Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2786: 289-300, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38814400

RESUMO

In this protocol, we outline how to produce a chimeric viral vaccine in a biosafety level 1 (BSL1) environment. An animal viral vector RNA encapsidated with tobacco mosaic virus (TMV) coat protein can be fully assembled in planta. Agrobacterium cultures containing each component are inoculated together into tobacco leaves and the self-assembled hybrid chimeric viral vaccine is harvested 4 days later and purified with a simple PEG precipitation. The viral RNA delivery vector is derived from the BSL1 insect virus, Flock House virus (FHV), and replicates in human and animal cells but does not spread systemically. A polyethylene glycol purification protocol is also provided to collect and purify these vaccines for immunological tests. In this update, we also provide a protocol for in trans co-inoculation of a modified FHV protein A, which significantly increased the yield of in planta chimeric viral vaccine.


Assuntos
Nicotiana , Replicon , Vírus do Mosaico do Tabaco , Vacinas Virais , Nicotiana/genética , Vacinas Virais/imunologia , Vacinas Virais/genética , Animais , Vírus do Mosaico do Tabaco/genética , Vírus do Mosaico do Tabaco/imunologia , Replicon/genética , RNA Viral/genética , Vetores Genéticos/genética , Nodaviridae/genética , Nodaviridae/imunologia , Plantas Geneticamente Modificadas/genética , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/imunologia , Agrobacterium/genética , Humanos
2.
Biology (Basel) ; 12(7)2023 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-37508413

RESUMO

In this study, we demonstrate that expression of viral latent membrane protein 1 (LMP1) in a mouse B cell line renders the animals responsive to protection from a 38C13-LMP1 tumor challenge with a novel vaccine. The Epstein-Barr virus (EBV) preferentially infects circulating B lymphocytes, has oncogenic potential, and is associated with a wide variety of B cell lymphomas. EBV is ectotrophic to human cells, and currently there are no B cell animal models of EBV-associated lymphoma that can be used to investigate vaccine immunotherapy. Since most EBV-infected human tumor cells express latent membrane protein 1 (LMP1) on their surface, this viral antigen was tested as a potential target for an anticancer vaccine in a mouse model. Here, we describe a new mouse model of LMP1-expressing B cell lymphoma produced with plasmid transduction of 38C13 into mouse B cells. The expression of LMP-1 was confirmed with a western blot analysis and immunocytochemistry. We then designed a novel LMP1 vaccine, by fusing viral antigen LMP1 surface loop epitopes to the surface of a viral antigen carrier, the Tobacco Mosaic virus (TMV). Vaccinated mice produced high titer antibodies against the TMV-LMP1 vaccine; however, cellular responses were at the baseline, as measured with IFNγ ELISpot. Despite this, the vaccine showed significant protection from a 38C13-LMP1 tumor challenge. To provide additional immune targets, we compared TMV-LMP1 peptide immunization with DNA immunization with the full-length LMP1 gene. Anti-LMP1 antibodies were significantly higher in TMV-LMP1-vaccinated mice compared to the DNA-immunized mice, but, as predicted, DNA-vaccinated mice had improved cellular responses using IFNγ ELISpot. Surprisingly, the TMV-LMP1 vaccine provided protection from a 38C13-LMP1 tumor challenge, while the DNA vaccine did not. Thus, we demonstrated that LMP1 expression in a mouse B cell line is responsive to antibody immunotherapy that may be applied to EBV-associated disease.

3.
Vaccines (Basel) ; 9(11)2021 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-34835278

RESUMO

Stable, effective, easy-to-manufacture vaccines are critical to stopping the COVID-19 pandemic resulting from the coronavirus SARS-CoV-2. We constructed a vaccine candidate CoV-RBD121-NP, which is comprised of the SARS-CoV-2 receptor-binding domain (RBD) of the spike glycoprotein (S) fused to a human IgG1 Fc domain (CoV-RBD121) and conjugated to a modified tobacco mosaic virus (TMV) nanoparticle. In vitro, CoV-RBD121 bound to the host virus receptor ACE2 and to the monoclonal antibody CR3022, a neutralizing antibody that blocks S binding to ACE2. The CoV-RBD121-NP vaccine candidate retained key SARS-CoV-2 spike protein epitopes, had consistent manufacturing release properties of safety, identity, and strength, and displayed stable potency when stored for 12 months at 2-8 °C or 22-28 °C. Immunogenicity studies revealed strong antibody responses in C57BL/6 mice with non-adjuvanted or adjuvanted (7909 CpG) formulations. The non-adjuvanted vaccine induced a balanced Th1/Th2 response and antibodies that recognized both the S1 domain and full S protein from SARS2-CoV-2, whereas the adjuvanted vaccine induced a Th1-biased response. Both adjuvanted and non-adjuvanted vaccines induced virus neutralizing titers as measured by three different assays. Collectively, these data showed the production of a stable candidate vaccine for COVID-19 through the association of the SARS-CoV-2 RBD with the TMV-like nanoparticle.

