Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Gynecol Oncol ; 170: 221-228, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36709663

RESUMO

BACKGROUND: IGF signaling has been implicated in the pathogenesis and progression of ovarian carcinoma (OC). Single agent activity and safety of ganitumab (AMG 479), a fully human monoclonal antibody against IGF1R that blocks binding of IGF1 and IGF2, were evaluated in patients with platinum-sensitive recurrent OC. METHODS: Patients with CA125 progression (GCIG criteria) or measurable disease per RECIST following primary platinum-based therapy received 18 mg/kg of ganitumab q3w. The primary endpoint was objective response rate (ORR) assessed per RECIST 1.1 by an independent radiology review committee (IRC) and/or GCIG CA125 criteria. Secondary endpoints included clinical benefit rate (CBR), progression free survival (PFS) and overall survival (OS). RESULTS: 61 pts. were accrued. Objective responses were seen in 5/61 patients (ORR 8.2%, 95% CI, 3.1-18.8) with 1 partial response (PR) by RECIST and 2 complete responses (CR) as well as 2 PR by CA125 criteria. CBR was 80.3% (95% CI, 67.8-89.0%). The median PFS according to RECIST by IRC was 2.1 months (95% CI, 2.0-3.1). The median PFS per RECIST IRC and/or CA125 was 2.0 months (95% CI, 1.8-2.2). The median OS was 21 months (95% CI, 19.5-NA). The most common overall adverse events were fatigue (36.1%) and hypertension (34.4%). Grade 1/2 hyperglycemia occurred in 30.4% of patients. Hypertension (11.5%) and hypersensitivity (8.2%) were the most frequent grade 3 adverse events. CONCLUSIONS: IGF1R inhibition with ganitumab was well-tolerated, however, our results do not support further study of ganitumab as a single agent in unselected OC patients.


Assuntos
Anticorpos Monoclonais Humanizados , Neoplasias Ovarianas , Humanos , Feminino , Anticorpos Monoclonais/efeitos adversos , Neoplasias Ovarianas/tratamento farmacológico
2.
Gynecol Oncol ; 163(3): 465-472, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34642026

RESUMO

PURPOSE: Insulin-like growth factor (IGF) signaling is implicated in pathogenesis and chemotherapy resistance of epithelial ovarian cancer (EOC). We explored efficacy and safety of adding ganitumab, a monoclonal antibody targeting IGF-1R, to carboplatin/paclitaxel (CP) chemotherapy in patients with primary EOC. DESIGN: Patients were randomly assigned to receive CP/ganitumab (18 mg/kg q3w) or CP/placebo for 6 cycles followed by 6 cycles of single agent ganitumab/placebo maintenance therapy as front-line therapy. Primary endpoint was progression free survival. Secondary endpoints were time to progression and overall survival. Pretreatment samples were prospectively collected for retrospective biomarker analyses. RESULTS: 170 patients enrolled. 165 patients assessable for toxicity. Median PFS was 15.7 months with CP/ganitumab and 16.7 months with CP/placebo (HR 1.23; 95% CI 0.82-1.83, P = 0.313). All grade neutropenia (84.1% vs 71.4%), thrombocytopenia (75.3% vs 57.1%) and hyperglycemia (15.9% vs 2.6%) were more common in the ganitumab group compared to the placebo group. Ganitumab/placebo related serious adverse events were reported in 26.1% of the patients with ganitumab and in 6.5% with placebo. Non-progression related fatal events were more common with ganitumab (5 versus 2 patients). The ganitumab group experienced more dose delays which resulted in lower relative dose intensity of chemotherapy in the experimental group. In an exploratory model IGFBP2 expression was predictive of ganitumab response (treatment interaction; PFS, P = 0.03; OS, P = 0.01). CONCLUSION: Addition of ganitumab to CP chemotherapy in primary EOC did not improve PFS. Our results do not support further study of ganitumab in unselected EOC patients.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Epitelial do Ovário/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Biomarcadores Tumorais/metabolismo , Carboplatina/administração & dosagem , Carboplatina/efeitos adversos , Carcinoma Epitelial do Ovário/metabolismo , Carcinoma Epitelial do Ovário/patologia , Feminino , Humanos , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Paclitaxel/administração & dosagem , Paclitaxel/efeitos adversos , Intervalo Livre de Progressão , Somatomedinas/metabolismo
3.
Gynecol Oncol ; 143(1): 152-158, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27461360

RESUMO

Cyclin E1 (CCNE1) gene amplification occurs in approximately 20% of ovarian high grade serous carcinoma (HGSC) and is associated with chemotherapy resistance and, in some studies, overall poor prognosis. The role of cyclin E1 in inducing S phase entry relies upon its interactions with cyclin dependent kinases (CDK), specifically CDK2. Therapies to target cyclin E1-related functions have centered on CDK inhibitors and proteasome inhibitors. While many studies have helped elucidate the functions and regulatory mechanisms of cyclin E1, further research utilizing cyclin E1 as a therapeutic target in ovarian cancer is warranted. This review serves to present the scientific background describing the role and function of cyclin E1 in cancer development and progression, to distinguish cyclin E1-amplified HGSC as a unique subset of ovarian cancer deserving of further therapeutic investigation, and to provide an updated overview on the studies which have utilized cyclin E1 as a target for therapy in ovarian cancer.


Assuntos
Ciclina E/fisiologia , Cistadenocarcinoma Seroso/etiologia , Proteínas Oncogênicas/fisiologia , Neoplasias Ovarianas/etiologia , Ciclina E/antagonistas & inibidores , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Quinase 2 Dependente de Ciclina/fisiologia , Cistadenocarcinoma Seroso/terapia , Feminino , Humanos , Proteínas Oncogênicas/antagonistas & inibidores , Neoplasias Ovarianas/terapia
4.
Gynecol Oncol ; 129(2): 364-71, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23391663

RESUMO

OBJECTIVES: The reported incidence of neoplasia identified at the time of risk-reducing salpingo-oophorectomy (RRSO) in germline BRCA1/2 mutation carriers ranges from 4 to 12% but long-term outcomes have not been described. We evaluated recurrence and survival outcomes of mutation carriers with neoplastic lesions identified at RRSO. METHODS: We identified BRCA1/2 mutation carriers with neoplasia at RRSO at three institutions. Data was collected on clinical variables, adjuvant treatment and follow-up. RESULTS: We identified 32 mutation carriers with invasive carcinomas (n=15) or high-grade intraepithelial neoplasia (n=17) that were not suspected prior to surgery. 26 occurred in BRCA1 and 6 in BRCA2 mutation carriers. Median and mean age for carcinomas were 50 years and 49.3 respectively, significantly younger than for intraepithelial neoplasm, median 53 years, and mean 55 years (p=0.04). For the 15 invasive carcinomas, median follow up was 88 months (range 45-172 months), 7 recurred (47%), median time to recurrence was 32.5 months and 3 have died of disease; 1 additional patient died of breast cancer. Overall survival was 73%, disease specific overall survival was 80% and disease free survival was 66%. For the 17 high-grade intraepithelial neoplasms, median follow up was 80 months (range 40-150), 4 were treated with chemotherapy. One recurred at 43 months and is currently not on therapy with a normal CA125, 16 months later. All patients with noninvasive neoplasia are alive. CONCLUSIONS: BRCA1 and BRCA2 mutation carriers with unsuspected invasive carcinoma at RRSO have a relatively high rate of recurrence despite predominantly early stage, small volume disease. High-grade intraepithelial neoplasms rarely recur as carcinoma and may not require adjuvant chemotherapy.


Assuntos
Neoplasias das Tubas Uterinas/terapia , Genes BRCA1 , Genes BRCA2 , Predisposição Genética para Doença , Neoplasias Ovarianas/terapia , Ovariectomia , Salpingectomia , Adulto , Idoso , Carcinoma in Situ/diagnóstico , Carcinoma in Situ/genética , Carcinoma in Situ/mortalidade , Carcinoma in Situ/terapia , Quimioterapia Adjuvante , Neoplasias das Tubas Uterinas/diagnóstico , Neoplasias das Tubas Uterinas/genética , Neoplasias das Tubas Uterinas/mortalidade , Feminino , Seguimentos , Humanos , Pessoa de Meia-Idade , Mutação , Invasividade Neoplásica , Metástase Neoplásica , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/mortalidade , Neoplasias Ovarianas/diagnóstico , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/mortalidade , Análise de Sobrevida , Resultado do Tratamento
5.
Br J Cancer ; 106(2): 333-43, 2012 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-22166800

RESUMO

BACKGROUND: Bortezomib is a proteasome inhibitor with minimal clinical activity as a monotherapy in solid tumours, but its combination with other targeted therapies is being actively investigated as a way to increase its anticarcinogenic properties. Here, we evaluate the therapeutic potential of co-treatment with bortezomib and indole-3-carbinol (I3C), a natural compound found in cruciferous vegetables, in human ovarian cancer. METHODS: We examined the effects of I3C, bortezomib and cisplatin in several human ovarian cancer cell lines. Synergy was determined using proliferation assays and isobologram analysis. Cell cycle and apoptotic effects were assessed by flow cytometry. The mechanism of I3C and bortezomib action was determined by RNA microarray studies, quantitative RT-PCR and western blotting. Antitumour activity of I3C and bortezomib was evaluated using an OVCAR5 xenograft mouse model. RESULTS: I3C sensitised ovarian cancer cell lines to bortezomib treatment through potent synergistic mechanisms. Combination treatment with bortezomib and I3C led to profound cell cycle arrest and apoptosis as well as disruptions to multiple pathways, including those regulating endoplasmic reticulum stress, cytoskeleton, chemoresistance and carcinogen metabolism. Moreover, I3C and bortezomib co-treatment sensitised ovarian cancer cells to the standard chemotherapeutic agents, cisplatin and carboplatin. Importantly, in vivo studies demonstrated that co-treatment with I3C and bortezomib significantly inhibited tumour growth and reduced tumour weight compared with either drug alone. CONCLUSION: Together, these data provide a novel rationale for the clinical application of I3C and bortezomib in the treatment of ovarian cancer.


Assuntos
Antineoplásicos/farmacologia , Ácidos Borônicos/farmacologia , Indóis/farmacologia , Neoplasias Ovarianas/patologia , Pirazinas/farmacologia , Animais , Bortezomib , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sinergismo Farmacológico , Feminino , Citometria de Fluxo , Humanos , Camundongos , Camundongos Nus , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase em Tempo Real , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Gynecol Oncol Rep ; 38: 100881, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34926762

RESUMO

BACKGROUND: Vaginal cancer is a rare malignancy making up 1-2% of all female genital tract cancers. Among vaginal cancers, sarcomas constitute 2% of malignant vaginal lesions, with leiomyosarcomas being the most common type of sarcoma. There is a paucity of data to guide treatment of vaginal sarcomas. This case report details a patient diagnosed with a gynecologic sarcoma during pregnancy who is subsequently treated for residual vaginal disease in the postpartum period with local resection and adjuvant vaginal brachytherapy. CASE: A 31-year-old gravida 4 para 0 who presented at 22-weeks gestation with vaginal bleeding to an outside hospital and expelled a mass 11 cm in diameter from the vagina during her admission. Findings were consistent with a high grade gynecologic sarcoma. She underwent planned cesarean section at 36 weeks gestational age with uterine pathology showing no sarcoma. At her 3 month postpartum visit she was found to have a 1 cm posterior vaginal wall lesion which was resected and consistent with vaginal sarcoma. She underwent adjuvant brachytherapy. CONCLUSION: This case demonstrates the challenges with obtaining a correct pathological diagnosis for pregnant patients with vaginal sarcoma during pregnancy. Surgical resection with negative margins remains an important treatment component. Given the low incidence of disease occurrence in pregnancy and rare number of cases reported in literature, further elucidation of timing of delivery and adjuvant treatment is warranted.

7.
Gynecol Oncol ; 115(3): 466-71, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19767065

RESUMO

OBJECTIVES.: The utility of hormone therapy in the management of uterine sarcomas is poorly defined. We hypothesize that estrogen receptor (ER) expression is common in uterine sarcomas, and carries prognostic significance. Further, we hypothesize that ER-positive uterine sarcomas respond to hormone therapy. METHODS.: We retrospectively reviewed charts of patients with uterine sarcomas. Stepwise Cox proportional hazards regression model was used to evaluate variables related to the risk of death: age, histology, stage, use of pelvic radiotherapy, and ER expression. In addition, we examined clinical outcomes in patients treated with aromatase inhibitors, megestrol acetate, depot medroxyprogesterone acetate, and tamoxifen. RESULTS.: Fifty-four patients underwent immunohistochemical staining, and 34 (63%) were ER-positive. Kaplan-Meier survival analysis and log-rank test indicated that patients with ER-positive sarcomas demonstrated improved overall survival when compared with ER-negative patients (median OS 36 vs. 16 months, p=0.004). Upon multivariate analysis, ER positivity retained significance as an independent predictor of survival (HR=0.32, CI 0.12-0.89, p=0.03). Four patients received hormonal treatment in the adjuvant setting and remained in remission (range of follow up: 18-68 months). Eighteen patients received hormone therapy in the setting of recurrent or progressive disease: fourteen (78%) demonstrated stable disease or complete or partial response (range of follow up: 6-124 months). CONCLUSIONS.: ER expression is common and is associated with improved overall survival in uterine sarcomas. Conducting immunohistochemical staining to ascertain ER status may aid with prognostication in this disease. Hormone therapy should be considered in patients with primary and recurrent ER-positive uterine sarcomas.


Assuntos
Receptores de Estrogênio/biossíntese , Sarcoma/metabolismo , Neoplasias Uterinas/metabolismo , Adenossarcoma/tratamento farmacológico , Adenossarcoma/metabolismo , Adenossarcoma/patologia , Antineoplásicos Hormonais/farmacologia , Inibidores da Aromatase/farmacologia , Carcinossarcoma/tratamento farmacológico , Carcinossarcoma/metabolismo , Carcinossarcoma/patologia , Feminino , Humanos , Estudos Retrospectivos , Sarcoma/tratamento farmacológico , Sarcoma do Estroma Endometrial/tratamento farmacológico , Sarcoma do Estroma Endometrial/metabolismo , Sarcoma do Estroma Endometrial/patologia , Neoplasias Uterinas/tratamento farmacológico , Neoplasias Uterinas/patologia
8.
Curr Oncol Rep ; 9(6): 485-93, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17991357

RESUMO

Our limited understanding of the natural biology of ovarian cancer, along with its low prevalence in the general population make early detection especially challenging. To be successful at the population level, an ovarian cancer screening test must prove its beneficial effect on ovarian cancer-specific mortality while achieving near-perfect specificity in order to minimize the harms resulting from false-positive results. No current screening tests for ovarian cancer fulfill these expectations. We review the current status and the challenges of ovarian cancer screening in the context of evidence-based principles that define a valuable cancer screening program.


Assuntos
Neoplasias Ovarianas/diagnóstico , Biomarcadores Tumorais/sangue , Ensaios Clínicos como Assunto , Feminino , Humanos , Programas de Rastreamento/métodos , Estadiamento de Neoplasias , Neoplasias Ovarianas/sangue , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/prevenção & controle , Sensibilidade e Especificidade
9.
Cancer Res ; 47(22): 6040-5, 1987 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-3117361

RESUMO

OVCA 433 human ovarian carcinoma cells secrete large amounts of plasminogen activator (PA), which consists of immunologically identifiable urokinase (UK) and tissue-type PA (tPA). Total extracellular PA activity is 95% inhibited by treatment of cells with 1 X 10(-7) M dexamethasone (Dex) for 3 days. This inhibition is both time and concentration dependent, with half-maximal inhibition occurring after 1.5 days with 1 X 10(-7) M Dex, or with 1 X 10(-9) M Dex for 3 days, respectively. Interestingly, the loss of UK activity precedes the loss of tPA activity, such that half-maximal inhibition of the two PA types occurs at 1 and 2 days, respectively. Dex treatment leads to approximately 50% inhibition of cell growth and pronounced morphological alterations, including marked enlargement, flattening, and multinucleation. Treatment of the cells with other classes of steroid hormones, i.e., estrogens, progestins, androgens, and mineralocorticoids, is without effect on UK and tPA activities, cell growth, or morphology. OVCA 433 cells contain about 14,000 nuclear glucocorticoid receptors (GR) per cell (measured at 37 degrees C), with an average affinity (Kd) for [3H]Dex of 6.6 X 10(-9) M. Only active glucocorticoids compete with [3H]Dex for nuclear GR binding sites. Our results demonstrate steroid-specific glucocorticoid responsiveness of ovarian carcinoma cells, a tumor cell type not usually considered hormonally responsive. Since almost 90% of ovarian carcinoma tumor biopsies contain GR (M. C. Galli, et al., Cancer (Phila.), 47: 1297-1302, 1981), it is possible that glucocorticoid sensitivity could be exploited clinically, particularly following the almost universal development of resistance to the chemotherapeutic drugs commonly used in this disease.


Assuntos
Aldosterona/farmacologia , Dexametasona/farmacologia , Hidrocortisona/farmacologia , Neoplasias Ovarianas/patologia , Esteroides/farmacologia , Ativador de Plasminogênio Tecidual/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Feminino , Humanos , Cinética , Neoplasias Ovarianas/metabolismo
10.
Cancer Res ; 48(12): 3502-6, 1988 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-3285997

RESUMO

The CA125 tumor marker is an antigenic determinant present on a high-molecular-weight glycoprotein expressed by more than 80% of newly diagnosed nonmucinous epithelial ovarian cancers. OVCA 433 human ovarian carcinoma cells express the CA125 marker at the cell surface and release large quantities of this antigen into culture medium. Here we show that release of CA125 by OVCA 433 cells is 90 to 95% inhibited by treatment with 1 x 10(-7) M dexamethasone, as determined using a biotin-based enzyme-linked immunosorbent assay utilizing OC125 monoclonal antibodies to CA125. The relative cell surface density of CA125 is also decreased following dexamethasone treatment as determined by immunofluorescence techniques using OC125 monoclonal antibodies. Inhibition of CA125 expression is specific for glucocorticoids, such as cortisol and dexamethasone, and does not occur with estrogens, progestins, androgens, or mineralocorticoids. CA125 inhibition is also dependent on the concentration of steroid used, with half-maximal and maximal inhibition by dexamethasone occurring at about 3 x 10(-9) M and 1 x 10(-7) M, respectively. Previous work has shown that OVCA 433 cells are growth inhibited by glucocorticoids and contain 14,000 glucocorticoid receptors per cell with an affinity for dexamethasone (Kd = 6.6 x 10(-9) M) which corresponds well with the concentration required for half-maximal CA125 inhibition. This correspondence, together with the specificity of CA125 inhibition for glucocorticoids, suggests that this effect is mediated by glucocorticoid receptors and is a specific biological effect of glucocorticoids on this cell type. Our results demonstrate glucocorticoid inhibition of CA125 expression by ovarian carcinoma cells and suggest that endogenous or therapeutically administered glucocorticoids can influence CA125 production by tumors in vivo.


Assuntos
Antígenos de Neoplasias/análise , Carcinoma/imunologia , Glucocorticoides/farmacologia , Neoplasias Ovarianas/imunologia , Antígenos Glicosídicos Associados a Tumores , Dexametasona/farmacologia , Relação Dose-Resposta a Droga , Feminino , Imunofluorescência , Humanos , Células Tumorais Cultivadas
11.
Cancer Res ; 60(19): 5329-33, 2000 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11034065

RESUMO

There is a clear association between germ-line BRCA1 mutations and inherited ovarian cancer; however, the association between BRCA1 mutations and sporadic ovarian cancer remains ambiguous. The frequency of BRCA1 promoter hypermethylation as an epigenetic means of BRCA1 inactivation was determined for a large, population-based cohort of ovarian cancer patients. BRCA1 promoter hypermethylation was determined by methylation-specific restriction digestion of tumor DNA, followed by Southern blot analysis and confirmed by methylation-specific PCR. BRCA1 promoter hypermethylation was observed in 12 of 98 ovarian tumors. BRCA1 methylation status of the primary tumor was conserved in six recurrent tumors after interim chemotherapy. None of the 12 tumors with BRCA1 promoter hypermethylation demonstrated BRCA1 protein expression by immunohistochemistry. BRCA1 methylation was only seen in ovarian cancer patients without a family history suggestive of a breast/ ovarian cancer syndrome. Therefore, the 12 BRCA1 methylated tumors represented 15% (12 of 81) of the sporadic cancers analyzed in this study. Although the clinical significance of BRCA1 promoter hypermethylation is yet to be determined, promoter hypermethylation may be an alternative to mutation in causing the inactivation of the BRCA1 tumor suppressor gene in sporadic ovarian cancer.


Assuntos
Metilação de DNA , Genes BRCA1/genética , Neoplasias Ovarianas/genética , Alelos , Southern Blotting , Cromossomos Humanos Par 17/genética , Estudos de Coortes , Feminino , Regulação Neoplásica da Expressão Gênica/fisiologia , Inativação Gênica/fisiologia , Mutação em Linhagem Germinativa , Humanos , Perda de Heterozigosidade , Pessoa de Meia-Idade , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Reação em Cadeia da Polimerase/métodos , Regiões Promotoras Genéticas/genética
12.
Cancer Res ; 60(23): 6744-9, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11118061

RESUMO

The majority of ovarian tumors arise from the transformation of the ovarian surface epithelial cells, a single layer of cells surrounding the ovary. To identify genes that may contribute to the malignant phenotype of ovarian cancers, cDNA representational difference analysis was used to compare expressed genes in primary cultures of normal human ovarian surface epithelium (HOSE) and ovarian tumor-derived epithelial cells from the Cedars-Sinai Ovarian Cancer (CSOC) repository. A total of 255 differentially expressed genes were identified, of which 160 and 95 were specifically expressed in HOSE and CSOC cells, respectively. Using cDNA array hybridization, the expression profiles of the genes identified by cDNA-representational difference analysis were examined in an additional 5 HOSE and 10 CSOC lines. The comparison of average signal of each gene revealed 44 HOSE-specific and 16 CSOC-specific genes that exhibited at least a 2.5-fold difference in expression. A large number of genes identified in this study encode membrane-associated or secreted proteins and, hence, may be useful as targets in the development of serum-based diagnostic markers for ovarian cancer. Very few genes associated with protein synthesis or metabolism were identified in this study, reflecting the lack of observable differences in phenotypic or growth characteristics between HOSE and CSOC cells. Northern blot analysis on a subset of these genes demonstrated comparable levels of gene expression as observed in the cDNA array hybridization.


Assuntos
Perfilação da Expressão Gênica , Neoplasias Ovarianas/genética , Ovário/fisiologia , Northern Blotting , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Feminino , Regulação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Hibridização de Ácido Nucleico , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Ovário/metabolismo , Células Tumorais Cultivadas
13.
Mol Endocrinol ; 3(6): 1006-13, 1989 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-2500590

RESUMO

Previous studies from our laboratory have demonstrated that OVCA 433 human ovarian carcinoma cells are glucocorticoid responsive by several criteria and contain high affinity, saturable, steroid-specific glucocorticoid receptors. These cells secrete both mammalian plasminogen activators (PAs), urokinase (uPA) and tissue-type PA (tPA). Treatment of OVCA 433 cells with 1 x 10(-7) M dexamethasone (Dex) for 4 days led to 77% and 83% reductions in the extracellular activities of uPA and tPA, respectively, released into serum-free conditioned medium during a 1-h period. Dex treatment led to a 71% decrease in the rate of extracellular uPA antigen accumulation, as determined by enzyme-linked immunosorbent assay, as well as a 73% reduction in steady state uPA mRNA levels. In contrast, Dex treatment led to only a 42% decrease in the rate of extracellular tPA antigen accumulation and a 48% decrease in tPA mRNA levels; such decreases were insufficient to account for the 83% reduction in tPA activity. Thus, while Dex-induced decreases in uPA antigen and mRNA levels accounted for all but 6% of the decrease in uPA activity, a large discrepancy existed between the magnitudes of decreased tPA activity and decreased tPA antigen and mRNA levels. OVCA 433 cells produce both PAI-1 and PAI-2, two specific PA inhibitors. Treatment of cells with 1 x 10(-7) M Dex for 4 days led to a 3.3-fold increase in the rate of extracellular PAI-1 accumulation, with little or no effect on PAI-2 accumulation.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Carcinoma/metabolismo , Dexametasona/farmacologia , Neoplasias Ovarianas/metabolismo , Ativador de Plasminogênio Tecidual/antagonistas & inibidores , Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Carcinoma/enzimologia , Células Cultivadas , Feminino , Humanos , Neoplasias Ovarianas/enzimologia
14.
Gene ; 238(2): 375-85, 1999 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-10570965

RESUMO

Comparative hybridization of cDNA arrays is a powerful tool for the measurement of differences in gene expression between two or more tissues. We optimized this technique and employed it to discover genes with potential for the diagnosis of ovarian cancer. This cancer is rarely identified in time for a good prognosis after diagnosis. An array of 21,500 unknown ovarian cDNAs was hybridized with labeled first-strand cDNA from 10 ovarian tumors and six normal tissues. One hundred and thirty-four clones are overexpressed in at least five of the 10 tumors. These cDNAs were sequenced and compared to public sequence databases. One of these, the gene HE4, was found to be expressed primarily in some ovarian cancers, and is thus a potential marker of ovarian carcinoma.


Assuntos
Biomarcadores Tumorais/genética , Hibridização de Ácido Nucleico , Neoplasias Ovarianas/genética , Ovário/metabolismo , Células Cultivadas , Células Clonais , DNA Complementar , Feminino , Humanos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sensibilidade e Especificidade
15.
Cancer Epidemiol Biomarkers Prev ; 9(10): 1107-11, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11045795

RESUMO

Ovarian cancer screening protocols generally have been limited by inadequate recognition of the normal behavior of tumor markers in women at risk of ovarian cancer. We have characterized the behavior of five serum tumor markers in a large cohort of healthy women and examined the implications for screening. Serial measurements of CA125, HER-2/neu, urinary gonadotropin peptide, lipid-associated sialic acid, and Dianon marker 70/K were obtained during 6 years of follow-up of 1257 healthy women at high risk of ovarian cancer. We analyzed individual-specific tumor marker behavior and explored methods that can exploit this information to develop individual-specific screening rules. These five tumor markers behaved approximately independently. Substantial heterogeneity was observed among women in the behavior of each tumor marker, particularly CA125. Intraclass correlation (ICC), or the proportion of total variability that occurs between individuals, was approximately 0.6 for log-transformed CA125 and urinary gonadotropin peptide, and less than 0.4 for the other markers. This degree of tumor marker heterogeneity among healthy women, and the relative independence of these markers, has important implications for screening and diagnostic tests. Independence of markers results in the clinically useful fact that the combined false positive rate from screening with multiple markers may be estimated by the sum of individual false positive rates. Heterogeneity of tumor marker patterns in healthy women implies that a fixed screening cutoff level is suboptimal to a degree that depends strongly on ICC. Using information on longitudinal measurements and ICC, individual-specific screening rules may be developed with the potential to improve early detection of ovarian cancer.


Assuntos
Biomarcadores Tumorais/análise , Programas de Rastreamento , Neoplasias Ovarianas/diagnóstico , Adulto , Idoso , Antígeno Ca-125/análise , Estudos de Coortes , Diagnóstico Diferencial , Reações Falso-Positivas , Feminino , Nível de Saúde , Humanos , Pessoa de Meia-Idade , Neoplasias Ovarianas/patologia , Valores de Referência , Sensibilidade e Especificidade
16.
Obstet Gynecol ; 85(5 Pt 1): 729-34, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7724103

RESUMO

OBJECTIVE: To compare the prognostic importance of certain molecular biologic characteristics (HER-2/neu and p53 gene overexpression, DNA ploidy, and the S-phase fraction) to standard clinical-pathologic factors used to predict survival in patients with endometrial carcinoma. METHODS: We reviewed archival specimens from 128 patients with endometrial cancer diagnosed during the period 1985-1987. One hundred four cases were eligible for inclusion in the study. Immunohistochemistry was used to detect p53 and HER-2/neu overexpression. We used flow cytometry to calculate DNA ploidy and the S-phase fraction. Life-table analysis and Cox multiple regression were used to analyze clinical and molecular data with respect to survival. RESULTS: International Federation of Obstetrics and Gynecology stage, nuclear grade, lymph-vascular space invasion, and adverse histopathologic features each significantly correlated with poor outcome (each at P < or = .001). Overexpression of p53 was demonstrated in 15% of the tumors and was associated with a 12% probability of 5-year survival, compared to a 90% probability of 5-year survival for the p53-negative cohort (P = .0001). Thirty percent of the tumors were aneuploid, which was also associated with poor prognosis (P = .0003). HER-2/neu overexpression and an S-phase fraction greater than 10% showed similar trends, but were not statistically significant. On multivariate analysis, p53 overexpression was the strongest independent prognosticator of survival (P = .0001). CONCLUSION: Molecular characteristics provide objective data that may be useful in predicting prognosis in patients with endometrial cancer. Further investigation of molecular and genetic characteristics are needed to refine our diagnostic and treatment modalities.


Assuntos
Carcinoma/genética , DNA de Neoplasias/genética , Neoplasias do Endométrio/genética , Regulação Neoplásica da Expressão Gênica , Genes erbB-2 , Genes p53 , Idoso , Carcinoma/mortalidade , Carcinoma/patologia , Neoplasias do Endométrio/mortalidade , Neoplasias do Endométrio/patologia , Feminino , Seguimentos , Humanos , Estadiamento de Neoplasias , Ploidias , Prognóstico , Taxa de Sobrevida
17.
Obstet Gynecol ; 78(6): 1023-6, 1991 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-1945201

RESUMO

We report four sisters whose maternal pedigree suggested a site-specific ovarian cancer syndrome, whereas their paternal pedigree closely fit the Cancer Family Syndrome (Lynch II). Eliciting a complete family history, both maternal and paternal, is important for defining the correct ovarian cancer syndrome. Once the definition is made, the patient and other family members at risk must be counseled and encouraged to begin the appropriate schedule of screening and intervention. These recommendations may be summarized as follows: 1) site-specific ovarian carcinoma: screening with physical examination, CA 125, and ultrasound, and bilateral oophorectomy after childbearing has been completed; 2) breast/ovary syndrome: screening for ovarian cancer as above, mammography and bilateral oophorectomy as above, and possible prophylactic mastectomy; and 3) Lynch Cancer Family Syndrome: screening for ovarian cancer as above, colonoscopy and endometrial biopsy, and prophylactic hysterectomy and bilateral oophorectomy once childbearing is complete.


Assuntos
Síndromes Neoplásicas Hereditárias/diagnóstico , Neoplasias Ovarianas/diagnóstico , Adulto , Feminino , Humanos , Pessoa de Meia-Idade , Síndromes Neoplásicas Hereditárias/genética , Neoplasias Ovarianas/genética , Linhagem
18.
Fertil Steril ; 66(4): 499-507, 1996 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8816606

RESUMO

OBJECTIVE: To review critically the published data regarding the proposed association of ovulation induction, infertility, and an increased risk of ovarian cancer. DESIGN: A medline search was conducted to identify all case reports, epidemiologic studies, and clinical investigations containing data relevant to infertility, treatment of infertility, and the associated risk of ovarian cancer. Additional sources were obtained from reference lists of original research and review articles. Particular emphasis was placed on the most recently published reports examining these associations. RESULTS: Four case-control studies and three retrospective cohort studies, as well as a large meta-analysis of three additional case-control studies were identified as presenting the most pertinent clinical data. CONCLUSION: Currently available data in the literature suggest that an association between ovulation induction and ovarian cancer does not indicate necessarily a causal effect. Infertility alone is an independent risk factor for the development of ovarian cancer. Nulliparous women with refractory infertility may harbor a particularly high risk of ovarian cancer, irrespective of their use of fertility drugs. Furthermore, the apparent association between fertility drug use and ovarian cancer may arise because these women are the most likely to have used ovulation-stimulating agents as part of their infertility treatment. Close clinical surveillance of patients before, during, and after treatment of infertility is warranted.


Assuntos
Infertilidade Feminina/complicações , Neoplasias Ovarianas/etiologia , Indução da Ovulação/efeitos adversos , Feminino , Humanos , Fatores de Risco
19.
J Soc Gynecol Investig ; 3(3): 99-105, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8796816

RESUMO

OBJECTIVE: The HER-2/neu proto-oncogene (also known as c-erbB2, neu, and HER2) encodes a 185-kDa transmembrane glycoprotein with intrinsic tyrosine kinase activity that resembles the receptor for epidermal growth factor. Aberrant HER-2/neu protein overexpression occurs in human gynecologic adenocarcinomas, including those of the ovary, endometrium, breast, fallopian tube, and cervix, and is secondary to gene amplification and/or overexpression of the p185HER2 protein. METHODS: A Medline literature search revealed numerous studies on HER-2/neu and tumor biology, cancer prognosis, and therapeutic targeting. We present a review of the literature pertinent to gynecologic malignancies. RESULTS: Overexpression of HER-2/neu was found to be a poor prognostic factor for survival from advanced-stage ovarian cancer, node-positive breast cancer, and endometrial cancer. Although a specific ligand has not been definitively identified, HER-2/neu may have unusually complex activation pathways because it can form both homodimeric and heterodimeric associations with other related receptor proteins. Preliminary findings suggest that serum HER-2/neu levels may be used as a tumor marker in a subset of patients with tumors that overexpress the HER-2/neu receptor. Receptor-targeted therapeutics currently being studied include the use of receptor antibodies, liposomally delivered antisense DNA, antigen-activated cytotoxic lymphocytes, and adenovirus-mediated E1A delivery to overexpressing tumor cells. CONCLUSION: HER-2/neu appears to play an important role in the biologic behavior of ovarian, endometrial, and breast cancers and holds potential as a target for oncogene-directed therapies.


Assuntos
Genes erbB-2 , Neoplasias dos Genitais Femininos/genética , Biomarcadores Tumorais/análise , Neoplasias da Mama/genética , Neoplasias da Mama/mortalidade , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/mortalidade , Neoplasias das Tubas Uterinas/genética , Neoplasias das Tubas Uterinas/mortalidade , Feminino , Regulação Neoplásica da Expressão Gênica , Neoplasias dos Genitais Femininos/metabolismo , Neoplasias dos Genitais Femininos/mortalidade , Neoplasias dos Genitais Femininos/patologia , Humanos , MEDLINE , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/mortalidade , Prognóstico , Proto-Oncogene Mas , Receptor ErbB-2/análise , Receptor ErbB-2/biossíntese , Taxa de Sobrevida , Transcrição Gênica , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/mortalidade
20.
Int J Gynecol Cancer ; 3(5): 304-310, 1993 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11578362

RESUMO

Patients with unresectable locally recurrent gynecologic malignancies pose a difficult therapeutic challenge. Conventional therapies are frequently unsuccessful and offer only marginal palliation. In this study, interstitial 192iridium-needle implants and concomitant infusional 5-fluorouracil (5FU) and cisplatin (CDDP) or carboplatin (CBDCA) chemotherapy were used to treat 14 women with recurrent pelvic tumors. Malignancies of the cervix, endometrium ovary, tube and vulva are represented; all patients were heavily pretreated. Twenty interstitial implants were performed in these 14 patients. Needle distributions and doses were individualized to accommodate the recurrent tumor volumes. Tumor responses were seen in 12 patients (six complete and six partial responses). Four women remain clinically free of disease and four are alive with disease at 18-34 months of follow-up. There were no severe acute toxicities, however, four patients have subsequently developed fistulae associated with tumor progression. Although longer follow-up is required, the high response rate, wide applicability and acceptable toxicity observed in this heavily pretreated patient population warrant further study of combined interstitial radiation and chemotherapy.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA