Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Cancer ; 147(9): 2515-2525, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32488909

RESUMEN

Antiangiogenic drugs are potentially a useful supplement to neoadjuvant chemotherapy for a subgroup of patients with human epidermal growth factor receptor 2 (HER2) negative breast cancer, but reliable biomarkers for improved response are lacking. Here, we report on a randomized phase II clinical trial to study the added effect of bevacizumab in neoadjuvant chemotherapy with FEC100 (5-fluorouracil, epirubicin and cyclophosphamide) and taxanes (n = 132 patients). Gene expression from the tumors was obtained before neoadjuvant treatment, and treatment response was evaluated by residual cancer burden (RCB) at time of surgery. Bevacizumab increased the proportion of complete responders (RCB class 0) from 5% to 20% among patients with estrogen receptor (ER) positive tumors (P = .02). Treatment with bevacizumab was associated with improved 8-year disease-free survival (P = .03) among the good responders (RCB class 0 or I). Patients treated with paclitaxel (n = 45) responded better than those treated with docetaxel (n = 21; P = .03). Improved treatment response was associated with higher proliferation rate and an immune phenotype characterized by high presence of classically activated M1 macrophages, activated NK cells and memory activated CD4 T cells. Treatment with bevacizumab increased the number of adverse events, including hemorrhage, hypertension, infection and febrile neutropenia, but despite this, the ECOG status was not affected.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Bevacizumab/farmacología , Neoplasias de la Mama/terapia , Terapia Neoadyuvante/métodos , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Bevacizumab/uso terapéutico , Mama/citología , Mama/patología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Quimioterapia Adyuvante/métodos , Ciclofosfamida/farmacología , Ciclofosfamida/uso terapéutico , Supervivencia sin Enfermedad , Epirrubicina/farmacología , Epirrubicina/uso terapéutico , Femenino , Fluorouracilo/farmacología , Fluorouracilo/uso terapéutico , Estudios de Seguimiento , Humanos , Células Asesinas Naturales/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Macrófagos/inmunología , Mastectomía , Persona de Mediana Edad , Neoplasia Residual , Noruega/epidemiología , Receptor ErbB-2/análisis , Receptor ErbB-2/metabolismo , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
2.
Biomed Microdevices ; 21(2): 39, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30949852

RESUMEN

Effective migration of dendritic cells into the lymphatic system organs is the prerequisite for a functional dendritic cell vaccine. We have previously developed a porous silicon microparticle (PSM)-based therapeutic dendritic cell vaccine (Nano-DC vaccine) where PSM serves both as the vehicle for antigen peptides and an adjuvant. Here, we analyzed parameters that determined dendritic cell uptake of PSM particles and Nano-DC vaccine accumulation in lymphatic tissues in a murine model of HER2-positive breast cancer. Our study revealed a positive correlation between sphericity of the PSM particles and their cellular uptake by circulating dendritic cells. In addition, the intravenously administered vaccines accumulated more in the spleens and inguinal lymph nodes, while the intradermally inoculated vaccines got enriched in the popliteal lymph nodes. Furthermore, mice with large tumors received more vaccines in the lymph nodes than those with small to medium size tumors. Information from this study will provide guidance on design and optimization of future therapeutic cancer vaccines.


Asunto(s)
Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/metabolismo , Células Dendríticas/metabolismo , Nanomedicina , Animales , Transporte Biológico , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/farmacocinética , Línea Celular Tumoral , Células Dendríticas/inmunología , Ratones , Microesferas , Fagocitos/inmunología , Silicio/química , Distribución Tisular , Carga Tumoral/inmunología
3.
Prostate ; 79(1): 9-20, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30073695

RESUMEN

BACKGROUND: Several lines of evidence suggest effects of dietary fat on prostate cancer (PCa) development and progression. Targeting omega (ω)-3:ω6 fatty acids (FA) ratio could be beneficial against PCa by favorably modulating inflammation. Here, we studied the effects of ω3- and ω6-enriched diets on prostate tumor growth and inflammatory response in androgen-deprived and non-deprived conditions. METHODS: Immune-competent eugonadal and castrated C57BL/6 mice were injected with TRAMP-C2 prostate tumor cells and daily fed with ω3- or ω6-enriched diet. FA and cytokine profiles were measured in blood and tumors using gas chromatography and multiplex immunoassay, respectively. Immune cell infiltration in tumors was profiled by multicolor flow cytometry. RESULTS: ω3-enriched diet decreased prostate TRAMP-C2 tumor growth in immune-competent eugonadal and castrated mice. Cytokines associated with Th1 immune response (IL-12 [p70], IFN-γ, GM-CSF) and eosinophil recruitment (eotaxin-1, IL-5, and IL-13) were significantly elevated in tumors of ω3-fed mice. Using in vitro experiments, we confirmed ω3 FA-induced eotaxin-1 secretion by tumor cells and that eotaxin-1 secretion was regulated by androgens. Analysis of immune cell infiltrating tumors showed no major difference of immune cells' abundance between ω3- and ω6-enriched diets. CONCLUSIONS: ω3-enriched diet reduces prostate tumor growth independently of androgen levels. ω3 FA can inhibit tumor cell growth and induce a local anti-tumor inflammatory response. These findings warrant further examination of dietary ω3's potential to slow down the progression of androgen-sensitive and castrate-resistant PCa by modulating immune cell function in tumors.


Asunto(s)
Progresión de la Enfermedad , Ácidos Grasos Omega-3/administración & dosificación , Inmunidad Celular/inmunología , Orquiectomía , Neoplasias de la Próstata/dietoterapia , Neoplasias de la Próstata/inmunología , Animales , Quimiocina CCL11/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Orquiectomía/tendencias , Neoplasias de la Próstata/patología , Carga Tumoral/inmunología , Células Tumorales Cultivadas
4.
J Immunol Res ; 2018: 3431782, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29967800

RESUMEN

Lycium barbarum polysaccharide (LBP) is isolated from the fruit of Chinese herbal Lycium barbarum. Previous studies had demonstrated that LBP could inhibit tumor growth and enhance the immunity in mice. However, the effect of LBP on systemic and local immune responses in vivo, especially on phenotypic and functional changes of T cells, is still largely unknown. In the present study, we investigated the effects of LBP on systemic and local T cell-dependent antitumor immune responses in H22 tumor-bearing mice. The results showed that LBP could inhibit the solid tumor growth in mice, but showed little effect on the body weight or spleen index. Furthermore, LBP could maintain high levels of T cells in peripheral blood (PB), tumor draining lymph node (TDLN), and tumor tissue, prevent the increase of Tregs while promote infiltration of CD8+ T cells in tumor tissue, inhibit the production of TGF-ß1 and IL-10 in serum, decrease the exhaustion phenotype of T cells, and maintain cytotoxicity of lymphocytes. Taken together, our results demonstrated that LBP simultaneously induced systemic and local immune responses in H22 tumor-bearing mice by alleviating immunosuppression and maintaining antitumor immune responses in mice.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Inmunidad/efectos de los fármacos , Neoplasias/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología
5.
Sci Rep ; 7: 43796, 2017 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-28272545

RESUMEN

Licorice has been used as herbal medicine and natural sweetener. Here, we prepared Glycyrrhiza uralensis water extract (GUWE) and investigated the effect of GUWE on the maturation and function of dendritic cells (DCs) and its adjuvant effect on DC-based vaccine. We observed that GUWE dose-dependently promoted DC maturation and cytokine secretion through TLR4 signaling pathway. The capacity of DC to stimulate allogenic splenocyte proliferation was also enhanced by GUWE treatment. Compared with control group, GUWE treated DCs pulsed with human papillomavirus (HPV)-16 E6/E7 peptides significantly inhibited the tumor growth in both early and late therapeutic groups. In early therapeutic group, the frequencies of induced regulatory T cells (iTregs: CD4+CD25-Fopx3+) and CD4+ and CD8+ T cells were significantly decreased and increased, respectively. HPV-16-specific CD8+ T cell responses were significantly induced and negatively correlated with iTreg frequencies and tumor weight. These results indicated the immunoregulatory activities of licorice.


Asunto(s)
Vacunas contra el Cáncer/farmacología , Diferenciación Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Glycyrrhiza uralensis/química , Neoplasias Experimentales/tratamiento farmacológico , Extractos Vegetales/farmacología , Alphapapillomavirus/inmunología , Animales , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Diferenciación Celular/inmunología , Línea Celular Transformada , Células Dendríticas/inmunología , Femenino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias Experimentales/inmunología , Proteínas Oncogénicas Virales/inmunología , Extractos Vegetales/química , Extractos Vegetales/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología , Agua/química
6.
Cancer Gene Ther ; 24(1): 13-19, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27934883

RESUMEN

The incidence of melanoma in the United States continues to rise, with metastatic lesions notoriously recalcitrant to therapy. There are limited effective treatment options available and a great need for more effective therapies that can be rapidly integrated in the clinic. In this study, we demonstrate that the combination of RGD-targeted adeno-associated virus phage (RGD-AAVP-TNF) with hypofractionated radiation therapy results in synergistic inhibition of primary syngeneic B16 melanoma in a C57 mouse model. Furthermore, this combination appeared to modify the tumor microenvironment, resulting in decreased Tregs in the draining LN and increased tumor-associated macrophages within the primary tumor. Finally, there appeared to be a reduction in metastatic potential and a prolongation of overall survival in the combined treatment group. These results indicate the use of targeted TNF gene therapy vector with radiation treatment could be a valuable treatment option for patients with metastatic melanoma.


Asunto(s)
Dependovirus/genética , Dependovirus/metabolismo , Vectores Genéticos/genética , Melanoma/genética , Melanoma/patología , Oligopéptidos/metabolismo , Factor de Necrosis Tumoral alfa/genética , Animales , Línea Celular Tumoral , Terapia Combinada , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Melanoma/metabolismo , Melanoma/terapia , Melanoma Experimental , Ratones , Metástasis de la Neoplasia , Neovascularización Patológica/genética , Neovascularización Patológica/inmunología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/terapia , Radioterapia/métodos , Radioterapia Guiada por Imagen , Análisis de Supervivencia , Resultado del Tratamiento , Carga Tumoral/genética , Carga Tumoral/inmunología , Carga Tumoral/efectos de la radiación , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
7.
Integr Cancer Ther ; 15(3): 368-75, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26699805

RESUMEN

Objective Both the Chinese herbal compound Songyou Yin (SYY) and swimming exercise have been shown to have protective effects against liver cancer in animal models. In this study, we investigated whether SYY and moderate swimming (MS) have enhanced effect on suppressing progression of liver cancer by immunomodulation. Methods C57BL/6 mice were transplanted with Hepa1-6 murine liver cancer cell lines and received treatment with SYY alone or SYY combined with MS. The green fluorescent protein (GFP)-positive metastatic foci in lungs were imaged with a stereoscopic fluorescence microscope. Flow cytometry was used to test the proportion of CD4 +, CD8 + T cells in peripheral blood and the proportions of CD4 + CD25 + Foxp3 + Treg cells in peripheral blood, spleen, and tumor tissues. Cytokine transforming growth factor (TGF)-ß1 level in serum was detected by ELISA. Results SYY plus MS significantly suppressed the growth and lung metastasis of liver cancer and prolonged survival in tumor-burdened mice. SYY plus MS markedly raised the CD4 to CD8 ratio in peripheral blood and lowered the serum TGF-ß1 level and the proportions of Treg cells in peripheral blood, spleen, and tumor tissue. The effects of the combined intervention were significantly superior to SYY or MS alone. Conclusion The combined application of SYY and MS exerted an enhanced effect on suppressing growth and metastasis of liver cancer by strengthening immunity.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/inmunología , Medicamentos Herbarios Chinos/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/inmunología , Natación/fisiología , Animales , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Citocinas/inmunología , Citocinas/metabolismo , Citometría de Flujo/métodos , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta1/inmunología , Factor de Crecimiento Transformador beta1/metabolismo , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología
8.
Cancer Immunol Immunother ; 62(8): 1369-80, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23715927

RESUMEN

Combinatory strategies are becoming of increasing interest in cancer immunotherapy. Costimulation by individual members of the immunoglobulin-like (Ig)- and TNF superfamily have already shown promising antitumor potential, thus prompting the exploration of their synergistic abilities in combinatorial approaches. Here, we pursued a targeted strategy with antibody-fusion proteins composed of a tumor-directed antibody and the extracellular domain of the costimulatory ligand B7.1, 4-1BBL, OX40L, GITRL or LIGHT, respectively. Costimulatory activity was assessed in an experimental setting where initial T cell activation was induced by a bispecific antibody (tumor-related antigen × CD3). Advantage of combined targeted costimulation was shown for either B7.1 or 4-1BBL with OX40L, GITRL, LIGHT and 4-1BBL in terms of T cell proliferation and IFN-γ release. Since encouraging results were obtained by the combination of B7.1 and 4-1BBL, we adapted the model system for a time-shift setting. Here, enhanced proliferation and granzyme B expression as well as reduced PD-1 expression on the T cell population demonstrated the benefit of costimulation-assisted restimulation. Finally, the antitumor potential of this combinatorial setting was confirmed in vivo in a lung metastasis mouse model. Thus, combinatorial approaches with costimulatory antibody-ligand fusion proteins seem a promising strategy to be further investigated for cancer immunotherapy.


Asunto(s)
Inmunoterapia/métodos , Neoplasias Experimentales/terapia , Proteínas Recombinantes de Fusión/uso terapéutico , Ligando 4-1BB/genética , Ligando 4-1BB/inmunología , Animales , Anticuerpos Biespecíficos/genética , Anticuerpos Biespecíficos/inmunología , Antígeno B7-1/genética , Antígeno B7-1/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Evaluación Preclínica de Medicamentos , Granzimas/inmunología , Granzimas/metabolismo , Células HEK293 , Humanos , Interferón gamma/inmunología , Interferón gamma/metabolismo , Ligandos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Receptor de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacocinética , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología
9.
J Immunother ; 34(8): 581-7, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21904218

RESUMEN

LIGHT is a tumor necrosis factor superfamily ligand that is considered as a promising candidate for cancer therapy. It has a potent antitumor activity through establishing lymphoid-like tissues inside tumor sites and recruiting naive T cells into the tumor. In this study, we examined the possibility of antitumor activity by expressing LIGHT in cervical cancer (CC) model. A recombinant adeno-associated virus (AAV) vector was chosen for the transfer, based on its transfection efficiency and lack of detectable pathology. In vitro transfer of recombinant AAV vector expressing LIGHT (AAV-LIGHT) stimulated T-lymphocyte proliferation and activation. AAV-mediated gene transfer of LIGHT by intratumoral injection exerted a very potent antitumor effect against preexisting TC-1 cell CC in C57BL/6 mice. This study confirmed that AAV-LIGHT regressed tumor growth by activating cytotoxic T lymphocyte, enhancing infiltration of inflammatory cells in tumor and increasing stimulatory cytokine expression in tumor microenvironment. Therefore, AAV-LIGHT therapy might have potential utility for the treatment of CC.


Asunto(s)
Transformación Celular Neoplásica , Dependovirus/genética , Vectores Genéticos/genética , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Neoplasias del Cuello Uterino/terapia , Animales , Células CHO , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Cricetinae , Modelos Animales de Enfermedad , Femenino , Técnicas de Transferencia de Gen , Células HEK293 , Humanos , Activación de Linfocitos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Bazo/inmunología , Bazo/metabolismo , Transducción Genética , Carga Tumoral/genética , Carga Tumoral/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Neoplasias del Cuello Uterino/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Cell Mol Immunol ; 7(4): 296-305, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20418899

RESUMEN

Here, we investigated the antitumor effect of adenovirus-mediated gene transfer of LIGHT, the tumor-necrosis factor (TNF) superfamily member also known as TNFSF14, in the murine A20 B-cell lymphoma. LIGHT gene modification resulted in upregulated expression of Fas and the accessory molecule--intercellular adhesion molecule-1 (ICAM-1) on A20 cells and led to enhanced A20 cell apoptosis. LIGHT-modified A20 cells effectively stimulated the proliferation of T lymphocytes and interferon (IFN)-gamma production in vitro. Immunization of BALB/c mice with a LIGHT-modified A20 cell vaccine efficiently elicited protective immunity against challenge with the parental tumor cell line. Adenovirus-mediated gene transfer of LIGHT by intratumoral injection exerted a very potent antitumor effect against pre-existing A20 cell lymphoma in BALB/c mice. This adenovirus-mediated LIGHT therapy induced substantial splenic natural killer (NK) and cytotoxic T lymphocyte (CTL) activity, enhanced tumor infiltration by inflammatory cells and increased chemokine expression of CC chemokine ligand 21 (CCL21), IFN-inducible protein-10 (IP-10) and monokine induced by IFN-gamma (Mig) from tumor tissues. Thus, adenovirus-mediated LIGHT therapy might have potential utility for the prevention and treatment of B-cell lymphoma.


Asunto(s)
Adenoviridae/genética , Terapia Genética , Inmunidad/inmunología , Linfoma de Células B/genética , Linfoma de Células B/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/uso terapéutico , Animales , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Quimiocinas/metabolismo , Femenino , Técnicas de Transferencia de Gen , Humanos , Inmunización , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Linfoma de Células B/prevención & control , Linfoma de Células B/terapia , Ratones , Ratones Endogámicos BALB C , Bazo/citología , Bazo/inmunología , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología , Carga Tumoral/inmunología
11.
Immunol Invest ; 38(8): 820-38, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19860591

RESUMEN

The influence of tumor burden on the generation of tumor antigen-specific cytotoxic T-lymphocytes (CTL) was investigated in a phase I/II clinical adoptive immunotherapy trial. Four previously treated metastatic breast cancer patients, two with macroscopic disease and two with no evidence of disease, in complete remission (CR), were enrolled. Each apheretic peripheral blood mononuclear cell (PBMC) sample was stimulated twice with MUC-1 before infusion back into the patients. CTL responses against MCF-7 cell line and cytokine production were measured before infusion. Patients received two monthly CTL infusions and were monitored for toxicity, tumor response as well as tumor marker levels. The CTL generated from patients with high tumor burdens had less cytokine production and lower cytotoxicity of MCF-7 than the CTL of patients in CR. The differences between the two groups were observed after the two MUC1 in vitro stimulations of the cells obtained in first apheresis. This difference increased after the two MUC1 stimulations of the cells obtained in the second apheresis. The cytotoxicity function was sustained from the first infusion to the second apheresis only for the patients in CR. This suggests that tumor burden had an inverse effect on the function of the generated CTL.


Asunto(s)
Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Inmunoterapia Adoptiva , Transfusión de Leucocitos , Mucina-1/inmunología , Linfocitos T Citotóxicos/metabolismo , Carga Tumoral/inmunología , Biomarcadores de Tumor/metabolismo , Transfusión de Sangre Autóloga , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Citocinas/biosíntesis , Citotoxicidad Inmunológica , Femenino , Humanos , Leucocitos Mononucleares , Persona de Mediana Edad , Metástasis de la Neoplasia , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/patología
12.
Eur Urol ; 56(1): 123-31, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18550267

RESUMEN

BACKGROUND AND OBJECTIVE: In a phase-III trial it was recently shown that an adjuvant renal cell carcinoma (RCC) vaccine (Reniale) reduces the risk of tumour progression following nephrectomy. This clinical trial focused on efficacy and did not investigate end-points relating to mode-of-action of the vaccine. In a murine model we investigated mode-of-action, efficacy, and safety of a homologous RENCA cell-based vaccine. DESIGN, SETTING, AND PARTICIPANTS: Six groups with 12 BALB/c mice per group received five vaccinations (lysate of 1x10(6)-1x10(7) RENCA cells, manufactured with or without prior IFN-gamma incubation) at 3-wk intervals before tumour transplantation and one vaccination 14 d afterwards. Controls (12 mice) received only solvent. All mice were sacrificed 21 d after tumour transplantation. MEASUREMENTS: Animal welfare, tumour growth, number of metastases, and the presence of cytotoxic T-lymphocytes as determined by a (51)chromium-release assay. Adoptive immune transfer experiments (vaccination of nine mice with the RENCA vaccine or saline and transfer of serum, spleen cells, and CD4 and/or CD8 depleted spleen cells into five recipient mice each) were carried out to demonstrate involvement of different immune mechanisms. RESULTS: All controls developed a renal tumour, compared to 7/72 animals (9.7%) in the vaccine groups. The mean number of lung metastases was 100 (range 3-750) in controls and 4 (range 0-196) in the vaccine groups, respectively. Tumour uptake and number of metastases were not related to the vaccine dose. The (51)chromium-release assay confirmed a significant tumour-specific cytolytic activity and marginally increased NK activity of splenocytes from vaccinated mice against RENCA cells compared to controls. Adoptive immune transfer experiments showed that the antitumoural effective immune mechanisms are cell-based. CONCLUSIONS: We could demonstrate the mode-of-action, efficacy, and safety of a homologous tumour vaccine in a RENCA model. These findings support the positive results from a phase-III trial with Reniale.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Carcinoma de Células Renales/prevención & control , Neoplasias Renales/prevención & control , Inmunidad Adaptativa , Traslado Adoptivo , Animales , Vacunas contra el Cáncer/farmacología , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/patología , Carcinoma de Células Renales/secundario , Carcinoma de Células Renales/cirugía , Línea Celular Tumoral , Terapia Combinada , Modelos Animales de Enfermedad , Femenino , Neoplasias Renales/inmunología , Neoplasias Renales/patología , Neoplasias Renales/cirugía , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Recuento de Linfocitos , Ratones , Ratones Endogámicos BALB C , Nefrectomía , Bazo/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/patología , Carga Tumoral/inmunología
13.
Clin Cancer Res ; 14(12): 3966-74, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18559619

RESUMEN

PURPOSE: Adoptive cellular immunotherapy is a promising approach to eradicate established tumors. However, a significant hurdle in the success of cellular immunotherapy involves recently identified mechanisms of immune suppression on cytotoxic T cells at the effector phase. Transforming growth factor-beta (TGF-beta) is one of the most important of these immunosuppressive factors because it affects both T-cell and macrophage functions. We thus hypothesized that systemic blockade of TGF-beta signaling combined with adoptive T-cell transfer would enhance the effectiveness of the therapy. EXPERIMENTAL DESIGN: Flank tumors were generated in mice using the chicken ovalbumin-expressing thymoma cell line, EG7. Splenocytes from transgenic OT-1 mice (whose CD8 T cells recognize an immunodominant peptide in chicken ovalbumin) were activated in vitro and adoptively transferred into mice bearing large tumors in the presence or absence of an orally available TGF-beta receptor-I kinase blocker (SM16). RESULTS: We observed markedly smaller tumors in the group receiving the combination of SM16 chow and adoptive transfer. Additional investigation revealed that TGF-beta receptor blockade increased the persistence of adoptively transferred T cells in the spleen and lymph nodes, increased numbers of adoptively transferred T cells within tumors, increased activation of these infiltrating T cells, and altered the tumor microenvironment with a significant increase in tumor necrosis factor-alpha and decrease in arginase mRNA expression. CONCLUSIONS: We found that systemic blockade of TGF-beta receptor activity augmented the antitumor activity of adoptively transferred T cells and may thus be a useful adjunct in future clinical trials.


Asunto(s)
Compuestos de Azabiciclo/uso terapéutico , Inmunoterapia Adoptiva , Neoplasias/terapia , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Animales , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Terapia Combinada , Evaluación Preclínica de Medicamentos , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Trasplante de Neoplasias , Neoplasias/inmunología , Neoplasias/patología , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología , Células Tumorales Cultivadas
14.
J Natl Cancer Inst ; 97(20): 1516-24, 2005 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-16234565

RESUMEN

BACKGROUND: Epithelial ovarian cancer often develops resistance to standard treatments, which is a major reason for the high mortality associated with the disease. We examined the efficacy of a treatment regimen that combines immunotherapy to block the activity of epidermal growth factor receptor (EGFR), overexpression of which is associated with the development of resistant ovarian cancer, and photodynamic therapy (PDT), a mechanistically distinct photochemistry-based modality that is effective against chemo- and radioresistant ovarian tumors. METHODS: We tested a combination regimen consisting of C225, a monoclonal antibody that inhibits the receptor tyrosine kinase activity of EGFR, and benzoporphyrin derivative monoacid A (BPD)-based PDT in a mouse model of human ovarian cancer. Therapeutic efficacy was evaluated in acute treatment response and survival studies that used 9-19 mice per group. Analysis of variance and Wilcoxon statistics were used to analyze the data. All statistical tests were two-sided. RESULTS: Mice treated with PDT + C225 had the lowest mean tumor burden compared with that in the no-treatment control mice (mean percent tumor burden = 9.8%, 95% confidence interval [CI] = 2.3% to 17.3%, P < .001). Mean percent tumor burden for mice treated with C225 only or PDT only was 66.6% (95% CI = 58.7% to 74.4%, P < .001) and 38.2% (95% CI = 29.3% to 47.0%, P < .001), respectively. When compared with PDT only or C225 only, PDT + C225 produced synergistic reductions in mean tumor burden (P < .001, analysis of variance) and improvements in survival (P = .0269, Wilcoxon test). Median survival was approximately threefold greater for mice in the PDT + C225 group than for mice in the no-treatment control group (80 days versus 28 days), and more mice in the PDT + C225 group were alive at 180 days (3/9; 33% [95% CI = 7% to 70%]) than mice in the C225-only (0/12; 0% [95% CI = 0% to 22%]) or PDT-only (1/10; 10% [95% CI = 0.2% to 44%]) groups. CONCLUSION: A mechanistically nonoverlapping combination modality consisting of receptor tyrosine kinase inhibition with C225 and BPD-PDT is well tolerated, effective, and synergistic in mice.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Carcinoma/terapia , Receptores ErbB/efectos de los fármacos , Fotorradiación con Hematoporfirina , Inmunoterapia , Terapia por Luz de Baja Intensidad , Neoplasias Ováricas/terapia , Neoplasias Peritoneales/tratamiento farmacológico , Porfirinas/farmacología , Análisis de Varianza , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales Humanizados , Antineoplásicos/farmacología , Carcinoma/tratamiento farmacológico , Carcinoma/inmunología , Carcinoma/metabolismo , Carcinoma/secundario , Cetuximab , Terapia Combinada , Intervalos de Confianza , Sinergismo Farmacológico , Receptores ErbB/inmunología , Receptores ErbB/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/inmunología , Fotorradiación con Hematoporfirina/métodos , Humanos , Inmunoterapia/métodos , Inyecciones Intraperitoneales , Terapia por Luz de Baja Intensidad/métodos , Ratones , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Neoplasias Peritoneales/inmunología , Neoplasias Peritoneales/metabolismo , Neoplasias Peritoneales/patología , Neoplasias Peritoneales/secundario , Fármacos Fotosensibilizantes/farmacología , Porfirinas/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Trasplante Heterólogo , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA