Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Nat Immunol ; 25(3): 537-551, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38337035

RESUMO

A nasally delivered chimpanzee adenoviral-vectored severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine (ChAd-SARS-CoV-2-S) is currently used in India (iNCOVACC). Here, we update this vaccine by creating ChAd-SARS-CoV-2-BA.5-S, which encodes a prefusion-stabilized BA.5 spike protein. Whereas serum neutralizing antibody responses induced by monovalent or bivalent adenoviral vaccines were poor against the antigenically distant XBB.1.5 strain and insufficient to protect in passive transfer experiments, mucosal antibody and cross-reactive memory T cell responses were robust, and protection was evident against WA1/2020 D614G and Omicron variants BQ.1.1 and XBB.1.5 in mice and hamsters. However, depletion of memory CD8+ T cells before XBB.1.5 challenge resulted in loss of protection against upper and lower respiratory tract infection. Thus, nasally delivered vaccines stimulate mucosal immunity against emerging SARS-CoV-2 strains, and cross-reactive memory CD8+ T cells mediate protection against lung infection by antigenically distant strains in the setting of low serum levels of cross-reactive neutralizing antibodies.


Assuntos
COVID-19 , Infecções Respiratórias , Vacinas , Cricetinae , Animais , Camundongos , Linfócitos T CD8-Positivos , SARS-CoV-2 , COVID-19/prevenção & controle , Anticorpos Neutralizantes , Anticorpos Amplamente Neutralizantes , Pan troglodytes
2.
Nat Immunol ; 25(10): 1913-1927, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39227514

RESUMO

A mucosal route of vaccination could prevent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication at the site of infection and limit transmission. We compared protection against heterologous XBB.1.16 challenge in nonhuman primates (NHPs) ~5 months following intramuscular boosting with bivalent mRNA encoding WA1 and BA.5 spike proteins or mucosal boosting with a WA1-BA.5 bivalent chimpanzee adenoviral-vectored vaccine delivered by intranasal or aerosol device. NHPs boosted by either mucosal route had minimal virus replication in the nose and lungs, respectively. By contrast, protection by intramuscular mRNA was limited to the lower airways. The mucosally delivered vaccine elicited durable airway IgG and IgA responses and, unlike the intramuscular mRNA vaccine, induced spike-specific B cells in the lungs. IgG, IgA and T cell responses correlated with protection in the lungs, whereas mucosal IgA alone correlated with upper airway protection. This study highlights differential mucosal and serum correlates of protection and how mucosal vaccines can durably prevent infection against SARS-CoV-2.


Assuntos
Anticorpos Antivirais , Vacinas contra COVID-19 , COVID-19 , Imunização Secundária , Imunoglobulina A , SARS-CoV-2 , Animais , Imunoglobulina A/imunologia , SARS-CoV-2/imunologia , COVID-19/prevenção & controle , COVID-19/imunologia , COVID-19/virologia , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Vacinas contra COVID-19/imunologia , Vacinas contra COVID-19/administração & dosagem , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/genética , Macaca mulatta , Adenoviridae/imunologia , Adenoviridae/genética , Imunidade nas Mucosas , Vacinas contra Adenovirus/imunologia , Vacinas contra Adenovirus/administração & dosagem , Feminino , Pulmão/virologia , Pulmão/imunologia , Linfócitos B/imunologia , Imunoglobulina G/imunologia , Imunoglobulina G/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/sangue , Administração Intranasal , Vacinação/métodos , Humanos
4.
Mol Ther ; 31(9): 2600-2611, 2023 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-37452494

RESUMO

B cells are the antibody-producing arm of the adaptive immune system and play a critical role in controlling pathogens. Several groups have now demonstrated the feasibility of using engineered B cells as a therapy, including infectious disease control and gene therapy of serum deficiencies. These studies have largely utilized ex vivo modification of the cells. Direct in vivo engineering would be of utility to the field, particularly in infectious disease control where the infrastructure needs of ex vivo cell modification would make a broad vaccination campaign highly challenging. In this study we demonstrate that engineered adenoviral vectors are capable of efficiently transducing murine and human primary B cells both ex vivo and in vivo. We found that unmodified human adenovirus C5 was capable of infecting B cells in vivo, likely due to interactions between the virus penton base protein and integrins. We further describe vector modification with B cell-specific gene promoters and successfully restrict transgene expression to B cells, resulting in a strong reduction in gene expression from the liver, the main site of human adenovirus C5 infection in vivo.


Assuntos
Adenoviridae , Doenças Transmissíveis , Camundongos , Humanos , Animais , Adenoviridae/genética , Vetores Genéticos/genética , Terapia Genética/métodos , Proteínas Virais/genética , Linfócitos B
5.
J Virol ; 94(10)2020 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-32102889

RESUMO

Human adenoviruses have many attractive features for gene therapy applications. However, the high prevalence of preexisting immunity against these viruses in general populations worldwide has greatly limited their clinical utility. In addition, the most commonly used human adenovirus, human adenovirus subgroup C serotype 5 (HAd5), when systemically administered, triggers systemic inflammation and toxicity, with the liver being the most severely affected organ. Here, we evaluated the utility and safety of a new low-seroprevalence gorilla adenovirus (GAd; GC46) as a gene transfer vector in mice. Biodistribution studies revealed that systemically administered GAd had a selective and robust lung endothelial cell (EC) tropism with minimal vector expression throughout many other organs and tissues. Administration of a high dose of GAd accomplished extensive transgene expression in the lung yet elicited no detectable inflammatory histopathology in this organ. Furthermore, GAd, unlike HAd5, did not exhibit hepatotropism or induce liver inflammatory toxicity in mice, demonstrating the exceptional safety profile of the vector vis-à-vis systemic utility. We further demonstrated that the GAd capsid fiber shared the flexibility of the HAd5 equivalent for permitting genetic modification; GAd with the pan-EC-targeting ligand myeloid cell-binding peptide (MBP) incorporated in the capsid displayed a reduced lung tropism and efficiently retargeted gene expression to vascular beds in other organs.IMPORTANCE In the aggregate, our mouse studies suggest that GAd is a promising gene therapy vector that utilizes lung ECs as a source of therapeutic payload production and a highly desirable toxicity profile. Further genetic engineering of the GAd capsid holds the promise of in vivo vector tropism modification and targeting.


Assuntos
Adenoviridae/genética , Capsídeo/metabolismo , Vetores Genéticos , Gorilla gorilla/virologia , Pulmão/metabolismo , Tropismo/imunologia , Infecções por Adenoviridae/patologia , Infecções por Adenoviridae/virologia , Adenovírus Humanos/genética , Animais , Proteínas do Capsídeo/genética , Células Endoteliais , Expressão Gênica , Terapia Genética , Fígado , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos C57BL , Estudos Soroepidemiológicos , Transdução Genética , Transgenes , Vírion
7.
Infect Immun ; 83(1): 286-91, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25368111

RESUMO

Hemolytic-uremic syndrome (HUS), caused by Shiga toxin (Stx)-producing Escherichia coli (STEC), remains untreatable. Production of human monoclonal antibodies against Stx, which are highly effective in preventing Stx sequelae in animal models, is languishing due to cost and logistics. We reported previously that the production and evaluation of a camelid heavy-chain-only VH domain (VHH)-based neutralizing agent (VNA) targeting Stx1 and Stx2 (VNA-Stx) protected mice from Stx1 and Stx2 intoxication. Here we report that a single intramuscular (i.m.) injection of a nonreplicating adenovirus (Ad) vector carrying a secretory transgene of VNA-Stx (Ad/VNA-Stx) protected mice challenged with Stx2 and protected gnotobiotic piglets infected with STEC from fatal systemic intoxication. One i.m. dose of Ad/VNA-Stx prevented fatal central nervous system (CNS) symptoms in 9 of 10 animals when it was given to piglets 24 h after bacterial challenge and in 5 of 9 animals when it was given 48 h after bacterial challenge, just prior to the onset of CNS symptoms. All 6 placebo animals died or were euthanized with severe CNS symptoms. Ad/VNA-Stx treatment had no impact on diarrhea. In conclusion, Ad/VNA-Stx treatment is effective in protecting piglets from fatal Stx2-mediated CNS complications following STEC challenge. With a low production cost and further development, this could presumably be an effective treatment for patients with HUS and/or individuals at high risk of developing HUS due to exposure to STEC.


Assuntos
Adenovírus Humanos/genética , Anticorpos Neutralizantes/uso terapêutico , Infecções por Escherichia coli/tratamento farmacológico , Escherichia coli O157/imunologia , Síndrome Hemolítico-Urêmica/tratamento farmacológico , Toxina Shiga I/antagonistas & inibidores , Toxina Shiga II/antagonistas & inibidores , Animais , Anticorpos Neutralizantes/genética , Modelos Animais de Doenças , Portadores de Fármacos/administração & dosagem , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Escherichia coli O157/genética , Feminino , Vetores Genéticos , Síndrome Hemolítico-Urêmica/imunologia , Síndrome Hemolítico-Urêmica/microbiologia , Injeções Intramusculares , Camundongos , Toxina Shiga I/imunologia , Toxina Shiga II/imunologia , Análise de Sobrevida , Suínos , Fatores de Tempo
8.
Mol Imaging ; 132014.
Artigo em Inglês | MEDLINE | ID: mdl-25249483

RESUMO

A significant limiting factor to the human clinical application of conditionally replicative adenovirus (CRAd)-based virotherapy is the inability to noninvasively monitor these agents and their potential persistence. To address this issue, we proposed a novel imaging approach that combines transient expression of the human somatostatin receptor (SSTR) subtype 2 reporter gene with genetic labeling of the viral capsid with mCherry fluorescent protein. To test this dual modality system, we constructed the Ad5/3Δ24pIXcherry/SSTR CRAd and validated its capacity to generate fluorescent and nuclear signals in vitro and following intratumoral injection. Analysis of 64Cu-CB-TE2A-Y3-TATE biodistribution in mice revealed reduced uptake in tumors injected with the imaging CRAd relative to the replication-incompetent, Ad-expressing SSTR2 but significantly greater uptake compared to the negative CRAd control. Optical imaging demonstrated relative correlation of fluorescent signal with virus replication as determined by viral genome quantification in tumors. Positron emission tomography/computed tomography studies demonstrated that we can visualize radioactive uptake in tumors injected with imaging CRAd and the trend for greater uptake by standardized uptake value analysis compared to control CRAd. In the aggregate, the plasticity of our dual imaging approach should provide the technical basis for monitoring CRAd biodistribution and persistence in preclinical studies while offering potential utility for a range of clinical applications.


Assuntos
Adenoviridae/fisiologia , Capsídeo/fisiologia , Substâncias Luminescentes/metabolismo , Proteínas Luminescentes/metabolismo , Neoplasias Ovarianas/virologia , Receptores de Somatostatina/metabolismo , Animais , Capsídeo/química , Linhagem Celular Tumoral , Complexos de Coordenação/farmacocinética , Feminino , Células HEK293 , Humanos , Camundongos , Transplante de Neoplasias , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Peptídeos/farmacocinética , Receptores de Somatostatina/genética , Replicação Viral , Proteína Vermelha Fluorescente
9.
Mol Ther ; 21(4): 758-66, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23319057

RESUMO

Hepatocellular carcinoma (HCC) is a highly fatal disease mandating development of novel, targeted therapies to elicit prolonged survival benefit to the patients. Insulin-like growth factor-binding protein-7 (IGFBP7), a secreted protein belonging to the IGFBP family, functions as a potential tumor suppressor for HCC. In the present study, we evaluated the therapeutic efficacy of a replication-incompetent adenovirus expressing IGFBP7 (Ad.IGFBP7) in human HCC. Ad.IGFBP7 profoundly inhibited viability and induced apoptosis in multiple human HCC cell lines by inducing reactive oxygen species (ROS) and activating a DNA damage response (DDR) and p38 MAPK. In orthotopic xenograft models of human HCC in athymic nude mice, intravenous administration of Ad.IGFBP7 profoundly inhibited primary tumor growth and intrahepatic metastasis. In a nude mice subcutaneous model, xenografts from human HCC cells were established in both flanks and only left-sided tumors received intratumoral injection of Ad.IGFBP7. Growth of both left-sided injected tumors and right-sided uninjected tumors were markedly inhibited by Ad.IGFBP7 with profound suppression of angiogenesis. These findings indicate that Ad.IGFBP7 might be a potent therapeutic eradicating both primary HCC and distant metastasis and might be an effective treatment option for terminal HCC patients.


Assuntos
Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/terapia , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/terapia , Adenoviridae/genética , Animais , Carcinoma Hepatocelular/genética , Humanos , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/genética , Neoplasias Hepáticas/genética , Masculino , Camundongos , Camundongos Nus , Espécies Reativas de Oxigênio/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Gynecol Oncol ; 130(3): 518-24, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23756180

RESUMO

OBJECTIVE: The conditionally replicative adenovirus Ad5/3-Δ24 has a type-3 knob incorporated into the type-5 fiber that facilitates enhanced ovarian cancer infectivity. Preclinical studies have shown that Ad5/3-Δ24 achieves significant oncolysis and anti-tumor activity in ovarian cancer models. The purpose of this study was to evaluate in a phase I trial the feasibility and safety of intraperitoneal (IP) Ad5/3-Δ24 in recurrent ovarian cancer patients. METHODS: Eligible patients were treated with IP Ad5/3-Δ24 for 3 consecutive days in one of three dose cohorts ranging 1 × 10(10)-1 × 10(12)vp. Toxicity was assessed utilizing CTC grading and efficacy with RECIST. Ascites, serum, and other samples were obtained to evaluate gene transfer, generation of wildtype virus, viral shedding, and antibody response. RESULTS: Nine of 10 patients completed treatment per protocol. A total of 15 vector-related adverse events were experienced in 5 patients. These events included fever or chills, nausea, fatigue, and myalgia. All were grades 1-2 in nature, transient, and medically managed. Of the 8 treated patients evaluable for response, six patients had stable disease and 2 patients had progressive disease. Three patients had decreased CA-125 from pretreatment levels one month after treatment. Ancillary biologic studies indicated Ad5/3-Δ24 replication in patients in the higher dose cohorts. All patients experienced an anti-adenoviral neutralizing antibody effect. CONCLUSIONS: This study suggests the feasibility and safety of a serotype chimeric infectivity-enhanced CRAd, Ad5/3-Δ24, as a potential therapeutic option for recurrent ovarian cancer patients.


Assuntos
Adenoviridae , Anticorpos Antivirais/sangue , Terapia Viral Oncolítica , Vírus Oncolíticos , Neoplasias Ovarianas/terapia , Adenoviridae/genética , Adenoviridae/fisiologia , Idoso , Idoso de 80 Anos ou mais , Ascite/virologia , Antígeno Ca-125/sangue , Calafrios/virologia , Progressão da Doença , Fadiga/virologia , Feminino , Febre/virologia , Expressão Gênica , Humanos , Dose Máxima Tolerável , Pessoa de Meia-Idade , Náusea/virologia , Terapia Viral Oncolítica/efeitos adversos , Vírus Oncolíticos/genética , Vírus Oncolíticos/fisiologia , Neoplasias Ovarianas/sangue , Neoplasias Ovarianas/virologia , Tomografia Computadorizada por Raios X , Replicação Viral , Eliminação de Partículas Virais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA