Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 196(8): 3354-63, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26944930

RESUMO

The establishment of robust T cell memory is critical for the development of novel vaccines for infections and cancers. Classical memory generated by CD8(+)T cells is characterized by contracted populations homing to lymphoid organs. T cell memory inflation, as seen for example after CMV infection, is the maintenance of expanded, functional, tissue-associated effector memory cell pools. Such memory pools may also be induced after adenovirus vaccination, and we recently defined common transcriptional and phenotypic features of these populations in mice and humans. However, the rules that govern which epitopes drive memory inflation compared with classical memory are not fully defined, and thus it is not currently possible to direct this process. We used our adenoviral model of memory inflation to first investigate the role of the promoter and then the role of the epitope context in determining memory formation. Specifically, we tested the hypothesis that conventional memory could be converted to inflationary memory by simple presentation of the Ag in the form of minigene vectors. When epitopes from LacZ and murine CMV that normally induce classical memory responses were presented as minigenes, they induced clear memory inflation. These data demonstrate that, regardless of the transgene promoter, the polypeptide context of a CD8(+)T cell epitope may determine whether classical or inflating memory responses are induced. The ability to direct this process by the use of minigenes is relevant to the design of vaccines and understanding of immune responses to pathogens.


Assuntos
Adenoviridae/imunologia , Antígenos/imunologia , Linfócitos T CD8-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Memória Imunológica/imunologia , Adenoviridae/genética , Animais , Citomegalovirus/imunologia , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Epitopos de Linfócito T/genética , Feminino , Óperon Lac/genética , Óperon Lac/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas/genética , Transgenes/genética , Vacinação
2.
Eur Child Adolesc Psychiatry ; 26(11): 1283-1307, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28429134

RESUMO

Children, adolescents and adults with attention-deficit/hyperactivity disorder (ADHD) experience functional impairment and poor health-related quality of life (HRQoL) in addition to symptoms of inattention/hyperactivity-impulsivity. To synthesize qualitatively the published evidence from randomized, double-blind, placebo-controlled trials of the effectiveness of pharmacotherapy on functional impairment or HRQoL in patients with ADHD, a systematic PubMed searching and screening strategy was designed to identify journal articles meeting pre-specified criteria. Post hoc analyses and meta-analyses were excluded. HRQoL outcomes, functional outcomes and the principal ADHD symptom-based outcome were extracted from included studies. An effect size of 0.5 versus placebo was used as a threshold for potential clinical relevance (unreported effect sizes were calculated when possible). Of 291 records screened, 35 articles describing 34 studies were included. HRQoL/functioning was usually self-rated in adults and proxy-rated in children/adolescents. Baseline data indicated substantial HRQoL deficits in children/adolescents. Placebo-adjusted effects of medication on ADHD symptoms, HRQoL and functioning, respectively, were statistically or nominally significant in 18/18, 10/12 and 7/9 studies in children/adolescents and 14/16, 9/11 and 9/10 studies in adults. Effect sizes were ≥0.5 versus placebo for symptoms, HRQoL and functioning, respectively, in 14/16, 7/9 and 4/8 studies in children/adolescents; and 6/12, 1/6 and 1/8 studies in adults. Effect sizes were typically larger for stimulants than for non-stimulants, for symptoms than for HRQoL/functioning, and for children/adolescents than for adults. The efficacy of ADHD medication extends beyond symptom control and may help reduce the related but distinct functional impairments and HRQoL deficits in patients with ADHD.


Assuntos
Transtorno do Deficit de Atenção com Hiperatividade/tratamento farmacológico , Qualidade de Vida/psicologia , Criança , Pré-Escolar , Humanos , Ensaios Clínicos Controlados Aleatórios como Assunto , Resultado do Tratamento
3.
Virol J ; 10: 349, 2013 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-24304565

RESUMO

BACKGROUND: Rift Valley Fever (RVF) is a viral zoonosis that historically affects livestock production and human health in sub-Saharan Africa, though epizootics have also occurred in the Arabian Peninsula. Whilst an effective live-attenuated vaccine is available for livestock, there is currently no licensed human RVF vaccine. Replication-deficient chimpanzee adenovirus (ChAd) vectors are an ideal platform for development of a human RVF vaccine, given the low prevalence of neutralizing antibodies against them in the human population, and their excellent safety and immunogenicity profile in human clinical trials of vaccines against a wide range of pathogens. METHODS: Here, in BALB/c mice, we evaluated the immunogenicity and efficacy of a replication-deficient chimpanzee adenovirus vector, ChAdOx1, encoding the RVF virus envelope glycoproteins, Gn and Gc, which are targets of virus neutralizing antibodies. The ChAdOx1-GnGc vaccine was assessed in comparison to a replication-deficient human adenovirus type 5 vector encoding Gn and Gc (HAdV5-GnGc), a strategy previously shown to confer protective immunity against RVF in mice. RESULTS: A single immunization with either of the vaccines conferred protection against RVF virus challenge eight weeks post-immunization. Both vaccines elicited RVF virus neutralizing antibody and a robust CD8+ T cell response. CONCLUSIONS: Together the results support further development of RVF vaccines based on replication-deficient adenovirus vectors, with ChAdOx1-GnGc being a potential candidate for use in future human clinical trials.


Assuntos
Adenoviridae/genética , Portadores de Fármacos , Vetores Genéticos , Febre do Vale de Rift/prevenção & controle , Vírus da Febre do Vale do Rift/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Febre do Vale de Rift/imunologia , Vírus da Febre do Vale do Rift/genética , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
4.
Mol Ther ; 20(8): 1633-47, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22354374

RESUMO

Substantial protection can be provided against the pre-erythrocytic stages of malaria by vaccination first with an adenoviral and then with an modified vaccinia virus Ankara (MVA) poxviral vector encoding the same ME.TRAP transgene. We investigated whether the two vaccine components adenovirus (Ad) and MVA could be coinjected as a mixture to enhance protection against malaria. A single-shot mixture at specific ratios of Ad and MVA (Ad+MVA) enhanced CD8(+) T cell-dependant protection of mice against challenge with Plasmodium berghei. Moreover, the degree of protection could be enhanced after homologous boosting with the same Ad+MVA mixture to levels comparable with classic heterologous Ad prime-MVA boost regimes. The mixture increased transgene-specific responses while decreasing the CD8(+) T cell antivector immunity compared to each vector used alone, particularly against the MVA backbone. Mixed vector immunization led to increased early circulating interferon-γ (IFN-γ) response levels and altered transcriptional microarray profiles. Furthermore, we found that sequential immunizations with the Ad+MVA mixture led to consistent boosting of the transgene-specific CD8(+) response for up to three mixture immunizations, whereas each vector used alone elicited progressively lower responses. Our findings offer the possibility of simplifying the deployment of viral vectors as a single mixture product rather than in heterologous prime-boost regimens.


Assuntos
Adenoviridae/imunologia , Imunização/métodos , Vaccinia virus/imunologia , Adenoviridae/genética , Animais , Linfócitos T CD8-Positivos/imunologia , Feminino , Vetores Genéticos/genética , Malária/imunologia , Malária/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Plasmodium berghei/imunologia , Vaccinia virus/genética
5.
Biotechnol Bioeng ; 109(3): 719-28, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22252512

RESUMO

First-generation, E1/E3-deleted adenoviral vectors with diverse transgenes are produced routinely in laboratories worldwide for development of novel prophylactics and therapies for a variety of applications, including candidate vaccines against important infectious diseases, such as HIV/AIDS, tuberculosis, and malaria. Here, we show, for two different transgenes (both encoding malarial antigens) inserted at the E1 locus, that rare viruses containing a transgene-inactivating mutation exhibit a selective growth advantage during propagation in E1-complementing HEK293 cells, such that they rapidly become the major or sole species in the viral population. For one of these transgenes, we demonstrate that viral yield and cytopathic effect are enhanced by repression of transgene expression in the producer cell line, using the tetracycline repressor system. In addition to these transgene-inactivating mutations, one of which occurred during propagation of the pre-viral genomic clone in bacteria, and the other after viral reconstitution in HEK293 cells, we describe two other types of mutation, a small deletion and a gross rearranging duplication, in one of the transgenes studied. These were of uncertain origin, and the effects on transgene expression and viral growth were not fully characterized. We demonstrate that, together with minor protocol modifications, repression of transgene expression in HEK293 cells during viral propagation enables production of a genetically stable chimpanzee adenovirus vector expressing a malarial antigen which had previously been impossible to derive. These results have important implications for basic and pre-clinical studies using adenoviral vectors and for derivation of adenoviral vector products destined for large-scale amplification during biomanufacture.


Assuntos
Adenoviridae/genética , Rearranjo Gênico , Vetores Genéticos , Adenoviridae/crescimento & desenvolvimento , Proteínas E1 de Adenovirus/genética , Linhagem Celular , Efeito Citopatogênico Viral , Genoma Viral , Instabilidade Genômica , Humanos , Recombinação Genética , Carga Viral , Replicação Viral
6.
Sci Rep ; 8(1): 15020, 2018 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-30301933

RESUMO

Despite promising progress in malaria vaccine development, an efficacious subunit vaccine against P. falciparum remains to be licensed and deployed. This study aimed to improve on the immunogenicity of the leading liver-stage vaccine candidate (ChAd63-MVA ME-TRAP), known to confer protection by eliciting high levels of antigen-specific CD8+ T cells. We previously showed fusion of ME-TRAP to the human MHC class II invariant chain (Ii) could enhance CD8+ T cell responses in non-human primates, but did not progress to clinical testing due to potential risk of auto-immunity by vaccination of humans with a self-antigen. Initial immunogenicity analyses of ME-TRAP fused to subdomains of the Ii showed that the Ii transmembrane domain alone can enhance CD8+ T cell responses. Subsequently, truncated Ii sequences with low homology to human Ii were developed and shown to enhance CD8+ T cell responses. By systematically mutating the TM domain sequence, multimerization of the Ii chain was shown to be important for immune enhancement. We subsequently identified several proteins from a variety of microbial pathogens with similar characteristics, that also enhance the CD8+ T cell response and could therefore be used in viral vector vaccines when potent cell mediated immunity is required.


Assuntos
Antígenos de Protozoários/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Farmacêuticos/administração & dosagem , Animais , Anticorpos Antiprotozoários/imunologia , Antígenos de Diferenciação de Linfócitos B/imunologia , Autoimunidade/efeitos dos fármacos , Linfócitos T CD8-Positivos/virologia , Epitopos/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Malária Falciparum/parasitologia , Malária Falciparum/prevenção & controle , Plasmodium falciparum/imunologia , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/imunologia , Vaccinia virus/genética
7.
Sci Rep ; 7: 46621, 2017 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-28422178

RESUMO

The leading malaria vaccine in development is the circumsporozoite protein (CSP)-based particle vaccine, RTS,S, which targets the pre-erythrocytic stage of Plasmodium falciparum infection. It induces modest levels of protective efficacy, thought to be mediated primarily by CSP-specific antibodies. We aimed to enhance vaccine efficacy by generating a more immunogenic CSP-based particle vaccine and therefore developed a next-generation RTS,S-like vaccine, called R21. The major improvement is that in contrast to RTS,S, R21 particles are formed from a single CSP-hepatitis B surface antigen (HBsAg) fusion protein, and this leads to a vaccine composed of a much higher proportion of CSP than in RTS,S. We demonstrate that in BALB/c mice R21 is immunogenic at very low doses and when administered with the adjuvants Abisco-100 and Matrix-M it elicits sterile protection against transgenic sporozoite challenge. Concurrent induction of potent cellular and humoral immune responses was also achieved by combining R21 with TRAP-based viral vectors and protective efficacy was significantly enhanced. In addition, in contrast to RTS,S, only a minimal antibody response to the HBsAg carrier was induced. These studies identify an anti-sporozoite vaccine component that may improve upon the current leading malaria vaccine RTS,S. R21 is now under evaluation in Phase 1/2a clinical trials.


Assuntos
Antígenos de Superfície da Hepatite B , Imunogenicidade da Vacina , Vacinas Antimaláricas , Malária Falciparum , Plasmodium falciparum/imunologia , Vacinas de Partículas Semelhantes a Vírus , Animais , Anticorpos Antiprotozoários/imunologia , Feminino , Antígenos de Superfície da Hepatite B/química , Antígenos de Superfície da Hepatite B/imunologia , Antígenos de Superfície da Hepatite B/farmacologia , Vacinas Antimaláricas/química , Vacinas Antimaláricas/imunologia , Vacinas Antimaláricas/farmacologia , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Vacinas de Partículas Semelhantes a Vírus/química , Vacinas de Partículas Semelhantes a Vírus/imunologia , Vacinas de Partículas Semelhantes a Vírus/farmacologia
8.
Sci Rep ; 5: 16756, 2015 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-26576856

RESUMO

Replication defective adenoviruses are promising vectors for the delivery of vaccine antigens. However, the potential of a vector to elicit transgene-specific adaptive immune responses is largely dependent on the viral serotype used. HAdV-5 (Human adenovirus C) vectors are more immunogenic than chimpanzee adenovirus vectors from species Human adenovirus E (ChAdOx1 and AdC68) in mice, though the mechanisms responsible for these differences in immunogenicity remain poorly understood. In this study, superior immunogenicity was associated with markedly higher levels of transgene expression in vivo, particularly within draining lymph nodes. To investigate the viral factors contributing to these phenotypes, we generated recombinant ChAdOx1 vectors by exchanging components of the viral capsid reported to be principally involved in cell entry with the corresponding sequences from HAdV-5. Remarkably, pseudotyping with the HAdV-5 fiber and/or penton RGD loop had little to no effect on in vivo transgene expression or transgene-specific adaptive immune responses despite considerable species-specific sequence heterogeneity in these components. Our results suggest that mechanisms governing vector transduction after intramuscular administration in mice may be different from those described in vitro.


Assuntos
Adenoviridae/imunologia , Adenovírus Humanos/imunologia , Proteínas do Capsídeo/imunologia , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Receptores de Orexina/genética , Adenoviridae/genética , Adenovírus Humanos/genética , Animais , Anticorpos Antivirais/imunologia , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Proteínas do Capsídeo/química , Expressão Gênica , Vetores Genéticos/administração & dosagem , Humanos , Imunidade , Imunização , Injeções Intramusculares , Camundongos , Pan troglodytes , Transdução Genética , Transgenes , Vacinas/genética , Vacinas/imunologia
9.
Vaccine ; 33(9): 1121-8, 2015 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-25629523

RESUMO

Adenovirus vaccine vectors generated from new viral serotypes are routinely screened in pre-clinical laboratory animal models to identify the most immunogenic and efficacious candidates for further evaluation in clinical human and veterinary settings. Here, we show that studies in a laboratory species do not necessarily predict the hierarchy of vector performance in other mammals. In mice, after intramuscular immunization, HAdV-5 (Human adenovirus C) based vectors elicited cellular and humoral adaptive responses of higher magnitudes compared to the chimpanzee adenovirus vectors ChAdOx1 and AdC68 from species Human adenovirus E. After HAdV-5 vaccination, transgene specific IFN-γ(+) CD8(+) T cell responses reached peak magnitude later than after ChAdOx1 and AdC68 vaccination, and exhibited a slower contraction to a memory phenotype. In cattle, cellular and humoral immune responses were at least equivalent, if not higher, in magnitude after ChAdOx1 vaccination compared to HAdV-5. Though we have not tested protective efficacy in a disease model, these findings have important implications for the selection of candidate vectors for further evaluation. We propose that vaccines based on ChAdOx1 or other Human adenovirus E serotypes could be at least as immunogenic as current licensed bovine vaccines based on HAdV-5.


Assuntos
Adenoviridae/genética , Portadores de Fármacos , Vetores Genéticos , Proteínas Recombinantes/imunologia , Transgenes , Vacinas Virais/imunologia , Animais , Animais de Laboratório , Linfócitos T CD8-Positivos/imunologia , Bovinos , Interferon gama/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/genética , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
10.
Sci Rep ; 5: 11820, 2015 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-26139288

RESUMO

The development of an efficacious Plasmodium falciparum malaria vaccine remains a top priority for global health. Vaccination with irradiated sporozoites is able to provide complete sterile protection through the action of CD8(+) T cells at the liver-stage of infection. However, this method is currently unsuitable for large-scale deployment and focus has instead turned to the development of sub-unit vaccines. Sub-unit vaccine efforts have traditionally focused on two well-known pre-erythrocytic antigens, CSP and TRAP, yet thousands of genes are expressed in the liver-stage. We sought to assess the ability of eight alternative P. falciparum pre-erythrocytic antigens to induce a high proportion of CD8(+) T cells. We show that all antigens, when expressed individually in the non-replicating viral vectors ChAd63 and MVA, are capable of inducing an immune response in mice. Furthermore, we also developed chimeric P. berghei parasites expressing the cognate P. falciparum antigen to enable assessment of efficacy in mice. Our preliminary results indicate that vectors encoding either PfLSA1 or PfLSAP2 are capable of inducing sterile protection dependent on the presence of CD8(+) T cells. This work has identified two promising P. falciparum liver-stage candidate antigens that will now undergo further testing in humans.


Assuntos
Antígenos de Protozoários/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/prevenção & controle , Animais , Linfócitos T CD8-Positivos/imunologia , Vetores Genéticos , Antígenos de Histocompatibilidade Classe II/administração & dosagem , Humanos , Fígado/imunologia , Vacinas Antimaláricas/administração & dosagem , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Camundongos , Plasmodium falciparum/imunologia , Plasmodium falciparum/patogenicidade , Vacinação
11.
PLoS One ; 9(8): e105520, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25140889

RESUMO

T cells play a central role in the immune response to many of the world's major infectious diseases. In this study we investigated the tumour necrosis factor receptor superfamily costimulatory molecule, 4-1BBL (CD137L, TNFSF9), for its ability to increase T cell immunogenicity induced by a variety of recombinant vectored vaccines. To efficiently test this hypothesis, we assessed a number of promoters and developed a stable bi-cistronic vector expressing both the antigen and adjuvant. Co-expression of 4-1BBL, together with our model antigen TIP, was shown to increase the frequency of murine antigen-specific IFN-γ secreting CD8(+) T cells in three vector platforms examined. Enhancement of the response was not limited by co-expression with the antigen, as an increase in CD8(+) immunogenicity was also observed by co-administration of two vectors each expressing only the antigen or adjuvant. However, when this regimen was tested in non-human primates using a clinical malaria vaccine candidate, no adjuvant effect of 4-1BBL was observed limiting its potential use as a single adjuvant for translation into a clinical vaccine.


Assuntos
Ligante 4-1BB/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Sintéticas/imunologia , Animais , Feminino , Macaca mulatta , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Especificidade da Espécie
12.
PLoS One ; 9(6): e100538, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24945248

RESUMO

The orthodox role of the invariant chain (CD74; Ii) is in antigen presentation to CD4+ T cells, but enhanced CD8+ T cells responses have been reported after vaccination with vectored viral vaccines encoding a fusion of Ii to the antigen of interest. In this study we assessed whether fusion of the malarial antigen, ME-TRAP, to Ii could increase the vaccine-induced CD8+ T cell response. Following single or heterologous prime-boost vaccination of mice with a recombinant chimpanzee adenovirus vector, ChAd63, or recombinant modified vaccinia virus Ankara (MVA), higher frequencies of antigen-specific CD4+ and CD8+ T cells were observed, with the largest increases observed following a ChAd63-MVA heterologous prime-boost regimen. Studies in non-human primates confirmed the ability of Ii-fusion to augment the T cell response, where a 4-fold increase was maintained up to 11 weeks after the MVA boost. Of the numerous different approaches explored to increase vectored vaccine induced immunogenicity over the years, fusion to the invariant chain showed a consistent enhancement in CD8+ T cell responses across different animal species and may therefore find application in the development of vaccines against human malaria and other diseases where high levels of cell-mediated immunity are required.


Assuntos
Antígenos de Diferenciação de Linfócitos B/imunologia , Antígenos de Protozoários/imunologia , Linfócitos T CD8-Positivos/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Vacinas Antimaláricas/imunologia , Proteínas Recombinantes de Fusão/imunologia , Animais , Galinhas , Feminino , Vetores Genéticos/metabolismo , Humanos , Imunização , Macaca mulatta/imunologia , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR
13.
Vaccine ; 31(39): 4247-51, 2013 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-23523407

RESUMO

Diseases such as HIV/AIDS, tuberculosis, malaria and cancer are prime targets for prophylactic or therapeutic vaccination, but have proven partially or wholly resistant to traditional approaches to vaccine design. New vaccines based on recombinant viral vectors expressing a foreign antigen are under intense development for these and other indications. One of the most advanced and most promising vectors is the attenuated, non-replicating poxvirus MVA (modified vaccinia virus Ankara), a safer derivative of the uniquely successful smallpox vaccine. Despite the ability of recombinant MVA to induce potent humoral and cellular immune responses against transgenic antigen in humans, especially when used as the latter element of a heterologous prime-boost regimen, doubts are occasionally expressed about the ultimate feasibility of this approach. In this review, five common misconceptions over recombinant MVA are discussed, and evidence is cited to show that recombinant MVA is at least sufficiently genetically stable, manufacturable, safe, and immunogenic (even in the face of prior anti-vector immunity) to warrant reasonable hope over the feasibility of large-scale deployment, should useful levels of protection against target pathogens, or therapeutic benefit for cancer, be demonstrated in efficacy trials.


Assuntos
Vacinas de DNA/imunologia , Vacinas Virais/imunologia , Animais , Vetores Genéticos , Humanos , Neoplasias/terapia , Vacinação , Vacinas Atenuadas , Vacinas de DNA/efeitos adversos , Vacinas de DNA/genética , Vacinas de DNA/uso terapêutico , Vaccinia virus/genética , Vaccinia virus/imunologia , Vacinas Virais/efeitos adversos , Vacinas Virais/genética
14.
Vaccine ; 31(39): 4223-30, 2013 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-23746455

RESUMO

Over the last decade, poxviral vectors emerged as a mainstay approach for the induction of T cell-mediated immunity by vaccination, and their suitability for human use has led to widespread clinical testing of candidate vectors against infectious intracellular pathogens and cancer. In contrast, poxviruses have been widely perceived in the vaccine field as a poor choice of vector for the induction of humoral immunity. However, a growing body of data, from both animal models and recent clinical trials, now suggests that these vectors can be successfully utilized to prime and boost B cells and effective antibody responses. Significant progress has been made in the context of heterologous prime-boost immunization regimes, whereby poxviruses are able to boost responses primed by other vectors, leading to the induction of high-titre antigen-specific antibody responses. In other cases, poxviral vectors have been shown to stimulate humoral immunity against both themselves and encoded transgenes, in particular viral surface proteins such as influenza haemagglutinin. In the veterinary field, recombinant poxviral vectors have made a significant impact with numerous vectors licensed for use against a variety of animal viruses. On-going studies continue to explore the potential of poxviral vectors to modulate qualitative aspects of the humoral response, as well as their amenability to adjuvantation seeking to improve quantitative antibody immunogenicity. Nevertheless, the underlying mechanisms of B cell induction by recombinant poxviruses remain poorly defined, and further work is necessary to help guide the rational optimization of future poxviral vaccine candidates aiming to induce antibodies.


Assuntos
Vetores Genéticos , Poxviridae/imunologia , Vacinas de DNA/imunologia , Animais , Formação de Anticorpos , Linfócitos B/imunologia , Ensaios Clínicos como Assunto , Humanos , Imunidade Celular/genética , Imunidade Celular/imunologia , Imunidade Humoral , Modelos Animais , Poxviridae/genética , Vacinação
15.
Methods Mol Biol ; 890: 37-57, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22688760

RESUMO

Traditional methods for genetic manipulation of poxviruses rely on low-frequency natural recombination in virus-infected cells. Although these powerful systems represent the technical foundation of current knowledge and applications of poxviruses, they require long (≥ 500 bp) flanking sequences for homologous recombination, an efficient viral selection method, and burdensome, time-consuming plaque purification. The beginning of the twenty-first century has seen the application of bacterial artificial chromosome (BAC) technology to poxviruses as an alternative method for their genetic manipulation, following the invention of a long-sought-after method for deriving a BAC clone of vaccinia virus (VAC-BAC) by Arban Domi and Bernard Moss. The key advantages of the BAC system are the ease and versatility of performing genetic manipulation using bacteriophage λ Red recombination (recombineering), which requires only ∼50 bp homology arms that can be easily created by PCR, and which allows seamless mutations lacking any marker gene without having to perform transient-dominant selection. On the other hand, there are disadvantages, including the significant setup time, the risk of contamination of the cloned genome with bacterial insertion sequences, and the nontrivial issue of removal of the BAC cassette from derived viruses. These must be carefully weighed to decide whether the use of BACs will be advantageous for a particular application, making pox-BAC systems likely to complement, rather than supplant, traditional methods in most laboratories.


Assuntos
Cromossomos Artificiais Bacterianos/genética , Vírus da Varíola das Aves Domésticas/crescimento & desenvolvimento , Engenharia Genética , Vírus Auxiliares/crescimento & desenvolvimento , Vaccinia virus/genética , Animais , Sequência de Bases , Linhagem Celular , Clonagem Molecular , DNA Recombinante/genética , DNA Recombinante/isolamento & purificação , DNA Viral/genética , DNA Viral/isolamento & purificação , Escherichia coli , Humanos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Cultura de Vírus
16.
PLoS One ; 7(6): e40167, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22761956

RESUMO

CD8(+) T cell responses to vaccinia virus are directed almost exclusively against early gene products. The attenuated strain modified vaccinia virus Ankara (MVA) is under evaluation in clinical trials of new vaccines designed to elicit cellular immune responses against pathogens including Plasmodium spp., M. tuberculosis and HIV-1. All of these recombinant MVAs (rMVA) utilize the well-established method of linking the gene of interest to a cloned poxviral promoter prior to insertion into the viral genome at a suitable locus by homologous recombination in infected cells. Using BAC recombineering, we show that potent early promoters that drive expression of non-functional or non-essential MVA open reading frames (ORFs) can be harnessed for immunogenic expression of recombinant antigen. Precise replacement of the MVA orthologs of C11R, F11L, A44L and B8R with a model antigen positioned to use the same translation initiation codon allowed early transgene expression similar to or slightly greater than that achieved by the commonly-used p7.5 or short synthetic promoters. The frequency of antigen-specific CD8(+) T cells induced in mice by single shot or adenovirus-prime, rMVA-boost vaccination were similarly equal or marginally enhanced using endogenous promoters at their authentic genomic loci compared to the traditional constructs. The enhancement in immunogenicity observed using the C11R or F11L promoters compared with p7.5 was similar to that obtained with the mH5 promoter compared with p7.5. Furthermore, the growth rates of the viruses were unimpaired and the insertions were genetically stable. Insertion of a transgenic ORF in place of a viral ORF by BAC recombineering can thus provide not only a potent promoter, but also, concomitantly, a suitable insertion site, potentially facilitating development of MVA vaccines expressing multiple recombinant antigens.


Assuntos
Antígenos/imunologia , Poxviridae/imunologia , Regiões Promotoras Genéticas , Animais , Sequência de Bases , Cromossomos Artificiais Bacterianos , Primers do DNA , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Fases de Leitura Aberta
17.
PLoS One ; 7(7): e40385, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22808149

RESUMO

Recombinant adenoviruses are among the most promising tools for vaccine antigen delivery. Recently, the development of new vectors has focused on serotypes to which the human population is less exposed in order to circumvent pre-existing anti vector immunity. This study describes the derivation of a new vaccine vector based on a chimpanzee adenovirus, Y25, together with a comparative assessment of its potential to elicit transgene product specific immune responses in mice. The vector was constructed in a bacterial artificial chromosome to facilitate genetic manipulation of genomic clones. In order to conduct a fair head-to-head immunological comparison of multiple adenoviral vectors, we optimised a method for accurate determination of infectious titre, since this parameter exhibits profound natural variability and can confound immunogenicity studies when doses are based on viral particle estimation. Cellular immunogenicity of recombinant E1 E3-deleted vector ChAdY25 was comparable to that of other species E derived chimpanzee adenovirus vectors including ChAd63, the first simian adenovirus vector to enter clinical trials in humans. Furthermore, the prevalence of virus neutralizing antibodies (titre >1:200) against ChAdY25 in serum samples collected from two human populations in the UK and Gambia was particularly low compared to published data for other chimpanzee adenoviruses. These findings support the continued development of new chimpanzee adenovirus vectors, including ChAdY25, for clinical use.


Assuntos
Adenovirus dos Símios/genética , Adenovirus dos Símios/imunologia , Vetores Genéticos/genética , Pan troglodytes/imunologia , Pan troglodytes/virologia , Vacinas contra Adenovirus/imunologia , Adenovírus Humanos/genética , Adenovírus Humanos/imunologia , Adenovirus dos Símios/patogenicidade , Animais , Anticorpos Neutralizantes/imunologia , Sequência de Bases , Feminino , Gâmbia/epidemiologia , Genes Virais/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Filogenia , Estudos Soroepidemiológicos , Titulometria , Reino Unido/epidemiologia , Vírion/genética
18.
PLoS One ; 7(3): e33555, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22470455

RESUMO

To prevent important infectious diseases such as tuberculosis, malaria and HIV, vaccines inducing greater T cell responses are required. In this study, we investigated whether fusion of the M. tuberculosis antigen 85A to recently described adjuvant IMX313, a hybrid avian C4bp oligomerization domain, could increase T cell responses in pre-clinical vaccine model species. In mice, the fused antigen 85A showed consistent increases in CD4(+) and CD8(+) T cell responses after DNA and MVA vaccination. In rhesus macaques, higher IFN-γ responses were observed in animals vaccinated with MVA-Ag85A IMX313 after both primary and secondary immunizations. In both animal models, fusion to IMX313 induced a quantitative enhancement in the response without altering its quality: multifunctional cytokines were uniformly increased and differentiation into effector and memory T cell subsets was augmented rather than skewed. An extensive in vivo characterization suggests that IMX313 improves the initiation of immune responses as an increase in antigen 85A specific cells was observed as early as day 3 after vaccination. This report demonstrates that antigen multimerization using IMX313 is a simple and effective cross-species method to improve vaccine immunogenicity with potentially broad applicability.


Assuntos
Aciltransferases/imunologia , Antígenos de Bactérias/imunologia , Proteína de Ligação ao Complemento C4b/imunologia , Mycobacterium tuberculosis/imunologia , Vacinas contra a Tuberculose/imunologia , Aciltransferases/genética , Adjuvantes Imunológicos , Animais , Antígenos de Bactérias/genética , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Proteína de Ligação ao Complemento C4b/genética , Feminino , Humanos , Interferon gama/metabolismo , Macaca mulatta , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mycobacterium tuberculosis/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Ativador de Plasminogênio Tecidual/genética , Vacinas contra a Tuberculose/genética , Vacinação , Vacinas de DNA/imunologia , Vacinas Virais/imunologia
19.
PLoS One ; 7(9): e44943, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22984589

RESUMO

Viral vectored vaccines have been shown to induce both T cell and antibody responses in animals and humans. However, the induction of even higher level T cell responses may be crucial in achieving vaccine efficacy against difficult disease targets, especially in humans. Here we investigate the oligomerization domain of the α-chain of C4b-binding protein (C4 bp) as a candidate T cell "molecular adjuvant" when fused to malaria antigens expressed by human adenovirus serotype 5 (AdHu5) vectored vaccines in BALB/c mice. We demonstrate that i) C-terminal fusion of an oligomerization domain can enhance the quantity of antigen-specific CD4(+) and CD8(+) T cell responses induced in mice after only a single immunization of recombinant AdHu5, and that the T cells maintain similar functional cytokine profiles; ii) an adjuvant effect is observed for AdHu5 vectors expressing either the 42 kDa C-terminal domain of Plasmodium yoelii merozoite surface protein 1 (PyMSP1(42)) or the 83 kDa ectodomain of P. falciparum strain 3D7 apical membrane antigen 1 (PfAMA1), but not a candidate 128kDa P. falciparum MSP1 biallelic fusion antigen; iii) following two homologous immunizations of AdHu5 vaccines, antigen-specific T cell responses are further enhanced, however, in both BALB/c mice and New Zealand White rabbits no enhancement of functional antibody responses is observed; and iv) that the T cell adjuvant activity of C4 bp is not dependent on a functional Fc-receptor γ-chain in the host, but is associated with the oligomerization of small (<80 kDa) antigens expressed by recombinant AdHu5. The oligomerization domain of C4 bp can thus adjuvant T cell responses induced by AdHu5 vectors against selected antigens and its clinical utility as well as mechanism of action warrant further investigation.


Assuntos
Adenoviridae/genética , Proteína de Ligação ao Complemento C4b/metabolismo , Malária/prevenção & controle , Linfócitos T/citologia , Adjuvantes Imunológicos/genética , Animais , Antígenos de Protozoários/genética , Feminino , Vetores Genéticos , Vacinas Antimaláricas/genética , Proteína 1 de Superfície de Merozoito/genética , Camundongos , Camundongos Endogâmicos BALB C , Plasmodium falciparum/genética , Plasmodium yoelii/genética , Estrutura Terciária de Proteína , Coelhos , Receptores de IgG/metabolismo , Linfócitos T/metabolismo , Linfócitos T/virologia , Vacinas/genética
20.
Curr Opin Immunol ; 23(3): 377-82, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21514130

RESUMO

A little more than a decade after the explosion of research into recombinant live-attenuated or replication-deficient viruses as vaccine platforms, many viral vector-based vaccines have been licensed for animals. Progress has been slower for humans but 2011 will see the licensure of the first viral-vectored vaccine for humans, against Japanese Encephalitis. In addition a vaccine with a viral-vectored component showed efficacy against HIV infection in humans. Viral-based vaccines have an excellent safety profile but must deal with the potential problem of pre-existing anti-vector immunity. Recent successes reflect diverse improvements such as development of new adenovirus serotypes and better prime-boost approaches, suggesting that many viral vectors are approaching their final years as vaccine 'candidates' rather than vaccines.


Assuntos
Vetores Genéticos/genética , Vacinas Virais/genética , Viroses/prevenção & controle , Animais , Ensaios Clínicos como Assunto , Humanos , Resultado do Tratamento , Vacinas Virais/imunologia , Viroses/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA