Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Am J Trop Med Hyg ; 76(4): 743-51, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17426182

RESUMEN

There are approximately 100 million new cases of dengue (DEN) virus infection each year. Infection can result in illness ranging from a mild fever to hemorrhaging, shock, or even death. There are four serotypes of dengue virus (DEN1-4), and immunity to one serotype does not cross protect from infection with other serotypes. Currently there are no approved vaccines for dengue fever. In this report, we describe the construction of a bivalent dengue virus vaccine using a complex recombinant adenovirus approach to express multiple genes of DEN1 and DEN2 serotypes. In vaccinated mice, this vector induced humoral immune responses against all four dengue serotypes as measured by enzyme-linked immunosorbent assay. However, the neutralizing antibody responses were specific for DEN1 and DEN2 serotypes. Expansion of this vaccine development platform towards the DEN3 and DEN4 serotypes can lead towards the development of an adenovirus-based tetravalent dengue vaccine.


Asunto(s)
Adenoviridae/genética , Antígenos Virales/genética , Antígenos Virales/inmunología , Virus del Dengue/genética , Virus del Dengue/inmunología , Dengue/inmunología , Dengue/virología , Vacunas Virales/genética , Vacunas Virales/inmunología , Animales , Línea Celular , Chlorocebus aethiops , Expresión Génica , Humanos , Ratones , Células Vero
2.
Vaccine ; 28(31): 5135-44, 2010 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-19737527

RESUMEN

Plasmodium falciparum Liver Stage Antigen 1 (LSA-1) is a pre-erythrocytic stage antigen. Our LSA-1 vaccine candidate is a recombinant protein with full-length C- and N-terminal flanking domains and two of the 17 amino acid repeats from the central repeat region termed "LSA-NRC." We describe the first Phase I/II study of this recombinant LSA-NRC protein formulated with either the AS01 or AS02 adjuvant system. We conducted an open-label Phase I/II study. Thirty-six healthy malaria-naïve adults received one of four formulations by intra-deltoid injection on a 0 and 1 month schedule; low dose (LD) LSA-NRC/AS01:10microg LSA-NRC/0.5ml AS01 (n=5), high dose (HD) LSA-NRC/AS01: 50microg LSA-NRC/0.5ml AS01 (n=13); LD LSA-NRC/AS02: 10microg LSA-NRC/0.5ml AS02 (n=5) and HD LSA-NRC/AS02: 50microg LSA-NRC/0.5ml AS02 (n=13). Two weeks post-second immunization, the high dose vaccinees and 6 non-immunized infectivity controls underwent experimental malaria sporozoite challenge. The vaccines showed a reassuring safety profile but were moderately reactogenic. There were no serious adverse events. All subjects seroconverted after the first immunization. Following the second immunization, LSA-1-specific CD4+ T cells producing two cytokines (IL-2 and IFN-gamma) were found by intra-cellular staining in all subjects in the LD LSA-NRC/AS01B group and in 3 of 5 subjects in the LD LSA-NRC/AS02 group. In contrast, the HD LSA-NRC/AS01 and HD LSA-NRC/AS02 group subjects had fewer LSA-1-specific CD4+ T cells, and minimal to no IFN-gamma responses. There was no increase in LSA-1-specific CD8+ T cells found in any group. Per protocol, 22 high dose vaccinees, but no low dose vaccinees, underwent P. falciparum homologous malaria challenge (3D7 clone). All vaccinees became parasitemic and there was no delay in their pre-patent period versus controls (p=0.95). LSA-NRC/AS01 and LSA-NRC/AS02 elicited antigen-specific antibody and CD4+ T cell responses, but elicited no protective immunity. Although the optimal antigen dose of LSA-NRC may not have been selected for the challenge portion of the protocol, further vaccine development based upon LSA-1 should not be excluded and should include alternative vaccine platforms able to elicit additional effector mechanisms such as CD8+ T cells.


Asunto(s)
Antígenos de Protozoos/inmunología , Linfocitos T CD4-Positivos/inmunología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/prevención & control , Adyuvantes Inmunológicos/farmacología , Adulto , Anticuerpos Antiprotozoarios/sangre , Formación de Anticuerpos , Femenino , Humanos , Inmunidad Celular , Inmunidad Humoral , Esquemas de Inmunización , Inmunización Secundaria , Interferón gamma/inmunología , Interleucina-2/inmunología , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/efectos adversos , Malaria Falciparum/inmunología , Masculino , Parasitemia/inmunología , Plasmodium falciparum/inmunología , Proteínas Recombinantes/inmunología , Esporozoítos/inmunología , Adulto Joven
3.
AIDS Res Hum Retroviruses ; 24(4): 573-86, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18426337

RESUMEN

The mechanisms of resistance to AIDS development in HIV-1-infected chimpanzees have remained elusive. Unique among chimpanzees naturally or experimentally infected with HIV, several animals of the Yerkes cohort have progressed to clinical AIDS with selection of isolates showing increased pathogenicity for chimpanzees. We compared progressors vs. nonprogressors among the HIV-infected chimpanzees that made up this cohort, eight of which have been infected with HIV-1 for over 14 years. The additional two progressors were infected de novo with chimpanzee-pathogenic HIV, rapidly leading to a progressor status. Nonprogressors were characterized by normal CD4(+) T cell counts and the absence of detectable viremia. In contrast, progressor chimpanzees had relatively high plasma viral loads associated with a dramatic loss of CD4(+) T cells. The analysis of immune responses showed a similar amplitude and breadth of ELISPOT T cell responses in both groups. HIV-specific proliferative responses were, however, absent in the progressor animals, which also exhibited increased levels of immune activation characterized by elevated levels of the circulating chemokines IP-10 and MCP-1. Of interest was the conservation of potent NK cell activity in all animals, potentially contributing to the extended symptom-free survival of progressor animals. Modest anti-HIV antibody titers were detectable in the nonprogressor group, but these antibodies exhibited good neutralizing activity. In progressors, however, two sets of data were noted: in animals that gradually selected for pathogenic isolates, or that were superinfected, very high neutralizing antibody titers were observed, although none to the pathogenic HIV. In contrast, two animals infected de novo with chimpanzee pathogenic HIV failed to mount an extensive humoral response and both failed to develop neutralizing antibodies to the virus. Taken together, pathogenic HIV infection in chimpanzees is associated with rapid loss of CD4(+) T cells and proliferative responses as well as higher levels of immune activation.


Asunto(s)
Infecciones por VIH/inmunología , VIH-1 , Animales , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Células Cultivadas , Quimiocinas/sangre , Quimiocinas/metabolismo , Anticuerpos Anti-VIH/sangre , Anticuerpos Anti-VIH/inmunología , Infecciones por VIH/sangre , Humanos , Células Asesinas Naturales/inmunología , Leucocitos Mononucleares , Activación de Linfocitos , Pruebas de Neutralización , Pan troglodytes , Receptores de Citocinas/sangre , Receptores de Citocinas/metabolismo
4.
Virology ; 353(2): 324-32, 2006 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-16820184

RESUMEN

The Marburg virus (MARV), an African filovirus closely related to the Ebola virus, causes a deadly hemorrhagic fever in humans, with up to 90% mortality. Currently, treatment of disease is only supportive, and no vaccines are available to prevent spread of MARV infections. In order to address this need, we have developed and characterized a novel recombinant vaccine that utilizes a single complex adenovirus-vectored vaccine (cAdVax) to overexpress a MARV glycoprotein (GP) fusion protein derived from the Musoke and Ci67 strains of MARV. Vaccination with the cAdVaxM(fus) vaccine led to efficient production of MARV-specific antibodies in both mice and guinea pigs. Significantly, guinea pigs vaccinated with at least 5 x 10(7) pfu of cAdVaxM(fus) vaccine were 100% protected against lethal challenges by the Musoke, Ci67 and Ravn strains of MARV, making it a vaccine with trivalent protective efficacy. Therefore, the cAdVaxM(fus) vaccine serves as a promising vaccine candidate to prevent and contain multi-strain infections by MARV.


Asunto(s)
Adenoviridae/metabolismo , Antígenos Virales/biosíntesis , Vacunas contra el Virus del Ébola/administración & dosificación , Vectores Genéticos/metabolismo , Enfermedad del Virus de Marburg/prevención & control , Marburgvirus/inmunología , Vacunación , Proteínas del Envoltorio Viral/biosíntesis , Proteínas Virales de Fusión/biosíntesis , Adenoviridae/genética , Secuencia de Aminoácidos , Animales , Anticuerpos Antivirales/sangre , Antígenos Virales/genética , Línea Celular , Chlorocebus aethiops , Relación Dosis-Respuesta Inmunológica , Vacunas contra el Virus del Ébola/genética , Terapia Genética/métodos , Cobayas , Humanos , Inyecciones Intraperitoneales , Inyecciones Subcutáneas , Enfermedad del Virus de Marburg/sangre , Enfermedad del Virus de Marburg/inmunología , Ratones , Datos de Secuencia Molecular , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Alineación de Secuencia , Vacunas Sintéticas/administración & dosificación , Proteínas del Envoltorio Viral/genética , Proteínas Virales de Fusión/genética
5.
J Virol ; 80(6): 2738-46, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16501083

RESUMEN

Ebola virus (EBOV) causes a severe hemorrhagic fever for which there are currently no vaccines or effective treatments. While lethal human outbreaks have so far been restricted to sub-Saharan Africa, the potential exploitation of EBOV as a biological weapon cannot be ignored. Two species of EBOV, Sudan ebolavirus (SEBOV) and Zaire ebolavirus (ZEBOV), have been responsible for all of the deadly human outbreaks resulting from this virus. Therefore, it is important to develop a vaccine that can prevent infection by both lethal species. Here, we describe the bivalent cAdVaxE(GPs/z) vaccine, which includes the SEBOV glycoprotein (GP) and ZEBOV GP genes together in a single complex adenovirus-based vaccine (cAdVax) vector. Vaccination of mice with the bivalent cAdVaxE(GPs/z) vaccine led to efficient induction of EBOV-specific antibody and cell-mediated immune responses to both species of EBOV. In addition, the cAdVax technology demonstrated induction of a 100% protective immune response in mice, as all vaccinated C57BL/6 and BALB/c mice survived challenge with a lethal dose of ZEBOV (30,000 times the 50% lethal dose). This study demonstrates the potential efficacy of a bivalent EBOV vaccine based on a cAdVax vaccine vector design.


Asunto(s)
Adenoviridae/genética , Vacunas contra el Virus del Ébola/administración & dosificación , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/prevención & control , Proteínas del Envoltorio Viral/inmunología , Adenoviridae/metabolismo , Animales , Anticuerpos Antivirales/sangre , Línea Celular , Vacunas contra el Virus del Ébola/genética , Vacunas contra el Virus del Ébola/inmunología , Ebolavirus/genética , Ebolavirus/inmunología , Ebolavirus/patogenicidad , Células HeLa , Fiebre Hemorrágica Ebola/virología , Humanos , Inmunidad Celular , Inmunización , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Especificidad de la Especie , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo
6.
Vaccine ; 24(15): 2975-86, 2006 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-16530297

RESUMEN

Marburg virus (MARV) is an African filovirus that causes a deadly hemorrhagic fever in humans, with up to 90% mortality. Currently, there are no MARV vaccines or therapies approved for human use. We hypothesized that developing a vaccine that induces a de novo synthesis of MARV antigens in vivo will lead to strong induction of both a humoral and cell-mediated immune response against MARV. Here, we develop and characterize three novel gene-based vaccine candidates which express the viral glycoprotein (GP) from either the Ci67, Ravn or Musoke strain of MARV. Immunization of mice with complex adenovirus (Ad)-based vaccine candidates (cAdVax vaccines), led to efficient production of both antibodies and cytotoxic T lymphocytes (CTL) specific to Musoke strain GP and Ci67 strain GP, respectively. Antibody responses were also shown to be cross-reactive across the MARV strains, but not cross-reactive to Ebola virus, a related filovirus. Additionally, three 1 x 10(8)pfu doses of vaccine vector were demonstrated to be safe in mice, as this did not lead to any detectable toxicity in liver or spleen. These promising results indicate that a cAdVax-based vaccine could be effective for induction of both humoral and cell-mediated immune responses to multiple strains of the Marburg virus.


Asunto(s)
Adenoviridae/genética , Enfermedad del Virus de Marburg/prevención & control , Marburgvirus/genética , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Antígenos Virales/biosíntesis , Antígenos Virales/genética , Antígenos Virales/inmunología , Reacciones Cruzadas , Pruebas Inmunológicas de Citotoxicidad , Ensayo de Inmunoadsorción Enzimática , Vectores Genéticos , Humanos , Hígado/patología , Enfermedad del Virus de Marburg/inmunología , Marburgvirus/inmunología , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Bazo/patología , Linfocitos T Citotóxicos/inmunología , Proteínas del Envoltorio Viral/biosíntesis , Proteínas del Envoltorio Viral/genética , Vacunas Virales/administración & dosificación , Vacunas Virales/efectos adversos , Vacunas Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA