should be used in the future. Epitope mapping of p15E-specific antibodies induced by simultaneous immunization with Leucogen and p15E revealed the same pattern of response as obtained after immunization with p15E alone: one epitope was localized in the membrane-proximal external region (MPER) and the other in the fusion peptide-proximal region, and they are related to the epitopes detected after immunization with p15E of the porcine endogenous retrovirus and the koala retrovirus. The data indicate that these epitopes in the MPER are an effective target for neutralization and that antigens containing them may therefore prove to be a useful component of vaccines against retroviruses, including HIV-1.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Imunização/métodos , Proteínas Oncogênicas de Retroviridae/imunologia , Vacinas Virais/imunologia , Animais , Mapeamento de Epitopos , Testes de Neutralização , Ratos , Ratos Wistar , Proteínas Oncogênicas de Retroviridae/administração & dosagem , Vacinas Virais/administração & dosagem
9.
J Am Vet Med Assoc ; 228(5): 726-7, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16506935

RESUMO

OBJECTIVE: To compare protection against FeLV challenge obtained following administration of 2 doses of an adjuvanted, chemically inactivated, whole FeLV (FeLV-k) vaccine with protection obtained following administration of 1 dose of an FeLV-k vaccine followed by 1 dose of a canarypox virus-vectored recombinant FeLV (rCP-FeLV) vaccine. DESIGN: Prospective study. ANIMALS: Thirty-two 9-week-old domestic shorthair cats. PROCEDURE: Cats received 2 doses of the FeLV-k vaccine SC, 21 days apart (n = 11); 1 dose of the FeLV-k vaccine SC and, 21 days later, 1 dose of the rCP-FeLV vaccine transdermally (11); or 2 doses of physiologic saline (0.9% NaCl) solution (control; 10). Four weeks after the second vaccine dose, all cats were challenged with FeLV by means of oronasal administration. Blood samples were collected at weekly intervals beginning 21 days after challenge, and serum was tested for FeLV antigen. RESULTS: All 10 control cats became persistently infected (ie, FeLV antigen detected in > or = 3 consecutive serum samples) following FeLV challenge, whereas only 1 of 11 cats that received 2 doses of the FeLV-k vaccine and none of the 11 cats that received 1 dose of the FeLV-k vaccine and 1 dose of the rCP-FeLV vaccine did. CONCLUSIONS AND CLINICAL RELEVANCE: Results suggest that protection against FeLV challenge obtained following SC administration of a single dose of an FeLV-k vaccine followed, 21 days later, by transdermal administration of a single dose of an rCP-FeLV vaccine was similar to that obtained following SC administration of 2 doses of the FeLV-k vaccine 21 days apart.


Assuntos
Vírus da Leucemia Felina/imunologia , Leucemia Felina/prevenção & controle , Proteínas Oncogênicas de Retroviridae/imunologia , Vacinação/veterinária , Vacinas Virais/imunologia , Animais , Vírus da Varíola dos Canários , Gatos , Esquemas de Imunização , Injeções Subcutâneas/veterinária , Estudos Prospectivos , Proteínas Oncogênicas de Retroviridae/administração & dosagem , Organismos Livres de Patógenos Específicos , Resultado do Tratamento , Vacinação/métodos , Vacinas de Produtos Inativados , Vacinas Sintéticas , Vacinas Virais/administração & dosagem
10.
Vaccine ; 24(11): 1838-46, 2006 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-16343700

RESUMO

The compatibility, safety and interaction on antibody induction of a combined vaccine application were assessed. Specific pathogen-free cats were vaccinated with either a modified live virus vaccine containing feline calici- (FCV), herpes- (FHV-1), parvovirus (FPV) and Chlamydophila felis (C. felis), an adjuvanted recombinant feline leukaemia virus (FeLV) vaccine or both vaccines in one syringe. After combined application, FeLV ELISA antibody titres were unaltered, However antibody production based on indirect immunofluorescence assay was remarkably enhanced for FCV and was at selected time points also enhanced for FHV-1 and C. felis but diminished for FPV. The use of these vaccines in combination was safe and will simplify vaccination schedules in veterinary practice.


Assuntos
Anticorpos Antivirais/sangue , Vacinas Bacterianas/imunologia , Gatos/imunologia , Chlamydophila/imunologia , Proteínas Oncogênicas de Retroviridae/imunologia , Vacinas Virais/imunologia , Adjuvantes Imunológicos , Animais , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/imunologia , Vacinas Bacterianas/administração & dosagem , Calicivirus Felino/imunologia , Citocinas/biossíntese , Citocinas/genética , Ensaio de Imunoadsorção Enzimática , Vírus da Panleucopenia Felina/imunologia , Técnica Indireta de Fluorescência para Anticorpo , RNA Mensageiro/análise , Proteínas Oncogênicas de Retroviridae/administração & dosagem , Proteínas Oncogênicas de Retroviridae/efeitos adversos , Proteínas Oncogênicas de Retroviridae/genética , Organismos Livres de Patógenos Específicos , Linfócitos T/imunologia , Vacinas Combinadas , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Varicellovirus/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/efeitos adversos , Vacinas Virais/genética
11.
J Virol ; 79(8): 4886-95, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15795274

RESUMO

To characterize the occurrence, frequency, and kinetics of retroviral recombination in vivo, we intravaginally inoculated rhesus macaques, either simultaneously or sequentially, with attenuated simian immunodeficiency virus (SIV) strains having complementary deletions in their accessory genes and various degrees of replication impairment. In monkeys inoculated simultaneously with SIVmac239Deltavpx/Deltavpr and SIVmac239Deltanef, recombinant wild-type (wt) virus and wild-type levels of plasma viral RNA (vRNA) were detected in blood by 2 weeks postinoculation. In monkeys inoculated first with SIVmac239Deltavpx/Deltavpr and then with SIVmac239Deltanef, recombination occurred but was associated with lower plasma vRNA levels than plasma vRNA levels seen for monkeys inoculated intravaginally with wt SIVmac239. In one monkey, recombination occurred 6 weeks after the challenge with SIVmac239Deltanef when plasma SIVmac239Deltavpx/Deltavpr RNA levels were undetectable. In monkeys inoculated first with the more highly replicating strain, SIVmac239Deltanef, and then with SIVmac239Deltavpx/Deltavpr, wild-type recombinant virus was not detected in blood or tissues. Instead, a virus that had repaired the deletion in the nef gene by a compensatory mutation was found in one animal. Overall, recombinant SIV was eventually found in four of six animals intravaginally inoculated with the two SIVmac239 deletion mutants. These findings show that recombination can occur readily in vivo after mucosal SIV exposure and thus contributes to the generation of viral genetic diversity and enhancement of viral fitness.


Assuntos
Produtos do Gene env/imunologia , Produtos do Gene vpr/genética , Genes Virais , Genes nef , Proteínas Oncogênicas de Retroviridae/imunologia , Vírus da Imunodeficiência Símia/genética , Proteínas Virais de Fusão/imunologia , Replicação Viral/genética , Animais , Feminino , Produtos do Gene env/administração & dosagem , Variação Genética , Genótipo , Injeções , Interferon gama/imunologia , Macaca mulatta , RNA Viral/genética , Proteínas Oncogênicas de Retroviridae/administração & dosagem , Deleção de Sequência , Vagina , Proteínas Virais de Fusão/administração & dosagem
12.
J Immunol ; 174(5): 2645-52, 2005 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15728471

RESUMO

Most tumor-associated Ags are self proteins that fail to elicit a T cell response as a consequence of immune tolerance. Dendritic cells (DCs) generated ex vivo have been used to break tolerance against such self Ags; however, in vitro manipulation of DCs is cumbersome and difficult to control, resulting in vaccines of variable potency. To address this problem we developed a method for loading and activating DCs, in situ, by first directing sufficient numbers of DCs to peripheral tissues using Flt3 ligand and then delivering a tumor-associated Ag and oligonucleotide containing unmethylated CG motifs to these tissues. In this study, we show in three different tumor models that this method can overcome tolerance and induce effective antitumor immunity. Vaccination resulted in the generation of CD8(+) T and NK cell effectors that mediated durable tumor responses without attacking normal tissues. These findings demonstrate that unmodified tumor-associated self Ags can be targeted to DCs in vivo to induce potent systemic antitumor immunity.


Assuntos
Antígenos de Neoplasias/imunologia , Autoantígenos/imunologia , Neoplasias do Colo/imunologia , Neoplasias do Colo/terapia , Células Dendríticas/imunologia , Tolerância Imunológica , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Animais , Antígenos de Neoplasias/administração & dosagem , Autoantígenos/administração & dosagem , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Antígeno Carcinoembrionário/administração & dosagem , Antígeno Carcinoembrionário/imunologia , Linhagem Celular Tumoral , Movimento Celular/imunologia , Proliferação de Células , Neoplasias do Colo/patologia , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Feminino , Células Matadoras Naturais/imunologia , Masculino , Melanoma Experimental/patologia , Proteínas de Membrana/administração & dosagem , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oligodesoxirribonucleotídeos/administração & dosagem , Oligodesoxirribonucleotídeos/imunologia , Oxirredutases/administração & dosagem , Oxirredutases/imunologia , Proteínas Oncogênicas de Retroviridae/administração & dosagem , Proteínas Oncogênicas de Retroviridae/imunologia , Proteínas do Envelope Viral/administração & dosagem , Proteínas do Envelope Viral/imunologia
13.
Vet Ther ; 5(4): 258-62, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15719325

RESUMO

The efficacy of a new recombinant FeLV vaccine (rFeLV), delivered transdermally via a needle-free delivery device was compared to that of an inactivated FeLV vaccine (FeLV-k), administered subcutaneously, with a conventional needle and syringe. Kittens were immunized with either rFeLV (0.25 ml, transdermal) or FeLV-k (1 ml, subcutaneous); or they were sham-vaccinated with physiologic saline (0.25 ml, transdermal). Two vaccinations were administered 21 days apart. Injection sites were monitored for any acute or subacute reactions relative to vaccine administration. Four weeks following the final vaccination, all cats were subject to oro-nasal FeLV challenge. Blood was collected for determination of FeLV antigenemia (p27) at weekly intervals beginning three weeks post-challenge. All of the vaccinated cats from both groups resisted FeLV challenge; and 90% of the control cats developed persistent FeLV antigenemia in response to challenge. No acute or persistent injection site reactions were observed. The rFeLV, delivered transdermally, provides protection against persistent FeLV antigenemia following a robust challenge that is equivalent to that of FeLV-k.


Assuntos
Vírus da Leucemia Felina/imunologia , Leucemia Felina/prevenção & controle , Proteínas Oncogênicas de Retroviridae , Vacinação/veterinária , Vacinas Virais , Administração Cutânea , Animais , Antígenos Virais/sangue , Gatos , Injeções Subcutâneas/veterinária , Distribuição Aleatória , Proteínas Oncogênicas de Retroviridae/administração & dosagem , Proteínas Oncogênicas de Retroviridae/normas , Segurança , Organismos Livres de Patógenos Específicos , Resultado do Tratamento , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/normas , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/normas , Vacinas Virais/administração & dosagem , Vacinas Virais/normas
14.
J Immunol ; 169(7): 3908-13, 2002 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-12244189

RESUMO

DNA vaccines can activate immunity against tumor Ags expressed as MHC class I-associated peptides. However, priming of CD8(+) CTL against weak tumor Ags may require adjuvant molecules. We have used a pathogen-derived sequence from tetanus toxin (fragment C (FrC)) fused to tumor Ag sequences to promote Ab and CD4(+) T cell responses. For induction of CD8(+) T cell responses, the FrC sequence has been engineered to remove potentially competitive MHC class I-binding epitopes and to improve presentation of tumor epitopes. The colon carcinoma CT26 expresses an endogenous retroviral gene product, gp70, containing a known H2-L(d)-restricted epitope (AH1). A DNA vaccine encoding gp70 alone was a poor inducer of CTL, and performance was not significantly improved by fusion of full-length FrC. However, use of a minimized domain of FrC, with the AH1 sequence fused to the 3' position, led to rapid induction of high levels of CTL. IFN-gamma-producing epitope-specific CTL were detectable ex vivo and these killed CT26 targets in vitro. The single epitope vaccine was more effective than GM-CSF-transfected CT26 tumor cells in inducing an AH1-specific CTL response and equally effective in providing protection against tumor challenge. Levels of AH1-specific CTL in vivo were increased following injection of tumor cells, and CTL expanded in vitro were able to kill CT26 cells in tumor bearers. Pre-existing immunity to tetanus toxoid had no effect on the induction of AH1-specific CTL. These data demonstrate the power of epitope-specific CTL against tumor cells and illustrate a strategy for priming immunity via a dual component DNA vaccine.


Assuntos
Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/imunologia , Citotoxicidade Imunológica , Epitopos de Linfócito T/imunologia , Ativação Linfocitária , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T Citotóxicos/imunologia , Vacinas de DNA/imunologia , Animais , Antígenos de Neoplasias/administração & dosagem , Antígenos de Neoplasias/genética , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/síntese química , Vacinas Anticâncer/genética , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Neoplasias do Colo/prevenção & controle , Citotoxicidade Imunológica/genética , Epitopos de Linfócito T/administração & dosagem , Epitopos de Linfócito T/genética , Inibidores do Crescimento/administração & dosagem , Inibidores do Crescimento/síntese química , Inibidores do Crescimento/genética , Inibidores do Crescimento/imunologia , Injeções Intramusculares , Interferon gama/biossíntese , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/síntese química , Proteínas Recombinantes de Fusão/genética , Proteínas Oncogênicas de Retroviridae/administração & dosagem , Proteínas Oncogênicas de Retroviridae/genética , Proteínas Oncogênicas de Retroviridae/imunologia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Citotóxicos/transplante , Toxoide Tetânico/administração & dosagem , Toxoide Tetânico/imunologia , Vacinas de DNA/administração & dosagem , Vacinas de DNA/síntese química , Vacinas de DNA/genética , Proteínas do Envelope Viral/administração & dosagem , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia
15.
Vaccine ; 20(23-24): 2866-72, 2002 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-12126896

RESUMO

The duration of immunity provided by a feline leukemia virus (FeLV) vaccine, Leukocell 2, was determined. Kittens were vaccinated when 9 and 12 weeks of age and were challenged 12 months later with FeLV-A/Glasgow-1. An oronasal challenge protocol without corticosteroid enhancement was developed in order to induce a persistent viraemia in a high proportion of adult cats. Fourteen of 18 (80%) of the vaccinated cats challenged in this way remained non-viraemic while 9/15 (60%) of age-matched controls became persistently infected, a preventable fraction of 63%. This difference was statistically significant (P=0.038). For comparison, 10 of 12 (83%) 15-17-week-old kittens challenged in the same way became persistently infected, confirming the relative resistance of adult animals to FeLV. Tests for virus neutralising and anti-feline oncornavirus-associated cell membrane antigen (FOCMA) antibodies suggested that the former were more important than the latter in protection. Thus, Leukocell 2 protected a significant proportion of cats from FeLV challenge 1 year after primary vaccination as kittens.


Assuntos
Doenças do Gato/prevenção & controle , Vírus da Leucemia Felina/imunologia , Infecções por Retroviridae/veterinária , Proteínas Oncogênicas de Retroviridae/administração & dosagem , Infecções Tumorais por Vírus/veterinária , Vacinas Virais/administração & dosagem , Animais , Anticorpos Antivirais/sangue , Antígenos Virais/sangue , Doenças do Gato/imunologia , Gatos , Feminino , Produtos do Gene gag/sangue , Produtos do Gene gag/imunologia , Vírus da Leucemia Felina/patogenicidade , Masculino , Boca , Testes de Neutralização , Nariz , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/prevenção & controle , Proteínas dos Retroviridae/sangue , Proteínas dos Retroviridae/imunologia , Fatores de Tempo , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/prevenção & controle , Viremia/imunologia , Viremia/prevenção & controle , Viremia/veterinária , Virulência
17.
J Am Vet Med Assoc ; 203(3): 396-405, 1993 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-8257557

RESUMO

Within the past 2 years, a putative causal relationship has been reported between vaccination against rabies and the development of fibrosarcomas at injection sites in cats. A retrospective study was undertaken, involving 345 cats with fibrosarcomas diagnosed between January 1991 and May 1992, to assess the causal hypothesis. Cats with fibrosarcomas developing at body locations where vaccines are typically administered (n = 185) were compared with controls (n = 160) having fibrosarcomas at locations not typically used for vaccination. In cats receiving FeLV vaccination within 2 years of tumorigenesis, the time between vaccination and tumor development was significantly (P = 0.005) shorter for tumors developing at sites where vaccines are typically administered than for tumors at other sites. Univariate analysis, adjusted for age, revealed associations between FeLV vaccination (odds ratio [OR] = 2.82; 95% confidence interval [CI] = 1.54 to 5.15), rabies vaccination at the cervical/interscapular region (OR = 2.09; 95% CI = 1.01 to 4.31), and rabies vaccination at the femoral region (OR = 1.83; 95% CI = 0.65 to 5.10) with fibrosarcoma development at the vaccination site within 1 year of vaccination. Multivariate analysis, adjusted for age and other vaccines, also revealed increased risks after FeLV (OR = 5.49; 95% CI = 1.98 to 15.24) and rabies (OR = 1.99; 95% CI = 0.72 to 5.54) vaccination.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Doenças do Gato/etiologia , Fibrossarcoma/veterinária , Neoplasias de Tecidos Moles/veterinária , Vacinação/veterinária , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/efeitos adversos , Fatores Etários , Animais , California/epidemiologia , Estudos de Casos e Controles , Doenças do Gato/epidemiologia , Gatos , Estudos de Coortes , Feminino , Fibrossarcoma/epidemiologia , Fibrossarcoma/etiologia , Havaí/epidemiologia , Incidência , Injeções Intramusculares/efeitos adversos , Injeções Intramusculares/veterinária , Injeções Subcutâneas/efeitos adversos , Injeções Subcutâneas/veterinária , Vírus da Leucemia Felina/imunologia , Masculino , Análise Multivariada , Vacina Antirrábica/administração & dosagem , Vacina Antirrábica/efeitos adversos , Estudos Retrospectivos , Proteínas Oncogênicas de Retroviridae/administração & dosagem , Proteínas Oncogênicas de Retroviridae/efeitos adversos , Fatores de Risco , Neoplasias de Tecidos Moles/epidemiologia , Neoplasias de Tecidos Moles/etiologia , Vacinação/efeitos adversos , Vacinas Virais/administração & dosagem , Vacinas Virais/efeitos adversos
18.
Vet Immunol Immunopathol ; 35(1-2): 191-7, 1992 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-1337397

RESUMO

Cats were vaccinated with one of the three preparations: purified feline immunodeficiency virus (FIV) incorporated into immune stimulating complexes (ISCOMs), recombinant FIV p24 ISCOMs, or a fixed, inactivated cell vaccine in quil A. Cats inoculated with the FIV ISCOMs or the recombinant p24 ISCOMs developed high titres of antibodies against the core protein p24 but had no detectable antibodies against the env protein gp120 or virus neutralising antibodies. In contrast, all of the cats inoculated with the fixed, inactivated cell vaccine developed anti-env antibodies and four of five had detectable levels of neutralising antibody. However, none of the vaccinated cats were protected from infection after intraperitoneal challenge with 20 infectious units of FIV. Indeed there appeared to be enhancement of infection after vaccination as the vaccinated cats become viraemic sooner than the unvaccinated controls, and 100% of the vaccinated cats became viraemic compared with 78% of the controls. The mechanism responsible for this enhancement remains unknown.


Assuntos
Anticorpos Antivirais/sangue , Síndrome de Imunodeficiência Adquirida Felina/imunologia , Produtos do Gene gag/imunologia , Vírus da Imunodeficiência Felina/imunologia , Proteínas Oncogênicas de Retroviridae/imunologia , Vacinação/veterinária , Vacinas Virais/imunologia , Adjuvantes Imunológicos , Animais , Gatos , Síndrome de Imunodeficiência Adquirida Felina/microbiologia , Síndrome de Imunodeficiência Adquirida Felina/prevenção & controle , Produtos do Gene gag/administração & dosagem , Vírus da Imunodeficiência Felina/crescimento & desenvolvimento , Testes de Neutralização , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Proteínas Oncogênicas de Retroviridae/administração & dosagem , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/imunologia , Vacinas Virais/administração & dosagem , Viremia/veterinária
19.
Vet Immunol Immunopathol ; 35(1-2): 61-9, 1992 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-1337403

RESUMO

Tumor necrosis factor alpha (TNF alpha) levels were determined by enzyme-linked immunosorbent assay (ELISA) and by cell culture bioassay in supernatants of lipopolysaccharide-stimulated feline monocyte cultures and in cat serum samples. There was a good correlation between the results obtained by the two methods. From the fact that TNF alpha was neutralized quantitatively by antibodies to human TNF alpha in feline monocyte supernatants and in feline sera, it was concluded that feline TNF alpha immunologically cross-reacts with human TNF alpha and that the human TNF alpha ELISA can be used to quantitate feline TNF alpha. During the first 6 months after experimental feline immunodeficiency virus (FIV) infection no differences in serum TNF alpha values were observed between infected and non-infected cats. TNF alpha levels increased significantly after primary vaccination with a feline leukemia virus (FeLV) vaccine in FIV infected cats over those in the non-infected controls. During secondary immune response TNF alpha levels rose transiently for a period of a few days in both the FIV positive and the FIV negative cats. After FeLV challenge, TNF alpha levels increased in all animals challenged with virulent FeLV for a period of 3 weeks. This period corresponded to the time necessary to develop persistent FeLV viremia in the control cats. It was concluded from these experiments that in the asymptomatic phase of FIV infection no increased levels of TNF alpha are present, similar to the situation in asymptomatic HIV infected humans. Activation of monocytes/macrophages in FIV infected cats by stimuli such as vaccination or FeLV challenge readily leads to increased levels of TNF alpha.


Assuntos
Síndrome de Imunodeficiência Adquirida Felina/imunologia , Vírus da Imunodeficiência Felina/imunologia , Vírus da Leucemia Felina/imunologia , Leucemia Experimental/imunologia , Proteínas Oncogênicas de Retroviridae/imunologia , Fator de Necrose Tumoral alfa/análise , Vacinas Virais/imunologia , Animais , Gatos , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Feminino , Leucemia Experimental/complicações , Ativação de Macrófagos/imunologia , Masculino , Monócitos/imunologia , Proteínas Oncogênicas de Retroviridae/administração & dosagem , Organismos Livres de Patógenos Específicos , Vacinas Virais/administração & dosagem
20.
J Gen Virol ; 73 ( Pt 7): 1811-8, 1992 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-1321214

RESUMO

The env and gag genes from feline leukaemia virus were expressed in a thymidine kinase-negative feline herpes-virus and a baculovirus. Cats were vaccinated with various combinations of these recombinant viruses and 100% protection against feline leukaemia virus challenge was achieved using an immunization schedule which utilized both env and gag products delivered at both a mucosal and systemic site.


Assuntos
Baculoviridae/genética , Genes env , Genes gag , Vetores Genéticos , Herpesviridae/genética , Vírus da Leucemia Felina/imunologia , Recombinação Genética , Proteínas Oncogênicas de Retroviridae , Vacinas Virais , Animais , Anticorpos Antivirais/análise , Gatos , Vírus da Leucemia Felina/genética , Proteínas Oncogênicas de Retroviridae/administração & dosagem , Proteínas Oncogênicas de Retroviridae/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...