Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Hepatol ; 80(1): 31-40, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37827470

RESUMO

BACKGROUND & AIMS: Immunotherapy for chronic hepatitis B virus (HBV) infection has not yet demonstrated sufficient efficacy. We developed a non-integrative lentiviral-vectored therapeutic vaccine for chronic hepatitis B and tested its antiviral effects in HBV-persistent mice and two inactive HBsAg carriers. METHODS: Lentiviral vectors (LVs) encoding the core, preS1, or large HBsAg (LHBs) proteins of HBV were evaluated for immunogenicity in HBV-naïve mice and therapeutic efficacy in a murine model of chronic HBV infection. In addition, two inactive HBsAg carriers each received two doses of 5×107 transduction units (TU) or 1×108 TU of lentiviral-vectored LHBs (LV-LHBs), respectively. The endpoints were safety, LHBs-specific T-cell responses, and serum HBsAg levels during a 24-week follow-up. RESULTS: In the mouse models, LV-LHBs was the most promising in eliciting robust antigen-specific T cells and in reducing the levels of serum HBsAg and viral load. By the end of the 34-week observation period, six out of ten (60%) HBV-persistent mice vaccinated with LV-LHBs achieved serum HBsAg loss and significant depletion of HBV-positive hepatocytes in the liver. In the two inactive HBsAg carriers, vaccination with LV-LHBs induced a considerable increase in the number of peripheral LHBs-specific T cells in one patient, and a weak but detectable response in the other, accompanied by a sustained reduction of HBsAg (-0.31 log10 IU/ml and -0.46 log10 IU/ml, respectively) from baseline to nadir. CONCLUSIONS: A lentiviral-vectored therapeutic vaccine for chronic HBV infection demonstrated the potential to improve HBV-specific T-cell responses and deplete HBV-positive hepatocytes, leading to a sustained loss or reduction of serum HBsAg. IMPACT AND IMPLICATIONS: Chronic HBV infection is characterized by an extremely low number and profound hypo-responsiveness of HBV-specific T cells. Therapeutic vaccines are designed to improve HBV-specific T-cell responses. We show that immunization with a lentiviral-vectored therapeutic HBV vaccine was able to expand HBV-specific T cells in vivo, leading to reductions of HBV-positive hepatocytes and serum HBsAg.


Assuntos
Hepatite B Crônica , Humanos , Camundongos , Animais , Hepatite B Crônica/prevenção & controle , Hepatite B Crônica/tratamento farmacológico , Vírus da Hepatite B , Antígenos de Superfície da Hepatite B , Lentivirus/genética , Vacinas contra Hepatite B/uso terapêutico , Vacinação
2.
EMBO Rep ; 23(2): e54341, 2022 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-34914162

RESUMO

SARS-CoV-2 infection results in impaired interferon response in patients with severe COVID-19. However, how SARS-CoV-2 interferes with host immune responses is incompletely understood. Here, we sequence small RNAs from SARS-CoV-2-infected human cells and identify a microRNA (miRNA) derived from a recently evolved region of the viral genome. We show that the virus-derived miRNA produces two miRNA isoforms in infected cells by the enzyme Dicer, which are loaded into Argonaute proteins. Moreover, the predominant miRNA isoform targets the 3'UTR of interferon-stimulated genes and represses their expression in a miRNA-like fashion. Finally, the two viral miRNA isoforms were detected in nasopharyngeal swabs from COVID-19 patients. We propose that SARS-CoV-2 can potentially employ a virus-derived miRNA to hijack the host miRNA machinery, which could help to evade the interferon-mediated immune response.


Assuntos
COVID-19 , MicroRNAs , RNA Viral/genética , SARS-CoV-2/genética , Regiões 3' não Traduzidas , COVID-19/imunologia , Humanos , Imunidade , MicroRNAs/genética
3.
Mol Ther ; 30(9): 2984-2997, 2022 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-35484842

RESUMO

As the coronavirus disease 2019 (COVID-19) pandemic continues and new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern emerge, the adaptive immunity initially induced by the first-generation COVID-19 vaccines starts waning and needs to be strengthened and broadened in specificity. Vaccination by the nasal route induces mucosal, humoral, and cellular immunity at the entry point of SARS-CoV-2 into the host organism and has been shown to be the most effective for reducing viral transmission. The lentiviral vaccination vector (LV) is particularly suitable for this route of immunization owing to its non-cytopathic, non-replicative, and scarcely inflammatory properties. Here, to set up an optimized cross-protective intranasal booster against COVID-19, we generated an LV encoding stabilized spike of SARS-CoV-2 Beta variant (LV::SBeta-2P). mRNA vaccine-primed and -boosted mice, with waning primary humoral immunity at 4 months after vaccination, were boosted intranasally with LV::SBeta-2P. A strong boost effect was detected on cross-sero-neutralizing activity and systemic T cell immunity. In addition, mucosal anti-spike IgG and IgA, lung-resident B cells, and effector memory and resident T cells were efficiently induced, correlating with complete pulmonary protection against the SARS-CoV-2 Delta variant, demonstrating the suitability of the LV::SBeta-2P vaccine candidate as an intranasal booster against COVID-19. LV::SBeta-2P vaccination was also fully protective against Omicron infection of the lungs and central nervous system, in the highly susceptible B6.K18-hACE2IP-THV transgenic mice.


Assuntos
COVID-19 , Vacinas Virais , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , COVID-19/prevenção & controle , Vacinas contra COVID-19 , Humanos , Pulmão , Camundongos , Mucosa , SARS-CoV-2/genética , Vacinação , Vacinas Sintéticas , Vacinas de mRNA
4.
Mol Ther ; 28(8): 1772-1782, 2020 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-32485138

RESUMO

Zika virus, a member of the Flaviviridae family, is primarily transmitted by infected Aedes species mosquitoes. In 2016, Zika infection emerged as a global health emergency for its explosive spread and the remarkable neurological defects in the developing fetus. Development of a safe and effective Zika vaccine remains a high priority owing to the risk of re-emergence and limited understanding of Zika virus epidemiology. We engineered a non-integrating lentiviralvector(NILV)-based Zika vaccine encoding the consensus pre-membrane and envelope glycoprotein of circulating Zika virus strains. We further evaluated the immunogenicity and protective efficacy of this vaccine in both immunocompromised and immunocompetent mouse models. A single immunization in both mouse models elicited a robust neutralizing antibody titer and afforded full protection against Zika challenge as early as 7 days post-immunization. This NILV-based vaccine also induced a long-lasting immunity when immunized mice were challenged 6 months after immunization. Altogether, our NILV Zika vaccine provides a rapid yet durable protection through a single dose of immunization without extra adjuvant formulation. Our data suggest a promising Zika vaccine candidate for an emergency situation, and demonstrate the capacity of lentiviral vector as an efficient vaccine delivery platform.


Assuntos
Vetores Genéticos , Lentivirus , Vacinas de DNA/imunologia , Vacinas Virais/imunologia , Infecção por Zika virus/prevenção & controle , Zika virus/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Reações Cruzadas/imunologia , Modelos Animais de Doenças , Vetores Genéticos/genética , Interações Hospedeiro-Patógeno/imunologia , Imunização , Imunogenicidade da Vacina , Lentivirus/genética , Camundongos , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Proteínas do Envelope Viral/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
5.
J Immunol ; 193(3): 1504-11, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24973440

RESUMO

We generated a new humanized mouse model to study HLA-restricted immune responses. For this purpose, we created unique murine hosts by enforcing the expression of human SIRPα by murine phagocytes in murine MHC-deficient HLA-transgenic alymphoid hosts, an approach that allowed the immune reconstitution of nonpermissive mice following injection of human hematopoietic stem cells. We showed that these mouse/human chimeras were able to generate HLA-restricted responses to immunization. These new humanized mice may offer attractive models to study immune responses to human diseases, such as HIV and EBV infections, as well as to assay new vaccine strategies.


Assuntos
Antígenos HLA/administração & dosagem , Antígenos HLA/imunologia , Transplante de Células-Tronco Hematopoéticas/métodos , Quimera por Radiação/imunologia , Animais , Animais Recém-Nascidos , Antígenos de Diferenciação/administração & dosagem , Antígenos de Diferenciação/sangue , Antígenos de Diferenciação/genética , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Modelos Animais de Doenças , Feminino , Antígenos HLA/genética , Humanos , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Quimera por Radiação/genética , Receptores Imunológicos/administração & dosagem , Receptores Imunológicos/sangue , Receptores Imunológicos/genética
6.
Gut ; 64(12): 1961-71, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25429051

RESUMO

OBJECTIVE: To assess a new adenovirus-based immunotherapy as a novel treatment approach to chronic hepatitis B (CHB). METHODS: TG1050 is a non-replicative adenovirus serotype 5 encoding a unique large fusion protein composed of a truncated HBV Core, a modified HBV Polymerase and two HBV Envelope domains. We used a recently described HBV-persistent mouse model based on a recombinant adenovirus-associated virus encoding an over length genome of HBV that induces the chronic production of HBsAg, HBeAg and infectious HBV particles to assess the ability of TG1050 to induce functional T cells in face of a chronic status. RESULTS: In in vitro studies, TG1050 was shown to express the expected large polyprotein together with a dominant, smaller by-product. Following a single administration in mice, TG1050 induced robust, multispecific and long-lasting HBV-specific T cells detectable up to 1 year post-injection. These cells target all three encoded immunogens and display bifunctionality (i.e., capacity to produce both interferon γ and tumour necrosis factor α as well as cytolytic functions). In addition, control of circulating levels of HBV DNA and HBsAg was observed while alanine aminotransferase levels remain in the normal range. CONCLUSIONS: Injection of TG1050 induced both splenic and intrahepatic functional T cells producing cytokines and displaying cytolytic activity in HBV-naïve and HBV-persistent mouse models together with significant reduction of circulating viral parameters. These results warrant clinical evaluation of TG1050 in the treatment of CHB.


Assuntos
Adenoviridae/metabolismo , Linfócitos T CD8-Positivos/metabolismo , DNA Viral/sangue , Vírus da Hepatite B/imunologia , Hepatite B Crônica/terapia , Imunoterapia/métodos , Proteínas Virais de Fusão/imunologia , Adenoviridae/classificação , Alanina Transaminase/sangue , Animais , DNA Polimerase Dirigida por DNA/genética , DNA Polimerase Dirigida por DNA/imunologia , Modelos Animais de Doenças , Produtos do Gene env/genética , Produtos do Gene env/imunologia , Vetores Genéticos , Antígeno HLA-A2/genética , Antígenos do Núcleo do Vírus da Hepatite B/genética , Antígenos do Núcleo do Vírus da Hepatite B/imunologia , Antígenos de Superfície da Hepatite B/sangue , Hepatite B Crônica/sangue , Interferon gama/sangue , Contagem de Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fatores de Tempo , Fator de Necrose Tumoral alfa/sangue , Proteínas Virais de Fusão/genética , Carga Viral
7.
J Virol ; 88(5): 3004-15, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24371056

RESUMO

UNLABELLED: We previously reported a proof-of-concept study for curing chronic hepatitis B virus (HBV) infection using a foreign-antigen recombinant HBV (rHBV) as a gene therapy vector. Targeted elimination of wild-type HBV (wtHBV)-infected cells could be achieved by functionally activating an in situ T-cell response against the foreign antigen. However, as chronic HBV infection spreads to all hepatocytes, specific targeting of virus-infected cells is thought to be less critical. It is also feared that rHBV may not induce active immunization in a setting resembling natural infection. For this immunotherapeutic approach to be practically viable, in the present study, we used a recombinant adenovirus (rAd) vector for rHBV delivery. The rAd vector allowed efficient transduction of wtHBV-producing HepG2 cells, with transferred rHBV undergoing dominant viral replication. Progeny rHBV virions proved to be infectious, as demonstrated in primary tupaia hepatocytes. These results greatly expanded the antiviral capacity of the replication-defective rAd/rHBV in wtHBV-infected liver tissue. With prior priming in the periphery, transduction with rAd/rHBV attracted a substantial influx of the foreign-antigen-specific T-effector cells into the liver. Despite the fully activated T-cell response, active expression of rHBV was observed for a prolonged time, which is essential for rHBV to achieve sustained expansion. In a mouse model of HBV persistence established by infection with a recombinant adeno-associated virus carrying the wtHBV genome, rAd/rHBV-based immunotherapy elicited a foreign-antigen-specific T-cell response that triggered effective viral clearance and subsequent seroconversion to HBV. It therefore represents an efficient strategy to overcome immune tolerance, thereby eliminating chronic HBV infection. IMPORTANCE: Adenovirus-delivered rHBV activated a foreign-antigen-specific T-cell response that abrogated HBV persistence in a mouse model. Our study provides further evidence of the potential of foreign-antigen-based immunotherapy for the treatment of chronic HBV infection.


Assuntos
Adenoviridae/genética , Epitopos/genética , Vetores Genéticos/genética , Vírus da Hepatite B/genética , Hepatite B Crônica/genética , Hepatite B Crônica/imunologia , Adenoviridae/imunologia , Administração Intravenosa , Animais , Linhagem Celular , Modelos Animais de Doenças , Epitopos/imunologia , Expressão Gênica , Ordem dos Genes , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Vírus da Hepatite B/imunologia , Vírus da Hepatite B/fisiologia , Hepatite B Crônica/terapia , Hepatócitos/metabolismo , Hepatócitos/virologia , Humanos , Imunização , Imunoterapia , Fígado/imunologia , Fígado/metabolismo , Fígado/virologia , Ativação Linfocitária , Masculino , Camundongos , Camundongos Transgênicos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Subpopulações de Linfócitos T/imunologia , Transdução Genética , Proteínas do Core Viral/genética , Proteínas do Core Viral/imunologia , Vírion/fisiologia , Montagem de Vírus , Replicação Viral
8.
Med Microbiol Immunol ; 204(1): 121-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25511871

RESUMO

The antiviral treatment of chronic hepatitis B virus (HBV) infection has greatly improved over the last 20 years since it has allowed a disappearance of cirrhosis decompensation and a significant reduction of the incidence of hepatocellular carcinoma. However, a complete HBV cure has not been achieved, and alternative treatments are still needed to optimize the current treatments. Therapeutic vaccination is a promising new strategy for controlling persistent infections and tumors. However, this approach has not been as successful as initially anticipated for chronic hepatitis B. General impairment of the immune responses generated during persistent HBV infection, with exhausted T cells not responding correctly to therapeutic vaccination, is most likely responsible for the poor clinical responses observed to date. We describe here the past approaches of therapeutic vaccination, in the hope that useful lessons will emerge from these previous clinical trials. Intensive research efforts are now focusing on a better understanding of immune responses in liver, on mechanisms by which HBV escapes innate immunity and on an accurate selection of the patients susceptible to benefit of immune therapy, which could increase the efficacy of therapeutic vaccination.


Assuntos
Vacinas contra Hepatite B/uso terapêutico , Hepatite B Crônica/terapia , Imunoterapia/métodos , Ensaios Clínicos como Assunto , Hepatite B Crônica/imunologia , Humanos , Resultado do Tratamento
9.
J Virol ; 87(10): 5554-63, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23468504

RESUMO

Hepatitis B virus (HBV) persistence may be due to impaired HBV-specific immune responses being unable to eliminate efficiently or cure infected hepatocytes. The immune mechanisms that lead to HBV persistence have not been completely identified, and no appropriate animal model is available for such studies. Therefore, we established a chronic HBV infection model in a mouse strain with human leukocyte antigen A2/DR1 (HLA-A2/DR1) transgenes and an H-2 class I/class II knockout. The liver of these mice was transduced with adeno-associated virus serotype 2/8 (AAV2/8) carrying a replication-competent HBV DNA genome. In all AAV2/8-transduced mice, hepatitis B virus surface antigen, hepatitis B virus e antigen, and HBV DNA persisted in serum for at least 1 year. Viral replication intermediates and transcripts were detected in the livers of the AAV-injected mice. The hepatitis B core antigen was expressed in 60% of hepatocytes. No significant inflammation was observed in the liver. This was linked to a higher number of regulatory T cells in liver than in controls and a defect in HBV-specific functional T-cell responses. Despite the substantial tolerance resulting from expression of HBV antigens in hepatocytes, we succeeded in priming functional HBV-specific T-cell responses in peripheral tissues, which subsequently reached the liver. This AAV2/8-HBV-transduced HLA-A2/DR1 murine model recapitulates virological and immunological characteristics of chronic HBV infection, and it could be useful for the development of new treatments and immune-based therapies or therapeutic vaccines for chronic HBV infections.


Assuntos
Modelos Animais de Doenças , Antígeno HLA-A2/metabolismo , Antígeno HLA-DR1/metabolismo , Vírus da Hepatite B/patogenicidade , Replicação Viral , Animais , DNA Viral/sangue , Dependovirus/genética , Feminino , Deleção de Genes , Vetores Genéticos , Antígenos H-2/genética , Antígeno HLA-A2/genética , Antígeno HLA-DR1/genética , Antígenos de Superfície da Hepatite B/sangue , Antígenos E da Hepatite B/sangue , Vírus da Hepatite B/fisiologia , Humanos , Fígado/virologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Transgenes
10.
Pharmaceutics ; 15(3)2023 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-36986707

RESUMO

Lentiviral vectors are among the most effective viral vectors for vaccination. In clear contrast to the reference adenoviral vectors, lentiviral vectors have a high potential for transducing dendritic cells in vivo. Within these cells, which are the most efficient at activating naive T cells, lentiviral vectors induce endogenous expression of transgenic antigens that directly access antigen presentation pathways without the need for external antigen capture or cross-presentation. Lentiviral vectors induce strong, robust, and long-lasting humoral, CD8+ T-cell immunity and effective protection against several infectious diseases. There is no pre-existing immunity to lentiviral vectors in the human population and the very low pro-inflammatory properties of these vectors pave the way for their use in mucosal vaccination. In this review, we have mainly summarized the immunological aspects of lentiviral vectors, their recent optimization to induce CD4+ T cells, and our recent data on lentiviral vector-based vaccination in preclinical models, including prophylaxis against flaviviruses, SARS-CoV-2, and Mycobacterium tuberculosis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA