Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Cancer Res ; 67(14): 7037-44, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17638917

RESUMO

The interaction between CD40 ligand (CD40L, CD154) and its receptor CD40 on antigen-presenting cells is essential for the initiation of cell-mediated and humoral immune responses. Malignant B cells also express CD40 and respond to CD40L by enhancing expression of costimulatory molecules. In this study, we investigated the therapeutic antitumor effect of intratumoral administration of recombinant fowlpox virus encoding murine CD40L (rF-mCD40L) in a murine B-cell lymphoma model. BALB/c mice with established s.c. and widely metastatic A20 lymphoma tumors were treated with intratumoral injections of rF-mCD40L together with systemic chemotherapy. This combined chemoimmunotherapy resulted in complete tumor regression and long-term survival of the mice. Some tumor cells in the injected sites expressed the CD40L transgene and had increased expression of the CD80 and CD86 costimulatory molecules. The therapeutic effect was dependent on CD8 but not on CD4 T cells. Moreover, there was a requirement that the recombinant CD40L virus be injected directly into the tumor, as opposed to peritumoral or distant sites. Thus, rF-mCD40L injected directly into the tumor microenvironment enhances the immunogenicity of tumor B cells. The results support future plans for intratumoral injection of rF-mCD40L in patients with lymphoma.


Assuntos
Ligante de CD40/química , Vírus da Varíola das Aves Domésticas/genética , Linfoma/terapia , Linfoma/virologia , Animais , Antígenos CD4/imunologia , Antígenos CD8/imunologia , Sobrevivência Celular , Terapia Combinada/métodos , Imunoterapia/métodos , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Transplante de Neoplasias , Neoplasias/metabolismo , Linfócitos T/metabolismo
2.
J Clin Invest ; 115(7): 1903-12, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15937544

RESUMO

Immunotherapy for the treatment of metastatic melanoma remains a major clinical challenge. The melanoma microenvironment may lead to local T cell tolerance in part through downregulation of costimulatory molecules, such as B7.1 (CD80). We report the results from the first clinical trial, to our knowledge, using a recombinant vaccinia virus expressing B7.1 (rV-B7.1) for monthly intralesional vaccination of accessible melanoma lesions. A standard 2-dose-escalation phase I clinical trial was conducted with 12 patients. The approach was well tolerated with only low-grade fever, myalgias, and fatigue reported and 2 patients experiencing vitiligo. An objective partial response was observed in 1 patient and disease stabilization in 2 patients, 1 of whom is alive without disease 59 months following vaccination. All patients demonstrated an increase in postvaccination antibody and T cell responses against vaccinia virus. Systemic immunity was tested in HLA-A*0201 patients who demonstrated an increased frequency of gp100 and T cells specific to melanoma antigen recognized by T cells 1 (MART-1), also known as Melan-A, by ELISPOT assay following local rV-B7.1 vaccination. Local immunity was evaluated by quantitative real-time RT-PCR, which suggested that tumor regression was associated with increased expression of CD8 and IFN-gamma. The local delivery of vaccinia virus expressing B7.1 was well tolerated and represents an innovative strategy for altering the local tumor microenvironment in patients with melanoma.


Assuntos
Antígeno B7-1/genética , Terapia Genética , Imunoterapia , Melanoma/terapia , Vaccinia virus/genética , Vaccinia virus/imunologia , Adulto , Idoso , Antígenos de Neoplasias , Antígeno B7-1/uso terapêutico , Antígenos CD8/genética , Feminino , Expressão Gênica , Antígenos HLA-A , Antígeno HLA-A2 , Humanos , Injeções Intralesionais , Interferon gama/genética , Interleucina-10/genética , Antígeno MART-1 , Masculino , Melanoma/genética , Melanoma/imunologia , Melanoma/secundário , Pessoa de Meia-Idade , Proteínas de Neoplasias/imunologia , Linfócitos T/imunologia
3.
Clin Cancer Res ; 12(4): 1260-9, 2006 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-16489082

RESUMO

PURPOSE: Docetaxel has activity against androgen-independent prostate cancer and preclinical studies have shown that taxane-based chemotherapy can enhance antitumor response of vaccines. The primary objective of this study was to determine if concurrent docetaxel (with dexamethasone) had any effect on generating an immune response to the vaccine. Secondary end points were whether vaccine could be given safely with docetaxel and the clinical outcome of the treatment regimen. EXPERIMENTAL DESIGN: The vaccination regimen was composed of (a) recombinant vaccinia virus (rV) that expresses the prostate-specific antigen gene (rV-PSA) admixed with (b) rV that expresses the B7.1 costimulatory gene (rV-B7.1), and (c) sequential booster vaccinations with recombinant fowlpox virus (rF-) containing the PSA gene (rF-PSA). Patients received granulocyte macrophage colony-stimulating factor with each vaccination. Twenty-eight patients with metastatic androgen-independent prostate cancer were randomized to receive either vaccine and weekly docetaxel or vaccine alone. Patients on the vaccine alone arm were allowed to cross over to receive docetaxel alone at time of disease progression. The ELISPOT assay was used to monitor immune responses for PSA-specific T cells. RESULTS: The median increase in these T-cell precursors to PSA was 3.33-fold in both arms following 3 months of therapy. In addition, immune responses to other prostate cancer-associated tumor antigens were also detected postvaccination. Eleven patients who progressed on vaccine alone crossed over to receive docetaxel at time of progression. Median progression-free survival on docetaxel was 6.1 months after receiving vaccine compared with 3.7 months with the same regimen in a historical control. CONCLUSION: This is the first clinical trial to show that docetaxel can be administered safely with immunotherapy without inhibiting vaccine specific T-cell responses. Furthermore, patients previously vaccinated with an anticancer vaccine may respond longer to docetaxel compared with a historical control of patients receiving docetaxel alone. Larger prospective clinical studies will be required to validate these findings.


Assuntos
Vacinas Anticâncer/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Taxoides/uso terapêutico , Fosfatase Ácida/sangue , Idoso , Idoso de 80 Anos ou mais , Androgênios/fisiologia , Antígenos/sangue , Antígenos de Neoplasias , Antígenos de Superfície/sangue , Antineoplásicos Fitogênicos/uso terapêutico , Vacinas Anticâncer/imunologia , Terapia Combinada , Estudos Cross-Over , Intervalo Livre de Doença , Docetaxel , Glutamato Carboxipeptidase II/sangue , Glicoproteínas/sangue , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Masculino , Pessoa de Meia-Idade , Mucina-1 , Mucinas/sangue , Metástase Neoplásica , Antígeno Prostático Específico/sangue , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/fisiopatologia , Fatores de Tempo , Resultado do Tratamento , Vacinação/métodos
4.
Clin Cancer Res ; 12(8): 2526-37, 2006 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-16638862

RESUMO

PURPOSE: Two clinical trials were conducted to evaluate the clinical efficacy and immunologic impact of vaccination against the tyrosinase protein plus systemic interleukin 2 (IL-2) administration in patients with advanced metastatic melanoma. EXPERIMENTAL DESIGN: Full-length tyrosinase was employed as an immunogen to induce diverse immunologic responses against a commonly expressed melanoma antigen. Heterologous prime/boost vaccination with recombinant vaccinia and fowlpox vectors encoding tyrosinase was first explored in a randomized three-arm phase II trial, in which vaccines were administered alone or concurrently with low-dose or high-dose IL-2. In a subsequent single cohort phase II trial, all patients received the same vaccines and high-dose IL-2 sequentially rather than concurrently. RESULTS: Among a total of 64 patients treated on these trials, 8 objective partial responses (12.5%) were observed, all in patients receiving high-dose IL-2. Additional patients showed evidence of lesional regression (mixed tumor response) or overall regression that did not achieve partial response status (minor response). In vitro evidence of enhanced immunity against tyrosinase following protocol treatments was documented in 3 of 49 (6%) patients tested serologically, 3 of 23 (13%) patients tested for T-cell recognition of individual tyrosinase peptides, and 4 of 16 (25%) patients tested for T-cell recognition of full-length tyrosinase protein with real-time reverse transcription-PCR techniques. CONCLUSIONS: Whereas prime/boost immunization with recombinant vaccinia and fowlpox viruses enhanced antityrosinase immunity in some patients with metastatic melanoma, it was ineffective alone in mediating clinical benefit, and in combination with IL-2 did not mediate clinical benefit significantly different from that expected from treatment with IL-2 alone.


Assuntos
Vacinas Anticâncer/imunologia , DNA Recombinante/imunologia , Imunização Secundária/métodos , Interleucina-2/uso terapêutico , Melanoma/terapia , Vacinação/métodos , Terapia Combinada , DNA Recombinante/genética , Vetores Genéticos/genética , Humanos , Esquemas de Imunização , Imunoglobulina G/sangue , Interferon gama/genética , Interferon gama/metabolismo , Interleucina-2/administração & dosagem , Melanoma/imunologia , Melanoma/patologia , Monofenol Mono-Oxigenase/genética , Monofenol Mono-Oxigenase/imunologia , Monofenol Mono-Oxigenase/metabolismo , Metástase Neoplásica , Poxviridae/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Resultado do Tratamento
5.
J Clin Oncol ; 23(4): 720-31, 2005 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15613691

RESUMO

PURPOSE: Our previous clinical experience with vaccinia and replication-defective avipox recombinant carcinoembryonic antigen (CEA) vaccines has demonstrated safety and clinical activity with a correlation between CEA-specific immune response and survival. Preclinical evidence demonstrated that the addition of the transgenes for three T-cell costimulatory molecules (B7-1, ICAM-1, LFA-3, designated TRICOM) results in a significant improvement in antigen-specific T-cell responses and antitumor activity. We describe here the first trial in humans of the CEA-TRICOM vaccines (also including an enhancer agonist epitope within the CEA gene). PATIENTS AND METHODS: Fifty-eight patients with advanced CEA-expressing cancers were accrued to eight cohorts that involved vaccinations with the following: replication-defective fowlpox recombinant (rF)-CEA(6D)-TRICOM; primary vaccination with recombinant vaccinia (rV)-CEA(6D)-TRICOM plus rF-CEA(6D)-TRICOM booster vaccinations; and rV-CEA(6D)-TRICOM and then rF-CEA(6D)-TRICOM, plus granulocyte-macrophage colony-stimulating factor (GM-CSF) with vaccines, or with divided doses of vaccine with GM-CSF. Vaccines were administered every 28 days for six doses and then once every 3 months. Reverting to treatments every 28 days was allowed if patients progressed on the 3-month schedule. RESULTS: In this phase I study, no significant toxicity was observed. Twenty-three patients (40%) had stable disease for at least 4 months, with 14 of these patients having prolonged stable disease (> 6 months). Eleven patients had decreasing or stable serum CEA, and one patient had a pathologic complete response. Enhanced CEA-specific T-cell responses were observed in the majority of patients tested. CONCLUSION: We demonstrated that the CEA-TRICOM vaccines are safe and can generate significant CEA-specific immune responses, and they seem to have clinical benefit in some patients with advanced cancer.


Assuntos
Antígeno B7-1/genética , Antígenos CD58/genética , Vacinas Anticâncer/imunologia , Antígeno Carcinoembrionário/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Molécula 1 de Adesão Intercelular/genética , Neoplasias/terapia , Vacinas Sintéticas/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Vírus da Varíola das Aves Domésticas/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Humanos , Imunoglobulina G/sangue , Masculino , Pessoa de Meia-Idade , Neoplasias/imunologia , Linfócitos T/imunologia , Vacinação , Vaccinia virus/genética
6.
Front Biosci ; 11: 788-803, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16146771

RESUMO

Tumor-associated antigens (TAAs) are by definition either weakly immunogenic or functionally nonimmunogenic. Therefore, efforts have concentrated on the development of vaccine strategies in which the presentation of TAAs to the immune system results in far greater activation of T cells than that occurring naturally in the host. Several strategies are being explored in our laboratory and others to enhance the immunogenicity of TAAs. These are: (a) placing the gene coding for the tumor antigen, as a transgene, into poxvirus vectors. (b) The use of diversified prime and boost vaccine strategies employing two different types of poxvirus vectors. (c) The use of T-cell costimulation; accomplished by placing transgenes for different T-cell costimulation molecules into viral vectors along with the transgenes for the TAA. (d) Altering the amino acid sequence of the TAA to enhance the host immune response. (e) The use of cytokines, and in particular GM-CSF, as a biologic adjuvant. This review will focus on the current state of the use of costimulatory molecules as adjuvants for immunotherapy, and in particular, as immunomodulators for cancer vaccines.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Imunoterapia/métodos , Neoplasias/prevenção & controle , Animais , Antígeno B7-1/metabolismo , Antígenos CD58/metabolismo , Vacinas Anticâncer , Ensaios Clínicos como Assunto , Citocinas/metabolismo , Vetores Genéticos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Neoplasias/terapia , Poxviridae/genética , Transdução de Sinais , Linfócitos T/metabolismo , Transgenes
7.
Clin Cancer Res ; 11(8): 3017-24, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15837756

RESUMO

PURPOSE: To determine the safety and immunologic and clinical efficacy of a dendritic cell vaccine modified to hyperexpress costimulatory molecules and tumor antigen. EXPERIMENTAL DESIGN: In this phase I study, we administered one or two cycles of four triweekly s.c./intradermal injections of ex vivo generated dendritic cells modified with a recombinant fowlpox vector encoding carcinoembryonic antigen (CEA) and a triad of costimulatory molecules [rF-CEA(6D)-TRICOM]. Controls consisted of immature dendritic cells loaded with tetanus toxoid and a HLA A2-restricted peptide derived from cytomegalovirus pp65 protein. RESULTS: Fourteen patients (11 with colorectal cancer and 3 with non-small cell lung cancer) were enrolled and 12 completed at least one cycle of immunization. There were no grade 3/4 toxicities directly referable to the immunizations. One patient had a decrease in the CEA level from 46 to 6.8 and a minor regression in adenopathy that occurred several months after completion of the immunizations. Five other patients were stable through at least one cycle of immunization (3 months). Direct analysis of peripheral blood mononuclear cells using the ELISpot assay showed an increase in the frequency of CEA-specific T cells in 10 patients (range, 10-541 CEA-specific cells/10(5) peripheral blood mononuclear cells). There was a trend for a greater peak frequency of CEA-specific T cells among those with either a minor response or a stable disease following at least one cycle of therapy. A second cycle was not associated with higher T-cell frequencies. Cytokine flow cytometry showed CEA-specific immune response among both CD4(+) and CD8(+) T cells in all immune responders. CONCLUSION: This immunization strategy is safe and activates potent CEA-specific immune responses.


Assuntos
Vacinas Anticâncer/imunologia , Carcinoma Pulmonar de Células não Pequenas/imunologia , Neoplasias Colorretais/imunologia , Células Dendríticas/imunologia , Neoplasias Pulmonares/imunologia , Antígeno B7-1/genética , Antígeno B7-1/imunologia , Antígenos CD58/genética , Antígenos CD58/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/uso terapêutico , Antígeno Carcinoembrionário/genética , Antígeno Carcinoembrionário/imunologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/tratamento farmacológico , Citocinas/metabolismo , Células Dendríticas/metabolismo , Feminino , Citometria de Fluxo , Vírus da Varíola das Aves Domésticas/genética , Antígeno HLA-A2/imunologia , Humanos , Imunoterapia Adotiva/métodos , Injeções Intradérmicas , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/imunologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Masculino , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Toxoide Tetânico/imunologia , Resultado do Tratamento
8.
Clin Cancer Res ; 11(4): 1597-607, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15746065

RESUMO

PURPOSE: The poor immunogenicity of tumor antigens and the antigenic heterogeneity of tumors call for vaccine strategies to enhance T-cell responses to multiple antigens. Two antigens expressed noncoordinately on most human carcinomas are carcinoembryonic antigen (CEA) and MUC-1. We report here the construction and characterization of two viral vector vaccines to address these issues. EXPERIMENTAL DESIGN: The two viral vectors analyzed are the replication-competent recombinant vaccinia virus (rV-) and the avipox vector, fowlpox (rF-), which is replication incompetent in mammalian cells. Each vector encodes the transgenes for three human costimulatory molecules (B7-1, ICAM-1, and LFA-3, designated TRICOM) and the CEA and MUC-1 transgenes (which also contain agonist epitopes). The vectors are designated rV-CEA/MUC/TRICOM and rF-CEA/MUC/TRICOM. RESULTS: Each of the vectors is shown to be capable of faithfully expressing all five transgenes in human dendritic cells (DC). DCs infected with either vector are shown to activate both CEA- and MUC-1-specific T-cell lines to the same level as DCs infected with CEA-TRICOM or MUC-1-TRICOM vectors. Thus, no evidence of antigenic competition between CEA and MUC-1 was observed. Human DCs infected with rV-CEA/MUC/TRICOM or rF-CEA/MUC/TRICOM are also shown to be capable of generating both MUC-1- and CEA-specific T-cell lines; these T-cell lines are in turn shown to be capable of lysing targets pulsed with MUC-1 or CEA peptides as well as human tumor cells endogenously expressing MUC-1 and/or CEA. CONCLUSION: These studies provide the rationale for the clinical evaluation of these multigene vectors in patients with a range of carcinomas expressing MUC-1 and/or CEA.


Assuntos
Vacinas Anticâncer/genética , Vetores Genéticos/genética , Transgenes/genética , Animais , Antígeno B7-1/genética , Antígeno B7-1/imunologia , Antígenos CD58/genética , Antígenos CD58/imunologia , Vacinas Anticâncer/imunologia , Antígeno Carcinoembrionário/genética , Antígeno Carcinoembrionário/imunologia , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Citotoxicidade Imunológica/imunologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Citometria de Fluxo , Vírus da Varíola das Aves Domésticas/genética , Humanos , Immunoblotting , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/imunologia , Interferon gama/biossíntese , Mucina-1/genética , Mucina-1/imunologia , Recombinação Genética , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Transfecção , Transgenes/imunologia , Vaccinia virus/genética
9.
Clin Cancer Res ; 9(8): 2973-80, 2003 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12912944

RESUMO

PURPOSE: The purpose of this study was to evaluate the immunological responses and therapeutic effectiveness of immunization with fowlpox vaccines encoding the gp100 melanoma antigen in patients with metastatic melanoma. EXPERIMENTAL DESIGN: In three consecutive clinical trials, patients were immunized with recombinant fowlpox viruses encoding three different forms of the melanoma/melanocyte-associated antigen gp100: (a) the native, full-length gp100 molecule; (b) the gp100 molecule with two amino acids modified to increase binding to HLA-A*0201 molecules; and (c) a "minigene" construct encoding a single, modified epitope gp100:209-217(210M) targeted to the endoplasmic reticulum. The immunogenicity of these constructs was studied using peripheral blood mononuclear cells to measure epitope-specific release of IFN-gamma. RESULTS: Reactivity against gp100 was not seen in any patient before receiving fowlpox immunization. Whereas just one of seven patients developed reactivity after receiving fowlpox encoding native gp100, 10 of 14 patients who received fowlpox encoding the anchor modified full-length gp100 exhibited reactivity against the native gp100 molecule, and 12 of 16 patients were successfully immunized after inoculation with the modified minigene construct (p2 = 0.02). There was no difference in the latter group between those randomized to vaccination by i.v. or i.m. routes. There was one partial cancer regression in the group of 46 patients receiving virus in the absence of interleukin (IL)-2. Once patients showed evidence of progressive disease, they were eligible for "cross-over" treatment to IL-2 alone or with the fowlpox virus. None of the 13 patients receiving the full-length or modified full-length forms of gp100 responded when receiving IL-2, whereas 6 of 12 patients who received the fowlpox containing the minigene construct and then received IL-2 showed objective cancer regressions, including three patients with complete regression. CONCLUSIONS: These data underscore the importance of modifying anchor residues of nonmutated self-antigen peptides to generate cellular immune responses after immunization and support the further investigation of recombinant fowlpox viruses encoding modified epitopes administered in combination with IL-2.


Assuntos
Vírus da Varíola das Aves Domésticas/química , Melanoma/metabolismo , Glicoproteínas de Membrana/química , Proteínas de Neoplasias/química , Adulto , Idoso , Antígenos de Neoplasias/química , Linfócitos T CD8-Positivos/metabolismo , Vacinas Anticâncer , Linhagem Celular Tumoral , Epitopos/química , Feminino , Antígenos HLA-A/genética , Antígeno HLA-A2 , Humanos , Imunoglobulina G/química , Interferon gama/metabolismo , Interleucina-2/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Proteínas de Neoplasias/metabolismo , Peptídeos/química , Vacinas/química , Vaccinia virus/metabolismo , Antígeno gp100 de Melanoma
10.
Vaccine ; 29(10): 1948-58, 2011 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-21216311

RESUMO

We evaluated replication-defective poxvirus vectors (modified vaccinia Ankara [MVA] and fowlpox [FPV]) in a homologous and heterologous vector prime-boost vaccination regimen containing matching HIV inserts (MVA-HIV and FPV-HIV) given at months 0, 1, 3, 5 and 7 in 150 healthy HIV-negative vaccinia-naïve participants. FPV-HIV alone was poorly immunogenic, while the high dose (10(9)pfu/2 ml) of MVA-HIV alone elicited maximal responses after two injections: CD4+ and CD8+ T-cell responses in 26/55 (47.3%) and 5/60 (8.3%) of participants, respectively, and IFN-γ ELISpot responses in 28/62 (45.2%). The infrequent CD8+ T-cell responses following MVA-HIV priming were boosted only by the heterologous (FPV-HIV) construct in 14/27 (51.9%) of participants post 4th vaccination. Alternatively, HIV envelope-specific binding antibodies were demonstrated in approximately two-thirds of recipients of the homologous boosting regimen, but in less than 20% of subjects after the heterologous vector boost. Thus, a heterologous poxvirus vector prime-boost regimen can induce HIV-specific CD8+ T-cell and CD4+ T-cell responses, which may be an important feature of an optimal regimen for preventive HIV vaccination.


Assuntos
Vacinas contra a AIDS/imunologia , Portadores de Fármacos , Vírus da Varíola das Aves Domésticas/genética , Vetores Genéticos , Infecções por HIV/prevenção & controle , HIV-1/imunologia , Vaccinia virus/genética , Vacinas contra a AIDS/genética , Adolescente , Adulto , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Feminino , Anticorpos Anti-HIV/sangue , HIV-1/genética , Humanos , Imunização Secundária/métodos , Masculino , Fatores de Tempo , Vacinação/métodos , Adulto Jovem
11.
J Clin Oncol ; 28(7): 1099-105, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-20100959

RESUMO

PURPOSE: Therapeutic prostate-specific antigen (PSA) -targeted poxviral vaccines for prostate cancer have been well tolerated. PROSTVAC-VF treatment was evaluated for safety and for prolongation of progression-free survival (PFS) and overall survival (OS) in a randomized, controlled, and blinded phase II study. PATIENTS AND METHODS: In total, 125 patients were randomly assigned in a multicenter trial of vaccination series. Eligible patients had minimally symptomatic castration-resistant metastatic prostate cancer (mCRPC). PROSTVAC-VF comprises two recombinant viral vectors, each encoding transgenes for PSA, and three immune costimulatory molecules (B7.1, ICAM-1, and LFA-3). Vaccinia-based vector was used for priming followed by six planned fowlpox-based vector boosts. Patients were allocated (2:1) to PROSTVAC-VF plus granulocyte-macrophage colony-stimulating factor or to control empty vectors plus saline injections. RESULTS: Eighty-two patients received PROSTVAC-VF and 40 received control vectors. Patient characteristics were similar in both groups. The primary end point was PFS, which was similar in the two groups (P = .6). However, at 3 years post study, PROSTVAC-VF patients had a better OS with 25 (30%) of 82 alive versus 7 (17%) of 40 controls, longer median survival by 8.5 months (25.1 v 16.6 months for controls), an estimated hazard ratio of 0.56 (95% CI, 0.37 to 0.85), and stratified log-rank P = .0061. CONCLUSION: PROSTVAC-VF immunotherapy was well tolerated and associated with a 44% reduction in the death rate and an 8.5-month improvement in median OS in men with mCRPC. These provocative data provide preliminary evidence of clinically meaningful benefit but need to be confirmed in a larger phase III study.


Assuntos
Vacinas Anticâncer/imunologia , Poxviridae/imunologia , Antígeno Prostático Específico/imunologia , Neoplasias da Próstata/terapia , Idoso , Idoso de 80 Anos ou mais , Método Duplo-Cego , Vetores Genéticos/imunologia , Humanos , Imunização , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Orquiectomia , Antígeno Prostático Específico/antagonistas & inibidores , Neoplasias da Próstata/mortalidade , Neoplasias da Próstata/patologia , Vacinas Sintéticas/imunologia
13.
AIDS Res Hum Retroviruses ; 25(11): 1107-16, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19943789

RESUMO

A recombinant modified vaccinia Ankara virus vaccine candidate (TBC-M4) expressing HIV-1 subtype C env, gag, tat-rev, and nef-RT genes was tested in a randomized, double-blind, dose escalation Phase I trial in 32 HIV-uninfected healthy volunteers who received three intramuscular injections of TBC-M4 at 0, 1, and 6 months of 5 x 10(7) plaque-forming units (pfu) (low dosage, LD) (n = 12) or 2.5 x 10(8) pfu (high dosage, HD) (n = 12) or placebo (n = 8). Local and systemic reactogenicity was experienced by approximately 67% and 83% of vaccine recipients, respectively. The reactogenicity events were mostly mild in severity. Severe but transient systemic reactogenicity was seen in one volunteer of the HD group. No vaccine-related serious adverse events or events suggesting perimyocarditis were seen. A higher frequency of local reactogenicity events was observed in the HD group. Cumulative HIV-specific IFN-gamma ELISPOT responses were detected in frozen PBMCs from 9/11 (82%), 12/12 (100%), and 1/8 (13%) volunteers after the third injection of the LD, HD, and placebo groups, respectively. Most of the responses were to gag and env proteins (maximum of 430 SFU/10(6) PBMCs) persisting across multiple time points. HIV-specific ELISA antibody responses were detected in 10/11, 12/12, and 0/8 volunteers post-third vaccination, in the LD, HD, and placebo groups, respectively. No neutralizing activity against HIV-1 subtype C isolates was detected. TBC-M4 appears to be generally safe and well-tolerated. The immune response detected was dose dependent, modest in magnitude, and directed mostly to env and gag proteins, suggesting further evaluation of this vaccine in a prime-boost regimen.


Assuntos
Vacinas contra a AIDS/efeitos adversos , Vacinas contra a AIDS/imunologia , Infecções por HIV/prevenção & controle , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Vacinas de DNA/efeitos adversos , Vacinas de DNA/imunologia , Vaccinia virus/genética , Vacinas contra a AIDS/administração & dosagem , Adolescente , Adulto , Método Duplo-Cego , Feminino , Anticorpos Anti-HIV/sangue , Infecções por HIV/imunologia , HIV-1/imunologia , Proteínas do Vírus da Imunodeficiência Humana/genética , Humanos , Índia , Interferon gama/metabolismo , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento , Vacinas de DNA/administração & dosagem , Adulto Jovem
14.
Vaccine ; 26(52): 6883-93, 2008 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-18940219

RESUMO

A trial to evaluate the safety and immunogenicity of recombinant modified vaccinia Ankara (MVA) and fowlpox (FP) vectors expressing multiple HIV-1 proteins was conducted in twenty HIV-1 infected youth with suppressed viral replication on HAART. The MVA and FP-based multigene HIV-1 vaccines were safe and well tolerated. Increased frequencies of HIV-1 specific CD4+ proliferative responses and cytokine secreting cells were detected following immunization. Increased frequencies and breadth of HIV-1 specific CD8 T-cell responses were also detected. Plasma HIV-1-specific antibody levels and neutralizing activity were unchanged following vaccination. Poxvirus-based vaccines may merit further study in therapeutic vaccine protocols.


Assuntos
Vacinas contra a AIDS/efeitos adversos , Vacinas contra a AIDS/imunologia , Terapia Antirretroviral de Alta Atividade , Vírus da Varíola das Aves Domésticas/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , Adolescente , Contagem de Linfócito CD4 , Relação CD4-CD8 , Citocinas/biossíntese , Feminino , Citometria de Fluxo , Produtos do Gene gag/biossíntese , Produtos do Gene gag/genética , Anticorpos Anti-HIV/análise , Anticorpos Anti-HIV/biossíntese , Infecções por HIV/tratamento farmacológico , HIV-1/genética , Humanos , Interferon gama/biossíntese , Masculino , Estudos Prospectivos , RNA Viral/análise , RNA Viral/biossíntese , Linfócitos T/imunologia , Vacinas Sintéticas/imunologia , Vacínia/imunologia , Adulto Jovem
15.
J Virol ; 81(16): 8563-70, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17553898

RESUMO

As the diversity of potential immunogens increases within certain classes of vectors, the possibility has arisen of employing heterologous prime/boost immunizations using diverse members of the same family of vectors. The present study was initiated to explore the use of divergent pox vectors in a prime/boost regimen to elicit high-frequency cellular immune responses to human immunodeficiency virus type 1 envelope and simian immunodeficiency virus gag in rhesus monkeys. We demonstrated that monkeys vaccinated with a recombinant modified vaccinia virus Ankara (rMVA) prime/recombinant fowlpox virus (rFPV) boost regimen and monkeys vaccinated with a recombinant vaccinia virus prime/rFPV boost regimen developed comparable cellular immune responses that were greater in magnitude than those elicited by a homologous prime/boost with rMVA. Nevertheless, comparable magnitude recall cellular immune responses were observed in monkeys vaccinated with heterologous and homologous recombinant poxvirus following challenge with the CXCR4-tropic SHIV-89.6P. Consistent with this finding, comparable levels of containment of viral replication and CD4(+) T-lymphocyte preservation were seen in these groups of recombinant poxvirus-vaccinated monkeys. This study supports further exploration of combining recombinant vectors of the same family in prime/boost immunization strategies to optimize vaccine-elicited cellular immune responses.


Assuntos
Vetores Genéticos/imunologia , Imunização Secundária/métodos , Poxviridae/imunologia , Vacinação/métodos , Vaccinia virus/imunologia , Animais , Anticorpos Antivirais/sangue , Contagem de Linfócito CD4 , Vetores Genéticos/genética , Imunidade Celular , Macaca mulatta , Poxviridae/genética , RNA Viral/sangue , Vaccinia virus/genética , Replicação Viral
16.
Proc Natl Acad Sci U S A ; 103(39): 14453-8, 2006 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-16984998

RESUMO

NY-ESO-1 is a cancer/testis antigen expressed in a range of human malignancies, and a number of vaccine strategies targeting NY-ESO-1 are being developed. In the present study, the safety and immunogenicity of recombinant vaccinia-NY-ESO-1 and recombinant fowlpox-NY-ESO-1 were analyzed in a series of 36 patients with a range of different tumor types. Each construct was first tested individually at two different dose levels and then in a prime-boost setting with recombinant vaccinia-NY-ESO-1 followed by recombinant fowlpox-NY-ESO-1. The vaccines were well tolerated either individually or together. NY-ESO-1-specific antibody responses and/or specific CD8 and CD4 T cell responses directed against a broad range of NY-ESO-1 epitopes were induced by a course of at least four vaccinations at monthly intervals in a high proportion of patients. CD8 T cell clones derived from five vaccinated patients were shown to lyse NY-ESO-1-expressing melanoma target cells. In several patients with melanoma, there was a strong impression that the natural course of the disease was favorably influenced by vaccination.


Assuntos
Formação de Anticorpos/imunologia , Vacinas Anticâncer/imunologia , Imunidade Celular/imunologia , Proteínas de Neoplasias/imunologia , Neoplasias/imunologia , Fragmentos de Peptídeos/imunologia , Vacinas Sintéticas/imunologia , Anticorpos/sangue , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/efeitos adversos , Vacinas Anticâncer/genética , Células Clonais , Estudos de Coortes , Citotoxicidade Imunológica/imunologia , Epitopos/imunologia , Vírus da Varíola das Aves Domésticas/metabolismo , Humanos , Proteínas de Membrana/imunologia , Vacinação , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/genética , Vaccinia virus/genética
17.
J Immunol ; 176(9): 5338-45, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16622001

RESUMO

Functional impairment of virus-specific memory CD8(+) T lymphocytes has been associated with clinical disease progression following HIV, SIV, and simian human immunodeficiency virus infection. These lymphocytes have a reduced capacity to produce antiviral cytokines and mediators involved in the lysis of virally infected cells. In the present study, we used polychromatic flow cytometry to assess the frequency and functional capacity of central memory (CD28(+)CD95(+)) and effector memory (CD28(-)CD95(+)) subpopulations of Gag-specific CD8(+) T cells in SIV/simian human immunodeficiency virus-infected rhesus monkeys. The aim of this study was to determine whether Ag-specific, memory CD8(+) T cell function could be preserved in infected monkeys that had been immunized before infection with a vaccine regimen consisting of a plasmid DNA prime followed by a recombinant viral vector boost. We observed that vaccination was associated with the preservation of Gag-specific central memory CD8(+) T cells that were functionally capable of producing IFN-gamma, and effector memory CD8(+) T cells that were capable of producing granzyme B following viral Ag exposure.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Memória Imunológica/imunologia , Macaca mulatta/imunologia , Macaca mulatta/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Vacinas Virais/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Comunicação Celular/imunologia , Células Cultivadas , Progressão da Doença , Epitopos de Linfócito T/imunologia , Regulação da Expressão Gênica , Produtos do Gene gag/imunologia , Granzimas , Interferon gama/biossíntese , Macaca mulatta/metabolismo , Peptídeos/imunologia , Serina Endopeptidases/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia
18.
Blood ; 106(10): 3515-23, 2005 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16081691

RESUMO

Chronic lymphocytic leukemia (CLL) is a disease of CD5(+) B lymphocytes (designated as CLL cells) that are inefficient antigen-presenting cells. Their poor ability to present antigens to the T cells, largely due to an inadequate costimulatory capacity, is manifested as a failure to stimulate proliferation of both allogeneic and autologous T cells. We have investigated the ability of in vitro manipulated CLL cells, via hyperexpression of a triad of costimulatory molecules (B7-1, intercellular adhesion molecule 1 [ICAM-1], and leukocyte-function-associated antigen 3 [LFA-3], designated TRICOM), to stimulate effective antitumor T-cell responses. A recombinant modified vaccinia virus strain Ankara (MVA), which is a highly attenuated, replication-impaired virus variant, was successfully used to infect and deliver the simultaneous expression of the 3 human costimulatory molecules in TRICOM on the surface of the CLL cells. Proliferation of allogeneic and autologous T cells was observed when MVA-TRICOM-infected CLL cells were used as stimulators in proliferation assays. Cytotoxic T lymphocytes, generated in vitro by stimulation of autologous T cells with MVA-TRICOM-infected CLL cells, showed cytotoxicity against unmodified/uninfected CLL cells. Therefore, our findings suggest that the use of CLL cells infected ex vivo with MVA-TRICOM or direct injection of MVA-TRICOM in patients with CLL has potential for the immunotherapy of CLL.


Assuntos
Antígenos CD/imunologia , Vacinas Anticâncer/imunologia , Vetores Genéticos , Imunoterapia , Leucemia Linfocítica Crônica de Células B/imunologia , Vaccinia virus , Idoso , Idoso de 80 Anos ou mais , Antígenos CD/genética , Vacinas Anticâncer/genética , Proliferação de Células , Citotoxicidade Imunológica/genética , Citotoxicidade Imunológica/imunologia , Feminino , Humanos , Imunoterapia/métodos , Leucemia Linfocítica Crônica de Células B/terapia , Masculino , Pessoa de Meia-Idade , Linfócitos T Citotóxicos/imunologia , Células Tumorais Cultivadas
19.
J Virol ; 79(8): 4580-8, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15795244

RESUMO

The expression of particular major histocompatibility complex (MHC) class I alleles can influence the rate of disease progression following lentiviral infections. This effect is a presumed consequence of potent cytotoxic T-lymphocyte (CTL) responses that are restricted by these MHC class I molecules. The present studies have examined the impact of the MHC class I allele Mamu-A*01 on simian/human immunodeficiency virus 89.6P (SHIV-89.6P) infection in unvaccinated and vaccinated rhesus monkeys by exploring the contribution of dominant-epitope specific CTL in this setting. Expression of Mamu-A*01 in immunologically naive monkeys was not associated with improved control of viral replication, CD4+ T-lymphocyte loss, or survival. In contrast, Mamu-A*01+ monkeys that had received heterologous prime/boost immunizations prior to challenge maintained higher CD4+ T-lymphocyte levels and better control of SHIV-89.6P replication than Mamu-A*01- monkeys. This protection was associated with the evolution of high-frequency anamnestic CTL responses specific for a dominant Mamu-A*01-restricted Gag epitope following infection. These data indicate that specific MHC class I alleles can confer protection in the setting of a pathogenic SHIV infection by their ability to elicit memory CTL following vaccination.


Assuntos
Vacinas contra a AIDS/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , HIV/fisiologia , Antígenos de Histocompatibilidade Classe I/imunologia , Memória Imunológica , Vírus da Imunodeficiência Símia/fisiologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/virologia , Animais , Primers do DNA , Humanos , Macaca mulatta , Reação em Cadeia da Polimerase , RNA Viral/sangue , Replicação Viral
20.
J Virol ; 79(10): 6554-9, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15858042

RESUMO

Poxvirus vaccine vectors, although capable of eliciting potent immune responses, pose serious health risks in immunosuppressed individuals. We therefore constructed five novel recombinant vaccinia virus vectors which contained overlapping deletions of coding regions for the B5R, B8R, B12R, B13R, B14R, B16R, B18R, and B19R immunomodulatory gene products and assessed them for both immunogenicity and pathogenicity. All five of these novel vectors elicited both cellular and humoral immunity to the inserted HIV-BH10 env comparable to that induced by the parental Wyeth strain vaccinia virus. However, deletion of these immunomodulatory genes did not increase the immunogenicity of these vectors compared with the parental vaccinia virus. Furthermore, four of these vectors were slightly less virulent and one was slightly more virulent than the Wyeth strain virus in neonatal mice. Attenuated poxviruses have potential use as safer alternatives to current replication-competent vaccinia virus. Improved vaccinia virus vectors can be generated by deleting additional genes to achieve a more significant viral attenuation.


Assuntos
Vacinas contra a AIDS/imunologia , Vetores Genéticos/genética , Infecções por HIV/imunologia , Vaccinia virus/genética , Vaccinia virus/patogenicidade , Animais , Feminino , Deleção de Genes , Anticorpos Anti-HIV/sangue , Proteína gp120 do Envelope de HIV/genética , Imunização , Imunização Secundária , Dose Letal Mediana , Camundongos , Camundongos Endogâmicos BALB C , Vacinas Sintéticas/imunologia , Virulência
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa