Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Gynecol Oncol ; 152(2): 328-333, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30528888

RESUMEN

OBJECTIVE: The universal genetic testing initiative (UGTI) is a quality improvement effort to increase rates of guideline-based genetic counseling (GC) and genetic testing (GT) of patients with potentially hereditary cancers. The UGTI was disseminated to a county hospital gynecologic oncology clinic that serves a diverse, indigent patient population. METHODS: Using the Model for Improvement quality improvement framework, interventions including integrated GC, clinic tracking, assisted GC referrals, and provider education were tested over 26 months. A retrospective data review included patients with high-grade, non-mucinous epithelial ovarian, fallopian tube, and primary peritoneal cancers (HGOC) and endometrial cancers (EC) diagnosed between 9/1/12-8/31/16. Statistical analyses were performed to describe the population and to evaluate rates of recommendation and use of immunohistochemistry tumor testing (IHC), GC, and GT. RESULTS: A cohort of 241 patients (57 HGOC, 184 EC) were included. At the conclusion of the study 84.2% of HGOC patients were referred for GC, 89.6% (43/48) completed GC, and 90.7% (39/43) completed GT. Of EC patients, 81.0% were recommended to have IHC and 62.4% (93/149) completed IHC. Patients with HGOC diagnosed during dissemination of UGTI were significantly more likely to receive a recommendation for GC (p = 0.02) and to complete GT (p = 0.03) than those diagnosed before UGTI. Patients with EC were significantly more likely to complete IHC if diagnosed after UGTI than those diagnosed prior to dissemination (p < 0.001). CONCLUSIONS: The UGTI can be adapted to increase use of guideline-based cancer genetics services in a diverse, indigent, gynecologic cancer patient population.


Asunto(s)
Pruebas Genéticas/métodos , Neoplasias de los Genitales Femeninos/genética , Adulto , Anciano , Carcinoma Epitelial de Ovario/genética , Estudios de Cohortes , Neoplasias de las Trompas Uterinas/genética , Femenino , Asesoramiento Genético/economía , Asesoramiento Genético/métodos , Pruebas Genéticas/economía , Neoplasias de los Genitales Femeninos/economía , Hospitales de Condado/economía , Hospitales de Condado/organización & administración , Humanos , Persona de Mediana Edad , Neoplasias Peritoneales/genética , Pobreza , Estudios Retrospectivos , Adulto Joven
2.
Ann Stat ; 6(2): 542-560, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-28473730

RESUMEN

In this paper we propose a Bayesian approach for inference about dependence of high throughput gene expression. Our goals are to use prior knowledge about pathways to anchor inference about dependence among genes; to account for this dependence while making inferences about differences in mean expression across phenotypes; and to explore differences in the dependence itself across phenotypes. Useful features of the proposed approach are a model-based parsimonious representation of expression as an ordinal outcome, a novel and flexible representation of prior information on the nature of dependencies, and the use of a coherent probability model over both the structure and strength of the dependencies of interest. We evaluate our approach through simulations and in the analysis of data on expression of genes in the Complement and Coagulation Cascade pathway in ovarian cancer.

3.
Cancer Immunol Immunother ; 58(2): 247-58, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18618111

RESUMEN

Tumor associated monocytes/macrophages (MO/MA) are known contributors to the immune-inflammatory cell environment of advanced epithelial ovarian carcinoma (EOC). The secreted proteome of ascitic MO/MA was examined as an aid to the discovery of novel proteins in EOC that are likely to have biological relevance in the inflammatory pathways of EOC. Ascitic fluid MO/MA were isolated from EOC patients, grown short-term in serum-free media. MO/MA supernatants were analyzed for secreted proteins by HPLC fractionation followed by LC-tandem mass spectrometric analysis. The 14-3-3 zeta adaptor protein was identified in supernatants of three of three EOC patients but not in supernatants of buffy coat monocytes isolated from normal donors or the established monocyte cell line THP1. Moreover, 14-3-3 zeta was identified in ascitic fluids in eight of eight chemotherapy-naïve patients by both immunoblot and mass spectrometric analysis. Immunofluorescent staining for 14-3-3 zeta demonstrated expression of the protein on ascitic and peritumoral macrophages in EOC patients. 14-3-3 zeta was also expressed on endothelial cells in the peritumoral stroma and partially on tumor cells. Uptake of 14-3-3 zeta was observed in EOC cell lines co-cultured with the recombinant protein expressed in E. coli. It is demonstrated for the first time that the important adaptor protein 14-3-3 zeta is common to the secretome of ascitic MO/MA and the ascites of advanced EOC patients.


Asunto(s)
Proteínas 14-3-3/metabolismo , Ascitis/metabolismo , Macrófagos/metabolismo , Monocitos/metabolismo , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Ováricas/metabolismo , Proteínas 14-3-3/química , Proteínas 14-3-3/genética , Secuencia de Aminoácidos , Anticuerpos Monoclonales/inmunología , Ascitis/inmunología , Línea Celular Tumoral , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Macrófagos/inmunología , Espectrometría de Masas , Datos de Secuencia Molecular , Monocitos/inmunología , Neoplasias Glandulares y Epiteliales/inmunología , Neoplasias Ováricas/inmunología , Proteoma , Alineación de Secuencia
4.
Gynecol Oncol ; 113(2): 210-5, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19264351

RESUMEN

OBJECTIVE: To evaluate the efficacy and toxicity of carboplatin, granulocyte-macrophage colony-stimulating factor (GM-CSF) and recombinant interferon gamma 1b (rIFN-gamma1b) in women with recurrent, platinum-sensitive ovarian, fallopian tube and primary peritoneal cancer. METHODS: In this phase II study, patients with recurrent, platinum-sensitive ovarian, fallopian tube or primary peritoneal cancer were treated with subcutaneous GM-CSF and rIFN-gamma1b before and after intravenous carboplatin until disease progression or unacceptable toxicity. All patients had measurable disease and a chemotherapy-free interval >6 months. Response was determined using RECIST criteria and CA 125 levels. RESULTS: Between 2003 and 2007, 59 patients received a median of 6 cycles of therapy (range, 1 to 13 cycles). Median age at enrollment was 61 years (range, 35 to 79 years). Median time to progression prior to enrollment was 11 months (range, 6 to 58 months). Of 54 patients evaluable for response, 9 (17%) had a complete response, 21 (39%) had a partial response, and 24 (44%) had progressive disease. The overall response rate was 56% (95% CI: 41% to 69%). With a median follow-up of 6.4 months, median time to progression was 6 months. Myeloid derived cells and platelets increased on day 9 of each chemotherapy cycle. The most common adverse effects were bone marrow suppression, carboplatin hypersensitivity, and fatigue. Responders reported improved quality of life. CONCLUSION: This pre and post-carboplatin cytokine regimen resulted in a reasonable response and a hematologic profile that could invite further evaluation of its components in the treatment of patients with ovarian cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de las Trompas Uterinas/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Peritoneales/tratamiento farmacológico , Adulto , Anciano , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Carboplatino/administración & dosificación , Carboplatino/efectos adversos , Supervivencia sin Enfermedad , Neoplasias de las Trompas Uterinas/sangre , Neoplasias de las Trompas Uterinas/inmunología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Factor Estimulante de Colonias de Granulocitos y Macrófagos/efectos adversos , Humanos , Interferón gamma/administración & dosificación , Interferón gamma/efectos adversos , Persona de Mediana Edad , Recurrencia Local de Neoplasia/sangre , Recurrencia Local de Neoplasia/inmunología , Neoplasias Ováricas/sangre , Neoplasias Ováricas/inmunología , Neoplasias Peritoneales/sangre , Neoplasias Peritoneales/inmunología , Calidad de Vida , Proteínas Recombinantes
5.
Nat Clin Pract Oncol ; 5(3): 171-5, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18227827

RESUMEN

Background A 52-year-old woman whose last known pregnancy was 12 years before presentation was diagnosed with mixed trophoblastic tumor that included placental-site trophoblastic tumor, epithelioid trophoblastic tumor, and focal choriocarcinoma. There was no clear evidence of metastatic disease on initial evaluation. Investigations Histopathology, laboratory tests, immunohistochemistry, chest X-ray, CT scan of the chest, abdomen, and pelvis, fine-needle aspiration biopsy, PET-CT scan.Diagnosis Metastatic chemoresistant placental-site trophoblastic tumor positive for EGFR, VEGF receptor, and platelet-derived growth factor receptor. Management Abdominal hysterectomy, bilateral salpingo-oophorectomy, pelvic radiation, etoposide, methotrexate, actinomycin D/cyclophosphamide and vincristine chemotherapy, left thoracotomy with wedge resection, taxol, etoposide, cisplatin therapy, right thoracotomy with wedge resection.


Asunto(s)
Resistencia a Antineoplásicos , Neoplasias Pulmonares/secundario , Tumor Mixto Maligno/secundario , Tumor Trofoblástico Localizado en la Placenta/secundario , Neoplasias Uterinas/patología , Terapia Combinada , Femenino , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Persona de Mediana Edad , Tumor Mixto Maligno/patología , Tumor Mixto Maligno/terapia , Embarazo , Tumor Trofoblástico Localizado en la Placenta/patología , Tumor Trofoblástico Localizado en la Placenta/terapia , Neoplasias Uterinas/terapia
6.
Clin Cancer Res ; 13(19): 5736-44, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17908963

RESUMEN

PURPOSE: To describe the eicosanoid profile and differentially expressed eicosanoid and arachidonic acid pathway genes in tissues from patients with advanced epithelial ovarian cancer (EOC). EXPERIMENTAL DESIGN: We first employed electrospray tandem mass spectrometry to determine tissue-specific concentrations of the eicosanoids prostaglandin E2 (PGE2), the hydroxyeicosatetraenoic acids (12-HETE and 5-HETE), and leukotriene (LTB4), selected for tumor growth potential, and two other bioactive lipids (15-HETE and 13-HODE) with tumor cell proliferation interference potential. The cellular location of eicosanoid activity was identified by immunofluorescence antibody costaining and confocal microscopy. Differential analysis of eicosanoid and arachidonic pathway genes was done using a previously validated cDNA microarray platform. Tissues used included EOC tumor, tumor-free malignant peritoneum (MP), and benign peritoneum (BP) from patients with benign pelvic disease. RESULTS: (a) Eicosanoid products were detected in tumor, MP, and BP specimens. PGE2 levels were significantly elevated in tumors in an overall comparison with MP or BP (P < 0.001). Combined levels of PGE2, 12-HETE, 5-HETE, and LTB4 increased progressively from low to high concentrations in BP, MP, and tumors (P = 0.012). Neither 15-HETE nor 13-HODE showed a significant opposite trend toward levels found in BP. (b) Tissue specimens representing common EOC histotypes showed strong coexpressions of cyclooxygenases (COX-1) and prostaglandin E synthases (PGES-1) on tumor cells, whereas intratumoral or peritumoral MO/MA coexpressed COX-1 and COX-2 and PGES-1 and PGES-2, respectively. (c) cDNA microarray analysis of MP, BP, and tumor showed that a number of eicosanoid and arachidonic acid pathway genes were differentially expressed in MP and BP compared with tumor, except for CYP2J2, which was increased in tumors. CONCLUSIONS: Elevated levels of eicosanoid metabolites in tumors and differential expression of eicosanoid and arachidonic acid pathway genes in the peritoneum support the involvement of bioactive lipids in the inflammatory tumor environment of EOC.


Asunto(s)
Eicosanoides/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Peritoneo/metabolismo , Antígenos CD/biosíntesis , Antígenos de Diferenciación Mielomonocítica/biosíntesis , Ácido Araquidónico/química , Dinoprostona/metabolismo , Eicosanoides/química , Femenino , Perfilación de la Expresión Génica , Humanos , Inflamación , Lípidos/química , Microscopía Confocal , Modelos Biológicos , Receptores de Superficie Celular/biosíntesis , Espectrometría de Masa por Ionización de Electrospray
7.
Cancer Res ; 66(6): 2953-61, 2006 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-16540643

RESUMEN

There is an increasing interest in complementing conventional histopathologic evaluation with molecular tools that could increase the sensitivity and specificity of cancer staging for diagnostic and prognostic purposes. This study strove to identify cancer-specific markers for the molecular detection of a broad range of cancer types. We used 373 archival samples inclusive of normal tissues of various lineages and benign or malignant tumors (predominantly colon, melanoma, ovarian, and esophageal cancers). All samples were processed identically and cohybridized with an identical reference RNA source to a custom-made cDNA array platform. The database was split into training (n = 201) and comparable prediction (n = 172) sets. Leave-one-out cross-validation and gene pairing analysis identified putative cancer biomarkers overexpressed by malignant lesions independent of tissue of derivation. In particular, seven gene pairs were identified with high predictive power (87%) in segregating malignant from benign lesions. Receiver operator characteristic curves based on the same genes could segregate malignant from benign tissues with 94% accuracy. The relevance of this study rests on the identification of a restricted number of biomarkers ubiquitously expressed by cancers of distinct histology. This has not been done before. These biomarkers could be used broadly to increase the sensitivity and accuracy of cancer staging and early detection of locoregional or systemic recurrence. Their selective expression by cancerous compared with paired normal tissues suggests an association with the oncogenic process resulting in stable expression during disease progression when the presently used differentiation markers are unreliable.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias/genética , Biomarcadores de Tumor/biosíntesis , Análisis por Conglomerados , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Valor Predictivo de las Pruebas , Sensibilidad y Especificidad , Regulación hacia Arriba
8.
Cancers (Basel) ; 10(12)2018 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-30469497

RESUMEN

Increased number of tumor-infiltrating CD8+ lymphocytes is associated with improved survival in patients with advanced stage high grade serous ovarian cancer (HGSOC) but the underlying molecular mechanism has not been thoroughly explored. Using transcriptome profiling of microdissected HGSOC tissue with high and low CD8+ lymphocyte count and subsequent validation studies, we demonstrated that significantly increased ISG15 (Interferon-stimulated gene 15) expression in HGSOC was associated with high CD8+ lymphocyte count and with the improvement in median overall survival in both univariate and multivariate analyses. Further functional studies showed that endogenous and exogenous ISG15 suppressed ovarian cancer progression through ISGylation of ERK in HGSOC, and activation of NK cells and CD8+ T lymphocytes. These data suggest that the development of treatment strategies based on up-regulating ISG15 in ovarian cancer cells or increased circulating ISG15 in ovarian cancer patients is warranted.

9.
J Transl Med ; 5: 66, 2007 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-18076766

RESUMEN

BACKGROUND: Pharmacokinetic advantages of intraperitoneal (IP) rhIL-12, tumor response to IP delivery of other cytokines as well as its potential anti-angiogenic effect provided the rationale for further evaluation of IPrhIL-12 in patients with persistent ovarian or peritoneal carcinoma. METHODS: A phase 2 multi-institutional trial (NCI Study #2251) of IP rIL-12 (300 nanogram/Kg weekly) was conducted in patients with ovarian carcinoma or primary peritoneal carcinoma. Patients treated with primary therapy for ovarian cancer who had no extraabdominal/parenchymal disease or bulky peritoneal disease were eligible. Four to 8 weeks from last chemotherapy, eligible patients underwent a laparotomy/laparoscopy. Patients with residual disease < or = 1 cm were registered for the treatment phase 2-5 weeks post surgery. The effect of IP rIL-12 on the expression of TNFalpha , INFalpha , IL-10, IP-10, IL-8, FGF, VEGF was also studied. RESULTS: Thirty-four patients were registered for the first screening phase of the study. Median age was 56.6 years (range: 31-71); 12 completed the second phase and were evaluable for response/toxicity. Performance scores of IL-12 treated patients were 0 (11 pts) and 1 (1 pt). There were no treatment related deaths, peritonitis or significant catheter related complications. Toxicities included grade 4 neutropenia (1), grade 3 fatigue (4), headache (2), myalgia (2), non-neutropenic fever (1), drug fever (1), back pain (1), and dizziness (1). The best response observed was SD. Two patients had SD and 9 had PD, and 1 was evaluable for toxicity only. Peritoneal fluid cytokine measurements demonstrated a > or = 3 fold relative increase post-rhIL-12: IFN-gamma, 5/5 pts; TNF-alpha , 1/5; IL-10, 4/5; IL-8, 5/5; and VEGF, 3/5. IP10 levels were increased in 5/5 patients. Cytokine response profiles suggest either NK or T-cell mediated effects of IP rhIL-12. Cytokine/chemokine results also suggest a pleiotropic response since proteins with potential for either anti-tumor (IFN-gamma , IP-10) or pro-tumor growth effects (VEGF, IL-8) were detected. CONCLUSION: IP IL-12 can safely be administered at this dose and schedule to patients after first line chemotherapy for ovarian/peritoneal carcinoma. The maximum response was stable disease. Future IP therapies with rhIL-12 will require better understanding and control of pleiotropic effects of IL-12.


Asunto(s)
Interleucina-12/toxicidad , Interleucina-12/uso terapéutico , Neoplasia Residual/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Peritoneales/tratamiento farmacológico , Adulto , Anciano , Líquido Ascítico/química , Ensayos Clínicos Fase I como Asunto , Citocinas/análisis , Femenino , Humanos , Persona de Mediana Edad , Proteínas Recombinantes/uso terapéutico , Proteínas Recombinantes/toxicidad , Seguridad
10.
Gynecol Oncol ; 107(2): 173-6, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17950384

RESUMEN

OBJECTIVE: The unique characteristics of cancer, particularly issues involving the use of surrogate endpoints in clinical trials, present special challenges in the development of cancer drugs. In response, the U.S. Food and Drug Administration (FDA) has partnered with the American Society of Clinical Oncology, the American Association for Cancer Research, and the American Society of Hematology to conduct public workshops evaluating potential endpoints for drug approvals for the most common tumor types. METHODS: A workshop evaluating potential endpoints in ovarian cancer drug research was held in Bethesda, Maryland, in April 2006. Invited experts presented research findings and discussed endpoints in trials of drugs for treatment of Stage III and IV ovarian cancer. RESULTS: The panel responded to specific questions from FDA, discussing use of progression-free survival as a surrogate for overall survival and use of CA-125 levels as an indicator of response. Panel members also addressed endpoints in first-line therapy, second-line and subsequent therapy, and maintenance therapy. CONCLUSION: Expert commentary provided by panel members will inform FDA's draft guidance on clinical endpoints for cancer drug approvals and will be discussed at meetings of the FDA's Oncologic Drugs Advisory Committee. FDA intends to develop a set of principles that can be used to define efficacy standards for drugs used to treat ovarian and other cancers.


Asunto(s)
Biomarcadores de Tumor/sangre , Determinación de Punto Final , Neoplasias Ováricas/tratamiento farmacológico , Satisfacción del Paciente , Antineoplásicos , Investigación Biomédica , Antígeno Ca-125/sangre , Ensayos Clínicos como Asunto , Supervivencia sin Enfermedad , Aprobación de Drogas , Femenino , Ginecología , Estado de Salud , Humanos , Oncología Médica , Calidad de Vida , Sociedades Médicas , Resultado del Tratamiento , Estados Unidos , United States Food and Drug Administration
11.
Clin Cancer Res ; 12(5): 1515-24, 2006 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-16533776

RESUMEN

PURPOSE: Monocytes/macrophages (MO/MA) are an important but heterogeneous population of immune inflammatory cells that have diverse effector functions. We examined and compared these differences in peripheral blood and ascites of epithelial ovarian cancer patients with peripheral blood of normal donors. EXPERIMENTAL DESIGN: Comparisons were made of cell surface subsets, cytokine production, and FcR-dependent cytotoxicity of CD14+ MO/MA and the CD14brightCD16-HLA-DR+ MO/MA subset in normal donor peripheral blood and peripheral blood and ascites from epithelial ovarian cancer patients. Studies were done on monocyte-derived macrophages cultured with macrophage colony-stimulating factor and activated with lipopolysaccharide or a combination of lipopolysaccharide plus recombinant IFN-gamma. RESULTS: We determined that MO/MA or its subset from epithelial ovarian cancer patients had altered morphology and significantly less antibody-dependent cell-mediated cytotoxicity and phagocytic activity than did MO/MA from normal donors. Our findings also showed that monocyte-derived macrophages from both epithelial ovarian cancer patients and normal donors produce macrophage colony-stimulating factor-stimulated cytokines, including interleukin-8, tumor necrosis factor-alpha, and interleukin-6. CONCLUSIONS: These findings highlight for the first time the defective antibody-dependent cell-mediated cytotoxicity and phagocyte functions of epithelial ovarian cancer-associated MO/MA, which could have implications for immunobiotherapeutic strategies.


Asunto(s)
Macrófagos/fisiología , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Ováricas/metabolismo , Fagocitosis , Adenocarcinoma de Células Claras/metabolismo , Adenocarcinoma de Células Claras/patología , Citotoxicidad Celular Dependiente de Anticuerpos , Líquido Ascítico/citología , Carcinoma Endometrioide/metabolismo , Carcinoma Endometrioide/patología , Carcinoma Papilar/metabolismo , Carcinoma Papilar/patología , Femenino , Antígenos HLA-DR/metabolismo , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Neoplasias Glandulares y Epiteliales/inmunología , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Neoplasias Peritoneales/metabolismo , Neoplasias Peritoneales/patología , Receptores de IgG/metabolismo , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/metabolismo
12.
J Transl Med ; 4: 30, 2006 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-16824216

RESUMEN

BACKGROUND: We previously showed that tumor-free peritoneum of patients with epithelial ovarian cancer (EOC) exhibited enhanced expression of several inflammatory response genes compared to peritoneum of benign disease. Here, we examined peritoneal inflammatory cell patterns to determine their concordance with selected enhanced genes. METHODS: Expression patterns of selected inflammatory genes were mined from our previously published data base. Bilateral pelvic peritoneal and subjacent stromal specimens were obtained from 20 women with EOC and 7 women with benign pelvic conditions. Sections were first stained by indirect immunoperoxidase and numbers of monocytes/macrophages (MO/MA), T cells, B cells, and NK cells counted. Proportions of CD68+ cells and CD3+ cells that coexpressed MO/MA differentiation factors (CD163, CCR1, CXCR8, VCAM1, and phosphorylated cytosolic phospholipase A2 [pcPLA2]), which had demonstrated expression in EOC peritoneal samples, were determined by multicolor immunofluorescence. RESULTS: MO/MA were present on both sides of the pelvic peritoneum in EOC patients, with infiltration of the subjacent stroma and mesothelium. CD68+ MO/MA, the most commonly represented population, and CD3+ T cells were present more often in EOC than in benign pelvic tumors. NK cells, B cells, and granulocytes were rare. CXCL8 (IL-8) and the chemokine receptor CCR1 were coexpressed more frequently on MO/MA than on CD3+ cells contrasting with CD68+/CD163+ cells that coexpressed CXCL8 less often. An important activated enzyme in the eicosanoid pathway, pcPLA2, was highly expressed on both CD68+ and CD163+ cells. The adherence molecule Vascular Cell Adhesion Molecule-1 (VCAM1) was expressed on CD31+ endothelial cells and on a proportion of CD68+ MO/MA but rarely on CD3+ cells. CONCLUSION: The pelvic peritoneum in EOC exhibits a general pattern of chronic inflammation, represented primarily by differentiated MO/MA, and distinct from that in benign conditions concordant with previous profiling results.

13.
J Transl Med ; 4: 16, 2006 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-16603073

RESUMEN

BACKGROUND: Monocyte/macrophages (MO/MA), a polymorphic population of innate immune cells, have the potential to mediate antitumor effects, and may also contribute to protumor effects. A priming and post-chemotherapy schedule of the myeloid cell mobilizing and immune stimulatory growth factor, granulocyte monocyte stimulating factor (GM-CSF, Leukine) and the MO/MA activating cytokine recombinant interferon gamma 1b (rIFN-gamma1b, Actimmune) has been developed. The pre- and post-chemotherapy design is based upon known in vivo kinetics and immune modulatory effects of these molecules. Carboplatin (Paraplatin) was selected as the cornerstone of treatment of epithelial ovarian cancer (EOC). METHODS: We studied hematopoietic and immunologic effects of GM-CSF and rIFN-gamma1b before and after carboplatin in patients with recurrent EOC. Potentially chemotherapy-sensitive patients with recurrent measurable tumors received subcutaneous GM-CSF (starting at 400 mug/day) for 7 days plus subcutaneous rIFN-gamma1b (100 mug) on days 5 and 7, before and after intravenous carboplatin (area under the curve of 5). We performed standard hematologic assessment and monitored monocyte (MO), dendritic cell, major cell subset counts, and antibody-dependent cell-mediated cytotoxicity (ADCC) against a Her2neu+ tumor cell line, as well as selected plasma inflammatory cytokine, chemokine and growth factor levels. RESULTS: Our analysis comprised only the first 3 months of treatment in the initial 25 patients. Relative to pretreatment baseline values, white blood cell, neutrophil, MO, and eosinophil counts increased (P

14.
Clin Cancer Res ; 11(1): 113-22, 2005 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-15671535

RESUMEN

PURPOSE: Epithelial ovarian cancer (EOC) is characterized by early peritoneal involvement ultimately contributing to morbidity and mortality. To study the role of the peritoneum in fostering tumor invasion, we analyzed differences between the transcriptional repertoires of peritoneal tissue lacking detectable cancer in patients with EOC versus benign gynecologic disease. EXPERIMENTAL DESIGN: Specimens were collected at laparotomy from patients with benign disease (b) or malignant (m) ovarian pathology and comprised primary ovarian tumors, paired bilateral specimens from adjacent peritoneum and attached stroma (PE), subjacent stroma (ST), peritoneal washes, ascites, and peripheral blood mononuclear cells. Specimens were immediately frozen. RNA was amplified by in vitro transcription and cohybridized with reference RNA to a custom-made 17.5k cDNA microarray. RESULTS: Principal component analysis and unsupervised clustering did not segregate specimens from patients with benign or malignant pathology. Class comparison identified differences between benign and malignant PE and ST specimens deemed significant by permutation test (P = 0.027 and 0.012, respectively). A two-tailed Student's t test identified 402 (bPE versus mPE) and 663 (mST versus bST) genes differentially expressed at a significance level of P2 < or = 0.005 when all available paired samples from each patient were analyzed. The same comparison using one sample per patient reduced the pool of differentially expressed genes but retained permutation test significance for bST versus mST (P = 0.031) and borderline significance for bPE versus mPE (P = 0.056) differences. CONCLUSIONS: The presence of EOC may foster peritoneal implantation and growth of cancer cells by inducing factors that may represent molecular targets for disease control.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias Ováricas/patología , Células del Estroma/patología , Biología Computacional , ADN Complementario/metabolismo , Matriz Extracelular/metabolismo , Femenino , Humanos , Invasividad Neoplásica , Metástasis de la Neoplasia , Análisis de Secuencia por Matrices de Oligonucleótidos , Ovario/patología , Peritoneo/patología , ARN/metabolismo , Transcripción Genética , Regulación hacia Arriba
15.
Clin Cancer Res ; 22(3): 533-9, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26832744

RESUMEN

In 2013, the U.S. Department of Health and Human Services modified the Health Insurance Portability and Accountability Act Privacy Rule to "strengthen privacy and security protections" while "improving workability and effectiveness to increase flexibility for and decrease burden on regulated entities." In this article, we attempt to translate these generalized goals into the real-world implications of these changes. Under the new rules, researchers can obtain participants' permission to use their protected health information for more research activities with a single, upfront authorization (thereby reducing paperwork for participants, researchers, and institutional review boards) while providing potential participants with more information upon which to base their decisions about participation. The combined authorizations can be used in clinical trials and their optional substudies and in stand-alone biospecimen-banking research that includes authorization to permit future research use. We also suggest best practices for taking advantage of the flexibility offered by the new rules while maintaining strong privacy protections for human subjects.


Asunto(s)
Centros Médicos Académicos , Instituciones Oncológicas , Health Insurance Portability and Accountability Act/legislación & jurisprudencia , Investigadores , Investigación Biomédica/legislación & jurisprudencia , Humanos , Estados Unidos
16.
J Transl Med ; 3: 25, 2005 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-15969748

RESUMEN

Epithelial ovarian cancer (EOC) represents the most frequent cause of death in the United States from a cancer involving the female genital tract. Contributing to the overall poor outcome in EOC patients, are the metastases to the peritoneum and stroma that are common in this cancer. In one study, cDNA microarray analysis was performed on fresh tissue to profile gene expression in patients with EOC. This study showed a number of genes with significantly altered expression in the pelvic peritoneum and stroma, and in the vicinity of EOC implants. These genes included those encoding coagulation factors and regulatory proteins in the coagulation cascade and genes encoding proteins associated with inflammatory responses. In addition to promoting the formation of blood clots, coagulation factors exhibit many other biologic functions as well as tumorigenic functions, the later including tumor cell proliferation, angiogenesis, invasion, and metastasis. Coagulation pathway proteins involved in tumorigenesis consist of factor II (thrombin), thrombin receptor (protease-activated receptors), factor III (tissue factor), factor VII, factor X and factor I (fibrinogen), and fibrin and factor XIII. In a recent study we conducted, we found that factor XII, factor XI, and several coagulation regulatory proteins, including heparin cofactor-II and epithelial protein C receptor (EPCR), were also upregulated in the peritoneum of EOC. In this review, we summarize evidence in support of a role for these factors in promoting tumor cell progression and the formation of ascites. We also discuss the different roles of coagulation factor pathways in the tumor and peritumoral microenvironments as they relate to angiogenesis, proliferation, invasion, and metastasis. Since inflammatory responses are another characteristic of the peritoneum in EOC, we also discuss the linkage between the coagulation cascade and the cytokines/chemokines involved in inflammation. Interleukin-8, which is considered an important chemokine associated with tumor progression, appears to be a linkage point for coagulation and inflammation in malignancy. Lastly, we review findings regarding the inflammatory process yielded by certain clinical trials of agents that target members of the coagulation cascade in the treatment of cancer. Current data suggest that disrupting certain elements of the coagulation and inflammation processes in the tumor microenvironment could be a new biologic approach to cancer therapeutics.

17.
Hum Immunol ; 66(5): 554-62, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15935893

RESUMEN

Substantial evidence has been accumulated suggesting that T cells in patients with epithelial ovarian carcinoma (EOC) exhibit an antigen-driven immune response directed against the tumor cells. In the context of human leukocyte antigen (HLA), this suggests its possible involvement in the disease. Therefore, we examined the distribution of the HLA-DRB1*, -DQA1*, and -DQB1* alleles in 47 patients with EOC and 67 healthy Caucasian women. The frequency of D(70) and E(71) polymorphic residues of the DRB1 alleles was significantly reduced in EOC patients versus controls (pD(70)E(71) = 0.009), suggesting a protective role against the disease. The DQalpha residues R(52) and Y(11)R(55) were increased in the patients (p = 0.008 and 0.012, respectively). Because residues 11 and 55 participate in the formation of pocket 1, they may be functionally important amino acid positions that influence disease susceptibility. The frequency of the DQalpha susceptibility epitope (R(52)Y(11)R(55)) among the DRbetaD(70)E(71)-positive EOC patients was increased when compared with DRbetaD(70)E(71)-positive controls (EOC, 100%; control, 52%; p = 0.028). Among individuals without the DQalpha susceptibility epitope, the distribution of DRbetaD(70)E(71)-positive cases was significantly different between EOC patients and controls (EOC, 0%; control, 60%; p = 0.039). Therefore, it appears that the presence of DQalpha susceptibility elements overrides the protective effect of the DRbetaD(70)E(71) epitope and suggests an interactive relationship between DRbeta and DQalpha epitopes that may be of importance for disease susceptibility. Because positions DRbeta 70,71 and DQalpha 52 have been implicated in immunologic diseases, it is likely that besides being critical for T-cell recognition, they may also play a role in T-cell development and acquisition of the T-cell repertoire.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Antígenos HLA-DQ/genética , Antígenos HLA-DR/genética , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Ováricas/genética , Secuencia de Aminoácidos/genética , Susceptibilidad a Enfermedades/inmunología , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Femenino , Frecuencia de los Genes/genética , Frecuencia de los Genes/inmunología , Antígenos HLA-DQ/inmunología , Cadenas alfa de HLA-DQ , Antígenos HLA-DR/inmunología , Prueba de Histocompatibilidad , Humanos , Datos de Secuencia Molecular , Neoplasias Glandulares y Epiteliales/inmunología , Neoplasias Ováricas/inmunología , Polimorfismo Genético/genética , Polimorfismo Genético/inmunología
18.
Oncol Res ; 15(1): 49-57, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15839305

RESUMEN

The STK15 (also known as Aurora-A/BTAK) gene localized on chromosome 20q13 and encoding a centrosome-associated serine/threonine kinase is amplified and overexpressed in multiple human tumor cell types. Overexpression of this gene is involved in tumorigenic transformation, induction of centrosome duplication-distribution abnormalities, and aneuploidy in mammalian cells. To examine the potential role of STK15 in ovarian tumorigenesis, its mRNA and protein expression status were examined in cells grown in culture from 15 ovarian cancer specimens using semiquantitative RT-PCR and Western blot analysis. Normal ovarian surface tissues and the near diploid nontumorigenic breast epithelial cell line MCF10 were used as controls. The status of STK15 correlated with transformation-associated cellular phenotypes including tumorigenicity in nude mice, p53 expression level, and chromosomal ploidy. For chromosome ploidy analyses, FISH was carried out with direct fluorescence-labeled a-satellite probes for chromosome 3 and 17. STK15 mRNA was found overexpressed in 10 of the 15 ovarian cancer cell cultures. Five of these cell cultures revealed a truncated form of the STK15 protein with a molecular mass of 36 kDa. When tested for tumorigenicity in nude mice, 9 of the 10 cell cultures that overexpressed STK15 mRNA formed tumors in nude mice, while only one of the five cell cultures with no overexpression did. Cells overexpressing STK15 mRNA showed significant correlation with chromosome 3 polysomy. Six of the 13 (46%) cell cultures analyzed for p53 expression revealed overexpression of p53 and five of these six (83%) also overexpressed STK15. Four of the remaining seven cultures (57%) with overexpression of STK15 revealed minimal or no expression of p53. These results demonstrate that overexpression of STK15 significantly correlates with nude mice tumorigenicity and chromosomal aneuploidy in human ovarian cancer cells grown in vitro. Additionally, cells overexpressing STK15 also revealed frequent coordinate loss of wild-type p53 function manifested either as highly expressed intense staining reflective of a mutant form of p53 or almost complete absence of p53 staining. Overexpression of STK15 with coordinate loss of wild-type p53 function thus appears to play an important role in ovarian tumorigenesis and offers a novel molecular target in designing effective therapy of human ovarian cancer.


Asunto(s)
Transformación Celular Neoplásica/genética , Inestabilidad Cromosómica , Perfilación de la Expresión Génica , Genes p53 , Neoplasias Ováricas/genética , Proteínas Serina-Treonina Quinasas/biosíntesis , Aneuploidia , Animales , Aurora Quinasa A , Aurora Quinasas , Análisis Mutacional de ADN , Femenino , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Ratones , Ratones Desnudos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Regulación hacia Arriba
19.
Clin Cancer Res ; 8(6): 1754-60, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12060613

RESUMEN

PURPOSE: The purpose of this study was to examine ovarian cancer cells for the expression of decorin, a proteoglycan component of the cell matrix that can inhibit cancer cell growth. EXPERIMENTAL DESIGN: Cultured ovarian cancer cells and surgically excised tumors were examined by immunohistochemistry and Western blot analysis for decorin expression. Reverse transcription-polymerase chain reaction analysis was used to analyze cultured cells for decorin transcripts. RESULTS: We detected decorin transcripts in two ovarian cancer cell lines by reverse transcription-polymerase chain reaction analysis. However, no decorin was found in conditioned culture medium from those cell lines. Cells treated with the proteasome inhibitor MG132 showed strong perinuclear staining with a decorin-specific monoclonal antibody by immunohistochemistry. Also, Western blot analysis showed the presence of a ladder of decorin-specific bands that were intensified by treatment with MG132, suggesting that de novo synthesized decorin was degraded by the ubiquitination pathway. The decorin component of tumor stroma was previously shown to contain high levels of chondroitin sulfate as opposed to dermatan sulfate side chains, and those molecules contained unusually high levels of O- and 6-sulfate linkages. We provided immunohistochemical evidence that these chondroitin sulfate side chains may have been produced by myofibroblasts. CONCLUSIONS: Decorin protein expression was not detected in ovarian cancer cells. Decorin transcripts were produced and probably translated, but the protein was probably degraded by the ubiquitination pathway. We present evidence that stromal decorin of ovarian tumors was made by myofibroblasts. We also propose that decorin may be a tumor suppressor gene that is inactivated during epithelial cell development.


Asunto(s)
Neoplasias Ováricas/metabolismo , Proteoglicanos/metabolismo , Actinas/metabolismo , Antineoplásicos/farmacología , Western Blotting , Cisteína Endopeptidasas , Inhibidores de Cisteína Proteinasa/farmacología , Cartilla de ADN/química , Decorina , Endometriosis/metabolismo , Endometriosis/patología , Proteínas de la Matriz Extracelular , Femenino , Humanos , Técnicas para Inmunoenzimas , Leupeptinas/farmacología , Complejos Multienzimáticos/antagonistas & inhibidores , Músculo Liso/metabolismo , Neoplasias Ováricas/genética , Complejo de la Endopetidasa Proteasomal , Proteoglicanos/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Clin Cancer Res ; 9(2): 845-52, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12576458

RESUMEN

PURPOSE: The purpose of this study was to characterize cell cultures and xenografts derived from patients with ovarian cancer. EXPERIMENTAL DESIGN: Ninety specimens from 67 patients were plated in RPMI 1640 or inoculated in nude mice. Growth characteristics of cell cultures and xenografts were determined. Expression of receptors for estrogen, progesterone, androgen, epithelial growth factor, fibroblast growth factor, HER-2/erbB-2/c-neu proto-oncogene, and the P53 expression were characterized by immunocytochemistry in 28 cell cultures. RESULTS: Forty-nine percent of samples were cultured successfully in vitro. Ascitic and pleural effusion specimens were more likely to produce a cell culture or a xenograft than solid tissue specimens (P < 0.005). All of the cell cultures had an epithelial morphology, and 89% were aneuploid with a mean DNA index of 1.6 (range, 0.9-3.0). Of 54 and 61 specimens inoculated into nude mice i.p. and s.c., 15 (28%) and 18 (30%) produced a xenograft, respectively, with two-thirds of these xenografts being reproducibly tumorigenic. The median time to first passage was 21 weeks for cell cultures and 8-12 weeks for xenografts. Expression of epithelial growth factor receptor, HER-2/erbB-2/c-neu proto-oncogene, fibroblast growth factor receptor, estrogen, progesterone, and androgen was seen in 24, 21, 31, 17, 43, and 18%, respectively. P53 was overexpressed in 62% of cell cultures analyzed. CONCLUSIONS: Ovarian cancer cells collected from effusions are easier to grow in vitro than in vivo. The only characteristic that may be associated with tumorigenicity was abnormal P53 expression. This panel of ovarian cancer materials provides useful models for biological or therapeutical studies.


Asunto(s)
Tumor Mulleriano Mixto/patología , Neoplasias Ováricas/patología , Animales , Biomarcadores de Tumor/análisis , Técnicas de Cultivo de Célula/métodos , Ciclo Celular , Receptores ErbB/análisis , Femenino , Humanos , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Proto-Oncogenes Mas , Receptores de Esteroides/análisis , Trasplante Heterólogo/métodos , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA