Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Immunol Immunother ; 70(9): 2701-2719, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34244816

ABSTRACT

Recombinant agonists that activate co-stimulatory and cytokine receptors have shown limited clinical anticancer utility, potentially due to narrow therapeutic windows, the need for coordinated activation of co-stimulatory and cytokine pathways and the failure of agonistic antibodies to recapitulate signaling by endogenous ligands. RTX-240 is a genetically engineered red blood cell expressing 4-1BBL and IL-15/IL-15Rα fusion (IL-15TP). RTX-240 is designed to potently and simultaneously stimulate the 4-1BB and IL-15 pathways, thereby activating and expanding T cells and NK cells, while potentially offering an improved safety profile through restricted biodistribution. We assessed the ability of RTX-240 to expand and activate T cells and NK cells and evaluated the in vivo efficacy, pharmacodynamics and tolerability using murine models. Treatment of PBMCs with RTX-240 induced T cell and NK cell activation and proliferation. In vivo studies using mRBC-240, a mouse surrogate for RTX-240, revealed biodistribution predominantly to the red pulp of the spleen, leading to CD8 + T cell and NK cell expansion. mRBC-240 was efficacious in a B16-F10 melanoma model and led to increased NK cell infiltration into the lungs. mRBC-240 significantly inhibited CT26 tumor growth, in association with an increase in tumor-infiltrating proliferating and cytotoxic CD8 + T cells. mRBC-240 was tolerated and showed no evidence of hepatic injury at the highest feasible dose, compared with a 4-1BB agonistic antibody. RTX-240 promotes T cell and NK cell activity in preclinical models and shows efficacy and an improved safety profile. Based on these data, RTX-240 is now being evaluated in a clinical trial.


Subject(s)
4-1BB Ligand/genetics , Cell- and Tissue-Based Therapy , Erythrocytes/metabolism , Gene Expression , Genetic Therapy , Interleukin-15/genetics , 4-1BB Ligand/metabolism , Animals , Cell- and Tissue-Based Therapy/methods , Erythroid Precursor Cells/metabolism , Female , Flow Cytometry , Genes, Reporter , Genetic Engineering , Genetic Therapy/methods , Humans , Interleukin-15/metabolism , Mice , Models, Animal , Protein Binding , Treatment Outcome , Xenograft Model Antitumor Assays
2.
Mol Cancer Ther ; 16(3): 417-427, 2017 03.
Article in English | MEDLINE | ID: mdl-28148715

ABSTRACT

Selinexor, a selective inhibitor of nuclear export (SINE) compound targeting exportin-1, has previously been shown to inhibit melanoma cell growth in vivo We hypothesized that combining selinexor with antibodies that block or disrupt T-cell checkpoint molecule signaling would exert superior antimelanoma activity. In vitro, selinexor increased PDCD1 and CTLA4 gene expression in leukocytes and induced CD274 gene expression in human melanoma cell lines. Mice bearing syngeneic B16F10 melanoma tumors demonstrated a significant reduction in tumor growth rate in response to the combination of selinexor and anti-PD-1 or anti-PD-L1 antibodies (P < 0.05). Similar results were obtained in B16F10-bearing mice treated with selinexor combined with anti-CTLA4 antibody. Immunophenotypic analysis of splenocytes by flow cytometry revealed that selinexor alone or in combination with anti-PD-L1 antibody significantly increased the frequency of both natural killer cells (P ≤ 0.050) and CD4+ T cells with a Th1 phenotype (P ≤ 0.050). Further experiments indicated that the antitumor effect of selinexor in combination with anti-PD-1 therapy persisted under an alternative dosing schedule but was lost when selinexor was administered daily. These data indicate that the efficacy of selinexor against melanoma may be enhanced by disrupting immune checkpoint activity. Mol Cancer Ther; 16(3); 417-27. ©2017 AACRSee related article by Tyler et al., p. 428.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Hydrazines/pharmacology , Karyopherins/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Triazoles/pharmacology , Animals , B7-H1 Antigen/antagonists & inhibitors , CTLA-4 Antigen/antagonists & inhibitors , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Humans , Immunomodulation/drug effects , Melanoma, Experimental , Mice , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , T-Lymphocytes/immunology , Exportin 1 Protein
3.
Cancer Lett ; 383(2): 309-317, 2016 12 28.
Article in English | MEDLINE | ID: mdl-27693556

ABSTRACT

In previous studies we demonstrated that targeting the nuclear exporter protein exportin-1 (CRM1/XPO1) by a selective inhibitor of nuclear export (SINE) compound is a viable therapeutic strategy against Non-Hodgkin Lymphoma (NHL). Our studies along with pre-clinical work from others led to the evaluation of the lead SINE compound, selinexor, in a phase 1 trial in patients with CLL or NHL (NCT02303392). Continuing our previous work, we studied combinations of selinexor-dexamethasone (DEX) and selinexor-everolimus (EVER) in NHL. Combination of selinexor with DEX or EVER resulted in enhanced cytotoxicity in WSU-DLCL2 and WSU-FSCCL cells which was consistent with enhanced apoptosis. Molecular analysis showed enhancement in the activation of apoptotic signaling and down-regulation of XPO1. This enhancement is consistent with the mechanism of action of these drugs in that both selinexor and DEX antagonize NF-κB (p65) and mTOR (EVER target) is an XPO1 cargo protein. SINE compounds, KPT-251 and KPT-276, showed activities similar to CHOP (cyclophosphamide-hydroxydaunorubicin-oncovin-prednisone) regimen in subcutaneous and disseminated NHL xenograft models in vivo. In both animal models the anti-lymphoma activity of selinexor is enhanced through combination with DEX or EVER. The in vivo activity of selinexor and related SINE compounds relative to 'standard of care' treatment is consistent with the objective responses observed in Phase I NHL patients treated with selinexor. Our pre-clinical data provide a rational basis for testing these combinations in Phase II NHL trials.


Subject(s)
Active Transport, Cell Nucleus/drug effects , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Dexamethasone/pharmacology , Everolimus/pharmacology , Hydrazines/pharmacology , Karyopherins/antagonists & inhibitors , Lymphoma, Non-Hodgkin/drug therapy , Protein Kinase Inhibitors/pharmacology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Triazoles/pharmacology , Acrylamides/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cyclophosphamide/pharmacology , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Drug Synergism , Humans , Karyopherins/metabolism , Lymphoma, Non-Hodgkin/enzymology , Lymphoma, Non-Hodgkin/pathology , Mice, Inbred ICR , Mice, SCID , Oxadiazoles/pharmacology , Prednisone/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Thiazoles/pharmacology , Time Factors , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelA/metabolism , Tumor Burden/drug effects , Vincristine/pharmacology , Xenograft Model Antitumor Assays , Exportin 1 Protein
4.
Mol Cancer Res ; 12(3): 464-76, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24269953

ABSTRACT

UNLABELLED: The PI3K-AKT signaling pathway regulates all phenotypes that contribute to progression of human cancers, including breast cancer. AKT mediates signal relay by phosphorylating numerous substrates, which are causally implicated in biologic responses such as cell growth, survival, metabolic reprogramming, migration, and invasion. Here a new AKT substrate is identified, the adherens junction protein Afadin, which is phosphorylated by AKT at Ser1718. Importantly, under conditions of physiologic IGF-1 signaling and oncogenic PI3K and AKT, Afadin is phosphorylated by all AKT isoforms, and this phosphorylation elicits a relocalization of Afadin from adherens junctions to the nucleus. Also, phosphorylation of Afadin increased breast cancer cell migration that was dependent on Ser1718 phosphorylation. Finally, nuclear localization of Afadin was observed in clinical breast cancer specimens, indicating that regulation of Afadin by the PI3K-AKT pathway has pathophysiologic significance. IMPLICATIONS: Phosphorylation of the adhesion protein Afadin by AKT downstream of the PI3K pathway, leads to redistribution of Afadin and controls cancer cell migration.


Subject(s)
Adherens Junctions/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement/physiology , Microfilament Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Adherens Junctions/genetics , Breast Neoplasms/genetics , Cell Culture Techniques , Cell Growth Processes/physiology , Cell Line, Tumor , Female , Humans , Microfilament Proteins/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction
5.
Clin Exp Metastasis ; 27(3): 161-72, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20213325

ABSTRACT

The present study investigated the intracellular regulation of E-cadherin in ovarian carcinoma. E-cadherin expression and regulation by Snail and Pak1 were studied in ES-2 and OVCAR-3 ovarian cancer cells in vitro. Twist1, Zeb1 and Vimentin mRNA expression and HIF-1alpha protein expression were analyzed in 80 and 189 clinical specimens, respectively. OVCAR-3 cells incubated with an anti-E-cadherin antibody formed smaller and looser spheroids compared to controls. Snail silencing using Small Hairpin RNA in ES-2 cells reduced invasion and MMP-2 activity, with unaltered cellular morphology. Using dominant negative (DN) and constitutively active (CA) Pak1 constructs, we found that DN Pak1 ES-2 and OVCAR-3 clones had reduced attachment to matrix proteins, invasion and MMP-2 activity compared to CA and wild-type cells. DN Pak1 ES-2 cells also bound less to LP9 mesothelial cells. DN Pak1 OVCAR-3 cells had lower Vimentin levels. Snail expression was lower in cultured effusions compared to primary carcinomas, and was cytoplasmic rather than nuclear. Twist1 (P < 0.001), Zeb1 (P = 0.003) and Vimentin (P = 0.03) mRNA expression was significantly higher in solid metastases compared to primary carcinomas and effusions. HIF-1alpha protein expression was lower in effusions compared to primary carcinomas and solid metastases (P = 0.033). Our data suggest that the previously reported E-cadherin re-expression in ovarian carcinoma effusions is regulated by Pak1. The transient nature of E-cadherin expression during ovarian carcinoma progression is probably the result of partial epithelial-to-mesenchymal transition (EMT) and the reverse process of mesenchymal-to-epithelial-like transition (MET). Expression of the EMT-related molecules Twist, Zeb1, Vimentin and HIF-1alpha is anatomic site-dependent in ovarian carcinoma.


Subject(s)
Epithelial Cells/metabolism , Mesoderm/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Vimentin/metabolism , Base Sequence , Cadherins/metabolism , Cell Line, Tumor , Epithelial Cells/pathology , Epithelium/pathology , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Homeodomain Proteins/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mesoderm/pathology , Nuclear Proteins/metabolism , Ovarian Neoplasms/genetics , Pleural Effusion, Malignant/genetics , Pleural Effusion, Malignant/metabolism , Pleural Effusion, Malignant/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Snail Family Transcription Factors , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism , Twist-Related Protein 1/metabolism , Zinc Finger E-box-Binding Homeobox 1 , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
6.
Hum Pathol ; 41(6): 794-804, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20153512

ABSTRACT

The AKT signaling pathway is crucial for cancer cell survival. The objective of this study was to analyze the expression and clinical role of this pathway in serous ovarian carcinoma. Phospho-AKT and phospho-mammalian target of rapamycin protein expression was studied in 269 ovarian carcinomas (159 effusions, 38 primary carcinomas, 72 solid metastases) using immunohistochemistry. The association between AKT, mammalian target of rapamycin, and DJ-1 in effusions was quantitatively analyzed using flow cytometry. AKT phosphorylation status in effusions was further studied using Western blotting. Phospho-AKT and phospho-mammalian target of rapamycin were detected in the majority of tumors at all anatomical sites. Phospho-AKT expression in effusions was higher in grade 3 versus grades 1 and 2 tumors (P = .013). Flow cytometry analysis showed association between AKT, mammalian target of rapamycin, and DJ-1 expression (P < .001). Higher phospho-AKT Thr308/pan-AKT ratio by Western blotting was associated with more advanced International Federation of Gynecology and Obstetrics stage (P = .018) and a trend for poor response to chemotherapy at first disease recurrence (P = .051). Higher phospho-mammalian target of rapamycin protein expression in effusions by immunohistochemistry was associated with poor progression-free survival for patients with postchemotherapy effusions (P = .005). Phospho-mammalian target of rapamycin was an independent predictor of poor progression-free survival for patients with postchemotherapy effusions (P = .03). The association between activated AKT and mammalian target of rapamycin expression and clinicopathologic parameters of aggressive disease, including shorter patient survival, provides further evidence regarding the central role of this signaling pathway in ovarian carcinoma.


Subject(s)
Biomarkers, Tumor/biosynthesis , Cystadenoma, Serous/metabolism , Ovarian Neoplasms/metabolism , Protein Serine-Threonine Kinases/biosynthesis , Adult , Aged , Aged, 80 and over , Ascitic Fluid/metabolism , Cystadenoma, Serous/mortality , Cystadenoma, Serous/secondary , Female , Humans , Intracellular Signaling Peptides and Proteins , Middle Aged , Neoplasm Staging , Oncogene Proteins/biosynthesis , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/secondary , Phosphoproteins/biosynthesis , Phosphorylation , Pleural Effusion, Malignant/metabolism , Prognosis , Protein Deglycase DJ-1 , Proto-Oncogene Proteins c-akt/biosynthesis , Survival Rate , TOR Serine-Threonine Kinases
7.
Virchows Arch ; 449(5): 520-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17024425

ABSTRACT

Unlike most epithelial cancers, E-cadherin expression is upregulated in ovarian carcinoma effusions compared with corresponding primary tumors. In the present study, we analyzed the anatomic site-specific expression of transcription factors that negatively regulate E-cadherin in ovarian carcinoma. Using reverse-transcription polymerase chain reaction, mRNA in situ hybridization, and Western blotting, we analyzed the expression and localization of the Snail, Slug, and SIP1 transcription factors and E-cadherin in 78 effusions, 41 primary carcinomas, and 15 solid metastases. Slug mRNA and protein expression was highest in metastases (p=0.042 and p<0.001, respectively). Snail mRNA was comparable at all anatomic sites, but higher protein expression was found in primary tumors and solid metastases compared with effusions (p<0.001). SIP1 mRNA expression was higher in effusions (p<0.001) compared to other sites. Confocal microscopy analysis of fresh and cultured cells from effusion specimens revealed cytoplasmic localization of the Snail protein in primary tumor cells, with a nuclear shift following culturing of these cells. In conclusion, E-cadherin and its negative regulators show site-dependent expression in ovarian carcinoma. In solid tumors, E-cadherin is negatively regulated by Snail and Slug. In effusions, SIP1 may be the main regulator of E-cadherin, but with a lesser level of suppression compared with primary tumors and solid metastases.


Subject(s)
Cadherins/genetics , Ovarian Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Female , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization , Middle Aged , Molecular Sequence Data , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/genetics , RNA, Messenger/analysis , RNA-Binding Proteins/analysis , RNA-Binding Proteins/genetics , Repressor Proteins/genetics , Snail Family Transcription Factors , Transcription Factors/analysis , Transcription Factors/genetics
8.
Lung Cancer ; 54(3): 309-17, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16996643

ABSTRACT

Snail, Slug and Sip1 regulate cadherin and protease expression and mediate epithelial-mesenchymal transition in cancer. We analyzed the expression of cadherins and matrix metalloproteinases (MMP) and their transcriptional regulators in malignant mesothelioma (MM). One hundred and ten MM specimens (86 solid, 24 effusions) and 10 non-malignant effusions with reactive mesothelial cells (RMC) were analyzed for E-cadherin, N-cadherin and P-cadherin protein expression using immunhistochemistry. MM effusions were further analyzed for expression of Snail, Slug, Sip1, E-cadherin, MMP-2, MMP-9, MT1-MMP (MMP-14) and the MMP inhibitor TIMP-2, and for MMP-2 and MMP-9 activity using RT-PCR, Western blotting, immunhistochemistry and zymography. Results were analyzed for relationship with specimen type (biopsy versus effusion) and anatomic site (pleural versus peritoneal). E-cadherin, N-cadherin and P-cadherin expression was found in 69/110 (63%), 87/110 (79%) and 84/110 (76%) MM cases, respectively. Pleural and peritoneal MM showed comparable expression, but all three cadherins were upregulated in effusions compared to solid tumors (p<0.001). RMC were uniformly negative for E-cadherin and N-cadherin, and showed P-cadherin expression in 7/10 specimens. Immunohistochemistry localized MMP-2, MMP-9 and TIMP-2 to MM cells in 11/15, 14/15 and 8/15 effusions, respectively. RT-PCR showed direct association between MMP-2 mRNA expression level and the levels of MT1-MMP (p=0.027) and TIMP-2 (p=0.011). Snail protein expression showed positive association with MT1-MMP (p=0.016) and TIMP-2 (p=0.02) mRNA expression, but its expression was unrelated to MMP-2 and MMP-9 expression or activity. Snail, Slug and Sip1 levels did not show inverse association with E-cadherin levels. Our data show that E-cadherin and N-cadherin are selectively expressed in malignant mesothelial cells, and that P-cadherin and N-cadherin are expressed with similar frequency in MM. In agreement with our earlier data for ovarian carcinoma, cadherin expression is upregulated in effusions compared to solid lesions. The increased E-cadherin expression in effusions may be related to lack of negative regulation at the epigenetic level. The relationship between Snail and MMP in MM is uncertain at present.


Subject(s)
Cadherins/metabolism , Matrix Metalloproteinases/metabolism , Mesothelioma/metabolism , Nerve Tissue Proteins/metabolism , Pleural Neoplasms/metabolism , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Adult , Aged , Aged, 80 and over , Cadherins/analysis , Cadherins/genetics , Female , Humans , Immunohistochemistry , Male , Matrix Metalloproteinases/analysis , Matrix Metalloproteinases/genetics , Mesothelioma/chemistry , Mesothelioma/pathology , Middle Aged , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/genetics , Pleural Effusion, Malignant/chemistry , Pleural Effusion, Malignant/metabolism , Pleural Effusion, Malignant/pathology , Pleural Neoplasms/chemistry , Pleural Neoplasms/pathology , RNA, Messenger/analysis , RNA, Messenger/metabolism , RNA-Binding Proteins/analysis , RNA-Binding Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Snail Family Transcription Factors , Tissue Inhibitor of Metalloproteinase-2/analysis , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism , Transcription Factors/analysis , Transcription Factors/genetics
9.
Cancer ; 103(8): 1631-43, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15742334

ABSTRACT

BACKGROUND: It was demonstrated previously that the Snail family of transcription factors and Smad-interacting protein 1 (Sip1) regulate E-cadherin and matrix metalloproteinase 2 (MMP-2) expression, cellular morphology, and invasion in carcinoma. For the current study, the authors analyzed the relation between the expression of Snail, Slug, and Sip1; the expression of MMP-2 and E-cadherin; and clinical parameters in patients with metastatic ovarian and breast carcinoma. METHODS: One hundred one fresh-frozen, malignant effusions from patients who were diagnosed with gynecologic carcinomas (78 ovarian carcinomas and 23 breast carcinomas) were studied for mRNA expression of Snail, Slug, Sip1, MMP-2, and E-cadherin using reverse transcriptase-polymerase chain reaction analysis. Snail mRNA and E-cadherin protein expression levels also were studied in ovarian carcinoma effusions using in situ hybridization and immunocytochemistry. The results were analyzed for possible correlation with clinicopathologic parameters in both tumor types. RESULTS: E-cadherin mRNA expression was lower in breast carcinoma (P = 0.001), whereas Snail expression was higher (P = 0.003). The Snail/E-cadherin ratio (P < 0.001) and the Sip1/E-cadherin ratio (P = 0.002) were higher in breast carcinomas. Sip1 mRNA expression (P < 0.001) and Slug mRNA expression (P < 0.001) were correlated with the expression of MMP-2 in ovarian carcinomas. The Sip1/E-cadherin ratio was higher in primary ovarian carcinomas at the time of diagnosis compared with postchemotherapy ovarian carcinoma effusions (P = 0.003), higher in Stage IV tumors compared with Stage III tumors (P = 0.049), and higher in pleural effusions compared with peritoneal effusions (P = 0.044). In a univariate survival analysis of patients with ovarian carcinoma, a high Sip1/E-cadherin ratio predicted poor overall survival (P = 0.018). High E-cadherin mRNA expression predicted better disease-free survival (P = 0.023), with a similar trend for a low Slug/E-cadherin ratio (P = 0.07). High Snail mRNA expression predicted shorter effusion-free survival (P = 0.008), disease-free survival (P = 0.03), and overall survival (P = 0.008) in patients with breast carcinoma. CONCLUSIONS: Transcription factors that regulate E-cadherin were expressed differentially in metastatic ovarian and breast carcinoma. Snail may predict a poor outcome in patients who have breast carcinoma metastatic to effusions. E-cadherin expression generally was conserved in effusions from patients with ovarian carcinoma, but the subset of patients with postulated Sip1-induced repression of this adhesion molecule had a significantly worse outcome. This finding was in agreement with the stronger suppression of E-cadherin by Snail and Sip1 in breast carcinoma effusions, a clinical condition associated with extremely poor survival.


Subject(s)
Breast Neoplasms/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Ovarian Neoplasms/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Adenocarcinoma, Clear Cell/genetics , Adenocarcinoma, Clear Cell/metabolism , Adenocarcinoma, Clear Cell/secondary , Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/metabolism , Adenocarcinoma, Mucinous/secondary , Adult , Aged , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cadherins/genetics , Cadherins/metabolism , Carcinoma, Ductal/genetics , Carcinoma, Ductal/metabolism , Carcinoma, Ductal/secondary , Carcinoma, Lobular/genetics , Carcinoma, Lobular/metabolism , Carcinoma, Lobular/secondary , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/metabolism , Cystadenocarcinoma, Serous/secondary , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Female , Homeodomain Proteins/metabolism , Humans , Immunoenzyme Techniques , In Situ Hybridization , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Middle Aged , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Snail Family Transcription Factors , Survival Rate , Transcription Factors/metabolism , Zinc Finger E-box Binding Homeobox 2
SELECTION OF CITATIONS
SEARCH DETAIL
...