Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Cancer Immunol Immunother ; 60(7): 975-84, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21461842

RESUMEN

MUC1 (CA15-3) and MUC16 (CA125) tumor-associated antigens are upregulated in ovarian cancer and can be detected in patients' sera by standardized tests. We postulated that increased MUC1 and MUC16 antigens augment antibody responses in platinum-resistant ovarian cancer patients and that the frequency and intensity of these responses can be used as immune biomarkers of treatment response and disease outcome. We measured MUC1 and MUC16 tumor expression by immunohistochemistry (IHC), assessed serum antigenic levels and quantitated circulating antibodies by ELISA in a cohort of 28 ovarian cancer patients with platinum-resistant or platinum-refractory ovarian cancer, and treated with intraperitoneal (IP) interleukin-2 (IL-2). MUC1 and MUC16 were overexpressed in tumor samples and showed differential distribution profiles. Serum MUC1 (CA15-3) measurements were elevated in all patients and significantly correlated with increased risk of death (P = 0.003). MUC1-specific IgM and IgG anitbodies were found in 92 and 50% of cases, respectively. Patients with progressive disease had higher mean anti-MUC1 IgG than responders at both early (P = 0.025) and late (P = 0.022) time points during IP IL-2 treatment. Anti-MUC1 IgM antibodies inversely correlated with overall survival at both early (P = 0.052) and late (P = 0.009) time points. In contrast to MUC1, neither soluble MUC16 nor MUC16-specific antibodies were significantly associated with clinical response or overall survival in this study. Increased serum MUC1 and high anti-MUC1 antibody levels are prognostic for poor clinical response and reduced overall survival in platinum-resistant or platinum-refractory ovarian cancer.


Asunto(s)
Anticuerpos Antineoplásicos/sangre , Autoanticuerpos/sangre , Biomarcadores de Tumor/inmunología , Antígeno Ca-125/inmunología , Resistencia a Antineoplásicos , Proteínas de la Membrana/inmunología , Mucina-1/inmunología , Neoplasias Ováricas/inmunología , Adenocarcinoma de Células Claras/tratamiento farmacológico , Adenocarcinoma de Células Claras/inmunología , Estudios de Casos y Controles , Cistadenocarcinoma Seroso/tratamiento farmacológico , Cistadenocarcinoma Seroso/inmunología , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Técnicas para Inmunoenzimas , Inyecciones Intraperitoneales , Interleucina-2/uso terapéutico , Compuestos Organoplatinos/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Pronóstico
2.
Brain Behav Immun ; 25(6): 1187-96, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21426930

RESUMEN

Stress triggers complex response mechanisms designed to recognize and adapt to perturbations in homeostasis. The immune system is highly responsive to stress, although the complete mechanisms linking stress and immune mediators including T lymphocytes, are not fully understood. Stress exerts its effects on immune effectors through two primary pathways: the sympathetic-adrenal-medullary pathway, and the hypothalamic-pituitary-adrenal pathway which modulate adaptive immunity and lymphocyte migration. In this report we show that stress via release of stress hormones induces early T cell activation and greatly impacts the cytoskeleton by modulating numerous actin-regulating proteins. In particular, proteomic profiling revealed significant decreases in numerous key actin-binding proteins including moesin. Although confocal microscopy showed that moesin and actin were uniformly distributed on the surface of resting T cells, a remarkable polarization and redistribution of moesin and actin was observed following treatment with stress hormones with moesin localizing at the distal pole complex. In addition, the alteration in moesin localization and eventual decrease in expression were accompanied by a loss of CD43; a receptor involved in negatively regulating T cell activation. In conclusion, we have defined a novel molecular mechanism whereby stress hormones negatively impact T cell activation and migration through regulation of key cytoskeletal and plasma membrane factors.


Asunto(s)
Citoesqueleto/ultraestructura , Restricción Física/efectos adversos , Estrés Fisiológico/inmunología , Estrés Psicológico/inmunología , Linfocitos T/inmunología , Actinas/biosíntesis , Actinas/genética , Animales , Antígenos CD/biosíntesis , Antígenos CD/genética , Antígenos de Diferenciación de Linfocitos T/biosíntesis , Antígenos de Diferenciación de Linfocitos T/genética , Catecolaminas/fisiología , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas/inmunología , Células Cultivadas/metabolismo , Células Cultivadas/ultraestructura , Citoesqueleto/metabolismo , Femenino , Glucocorticoides/fisiología , Ionomicina/farmacología , Lectinas Tipo C/biosíntesis , Lectinas Tipo C/genética , Leucosialina/análisis , Lipopolisacáridos/farmacología , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Proteínas de Microfilamentos/biosíntesis , Proteínas de Microfilamentos/genética , Proteómica , Estrés Fisiológico/fisiología , Estrés Psicológico/fisiopatología , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Linfocitos T/ultraestructura , Acetato de Tetradecanoilforbol/farmacología
3.
Cancer Immunol Immunother ; 59(2): 293-301, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19690855

