Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cancer Immunol Immunother ; 65(5): 537-49, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26961085

RESUMEN

The dramatic clinical benefit of immune checkpoint blockade for a fraction of cancer patients suggests the potential for further clinical benefit in a broader cancer patient population by combining immune checkpoint inhibitors with active immunotherapies. The anti-tumor efficacy of MVA-BN-HER2 poxvirus-based active immunotherapy alone or in combination with CTLA-4 checkpoint blockade was investigated in a therapeutic CT26-HER-2 lung metastasis mouse model. MVA-BN-HER2 immunotherapy significantly improved the median overall survival compared to untreated controls or CTLA-4 blockade alone (p < 0.001). Robust synergistic efficacy was achieved with the combination therapy (p < 0.01). Improved survival following MVA-BN-HER2 administration was accompanied by increased tumor infiltration by HER-2-specific cytotoxic T lymphocytes (CTL). These tumor-specific CTL had characteristics similar to antiviral CTL, including strong expression of activation markers and co-expression of IFNγ and TNFα. Combination with CTLA-4 blockade significantly increased the magnitude of HER-2-specific T cell responses, with a higher proportion co-expressing TNFα and/or IL-2 with IFNγ. Furthermore, in mice treated with MVA-BN-HER2 (alone or in combination with CTLA-4 blockade), the inducible T cell co-stimulator (ICOS) protein was expressed predominantly on CD4 and CD8 effector T cells but not on regulatory T cells (T(reg)). In contrast, mice left untreated or treated solely with CTLA-4 blockade harbored elevated ICOS(+) Treg, a phenotype associated with highly suppressive activity. In conclusion, poxvirus-based active immunotherapy induced robust tumor infiltration by highly efficient effector T cells. Combination with CTLA-4 immune checkpoint blockade amplified this response resulting in synergistically improved efficacy. These hypothesis-generating data may help elucidate evidence of enhanced clinical benefit from combining CTLA-4 blockade with poxvirus-based active immunotherapy.


Asunto(s)
Antígeno CTLA-4/inmunología , Vacunas contra el Cáncer/inmunología , Neoplasias Experimentales/inmunología , Linfocitos T Citotóxicos/inmunología , Virus Vaccinia/inmunología , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Antígeno CTLA-4/antagonistas & inhibidores , Vacunas contra el Cáncer/farmacología , Línea Celular Tumoral , Citocinas/inmunología , Citocinas/metabolismo , Sinergismo Farmacológico , Femenino , Citometría de Flujo , Humanos , Inmunoterapia/métodos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Ratones Endogámicos BALB C , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Análisis de Supervivencia , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Virus Vaccinia/genética
2.
PLoS One ; 11(2): e0150084, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26910562

RESUMEN

Poxvirus-based active immunotherapies mediate anti-tumor efficacy by triggering broad and durable Th1 dominated T cell responses against the tumor. While monotherapy significantly delays tumor growth, it often does not lead to complete tumor regression. It was hypothesized that the induced robust infiltration of IFNγ-producing T cells into the tumor could provoke an adaptive immune evasive response by the tumor through the upregulation of PD-L1 expression. In therapeutic CT26-HER-2 tumor models, MVA-BN-HER2 poxvirus immunotherapy resulted in significant tumor growth delay accompanied by a robust, tumor-infiltrating T cell response that was characterized by low to mid-levels of PD-1 expression on T cells. As hypothesized, this response was countered by significantly increased PD-L1 expression on the tumor and, unexpectedly, also on infiltrating T cells. Synergistic benefit of anti-tumor therapy was observed when MVA-BN-HER2 immunotherapy was combined with PD-1 immune checkpoint blockade. Interestingly, PD-1 blockade stimulated a second immune checkpoint molecule, LAG-3, to be expressed on T cells. Combining MVA-BN-HER2 immunotherapy with dual PD-1 plus LAG-3 blockade resulted in comprehensive tumor regression in all mice treated with the triple combination therapy. Subsequent rejection of tumors lacking the HER-2 antigen by treatment-responsive mice without further therapy six months after the original challenge demonstrated long lasting memory and suggested that effective T cell immunity to novel, non-targeted tumor antigens (antigen spread) had occurred. These data support the clinical investigation of this triple therapy regimen, especially in cancer patients harboring PD-L1neg/low tumors unlikely to benefit from immune checkpoint blockade alone.


Asunto(s)
Antígenos CD/inmunología , Inmunidad Celular , Inmunoterapia , Neoplasias Experimentales/terapia , Poxviridae/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T/inmunología , Animales , Antígenos CD/genética , Línea Celular Tumoral , Ratones , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Poxviridae/genética , Receptor de Muerte Celular Programada 1/genética , Proteína del Gen 3 de Activación de Linfocitos
3.
J Immunother Cancer ; 2(1): 34, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25328681

RESUMEN

BACKGROUND: PROSTVAC®, an active immunotherapy currently studied for the treatment of metastatic castration-resistant prostate cancer (mCRPC), consists of a heterologous prime-boost regimen with two different poxvirus-based vectors to provoke productive immune responses against prostate specific antigen (PSA) as the target tumor antigen. A Phase 2 study of PROSTVAC immunotherapy showed significantly improved median overall survival by 8.5 months and is currently being validated in a global Phase 3 study (PROSPECT; NCT01322490). Here, preclinical models were explored to investigate the mechanism of action and immune signatures of anti-tumor efficacy with PROSTVAC immunotherapy with the goal to identify potential immune correlates of clinical benefit. METHODS: PROSTVAC-induced immune responses and anti-tumor efficacy were studied in male BALB/c mice. Functionality of the induced T cell response was characterized by interferon-gamma (IFNγ) ELISPOT, cytotoxic degranulation, multi-cytokine intracellular staining, and in vivo T cell depletion. Tumor infiltrating lymphocytes (TILs) were evaluated phenotypically by flow cytometry. RESULTS: The heterologous prime-boost regimen of the two PROSTVAC vectors significantly enhanced the magnitude and quality of activated PSA-specific CD4 and CD8 T cell responses compared to homologous, single vector regimens. PROSTVAC-activated CD4 and CD8 T cells were highly functional as evidenced by expression of activation markers, production of multiple cytokines, and amplified cytotoxic T cell activity. Importantly, PROSTVAC immunotherapy resulted in significant anti-tumor efficacy in a transplantable prostate cancer mouse model. Antigen-spreading occurred in PROSTVAC-treated animals that rejected PSA-expressing tumors, as shown by subsequent rejection of PSA-negative tumors. In vivo CD4 and CD8 depletion revealed that both T cell subsets contributed to anti-tumor efficacy. Characterization of TILs demonstrated that PROSTVAC immunotherapy greatly increased the intra-tumoral ratio of activated effector to regulatory T cells. CONCLUSIONS: PROSTVAC immunotherapy activates broad, highly functional T cell immunity to PSA and to endogenous tumor antigens via immune-mediated antigen spreading. These preclinical results further elucidate the mode of action of PROSTVAC immunotherapy and its potential causal relationship to extended overall survival as observed in the PROSTVAC Phase 2 study. The clinical validation is ongoing in the PROSPECT Phase 3 clinical study.

4.
Vaccine ; 28(37): 6028-35, 2010 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-20619375

RESUMEN

Chromosomal translocations generating the BCR-ABL oncogene cause chronic myeloid leukemia (CML) and a subset of acute lymphoblastic leukemia. The BCR-ABL(T315I) mutation confers drug resistance to FDA-approved targeted therapeutics imatinib mesylate, dasatinib, and nilotinib. We tested the ability of a recombinant yeast-based vaccine expressing the T315I-mutated BCR-ABL antigen to stimulate an anti-BCR-ABL(T315I) immune response. The yeast-based immunotherapy significantly reduced or eliminated BCR-ABL(T315I) leukemia cells from the peripheral blood of immunized animals and extended leukemia-free survival in a murine model of BCR-ABL(+) leukemia compared to animals receiving sham injection or yeast expressing ovalbumin. With immunization, leukemic cells harboring BCR-ABL(T315I) were selectively eliminated after challenge with a mixed population of BCR-ABL and BCR-ABL(T315I) leukemias. In summary, yeast-based immunotherapy represents a novel approach against the emergence of cancer drug resistance by the pre-emptive targeted ablation of tumor escape mutants.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Proteínas de Fusión bcr-abl/inmunología , Leucemia Mielógena Crónica BCR-ABL Positiva/terapia , Secuencia de Aminoácidos , Animales , Supervivencia sin Enfermedad , Genes MHC Clase I , Inmunoterapia , Leucemia Experimental/inmunología , Leucemia Experimental/terapia , Leucemia Mielógena Crónica BCR-ABL Positiva/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Saccharomyces cerevisiae , Vacunas Sintéticas/inmunología
5.
Cancer Immunol Immunother ; 59(3): 397-408, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19756595

RESUMEN

PURPOSE: Studies comparing two or more vaccine platforms have historically evaluated each platform based on its ability to induce an immune response and may conclude that one vaccine is more efficacious than the other(s), leading to a recommendation for development of the more effective vaccine for clinical studies. Alternatively, these studies have documented the advantages of a diversified prime and boost regimen due to amplification of the antigen-specific T-cell population. We hypothesize here that two vaccine platforms targeting the same antigen might induce shared and distinct antigen-specific T-cell populations, and examined the possibility that two distinct vaccines could be used concomitantly. EXPERIMENTAL DESIGN: Using recombinant poxvirus and yeast vaccines, we compared the T-cell populations induced by these two platforms in terms of serum cytokine response, T-cell gene expression, T-cell receptor phenotype, antigen-specific cytokine expression, T-cell avidity, and T-cell antigen-specific tumor cell lysis. RESULTS: These studies demonstrate for the first time that vaccination with a recombinant poxvirus platform (rV/F-CEA/TRICOM) or a heat-killed yeast vaccine platform (yeast-CEA) elicits T-cell populations with both shared and unique phenotypic and functional characteristics. Furthermore, both the antigen and the vector play a role in the induction of distinct T-cell populations. CONCLUSIONS: In this study, we demonstrate that concurrent administration of two vaccines targeting the same antigen induces a more diverse T-cell population that leads to enhanced antitumor efficacy. These studies provide the rationale for future clinical studies investigating concurrent administration of vaccine platforms targeting a single antigen to enhance the antigen-specific immune response.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Linfocitos T/inmunología , Vacunación , Animales , Antígenos/inmunología , Antígeno Carcinoembrionario/inmunología , Línea Celular , Línea Celular Tumoral , Quimiocinas/metabolismo , Citocinas/sangre , Femenino , Vectores Genéticos , Inmunización Secundaria , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Poxviridae/inmunología , Saccharomyces cerevisiae/inmunología
6.
Vaccine ; 27(7): 987-94, 2009 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-19110021

RESUMEN

Tumor-associated antigens are weakly immunogenic. Human carcinoembryonic antigen (CEA) is overexpressed on a wide range of human carcinomas and represents an attractive target for cancer immunotherapy. This study analyzes the ability of a Saccharomyces cerevisiae vector containing the transgene encoding CEA (yeast-CEA) to activate human dendritic cells (DCs) and stimulate CEA-specific T-cell responses. We demonstrate for the first time that treatment with yeast-CEA can activate human DCs, resulting in increases in surface expression of CD80, CD83, CD54, CD58, and MHC class II, and increased production by DCs of IL-12p70, TNF-alpha, IFN-gamma, IL-8, IL-2, IL-13, IL-10, and IL-1beta. We also show that human DCs treated with yeast-CEA can activate CEA-specific T-cell lines and can act as antigen-presenting cells (APCs) to generate CEA-specific T-cell lines capable of lysing CEA(+) human tumor cells. Gene expression profiles of human DCs treated with yeast-CEA show increased expression of numerous genes involved in the production of chemokines and cytokines and their receptors, and genes related to antigen uptake, antigen presentation, and signal transduction.


Asunto(s)
Antígeno Carcinoembrionario/biosíntesis , Antígeno Carcinoembrionario/inmunología , Células Dendríticas/inmunología , Saccharomyces cerevisiae/genética , Antígenos CD/análisis , Antígeno Carcinoembrionario/genética , Línea Celular Tumoral , Citocinas/biosíntesis , Pruebas Inmunológicas de Citotoxicidad , Células Dendríticas/química , Perfilación de la Expresión Génica , Humanos , Activación de Linfocitos , Saccharomyces cerevisiae/inmunología , Linfocitos T Citotóxicos/inmunología
7.
Clin Cancer Res ; 14(13): 4316-25, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18594015

RESUMEN

PURPOSE: Saccharomyces cerevisiae, a nonpathogenic yeast, has been used previously as a vehicle to elicit immune responses to foreign antigens, and tumor-associated antigens, and has been shown to reduce tumor burden in mice. Studies were designed to determine if vaccination of human carcinoembryonic antigen (CEA)-transgenic (CEA-Tg) mice (where CEA is a self-antigen) with a recombinant S. cerevisiae construct expressing human CEA (yeast-CEA) elicits CEA-specific T-cell responses and antitumor activity. EXPERIMENTAL DESIGN: CEA-Tg mice were vaccinated with yeast-CEA, and CD4(+) and CD8(+) T-cell responses were assessed after one and multiple administrations or vaccinations at multiple sites per administration. Antitumor activity was determined by tumor growth and overall survival in both pulmonary metastasis and s.c. pancreatic tumor models. RESULTS: These studies demonstrate that recombinant yeast can break tolerance and that (a) yeast-CEA constructs elicit both CEA-specific CD4(+) and CD8(+) T-cell responses; (b) repeated yeast-CEA administration causes increased antigen-specific T-cell responses after each vaccination; (c) vaccination with yeast-CEA at multiple sites induces a greater T-cell response than the same dose given at a single site; and (d) tumor-bearing mice vaccinated with yeast-CEA show a reduction in tumor burden and increased overall survival compared to mock-treated or control yeast-vaccinated mice in both pulmonary metastasis and s.c. pancreatic tumor models. CONCLUSIONS: Vaccination with a heat-killed recombinant yeast expressing the tumor-associated antigen CEA induces CEA-specific immune responses, reduces tumor burden, and extends overall survival in CEA-Tg mice. These studies thus form the rationale for the incorporation of recombinant yeast-CEA and other recombinant yeast constructs in cancer immunotherapy protocols.


Asunto(s)
Antígenos de Neoplasias/química , Antineoplásicos/farmacología , Antígeno Carcinoembrionario/química , Regulación de la Expresión Génica , Inmunoterapia/métodos , Saccharomyces cerevisiae/metabolismo , Vacunas de ADN/química , Animales , Antígenos de Neoplasias/metabolismo , Linfocitos T CD8-positivos/metabolismo , Vacunas contra el Cáncer , Antígeno Carcinoembrionario/metabolismo , Proliferación Celular , Femenino , Humanos , Linfocitos/citología , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/química , Vacunas de ADN/metabolismo
8.
Vaccine ; 26(4): 509-21, 2008 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-18155327

RESUMEN

Recombinant Saccharomyces cerevisiae (yeast) represents a unique and attractive vehicle to deliver antigens in vaccine immunotherapy protocols for cancer or infectious disease, in that it has been shown to be extremely safe and can be administered multiple times to hosts. In the studies reported here, we describe the effects of treatment with recombinant yeast on murine immature dendritic cells (DCs). Yeast expressing human carcinoembryonic antigen (CEA) as a model antigen was studied. Injection of mice subcutaneously with yeast-CEA resulted in rapid increases in MHC class II(+) cells and total antigen-presenting cells in draining lymph nodes. Post-treatment with yeast-CEA, DCs rapidly elevated both MHC class I and class II, numerous costimulatory molecules and other DC maturation markers, and secreted a range of Type I inflammatory cytokines. Gene expression arrays also revealed the rapid up-regulation of numerous cytokine and chemokine mRNAs, as well as genes involved in signal transduction and antigen uptake. Functional studies demonstrated enhanced allospecific reactivity of DCs following treatment with yeast-CEA or control yeast. Additionally, treatment of DCs with yeast-CEA resulted in specific activation of CEA-specific CD8(+) T cells in an MHC-restricted manner in vitro. Lastly, vaccination of CEA-transgenic mice with yeast-CEA elicited antigen-specific CD4(+) and CD8(+) immune responses in vivo. Thus, these studies taken together form a scientific rationale for the use of recombinant yeast in vaccination protocols for cancer or infectious diseases.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Vectores Genéticos/inmunología , Saccharomyces cerevisiae/inmunología , Vacunación , Animales , Células Presentadoras de Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Antígeno Carcinoembrionario/genética , Citocinas/biosíntesis , Femenino , Antígenos de Histocompatibilidad Clase I/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Inyecciones Subcutáneas , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Neoplasias/terapia , Saccharomyces cerevisiae/genética
9.
Exp Dermatol ; 16(10): 814-22, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17845213

RESUMEN

Malignant melanoma is a potentially deadly form of skin cancer and people at high-risk of developing melanoma will benefit from effective preventive intervention. Yeast can be used as an efficient vehicle of antigen loading and immunostimulation. Saccharomyces cerevisiae is not pathogenic to humans and can be easily engineered to express specific antigens. In this study, we have developed a melanoma vaccine using a yeast-based platform expressing a full-length melanocyte/melanoma protein to investigate its utility as a prophylactic melanoma vaccine in a transplantable mouse melanoma model. Yeast was engineered and expanded in vitro without technical difficulties, administered easily with subcutaneous injection, and did not show adverse effects, indicating its practical applicability and favourable safety profile. Despite the lack of knowledge of dominant epitopes of the protein recognized by mouse MHC-class I, the vaccine protected mice from tumor development and induced efficient immune responses, suggesting that the precise knowledge of epitopic sequences and the matched HLA type is not required when delivering a full-length protein using the yeast platform. In addition, the vaccine stimulated both CD4(+) T cells and CD8(+) T cells simultaneously. This study provides a 'proof of principle' that recombinant yeast can be utilized as an effective prophylactic vaccine to target patients at high-risk for melanoma.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Melanoma Experimental/prevención & control , Proteínas de Neoplasias/inmunología , Saccharomyces cerevisiae/genética , Animales , Antígenos de Neoplasias/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/genética , Línea Celular Tumoral , Pruebas Inmunológicas de Citotoxicidad , Citotoxicidad Inmunológica/inmunología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Interferón gamma/metabolismo , Estimación de Kaplan-Meier , Antígeno MART-1 , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Proteínas de Neoplasias/genética , Proteínas Recombinantes/inmunología , Bazo/citología , Bazo/inmunología , Bazo/metabolismo , Transfección , Vacunación
10.
J Neurooncol ; 81(2): 149-62, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16941073

RESUMEN

TGF-beta2 secretion by high grade gliomas has been implicated as one of the major factors contributing to tumor growth, alterations in the host immune response to tumor, and failure of gliomas to respond to current immunotherapy strategies. We hypothesized that targeted delivery and inhibition of TGF-beta2 by TGF-beta2 antisense oligonucleotides (AS-ODNs) would overcome tumor-induced immunosuppression and enhance the capacity of tumor vaccines to eradicate established brain tumors. Utilizing the mRNA sequences of TGF-beta2, specific AS-ODNs were constructed and tested for their ability to inhibit TGF-beta2 production in 9L glioma cells. The effect of combining local intracranial administration of antisense ODNs with systemic tumor vaccine was examined. Fisher 344 rats were vaccinated subcutaneously with irradiated 9L tumor cells 3 days after intracranial tumor implantation. Four days after vaccination, ODNs were administered into the tumor mass and survival was followed. ODNs delivered locally distributed widely within the brain tumor mass and inhibited TGF-beta2 expression. Survival of tumor-bearing rats treated with the combination of local antisense and systemic tumor vaccine was significantly enhanced (mean survival time (MST): 48.0 days). In contrast, MST for animals treated with nonsense plus vaccine, vaccine alone, antisense alone or PBS showed no survival advantage and no statistical differences between groups (33.5 days, 29.0 days, 37.5 days, and 31.5 days, respectively). Our data supports the hypothesis that local administration of antisense TGF-beta2 ODNs combined with systemic vaccination can increase efficacy of immunotherapy and is a novel, potentially clinically applicable, strategy for high-grade glioma treatment.


Asunto(s)
Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/uso terapéutico , Gliosarcoma/terapia , Inmunoterapia/métodos , Oligonucleótidos Antisentido/uso terapéutico , Factor de Crecimiento Transformador beta2/antagonistas & inhibidores , Animales , Neoplasias Encefálicas/mortalidad , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Vectores Genéticos , Gliosarcoma/mortalidad , Inmunoprecipitación , Inyecciones Subcutáneas , Masculino , Ratas , Ratas Endogámicas F344 , Tasa de Supervivencia , Células Tumorales Cultivadas , Vacunación
11.
Vaccine ; 25(8): 1452-63, 2007 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-17098335

RESUMEN

Control of primary infection with hepatitis C virus (HCV) is associated with robust and broad T cell immunity. In contrast, chronic infection is characterized by weak T cell responses suggesting that an approach that boosts these responses could be a therapeutic advance. Saccharomyces cerevisiae is an effective inducer of innate and adaptive cellular immunity and we have generated recombinant yeast cells (GI-5005) that produce an HCV NS3-Core fusion protein. Pre-clinical studies in mice showed that GI-5005 induced potent antigen-specific proliferative and cytotoxic T cell responses that were associated with Th1-type cytokine secretion. In studies in which GI-5005 was administered up to 13 times, no detectable vector neutralization or induction of tolerance was observed. Prophylactic as well as therapeutic administration of GI-5005 in mice led to eradication of tumor cells expressing HCV NS3 protein. Immunotherapy with GI-5005 is being evaluated in chronic HCV infected individuals in a Phase 1 clinical trial.


Asunto(s)
Hepacivirus/inmunología , Inmunoterapia/métodos , Proteínas Recombinantes de Fusión/inmunología , Saccharomyces cerevisiae/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Proteínas del Núcleo Viral/inmunología , Vacunas contra Hepatitis Viral/inmunología , Proteínas no Estructurales Virales/inmunología , Animales , Línea Celular Tumoral , Citocinas/inmunología , Citocinas/metabolismo , Células HeLa , Hepacivirus/genética , Hepatitis C/inmunología , Hepatitis C/prevención & control , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas Recombinantes de Fusión/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas del Núcleo Viral/biosíntesis , Proteínas del Núcleo Viral/genética , Vacunas contra Hepatitis Viral/genética , Proteínas no Estructurales Virales/biosíntesis , Proteínas no Estructurales Virales/genética
12.
Expert Opin Biol Ther ; 5(4): 565-75, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15934834

RESUMEN

Immunotherapy for cancer represents an attractive therapeutic target because of its specificity and lack of toxicity, but products investigated so far have been limited by neutralisation, complexity of manufacturing and requirement for patient-specific products. Recombinant yeast cells are capable of stimulating the immune system to produce highly specific and potent cellular responses against target protein antigens with little toxicity. Data from animal models suggest that Tarmogens (yeast-based immunotherapeutics) can elicit protective immunity against xenografted and chemically induced tumours. This concept is now being tested in a Phase I trial in patients with colorectal, pancreatic and non-small cell lung cancers.


Asunto(s)
Antígenos de Neoplasias/inmunología , Inmunoterapia/métodos , Neoplasias/inmunología , Levaduras/inmunología , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/uso terapéutico , Humanos , Neoplasias/genética , Neoplasias/terapia , Levaduras/genética
13.
Cancer Res ; 64(15): 5084-8, 2004 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-15289309

RESUMEN

Activating mutations in Ras oncoproteins represent attractive targets for cancer immunotherapy, but few vectors capable of generating immune responses required for tumor killing without vector neutralization have been described. Whole recombinant yeast heterologously expressing mammalian mutant Ras proteins were used to immunize mice in a carcinogen-induced lung tumor model. Therapeutic immunization with the whole recombinant yeast caused complete regression of established Ras mutation-bearing lung tumors in a dose-dependent, antigen-specific manner. In combination with the genomic sequencing of tumors in patients, the yeast-based immunotherapeutic approach could be applied to treat Ras mutation-bearing human cancers.


Asunto(s)
Inmunoterapia/métodos , Neoplasias Pulmonares/prevención & control , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/prevención & control , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/inmunología , Adenoma/inducido químicamente , Adenoma/inmunología , Adenoma/prevención & control , Animales , ADN de Neoplasias/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/inmunología , Masculino , Ratones , Ratones Endogámicos , Mutación , Neoplasias Experimentales/inducido químicamente , Proteínas Proto-Oncogénicas p21(ras)/farmacología , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/farmacología , Saccharomyces cerevisiae/fisiología , Uretano
14.
Clin Immunol ; 112(1): 54-65, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15207782

RESUMEN

Signals transmitted by binding of Fas ligand (FasL) to the Fas receptor (CD95/Apo-1) have pleiotropic effects on cellular function that present opportunities for therapeutic applications. For example, depending on the circumstances, overexpression of FasL can enhance, prevent, or reverse growth of spontaneous or transplantable tumors. Furthermore, local administration of FasL into a single paw in susceptible mice protects from or reduces the severity of collagen-induced arthritis (CIA) in all paws. Here, we define mechanisms that mediate systemic protection induced by locally delivered FasL. Protection is not solely dependent on local interactions between Fas and FasL, but rather requires induction of a paradoxical inflammatory response that not only destroys Fas-resistant tumors, but also recruits motile, activated, Fas-bearing T cells that are Fas sensitive. We demonstrate by following the antigen-specific recruitment and subsequent termination of transgenic T cells that activated T cells, including autoreactive cells responsible for CIA, are eliminated within this inflammatory environment through the overexpressed FasL. The nature of the inflammatory response, which depends on the Fas ligand being cell bound and not soluble, and the magnitude of FasL expression within the inflammatory milieu are essential for this effect, as arthritogenic inflammation alone resulting from CIA induction is insufficient to ameliorate the disease or eliminate antigen-specific T cells, even upon systemic delivery of soluble FasL. These data show that gene delivery of membrane-bound FasL can effectively recruit and eliminate autoreactive T cells.


Asunto(s)
Artritis Experimental/prevención & control , Autoinmunidad/efectos de los fármacos , Glicoproteínas de Membrana/farmacología , Linfocitos T/efectos de los fármacos , Adenoviridae/genética , Traslado Adoptivo , Animales , Artritis Experimental/inmunología , Artritis Experimental/patología , Autoinmunidad/inmunología , Muerte Celular/inmunología , Citotoxicidad Inmunológica , Proteína Ligando Fas , Femenino , Activación de Linfocitos , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Neoplasias Experimentales/inmunología , Linfocitos T/inmunología
15.
Cancer Biol Ther ; 2(5): 526-31, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14614320

RESUMEN

Silibinin, derived from milk thistle extract, has been shown to inhibit growth factor receptor-mediated mitogenic and cell survival signaling, and to alter cell cycle regulators. Alteration in pathways regulating cell growth likely account for silibinin's inhibition of tumor growth. Since the epidermal growth factor receptor (EGFR) is a key regulator in cell signaling pathways, in the present study we directly tested the hypothesis that the EGFR plays a key role in mediating silibinin cytotoxicity to cancer cells. We generated a cell line, 9L-EGFR, which stably expressed human EGFR; the parental rat glioma cell line, 9L, does not contain endogenous EGFR message or protein. Our results show that expression of EGFR was both necessary and sufficient for conferring toxicity in response to silibinin in 9L-EGFR cells. Addition of silibinin was shown to inhibit EGFR activation by EGF in 9L-EGFR cells. These studies support the hypothesis that silibinin toxicity to cancer cells involves the EGFR signaling pathway. The findings presented here provide a rationale for understanding the growth inhibition effect of silibinin in cancer cells, and warrant further investigation into the effect of silibinin on specific pathways of cell signaling mediated by the EGF receptor.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Receptores ErbB/fisiología , Glioma/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Silimarina/toxicidad , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , División Celular , Supervivencia Celular/efectos de los fármacos , Factor de Crecimiento Epidérmico/metabolismo , Glioma/metabolismo , Glioma/patología , Humanos , Silybum marianum , Fosforilación/efectos de los fármacos , Ratas , Silibina , Transfección , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo , Ensayo de Tumor de Célula Madre
16.
Proc Natl Acad Sci U S A ; 99(12): 8031-6, 2002 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-12060749

RESUMEN

The envelope glycoprotein (Env) of HIV-1 is incorporated into virions that bud from the cell surface of infected T cells. With immunofluorescence microscopy and subcellular membrane fractionation techniques, the intracellular fate of Env in the secretory pathway of HIV-1-infected T cells was evaluated. Rather than trafficking constitutively from the Golgi to the cell surface, Env is directed to intracellular CTLA-4-containing granules, whose recruitment to the cell surface is regulated. The use of the regulated pathway for intracellular Env storage before virion assembly holds implications for the staging of Env exposure at the cell surface of infected cells and of coordinating HIV virion assembly.


Asunto(s)
Antígenos de Diferenciación/metabolismo , VIH-1/genética , Inmunoconjugados , Vesículas Secretoras/fisiología , Proteínas del Envoltorio Viral/genética , Abatacept , Antígenos CD , Western Blotting , Antígeno CTLA-4 , Fraccionamiento Celular/métodos , Línea Celular , Membrana Celular/ultraestructura , Membrana Celular/virología , Centrifugación por Gradiente de Densidad/métodos , Regulación Viral de la Expresión Génica , Aparato de Golgi/ultraestructura , Aparato de Golgi/virología , Humanos , Inmunohistoquímica , Vesículas Secretoras/ultraestructura , Vesículas Secretoras/virología , Linfocitos T
17.
J Biol Chem ; 277(19): 16505-11, 2002 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-11861638

RESUMEN

Extracellular superoxide dismutase (EC-SOD) is an antioxidant enzyme that attenuates brain and lung injury from oxidative stress. A polybasic region in the carboxyl terminus distinguishes EC-SOD from other superoxide dismutases and determines EC-SOD's tissue half-life and affinity for heparin. There are two types of EC-SOD that differ based on the presence or absence of this heparin-binding region. It has recently been shown that proteolytic removal of the heparin-binding region is an intracellular event (Enghild, J. J., Thogersen, I. B., Oury, T. D., Valnickova, Z., Hojrup, P., and Crapo, J. D. (1999) J. Biol. Chem. 274, 14818-14822). By using mammalian cell lines, we have now determined that removal of the heparin-binding region occurs after passage through the Golgi network but before being secreted into the extracellular space. Specific protease inhibitors and overexpression of intracellular proteases implicate furin as a processing protease. In vitro experiments using furin and purified EC-SOD suggest that furin proteolytically cleaves EC-SOD in the middle of the polybasic region and then requires an additional carboxypeptidase to remove the remaining lysines and arginines. A mutation in Arg(213) renders EC-SOD resistant to furin processing. These results indicate that furin-dependent processing of EC-SOD is important for determining the tissue distribution and half-life of EC-SOD.


Asunto(s)
Aparato de Golgi/metabolismo , Heparina/metabolismo , Subtilisinas/metabolismo , Superóxido Dismutasa/metabolismo , Secuencia de Aminoácidos , Animales , Arginina/química , Western Blotting , Brefeldino A/farmacología , Células CHO , Línea Celular , Cricetinae , Electroforesis en Gel de Poliacrilamida , Inhibidores Enzimáticos/farmacología , Furina , Glicina/química , Heparina/química , Humanos , Espectrometría de Masas , Ratones , Datos de Secuencia Molecular , Mutación , Estrés Oxidativo , Pruebas de Precipitina , Unión Proteica , Estructura Terciaria de Proteína , Inhibidores de la Síntesis de la Proteína/farmacología , Ratas , Subtilisinas/química , Temperatura , Factores de Tiempo , Distribución Tisular , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...