4.
Vaccine ; 39(9): 1435-1444, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33531196

RESUMO

Yersinia pestis, the causative agent of plague, has killed millions throughout human history. Though public health initiatives have reduced the number of plague cases, it remains endemic in many areas of the world. It also remains a significant threat for use as a biological weapon. Naturally occurring multi-drug antibiotic resistance has been observed in Y. pestis, and resistant strains have been engineered for use as a biological weapon. Vaccines represent our best means of protection against the threat of antibiotic resistant plague. We have developed a vaccine consisting of two Y. pestis virulence factors, LcrV (V) and F1, conjugated to Tobacco Mosaic Virus (TMV), a safe, non-replicating plant virus that can be administered mucosally, providing complete protection against pneumonic plague, the deadliest form of the disease and the one most likely to be seen in a biological attack. A single intranasal (i.n.) dose of TMV-F1 + TMV-V (TMV-F1/V) protected 88% of mice against lethal challenge with 100 LD50 of Y. pestis CO92pgm-, while immunization with rF1 + rV without TMV was not protective. Serum and tissues were collected at various timepoints after challenge to assess bacterial clearance, histopathology, cytokine production, and antibody production. Overall, TMV-F1/V immunized mice showed a significant reduction in histopathology, bacterial burden, and inflammatory cytokine production following challenge compared to rF1 + rV vaccinated and unvaccinated mice. Pneumonic challenge resulted in systemic dissemination of the bacteria in all groups, but only TMV-F1/V immunized mice rapidly cleared bacteria from the spleen and liver. There was a direct correlation between pre-challenge serum F1 titers and recovery in all immunized mice, strongly suggesting a role for antibody in the neutralization and/or opsonization of Y. pestis in this model. Mucosal administration of a single dose of a Y. pestis TMV-based subunit vaccine, without any additional adjuvant, can effectively protect mice from lethal infection.


Assuntos
Vacina contra a Peste , Peste , Sepse , Yersinia pestis , Animais , Anticorpos Antibacterianos , Antígenos de Bactérias , Proteínas de Bactérias , Camundongos , Peste/prevenção & controle , Proteínas Citotóxicas Formadoras de Poros , Vacinas de Subunidades Antigênicas
5.
Front Microbiol ; 9: 1195, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29922267

RESUMO

Francisella tularensis, the causative agent of the fatal human disease known as tularemia is classified as a Category A Select Agent by the Centers for Disease Control. No licensed vaccine is currently available for prevention of tularemia in the United States. Previously, we published that a tri-antigen tobacco mosaic virus (TMV) vaccine confers 50% protection in immunized mice against respiratory tularemia caused by F. tularensis. In this study, we refined the TMV-vaccine formulation to improve the level of protection in immunized C57BL/6 mice against respiratory tularemia. We developed a tetra-antigen vaccine by conjugating OmpA, DnaK, Tul4, and SucB proteins of Francisella to TMV. CpG was also included in the vaccine formulation as an adjuvant. Primary intranasal (i.n.) immunization followed by two booster immunizations with the tetra-antigen TMV vaccine protected 100% mice against i.n. 10LD100 challenges dose of F. tularensis live vaccine strain (LVS). Mice receiving three immunization doses of tetra-antigen TMV vaccine showed only transient body weight loss, cleared the infection rapidly, and showed minimal histopathological lesions in lungs, liver, and spleen following a lethal respiratory challenge with F. tularensis LVS. Mice immunized with the tetra-antigen TMV vaccine also induced strong ex vivo recall responses and were protected against a lethal challenge as late as 163 days post-primary immunization. Three immunization with the tetra-antigen TMV vaccine also induced a stronger humoral immune response predominated by IgG1, IgG2b, and IgG2c antibodies than mice receiving only a single or two immunizations. Remarkably, a single dose protected 40% of mice, while two doses protected 80% of mice from lethal pathogen challenge. Immunization of Interferon-gamma (IFN-γ)-deficient mice with the tetra-antigen TMV vaccine demonstrated an absolute requirement of IFN-γ for the generation of protective immune response against a lethal respiratory challenge with F. tularensis LVS. Collectively, this study further demonstrates the feasibility of TMV as an efficient platform for the delivery of multiple F. tularensis antigens and that tetra-antigen TMV vaccine formulation provides complete protection, and induces long-lasting protective and memory immune responses against respiratory tularemia caused by F. tularensis LVS.

6.
PLoS One ; 13(4): e0194614, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29684046

RESUMO

Tularemia is a fatal human disease caused by Francisella tularensis, a Gram-negative encapsulated coccobacillus bacterium. Due to its low infectious dose, ease of aerosolized transmission, and lethal effects, the CDC lists F. tularensis as a Category A pathogen, the highest level for a potential biothreat agent. Previous vaccine studies have been conducted with live attenuated, inactivated, and subunit vaccines, which have achieved partial or full protection from F. tularensis live vaccine strain (LVS) challenge, but no vaccine has been approved for human use. We demonstrate the improved efficacy of a multi-antigen subunit vaccine by using Tobacco Mosaic virus (TMV) as an antigen carrier for the F. tularensis SchuS4 proteins DnaK, OmpA, SucB and Tul4 (DOST). The magnitude and quality of immune responses were compared after mice were immunized by subcutaneous or intranasal routes of administration with a TMV-DOST mixture, with or without four different adjuvants. Immune responses varied in magnitude and isotype profile, by antigen, by route of administration, and by protection in an F. tularensis LVS challenge model of disease. Interestingly, our analysis demonstrates an overwhelming IgG2 response to SucB after intranasal dosing, as well as a robust cellular response, which may account for the improved two-dose survival imparted by the tetravalent vaccine, compared to a previous study that tested efficacy of TMV-DOT. Our study provides evidence that potent humoral, cellular and mucosal immunity can be achieved by optimal antigen combination, delivery, adjuvant and appropriate route of administration, to improve vaccine potency and provide protection from pathogen challenge.


Assuntos
Antígenos de Bactérias/imunologia , Vacinas Bacterianas/imunologia , Vírus do Mosaico do Tabaco/genética , Tularemia/imunologia , Vacinas de Subunidades Antigênicas/imunologia , Adjuvantes Imunológicos , Administração Intranasal , Animais , Anticorpos Antibacterianos/análise , Anticorpos Antibacterianos/metabolismo , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Modelos Animais de Doenças , Feminino , Francisella tularensis/imunologia , Imunidade Celular , Imunoglobulina G/análise , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Isotipos de Imunoglobulinas/imunologia , Isotipos de Imunoglobulinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Taxa de Sobrevida , Vírus do Mosaico do Tabaco/metabolismo , Tularemia/microbiologia , Tularemia/prevenção & controle , Vacinas Conjugadas/imunologia
7.
Methods Mol Biol ; 1499: 77-86, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27987143

RESUMO

In this protocol, we outline how to produce a live viral nanoparticle vaccine in a biosafety level 1 (BSL1) environment. An animal viral vector RNA encapsidated with tobacco mosaic virus (TMV) coat protein can be fully assembled in planta. Agrobacterium cultures containing each component are inoculated together into tobacco leaves and the self-assembled hybrid nanoparticle vaccine is harvested 4 days later and purified with a simple PEG precipitation. The viral RNA delivery vector is derived from the BSL1 insect virus, Flock House virus (FHV), and replicates in human and animal cells but does not spread systemically. A polyethylene glycol purification protocol is also provided to collect and purify these vaccines for immunological tests.


Assuntos
Nanopartículas/virologia , Nicotiana/virologia , RNA Viral/genética , Replicon/genética , Vacinas Virais/genética , Animais , Vetores Genéticos/genética , Humanos , Vírus do Mosaico do Tabaco/imunologia , Replicação Viral/genética
8.
Vaccine ; 34(47): 5768-5776, 2016 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-27745954

RESUMO

Yersinia pestis, one of history's deadliest pathogens, has killed millions over the course of human history. It has attributes that make it an ideal choice to produce mass casualties and is a prime candidate for use as a biological weapon. When aerosolized, Y. pestis causes pneumonic plague, a pneumonia that is 100% lethal if not promptly treated with effective antibiotics. Currently, there is no FDA approved plague vaccine. The current lead vaccine candidate, a parenterally administered protein subunit vaccine comprised of the Y. pestis virulence factors, F1 and LcrV, demonstrated variable levels of protection in primate pneumonic plague models. As the most likely mode of exposure in biological attack with Y. pestis is by aerosol, this raises a question of whether this parenteral vaccine will adequately protect humans against pneumonic plague. In the present study we evaluated two distinct mucosal delivery platforms for the intranasal (IN) administration of LcrV and F1 vaccine proteins, a live bacterial vector, Lactobacillus plantarum, and a Tobacco Mosaic Virus (TMV) based delivery platform. IN administration of L. plantarum expressing LcrV, or TMV-conjugated to LcrV and F1 (TMV-LcrV+TMV-F1) resulted in the similar induction of high titers of IgG antibodies and evidence of proinflammatory cytokine secretion. However, only the TMV-conjugate delivery platform protected against subsequent lethal challenge with Y. pestis. TMV-LcrV+TMV-F1 co-vaccinated mice had no discernable morbidity and no mortality, while mice vaccinated with L. plantarum expressing LcrV or rLcrV+rF1 without TMV succumbed to infection or were only partially protected. Thus, TMV is a suitable mucosal delivery platform for an F1-LcrV subunit vaccine that induces complete protection against pneumonic infection with a lethal dose of Y. pestis in mice.


Assuntos
Antígenos de Bactérias/imunologia , Lactobacillus plantarum/imunologia , Vacina contra a Peste/imunologia , Peste/prevenção & controle , Pneumonia Bacteriana/prevenção & controle , Proteínas Citotóxicas Formadoras de Poros/imunologia , Vírus do Mosaico do Tabaco/imunologia , Yersinia pestis/imunologia , Administração Intranasal , Análise de Variância , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/imunologia , Antígenos de Bactérias/genética , Citocinas/análise , Modelos Animais de Doenças , Humanos , Lactobacillus plantarum/genética , Camundongos , Vacina contra a Peste/administração & dosagem , Vacina contra a Peste/genética , Proteínas Citotóxicas Formadoras de Poros/genética , Fatores de Tempo , Vírus do Mosaico do Tabaco/genética , Vacinação , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/genética , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Fatores de Virulência/genética , Fatores de Virulência/imunologia , Yersinia pestis/genética
9.
PLoS One ; 10(6): e0130858, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26098553

RESUMO

Francisella tularensis is a facultative intracellular pathogen, and is the causative agent of a fatal human disease known as tularemia. F. tularensis is classified as a Category A Biothreat agent by the CDC based on its use in bioweapon programs by several countries in the past and its potential to be used as an agent of bioterrorism. No licensed vaccine is currently available for prevention of tularemia. In this study, we used a novel approach for development of a multivalent subunit vaccine against tularemia by using an efficient tobacco mosaic virus (TMV) based delivery platform. The multivalent subunit vaccine was formulated to contain a combination of F. tularensis protective antigens: OmpA-like protein (OmpA), chaperone protein DnaK and lipoprotein Tul4 from the highly virulent F. tularensis SchuS4 strain. Two different vaccine formulations and immunization schedules were used. The immunized mice were challenged with lethal (10xLD100) doses of F. tularensis LVS on day 28 of the primary immunization and observed daily for morbidity and mortality. Results from this study demonstrate that TMV can be used as a carrier for effective delivery of multiple F. tularensis antigens. TMV-conjugate vaccine formulations are safe and multiple doses can be administered without causing any adverse reactions in immunized mice. Immunization with TMV-conjugated F. tularensis proteins induced a strong humoral immune response and protected mice against respiratory challenges with very high doses of F. tularensis LVS. This study provides a proof-of-concept that TMV can serve as a suitable platform for simultaneous delivery of multiple protective antigens of F. tularensis. Refinement of vaccine formulations coupled with TMV-targeting strategies developed in this study will provide a platform for development of an effective tularemia subunit vaccine as well as a vaccination approach that may broadly be applicable to many other bacterial pathogens.


Assuntos
Vacinas Bacterianas/biossíntese , Vacinas Bacterianas/imunologia , Sistemas de Liberação de Medicamentos/métodos , Vetores Genéticos/genética , Vírus do Mosaico do Tabaco/genética , Tularemia/prevenção & controle , Vacinas de Subunidades Antigênicas/imunologia , Animais , Formação de Anticorpos/imunologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Escherichia coli , Humanos , Estimativa de Kaplan-Meier , Camundongos , Camundongos Endogâmicos C57BL , Vírus do Mosaico do Tabaco/metabolismo
10.
Mol Biotechnol ; 57(4): 325-36, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25432792

RESUMO

We have developed a transencapsidated vaccine delivery system based on the insect virus, Flock House virus (FHV). FHV is attractive due to its small genome size, simple organization, and nonpathogenic characteristics. With the insertion of a Tobacco mosaic virus (TMV) origin of assembly (Oa), the independently replicating FHV RNA1 can be transencapsidated by TMV coat protein. In this study, we demonstrated that the Oa-adapted FHV RNA1 transencapsidation process can take place in planta, by using a bipartite plant expression vector system, where TMV coat protein is expressed by another plant virus vector, Foxtail mosaic virus (FoMV). Dual infection in the same cell by both FHV and FoMV was observed. Though an apparent classical coat protein-mediated resistance repressed FHV expression, this was overcome by delaying inoculation of the TMV coat protein vector by 3 days after FHV vector inoculation. Expression of the transgene marker in animals by these in vivo-generated transencapsidated nanoparticles was confirmed by mouse vaccination, which also showed an improved vaccine response compared to similar in vitro-produced vaccines.


Assuntos
Vetores Genéticos/genética , Nanopartículas/química , Nodaviridae/genética , RNA Viral/genética , Vírus do Mosaico do Tabaco/genética , Vacinas/química , Vírion/genética , Animais , Clonagem Molecular , Sistemas de Liberação de Medicamentos , Vetores Genéticos/química , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , RNA Viral/química , Nicotiana/genética , Nicotiana/metabolismo , Vacinas/imunologia , Vírion/química
11.
Int J Mol Sci ; 15(10): 18540-56, 2014 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-25318056

RESUMO

Tobacco Mosaic virus (TMV) coat protein is well known for its ability to self-assemble into supramolecular nanoparticles, either as protein discs or as rods originating from the ~300 bp genomic RNA origin-of-assembly (OA). We have utilized TMV self-assembly characteristics to create a novel Flock House virus (FHV) RNA nanoparticle. FHV encodes a viral polymerase supporting autonomous replication of the FHV genome, which makes it an attractive candidate for viral transgene expression studies and targeted RNA delivery into host cells. However, FHV viral genome size is strictly limited by native FHV capsid. To determine if this packaging restriction could be eliminated, FHV was adapted to express enhanced green fluorescent protein (GFP), to allow for monitoring of functional FHV RNA activity. Then TMV OA was introduced in six 3' insertion sites, with only site one supporting functional FHV GFP expression. To create nanoparticles, FHV GFP-OA modified genomic RNA was mixed in vitro with TMV coat protein and monitored for encapsidation by agarose electrophoresis and electron microscopy. The production of TMV-like rod shaped nanoparticles indicated that modified FHV RNA can be encapsidated by purified TMV coat protein by self-assembly. This is the first demonstration of replication-independent packaging of the FHV genome by protein self-assembly.


Assuntos
Proteínas do Capsídeo/química , Capsídeo/química , Nanopartículas/química , Nodaviridae/química , RNA Viral/química , Vírus do Mosaico do Tabaco/química , Animais , Linhagem Celular , Cricetinae , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Nodaviridae/genética , RNA Viral/genética , Transfecção
12.
Vaccine ; 32(33): 4228-33, 2014 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-24923637

RESUMO

Over the past 20 years, dendritic cells (DCs) have been utilized to activate immune responses capable of eliminating cancer cells. Currently, ex vivo DC priming has been the mainstay of DC cancer immunotherapies. However, cell-based treatment modalities are inherently flawed due to a lack of standardization, specialized facilities and personnel, and cost. Therefore, direct modes of DC manipulation, circumventing the need for ex vivo culture, must be investigated. To facilitate the development of next-generation, in vivo targeted DC vaccines, we characterized the DC interaction and activation potential of the Tobacco Mosaic virus (TMV), a plant virus that enjoys a relative ease of production and the ability to deliver protein payloads via surface conjugation. In this study we show that TMV is readily taken up by mouse bone marrow-derived DCs, in vitro. Footpad injection of fluorophore-labeled TMV reveals preferential uptake by draining lymph node resident DCs in vivo. Uptake leads to activation, as measured by the upregulation of key DC surface markers. When peptide antigen-conjugated TMV is injected into the footpad of mice, DC-mediated uptake and activation leads to robust antigen-specific CD8(+) T cell responses, as measured by antigen-specific tetramer analysis. Remarkably, TMV priming induced a greater magnitude T cell response than Adenovirus (Ad) priming. Finally, TMV is capable of boosting either Ad-induced or TMV-induced antigen-specific T cell responses, demonstrating that TMV, uniquely, does not induce neutralizing self-immunity. Overall, this study elucidates the in vivo DC delivery and activation properties of TMV and indicates its potential as a vaccine vector in stand alone or prime-boost strategies.


Assuntos
Apresentação de Antígeno , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Vírus do Mosaico do Tabaco/imunologia , Adenoviridae/imunologia , Animais , Células Dendríticas/metabolismo , Feminino , Linfonodos/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Baço/imunologia
13.
Hum Vaccin Immunother ; 10(3): 586-95, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24378714

RESUMO

Recombinant subunit vaccines are an efficient strategy to meet the demands of a possible influenza pandemic, because of rapid and scalable production. However, vaccines made from recombinant hemagglutinin (HA) subunit protein are often of low potency, requiring high dose or boosting to generate a sustained immune response. We have improved the immunogenicity of a plant-made HA vaccine by chemical conjugation to the surface of the Tobacco mosaic virus (TMV) which is non infectious in mammals. We have previously shown that TMV is taken up by mammalian dendritic cells and is a highly effective antigen carrier. In this work, we tested several TMV-HA conjugation chemistries, and compared immunogenicity in mice as measured by anti-HA IgG titers and hemagglutination inhibition (HAI). Importantly, pre-existing immunity to TMV did not reduce initial or boosted titers. Further optimization included dosing with and without alum or oil-in water adjuvants. Surprisingly, we were able to stimulate potent immunogenicity and HAI titers with a single 15 µg dose of HA as a TMV conjugate. We then evaluated the efficacy of the TMV-HA vaccine in a lethal virus challenge in mice. Our results show that a single dose of the TMV-HA conjugate vaccine is sufficient to generate 50% survival, or 100% survival with adjuvant, compared with 10% survival after vaccination with a commercially available H1N1 vaccine. TMV-HA is an effective dose-sparing influenza vaccine, using a single-step process to rapidly generate large quantities of highly effective flu vaccine from an otherwise low potency HA subunit protein.


Assuntos
Vírus da Influenza A/imunologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Vacinação/métodos , Adjuvantes Imunológicos/administração & dosagem , Animais , Anticorpos Antivirais/sangue , Modelos Animais de Doenças , Portadores de Fármacos/química , Testes de Inibição da Hemaglutinação , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Imunoglobulina G/sangue , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/prevenção & controle , Análise de Sobrevida , Tobamovirus/química , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia
14.
Antimicrob Agents Chemother ; 58(1): 120-7, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24145548

RESUMO

Griffithsin (GRFT) is a red-alga-derived lectin that binds the terminal mannose residues of N-linked glycans found on the surface of human immunodeficiency virus type 1 (HIV-1), HIV-2, and other enveloped viruses, including hepatitis C virus (HCV), severe acute respiratory syndrome coronavirus (SARS-CoV), and Ebola virus. GRFT displays no human T-cell mitogenic activity and does not induce production of proinflammatory cytokines in treated human cell lines. However, despite the growing evidence showing the broad-spectrum nanomolar or better antiviral activity of GRFT, no study has reported a comprehensive assessment of GRFT safety as a potential systemic antiviral treatment. The results presented in this work show that minimal toxicity was induced by a range of single and repeated daily subcutaneous doses of GRFT in two rodent species, although we noted treatment-associated increases in spleen and liver mass suggestive of an antidrug immune response. The drug is systemically distributed, accumulating to high levels in the serum and plasma after subcutaneous delivery. Further, we showed that serum from GRFT-treated animals retained antiviral activity against HIV-1-enveloped pseudoviruses in a cell-based neutralization assay. Overall, our data presented here show that GRFT accumulates to relevant therapeutic concentrations which are tolerated with minimal toxicity. These studies support further development of GRFT as a systemic antiviral therapeutic agent against enveloped viruses, although deimmunizing the molecule may be necessary if it is to be used in long-term treatment of chronic viral infections.


Assuntos
Fármacos Anti-HIV/sangue , Fármacos Anti-HIV/farmacocinética , Antivirais/sangue , Antivirais/farmacocinética , Lectinas de Plantas/sangue , Lectinas de Plantas/farmacocinética , Animais , Fármacos Anti-HIV/uso terapêutico , Antivirais/uso terapêutico , Feminino , Cobaias , Proteína gp120 do Envelope de HIV/metabolismo , Imunoensaio , Camundongos , Camundongos Endogâmicos BALB C , Lectinas de Plantas/uso terapêutico
15.
Mol Ther ; 20(7): 1462-71, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22434138

RESUMO

Recent modest successes in ex vivo dendritic cell (DC) immunotherapy have motivated continued innovation in the area of DC manipulation and activation. Although ex vivo vaccine approaches continue to be proving grounds for new DC manipulation techniques, the intrinsic limits of ex vivo therapy, including high cost, minimal standardization, cumbersome delivery, and poor accessibility, incentivizes the development of vaccines compatible with in vivo DC targeting. We describe here a method to co-deliver both tumor-specific antigen (TSA) and an iMyD88/CD40 adjuvant (iMC), to DCs that combines toll-like receptor (TLR) and CD40 signaling. In this study, we demonstrate that simple TSA delivery via adenoviral vectors results in strong antitumor immunity. Addition of iMC delivered in a separate vector is insufficient to enhance this effect. However, when delivered simultaneously with TSA in a single bicistronic vector (BV), iMC is able to significantly enhance antigen-specific cytotoxic T-cell (CTL) responses and inhibit established tumor growth. This study demonstrates the spatial-temporal importance of concurrent DC activation and TSA presentation. Further, it demonstrates the feasibility of in vivo molecular enhancement of DCs necessary for effective antitumor immune responses.


Assuntos
Antígenos de Neoplasias/imunologia , Antígenos CD40/imunologia , Citotoxicidade Imunológica , Células Dendríticas/imunologia , Melanoma Experimental/terapia , Fator 88 de Diferenciação Mieloide/imunologia , Linfócitos T Citotóxicos/imunologia , Adjuvantes Imunológicos , Animais , Antígenos de Neoplasias/biossíntese , Antígenos de Neoplasias/genética , Antígenos CD40/metabolismo , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/metabolismo , Dependovirus , Feminino , Imunoterapia , Interleucina-12/metabolismo , Ativação Linfocitária , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide/biossíntese
16.
Hum Vaccin ; 7(3): 305-12, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21346416

RESUMO

Everyone appreciates the irony of using tobacco plants to cure cancer. (1) Recently featured in a populist Wall Street Journal article, (2) the use of plants to produce medicinal products was presented as novel, even though we are decades into development of numerous products for specific medical applications (reviewed extensively in (3, 4)). Though the tobacco plant and its relatives offer a qualified set of advantages for producing complex biologicals, and in many cases overcome problems that plague traditional expression systems, FDA licensed products derived from bioengineered plants have yet to appear in the marketplace. Despite a difficult beginning, recent advances in plant biotechnology have been as cutting edge as those in the fields of molecular biology and chemical engineering, which now position the field for a new level of commercial relevance. The basis for this review is a description of the first FDA qualified parenterally administered vaccine clinical trial using a plant derived product. We have confirmed in this trial that plant proteins can be qualified to the same level as biologicals from other sources, and are safe when given as injected vaccines. Most importantly though, immune responses to plant proteins were seen in 66% of cancer patients, and these responses were to the desired antigenic determinants, not to xenogenic plant antigens. Problems and solutions that arose during the development of a safe and effective human vaccine are discussed.


Assuntos
Vacinas Anticâncer/imunologia , Linfoma não Hodgkin/imunologia , Nicotiana/imunologia , Humanos , Idiótipos de Imunoglobulinas/imunologia , Leucócitos Mononucleares/imunologia , Plantas Geneticamente Modificadas/imunologia , Anticorpos de Cadeia Única/imunologia , Vacinação
17.
Expert Rev Vaccines ; 7(1): 33-41, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18251692

RESUMO

Tobacco mosaic virus (TMV) is an RNA virus that typically infects plants but has recently been adapted for vaccine development, owing to the suitability of the virions for modifications as nanoparticles. TMV also has a simple functional structure of a 6.4 Kb (+)-strand RNA encapsidated by a single coat protein, which permits facile genetic manipulation. In this review, we describe recent advances in the manipulation of TMV for the development of several different types of vaccines, including ones that induce antibody and T-cell responses that are protective in pathogen and tumor challenge animal models. Lastly, we describe how TMV self-assembly properties are being used to make a new mammalian RNA pseudovirus, that has unique characteristics for RNA and protein antigen delivery to antigen-presenting cells.


Assuntos
Engenharia Genética , Nanopartículas , Vírus do Mosaico do Tabaco/genética , Vacinas/genética , Antígenos Virais/imunologia , Cisteína/genética , Proteínas de Fluorescência Verde/genética , Imunidade Celular , Lisina/genética , Vacinas/imunologia
18.
Virology ; 358(2): 321-33, 2007 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-17014881

RESUMO

RNA virus vectors are attractive vaccine delivery agents capable of directing high-level gene expression without integration into host cell DNA. However, delivery of non-encapsidated RNA viral vectors into animal cells is relatively inefficient. By introducing the tobacco mosaic virus (TMV) origin of assembly into the RNA genome of Semliki Forest virus (SFV), we generated an SFV expression vector that could be efficiently packaged (trans-encapsidated) in vitro by purified TMV coat protein (CP). Using cellular assays, pseudovirus disassembly, RNA replication and reporter gene expression were demonstrated. We also evaluated the immune response to trans-encapsidated recombinant SFV carrying a model antigen gene (beta-galactosidase) in C57/B6 mice. Relative to RNA alone, vector encapsidation significantly improved the humoral and cellular immune responses. Furthermore, reassembly with recombinant TMV CPs permitted the display of peptide epitopes on the capsid surface as either genetic fusions or through chemical conjugation, to complement the immunoreactivity of the encapsidated RNA genetic payload. The SFV vector/TMV CP system described provides an alternative nucleic acid delivery mechanism that is safe, easy to manufacture in vitro and that also facilitates the generation of unique nucleic acid/protein antigen compositions.


Assuntos
Vetores Genéticos/metabolismo , RNA Viral/metabolismo , Vírus da Floresta de Semliki/genética , Vírus da Floresta de Semliki/metabolismo , Vírus do Mosaico do Tabaco/fisiologia , Proteínas Virais/metabolismo , Vacinas Virais/imunologia , beta-Galactosidase/metabolismo , Animais , Anticorpos Antivirais/sangue , Proteínas do Capsídeo/metabolismo , Feminino , Imunização , Esquemas de Imunização , Injeções Subcutâneas , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Baço/imunologia , Linfócitos T/imunologia , Vírus do Mosaico do Tabaco/genética , Proteínas Virais/imunologia , Vacinas Virais/administração & dosagem , Replicação Viral , beta-Galactosidase/imunologia
19.
Bioconjug Chem ; 17(5): 1330-8, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16984144

RESUMO

Chemical conjugation of CTL peptides to tobacco mosaic virus (TMV) has shown promise as a molecular adjuvant scaffold for augmentation of cellular immune responses to peptide vaccines. This study demonstrates the ease of generating complex multipeptide vaccine formulations using chemical conjugation to TMV for improved vaccine efficacy. We have tested a model foreign antigen target-the chicken ovalbumin-derived CTL peptide (Ova peptide), as well as mouse melanoma-associated CTL epitopes p15e and tyrosinase-related protein 2 (Trp2) peptides that are self-antigen targets. Ova peptide fusions to TMV, as bivalent formulations with peptides encoding additional T-help or cellular uptake via the integrin-receptor binding RGD peptide, showed improved vaccine potency evidenced by significantly enhanced numbers of antigen-reactive T cells measured by in vitro IFNgamma cellular analysis. We measured the biologically relevant outcome of vaccination in protection of mice from EG.7-Ova tumor challenge, which was achieved with only two doses of vaccine ( approximately 600 ng peptide) given without adjuvant. The p15e peptide alone or Trp2 peptide alone, or as a bivalent formulation with T-help or RGD uptake epitopes, was unable to stimulate effective tumor protection. However, a vaccine with both CTL peptides fused together onto TMV generated significantly improved survival. Interestingly, different bivalent vaccine formulations were required to improve vaccine efficacy for Ova or melanoma tumor model systems.


Assuntos
Vacinas Anticâncer/biossíntese , Imunidade Celular/fisiologia , Neoplasias/prevenção & controle , Peptídeos/metabolismo , Vírus do Mosaico do Tabaco/metabolismo , Adjuvantes Imunológicos/metabolismo , Animais , Galinhas , Epitopos , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos/genética , Taxa de Sobrevida , Vírus do Mosaico do Tabaco/genética
20.
Vaccine ; 24(40-41): 6414-23, 2006 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-16860441

RESUMO

Fusion of peptides to viral carriers has proven an effective method for improving cellular immunity. In this study we explore the ability of a plant virus, Tobacco mosaic virus (TMV), to stimulate cellular immunity by interacting directly with immune cells. Fluorescently labeled TMV was incubated in vitro with murine spleen or lymph node cells, and near quantitative labeling of lymphocytes was achieved after 2 h, which persisted for up to 48 h. Direct TMV uptake and upregulation of the CD86 activation marker was measured in nearly all dendritic cells (DCs) by flow cytometry. To demonstrate that TMV can also provide functional antigen delivery and immune stimulation in vivo, two well-characterized T-cell epitopes that provide protection against tumor challenge in mice were fused to TMV coat protein by genetic manipulation, or by chemical conjugation. Vaccination of C57BL/6 mice elicited measurable cellular responses by interferon gamma (IFN gamma) ELISpot and resulted in significantly improved protection from tumor challenge in both the EG.7-Ova and B16 melanoma models. From these results we conclude that TMV was an effective antigen carrier for inducing cellular immune responses to less than 1 microg of peptide.


Assuntos
Vacinas Anticâncer/imunologia , Proteínas do Capsídeo/imunologia , Modelos Animais de Doenças , Engenharia Genética , Neoplasias/imunologia , Neoplasias/prevenção & controle , Vírus do Mosaico do Tabaco/genética , Animais , Medula Óssea/imunologia , Vacinas Anticâncer/química , Vacinas Anticâncer/genética , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Linhagem Celular Tumoral , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Transplante de Neoplasias/imunologia , Neoplasias/patologia , Estrutura Terciária de Proteína , Baço/metabolismo , Taxa de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...