Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
1.
Sci Rep ; 9(1): 20005, 2019 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-31882800

RESUMEN

Multiple approaches utilizing viral and DNA vectors have shown promise in the development of an effective vaccine against HIV. In this study, an alternative replication-defective flavivirus vector, RepliVax (RV), was evaluated for the delivery of HIV-1 immunogens. Recombinant RV-HIV viruses were engineered to stably express clade C virus Gag and Env (gp120TM) proteins and propagated in Vero helper cells. RV-based vectors enabled efficient expression and correct maturation of Gag and gp120TM proteins, were apathogenic in a sensitive suckling mouse neurovirulence test, and were similar in immunogenicity to recombinant poxvirus NYVAC-HIV vectors in homologous or heterologous prime-boost combinations in mice. In a pilot NHP study, immunogenicity of RV-HIV viruses used as a prime or boost for DNA or NYVAC candidates was compared to a DNA prime/NYVAC boost benchmark scheme when administered together with adjuvanted gp120 protein. Similar neutralizing antibody titers, binding IgG titers measured against a broad panel of Env and Gag antigens, and ADCC responses were observed in the groups throughout the course of the study, and T cell responses were elicited. The entire data demonstrate that RV vectors have the potential as novel HIV-1 vaccine components for use in combination with other promising candidates to develop new effective vaccination strategies.


Asunto(s)
Vacunas contra el SIDA/inmunología , Virus Defectuosos/genética , Flavivirus/genética , Vectores Genéticos , VIH-1/inmunología , Vacunas Sintéticas/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Chlorocebus aethiops , Reacciones Cruzadas , Femenino , Infecciones por VIH/virología , VIH-1/patogenicidad , Macaca mulatta , Ratones , Ratones Endogámicos BALB C , Células Vero , Virulencia
2.
Vaccine ; 27(14): 1997-2015, 2009 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-19200450

RESUMEN

Generating broadly neutralizing antibodies with candidate vaccines has remained an elusive goal. Consequently, vaccine candidates developed have aimed at eliciting cell-mediated immune effector activities (CMI) that could delay disease progression, and maybe also limit secondary transmission, by controlling virus replication. There is considerable discussion about what types of endpoints would constitute definable standardized clinical benefit to the individual that would result in licensure of these candidate vaccines. Identifying biomarkers that can be used as surrogates for clinical endpoints in randomized clinical trials would be useful, because it would shorten studies and reduce costs. Biological markers associated with disease progression and secondary transmission and that may be used as prognosis markers and surrogate endpoints in HIV vaccine trials have emerged from analyses of data from studies on natural history of HIV infection. Extensive literature is cited to support the use of plasma viral load as a primary endpoint for supporting licensure decisions. Overall, a significant result on viral load in a vaccine trial should be considered as a significant breakthrough for vaccines and be aggressively pursued with the caveat that such a result should rapidly be followed by well-defined studies to verify durable virological and immunological vaccine benefit, as well as ultimate clinical benefit. The review also provides perspectives on magnitude of viral load reduction, durability of viral load reduction for reduced progression of HIV disease.


Asunto(s)
Vacunas contra el SIDA/inmunología , Infecciones por VIH/terapia , VIH-1/inmunología , Carga Viral , Vacunas contra el SIDA/administración & dosificación , Animales , Biomarcadores/sangre , Ensayos Clínicos como Asunto , Progresión de la Enfermedad , Humanos , ARN Viral/sangre , Linfocitos T Citotóxicos/inmunología
3.
Gene Ther ; 10(26): 2126-32, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14625567

RESUMEN

We tested the canarypox virus vector ALVAC and the genetically attenuated vaccinia virus vector NYVAC as vehicles for achieving local immunomodulation in domestic animals bearing spontaneous tumours. Following intratumoral administration of ALVAC-, or NYVAC-luciferase in dogs with melanoma, it was demonstrated that viral recombinants remained localized along the needle track, with no virus detectable in the periphery of the tumour. Given these distribution characteristics and their well-documented safety profile, ALVAC- or NYVAC-based recombinants expressing feline or human IL2, respectively, were administered to domestic cats, in order to prevent the recurrence of spontaneous fibrosarcomas. In the absence of immunotherapy, tumour recurrence was observed in 61% of animals within a 12-month follow-up period after treatment with surgery and iridium-based radiotherapy. In contrast, only 39 and 28% of cats receiving either NYVAC-human IL2 or ALVAC-feline IL2, respectively, exhibited tumour recurrences. Based on such results, and in the context of ongoing clinical studies conducted in humans, we discuss the utilization of ALVAC- or NYVAC-based recombinants as viable therapeutic modalities for local immunotherapy or therapeutic vaccination against cancer, both in humans and companion animals.


Asunto(s)
Enfermedades de los Gatos/terapia , Fibrosarcoma/terapia , Inmunoterapia/métodos , Interleucina-2/metabolismo , Neoplasias Cutáneas/terapia , Vacunas Virales/uso terapéutico , Animales , Animales Domésticos , Virus de la Viruela de los Canarios/metabolismo , Enfermedades de los Gatos/enzimología , Gatos , Perros , Femenino , Fibrosarcoma/enzimología , Vectores Genéticos , Luciferasas/metabolismo , Masculino , Recurrencia Local de Neoplasia/prevención & control , Neoplasias Cutáneas/enzimología , Vacunas Virales/metabolismo , Vacunas Virales/farmacocinética
4.
Vet Rec ; 153(5): 141-5, 2003 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-12934796

RESUMEN

Canarypox virus recombinant vaccines have a unique efficacy and safety profile for the vaccinated host because the canarypox virus is non-replicative in mammalian hosts. After the vaccination of a mammalian species, recombinant canarypox viruses express the inserted genes but cannot multiply in the host. They stimulate a strong immune response in the absence of any virus amplification in the host or any viral spread into the environment. A new canarypox-based recombinant vaccine is the canarypox-feline leukaemia virus (FeLV) vaccine (EURIFEL FeLV; Merial) that expresses the FeLV env and gag protective genes. This paper describes experiments which demonstrate that it is effective against any oronasal FeLV challenge. The protection was shown to be solid against an oronasal challenge one year after the initial vaccination, and was effective against a very severe 'in-contact' challenge. Furthermore, the canarypox virus-FeLV vaccine was effective without an adjuvant.


Asunto(s)
Virus de la Viruela de los Canarios/genética , Virus de la Leucemia Felina/inmunología , Leucemia Felina/prevención & control , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/farmacología , Animales , Formación de Anticuerpos , Gatos , Embrión de Pollo , Regulación de la Expresión Génica , Leucemia Felina/inmunología
5.
Clin Cardiol ; 26(6): 287-90, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12839048

RESUMEN

BACKGROUND: Enhanced external counterpulsation (EECP) has been shown to improve treadmill times and myocardial perfusion. However, improvement in perfusion defects has been demonstrated only in patients exercised to the same cardiac workload on the post-EECP as the pre-EECP stress test. HYPOTHESIS: This study was to determine the effect of EECP on exercise capacity and myocardial perfusion by comparing results of maximal exercise radionuclide testing pre- and post-EECP treatment. METHODS: This prospective study included 25 patients with angina who had performed maximal symptom-limited exercise tolerance tests (ETT) with Bruce protocol and radionuclide perfusion single-photon emission computed tomography (SPECT) study prior to and at completion of EECP treatment. RESULTS: After 35 h of EECP, 23 patients (93%) improved by at least one functional angina class. There is a significant improvement in their total treadmill times (357 +/- 93 to 449 +/- 97 s, p < 0.001). There was a significant change in their peak double products, from 18,891 +/- 3,939 pre-EECP to 20,464 +/- 4,305 post-EECP ETT (p < 0.03). Pre EECP, 16 patients had ST-segment depression on their initial ETT. After EECP, 13 of these patients (80%) either no longer had ST depression or had a significant increase in their time to ST depression (229 +/- 52 to 315 +/- 60 s, p < 0.001). The radionuclide perfusion scores also showed a significant reduction in ischemic segments (16.36 +/- 10.52 to 14 +/- 10.9, p < 0.05). CONCLUSIONS: Patients treated with EECP demonstrated a reduction in angina symptoms, improvement in exercise capacity, increase in time to ST-segment depression, and decrease in perfusion defects despite performing at a higher workload.


Asunto(s)
Angina de Pecho/fisiopatología , Angina de Pecho/cirugía , Circulación Coronaria/fisiología , Contrapulsación , Tolerancia al Ejercicio/fisiología , Anciano , Angina de Pecho/clasificación , Angina de Pecho/diagnóstico por imagen , Presión Sanguínea/fisiología , Electrocardiografía , Prueba de Esfuerzo , Femenino , Humanos , Masculino , Estudios Prospectivos , Tomografía Computarizada de Emisión de Fotón Único , Resultado del Tratamiento
6.
DNA Cell Biol ; 21(9): 619-26, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12396604

RESUMEN

Most vaccine modalities for human immunodeficiency virus type 1 (HIV-1) tested for immunogenicity and efficacy in the SIVmac (simian immunodeficiency virus) macaque model do not include the viral regulatory proteins. Because viral regulatory proteins are expressed early during the virus life cycle and represent an additional source of antigens, their inclusion as a vaccine component may increase the overall virus-specific immune response in vaccinees. However, at least two of the early proteins, Tat and Nef, may be immunosuppressive, limiting their usefulness as components of an SIV vaccine. We have constructed a polyvalent chimeric protein in which the open reading frames for Tat and Nef have been reassorted and the nuclear localization sequence for Tat and Rev and the myristoylation site for Nef have been removed. The resulting DNA plasmid (pDNA-SIV-Retanef) (pDNA-SIV-RTN) encodes a protein of 55 kDa (Retanef) that localizes at the steady state in the cytoplasma of transfected cells. Both the DNA-SIV-RTN and the highly attenuated recombinant poxvirus vector NYVAC-SIV-RTN were demonstrated to be immunogenic in SIVmac251-infected macaques treated with ART as well as in naive macaques. An equivalent strategy may be used for the generation of polyvalent antigens encoding the regulatory proteins in a HIV-1 vaccine candidate.


Asunto(s)
Secuencias Reguladoras de Ácidos Nucleicos , Virus de la Inmunodeficiencia de los Simios/inmunología , Vacunas/inmunología , Animales , Productos del Gen gag/metabolismo , Genes nef/inmunología , Genes rev/inmunología , Genes tat/inmunología , Células HeLa , Humanos , Macaca mulatta , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Vacunas Sintéticas/inmunología , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Reguladoras y Accesorias Virales/inmunología
7.
J Virol ; 76(1): 292-302, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11739694

RESUMEN

T-cell-mediated immune effector mechanisms play an important role in the containment of human immunodeficiency virus/simian immunodeficiency virus (HIV/SIV) replication after infection. Both vaccination- and infection-induced T-cell responses are dependent on the host major histocompatibility complex classes I and II (MHC-I and MHC-II) antigens. Here we report that both inherent, host-dependent immune responses to SIVmac251 infection and vaccination-induced immune responses to viral antigens were able to reduce virus replication and/or CD4+ T-cell loss. Both the presence of the MHC-I Mamu-A*01 genotype and vaccination of rhesus macaques with ALVAC-SIV-gag-pol-env (ALVAC-SIV-gpe) contributed to the restriction of SIVmac251 replication during primary infection, preservation of CD4+ T cells, and delayed disease progression following intrarectal challenge exposure of the animals to SIV(mac251 (561)). ALVAC-SIV-gpe immunization induced cytotoxic T-lymphocyte (CTL) responses cumulatively in 67% of the immunized animals. Following viral challenge, a significant secondary virus-specific CD8+ T-cell response was observed in the vaccinated macaques. In the same immunized macaques, a decrease in virus load during primary infection (P = 0.0078) and protection from CD4 loss during both acute and chronic phases of infection (P = 0.0099 and P = 0.03, respectively) were observed. A trend for enhanced survival of the vaccinated macaques was also observed. Neither boosting the ALVAC-SIV-gpe with gp120 immunizations nor administering the vaccine by the combination of mucosal and systemic immunization routes increased significantly the protective effect of the ALVAC-SIV-gpe vaccine. While assessing the role of MHC-I Mamu-A*01 alone in the restriction of viremia following challenge of nonvaccinated animals with other SIV isolates, we observed that the virus load was not significantly lower in Mamu-A*01-positive macaques following intravenous challenge with either SIV(mac251 (561)) or SIV(SME660). However, a significant delay in CD4+ T-cell loss was observed in Mamu-A*01-positive macaques in each group. Of interest, in the case of intravenous or intrarectal challenge with the chimeric SIV/HIV strains SHIV(89.6P) or SHIV(KU2), respectively, MHC-I Mamu-A*01-positive macaques did not significantly restrict primary viremia. The finding of the protective effect of the Mamu-A*01 molecule parallels the protective effect of the B*5701 HLA allele in HIV-1-infected humans and needs to be accounted for in the evaluation of vaccine efficacy against SIV challenge models.


Asunto(s)
Productos del Gen env/administración & dosificación , Productos del Gen gag/administración & dosificación , Productos del Gen pol/administración & dosificación , Antígenos de Histocompatibilidad Clase I/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Virus de la Inmunodeficiencia de los Simios/inmunología , Vacunación , Vacunas Virales/administración & dosificación , Animales , Productos del Gen env/inmunología , Productos del Gen gag/inmunología , Productos del Gen pol/inmunología , Macaca , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus Vaccinia , Vacunas Virales/inmunología
8.
J Immunol ; 167(12): 7180-91, 2001 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11739541

RESUMEN

T cell-mediated immune responses play an important role in the containment of HIV-1 replication. Therefore, an effective vaccine against HIV-1 should be able to elicit high frequencies of virus-specific CD8(+) and CD4(+) T cells. The highly attenuated poxvirus-based vaccine candidate, NYVAC-SIV-gag-pol-env (NYVAC-SIV-gpe), has been shown to induce and/or expand SIV-specific CD4(+) and CD8(+) T cell responses in both naive and infected macaques. In this study, the immunogenicity of NYVAC-SIV-gpe alone was compared with a combination regimen where priming with an optimized DNA-SIV-gag-env vaccine candidate was followed by a NYVAC-SIV-gpe boost. In macaques immunized with the prime-boost regimen, the extent and durability of CD8(+) T cell response to an immunodominant SIV gag epitope was increased and these animals recognized a broader array of subdominant SIV epitopes in the cytolytic assay. In addition, the prime-boost regimen significantly enhanced the proliferative responses to both SIV gag and env proteins. Thus, the combination of these vaccine modalities may represent a valuable strategy in the development of a vaccine for HIV.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el SIDAS/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Virus de la Inmunodeficiencia de los Simios/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/inmunología , Antígenos Virales/inmunología , Células Cultivadas , Pruebas Inmunológicas de Citotoxicidad , Esquemas de Inmunización , Inmunización Secundaria , Interferón gamma/biosíntesis , Cinética , Activación de Linfocitos , Macaca mulatta , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/genética , Vacunas Atenuadas/inmunología , Vacunas de ADN/inmunología
9.
Cancer Immunol Immunother ; 50(7): 345-55, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11676394

RESUMEN

Natural attenuation of ALVAC virus in mammals makes it an attractive vector for cancer vaccine therapy of immunocompromised hosts, such as patients with lymphoid malignancies. However, the transduction efficiency of ALVAC constructs in lymphoid tumors has not yet been characterized. We studied a wide spectrum of human T- and B-cell leukemia and lymphomas and found significant heterogeneity of the ALVAC-mediated gene product expression in these tumors. While ALVAC-B7.1, ALVAC-B7.2, or ALVAC-luciferase vectors effectively expressed recombinant genes in malignancies arising from T- or early B-cell precursors, negative or low expression of ALVAC recombinant genes occurred in tumors arising from mature B-cells. We showed that ALVAC-encoded B7.1 or B7.2 was continuously expressed on the infected, and subsequently irradiated, leukemia cells, and only cells with ALVAC-mediated expression of costimulatory molecules (but not unmodified leukemia cells or those infected with the ALVAC-parental vector) induced significant proliferation and IFN-gamma production by alloreactive T-cells. These data provide the rationale for clinical studies using the ALVAC vector system for gene transfer into lymphoid tumors of T- and early B-cell origin to render them more immunogenic, while alternative strategies should be considered for immunotherapy of mature B-cell malignancies.


Asunto(s)
Avipoxvirus/genética , Linfocitos B/metabolismo , Transferencia de Gen Horizontal , Vectores Genéticos , Leucemia/metabolismo , Linfoma/metabolismo , Animales , Antígenos CD/genética , Antígenos CD/fisiología , Antígeno B7-1/genética , Antígeno B7-1/fisiología , Antígeno B7-2 , Humanos , Interferón gamma/biosíntesis , Activación de Linfocitos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Ratones , Linfocitos T/inmunología , Células Tumorales Cultivadas
10.
Cell Immunol ; 210(1): 1-4, 2001 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-11485346

RESUMEN

Profound alterations in humoral and cellular immune responses are a hallmark of aging, and understanding the immunobiology of aging is key to the success of preventive vaccination strategies. With aging, while recall or memory responses to influenza viral antigens for the most part remained unaltered, primary immune responses are severely impaired. The impaired primary responses are partly due to a lack of costimulation, as providing costimulation at the time of induction of primary immune responses against influenza virus vaccine partially reversed aged-related immune dysfunction and conferred enhanced protection. Inclusion of immunomodulators that up-regulate the expression of costimulatory molecules must be considered to improve the efficacy of vaccination in the elderly, particularly to novel immunogens.


Asunto(s)
Envejecimiento/inmunología , Antígenos Virales/inmunología , Memoria Inmunológica , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/farmacología , Infecciones por Orthomyxoviridae/inmunología , Adyuvantes Inmunológicos/biosíntesis , Adyuvantes Inmunológicos/genética , Animales , Anticuerpos Antivirales/biosíntesis , Antígenos CD/biosíntesis , Antígenos CD/genética , Antígeno B7-1/biosíntesis , Antígeno B7-1/genética , Antígeno B7-2 , Células Cultivadas , Femenino , Vectores Genéticos , Activación de Linfocitos , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos DBA , Análisis de Supervivencia , Linfocitos T Citotóxicos/inmunología , Regulación hacia Arriba , Virus Vaccinia/genética
11.
J Natl Cancer Inst ; 93(13): 998-1007, 2001 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-11438565

RESUMEN

BACKGROUND: Immunization with modified tumor cells carrying recombinant immunomodulatory genes is being explored as cancer immunotherapy. In this study, we examine whether canarypox ALVAC viruses carrying immunostimulatory cytokine genes (granulocyte-macrophage colony-stimulating factor, interleukin 2, interleukin 12, and tumor necrosis factor-alpha) can induce antitumor immunity (to rechallenge) in the RM-1 model of a highly aggressive, weakly immunogenic murine prostate cancer. METHODS: For antitumor activity studies, RM-1 murine prostate cancer cells were infected with the parental ALVAC virus or one or two recombinant ALVAC-cytokine viruses and then injected into male C57BL/6 mice. For rechallenge studies, other mice were first given an injection subcutaneously with irradiated (nonproliferating) recombinant ALVAC-infected RM-1 cells and then (10 days later) with untreated RM-1 cells. For the determination of which immune cells were required for antitumor activity, mice were immunodepleted of CD4, CD8, or natural killer (NK) NK1.1 cells with the corresponding monoclonal antibodies and were then given an injection of ALVAC-cytokine-infected RM-1 cells. For all experiments, tumor outgrowth and animal survival were monitored. RESULTS: After subcutaneous injection into mice, RM-1 cells infected with one (except ALVAC-interleukin 2) or two ALVAC-cytokine recombinants had statistically significantly greater antitumor activity than RM-1 cells infected with parental ALVAC (P<.001 for all; two-sided test). The antitumor activity of RM-1 cells infected with any two ALVAC-cytokine recombinants was greater than, but not statistically significantly different from, that of RM-1 cells infected with any one ALVAC-cytokine recombinant. NK1.1 cells were necessary for antitumor activity, but tumor-specific CD4(+) regulatory T cells were also induced that inhibited CD8(+) RM-1-specific cytotoxic T cells, resulting in the lack of immunity to a rechallenge by RM-1 cells. DISCUSSION: Canarypox viruses can transfer immunostimulatory cytokine genes into RM-1 prostate cancer cells. When such cells were injected into mice, the cytokines induced an antitumor response against this highly aggressive, weakly immunogenic tumor. This response, however, did not protect the mouse against a rechallenge with RM-1 cells because suppressor CD4(+) T cells were induced that inhibited tumor-specific CD8(+) cytotoxic T cells.


Asunto(s)
Avipoxvirus/genética , Neoplasias de la Próstata/terapia , Proteínas , Animales , Anticuerpos Monoclonales/metabolismo , Antígenos/biosíntesis , Antígenos Ly , Antígenos de Superficie , Antígenos CD4/biosíntesis , Antígenos CD8/biosíntesis , Citometría de Flujo , Técnicas de Transferencia de Gen , Terapia Genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Interleucina-12/genética , Interleucina-2/genética , Lectinas Tipo C , Masculino , Ratones , Ratones Endogámicos C57BL , Subfamilia B de Receptores Similares a Lectina de Células NK , Trasplante de Neoplasias , Neoplasias de la Próstata/inmunología , Biosíntesis de Proteínas , Proteínas Recombinantes/metabolismo , Linfocitos T Citotóxicos/metabolismo , Factores de Tiempo , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/genética
12.
Vaccine ; 19(30): 4267-75, 2001 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-11457554

RESUMEN

Poxviruses are large DNA viruses capable of infecting a broad range of animal species. Infection is generally accompanied by an inflammatory response in the host, the extent of which varies considerably with the specific poxvirus and host species. Regarding ALVAC, a poxvirus derived from the canarypox vaccine strain, Kanapox, and which represents a promising immunization vehicle in humans, nothing is known about its inflammatory capacity. The present study was aimed at documenting this issue in rodents, including mice and guinea pigs. It was then attempted to evaluate how such properties could influence the immunogenicity of an antigen concomitantly administered with ALVAC preparations using the HIV envelope subunit, rgp160, as the model immunogen. The results revealed that ALVAC, either infectious or heat-inactivated, induced in both animal species an early inflammatory response, as evidenced by a rapid migration of neutrophils to the site of inoculation. In parallel, the canarypoxvirus was shown to strongly adjuvant the co-administered immunogen, resulting in a marked increase in Env-specific IgG, IgG1 and particularly IgG2(a) serum titers. Of further interest, the heat-inactivated preparation of ALVAC retained this immunostimulatory activity. Whether or not a link between the inflammatory and immunomodulatory properties of ALVAC exists remains to be established, but such features are clearly interesting with respect to the potential use of ALVAC as an immunization vehicle.


Asunto(s)
Avipoxvirus/inmunología , Cobayas , Anticuerpos Anti-VIH/sangre , Proteínas gp160 de Envoltorio del VIH/inmunología , Animales , Pollos , Femenino , Inmunoglobulina G/sangre , Masculino , Ratones , Neutrófilos/fisiología , Proteínas Recombinantes/inmunología
13.
J Immunol ; 167(3): 1795-802, 2001 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-11466405

RESUMEN

We have investigated the possible usefulness of recombinant canarypox virus (ALVAC) encoding the melanoma-associated Ag, Melan-A/MART-1 (MART-1), in cancer immunotherapy, using a dendritic cell (DC)-based approach. ALVAC MART-1-infected DC express, and are able to process and present, the Ag coded by the viral vector. One consistent feature of infection by ALVAC is that these viruses induce apoptosis, and we show cross-presentation of Ag when uninfected DC are cocultured with ALVAC MART-1-infected DC. Uptake of apoptotic virally infected DC by uninfected DC and subsequent expression of tumor Ag in the latter were verified by flow cytometry analysis, image cytometry, and confocal microscopy. Functional activity was monitored in vitro by the stimulation of a MART-1-specific cytotoxic T cell clone. Heightened efficiency in Ag presentation is evidenced in the 2- to 3-fold increase in IFN-gamma production by the T cell clone, as compared with the ALVAC-infected DC alone. Cocultures of ALVAC MART-1-infected and uninfected DC are able to induce MART-1-specific T cell immune responses, as assessed by HLA class I/peptide tetramer binding, IFN-gamma ELISPOT assays, and cytotoxicity tests. Overall, our data indicate that DC infected with recombinant canarypox viruses may represent an efficient presentation platform for tumor Ags, which can be exploited in clinical studies.


Asunto(s)
Presentación de Antígeno/genética , Antígenos de Neoplasias/metabolismo , Apoptosis/genética , Avipoxvirus/genética , Células Dendríticas/inmunología , Células Dendríticas/virología , Proteínas de Neoplasias/inmunología , Vacunas Virales/genética , Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/genética , Apoptosis/inmunología , Avipoxvirus/inmunología , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Cultivadas , Células Clonales , Técnicas de Cocultivo , Citotoxicidad Inmunológica/genética , Células Dendríticas/metabolismo , Vectores Genéticos/inmunología , Humanos , Leucocitos Mononucleares/inmunología , Activación de Linfocitos/genética , Antígeno MART-1 , Melanoma/genética , Melanoma/inmunología , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fagocitosis/genética , Células Tumorales Cultivadas , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Virales/inmunología
14.
J Virol ; 75(13): 5879-90, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11390589

RESUMEN

The ability to generate antibodies that cross-neutralize diverse primary isolates is an important goal for human immunodeficiency virus type 1 (HIV-1) vaccine development. Most of the candidate HIV-1 vaccines tested in humans and nonhuman primates have failed in this regard. Past efforts have focused almost entirely on the envelope glycoproteins of a small number of T-cell line-adapted strains of the virus as immunogens. Here we assessed the immunogenicity of noninfectious virus-like particles (VLP) consisting of Gag, Pro (protease), and Env from R5 primary isolate HIV-1(Bx08). Immunogens were delivered to rhesus macaques in the form of either purified VLP, recombinant DNA and canarypox (ALVAC) vectors engineered to express VLP, or a combination of these products. Seroconversion to Gag and Pro was detected in all of the immunized animals. Antibodies that could neutralize HIV-1(Bx08) were detected in animals that received (i) coinoculations with DNA(Bx08) and VLP(Bx08), (ii) DNA(Bx08) followed by ALVAC(Bx08) boosting, and (iii) VLP(Bx08) alone. The neutralizing antibodies were highly strain specific despite the fact that they did not appear to be directed to linear epitopes in the V3 loop. Virus-specific cellular immune responses also were generated, as judged by the presence of Gag-specific gamma interferon (IFN-gamma)-producing cells. These cellular immune responses required the inclusion of DNA(Bx08) in the immunization modality, since few or no IFN-gamma-producing cells were detected in animals that received either VLP(Bx08) or ALVAC(Bx08) alone. The results demonstrate the feasibility of generating neutralizing antibodies and cellular immune responses that target an R5 primary HIV-1 isolate by vaccination in primates.


Asunto(s)
Productos del Gen gag/inmunología , Anticuerpos Anti-VIH/biosíntesis , VIH-1/inmunología , Animales , Reacciones Cruzadas , Seropositividad para VIH , Humanos , Inmunización , Interferón gamma/metabolismo , Macaca mulatta , Pruebas de Neutralización
15.
J Virol ; 75(13): 5939-48, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11390595

RESUMEN

We assessed the immunogenicities and efficacies of two highly attenuated vaccinia virus-derived NYVAC vaccine candidates encoding the human T-cell leukemia/lymphoma virus type 1 (HTLV-1) env gene or both the env and gag genes in prime-boost pilot regimens in combination with naked DNA expressing the HTLV-1 envelope. Three inoculations of NYVAC HTLV-1 env at 0, 1, and 3 months followed by a single inoculation of DNA env at 9 months protected against intravenous challenge with HTLV-1-infected cells in one of three immunized squirrel monkeys. Furthermore, humoral and cell-mediated immune responses against HTLV-1 Env could be detected in this protected animal. However, priming the animal with a single dose of env DNA, followed by immunization with the NYVAC HTLV-1 gag and env vaccine at 6, 7, and 8 months, protected all three animals against challenge with HTLV-1-infected cells. With this protocol, antibodies against HTLV-1 Env and cell-mediated responses against Env and Gag could also be detected in the protected animals. Although the relative superiority of a DNA prime-NYVAC boost regimen over addition of the Gag component as an immunogen cannot be assessed directly, our findings nevertheless show that an HTLV-1 vaccine approach is feasible and deserves further study.


Asunto(s)
Productos del Gen env/inmunología , Productos del Gen gag/inmunología , Virus Linfotrópico T Tipo 1 Humano/inmunología , Vacunas de ADN/inmunología , Vacunas Sintéticas/inmunología , Vacunas Virales/inmunología , Animales , Inmunización , Masculino , Saimiri
16.
Cancer Gene Ther ; 8(2): 87-98, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11263530

RESUMEN

The antitumor activity of a recombinant canarypox virus expressing wild type murine p53 (ALVAC-p53) was investigated in two murine syngeneic tumors harboring an endogenous p53 mutation (CMS4 and TS/A). Direct intratumor injections of ALVAC-p53 in CMS4 pre-established subcutaneous tumors induced total tumor regression in 66% of mice. Furthermore, 100% of the cured mice was protected against a contralateral subsequent challenge with the parental tumor cells. The intravenous treatment of experimental lung metastasis by ALVAC-p53 also induced significant tumor growth inhibition in both models. The antitumor effect of ALVAC-p53 was only observed in immunocompetent animals and was associated with the generation of a specific antitumor immune response. ALVAC-p53 induced the expression of a functional p53 wild type protein as demonstrated by up-regulation of p21waf1 and induction of apoptosis. A vaccine strategy using intravenous or subcutaneous ALVAC-p53/NYVAC-p53 prime boost protocol failed to induce CTL against p53 wild type used as target tumor antigen, and failed to protect mice against challenge with the mutated tumor cells. The mechanism of the curative and protective effects observed after direct intratumor injections results from the induction of a specific antitumor response directed against other antigens than p53. Our results suggest that the local induction of tumor apoptosis, combined with the adjuvant effect of ALVAC vector, enhances the immunogenicity of the intratumor environment and allows induction of specific antitumor immune response.


Asunto(s)
Avipoxvirus/genética , Genes p53/genética , Terapia Genética , Neoplasias Pulmonares/terapia , Neoplasias Cutáneas/terapia , Proteína p53 Supresora de Tumor/metabolismo , Animales , Anticuerpos Antineoplásicos/biosíntesis , Femenino , Expresión Génica , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Inyecciones Subcutáneas , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mutación , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/metabolismo , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/patología , Linfocitos T Citotóxicos/inmunología , Células Tumorales Cultivadas
17.
Vaccine ; 19(17-19): 2571-5, 2001 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-11257394

RESUMEN

Our overall strategy is to develop multivalent recombinant vaccines capable of eliciting broad immune responses in patients with malignant melanoma or colorectal cancer. We report herein results from initial studies conducted in cancer patients to evaluate the effect of intratumoral administration of recombinant canarypox viruses carrying cytokine genes. Our current focus is on the induction of tumor-specific T-cell responses using a prime/boost immunization schedule with a unique vector system derived from the canary pox virus called ALVAC, in which we incorporate genes encoding Tumor Associated Antigens (TAAs) of interest. Clinical studies in colorectal cancer evaluating an ALVAC CEA candidate vaccine have shown that this approach is safe and can induce tumor-specific T cell responses. Additional clinical studies evaluating candidate vaccines against melanoma and colorectal cancer, targeting either the gp100, Mage 1, Mage 3 or p53 molecules are ongoing.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Neoplasias Colorrectales/terapia , Melanoma/terapia , Antígenos de Neoplasias/genética , Avipoxvirus/genética , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Ensayos Clínicos como Asunto , Neoplasias Colorrectales/inmunología , Citocinas/genética , Vectores Genéticos , Humanos , Melanoma/inmunología , Seguridad , Linfocitos T/inmunología , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/uso terapéutico
18.
J Urol ; 165(2): 667-71, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11176455

RESUMEN

PURPOSE: In pre-clinical gene therapy studies of bladder cancer there is tremendous variation in the ability of viral vectors to deliver genetic material to bladder epithelium. Possible explanations for this variability may involve the physical parameters of delivering vectors in these experimental models. We examined the effects of intravesical volume and pressure during instillation as well as chemical modification of the bladder epithelium on subsequent gene expression in the bladder in mice. MATERIALS AND METHODS: Female C57B1/6 mice underwent intravesical instillation of the replication restricted canarypox virus (ALVAC) recombinant for the reporter genes luciferase or beta-galactosidase. Similar viral titers were instilled at different volumes and a pressure transducer measured intravesical pressure when the vector was instilled. Also, various agents, including 0.6 N hydrochloric acid, 0.4% oxychlorosene, poly-L-lysine and 0.25 M. ammonium chloride, were used to modify the bladder surface before vector instillation and then assayed for transgene expression. RESULTS: As expected, maximum intravesical pressure measured during instillation was significantly greater in mice instilled with a higher volume (33.1 versus 9.8 mm. Hg). Significantly more gene expression was detected in bladders instilled with a higher volume of viral vectors (p <0.05). Likewise, higher instillation pressures resulted in higher transgene expression in distant organs. Modification of the bladder epithelium with agents such as oxychlorosene and poly-L-lysine resulted in elevated gene expression with only minimal increases in systemic activity. CONCLUSIONS: Significant differences in gene expression are achieved by varying physical parameters during intravesical instillation. Increased gene expression associated with larger volume instillation may be responsible for some reported variability of gene transfer to the bladder. Alternate manipulations, such as modifying the bladder surface, may be done to enhance gene transfer to the urothelium without increasing systemic distribution.


Asunto(s)
Técnicas de Transferencia de Gen , Vejiga Urinaria , Animales , Avipoxvirus , Expresión Génica , Ratones , Ratones Endogámicos C57BL , Presión , Vejiga Urinaria/fisiología , Urotelio
19.
Vaccine ; 19(13-14): 1661-70, 2001 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-11166889

RESUMEN

p53 is over-expressed in approximately 50% of human cancers, and transfer of cytotoxic T lymphocytes (CTL) against wild-type p53 protects mice against p53-over-expressing tumors, suggesting that p53 might be an attractive target for immunotherapy. Immunization of mice with a recombinant canarypox virus, ALVAC, expressing human wild-type p53 (vCP207) prevented growth of p53-over-expressing tumors. Since intravenous administration induced better immune responses in mice than other routes, we have proposed to use this route in cancer patients. However, because this vector has never been administered intravenously to humans, and because of the possibility of inducing auto-immunity to a self-antigen, we felt it was necessary to first evaluate safety in rhesus macaques. We found that three intravenous administrations of vCP207 at proportional doses up to 10x those proposed for humans produced no abnormalities in hematologic or clinical chemistry parameters. Serologic markers of autoimmunity and inflammation were unaffected, despite the >95% amino acid identity between human and rhesus p53. Pathological examination of numerous tissues yielded findings comparable to those in animals given placebo. Some animals showed anti-p53 antibody responses following vaccination, indicating that tolerance could be broken to some extent. However, with the exception of one animal with a possible delayed type hypersensitivity reaction to p53 protein, we did not see evidence for a cell-mediated response. The safety profile in monkeys with ALVAC-p53 provides encouragement for using such live, modified vectors via the intravenous route for human immunotherapy.


Asunto(s)
Avipoxvirus/genética , Macaca mulatta/inmunología , Proteína p53 Supresora de Tumor/efectos adversos , Proteína p53 Supresora de Tumor/inmunología , Vacunas Sintéticas/efectos adversos , Vacunas Sintéticas/inmunología , Animales , Anticuerpos/inmunología , Autoinmunidad/inmunología , Temperatura Corporal , Peso Corporal , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Hipersensibilidad Tardía/inmunología , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/patología , Inyecciones Intravenosas , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/patología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Masculino , Piel/efectos de los fármacos , Piel/inmunología , Piel/patología , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/patología , Linfocitos T Citotóxicos/inmunología , Proteína p53 Supresora de Tumor/administración & dosificación , Proteína p53 Supresora de Tumor/genética , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética
20.
Virology ; 291(2): 272-84, 2001 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-11878896

RESUMEN

Poxviruses that are attenuated for growth in human cells provide a safe means of HIV antigen expression and are capable of eliciting HIV-specific immune responses, including CD8+ cytotoxic T-lymphocyte (CTL) responses. HIV-1 antigen expression in human cells by attenuated poxvirus vectors may be limited by interferon-mediated host defense mechanisms. To enhance HIV antigen expression in human cells, the vaccinia virus E3L and K3L genes were inserted into a canarypox vector that expresses HIV-1 Gag, Env, and a Nef/Pol polyepitope string. E3L and K3L markedly reduced the activation of the double-stranded RNA-dependent protein kinase, PKR, and led to a significant reduction in apoptosis in HeLa cells. Production and release of HIV-1 antigen in the form of pseudovirions was enhanced in both duration and magnitude by this vector modification. The addition of immunomodulatory genes to attenuated poxviruses represents a novel strategy for enhancing antigen production by live vector HIV vaccine candidates.


Asunto(s)
Apoptosis , Virus de la Viruela de los Canarios/genética , Expresión Génica , Vectores Genéticos/genética , VIH-1/fisiología , Proteínas de Unión al ARN/genética , Virus Vaccinia/genética , Proteínas Virales/genética , Ensamble de Virus , Genes Virales , Genes env , Genes gag , Genes nef , Genes pol , VIH-1/genética , Células HeLa , Humanos , Microscopía Electrónica , Fosforilación , Recombinación Genética , Virión/fisiología , eIF-2 Quinasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...