RESUMEN

Ovarian cancer patients with persistent (platinum-resistant) or progressive (platinum-refractory) disease respond poorly to second line chemotherapy and have low survival expectancy. New and improved therapeutic approaches are needed and immune biologics are one possibility. Interleukin-2 (IL-2) is a T-cell growth factor believed to be important in anti-tumor immunity. We performed a phase II clinical trial with intraperitoneal (IP) recombinant IL-2 administered in weekly infusions of 6 x 10(5) IU/m2. Thirty-one subjects were sequentially entered into the study and clinical responses were surgically confirmed in 24 patients. The primary end point of this study was clinical response with immunologic measurements as secondary end points. The IP regimen was generally well tolerated. Of the 24 patients assessed for response, there were 6 (4 complete, 2 partial) responses for an overall response rate of 25.0% [95% confidence interval (CI) of 11-45]. The median survival of the 31 patient cohort was 2.1 years (95% CI of 1.3-4.4), but for the 6 patients with responses the median survival has not been reached (range 24-120+ months). Eosinophil and lymphocyte numbers were continuously monitored during treatment. Peripheral blood eosinophils were markedly increased at the completion of treatment (p < 0.0001) and associated with increased circulating eotaxin (p = 0.03). We also found significant associations between changes in CD3 counts and survival (p = 0.05) and between IFNγ- secreting CD8 T cells at early time points and survival (p = 0.04). This study provides important evidence for IP IL-2 in platinum-resistant ovarian cancer and identifies several immune correlates of survival.


Asunto(s)
Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos , Interleucina-2/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Proteínas Recombinantes/uso terapéutico , Adulto , Anciano , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Eosinofilia/inmunología , Femenino , Humanos , Inyecciones Intraperitoneales , Interleucina-2/administración & dosificación , Interleucina-2/efectos adversos , Linfocitos/inmunología , Linfocitosis/inmunología , Persona de Mediana Edad , Neoplasias Ováricas/mortalidad , Compuestos de Platino/uso terapéutico , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Resultado del Tratamiento
4.
Cancer Clin Oncol ; 6(1): 12-24, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28603578

RESUMEN

The ability of stress to induce immune suppression is widely recognized, but the mechanisms underlying the effects of stress on the adaptive immune system during tumor progression are not completely understood. To study the effect of stress on the immune system in vivo, we used a preclinical immunocompetent mouse model bearing 4T1 mammary adenocarcinoma cells. Mice were randomized into 4 groups, including social isolation (SI), acute restraint stress (aRRS), chronic restraint stress (cRRS), or no stress (NS). We found that SI significantly decreased the number of tumor-bearing mice still alive at the end of protocol (28 days), compared to NS mice. Although we did not detect significant changes in primary tumor volume, we observed a significant increase in the endothelial marker CD31 in primary tumors of SI mice and in lung metastases in SI and RRS mice. Survival decline in SI mice was associated with significant decreases in splenic CD8 cells and in activated T cells. From a mechanistic standpoint, RRS increased expression of FOXP3, CXCL-10, and granzyme B in mouse tumors, and the effects were reversed by propranolol. Our data demonstrate that various forms of stress differentially impact adaptive immunity and tumor angiogenesis, and negatively impact survival.

5.
PLoS One ; 9(7): e102409, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25078979

RESUMEN

Activating mutations of Kras oncogene and deletions of Pten tumor suppressor gene play important roles in cancers of the female genital tract. We developed here new preclinical models for gynecologic cancers, using conditional (Cre-loxP) mice with floxed genetic alterations in Kras and Pten. The triple transgenic mice, briefly called MUC1KrasPten, express human MUC1 antigen as self and carry a silent oncogenic KrasG12D and Pten deletion mutation. Injection of Cre-encoding adenovirus (AdCre) in the ovarian bursa, oviduct or uterus activates the floxed mutations and initiates ovarian, oviductal, and endometrial cancer, respectively. Anatomical site-specific Cre-loxP recombination throughout the genital tract of MUC1KrasPten mice leads to MUC1 positive genital tract tumors, and the development of these tumors is influenced by the anatomical environment. Endometrioid histology was consistently displayed in all tumors of the murine genital tract (ovaries, oviducts, and uterus). Tumors showed increased expression of MUC1 glycoprotein and triggered de novo antibodies in tumor bearing hosts, mimicking the immunobiology seen in patients. In contrast to the ovarian and endometrial tumors, oviductal tumors showed higher nuclear grade. Survival for oviduct tumors was significantly lower than for endometrial tumors (p = 0.0015), yet similar to survival for ovarian cancer. Oviducts seem to favor the development of high grade tumors, providing preclinical evidence in support of the postulated role of fallopian tubes as the originating site for high grade human ovarian tumors.


Asunto(s)
Núcleo Celular/patología , Genes ras , Neoplasias de los Genitales Femeninos/genética , Mucina-1/genética , Fosfohidrolasa PTEN/genética , Animales , Femenino , Neoplasias de los Genitales Femeninos/patología , Humanos , Ratones , Microambiente Tumoral
6.
Clin Cancer Res ; 20(23): 6163-74, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25294912

RESUMEN

PURPOSE: Mechanisms of immune dysregulation associated with advanced tumors are relatively well understood. Much less is known about the role of immune effectors against cancer precursor lesions. Endometrioid and clear-cell ovarian tumors partly derive from endometriosis, a commonly diagnosed chronic inflammatory disease. We performed here a comprehensive immune gene expression analysis of pelvic inflammation in endometriosis and endometriosis-associated ovarian cancer (EAOC). EXPERIMENTAL DESIGN: RNA was extracted from 120 paraffin tissue blocks comprising of normal endometrium (n = 32), benign endometriosis (n = 30), atypical endometriosis (n = 15), and EAOC (n = 43). Serous tumors (n = 15) were included as nonendometriosis-associated controls. The immune microenvironment was profiled using Nanostring and the nCounter GX Human Immunology Kit, comprising probes for a total of 511 immune genes. RESULTS: One third of the patients with endometriosis revealed a tumor-like inflammation profile, suggesting that cancer-like immune signatures may develop earlier, in patients classified as clinically benign. Gene expression analyses revealed the complement pathway as most prominently involved in both endometriosis and EAOC. Complement proteins are abundantly present in epithelial cells in both benign and malignant lesions. Mechanistic studies in ovarian surface epithelial cells from mice with conditional (Cre-loxP) mutations show intrinsic production of complement in epithelia and demonstrate an early link between Kras- and Pten-driven pathways and complement upregulation. Downregulation of complement in these cells interferes with cell proliferation. CONCLUSIONS: These findings reveal new characteristics of inflammation in precursor lesions and point to previously unknown roles of complement in endometriosis and EAOC.


Asunto(s)
Activación de Complemento/inmunología , Proteínas del Sistema Complemento/inmunología , Endometriosis/complicaciones , Endometriosis/inmunología , Neoplasias Ováricas/etiología , Adulto , Anciano , Animales , Proliferación Celular , Transformación Celular Neoplásica/inmunología , Análisis por Conglomerados , Activación de Complemento/genética , Complemento C7/genética , Complemento C7/inmunología , Proteínas del Sistema Complemento/genética , Modelos Animales de Enfermedad , Endometriosis/genética , Endometriosis/patología , Células Epiteliales/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Vigilancia Inmunológica , Ratones , Persona de Mediana Edad , Neoplasias Ováricas/patología , Factores de Riesgo
7.
Dis Model Mech ; 2(11-12): 593-603, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19841240

RESUMEN

Endometriosis is defined by the presence of tissue implants resembling endometrial glands outside of the uterus, at ectopic sites, frequently on the ovarian surface. The ectopic lesions are often invasive, resistant to therapy, and may predispose to endometrioid and clear cell ovarian tumors. The complex mechanisms leading to chronic endometriosis are mediated partly by impaired immune surveillance in the host. Although innate immunity has been addressed previously, the response of adaptive immune effectors to specific antigens has not been characterized, mostly because very few endometriosis antigens have been defined to date. We postulated that the mucin 1 (MUC1) glycoprotein, which is normally present on eutopic human endometrial glands and overexpressed in endometrioid and clear cell ovarian tumors, is also present in ectopic lesions of ovarian endometriosis. Furthermore, changes in MUC1 expression in endometriosis could promote adaptive anti-MUC1 immunity that might play a role in the malignant progression. To test our hypothesis, we crossed MUC1 transgenic mice, which express human MUC1 under the endogenous promoter, with the loxP-Stop-loxP-Kras(G12D/+) (Kras) mice, in which endometriosis can be induced through Cre-loxP recombination. The double transgenic MUC1Kras mice develop benign, MUC1-positive ovarian lesions, closely resembling human endometriosis. Subsequent to disease induction, the mice generate high titers of IgM and IgG antibodies that are specific for MUC1. Antibodies appear early in disease and the predominance of the IgG1 subclass suggests Th2-driven immunity. Immune phenotyping revealed an accumulation of Foxp3+ CD4 regulatory T cells (Tregs) in the draining lymph nodes at late-stage disease. Furthermore, our observations in human endometriosis showed a similar recruitment of FOXP3+ CD4 T cells. Overall, our results reveal a Th2/Treg-dominant natural immunity in endometriosis with potential implications for cancer progression.


Asunto(s)
Endometriosis/metabolismo , Factores de Transcripción Forkhead/biosíntesis , Mucina-1/biosíntesis , Linfocitos T Reguladores/citología , Animales , Modelos Animales de Enfermedad , Endometriosis/inmunología , Femenino , Factores de Transcripción Forkhead/genética , Humanos , Inmunidad Innata , Inmunoglobulina G/metabolismo , Ratones , Ratones Transgénicos , Mucina-1/genética , Regiones Promotoras Genéticas , Células Th2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA