Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Ther ; 29(3): 1186-1198, 2021 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-33278563

RESUMO

Historically poor clinical results of tumor vaccines have been attributed to weakly immunogenic antigen targets, limited specificity, and vaccine platforms that fail to induce high-quality polyfunctional T cells, central to mediating cellular immunity. We show here that the combination of antigen selection, construct design, and a robust vaccine platform based on the Synthetically Modified Alpha Replicon RNA Technology (SMARRT), a self-replicating RNA, leads to control of tumor growth in mice. Therapeutic immunization with SMARRT replicon-based vaccines expressing tumor-specific neoantigens or tumor-associated antigen were able to generate polyfunctional CD4+ and CD8+ T cell responses in mice. Additionally, checkpoint inhibitors, or co-administration of cytokine also expressed from the SMARRT platform, synergized to enhance responses further. Lastly, SMARRT-based immunization of non-human primates was able to elicit high-quality T cell responses, demonstrating translatability and clinical feasibility of synthetic replicon technology for therapeutic oncology vaccines.


Assuntos
Antígenos de Neoplasias/imunologia , Linfócitos T CD4-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Neoplasias do Colo/terapia , Imunidade Celular/imunologia , Replicon , Animais , Vacinas Anticâncer/imunologia , Neoplasias do Colo/genética , Neoplasias do Colo/imunologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Primatas , Células Tumorais Cultivadas , Vacinação
2.
Expert Rev Vaccines ; 14(2): 283-312, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25382613

RESUMO

The advent of reverse genetic approaches to manipulate the genomes of both positive (+) and negative (-) sense RNA viruses allowed researchers to harness these genomes for basic research. Manipulation of positive sense RNA virus genomes occurred first largely because infectious RNA could be transcribed directly from cDNA versions of the RNA genomes. Manipulation of negative strand RNA virus genomes rapidly followed as more sophisticated approaches to provide RNA-dependent RNA polymerase complexes coupled with negative-strand RNA templates were developed. These advances have driven an explosion of RNA virus vaccine vector development. That is, development of approaches to exploit the basic replication and expression strategies of RNA viruses to produce vaccine antigens that have been engineered into their genomes. This study has led to significant preclinical testing of many RNA virus vectors against a wide range of pathogens as well as cancer targets. Multiple RNA virus vectors have advanced through preclinical testing to human clinical evaluation. This review will focus on RNA virus vectors designed to express heterologous genes that are packaged into viral particles and have progressed to clinical testing.


Assuntos
Vetores Genéticos/genética , Vírus de RNA/genética , RNA Viral/genética , Terapia Genética , Humanos , Genética Reversa , Vírion/genética
3.
J Virol ; 87(10): 5447-60, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23468490

RESUMO

We have previously shown that delivery of the porcine type I interferon gene (poIFN-α/ß) with a replication-defective human adenovirus vector (adenovirus 5 [Ad5]) can sterilely protect swine challenged with foot-and-mouth disease virus (FMDV) 1 day later. However, the need of relatively high doses of Ad5 limits the applicability of such a control strategy in the livestock industry. Venezuelan equine encephalitis virus (VEE) empty replicon particles (VRPs) can induce rapid protection of mice against either homologous or, in some cases, heterologous virus challenge. As an alternative approach to induce rapid protection against FMDV, we have examined the ability of VRPs containing either the gene for green fluorescent protein (VRP-GFP) or poIFN-α (VRP-poIFN-α) to block FMDV replication in vitro and in vivo. Pretreatment of swine or bovine cell lines with either VRP significantly inhibited subsequent infection with FMDV as early as 6 h after treatment and for at least 120 h posttreatment. Furthermore, mice pretreated with either 10(7) or 10(8) infectious units of VRP-GFP and challenged with a lethal dose of FMDV 24 h later were protected from death. Protection was induced as early as 6 h after treatment and lasted for at least 48 h and correlated with induction of an antiviral response and production of IFN-α. By 6 h after treatment several genes were upregulated, and the number of genes and the level of induction increased at 24 h. Finally, we demonstrated that the chemokine IP-10, which is induced by IFN-α and VRP-GFP, is directly involved in protection against FMDV.


Assuntos
Vírus da Encefalite Equina Venezuelana/genética , Vírus da Febre Aftosa/imunologia , Febre Aftosa/prevenção & controle , Terapia Genética/métodos , Vetores Genéticos , Interferon-alfa/genética , Interferon-alfa/imunologia , Animais , Modelos Animais de Doenças , Febre Aftosa/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sobrevida
4.
J Virol ; 87(9): 4952-64, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23408633

RESUMO

There are no vaccines or therapeutics currently approved for the prevention or treatment of ebolavirus infection. Previously, a replicon vaccine based on Venezuelan equine encephalitis virus (VEEV) demonstrated protective efficacy against Marburg virus in nonhuman primates. Here, we report the protective efficacy of Sudan virus (SUDV)- and Ebola virus (EBOV)-specific VEEV replicon particle (VRP) vaccines in nonhuman primates. VRP vaccines were developed to express the glycoprotein (GP) of either SUDV or EBOV. A single intramuscular vaccination of cynomolgus macaques with VRP expressing SUDV GP provided complete protection against intramuscular challenge with SUDV. Vaccination against SUDV and subsequent survival of SUDV challenge did not fully protect cynomolgus macaques against intramuscular EBOV back-challenge. However, a single simultaneous intramuscular vaccination with VRP expressing SUDV GP combined with VRP expressing EBOV GP did provide complete protection against intramuscular challenge with either SUDV or EBOV in cynomolgus macaques. Finally, intramuscular vaccination with VRP expressing SUDV GP completely protected cynomolgus macaques when challenged with aerosolized SUDV, although complete protection against aerosol challenge required two vaccinations with this vaccine.


Assuntos
Ebolavirus/imunologia , Vírus da Encefalite Equina Venezuelana/genética , Doença pelo Vírus Ebola/prevenção & controle , Replicon , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/imunologia , Ebolavirus/genética , Vírus da Encefalite Equina Venezuelana/fisiologia , Vetores Genéticos/genética , Vetores Genéticos/fisiologia , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/virologia , Humanos , Macaca fascicularis , Vacinação , Proteínas do Envelope Viral/administração & dosagem , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
5.
Virol J ; 10: 35, 2013 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-23356714

RESUMO

BACKGROUND: Bovine viral diarrhea virus is one of the most significant and costly viral pathogens of cattle worldwide. Alphavirus-derived replicon particles have been shown to be safe and highly effective vaccine vectors against a variety of human and veterinary pathogens. Replicon particles are non-propagating, DIVA compatible, and can induce both humoral and cell mediated immune responses. This is the first experiment to demonstrate that Alphavirus-based replicon particles can be utilized in a standard prime/boost vaccination strategy in calves against a commercially significant bovine pathogen. FINDINGS: Replicon particles that express bovine viral diarrhea virus sub-genotype 1b E2 glycoprotein were generated and expression was confirmed in vitro using polyclonal and monoclonal antibodies specific to E2. Vaccine made from particles was generated in Vero cells and administered to BVDV free calves in a prime/boost regimen at two dosage levels. Vaccination resulted in neutralizing antibody titers that cross-neutralized both type 1 and type 2 BVD genotypes following booster vaccination. Additionally, high dose vaccine administration demonstrated some protection from clinical disease and significantly reduced the degree of leukopenia caused by viral infection. CONCLUSIONS: Replicon particle vaccines administered in a prime/boost regimen expressing BVDV E2 glycoprotein can induce cross-neutralizing titers, reduce leukopenia post challenge, and mitigate clinical disease in calves. This strategy holds promise for a safe and effective vaccine to BVDV.


Assuntos
Alphavirus/genética , Vírus da Diarreia Viral Bovina/imunologia , Glicoproteínas/imunologia , Replicon , Proteínas Estruturais Virais/imunologia , Vacinas Virais/imunologia , Alphavirus/metabolismo , Animais , Anticorpos Antivirais/imunologia , Doença das Mucosas por Vírus da Diarreia Viral Bovina/imunologia , Doença das Mucosas por Vírus da Diarreia Viral Bovina/prevenção & controle , Doença das Mucosas por Vírus da Diarreia Viral Bovina/virologia , Bovinos , Vírus da Diarreia Viral Bovina/genética , Feminino , Expressão Gênica , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Glicoproteínas/administração & dosagem , Glicoproteínas/genética , Masculino , Vacinação , Proteínas Estruturais Virais/administração & dosagem , Proteínas Estruturais Virais/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
6.
PLoS Pathog ; 7(10): e1002308, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22028652

RESUMO

Epstein-Barr virus (EBV) is a human lymphocryptovirus that is associated with several malignancies. Elevated EBV DNA in the blood is observed in transplant recipients prior to, and at the time of post-transplant lymphoproliferative disease; thus, a vaccine that either prevents EBV infection or lowers the viral load might reduce certain EBV malignancies. Two major approaches have been suggested for an EBV vaccine- immunization with either EBV glycoprotein 350 (gp350) or EBV latency proteins (e.g. EBV nuclear antigens [EBNAs]). No comparative trials, however, have been performed. Rhesus lymphocryptovirus (LCV) encodes a homolog for each gene in EBV and infection of monkeys reproduces the clinical, immunologic, and virologic features of both acute and latent EBV infection. We vaccinated rhesus monkeys at 0, 4 and 12 weeks with (a) soluble rhesus LCV gp350, (b) virus-like replicon particles (VRPs) expressing rhesus LCV gp350, (c) VRPs expressing rhesus LCV gp350, EBNA-3A, and EBNA-3B, or (d) PBS. Animals vaccinated with soluble gp350 produced higher levels of antibody to the glycoprotein than those vaccinated with VRPs expressing gp350. Animals vaccinated with VRPs expressing EBNA-3A and EBNA-3B developed LCV-specific CD4 and CD8 T cell immunity to these proteins, while VRPs expressing gp350 did not induce detectable T cell immunity to gp350. After challenge with rhesus LCV, animals vaccinated with soluble rhesus LCV gp350 had the best level of protection against infection based on seroconversion, viral DNA, and viral RNA in the blood after challenge. Surprisingly, animals vaccinated with gp350 that became infected had the lowest LCV DNA loads in the blood at 23 months after challenge. These studies indicate that gp350 is critical for both protection against infection with rhesus LCV and for reducing the viral load in animals that become infected after challenge. Our results suggest that additional trials with soluble EBV gp350 alone, or in combination with other EBV proteins, should be considered to reduce EBV infection or virus-associated malignancies in humans.


Assuntos
Infecções por Herpesviridae/virologia , Lymphocryptovirus/patogenicidade , Macaca mulatta/virologia , Glicoproteínas de Membrana/imunologia , Infecções Tumorais por Vírus/virologia , Vacinas Virais/administração & dosagem , Animais , DNA Viral/sangue , Modelos Animais de Doenças , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/imunologia , Interações Hospedeiro-Patógeno , Lymphocryptovirus/imunologia , Infecções Tumorais por Vírus/genética , Infecções Tumorais por Vírus/imunologia , Carga Viral , Latência Viral , Replicação Viral
7.
J Virol ; 84(15): 7713-25, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20504925

RESUMO

Alphavirus-based replicon vector systems (family Togaviridae) have been developed as expression vectors with demonstrated potential in vaccine development against both infectious diseases and cancer. The single-cycle nature of virus-like replicon particles (VRP), generated by supplying the structural proteins from separate replicable helper RNAs, is an attractive safety component of these systems. MicroRNAs (miRNAs) have emerged as important cellular RNA regulation elements. Recently, miRNAs have been employed as a mechanism to attenuate or restrict cellular tropism of replication-competent viruses, such as oncolytic adenoviruses, vesicular stomatitis virus, and picornaviruses as well as nonreplicating lentiviral and adenoviral vectors. Here, we describe the incorporation of miRNA-specific target sequences into replicable alphavirus helper RNAs that are used in trans to provide the structural proteins required for VRP production. VRP were found to be efficiently produced using miRNA-targeted helper RNAs if miRNA-specific inhibitors were introduced into cells during VRP production. In the absence of such inhibitors, cellular miRNAs were capable of downregulating helper RNA replication in vitro. When miRNA targets were incorporated into a replicon RNA, cellular miRNAs were capable of downregulating replicon RNA replication upon delivery of VRP into animals, demonstrating activity in vivo. These data provide the first example of miRNA-specific repression of alphavirus replicon and helper RNA replication and demonstrate the feasibility of miRNA targeting of expression vector helper functions that are provided in trans.


Assuntos
Alphavirus/crescimento & desenvolvimento , Alphavirus/genética , Marcação de Genes , Vetores Genéticos , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Animais , Chlorocebus aethiops , Feminino , Camundongos , Camundongos Endogâmicos BALB C , RNA Viral/genética , RNA Viral/metabolismo , Células Vero
8.
PLoS One ; 4(8): e6559, 2009 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-19668343

RESUMO

Using newer vaccine platforms which have been effective against malaria in rodent models, we tested five immunization regimens against Plasmodium knowlesi in rhesus monkeys. All vaccines included the same four P. knowlesi antigens: the pre-erythrocytic antigens CSP, SSP2, and erythrocytic antigens AMA1, MSP1. We used four vaccine platforms for prime or boost vaccinations: plasmids (DNA), alphavirus replicons (VRP), attenuated adenovirus serotype 5 (Ad), or attenuated poxvirus (Pox). These four platforms combined to produce five different prime/boost vaccine regimens: Pox alone, VRP/Pox, VRP/Ad, Ad/Pox, and DNA/Pox. Five rhesus monkeys were immunized with each regimen, and five Control monkeys received a mock vaccination. The time to complete vaccinations was 420 days. All monkeys were challenged twice with 100 P. knowlesi sporozoites given IV. The first challenge was given 12 days after the last vaccination, and the monkeys receiving the DNA/Pox vaccine were the best protected, with 3/5 monkeys sterilely protected and 1/5 monkeys that self-cured its parasitemia. There was no protection in monkeys that received Pox malaria vaccine alone without previous priming. The second sporozoite challenge was given 4 months after the first. All 4 monkeys that were protected in the first challenge developed malaria in the second challenge. DNA, VRP and Ad5 vaccines all primed monkeys for strong immune responses after the Pox boost. We discuss the high level but short duration of protection in this experiment and the possible benefits of the long interval between prime and boost.


Assuntos
Macaca mulatta/imunologia , Vacinas Antimaláricas/imunologia , Malária/veterinária , Plasmodium knowlesi/imunologia , Animais , Anticorpos Antiprotozoários/biossíntese , Antígenos de Protozoários/genética , Antígenos de Protozoários/imunologia , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Vetores Genéticos , Malária/parasitologia , Malária/prevenção & controle , Vacinas Antimaláricas/administração & dosagem , Linfócitos T/imunologia , Vírus/genética
9.
Clin Vaccine Immunol ; 14(3): 331-3, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17229882

RESUMO

Recombinant severe acute respiratory syndrome (SARS) nucleocapsid and spike protein-based immunoglobulin G immunoassays were developed and evaluated. Our assays demonstrated high sensitivity and specificity to the SARS coronavirus in sera collected from patients as late as 2 years postonset of symptoms. These assays will be useful not only for routine SARS coronavirus diagnostics but also for epidemiological and antibody kinetic studies.


Assuntos
Anticorpos Antivirais/sangue , Glicoproteínas de Membrana/imunologia , Proteínas do Nucleocapsídeo/imunologia , Síndrome Respiratória Aguda Grave/diagnóstico , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/imunologia , Proteínas do Envelope Viral/imunologia , Proteínas do Nucleocapsídeo de Coronavírus , Ensaio de Imunoadsorção Enzimática , Humanos , Proteínas Recombinantes/imunologia , Sensibilidade e Especificidade , Testes Sorológicos , Glicoproteína da Espícula de Coronavírus
10.
J Virol Methods ; 128(1-2): 21-8, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15885812

RESUMO

Monoclonal antibodies (Mabs) against the Urbani strain of the SARS-associated coronavirus (SARS-CoV) were developed and characterized for reactivity to SARS-CoV and SARS-CoV S, N, M, and E proteins using enzyme-linked immunoabsorbent (ELISA), radioimmunoprecipitation, immunofluorescence, Western Blot and microneutralization assays. Twenty-six mAbs were reactive to SARS-CoV by ELISA, and nine were chosen for detailed characterization. Five mAbs reacted against the S protein, two against the M protein, and one each against the N and E proteins. Two of five S protein mAbs neutralized SARS-CoV infection of Vero E6 cells and reacted to an epitope within amino acids 490-510 in the S protein. While two of the three non-neutralizing antibodies recognized at second epitope within amino acids 270-350. The mAbs characterized should prove useful for developing SARS-CoV diagnostic assays and for studying the biology of infection and pathogenesis of disease.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Especificidade de Anticorpos , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/imunologia , Proteínas Estruturais Virais/imunologia , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Antivirais/biossíntese , Linhagem Celular , Chlorocebus aethiops , Proteínas M de Coronavírus , Proteínas do Nucleocapsídeo de Coronavírus , Mapeamento de Epitopos , Humanos , Glicoproteínas de Membrana/imunologia , Testes de Neutralização , Proteínas do Nucleocapsídeo/imunologia , Síndrome Respiratória Aguda Grave/virologia , Glicoproteína da Espícula de Coronavírus , Células Vero , Proteínas do Envelope Viral/imunologia , Proteínas da Matriz Viral/imunologia , Proteínas Viroporinas
11.
Breast Cancer Res Treat ; 82(3): 169-83, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14703064

RESUMO

Many tumor-associated antigens (TAAs) represent 'self' antigens and as such, are subject to the constraints of immunologic tolerance. There are significant barriers to eliciting anti-tumor immune responses of sufficient magnitude. We have taken advantage of a Venezuelan equine encephalitis-derived alphavirus replicon vector system with documented in vivo tropism for immune system dendritic cells. We have overcome the intrinsic tolerance to the 'self' TAA rat neu and elicited an effective anti-tumor immune response using this alphavirus replicon vector system and a designed target antigen in a rigorous rat mammary tumor model. We have demonstrated the capacity to generate 50% protection in tumor challenge experiments (p = 0.004) and we have confirmed the establishment of immunologic memory by both second tumor challenge and Winn Assay (p = 0.009). Minor antibody responses were identified and supported the establishment of T helper type 1 (Th1) anti-tumor immune responses by isotype. Animals surviving in excess of 300 days with established effective anti-tumor immunity showed no signs of autoimmune phenomena. Together these experiments support the establishment of T lymphocyte dependent, Th1-biased anti-tumor immune responses to a non-mutated 'self' TAA in an aggressive tumor model. Importantly, this tumor model is subject to the constraints of immunologic tolerance present in animals with normal developmental, temporal, and anatomical expression of a non-mutated TAA. These data support the continued development and potential clinical application of this alphaviral replicon vector system and the use of appropriately designed target antigen sequences for anti-tumor immunotherapy.


Assuntos
Antígenos de Neoplasias/imunologia , Modelos Animais de Doenças , Vírus da Encefalite Equina Venezuelana/genética , Neoplasias Mamárias Experimentais/imunologia , Replicon/imunologia , Vacinas Sintéticas/imunologia , Sequência de Aminoácidos , Animais , Feminino , Vetores Genéticos/imunologia , Vetores Genéticos/uso terapêutico , Humanos , Imunização , Neoplasias Mamárias Experimentais/terapia , Dados de Sequência Molecular , Proteínas de Neoplasias/imunologia , Ratos , Ratos Endogâmicos F344 , Receptor ErbB-2/química , Receptor ErbB-2/genética , Receptor ErbB-2/imunologia , Tolerância a Antígenos Próprios
12.
Rev Med Virol ; 12(5): 279-96, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12211042

RESUMO

Alphaviruses are positive-stranded RNA viruses that have a broad host range and therefore are capable of replicating in many vertebrate and invertebrate cells. The single-stranded alphavirus genome is divided into two ORFs. The first ORF encodes the nonstructural proteins that are translated upon entry of the virus into the cytoplasm and are responsible for transcription and replication of viral RNA. The second ORF is under the control of a subgenomic promoter and normally encodes the structural proteins, which are responsible for encapsidation of viral RNA and final assembly into enveloped particles. Expression vectors have been engineered from at least three alphaviruses in which the structural protein gene region has been replaced by heterologous genes and have been shown to express high levels of the heterologous protein in cultured cells. These RNA vectors, known as replicons, are capable of replicating on their own but are not packaged into virus-like particles unless the structural proteins are provided in trans. Thus, replicons are single cycle vectors incapable of spreading from infected to noninfected cells. Because of these features, alphavirus replicon vectors are being developed as a platform vaccine technology for numerous viral, bacterial, protozoan and tumour antigens where they have been shown to be efficient inducers of both humoral and T cell responses. In addition, as the alphavirus structural proteins are not expressed in vaccine recipients, antivector immune responses are generally minimal, allowing for multiple effective immunisations of the same individual.


Assuntos
Alphavirus/genética , Vetores Genéticos , Vacinas de DNA , Vacinas Sintéticas , Animais , Controle de Doenças Transmissíveis , Humanos , Replicon , Vacinação/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA