Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Artículo en Alemán | MEDLINE | ID: mdl-26998822

RESUMEN

Paramunity inducers have been used to treat small animals for decades. Paramunity inducers are based on attenuated and inactivated poxviruses (avipox virus and parapox virus). Their applications include both therapeutic and prophylactic use in various diseases. Despite their wide and variable use, only a very small number of placebo-controlled studies has been published. Positive effects in preventing kitten mortality and in treating feline stomatitis have been reported, however, no statistically significant effect of their therapeutic use in canine parvovirus infection, feline leukemia infection virus infection or canine papillomavirus infection could be demonstrated. For these infectious diseases, paramunity inducers do not appear to be effective.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Productos Biológicos/uso terapéutico , Enfermedades de los Gatos/terapia , Enfermedades de los Perros/terapia , Poxviridae/inmunología , Animales , Avipoxvirus/inmunología , Gatos , Perros , Vacunas Atenuadas/uso terapéutico , Vacunas de Productos Inactivados/uso terapéutico
2.
Anticancer Agents Med Chem ; 14(2): 183-9, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24237219

RESUMEN

Poxvirus-based vaccines have a long record of efficacy as both anti-tumour agents and vectors for gene therapy in different human tumour models. Interestingly, several studies of these vaccines have now entered the clinical evaluation phase for safety and effectiveness. A desirable outcome of antigen specific cancer immunotherapy is the disruption of host self-tolerance against endogenous tumour-associated antigens (TAAs). Nonetheless, recent studies have found reductions in vaccine efficacy due to host anti-vaccine immune reactions. Thus, newer approaches bringing together poxvirus-based vaccination and immunostimulation are being developed, and new poxvirus strains are being examined in tumour therapy studies. Our review summarizes the current knowledge on the efficacy of poxvirus-based vaccination on human tumours, with a particular focus on approaches aimed at increasing innate and specific immune responses. Special attention will be devoted to the new poxvirus strains that are currently under consideration for tumour therapy; the current knowledge on clinical trials and outcomes will also be reviewed.


Asunto(s)
Avipoxvirus/inmunología , Vacunas contra el Cáncer/uso terapéutico , Citocinas/metabolismo , Inmunoterapia/métodos , Neoplasias/terapia , Inmunidad Adaptativa , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Avipoxvirus/genética , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Ensayos Clínicos como Asunto , Citocinas/genética , Citocinas/inmunología , Vectores Genéticos , Humanos , Inmunidad Innata , Inmunización , Factores Inmunológicos/uso terapéutico , Neoplasias/inmunología , Microambiente Tumoral/inmunología , Vacunas Atenuadas
3.
J Avian Med Surg ; 21(1): 50-5, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18069171

RESUMEN

A natural outbreak of avipoxvirus occurred in recently purchased stone curlews (Burhinus oedicnemus) at a breeding farm and subsequently spread to other stone curlews residing at the farm. The initial outbreak was characterized by mild vesicular skin lesions on the legs, which then developed crusts and bled. The overall morbidity rate was 100%, but none of the birds died, and all recovered without complication. Four gallinaceous species, also kept on the farm, did not develop lesions. Avipoxvirus was identified from the skin lesions by virus isolation, electron microscopy, and monoclonal antibody testing, as well as by polymerase chain reaction testing. Eight months after this outbreak, 7 male stone curlews developed large, round, crusty lesions on their legs. Although poxvirus virions were identified in the lesions, results of virus isolation were negative. These lesions possibly were the result of a recrudescence of the original infection in male birds that were stressed because they were housed together during the breeding season. This is the first clinical description of an avipoxvirus infection in stone curlews.


Asunto(s)
Avipoxvirus/aislamiento & purificación , Enfermedades de las Aves/epidemiología , Charadriiformes , Brotes de Enfermedades/veterinaria , Infecciones por Poxviridae/veterinaria , Animales , Avipoxvirus/genética , Avipoxvirus/inmunología , Avipoxvirus/ultraestructura , Enfermedades de las Aves/etiología , Enfermedades de las Aves/virología , Cartilla de ADN , ADN Viral/análisis , Femenino , Masculino , Microscopía Electrónica/veterinaria , Reacción en Cadena de la Polimerasa/veterinaria , Infecciones por Poxviridae/epidemiología , Emiratos Árabes Unidos/epidemiología
4.
Expert Rev Vaccines ; 3(4 Suppl): S75-88, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15285707

RESUMEN

Poxvirus vectors have emerged as important vectors for licensed veterinary vaccines and candidate vaccines for humans. Vaccinia, highly-attenuated vaccinia strains and avipoxviruses have been assessed extensively in preclinical models, as well as in humans, to determine their immunogenicity and protective efficacy against HIV. The attenuated vaccinia strains and avipoxviruses have been shown to be safe and able to carry HIV genes and express their proteins to induce both antibodies and cellular immune responses. Preclinical studies show protection against HIV challenge. When using a live attenuated vector system, one must be cognizant of the potential for immune dampening because of vector-specific immunity. In this regard, avipoxviruses, such as canarypox, appear free of the inhibitory effects of vector immunity and repeated use. Unlike vaccinia-based vectors derived from classical vaccine strains, NYVAC and modified vaccinia Ankara may be less susceptible to this effect. In the coming 5 to 10 years, we will certainly know whether this class of vaccine candidates, either alone or in a prime-boost format with other vectors or proteins, will contribute to HIV disease management either from a preventive or therapeutic perspective. Additional Phase I and II studies, as well as human efficacy trials will provide new information. Furthermore, it is hoped that this body of data will contribute to a better understanding of the relevance of specific immunogenicity end points to protection and the predictive value of available animal models in HIV vaccine development.


Asunto(s)
Vacunas contra el SIDA/administración & dosificación , Avipoxvirus/genética , Infecciones por VIH/inmunología , VIH/genética , Vacunas contra el SIDA/inmunología , Animales , Avipoxvirus/inmunología , Modelos Animales de Enfermedad , VIH/inmunología , Infecciones por VIH/genética , Infecciones por VIH/prevención & control , VIH-1/genética , VIH-1/inmunología , VIH-2/genética , VIH-2/inmunología , Humanos , Virus Reordenados , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/inmunología
5.
Vaccine ; 22(5-6): 597-606, 2004 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-14741150

RESUMEN

The ability of ALVAC- or fowlpox-based simian immunodeficiency virus (SIV) vaccines to boost SIV-specific CD4+ and CD8+ T-cell responses was tested in 10 vaccinia-experienced macaques infected with SIVmac251. The CD8+ T-cell response to the dominant Gag(181-189) CM9 was quantitated in seven Mamu-A*01-positive macaques by tetramer staining, by ex vivo cytotoxic T-lymphocyte (CTL) activity, and by intracellular cytokine staining (ICS) with the specific Gag(181-189) CM9 peptide. The overall CD8+ T-cell response to Gag was assessed using a peptide pool encompassing the entire Gag protein followed by measurement of TNF-alpha production in ICS assay. Similarly, virus-specific CD4+ T-cell responses were measured by ICS for TNF-alpha following stimulation with the Gag-overlapping peptide and by proliferative response following stimulation with purified p27 Gag. The two vaccine modalities effectively boosted both CD4+ and CD8+ SIV-specific T-cell response despite prior exposure to the vaccinia-derivative NYVAC vector, suggesting that sequential boosting with either avipox-based vector vaccine candidate is a realistic approach in immune therapy of human immunodeficiency virus type 1 (HIV-1)-infected individuals.


Asunto(s)
Avipoxvirus/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunidad Celular/fisiología , Vacunas contra el SIDAS/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Vaccinia/inmunología , Animales , Anticuerpos Antivirales/biosíntesis , Citocinas/biosíntesis , Productos del Gen gag/biosíntesis , Productos del Gen gag/genética , Productos del Gen pol/biosíntesis , Productos del Gen pol/genética , Genotipo , Esquemas de Inmunización , Recuento de Linfocitos , Macaca mulatta , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , ARN Viral/análisis , ARN Viral/biosíntesis , Linfocitos T Citotóxicos/inmunología
6.
Infect Immun ; 69(9): 5565-72, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11500430

RESUMEN

A nonhuman primate model for malaria vaccine development allowing reliable, stringent sporozoite challenge and evaluation of both cellular and antibody responses is needed. We therefore constructed a multicomponent, multistage DNA vaccine for the simian malaria species Plasmodium knowlesi including two preerythrocytic-stage antigens, the circumsporozoite protein (PkCSP) and sporozoite surface protein 2 (PkSSP2), and two blood stage antigens, apical merozoite antigen 1 (PkAMA1) and merozoite surface protein 1 (PkMSP1p42), as well as recombinant canarypox viruses encoding the four antigens (ALVAC-4). The DNA vaccine plasmids expressed the corresponding antigens in vitro and induced antiparasite antibodies in mice. Groups of four rhesus monkeys received three doses of a mixture of the four DNA vaccine plasmids and a plasmid encoding rhesus granulocyte-monocyte colony-stimulating factor, followed by boosting with a single dose of ALVAC-4. Three groups received the priming DNA doses by different routes, either by intramuscular needle injection, by intramuscular injection with a needleless injection device, the Biojector, or by a combination of intramuscular and intradermal routes by Biojector. Animals immunized by any route developed antibody responses against sporozoites and infected erythrocytes and against a recombinant PkCSP protein, as well as gamma interferon-secreting T-cell responses against peptides from PkCSP. Following challenge with 100 P. knowlesi sporozoites, 1 of 12 experimental monkeys was completely protected and the mean parasitemia in the remaining monkeys was significantly lower than that in 4 control monkeys. This model will be important in preclinical vaccine development.


Asunto(s)
Antígenos de Protozoos/inmunología , Avipoxvirus/genética , Vacunas contra la Malaria , Malaria/prevención & control , Plasmodium knowlesi/inmunología , Vacunas de ADN , Animales , Anticuerpos Antiprotozoarios/sangre , Antígenos de Protozoos/genética , Antígenos de Protozoos/metabolismo , Avipoxvirus/inmunología , Inmunización Secundaria/métodos , Interferón gamma/biosíntesis , Macaca mulatta , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/inmunología , Ratones , Parasitemia/prevención & control , Plásmidos/genética , Células Tumorales Cultivadas , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología
7.
J Infect Dis ; 184(5): 525-32, 2001 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-11474428

RESUMEN

Induction of human gammadelta T cells was investigated in subjects who were vaccinated with live recombinant canarypox virus expressing human immunodeficiency virus (HIV) proteins or soluble MN rgp120. Both canarypox and rgp120 induced antigen-specific lymphoproliferative and interferon (IFN)-gamma responses. However, only canarypox vaccination induced increased gammadelta T cell responses detectable after secondary in vitro expansion (P<.02). These enhanced gammadelta T cell responses were specific for canarypox but not HIV antigens. Canarypox-specific gammadelta T cells were predominantly Vgamma9(+) and produced intracellular and secreted IFN-gamma. gammadelta T cell lines generated from canarypox vaccinees responded to canarypox antigens but not to mycobacterial antigens shown previously to induce bacille Calmette-Guérin-specific gammadelta T cells. Furthermore, canarypox vaccinations were associated with significantly higher NK cell expansions (P=.02). Increased IFN-gamma production by gammadelta T and NK cells could enhance the induction of protective type 1 memory immunity. Thus, stimulation of gammadelta T cells might be an important feature of live vaccines.


Asunto(s)
Vacunas contra el SIDA/inmunología , Antígenos/inmunología , Avipoxvirus/inmunología , Interferón gamma/biosíntesis , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/inmunología , Vacunas contra el SIDA/genética , Avipoxvirus/genética , Proteína gp120 de Envoltorio del VIH/genética , Proteína gp120 de Envoltorio del VIH/inmunología , Humanos , Células Asesinas Naturales , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/inmunología , Activación de Linfocitos , Mycobacterium tuberculosis/inmunología , Proteínas Recombinantes/inmunología , Vacunación
8.
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
9.
J Infect Dis ; 183(8): 1171-9, 2001 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-11262198

RESUMEN

The major matrix phosphoprotein 65 (pp65) of cytomegalovirus (CMV) is an important target of HLA-restricted cytotoxic T cells (CTL) after natural infection. A canarypox-CMV pp65 recombinant was studied for its ability to induce CMV pp65-specific CTL, helper T lymphocytes, and antibodies in a phase I clinical trial. Twenty-one CMV-seronegative adult volunteers were randomized to receive immunizations at months 0, 1, 3, and 6 with either canarypox-CMV pp65 or placebo. In canarypox-CMV pp65-immunized subjects, pp65-specific CTL were elicited after only 2 vaccinations and were present at months 12 and 26 in all subjects tested. Cell-depletion studies indicated that the CTL were phenotype CD8(+). Peripheral blood mononuclear cells proliferated in response to stimulation with purified pp65, and antibodies specific for pp65 also were detected. Canarypox-CMV pp65 is the first recombinant vaccine to elicit CMV-specific CTL responses, which suggests the potential usefulness of this approach in preventing disease caused by CMV.


Asunto(s)
Anticuerpos Antivirales/sangre , Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Fosfoproteínas/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Vacunas Sintéticas/efectos adversos , Proteínas de la Matriz Viral/inmunología , Vacunas Virales/efectos adversos , Adolescente , Adulto , Formación de Anticuerpos , Especificidad de Anticuerpos , Avipoxvirus/genética , Avipoxvirus/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular , Citotoxicidad Inmunológica , Vectores Genéticos , Humanos , Inmunidad Celular , Fosfoproteínas/genética , Proteínas de la Matriz Viral/genética
10.
Hepatology ; 33(2): 448-54, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11172348

RESUMEN

There are about 200 million chronic hepatitis B virus (HBV) carriers at high risk of development of cirrhosis and hepatocellular carcinoma. Termination of the carrier state may avert these risks. We have investigated immunotherapy for chronic HBV infection in a chimpanzee HBV carrier using recombinant DNA-based immunization followed by a recombinant canarypox booster. One week after the booster, HBV DNA declined greater than 400-fold and remained undetectable by the quantitative polymerase chain reaction (PCR) assay for 186 weeks. Plasma levels of hepatitis B surface antigen (HBsAg) declined for only a short time. The decline in HBV DNA correlated with a boost in gamma interferon production without a corresponding boost in cytotoxic T lymphocyte levels, and decline in the transcriptional template or covalently closed circular DNA level. Confirmation of these findings requires further studies in chimpanzees and/or in humans.


Asunto(s)
Avipoxvirus/inmunología , ADN/uso terapéutico , Antígenos de Superficie de la Hepatitis B/genética , Hepatitis B Crónica/terapia , Inmunización Secundaria , Inmunoterapia , Alanina Transaminasa/sangre , Animales , Citocinas/genética , ADN/genética , ADN Complementario/sangre , ADN Viral/sangre , Antígenos de Superficie de la Hepatitis B/análisis , Antígenos e de la Hepatitis B/sangre , Virus de la Hepatitis B/genética , Hepatitis B Crónica/sangre , Hepatitis B Crónica/inmunología , Hepatitis B Crónica/virología , Inmunidad Celular , Inmunización , Interferón gamma/metabolismo , Hígado/metabolismo , Pan troglodytes , ARN Mensajero/metabolismo , Recombinación Genética
11.
Cancer Immunol Immunother ; 49(10): 517-29, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11129322

RESUMEN

An enzyme-linked immunosorbent spot (ELISPOT) assay for interferon gamma production has been used to analyze specific T cell responses to a Flu 9-mer peptide, and a 9-mer peptide of carcinoembryonic antigen (CEA). Assays were performed on peripheral blood mononuclear cells (PBMC) of HLA-A2-positive patients with CEA-expressing carcinomas, both before and after vaccination with CEA-based vaccines, and from HLA-A2-positive healthy blood donors. The ELISPOT assay utilized aliquots of frozen PBMC, and assays were performed after 24 h in culture with peptide to rule out any artifacts due to long-term in vitro stimulation cycles. An internal standard was used for each assay to define reproducibility of the assay, and all samples from a given patient (pre- and post-vaccination, with both the Flu and CEA peptides) were analyzed simultaneously. The results indicated a trend towards healthy blood donors having higher levels of Flu-specific T cell precursors than do colon carcinoma patients, but these results were not statistically significant (P = 0.06). On the other hand, slightly higher CEA-specific T cell responses were observed in cancer patients with CEA-expressing carcinomas than in healthy blood donors. PBMC from two CEA-based vaccine clinical trials were analyzed for T cell responses to the same CEA peptide and to the Flu control peptide. The first trial consisted of three monthly vaccinations of CEA peptide (designated PPP) in adjuvant. The second trial consisted of cohorts receiving three monthly vaccinations of avipox-CEA recombinant (designated AAA) or cohorts receiving a primary vaccination with recombinant vaccinia-CEA followed by two monthly vaccinations with avipox-CEA (designated VAA). Few, if any, CEA-specific T cell responses were seen in the PPP vaccinations, while the majority of patients receiving the poxvirus CEA recombinants demonstrated increases in CEA-specific T cell responses and no increases in Flu-specific responses. CEA-specific IgG responses were also demonstrated in patients following recombinant CEA poxvirus vaccinations. Statistical analyses of the T cell responses to the same CEA peptide demonstrated a P value of 0.028 for the recombinant poxvirus vaccines, as compared with the peptide vaccine. There were no differences seen (P = 0.37) in Flu-specific responses after these two types of CEA vaccination. These results thus provide the first evidence that poxvirus recombinant-based vaccines are more potent in the initiation of tumor-antigen-specific T cell responses than vaccines employing peptide in adjuvant, when assays are conducted in an identical manner, and in defining responses to the same peptide. These results also demonstrate for the first time that an ELISPOT assay, performed over a 24-h period and without in vitro sensitization, can be successfully used to monitor immune responses to a tumor-associated antigen in cancer patients.


Asunto(s)
Antígenos de Neoplasias/inmunología , Avipoxvirus/inmunología , Vacunas contra el Cáncer/uso terapéutico , Antígeno Carcinoembrionario/inmunología , Carcinoma/terapia , Ensayo de Inmunoadsorción Enzimática , Inmunoterapia Activa , Interferón gamma/biosíntesis , Fragmentos de Péptidos/inmunología , Subgrupos de Linfocitos T/metabolismo , Vacunas Sintéticas/uso terapéutico , Vacunas Virales/uso terapéutico , Adyuvantes Inmunológicos , Anticuerpos Antineoplásicos/biosíntesis , Anticuerpos Antivirales/biosíntesis , Células Presentadoras de Antígenos/inmunología , Antígenos de Neoplasias/genética , Antígenos Virales/genética , Antígenos Virales/inmunología , Donantes de Sangre , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/terapia , Vacunas contra el Cáncer/inmunología , Antígeno Carcinoembrionario/genética , Carcinoma/inmunología , Línea Celular , Estudios de Cohortes , Citotoxicidad Inmunológica , Neoplasias del Sistema Digestivo/inmunología , Neoplasias del Sistema Digestivo/terapia , Genes Sintéticos , Antígeno HLA-A2/análisis , Humanos , Inmunoglobulina G/biosíntesis , Interferón gamma/análisis , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Activación de Linfocitos , Fragmentos de Péptidos/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Subgrupos de Linfocitos T/inmunología , Vacunas Sintéticas/inmunología , Vacunas Virales/inmunología
12.
J Clin Oncol ; 18(23): 3964-73, 2000 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-11099326

RESUMEN

PURPOSE: This trial sought to determine, for the first time, the validity in human vaccinations of using two different recombinant vaccines in diversified prime-and-boost regimens to enhance T-cell responses to a tumor antigen. PATIENTS AND METHODS: Eighteen patients with advanced tumors expressing carcinoembryonic antigen (CEA) were randomized to receive either recombinant vaccinia (rV)-CEA followed by three avipox-CEA vaccinations, or avipox-CEA (three times) followed by one rV-CEA vaccination. Subsequent vaccinations in both cohorts were with avipox-CEA. Immunologic monitoring was performed using a CEA peptide and the enzyme-linked immunospot assay for interferon gamma production. RESULTS: rV-CEA followed by avipox-CEA was superior to the reverse order in the generation of CEA-specific T-cell responses. Further increases in CEA-specific T-cell precursors were seen when local granulocyte-macrophage colony-stimulating factor (GM-CSF) and low-dose interleukin (IL)-2 were given with subsequent vaccinations. The treatment was extremely well tolerated. Limited clinical activity was seen using vaccines alone in this patient population. Antibody production against CEA was also observed in some of the treated patients. CONCLUSION: rV-CEA was more effective in its role as a primer of the immune system; avipox-CEA could be given up to eight times with continued increases in CEA T-cell precursors. Future trials should use rV-CEA first followed by avipox-CEA. Vaccines specific to CEA are able to generate CEA-specific T-cell responses in patients without significant toxicity. T-cell responses using vaccines alone may be inadequate to generate significant anticancer objective responses in patients with advanced disease. Cytokines such as GM-CSF and IL-2 may play a key role in generating such responses.


Asunto(s)
Avipoxvirus/inmunología , Vacunas contra el Cáncer/administración & dosificación , Antígeno Carcinoembrionario/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Vacunas Sintéticas/administración & dosificación , Virus Vaccinia/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Vacunas contra el Cáncer/efectos adversos , Citocinas/inmunología , Ensayo de Inmunoadsorción Enzimática , Epítopos de Linfocito T/inmunología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Antígeno HLA-A2/genética , Antígeno HLA-A2/inmunología , Humanos , Esquemas de Inmunización , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/sangre , Interleucina-2/uso terapéutico , Masculino , Persona de Mediana Edad , Linfocitos T/inmunología , Vacunas Sintéticas/efectos adversos
13.
Cancer Immunol Immunother ; 49(9): 504-14, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11092617

RESUMEN

The generation of cytotoxic effector T cells requires delivery of two signals, one derived from a specific antigenic epitope and one from a costimulatory molecule. A phase I clinical trial was conducted with a non-replicating canarypoxvirus (ALVAC) constructed to express both human carcinoembryonic antigen (CEA) and the B7.1 costimulatory molecule. This was the first study in cancer patients to determine if the delivery of costimulation with a tumor vaccine was feasible and improved immune responses. Three cohorts of six patients, each with advanced CEA-expressing adenocarcinomas, were treated with increasing doses of an ALVAC-CEA-B7.1 vaccine (4.5 x 10(6), 4.5 x 10(7), and 4.5 x 10(8) plaque-forming units, PFU). Patients were vaccinated by intramuscular injection every 4 weeks for 3 months and monitored for side-effects, tumor growth and anti-CEA immune responses. ALVAC-CEA-B7.1 at doses up to 4.5 x 10(8) PFU was given without evidence of significant toxicity or autoimmune reactions. Three patients experienced clinically stable disease that correlated with increasing CEA-specific precursor T cells, as shown by in vitro interferon-gamma enzyme-linked immunoassay spot tests (ELISPOT). These three patients underwent repeated vaccination resulting in augmented CEA-specific T cell responses. This study represents the first use of costimulation to enhance antitumor vaccines in cancer patients. This approach resulted in CEA-specific immunity associated with stable diseases in three patients. This study also demonstrated that CEA-specific T cell responses could be sustained by repeated vaccinations. Although the number of patients was small, the addition of B7.1 to virus-based vaccines may improve immunological and stable diseases to vaccination against tumor-associated antigens with tolerable toxicity.


Asunto(s)
Avipoxvirus/genética , Avipoxvirus/inmunología , Antígeno B7-1/genética , Vacunas contra el Cáncer/uso terapéutico , Antígeno Carcinoembrionario/genética , Adenocarcinoma/inmunología , Adenocarcinoma/terapia , Adulto , Anciano , Antígeno B7-1/inmunología , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/toxicidad , Antígeno Carcinoembrionario/inmunología , Línea Celular , Células Cultivadas , Estudios de Cohortes , Neoplasias del Colon/inmunología , Neoplasias del Colon/terapia , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Epítopos/metabolismo , Femenino , Citometría de Flujo , Humanos , Interferón gamma/inmunología , Interferón gamma/metabolismo , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/terapia , Péptidos/uso terapéutico , Fenotipo , Neoplasias del Recto/inmunología , Neoplasias del Recto/terapia , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/terapia , Linfocitos T/inmunología , Factores de Tiempo
14.
Hum Gene Ther ; 11(9): 1289-301, 2000 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-10890739

RESUMEN

The immunogenicity of recombinant canarypox (ALVAC) viral vectors within murine whole-cell tumor vaccines was evaluated using the T cell thymic lymphoma STF10 and the B16 melanoma. Tumor cells were modified with the recombinant ALVAC vectors and injected into syngeneic mice. Control mice receiving cells alone all developed tumors, while mice injected with tumor variants bearing parental and recombinant vectors either completely rejected their tumors, or exhibited a significant delay in tumor formation. Rechallenge of mice receiving STF10-variant vaccines yielded a protective effect against parental tumor cells only when a modified regimen incorporating two vaccinations was utilized. Notably, the parental ALVAC virus was equivalent to all other recombinant ALVAC viruses in conferring antitumor immunity when using a prime-and-boost protocol. Tumorigenicity experiments in nude mice revealed that the effector mechanism mediating rejection of tumor cells bearing ALVAC vectors is multifactorial, in that the immunogenicity of STF10/ALVAC vaccines is reduced, but not completely abolished in these mice. Finally, in vitro experiments revealed that cytotoxic T cells specific for parental STF10 cells could be generated as a result of in vivo immunization with STF10/ALVAC vaccines.


Asunto(s)
Avipoxvirus/inmunología , Vacunas contra el Cáncer/inmunología , Vectores Genéticos/inmunología , Animales , Anticuerpos Antivirales/inmunología , Avipoxvirus/genética , Antígeno B7-1/biosíntesis , Antígeno B7-1/genética , Vacunas contra el Cáncer/genética , Citotoxicidad Inmunológica , Inmunización Secundaria , Interleucina-12/biosíntesis , Interleucina-12/genética , Linfoma de Células T/genética , Linfoma de Células T/inmunología , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Linfocitos T Citotóxicos/inmunología , Transducción Genética , Células Tumorales Cultivadas
15.
J Infect Dis ; 182(1): 18-27, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10882577

RESUMEN

DNA vaccination was employed to study immune responses to hepatitis C virus (HCV) proteins. As an immunizing strategy, we studied immune responses of BALB/c (H-2d) and C57BL/6 mice (H-2b) to HCV genes delivered intramuscularly as a polycistronic construct capsid/E1/E2/NS2/NS3 (pRC/C-NS3) encoding 5 structural and nonstructural proteins. We also evaluated canarypox virus containing the same HCV genes as a means for potentiating immune responses to naked DNA. Our results indicate that mice that received a polycistronic pRC/C-NS3 with canarypox booster had enhanced antibody and cellular responses to HCV proteins. Immunodominant CD8(+) T cell responses to several HCV structural and nonstructural proteins, characterized by cytotoxicity and interferon (IFN)-gamma production or IFN-gamma production without significant cytotoxicity, were observed in both strains of mice. The combination of naked DNA with a nonreplicating canarypox booster encoding HCV polycistronic pRC/C-NS3 genes appears to diversify and enhance T cell responses to HCV proteins.


Asunto(s)
Avipoxvirus/inmunología , Hepacivirus/inmunología , Vacunas de ADN , Vacunas contra Hepatitis Viral/inmunología , Proteínas no Estructurales Virales/inmunología , Proteínas Estructurales Virales/inmunología , Animales , Formación de Anticuerpos , Avipoxvirus/genética , Cartilla de ADN , ADN Viral/inmunología , Femenino , Hepacivirus/genética , Inmunidad Celular , Inmunización Secundaria , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fenotipo , Plásmidos/genética , Plásmidos/inmunología , Linfocitos T Citotóxicos/inmunología , Transfección , Vacunas contra Hepatitis Viral/genética
16.
FEMS Immunol Med Microbiol ; 27(3): 211-8, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10683465

RESUMEN

The fine immunoreactivity of the rabbit humoral response elicited by four env-recombinant avipoxviruses and their ability to stimulate a memory T-cell response and a protective immunity have been studied. The antibody specificity was compared with the serum neutralizing activity and virus-specific T-cell proliferative response. Resistance to challenge by cell-associated HIV-1 was monitored by PCR. Canarypox (CP) and fowlpox (FP) constructs, containing the complete env gene (IS(+)) from the HIV-1(SF2) strain, induced a higher profile of epitope recognition than their counterparts expressing the env gene deleted of the putative immunosuppressive region (IS(-)). Serum neutralizing activity was in agreement with fusion inhibition and lymphoproliferative response in rabbits immunized with CPIS(+), and only partially with FPIS(+). Rabbits failed to be infected, but anti- p55 gag-specific antibodies could be demonstrated by Western blot. This study confirms the ability of these non-replicative live recombinant viruses to elicit a complete immune response, capable of inhibiting specific HIV-1 functions.


Asunto(s)
Vacunas contra el SIDA/inmunología , Anticuerpos Anti-VIH/sangre , VIH-1/inmunología , Vacunas Sintéticas/inmunología , Vacunas contra el SIDA/administración & dosificación , Animales , Avipoxvirus/genética , Avipoxvirus/inmunología , Línea Celular , Mapeo Epitopo , Virus de la Viruela de las Aves de Corral/genética , Virus de la Viruela de las Aves de Corral/inmunología , Productos del Gen env/genética , Productos del Gen env/inmunología , Productos del Gen gag/inmunología , Infecciones por VIH/inmunología , Infecciones por VIH/prevención & control , VIH-1/genética , VIH-1/fisiología , Humanos , Inmunización , Activación de Linfocitos , Pruebas de Neutralización , Conejos , Vacunas Sintéticas/administración & dosificación
17.
Am Heart J ; 138(5 Pt 2): S484-7, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10539854

RESUMEN

Congenital cytomegalovirus disease is an unsolved public health problem, unlikely to be solved by means other than immune prophylaxis. Development of a vaccine has been hampered by low awareness of the problem, which is caused by the often delayed detection of abnormalities after birth. Nevertheless, cytomegalovirus vaccine development is active. An attenuated, live vaccine has been studied extensively, and an improved strain may result from genetic manipulation. An immunogenic viral glycoprotein (gB) vaccine is currently in clinical trial to determine if antibodies alone will be protective. The idea of a combined vaccine has been proposed, in which a canarypox recombinant containing several cytomegalovirus genes is used both to generate cellular immunity and to prime for augmented antibody responses to the viral glycoprotein. Finally, DNA plasmids containing cytomegalovirus genes are being investigated for their utility as vaccines.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/prevención & control , Citomegalovirus/inmunología , Vacunas Atenuadas/uso terapéutico , Vacunas Virales/uso terapéutico , Adyuvantes Inmunológicos/uso terapéutico , Adulto , Anticuerpos Antivirales/sangre , Avipoxvirus/genética , Avipoxvirus/inmunología , Ensayos Clínicos como Asunto , Citomegalovirus/genética , Infecciones por Citomegalovirus/sangre , Humanos , Plásmidos/genética , Plásmidos/inmunología , Recombinación Genética , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología
18.
J Infect Dis ; 180(4): 1122-32, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10479139

RESUMEN

Canarypox viruses undergo abortive replication in mammalian cells. Despite this restriction on replication in mammalian cells, significant immune responses have been shown in animals and in humans receiving recombinant canarypox vaccine vectors expressing heterologous immunogens. A recombinant canarypox vaccine candidate (vCP205), which expresses human immunodeficiency virus (HIV)-1 Gag, Env, and protease proteins, is presently under investigation in phase I and phase II human trials in the United States and elsewhere. In this study, the ability of vCP205 to elicit HIV Gag-Env pseudovirion formation in avian and mammalian cells was investigated. Gag-Env pseudovirions were produced from both avian and mammalian cell lines infected by this vaccine vector. A subset of mammalian cells was identified in which pseudovirion production and release was very efficient, surpassing the production from infected avian cells. The production of Gag-Env pseudovirions by canarypox HIV vaccine vectors may have important implications for future HIV vaccine design.


Asunto(s)
Vacunas contra el SIDA , Avipoxvirus/inmunología , Genes env , Genes gag , VIH-1/fisiología , Replicación Viral , Animales , Avipoxvirus/fisiología , Avipoxvirus/ultraestructura , Línea Celular , Células Cultivadas , Embrión de Pollo , Chlorocebus aethiops , Fibroblastos , Productos del Gen env/genética , Productos del Gen gag/genética , VIH-1/genética , VIH-1/ultraestructura , Humanos , Virión/genética , Virión/fisiología , Virión/ultraestructura
19.
J Immunol ; 163(5): 2928-36, 1999 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-10453041

RESUMEN

MAGE genes are expressed by many human tumors of different histological types but not by normal cells, except for male germline cells. The Ags encoded by MAGE genes and recognized by T cells are therefore strictly tumor-specific. Clinical trials involving therapeutic vaccination of cancer patients with MAGE antigenic peptides or proteins are in progress. To increase the range of patients eligible for therapy with peptides, it is important to identify additional MAGE epitopes recognized by CTL. Candidate peptides known to bind to a given HLA have been used to stimulate T lymphocytes in vitro. In some instances, CTL clones directed against these synthetic peptides have been obtained, but these clones often failed to recognize tumor cells expressing the relevant gene. Therefore, we designed a method to identify CTL epitopes that selects naturally processed peptides. Monocyte-derived dendritic cells infected with a recombinant canarypoxvirus (ALVAC) containing the entire MAGE-A1 gene were used to stimulate CD8+ T lymphocytes from the blood of individuals without cancer. Responder cell microcultures that specifically lysed autologous cells expressing MAGE-A1 were cloned using autologous stimulator cells either transduced with a retrovirus coding for MAGE-A1 or infected with recombinant Yersinia-MAGE-A1 bacteria. The CTL clones were tested for their ability to lyse autologous cells loaded with each of a set of overlapping MAGE-A1 peptides. This strategy led to the identification of five new MAGE-A1 epitopes recognized by CTL clones on HLA-A3, -A28, -B53, -Cw2, and -Cw3 molecules. All of these CTL clones recognized target cells expressing gene MAGE-A1.


Asunto(s)
Células Dendríticas/inmunología , Epítopos de Linfocito T/metabolismo , Activación de Linfocitos , Proteínas de Neoplasias/metabolismo , Linfocitos T Citotóxicos/inmunología , Transfección/inmunología , Secuencia de Aminoácidos , Animales , Presentación de Antígeno/genética , Antígenos de Neoplasias , Avipoxvirus/genética , Avipoxvirus/inmunología , Células Cultivadas , Células Clonales , Técnicas de Cocultivo , Citotoxicidad Inmunológica , Células Dendríticas/metabolismo , Células Dendríticas/virología , Epítopos de Linfocito T/inmunología , Antígenos HLA/inmunología , Antígenos HLA/metabolismo , Antígenos HLA-A/inmunología , Antígenos HLA-A/metabolismo , Antígenos HLA-C/inmunología , Antígenos HLA-C/metabolismo , Humanos , Activación de Linfocitos/genética , Prueba de Cultivo Mixto de Linfocitos , Antígenos Específicos del Melanoma , Ratones , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Péptidos/inmunología , Péptidos/metabolismo , Linfocitos T Citotóxicos/metabolismo
20.
J Infect Dis ; 180(3): 843-6, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10438376

RESUMEN

To develop a vaccine against cytomegalovirus (CMV), a canarypox virus (ALVAC) expressing CMV glycoprotein (gB) was evaluated alone or in combination with a live, attenuated CMV vaccine (Towne). Three doses of 106.5 TCID50 of ALVAC-CMV(gB) induced very low neutralizing or ELISA antibodies in most seronegative adults. However, to determine whether ALVAC-CMV(gB) could prime for antibody responses, 20 seronegative adults randomly received either 106.8 TCID50 of ALVAC-CMV(gB) or 106.8 TCID50 of ALVAC-RG, expressing the rabies glycoprotein, administered at 0 and 1 month, with all subjects receiving a dose of 103.5 pfu of the Towne vaccine at 90 days. For subjects primed with ALVAC-CMV(gB), neutralizing titers and ELISA antibodies to CMV(gB) developed sooner, were much higher, and persisted longer than for subjects primed with ALVAC-RG. All vaccines were well tolerated. These results demonstrate that ALVAC-CMV(gB) primes the immune system and suggest a combined-vaccine strategy to induce potentially protective levels of neutralizing antibodies.


Asunto(s)
Avipoxvirus/inmunología , Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Vacunas Atenuadas/inmunología , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/inmunología , Adulto , Anticuerpos Antivirales/sangre , Formación de Anticuerpos , Avipoxvirus/genética , Citomegalovirus/genética , Infecciones por Citomegalovirus/prevención & control , Método Doble Ciego , Ensayo de Inmunoadsorción Enzimática , Femenino , Vectores Genéticos , Humanos , Masculino , Persona de Mediana Edad , Vacunas Atenuadas/efectos adversos , Vacunas Virales/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA