Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Int J Cancer ; 133(3): 624-36, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23364915

RESUMEN

Certain chemotherapeutic regimens trigger cancer cell death while inducing dendritic cell maturation and subsequent immune responses. However, chemotherapy-induced immunogenic cell death (ICD) has thus far been restricted to select agents. In contrast, several chemotherapeutic drugs modulate antitumor immune responses, despite not inducing classic ICD. In addition, in many cases tumor cells do not die after treatment. Here, using docetaxel, one of the most widely used cancer chemotherapeutic agents, as a model, we examined phenotypic and functional consequences of tumor cells that do not die from ICD. Docetaxel treatment of tumor cells did not induce ATP or high-mobility group box 1 (HMGB1) secretion, or cell death. However, calreticulin (CRT) exposure was observed in all cell lines examined after chemotherapy treatment. Killing by carcinoembryonic antigen (CEA), MUC-1, or PSA-specific CD8(+) CTLs was significantly enhanced after docetaxel treatment. This killing was associated with increases in components of antigen-processing machinery, and mediated largely by CRT membrane translocation, as determined by functional knockdown of CRT, PERK, or CRT-blocking peptide. A docetaxel-resistant cell line was selected (MDR-1(+), CD133(+)) by continuous exposure to docetaxel. These cells, while resistant to direct cytostatic effects of docetaxel, were not resistant to the chemomodulatory effects that resulted in enhancement of CTL killing. Here, we provide an operational definition of "immunogenic modulation," where exposure of tumor cells to nonlethal/sublethal doses of chemotherapy alters tumor phenotype to render the tumor more sensitive to CTL killing. These observations are distinct and complementary to ICD and highlight a mechanism whereby chemotherapy can be used in combination with immunotherapy.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Citotoxicidad Inmunológica , Linfocitos T Citotóxicos/inmunología , Taxoides/farmacología , Adenosina Trifosfato/biosíntesis , Apoptosis/inmunología , Neoplasias de la Mama/terapia , Calreticulina/análisis , Antígeno Carcinoembrionario/inmunología , Línea Celular Tumoral , Neoplasias Colorrectales/terapia , Células Dendríticas/inmunología , Docetaxel , Femenino , Proteína HMGB1/biosíntesis , Humanos , Inmunoterapia , Masculino , Mucina-1/inmunología , Antígeno Prostático Específico/inmunología , Neoplasias de la Próstata/terapia , Interferencia de ARN , ARN Interferente Pequeño , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/metabolismo , eIF-2 Quinasa/genética
2.
Clin Cancer Res ; 14(11): 3536-44, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18519787

RESUMEN

PURPOSE: Taxanes comprise some of the most widely used cancer chemotherapeutic agents. Members of this drug family, including docetaxel, are commonly used to treat breast, prostate, and lung cancers, among others. This study was designed to determine if this taxane has the ability to modulate components of the immune system independent of antitumor activity and to investigate the potential synergistic activities of the combination of docetaxel and vaccine therapy. EXPERIMENTAL DESIGN: We examined the in vivo effects of docetaxel on immune-cell subsets and on the function of CD4+, CD8+, and regulatory T-cell (Treg) populations in response to antigen-specific vaccination. We also examined the antitumor effects of the combination of docetaxel and vaccine in a preclinical model in which docetaxel has no observable effect on tumor growth. RESULTS: These studies show for the first time that (a) docetaxel modulates CD4+, CD8+, CD19+, natural killer cell, and Treg populations in non-tumor-bearing mice; (b) unlike cyclophosphamide, docetaxel does not inhibit the function of Tregs; (c) docetaxel enhances CD8+ but not CD4+ response to CD3 cross-linking; (d) docetaxel given after vaccination provides optimal enhancement of immune response to recombinant viral vaccines; (e) docetaxel combined with recombinant viral vaccine is superior to either agent alone at reducing tumor burden; and (f) docetaxel plus vaccine increases antigen-specific T-cell responses to antigen in the vaccine, as well as to cascade antigens derived from the tumor. CONCLUSIONS: These findings suggest potential clinical benefit for the combined use of docetaxel and recombinant cancer vaccines.


Asunto(s)
Antineoplásicos/administración & dosificación , Vacunas contra el Cáncer/uso terapéutico , Neoplasias Experimentales/terapia , Linfocitos T/efectos de los fármacos , Taxoides/administración & dosificación , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Antígeno Carcinoembrionario/genética , Terapia Combinada , Docetaxel , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias Experimentales/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Vacunas Sintéticas/uso terapéutico
3.
Front Biosci ; 12: 4900-10, 2007 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-17569618

RESUMEN

Since its discovery more than a hundred years ago, radiation has been used to treat cancer. In recent decades, advances in radiation technology have expanded the role and value of radiation in imaging and treating many forms of cancer. Currently, there is a growing interest in combining radiation with other modalities, such as immunotherapy, to treat a broad range of malignancies. This article reviews the use of standard and novel combinations of radiation therapy and immunotherapy to eradicate tumor cells. The combination of radiation therapy and immunotherapy holds particular promise as a strategy for cancer therapeutics for a variety of reasons. First, there is evidence that immunotherapy is most beneficial when employed early in the disease process and in combination with standard therapies. In addition, radiation may act synergistically with immunotherapy to enhance immune responses, inhibit immunosuppression, and/or alter the phenotype of tumor cells, thus rendering them more susceptible to immune-mediated killing. Finally, as monotherapies, both immunotherapy and radiation may be insufficient to eliminate tumor masses. However, following immunization with a cancer vaccine, the destruction of even a small percentage of tumor cells by radiation could result in cross-priming and presentation of tumor antigens to the immune system, thereby potentiating antitumor responses.


Asunto(s)
Inmunoterapia , Neoplasias/terapia , Braquiterapia , Humanos , Metástasis de la Neoplasia , Neoplasias/radioterapia , Radioinmunodetección , Radioinmunoterapia , Radioterapia Adyuvante
4.
Clin Cancer Res ; 12(6): 1897-905, 2006 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-16551875

RESUMEN

PURPOSE: The combination of systemic multiagent chemotherapy (5-fluorouracil + cisplatin) and tumor irradiation is standard of care for head and neck squamous cell carcinoma (HNSCC). Furthermore, it has been shown that sublethal doses of radiation or chemotherapeutic drugs in diverse cancer types may alter the phenotype or biology of neoplastic cells, making them more susceptible to CTL-mediated cytotoxicity. However, little is known about the potential synergistic effect of drug plus radiation on CTL killing. Here, we examined whether the combination of two chemotherapeutics and ionizing radiation enhanced CTL-mediated destruction of HNSCC more so than either modality separately, as well as the basis for the enhanced tumor cell lysis. EXPERIMENTAL DESIGN: Several HNSCC cell lines with distinct biological features were treated with sublethal doses of cisplatin and 5-fluorouracil for 24 hours and with 10-Gy irradiation. Seventy-two hours postirradiation, tumor cells were exposed to an antigen-specific CD8+ CTL directed against carcinoembryonic antigen or MUC-1. RESULTS: In three of three tumor cell lines tested, enhanced CTL activity was observed when the two modalities (chemotherapy and radiation) were combined as compared with target cells exposed to either modality separately. CTL-mediated lysis was MHC restricted and antigen specific and occurred almost entirely via the perforin pathway. Moreover, the combination treatment regimen led to a 50% reduction in Bcl-2 expression whereas single modality treatment had little bearing on the expression of this antiapoptotic gene. CONCLUSIONS: Overall, these results reveal that (a) CTL killing can be enhanced by combining multiagent chemotherapy and radiation and (b) combination treatment enhanced or sensitized HNSCC to the perforin pathway, perhaps by down-regulating Bcl-2 expression. These studies thus form the rational basis for clinical trials of immunotherapy concomitant with the current standard of care of HNSCC.


Asunto(s)
Citotoxicidad Inmunológica/inmunología , Linfocitos T Citotóxicos/inmunología , Antígenos de Neoplasias/análisis , Antígenos de Neoplasias/inmunología , Antineoplásicos/farmacología , Antígeno Carcinoembrionario/análisis , Antígeno Carcinoembrionario/inmunología , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Supervivencia Celular/efectos de la radiación , Cisplatino/farmacología , Pruebas Inmunológicas de Citotoxicidad , Citometría de Flujo , Fluorouracilo/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/patología , Humanos , Molécula 1 de Adhesión Intercelular/análisis , Glicoproteínas de Membrana/análisis , Mucina-1 , Mucinas/análisis , Mucinas/inmunología , Perforina , Proteínas Citotóxicas Formadoras de Poros/análisis , Proteínas Proto-Oncogénicas c-bcl-2/genética , Receptores del Factor de Necrosis Tumoral/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Linfocitos T Citotóxicos/química , Linfocitos T Citotóxicos/citología , Factores de Tiempo
5.
Curr Pharm Des ; 12(3): 351-61, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16454749

RESUMEN

For the immune system to mount an effective antitumor T-cell response, an adequate number of T-cells specific for the antigens expressed by the malignancy must be activated [1]. Since most antigens expressed by tumors are "self"-antigens, tumor antigens often lack endogenous immunogenicity and thus do not sufficiently activate T-cells to levels that can mediate tumor eradication. In addition, virtually all solid tumor cells lack the costimulatory molecules necessary to activate tumor-specific T-cells. Approaches that stimulate immune responses to these tumor antigens have the potential to alter this poor responsiveness. This theory has promoted the use of active immunotherapy to generate immune responses against tumor-associated antigens (TAAs) for the treatment of cancer. As one such vaccine strategy, we have utilized poxviruses as delivery vehicles for TAAs in combination with T-cell costimulatory molecules. Initial studies have demonstrated that the insertion of costimulatory molecule trangenes into viral vectors, along with a TAA transgene, greatly enhances the immune response to the antigen. Using this approach, a TRIad of COstimulatory Molecules (TRICOM; B7-1, ICAM-1 and LFA-3) has been shown to enhance T-cell responses to TAAs to levels far greater than any one or two of the costimulatory molecules in combination. In this article, preclinical findings and recent clinical applications of TRICOM-based vaccines as a cancer immunotherapy are reviewed.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Inmunoterapia/métodos , Neoplasias/terapia , Animales , Ensayos Clínicos como Asunto , Terapia Combinada , Humanos , Neoplasias/fisiopatología , Neoplasias/prevención & control , Neoplasias/radioterapia , Transducción de Señal/efectos de los fármacos , Linfocitos T/inmunología , Vacunación , Vacunas Sintéticas/uso terapéutico
6.
Cancer Res ; 64(21): 7985-94, 2004 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-15520206

RESUMEN

Local radiation of tumor masses is an established modality for the therapy of a range of human tumors. It has recently been recognized that doses of radiation, lower than or equal to those that cause direct cytolysis, may alter the phenotype of target tissue by up-regulating gene products that may make tumor cells more susceptible to T-cell-mediated immune attack. Previously, we demonstrated that radiation increased Fas (CD95) gene expression in carcinoembryonic antigen (CEA)-expressing murine tumor cells, which consequently enhanced their susceptibility to CEA-specific CTL-mediated killing. The present study was designed to determine whether these phenomena also occur with human tumor cells. Here, 23 human carcinoma cell lines (12 colon, 7 lung, and 4 prostate) were examined for their response to nonlytic doses of radiation (10 or 20 Gy). Seventy-two hours postirradiation, changes in surface expression of Fas (CD95), as well as expression of other surface molecules involved in T-cell-mediated immune attack such as intercellular adhesion molecule 1, mucin-1, CEA, and MHC class I, were examined. Twenty-one of the 23 (91%) cell lines up-regulated one or more of these surface molecules postirradiation. Furthermore, five of five irradiated CEA(+)/A2(+) colon tumor cells lines demonstrated significantly enhanced killing by CEA-specific HLA-A2-restricted CD8(+) CTLs compared with nonirradiated counterparts. We then used microarray analysis to broaden the scope of observed changes in gene expression after radiation and found that many additional genes had been modulated. These up-regulated gene products may additionally enhance the tumor cells' susceptibility to T-cell-mediated immune attack or serve as additional targets for immunotherapy. Overall, the results of this study suggest that nonlethal doses of radiation can be used to make human tumors more amenable to immune system recognition and attack and form the rational basis for the combinatorial use of cancer vaccines and local tumor irradiation.


Asunto(s)
Citotoxicidad Inmunológica , Neoplasias/radioterapia , Linfocitos T Citotóxicos/inmunología , Antígeno Carcinoembrionario/análisis , Línea Celular Tumoral , Perfilación de la Expresión Génica , Humanos , Molécula 1 de Adhesión Intercelular/análisis , Mucina-1/análisis , Neoplasias/inmunología , Neoplasias/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Receptor fas/análisis
7.
Cancer Biother Radiopharm ; 29(4): 153-61, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24693958

RESUMEN

We sought to determine if single-dose external beam radiation therapy (EBRT) could modulate the expression signature of T-cell costimulatory and coinhibitory molecules in human prostate cancer (PCa) cell lines in vitro. We investigated the functional impact of irradiated PCa cells with a modulated costimulatory profile on responder T-cell activity. We used three PCa cell lines (DU145, PC3, and LNCaP) and two epithelial cell lines from noncancerous prostate and lung tissue. After 72 hours of EBRT, surface expression of four immunostimulatory molecules (CD70, CD275/ICOSL, CD134L/OX40L, and CD137L/41BBL) and two immunosuppressive markers (CTLA-4/CD152 and PD-L1/CD274) were evaluated by flow cytometry. We evaluated the impact of several radiation doses and the longevity of modulated expression. We examined the functional impact of radiation-induced modulation of cancer cells by cytotoxic T cells (CTL) cytotoxicity and ELISPOT assay for interferon-gamma (IFN-γ) production. Last, we evaluated whether IFN-γ-induced PD-L1 expression could be reversed by EBRT. After 10 Gy EBRT, expression of OX40L and 41BBL increased in all three PCa cell lines; expression of CD70 and ICOSL increased in PC3 cells. Conversely, a decrease in PD-L1 expression in DU145 and PC3 cells was detectable up to 144 hours after EBRT. No PD-L1 was detected in LNCaP. Epithelial cells from normal prostate were not modulated by radiation. CTL cytolytic activity and IFN-γ production were enhanced by interaction with irradiated PCa cells. Finally, EBRT failed to prevent IFN-γ-induced upregulation of PD-L1. We demonstrate that a single dose of EBRT increased surface expression of costimulatory molecules and decreased the expression of coinhibitory molecules in human PCa cell lines. Changes in irradiated tumor cells led to functional enhancement of T-cell activity, despite EBRT failing to reduce IFN-γ-induced expression of PD-L1. These data suggest that combining radiotherapy with T-cell stimulating immunotherapy may be an attractive strategy for cancer treatment.


Asunto(s)
Receptores Coestimuladores e Inhibidores de Linfocitos T/inmunología , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/radioterapia , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/efectos de la radiación , Animales , Línea Celular Tumoral , Humanos , Masculino , Ratones Endogámicos C57BL , Neoplasias de la Próstata/patología , Transducción de Señal
8.
Cancer Immunol Immunother ; 56(12): 1897-910, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17503041

RESUMEN

Systemic IL-2 is currently employed in the therapy of several tumor types, but at the price of often severe toxicities. Local vector mediated delivery of IL-2 at the tumor site may enhance local effector cell activity while reducing toxicity. To examine this, a model using CEA-transgenic mice bearing established CEA expressing tumors was employed. The vaccine regimen was a s.c. prime vaccination with recombinant vaccinia (rV) expressing transgenes for CEA and a triad of costimulatory molecules (TRICOM) followed by i.t. boosting with rF-CEA/TRICOM. The addition of intratumoral (i.t.) delivery of IL-2 via a recombinant fowlpox (rF) IL-2 vector greatly enhanced anti-tumor activity of a recombinant vaccine, resulting in complete tumor regression in 70-80% of mice. The anti-tumor activity was shown to be dependent on CD8(+) cells and NK1.1(+). Cellular immune assays revealed that the addition of rF-IL-2 to the vaccination therapy enhanced CEA-specific tetramer(+) cell numbers, cytokine release and CTL lysis of CEA(+) targets. Moreover, tumor-bearing mice vaccinated with the CEA/TRICOM displayed an antigen cascade, i.e., CD8(+) T cell responses to two other antigens expressed on the tumor and not the vaccine: wild-type p53 and endogenous retroviral antigen gp70. Mice receiving rF-IL-2 during vaccination demonstrated higher avidity CEA-specific, as well as higher avidity gp70-specific, CD8(+) T cells when compared with mice vaccinated without rF-IL-2. These studies demonstrate for the first time that the level and avidity of antigen specific CTL, as well as the therapeutic outcome can be improved with the use of i.t. rF-IL-2 with vaccine regimens.


Asunto(s)
Terapia Genética/métodos , Inmunoterapia/métodos , Interleucina-2/uso terapéutico , Neoplasias/inmunología , Neoplasias/terapia , Linfocitos T/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer , Citometría de Flujo , Viruela Aviar/microbiología , Vectores Genéticos , Sistema Inmunológico , Interleucina-2/metabolismo , Ratones , Ratones Endogámicos C57BL
9.
J Immunol ; 174(10): 5994-6004, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15879092

RESUMEN

Recent studies in both animal models and clinical trials have demonstrated that the avidity of T cells is a major determinant of antitumor and antiviral immunity. In this study, we evaluated several different vaccine strategies for their ability to enhance both the quantity and avidity of CTL responses. CD8(+) T cell quantity was measured by tetramer binding precursor frequency, and avidity was measured by both tetramer dissociation and quantitative cytolytic function. We have evaluated a peptide, a viral vector expressing the Ag transgene alone, with one costimulatory molecule (B7-1), and with three costimulatory molecules (B7-1, ICAM-1, and LFA-3), with anti-CTLA-4 mAb, with GM-CSF, and combinations of the above. We have evaluated these strategies in both a foreign Ag model using beta-galactosidase as immunogen, and in a "self" Ag model, using carcinoembryonic Ag as immunogen in carcinoembryonic Ag transgenic mice. The combined use of several of these strategies was shown to enhance not only the quantity, but, to a greater magnitude, the avidity of T cells generated; a combination strategy is also shown to enhance antitumor effects. The results reported in this study thus demonstrate multiple strategies that can be used in both antitumor and antiviral vaccine settings to generate higher avidity host T cell responses.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Pruebas Inmunológicas de Citotoxicidad/métodos , Activación de Linfocitos/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Adyuvantes Inmunológicos/genética , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Antígenos CD , Antígenos de Diferenciación/inmunología , Antígeno CTLA-4 , Vacunas contra el Cáncer/genética , Antígeno Carcinoembrionario/administración & dosificación , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/inmunología , Línea Celular Tumoral , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Neoplasias del Colon/prevención & control , Terapia Combinada , Femenino , Vectores Genéticos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Unión Proteica/inmunología , beta-Galactosidasa/administración & dosificación , beta-Galactosidasa/genética , beta-Galactosidasa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA