Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Infect Immun ; 82(2): 793-807, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24478093

RESUMO

Plasmodium vivax is the most widespread and the second most prevalent malaria-causing species in the world. Current measures used to control the transmission of this disease would benefit from the development of an efficacious vaccine. In the case of the deadly parasite P. falciparum, the recombinant RTS,S vaccine containing the circumsporozoite antigen (CSP) consistently protects 30 to 50% of human volunteers against infection and is undergoing phase III clinical trials in Africa with similar efficacy. These findings encouraged us to develop a P. vivax vaccine containing the three circulating allelic forms of P. vivax CSP. Toward this goal, we generated three recombinant bacterial proteins representing the CSP alleles, as well as a hybrid polypeptide called PvCSP-All-CSP-epitopes. This hybrid contains the conserved N and C termini of P. vivax CSP and the three variant repeat domains in tandem. We also generated simian and human recombinant replication-defective adenovirus vectors expressing PvCSP-All-CSP-epitopes. Mice immunized with the mixture of recombinant proteins in a formulation containing the adjuvant poly(I·C) developed high and long-lasting serum IgG titers comparable to those elicited by proteins emulsified in complete Freund's adjuvant. Antibody titers were similar in mice immunized with homologous (protein-protein) and heterologous (adenovirus-protein) vaccine regimens. The antibodies recognized the three allelic forms of CSP, reacted to the repeated and nonrepeated regions of CSP, and recognized sporozoites expressing the alleles VK210 and VK247. The vaccine formulations described in this work should be useful for the further development of an anti-P. vivax vaccine.


Assuntos
Vacinas Antimaláricas/imunologia , Malária Vivax/prevenção & controle , Plasmodium vivax/imunologia , Proteínas de Protozoários/imunologia , Vacinação/métodos , Adjuvantes Imunológicos/administração & dosagem , Animais , Anticorpos Antiprotozoários/sangue , Feminino , Imunoglobulina G/sangue , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/genética , Malária Vivax/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium vivax/genética , Poli I-C/administração & dosagem , Proteínas de Protozoários/genética , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
2.
J Leukoc Biol ; 93(3): 437-48, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23271702

RESUMO

In this study, we compared the immunogenicity and protection from repeated low-dose intrarectal SIVmac251 challenge in two groups of vaccinated RMs. Animals were immunized with live SIVmac239, which had been attenuated by a deletion of the nef sequence, or they were vaccinated twice with an E1-deleted AdHu5, expressing SIVmac239gag. The vaccinated animals and a cohort of unvaccinated control animals were then challenged 10 times in weekly intervals with low doses of SIVmac251 given rectally. Our results confirm previous studies showing that whereas SIVΔnef provides some degree of protection against viral acquisition after repeated low-dose rectal SIVmac251 challenges, vaccination with an AdHu5gag vaccine designed to induce only antiviral T cell responses is ineffective. As immunological analyses of prechallenge, vaccine-induced T and B cell responses failed to reveal correlates of protection that distinguished the more susceptible from the more resistant vaccinated animals, we carried out RNA-Seq studies of paired pre- and postvaccination samples to identify transcriptional patterns that correlated with the differences in response. We show that gene expression signatures associated with the delayed SIV infection seen in some AdHu5gag recipients were largely present in prevaccination samples of those animals. In contrast, the responding SIVΔnef-immunized animals showed a predominance of vaccine-induced changes, thus enabling us to define inherited and vaccine-induced gene expression signatures and their associated pathways that may play a role in preventing SIV acquisition.


Assuntos
Linfócitos B/imunologia , Imunidade Celular/efeitos dos fármacos , Vacinas contra a SAIDS/farmacologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia , Linfócitos T/imunologia , Vacinação , Animais , Linfócitos B/virologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Imunidade Celular/genética , Imunidade Celular/imunologia , Macaca mulatta , Masculino , Vacinas contra a SAIDS/genética , Vacinas contra a SAIDS/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/genética , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Linfócitos T/virologia
3.
Mol Ther ; 19(11): 2021-30, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21587208

RESUMO

Hepatic adeno-associated virus serotype 2 (AAV2)-mediated gene transfer failed to achieve sustained transgene product expression in human subjects. We formulated the hypothesis that rejection of AAV-transduced hepatocytes is caused by AAV capsid-specific CD8(+) T cells that become reactivated upon gene transfer. Although this hypothesis was compatible with clinical data, which showed a rise in circulating AAV capsid-specific T cells following injection of AAV vectors, it did not explain that AAV vectors achieved long-term transgene expression in rhesus macaques, which are naturally infected with AAV serotypes closely related to those of humans. To address this apparent contradiction, we tested human and rhesus macaque samples for AAV capsid-specific T cells by intracellular cytokine staining combined with staining for T-cell subset and differentiation markers. This highly sensitive method, which could provide a tool to monitor adverse T-cell responses in gene transfer trials, showed that AAV capsid-specific CD8(+) and CD4(+) T cells can be detected in blood of naturally infected humans and rhesus macaques. They are present at higher frequencies in rhesus macaques. Furthermore, T cells from humans and rhesus macaques exhibit striking differences in their differentiation status and in their functions, which may explain the disparate duration of AAV-mediated gene transfer in these two species.


Assuntos
Capsídeo/imunologia , Dependovirus/imunologia , Infecções por Parvoviridae/imunologia , Linfócitos T/imunologia , Adolescente , Adulto , Animais , Humanos , Imunofenotipagem , Macaca mulatta , Pessoa de Meia-Idade , Linfócitos T/metabolismo , Adulto Jovem
4.
Mol Ther ; 19(9): 1727-36, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21587210

RESUMO

Vaccines that aim to expand tumor-specific CD8(+) T cells have yielded disappointing results in cancer patients although they showed efficacy in transplantable tumor mouse models. Using a system that more faithfully mimics a progressing cancer and its immunoinhibitory microenvironment, we here show that in transgenic mice, which gradually develop adenocarcinomas due to expression of HPV-16 E7 within their thyroid, a highly immunogenic vaccine expressing E7 only induces low E7-specific CD8(+) T-cell responses, which fail to affect the size of the tumors. In contrast, the same type of vaccine expressing E7 fused to herpes simplex virus (HSV)-1 glycoprotein D (gD), an antagonist of the coinhibitory B- and T-lymphocyte attenuator (BTLA)/CD160-herpes virus entry mediator (HVEM) pathways, stimulates potent E7-specific CD8(+) T-cell responses, which can be augmented by repeated vaccination, resulting in initial regression of even large tumor masses in all mice with sustained regression in more than half of them. These results indicate that active immunization concomitantly with blockade of the immunoinhibitory HVEM-BTLA/CD160 pathways through HSV-1 gD may result in sustained tumor regression.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Imunoterapia Ativa/métodos , Neoplasias/terapia , Proteínas E7 de Papillomavirus/imunologia , Animais , Animais Geneticamente Modificados , Biomarcadores , Linfócitos T CD8-Positivos/metabolismo , Regulação da Expressão Gênica , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias/imunologia , Neoplasias/metabolismo , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/metabolismo , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Receptores Imunológicos/metabolismo , Membro 14 de Receptores do Fator de Necrose Tumoral/genética , Membro 14 de Receptores do Fator de Necrose Tumoral/imunologia , Membro 14 de Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais , Glândula Tireoide/imunologia , Vacinação/métodos
5.
Mol Ther ; 19(2): 417-26, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21081905

RESUMO

Despite enormous efforts by the scientific community, an effective HIV vaccine remains elusive. To further address to what degree T cells in absence of antibodies may protect against simian immunodeficiency virus (SIV) disease progression, rhesus macaques were vaccinated intramuscularly with a chimpanzee-derived Ad vector (AdC) serotype 6 and then boosted intramuscularly with a serologically distinct AdC vector of serotype 7 both expressing Gag of SIVmac239. Animals were subsequently boosted intramuscularly with a modified vaccinia Ankara (MVA) virus expressing Gag and Tat of the homologous SIV before mucosal challenge with a high dose of SIVmac239 given rectally. Whereas vaccinated animals showed only a modest reduction of viral loads, their overall survival was improved, in association with a substantial protection from the loss of CD4(+) T cells. In addition, the two vaccinated Mamu-A*01(+) macaques controlled viral loads to levels below detection within weeks after challenge. These data strongly suggest that T cells, while unable to affect SIV acquisition upon high-dose rectal infection, can reduce disease progression. Induction of potent T-cell responses should thus remain a component of our efforts to develop an efficacious vaccine to HIV-1.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Macaca mulatta/imunologia , Macaca mulatta/virologia , Vacinas contra a SAIDS/imunologia , Vírus da Imunodeficiência Símia/imunologia , Vírus da Imunodeficiência Símia/patogenicidade , Animais , Feminino , Masculino
6.
Mol Ther ; 18(12): 2182-9, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20877342

RESUMO

A universal influenza vaccine, designed to induce broadly cross-reactive immunity against current and future influenza A virus strains, is in critical demand to reduce the need for annual vaccinations with vaccines chosen upon predicting the predominant circulating viral strains, and to ameliorate the threat of cyclically occurring pandemics that have, in the past, killed tens of millions. Here, we describe a vaccine regimen based on sequential immunization with two serologically distinct chimpanzee-derived replication-defective adenovirus (Ad) vectors expressing the matrix-2 protein ectodomain (M2e) from three divergent strains of influenza A virus fused to the influenza virus nucleoprotein (NP) for induction of antibodies to M2e and virus-specific CD8(+) T cells to NP. In preclinical mouse models, the Ad vaccines expressing M2e and NP elicit robust NP-specific CD8(+) T-cell responses and moderate antibody responses to all three M2e sequences. Most importantly, vaccinated mice are protected against morbidity and mortality following challenge with high doses of different influenza virus strains. Protection requires both antibodies to M2e and cellular immune responses to NP.


Assuntos
Adenoviridae , Vírus da Influenza A , Vacinas contra Influenza , Influenza Humana/prevenção & controle , Nucleoproteínas/metabolismo , Proteínas da Matriz Viral/metabolismo , Adenoviridae/genética , Sequência de Aminoácidos , Animais , Humanos , Vírus da Influenza A/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Proteínas Recombinantes de Fusão/genética
7.
Clin Vaccine Immunol ; 17(10): 1576-83, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20739505

RESUMO

Recombinant adenovirus or DNA vaccines encoding herpes simplex virus type 1 (HSV-1) glycoprotein D (gD) genetically fused to human papillomavirus type 16 (HPV-16) oncoproteins (E5, E6, and E7) induce antigen-specific CD8(+) T-cell responses and confer preventive resistance to transplantable murine tumor cells (TC-1 cells). In the present report, we characterized some previously uncovered aspects concerning the induction of CD8(+) T-cell responses and the therapeutic anticancer effects achieved in C57BL/6 mice immunized with pgD-E7E6E5 previously challenged with TC-1 cells. Concerning the characterization of the immune responses elicited in mice vaccinated with pgD-E7E6E5, we determined the effect of the CD4(+) T-cell requirement, longevity, and dose-dependent activation on the E7-specific CD8(+) T-cell responses. In addition, we determined the priming/boosting properties of pgD-E7E6E5 when used in combination with a recombinant serotype 68 adenovirus (AdC68) vector encoding the same chimeric antigen. Mice challenged with TC-1 cells and then immunized with three doses of pgD-E7E6E5 elicited CD8(+) T-cell responses, measured by intracellular gamma interferon (IFN-γ) and CD107a accumulation, to the three HPV-16 oncoproteins and displayed in vivo antigen-specific cytolytic activity, as demonstrated with carboxyfluorescein diacetate succinimidyl ester (CFSE)-labeled target cells pulsed with oligopeptides corresponding to the H-2D(b)-restricted immunodominant epitopes of the E7, E6, or E5 oncoprotein. Up to 70% of the mice challenged with 5 × 10(5) TC-1 cells and immunized with pgD-E7E6E5 controlled tumor development even after 3 days of tumor cell challenge. In addition, coadministration of pgD-E7E6E5 with DNA vectors encoding pGM-CSF or interleukin-12 (IL-12) enhanced the therapeutic antitumor effects for all mice challenged with TC-1 cells. In conclusion, the present results expand our previous knowledge on the immune modulation properties of the pgD-E7E6E5 vector and demonstrate, for the first time, the strong antitumor effects of the DNA vaccine, raising promising perspectives regarding the development of immunotherapeutic reagents for the control of HPV-16-associated tumors.


Assuntos
Vacinas Anticâncer/imunologia , Papillomavirus Humano 16/imunologia , Neoplasias Experimentais/prevenção & controle , Proteínas Oncogênicas Virais/imunologia , Vacinas contra Papillomavirus/imunologia , Vacinas de DNA/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Relação Dose-Resposta Imunológica , Feminino , Papillomavirus Humano 16/genética , Imunização Secundária/métodos , Interferon gama/metabolismo , Leucócitos Mononucleares/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Experimentais/imunologia , Proteínas Oncogênicas Virais/genética , Vacinas contra Papillomavirus/genética , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/imunologia , Fatores de Tempo , Vacinação/métodos , Vacinas de DNA/administração & dosagem , Proteínas do Envelope Viral/genética
8.
Curr Opin Immunol ; 22(3): 385-90, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20466529

RESUMO

The clinical success of adaptive transfer of in vitro expanded antigen-specific CD8(+) T cells isolated from patients' tumors has demonstrated that effector cells of the adaptive immune system can effectively eliminate even large tumor masses. Nevertheless, cancer vaccines that aim to expand such CD8(+) T cells in situ have had remarkably little success in spite of numerous attempts. Recent advances in basic immunology have revealed layers of complexity controlling activation and maintenance of adaptive immune responses that are tightly controlled by immunoinhibitory pathways to avoid horror autotoxicus. During tumor progression the activities of negative pathways increase and together with cancer immune evasion tactics presumably prevent induction of an efficacious immune response by cancer vaccines that solely provide more antigen to an already suppressed system. Cancer vaccines may thus need to readjust the imbalance of the cancer patients' immune system by inhibiting immunoinhibitors; such regimens have shown preclinical efficacy and are now entering clinical trials hopefully ending the Kafkaesque futility of cancer vaccines.


Assuntos
Vacinas Anticâncer/imunologia , Imunoterapia/métodos , Neoplasias/imunologia , Neoplasias/terapia , Evasão Tumoral/imunologia , Animais , Antígenos de Neoplasias/imunologia , Humanos , Camundongos
9.
J Immunol ; 184(10): 5475-84, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20410485

RESUMO

Immune responses diminish with age resulting in an increased susceptibility of the elderly to infectious agents and an inability to mount protective immune responses to vaccines. Immunosenescence affects multiple aspects of the immune system, including CD8(+) T cells, which control viral infections and are assumed to prevent the development of cancers. In this study, we tested if CD8(+) T cell responses in aged mice could be enhanced through a vaccine that concomitantly expresses Ag and a molecule that blocks an immunoinhibitory pathway. Specifically, we tested a vaccine based on a replication-defective chimpanzee-derived adenovirus vector expressing the nucleoprotein (NP) of influenza A virus as a fusion protein with the HSV type 1 glycoprotein D, which through binding to the herpes virus entry mediator, blocks the immunoinhibitory herpes virus entry mediator B and T lymphocyte attenuator/CD160 pathways. Our results show that the vaccine expressing a fusion protein of NP and glycoprotein D induces significantly higher NP-specific CD8(+) T cell responses in young and aged mice compared with the vaccine expressing NP only.


Assuntos
Envelhecimento/imunologia , Linfócitos T CD8-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Vírus da Influenza A/imunologia , Membro 14 de Receptores do Fator de Necrose Tumoral/antagonistas & inibidores , Transdução de Sinais/imunologia , Regulação para Cima/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Linhagem Celular , Epitopos de Linfócito T/genética , Feminino , Vetores Genéticos/imunologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/imunologia , Humanos , Epitopos Imunodominantes/genética , Epitopos Imunodominantes/imunologia , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Nucleocapsídeo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/imunologia , Receptores Imunológicos/genética , Membro 14 de Receptores do Fator de Necrose Tumoral/fisiologia , Proteínas Recombinantes de Fusão/imunologia , Transdução de Sinais/genética , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/antagonistas & inibidores , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Proteínas do Core Viral/genética , Proteínas do Core Viral/imunologia
10.
PLoS One ; 5(12): e14385, 2010 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-21203546

RESUMO

BACKGROUND: Adenoviral (Ad) vaccine vectors represent both a vehicle to present a novel antigen to the immune system as well as restimulation of immune responses against the Ad vector itself. To what degree Ad-specific CD8(+) T cells are restimulated by Ad vector vaccination is unclear, although such knowledge would be important as vector-specific CD8(+) T cell expansion could potentially further limit Ad vaccine efficacy beyond Ad-specific neutralizing antibody alone. METHODOLOGY/PRINCIPAL FINDINGS: Here we addressed this issue by measuring human Adenovirus serotype 5 (Ad5)-specific CD8(+) T cells in recipients of the Merck Ad5 HIV-1 vaccine vector before, during, and after vaccination by multicolor flow cytometry. Ad5-specific CD8(+) T-cells were detectable in 95% of subjects prior to vaccination, and displayed primarily an effector-type functional profile and phenotype. Peripheral blood Ad5-specific CD8(+) T-cell numbers expanded after Ad5-HIV vaccination in all subjects, but differential expansion kinetics were noted in some baseline Ad5-neutralizing antibody (Ad5 nAb) seronegative subjects compared to baseline Ad5 nAb seropositive subjects. However, in neither group did vaccination alter polyfunctionality, mucosal targeting marker expression, or memory phenotype of Ad5-specific CD8(+) T-cells. CONCLUSIONS: These data indicate that repeat Ad5-vector administration in humans expands Ad5-specific CD8(+) T-cells without overtly affecting their functional capacity or phenotypic properties. This is a secondary analysis of samples collected during the 016 trial. Results of the Merck 016 trial safety and immunogenicity have been previously published in the journal of clinical infectious diseases [1]. TRIAL REGISTRATION: ClinicalTrials.gov NCT00849680[http://www.clinicaltrials.gov/show/NCT00849680].


Assuntos
Vacinas contra a AIDS/uso terapêutico , Adenoviridae/genética , Linfócitos T CD8-Positivos/citologia , Animais , Células CHO , Ensaios Clínicos como Assunto , Cricetinae , Cricetulus , Citometria de Fluxo/métodos , Vetores Genéticos , Humanos , Leucócitos Mononucleares/citologia , Modelos Biológicos , Fenótipo , Fatores de Tempo
11.
Mol Ther ; 17(8): 1333-9, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19513019

RESUMO

Adenovirus (Ad) vectors were initially developed for treatment of genetic diseases. Their usefulness for permanent gene replacement was limited by their high immunogenicity, which resulted in rapid elimination of transduced cells through induction of T and B cells to antigens of Ad and the transgene product. The very trait that excluded their use for sustained treatment of genetic diseases made them highly attractive as vaccine carriers. Recently though results showed that Ad vectors based on common human serotypes, such as serotype 5, may not be ideal as vaccine carriers. A recently conducted phase 2b trial, termed STEP trial, with an AdHu5-based vaccine expressing antigens of human immunodeficiency virus 1 (HIV-1) not only showed lack of efficacy in spite of the vaccine's immunogenicity, but also suggested an increased trend for HIV acquisition in individuals that had circulating AdHu5 neutralizing antibodies prior to vaccination. Alternative serotypes from humans or nonhuman primates (NHPs), to which most humans lack pre-existing immunity, have been vectored and may circumvent the problems encountered with the use of AdHu5 vectors in humans. In summary, although Ad vectors have seen their share of setbacks in recent years, they remain viable tools for prevention or treatment of a multitude of diseases.


Assuntos
Adenoviridae/genética , Vetores Genéticos/genética , Vacinas/genética , Adenoviridae/imunologia , Animais , Vetores Genéticos/imunologia , Humanos , Sorotipagem , Vacinas/imunologia
12.
J Immunol ; 182(10): 6587-99, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19414814

RESUMO

In the phase IIb STEP trial an HIV-1 vaccine based on adenovirus (Ad) vectors of the human serotype 5 (AdHu5) not only failed to induce protection but also increased susceptibility to HIV-1 infection in individuals with preexisting neutralizing Abs against AdHu5. The mechanisms underlying the increased HIV-1 acquisition rates have not yet been elucidated. Furthermore, it remains unclear if the lack of the vaccine's efficacy reflects a failure of the concept of T cell-mediated protection against HIV-1 or a product failure of the vaccine. Here, we compared two vaccine regimens based on sequential use of AdHu5 vectors or two different chimpanzee-derived Ad vectors in rhesus macaques that were AdHu5 seropositive or seronegative at the onset of vaccination. Our results show that heterologous booster immunizations with the chimpanzee-derived Ad vectors induced higher T and B cell responses than did repeated immunizations with the AdHu5 vector, especially in AdHu5-preexposed macaques.


Assuntos
Vacinas contra a AIDS/imunologia , Adenoviridae/imunologia , Anticorpos Antivirais/imunologia , Vetores Genéticos/imunologia , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Adenoviridae/genética , Animais , Anticorpos Antivirais/sangue , Ensaio de Imunoadsorção Enzimática , Vetores Genéticos/genética , Proteínas do Vírus da Imunodeficiência Humana/genética , Imunização Secundária/métodos , Interferon gama/biossíntese , Interleucina-2/biossíntese , Ativação Linfocitária/imunologia , Macaca mulatta , Pan troglodytes , Reação em Cadeia da Polimerase , Linfócitos T/imunologia
13.
J Virol ; 83(11): 5567-73, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19279092

RESUMO

A major obstacle to the use of adenovirus vectors derived from common human serotypes, such as human adenovirus 5 (AdHu5), is the high prevalence of virus-neutralizing antibodies in the human population. We previously constructed a variant of chimpanzee adenovirus 68 (AdC68) that maintained the fundamental properties of the carrier but was serologically distinct from AdC68 and resisted neutralization by AdC68 antibodies. In the present study, we tested whether this modified vector, termed AdCDQ, could induce transgene product-specific CD8(+) T cells in mice with preexisting neutralizing antibody to wild-type AdC68. Contrary to our expectation, the data show conclusively that antibodies that fail to neutralize the AdCDQ mutant vector in vitro nevertheless impair the vector's capacity to transduce cells and to stimulate a transgene product-specific CD8(+) T-cell response in vivo. The results thus suggest that in vitro neutralization assays may not reliably predict the effects of virus-specific antibodies on adenovirus vectors in vivo.


Assuntos
Adenoviridae/genética , Adenoviridae/imunologia , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/farmacologia , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Vacinas Virais/imunologia , Animais , Especificidade de Anticorpos , Linfócitos T CD8-Positivos/imunologia , Feminino , Genes Reporter/genética , Imunoensaio , Camundongos , Camundongos Endogâmicos BALB C , Receptores Fc/imunologia , Transgenes/genética
14.
Hum Vaccin ; 5(1): 6-14, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-18927514

RESUMO

Adaptive immune responses are tightly controlled by activating and inhibitory signals, which ensure an effective defense against pathogens while preventing detrimental reactions. Regulation is provided through sets of receptors and ligands expressed on lymphocytes and antigen-presenting cells. Expression of the regulatory molecules is up or downregulated during immune responses and some pathways provide positive or negative signals depending on the state of differentiation of the cells. Many of the ligands and receptors can engage multiple binding partners and thus have distinct effects on downstream signaling. Medicinal manipulations of such pathways are being explored to augment antigen-driven immune responses through activation of positive or blockade of negative signals. In this review we provide a brief description of the co-stimulatory and co-inhibitory receptors and ligands with focus on the pathways mediated by the herpes virus entry mediator (HVEM) and the B and T lymphocyte mediator (BTLA).


Assuntos
Receptores Imunológicos/imunologia , Membro 14 de Receptores do Fator de Necrose Tumoral/imunologia , Vacinas/imunologia , Humanos
15.
Nat Med ; 14(2): 205-12, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18193057

RESUMO

Interactions between the herpesvirus entry mediator (HVEM) and the B- and T-lymphocyte attenuator (BTLA) inhibit B and T cell activation. HVEM-BTLA interactions are blocked by herpes simplex virus (HSV) glycoprotein D (gD) through binding of its N-terminal domain to the BTLA binding site of HVEM. In this study, we inserted viral antigens into the C-terminal domain of gD and expressed these antigens with plasmid or E1-deleted (replication-defective) adenovirus vectors. Viral antigens fused to gD induced T and B cell responses to the antigen that were far more potent than those elicited by the same antigen expressed without gD. The immunopotentiating effect required binding of the gD chimeric protein to HVEM. Overall, the studies demonstrate that targeting of antigen to the BTLA binding site of HVEM augments the immunogenicity of vaccines.


Assuntos
Antígenos Virais/imunologia , Imunidade/imunologia , Membro 14 de Receptores do Fator de Necrose Tumoral/imunologia , Adenoviridae , Animais , Anticorpos Antivirais/imunologia , Formação de Anticorpos/imunologia , Especificidade de Anticorpos , Linfócitos T CD8-Positivos/imunologia , Células CHO , Cricetinae , Cricetulus , Produtos do Gene gag/imunologia , Vetores Genéticos , Humanos , Imunização , Ativação Linfocitária/imunologia , Camundongos , Fenótipo , Ligação Proteica , Proteínas Recombinantes de Fusão/imunologia , Proteínas do Envelope Viral/imunologia
16.
J Clin Invest ; 117(12): 3958-70, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18008010

RESUMO

Recombinant adeno-associated virus (rAAV) vectors were used in human trials as carriers of vaccines for HIV-1 after encouraging preclinical results. However, the clinical trials yielded disappointing results. Here we demonstrated that in mice, rAAV vectors expressing the gene encoding HIV-1 gag stimulated gag-specific CD8(+) T cells, but these T cells failed to expand after a booster immunization with a replication-defective adenoviral (Ad) vector also expressing gag. We tested rAAV vectors of different serotypes expressing HIV-1 gag for induction of transgene product-specific CD8(+) T cells and found that the immunoinhibitory effect of rAAV priming observed with different AAV serotypes was transgene product specific, was independent of the interval between prime and boost, and extended to boosts with vaccine modalities other than Ad vectors. rAAV vector-induced CD8(+) T cells proliferated poorly, produced low levels of IFN-gamma in response to gag stimulation, and upregulated immunoinhibitory molecules. These T cells did not protect efficiently against challenge with a surrogate pathogen. Finally, we showed that the impaired proliferative capacity of the T cells was caused by persistence of the antigen-encoding rAAV vectors and could be reversed by placing the CD8(+) T cells in an antigen-free environment. Our data suggest that rAAV vectors induce functionally impaired T cells and could dampen the immune response to a natural infection.


Assuntos
Vacinas contra a AIDS/imunologia , Adenoviridae/imunologia , Linfócitos T CD8-Positivos/imunologia , Dependovirus/imunologia , Vetores Genéticos/imunologia , HIV-1/imunologia , Produtos do Gene gag do Vírus da Imunodeficiência Humana/imunologia , Vacinas contra a AIDS/genética , Adenoviridae/genética , Animais , Proliferação de Células , Dependovirus/genética , Vetores Genéticos/genética , Humanos , Imunização Secundária , Camundongos , Camundongos Endogâmicos BALB C , Transgenes/imunologia , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética
17.
J Virol ; 81(12): 6594-604, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17428852

RESUMO

In this study we compared a prime-boost regimen with two serologically distinct replication-defective adenovirus (Ad) vectors derived from chimpanzee serotypes C68 and C1 expressing Gag, Pol, gp140, and Nef of human immunodeficiency virus type 1 with a regimen in which replication-defective Ad vectors of the human serotype 5 (AdHu5) were given twice. Experiments were conducted in rhesus macaques that had or had not been preexposed to antigens of AdHu5. There was no significant difference in T-cell responses tested from peripheral blood of the different groups, although responses were overall highest in nonpreexposed animals immunized with the chimpanzee Ad vectors. Preexisting immunity to AdHu5 completely inhibited induction of transgene product-specific antibodies by the AdHu5 vectors without affecting antibody responses to the chimpanzee vectors. Upon euthanasia, T-cell responses were tested from a number of tissues. Preexisting immunity to AdHu5, commonly found in humans, changed the homing pattern of vaccine-induced T cells. In AdHu5-preexposed animals vaccinated with the chimpanzee Ad vectors, frequencies of transgene-specific T cells were higher in spleens than in blood, and in most preexposed animals vaccinated either with AdHu5 vectors or chimpanzee adenovirus vectors, frequencies of such T cells were exceptionally high in livers. The latter results indicate that analysis of T-cell responses solely from blood mononuclear cells of vaccine recipients may not suffice to compare the potencies of different vaccine regimens.


Assuntos
Adenoviridae/genética , Adenoviridae/imunologia , Pan troglodytes/metabolismo , Vacinas/química , Animais , Antígenos/química , Linhagem Celular , Feminino , Vetores Genéticos , Humanos , Leucócitos Mononucleares/virologia , Linfócitos/virologia , Macaca mulatta , Masculino , Peptídeos/química , Especificidade da Espécie , Transgenes
18.
Mol Ther ; 15(2): 393-403, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17235319

RESUMO

Recent studies have indicated that type I interferon (IFN) enhances antibody responses and promotes isotype switching. In this study, we analyzed the role of type I IFN signaling during the generation of transgene product-specific antibody responses elicited by recombinant adenovirus (Ad) vectors. A vector derived from a human Ad serotype (AdHu5) induced low levels of type I IFN following infection of dendritic cells (DCs) and stimulated normal transgene product-specific antibody responses in mice that have a defective type I IFN receptor (IFNAR(-/-)). A vector derived from a chimpanzee Ad serotype (AdC68) induced very high levels of type I IFN following infection of DCs, and surprisingly, primed stronger transgene product-specific antibody responses in IFNAR(-/-) mice compared to wild-type mice. The increased antibody response in IFNAR(-/-) mice vaccinated with the AdC68 vector was mainly due to the generation of IgG1 antibodies that were not elicited in wild-type mice. The induction of IgG1 antibodies correlated with an increase in transgene product expression in IFNAR(-/-) mice and was not associated with an increase in T helper 2 responses. We conclude that type I IFN, when induced at high levels, can downregulate transgene product expression of Ad vectors and inhibit the formation of optimal antibody responses.


Assuntos
Adenoviridae/genética , Vetores Genéticos/genética , Interferon Tipo I/fisiologia , Pan troglodytes/virologia , Animais , Células da Medula Óssea/imunologia , Citocinas/metabolismo , Células Dendríticas/imunologia , Ensaio de Imunoadsorção Enzimática , Humanos , Imunoglobulina G/imunologia , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/fisiologia , Transdução de Sinais/fisiologia , Células Th2/imunologia
19.
Mol Ther ; 15(3): 608-17, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17228314

RESUMO

A replication-defective chimeric vector based on the chimpanzee adenovirus serotype C1 was developed and tested as a vaccine carrier in mice. The AdC1 virus is closely related to human adenoviruses of subgroup B2 and uses CD46 for cell attachment. To overcome poor growth of E1-deleted AdC1 vectors on cell lines that provide the E1 of adenovirus of the human serotype 5 (AdHu5) virus in trans, the inverted terminal repeats and some of the early genes of AdC1 were replaced with those from AdC5, a chimpanzee origin adenovirus of subfamily E. The chimeric AdC1/C5 vector efficiently transduces CD46-expressing mouse dendritic cells (DCs) in vitro and initiates their maturation. Transduction of DCs in vivo is inefficient in CD46 transgenic mice. The AdC1/C5 vector induces transgene product-specific B- and CD8(+) T-cell responses in mice. Responses are slightly higher in wild-type mice than in CD46 transgenic mice. Transgene product-specific T-cell responses elicited by the AdC1/C5 vector can be increased by priming or boosting with a heterologous adenovirus vector. Pre-existing immunity to adenovirus of the common human serotype 5 does not affect induction of cell-mediated immune responses by the AdC1/C5 vector. This vector provides an additional tool in a repertoire of adenovirus-based vaccine vectors.


Assuntos
Adenoviridae/genética , Vetores Genéticos/genética , Proteína Cofatora de Membrana/metabolismo , Pan troglodytes/metabolismo , Vacinas/imunologia , Animais , Anticorpos/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Humanos , Proteína Cofatora de Membrana/genética , Camundongos , Pan troglodytes/genética , Ligação Proteica , Sensibilidade e Especificidade , Transgenes/genética
20.
Mol Ther ; 15(4): 792-800, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17245353

RESUMO

The goal of these studies was to test whether adeno-associated virus (AAV) capsid-specific CD8(+) T cells cause loss of hepatic AAV-mediated gene expression in experimental animals. Mice immunized with adenoviral vectors expressing AAV capsid or with AAV vectors developed CD8(+) T cells in blood, lymphatic tissues, and liver to epitopes shared between AAV2 and AAV8, and serotype-specific neutralizing antibodies. At the height of the T cells' effector phase, mice were infused with a heterologous AAV vector expressing human factor IX under a hepatocyte-specific promoter. Despite the presence of lytic CD8(+) T cells in the liver, hepatic Factor IX expression was sustained and comparable in AAV-preimmune and naïve animals. These results suggest that, in mice, pre-existing CD8(+) T cells to AAV capsid do not affect the longevity of AAV-mediated hepatic gene transfer. These results are in contrast to the outcome of a recent gene therapy trial of hemophilia B patients who were treated by hepatic gene transfer of AAV2 vectors expressing Factor IX. The loss of Factor IX expression, accompanied by a rise in liver enzymes and detectable frequencies of circulating AAV capsid-specific T cells, suggested T-cell-mediated destruction of transduced hepatocytes following reactivation of AAV-specific T cells upon AAV transfer.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Capsídeo/imunologia , Dependovirus/genética , Dependovirus/imunologia , Hepatócitos/imunologia , Hepatócitos/virologia , Transdução Genética , Transferência Adotiva , Animais , Fator IX/genética , Expressão Gênica , Terapia Genética/efeitos adversos , Vetores Genéticos/imunologia , Hemofilia B/genética , Hemofilia B/imunologia , Hemofilia B/terapia , Humanos , Depleção Linfocítica , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA