Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Am J Surg Pathol ; 47(5): 580-588, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36939053

RESUMEN

Among gynecologic cancers, uterine serous carcinoma (USC) has been shown to be human epidermal growth factor receptor 2 (HER2) amplified and trastuzumab has been included in the recent National Comprehensive Cancer Network (NCCN) guidelines for treatment of advanced stage or recurrent USC with HER2 overexpression/amplification. There is limited literature suggesting that a subset of high-grade endometrioid carcinomas with aberrant p53 expression may also be HER2 amplified and these patients could benefit from the addition of targeted therapy. We identified 59 p53-aberrant (mismatch repair proficient) FIGO 3 endometrioid carcinomas of the uterus. HER2 immunohistochemistry was performed in all 59 tumors and HER2 fluorescence in situ hybridization (FISH) was performed in 52 of the 59 cases. Four of the 59 cases were HER2 3+ by immunohistochemistry (6.7%), using the American Society of Clinical Oncology/College of American Pathologists (ASCO/CAP) 2007, 2013, and 2018 criteria. HER2 FISH was performed in 3 of the 4 cases and was amplified in all 3. Nine, 8, and 7 tumors showed 2+ HER2 staining when applying 2018, 2013, and 2007 criteria, respectively, FISH was performed in 7 tumors and none were amplified. An additional 4 cases did not perfectly meet the 2018 ASCO/CAP criteria but were assigned a score of 2+, none were amplified by HER2 FISH. The remaining 42 cases showed 1+ or no staining for HER2, FISH was successfully performed in 38 tumors and none showed amplification. Approximately half of the tumors fulfilled criteria for HER2-low or HER2-very low (10 HER2-low and 20 HER2-very low). Our data shows that a subset of p53-aberrant high-grade endometrial endometrioid carcinoma express HER2 and these patients may benefit from the addition of targeted therapy. The role of targeted therapy in HER2-low gynecologic carcinoma is currently unexplored.


Asunto(s)
Neoplasias de la Mama , Carcinoma Endometrioide , Cistadenocarcinoma Seroso , Neoplasias Uterinas , Humanos , Femenino , Amplificación de Genes , Carcinoma Endometrioide/genética , Carcinoma Endometrioide/terapia , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Hibridación Fluorescente in Situ , Receptor ErbB-2 , Neoplasias Uterinas/patología , Cistadenocarcinoma Seroso/genética , Neoplasias de la Mama/genética , Biomarcadores de Tumor/genética
2.
Arch Pathol Lab Med ; 145(6): 687-691, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32649220

RESUMEN

CONTEXT.­: Endometrial serous carcinoma is an aggressive subtype of endometrial cancer with the highest rate of recurrence and mortality among all histotypes. A recent clinical trial showed prolonged progression-free survival in advanced-stage and recurrent human epidermal growth factor receptor 2 (HER2)-positive endometrial serous carcinoma when trastuzumab was added to the standard chemotherapy regimen. This targeted therapeutic approach was recently endorsed by the National Comprehensive Cancer Network clinical guidelines. There is a growing interest among clinicians to obtain HER2 testing in endometrial serous carcinoma, and pathologists need to be prepared to recognize the unique characteristics of HER2 protein expression and gene amplification in these tumors and apply specific HER2 scoring criteria. OBJECTIVE.­: To provide a historical overview of targeted HER2 therapy in endometrial serous carcinoma and to summarize key findings from recent studies on the specific features of HER2 protein expression and gene amplification relative to other tumor types. Endometrial carcinoma-specific HER2 testing criteria are proposed based on evidence in the existing literature. DATA SOURCES.­: Sources comprise review of the literature and personal experience of the author. CONCLUSIONS.­: HER2 protein overexpression and/or gene amplification is present in approximately 25% to 30% of endometrial serous carcinomas, providing an opportunity for targeted therapy. Pathologists play a key role in tumor HER2 testing and scoring to ensure appropriate patient selection and successful clinical outcome.


Asunto(s)
Cistadenocarcinoma Seroso/genética , Neoplasias Endometriales/genética , Amplificación de Genes , Pruebas Genéticas/métodos , Patología Clínica/métodos , Receptor ErbB-2/genética , Antineoplásicos Inmunológicos/uso terapéutico , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Cistadenocarcinoma Seroso/diagnóstico , Cistadenocarcinoma Seroso/metabolismo , Neoplasias Endometriales/diagnóstico , Neoplasias Endometriales/metabolismo , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Endometrio/patología , Femenino , Humanos , Terapia Molecular Dirigida/métodos , Patología Clínica/normas , Patología Clínica/tendencias , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/metabolismo , Estándares de Referencia , Trastuzumab/uso terapéutico
3.
Biomed Res Int ; 2020: 8965459, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33150184

RESUMEN

Reports increasingly suggest that Chinese herbal medicine (CHM) has been used to treat ovarian cancer (OvCa) with a good curative effect; however, the molecular mechanisms underlying CHM are still unclear. In this retrospective study, we explored CHM's molecular targets for the treatment of OvCa based on clinical data and network pharmacology. We used the Kaplan-Meier method and Cox regression analysis to verify the survival rate of 202 patients with CHM-treated OvCa. The association between CHM and survival time was analyzed by bivariate correlation. A target network of CHM active ingredients against OvCa was established via network pharmacology. Cox regression analysis showed that CHM is an independent favorable prognostic factor. The median survival time was 91 months in the CHM group and 65 months in the non-CHM group. The survival time of FIGO stage III patients in the two groups was 91 months and 52 months, and the median survival period of FIOG stage IV patients was 60 months and 22 months, respectively (p < 0.001). Correlation analysis demonstrated that 12 herbs were closely associated with prognosis, especially in regard to the long-term benefits. Bioinformatics analysis indicated that the anti-OvCa activity of these 12 herbs occurs mainly through the regulation of apoptosis-related protein expression, which promotes OvCa cell apoptosis and inhibits OvCa development. They also regulate the progress of OvCa treatment by promoting or inhibiting protein expression on the p53 signaling pathway and by inhibiting the NF-κB signaling pathway by directly inhibiting NF-κB.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Carcinoma Endometrioide/terapia , Cistadenocarcinoma Seroso/terapia , Medicamentos Herbarios Chinos/uso terapéutico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Ováricas/terapia , Adulto , Anciano , Pueblo Asiatico , Carcinoma Endometrioide/etnología , Carcinoma Endometrioide/genética , Carcinoma Endometrioide/mortalidad , Cistadenocarcinoma Seroso/etnología , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/mortalidad , Femenino , Humanos , Estimación de Kaplan-Meier , Medicina Tradicional China , Persona de Mediana Edad , Terapia Molecular Dirigida/métodos , FN-kappa B/genética , FN-kappa B/metabolismo , Estadificación de Neoplasias , Neoplasias Ováricas/etnología , Neoplasias Ováricas/genética , Neoplasias Ováricas/mortalidad , Pronóstico , Modelos de Riesgos Proporcionales , Estudios Retrospectivos , Transducción de Señal , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
4.
Cancer Biomark ; 28(1): 65-72, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32176632

RESUMEN

OBJECTIVE: LGR4 expression in serous ovarian cancer paraffin-embedded tissues and fresh tissues were investigated, and its expression associated with clinicopathological parameters and prognosis in serous ovarian cancer was explored. METHODS: From Dec, 2009 to Jan, 2020, 122 paraffin-embedded serous ovarian cancer patients and 41 paired paratumor tissues who were both diagnosed and operated at the memorial hospital of Sun Yat-sen University and Integrated Hospital of Traditional Chinese Medicine, Southern Medical University were selected in this research, respectively, and all of these tissues were performed by immunohistochemistry (IHC) with a polyclonal antibody for LGR4. Meanwhile, from Aug, 2013 to Mar, 2019, 15 cases of serous ovarian cancer fresh tissues and 15 cases of paratumor fresh tissues who were operated at Integrated Hospital of Traditional Chinese Medicine, Southern Medical University were performed with Quantitative Real-time PCR to detect the mRNA expression of LGR4, respectively. RESULTS: LGR4 expression was much higher both in paraffin-embedded and fresh cancer tissues than that in paratumor tissues, respectively, and its expression was associated with recurrence free survival and overall survival in serous ovarian cancer patients. Moreover, in a multivariate model LGR4 was an indeed independent predictor of poor survival in serous ovarian cancer patients. CONCLUSION: LGR4 is upregulated in serous ovarian cancer, and LGR4 is an indeed useful independent prognostic predictor in serous ovarian cancer, and it may provide important clinical value of serous ovarian cancer.


Asunto(s)
Carcinoma Epitelial de Ovario/metabolismo , Cistadenocarcinoma Seroso/metabolismo , Neoplasias Ováricas/metabolismo , Receptores Acoplados a Proteínas G/biosíntesis , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/metabolismo , Carcinoma Epitelial de Ovario/genética , Carcinoma Epitelial de Ovario/patología , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patología , Supervivencia sin Enfermedad , Femenino , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Adhesión en Parafina , Pronóstico , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Análisis de Supervivencia , Regulación hacia Arriba
5.
Cancer Med ; 8(9): 4315-4329, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31197964

RESUMEN

Here we sought to determine the relationship between STAT3 activity and Galectin-3 (Gal-3) and to investigate the cytotoxic effect of PectaSol-C Modified Citrus Pectin (Pect-MCP) as a specific competitive inhibitor of Galectin-3 (Gal-3) in combination with Paclitaxel (PTX) to kill the ovarian cancer cell SKOV-3 multicellular tumor spheroid (MCTS). To this order, SKOV-3 cells in 2D and 3D cultures were treated with exogenous Gal-3 for the assessment of STAT3 activity. Two-way ANOVA main effect and IC50 of each drug Paclitaxel (PTX) and Pect-MCP or in combination were obtained from MTT assay results. The phosphorylated STAT3 levels, migration, invasion, integrin mRNA and p-AKTser473 levels were assessed in the absence or presence of each drug alone or in combination. Gal-3 expression levels were assessed in human serous ovarian cancer (SOC) specimens and its correlation with different integrin mRNA levels was further assessed. Our results showed that Gal-3 expression level was significantly increased in MCTS compared to monolayer SKOV-3 cells which triggered STAT3 phosphorylation. Moreover, Pect-MCP synergized with PTX to kill SKOV3 MCTS through abrogation of STAT3 activity and reduced expression of its downstream target HIF-1α, reduced integrin mRNA levels, and subsequently decreased AKT activity. There were higher expression levels of Gal-3 in human high-grade SOC specimens compared to the normal ovary and borderline SOC which positively and significantly correlated with α5, ß2 and ß6 integrin mRNA levels. Together, these results revealed for the first time that Pect-MCP could be considered as a potential drug to enhance the PTX effect on ovarian cancer cells MCTS through inhibition of STAT3 activity.


Asunto(s)
Cistadenocarcinoma Seroso/metabolismo , Galectina 3/metabolismo , Neoplasias Ováricas/metabolismo , Paclitaxel/farmacología , Pectinas/farmacología , Factor de Transcripción STAT3/metabolismo , Proteínas Sanguíneas , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cistadenocarcinoma Seroso/tratamiento farmacológico , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patología , Sinergismo Farmacológico , Femenino , Galectinas , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Integrinas/genética , Clasificación del Tumor , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Esferoides Celulares/citología , Esferoides Celulares/efectos de los fármacos , Células Tumorales Cultivadas
6.
Int J Mol Sci ; 20(5)2019 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-30866519

RESUMEN

Nearly one-third of patients with high-grade serous ovarian cancer (HGSC) do not respond to initial treatment with platinum-based therapy. Genomic and clinical characterization of these patients may lead to potential alternative therapies. Here, the objective is to classify non-responders into subsets using clinical and molecular features. Using patients from The Cancer Genome Atlas (TCGA) dataset with platinum-resistant or platinum-refractory HGSC, we performed a genome-wide unsupervised cluster analysis that integrated clinical data, gene copy number variations, gene somatic mutations, and DNA promoter methylation. Pathway enrichment analysis was performed for each cluster to identify the targetable processes. Following the unsupervised cluster analysis, three distinct clusters of non-responders emerged. Cluster 1 had overrepresentation of the stage IV disease and suboptimal debulking, under-expression of miRNAs and mRNAs, hypomethylated DNA, "loss of function" TP53 mutations, and the overexpression of genes in the PDGFR pathway. Cluster 2 had low miRNA expression, generalized hypermethylation, MUC17 mutations, and significant activation of the HIF-1 signaling pathway. Cluster 3 had more optimally cytoreduced stage III patients, overexpression of miRNAs, mixed methylation patterns, and "gain of function" TP53 mutations. However, the survival for all clusters was similar. Integration of genomic and clinical data from patients that do not respond to chemotherapy has identified different subgroups or clusters. Pathway analysis further identified the potential alternative therapeutic targets for each cluster.


Asunto(s)
Biología Computacional/métodos , Cistadenocarcinoma Seroso/clasificación , Metilación de ADN , Dosificación de Gen , Mutación , Neoplasias Ováricas/clasificación , Análisis por Conglomerados , Cistadenocarcinoma Seroso/tratamiento farmacológico , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patología , Bases de Datos Genéticas , Epigénesis Genética , Femenino , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Platino (Metal)/uso terapéutico , Aprendizaje Automático no Supervisado
7.
Oncogene ; 36(28): 4014-4024, 2017 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-28288142

RESUMEN

High-grade serous (HGS) ovarian cancer accounts for 90% of all ovarian cancer-related deaths. However, factors that drive HGS ovarian cancer tumor growth have not been fully elucidated. In particular, comprehensive analysis of the metabolic requirements of ovarian cancer tumor growth has not been performed. By analyzing The Cancer Genome Atlas mRNA expression data for HGS ovarian cancer patient samples, we observed that six enzymes of the folic acid metabolic pathway were overexpressed in HGS ovarian cancer samples compared with normal ovary samples. Systematic knockdown of all six genes using short hairpin RNAs (shRNAs) and follow-up functional studies demonstrated that serine hydroxymethyl transferase 1 (SHMT1) was necessary for ovarian cancer tumor growth and cell migration in culture and tumor formation in mice. SHMT1 promoter analysis identified transcription factor Wilms tumor 1 (WT1) binding sites, and WT1 knockdown resulted in reduced SHMT1 transcription in ovarian cancer cells. Unbiased large-scale metabolomic analysis and transcriptome-wide mRNA expression profiling identified reduced levels of several metabolites of the amino sugar and nucleotide sugar metabolic pathways, including sialic acid N-acetylneuraminic acid (Neu5Ac), and downregulation of pro-oncogenic cytokines interleukin-6 and 8 (IL-6 and IL-8) as unexpected outcomes of SHMT1 loss. Overexpression of either IL-6 or IL-8 partially rescued SHMT1 loss-induced tumor growth inhibition and migration. Supplementation of culture medium with Neu5Ac stimulated expression of IL-6 and IL-8 and rescued the tumor growth and migratory phenotypes of ovarian cancer cells expressing SHMT1 shRNAs. In agreement with the ovarian tumor-promoting role of Neu5Ac, treatment with Neu5Ac-targeting glycomimetic P-3Fax-Neu5Ac blocked ovarian cancer growth and migration. Collectively, these results demonstrate that SHMT1 controls the expression of pro-oncogenic inflammatory cytokines by regulating sialic acid Neu5Ac to promote ovarian cancer tumor growth and migration. Thus, targeting of SHMT1 and Neu5Ac represents a precision therapy opportunity for effective HGS ovarian cancer treatment.


Asunto(s)
Carcinogénesis/genética , Proliferación Celular/genética , Cistadenocarcinoma Seroso/patología , Citocinas/genética , Glicina Hidroximetiltransferasa/fisiología , Ácido N-Acetilneuramínico/metabolismo , Neoplasias Ováricas/patología , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Línea Celular Tumoral , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/metabolismo , Citocinas/metabolismo , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Mediadores de Inflamación/metabolismo , Ratones , Ratones Desnudos , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo
8.
OMICS ; 15(5): 281-92, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21332407

RESUMEN

To identify specific proteins associated with chemotherapeutic responses, we analyzed protein expression patterns in stage IIIc primary serous epithelial ovarian cancer tissues displaying differential responses to first-line postoperative adjuvant chemotherapy. The expression profiles of five chemoresistant tissues [progression-free survival (PFS) ≤12 months] and five chemosensitive tissues (PFS ≥48 months) were analyzed with 2D electrophoresis, and the spot intensities of differentially expressed proteins were quantified. To validate these proteins as markers for chemoresistant disease, we analyzed tissues from an additional 17 patients. All the patients were allocated to the over- or underexpressing group according to protein spot intensity, and survival analysis was performed. In chemoresistant tissues, four proteins (thioredoxin domain containing four, similar to RIKEN cDNA 1700016G05, tubulin α 1A chain, and the pyruvate dehydrogenase E1-ß subunit precursor) were overexpressed, and seven proteins [keratin 1, vitamin D-binding protein, creatine kinase B, annexin V, SH3-containing guanine nucleotide exchange factor (SGEF), tryptophan-aspartate repeat protein-1 (WDR 1), and WDR 1 isoform 1] were underexpressed. The underexpression of keratin 1, creatine kinase B, annexin V, SGEF, WDR1, and WDR1 isoform 1 were significantly correlated with poor overall survival. A combination of keratin 1 and SGEF showed the highest sensitivity of 0.800, specificity of 0.917, PPV of 0.800, and NPV of 0.917 in predicting chemoresistant disease. These proteins may be useful as predictive markers of chemoresistant disease. However, further analyses in large-scale should be performed before they can be considered reliable predictive markers of chemoresistant disease.


Asunto(s)
Cistadenocarcinoma Seroso/metabolismo , Resistencia a Antineoplásicos/genética , Neoplasias Ováricas/metabolismo , Proteómica , Adulto , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patología , Electroforesis en Gel Bidimensional , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Pronóstico , Curva ROC , Análisis de Supervivencia
9.
Int J Cancer ; 112(1): 14-25, 2004 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-15305371

RESUMEN

With the goal of identifying genes with a differential pattern of expression between ovarian serous papillary carcinomas (OSPCs) and normal ovarian (NOVA) epithelium and using this knowledge for the development of novel diagnostic and therapeutic markers for ovarian cancer, we used oligonucleotide microarrays with probe sets complementary to 12,533 genes to analyze the gene expression profiles of 10 primary OSPC cell lines, 2 established OSPC cell lines (UCI-101, UCI-107) and 5 primary NOVA epithelial cultures. Unsupervised analysis of gene expression data identified 129 and 170 genes that exhibited >5-fold upregulation and downregulation, respectively, in primary OSPC compared to NOVA. Genes overexpressed in established OSPC cell lines had little correlation with those overexpressed in primary OSPC, highlighting the divergence of gene expression that occurs as a result of long-term in vitro growth. Hierarchical clustering of the expression data readily distinguished normal tissue from primary OSPC. Laminin, claudin 3, claudin 4, tumor-associated calcium signal transducers 1 and 2 (TROP-1/Ep-CAM, TROP-2), ladinin 1, S100A2, SERPIN2 (PAI-2), CD24, lipocalin 2, osteopontin, kallikrein 6 (protease M), kallikrein 10, matriptase (TADG-15) and stratifin were among the most highly overexpressed genes in OSPC compared to NOVA. Downregulated genes in OSPC included transforming growth factor-beta receptor III, platelet-derived growth factor receptor alpha, SEMACAP3, ras homolog gene family member I (ARHI), thrombospondin 2 and disabled-2/differentially expressed in ovarian carcinoma 2 (Dab2/DOC2). Differential expression of some of these genes, including claudin 3, claudin 4, TROP-1 and CD24, was validated by quantitative RT-PCR and flow cytometry on primary OSPC and NOVA. Immunohistochemical staining of formalin-fixed, paraffin-embedded tumor specimens from which primary OSPC cultures were derived further confirmed differential expression of CD24 and TROP-1/Ep-CAM markers on OSPC vs. NOVA. These results, obtained with highly purified primary cultures of ovarian cancer, highlight important molecular features of OSPC and may provide a foundation for the development of new type-specific therapies against this disease.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma Papilar/genética , Cistadenocarcinoma Seroso/genética , Perfilación de la Expresión Génica , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Ováricas/genética , Serina Endopeptidasas , Adulto , Anciano , Carcinoma Papilar/diagnóstico , Células Cultivadas , Cistadenocarcinoma Seroso/diagnóstico , Epitelio/metabolismo , Epitelio/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Persona de Mediana Edad , Neoplasias Ováricas/diagnóstico , Ovario/metabolismo , Ovario/patología , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Zhonghua Fu Chan Ke Za Zhi ; 31(3): 169-72, 1996 Mar.
Artículo en Chino | MEDLINE | ID: mdl-8758793

RESUMEN

OBJECTIVE: To explore the effects of antisense erbB2 on the biological behaviours and on chemotherapeutic drug sensitivity in human ovarian cancer cells. METHODS: A recombinant retroviral expression vector: pDOR-erbB-neo was constructed which contain the neomycin resistant gene and the erbB2 cDNA 3.8kb antisense fragment. pDOR-erbB-neo was introduced into human ovarian cancer cell line SKOV3 by lipofectin. The cells were selected with geneticin (G418). The cells transfected with pDOR-erbB-neo were named SKOV3-A2. RESULTS: Southern blot analysis confirmed the presence of the antisense erbB2 in transfected cells. Transfected cells showed no obvious changes in morphology. Compared with parental cells and with pDOR-neo transfected cells used as controls, the growth and DNA synthesis of antisense transfected cells, SKOV3-A2, were inhibited. Transfection with pDOR-erbB-neo rendered the cells significantly, more sensitive to chemotherapeutic drugs (5-fluorouracil, cisplatinum) than the parental cells. CONCLUSIONS: The effectiveness and the potentiality of antisense erbB2 in ovarian cancer gene therapy is demonstrated. c-erbB2 oncogene may be related to drug resistance in ovarian cancer cell.


Asunto(s)
Cistadenocarcinoma Seroso/patología , ADN sin Sentido , Genes erbB-2 , Neoplasias Ováricas/patología , Antineoplásicos/farmacología , Cisplatino/farmacología , Cistadenocarcinoma Seroso/genética , ADN Complementario , Resistencia a Antineoplásicos , Femenino , Fluorouracilo/farmacología , Humanos , Neoplasias Ováricas/genética , Transfección , Células Tumorales Cultivadas
11.
Genes Chromosomes Cancer ; 14(3): 220-2, 1995 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8589040

RESUMEN

Chromosome 6 has been shown to contain at band q27 a minimal region of deletion associated with epithelial ovarian cancers and AF6, a gene disrupted in acute myeloid leukemia with t(6;11)(q27;q23). Using rapid amplification of cDNA ends by polymerase chain reaction, the breakpoint in AF6 was confirmed and a cDNA clone identified. This clone was used as a probe to screen a chromosome 6 cosmid library, and a single cosmid C-109F0645 was isolated. By fluorescence in situ hybridization, C-109F0465 was found to map distal to the critically deleted region associated with ovarian malignancies. AF6 is therefore distinct from and lies telomeric to this region.


Asunto(s)
Cromosomas Humanos Par 6/genética , Cistadenocarcinoma Seroso/genética , Genes Supresores de Tumor , Cinesinas/genética , Miosinas/genética , Neoplasias Ováricas/genética , Proto-Oncogenes , Eliminación de Secuencia , Factores de Transcripción , Enfermedad Aguda , Mapeo Cromosómico , Cromosomas Humanos Par 11/genética , Cromosomas Humanos Par 11/ultraestructura , Cromosomas Humanos Par 6/ultraestructura , Cósmidos/genética , ADN Complementario/genética , ADN de Neoplasias/genética , Proteínas de Unión al ADN/genética , Femenino , N-Metiltransferasa de Histona-Lisina , Humanos , Hibridación Fluorescente in Situ , Leucemia Mieloide/genética , Proteína de la Leucemia Mieloide-Linfoide , Proteínas de Fusión Oncogénica/genética , Translocación Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA