Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Gynecol Oncol ; 165(3): 568-576, 2022 06.
Article in English | MEDLINE | ID: mdl-35370009

ABSTRACT

OBJECTIVE: Aberrant DNA methylation is an early event in carcinogenesis which could be leveraged to detect ovarian cancer (OC) in plasma. METHODS: DNA from frozen OC tissues, benign fallopian tube epithelium (FTE), and buffy coats from cancer-free women underwent reduced representation bisulfite sequencing (RRBS) to identify OC MDMs. Candidate MDM selection was based on receiver operating characteristic (ROC) discrimination, methylation fold change, and low background methylation among controls. Blinded biological validation was performed using methylated specific PCR on DNA extracted from independent OC and FTE FFPE tissues. MDMs were tested using Target Enrichment Long-probe Quantitative Amplified Signal (TELQAS) assays in pre-treatment plasma from women newly diagnosed with OC and population-sampled healthy women. A random forest modeling analysis was performed to generate predictive probability of disease; results were 500-fold in silico cross-validated. RESULTS: Thirty-three MDMs showed marked methylation fold changes (10 to >1000) across all OC subtypes vs FTE. Eleven MDMs (GPRIN1, CDO1, SRC, SIM2, AGRN, FAIM2, CELF2, RIPPLY3, GYPC, CAPN2, BCAT1) were tested on plasma from 91 women with OC (73 (80%) high-grade serous (HGS)) and 91 without OC; the cross-validated 11-MDM panel highly discriminated OC from controls (96% (95% CI, 89-99%) specificity; 79% (69-87%) sensitivity, and AUC 0.91 (0.86-0.96)). Among the 5 stage I/II HGS OCs included, all were correctly identified. CONCLUSIONS: Whole methylome sequencing, stringent filtering criteria, and biological validation yielded candidate MDMs for OC that performed with high sensitivity and specificity in plasma. Larger plasma-based OC MDM studies, including testing of pre-diagnostic specimens, are warranted.


Subject(s)
DNA Methylation , Ovarian Neoplasms , Biomarkers, Tumor/genetics , CELF Proteins/genetics , Carcinoma, Ovarian Epithelial/diagnosis , Carcinoma, Ovarian Epithelial/genetics , Feasibility Studies , Female , Genetic Markers , Humans , Nerve Tissue Proteins/genetics , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics , Transaminases/genetics
2.
Sci Transl Med ; 6(240): 240ra75, 2014 Jun 11.
Article in English | MEDLINE | ID: mdl-24920661

ABSTRACT

Many solid tumors contain an overabundance of phospholipid ethers relative to normal cells. Capitalizing on this difference, we created cancer-targeted alkylphosphocholine (APC) analogs through structure-activity analyses. Depending on the iodine isotope used, radioiodinated APC analog CLR1404 was used as either a positron emission tomography (PET) imaging ((124)I) or molecular radiotherapeutic ((131)I) agent. CLR1404 analogs displayed prolonged tumor-selective retention in 55 in vivo rodent and human cancer and cancer stem cell models. (131)I-CLR1404 also displayed efficacy (tumor growth suppression and survival extension) in a wide range of human tumor xenograft models. Human PET/CT (computed tomography) and SPECT (single-photon emission computed tomography)/CT imaging in advanced-cancer patients with (124)I-CLR1404 or (131)I-CLR1404, respectively, demonstrated selective uptake and prolonged retention in both primary and metastatic malignant tumors. Combined application of these chemically identical APC-based radioisosteres will enable personalized dual modality cancer therapy of using molecular (124)I-CLR1404 tumor imaging for planning (131)I-CLR1404 therapy.


Subject(s)
Neoplasms/diagnosis , Neoplasms/drug therapy , Phosphorylcholine/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Female , Humans , Mice , Phosphorylcholine/analogs & derivatives , Positron-Emission Tomography/methods , Tomography, Emission-Computed, Single-Photon/methods , Tomography, X-Ray Computed/methods , Xenograft Model Antitumor Assays
3.
Oncologist ; 12(8): 942-51, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17766653

ABSTRACT

Carcinoid tumors are neuroendocrine malignancies that frequently metastasize and secrete hormones that cause debilitating symptoms in patients. In this study we report the effects of valproic acid (VPA), a drug long used for the treatment of epilepsy, on the growth and neuroendocrine phenotype of human carcinoid cancer cells. VPA treatment of gastrointestinal and pulmonary carcinoid cells resulted in a dose-dependent inhibition of cancer cell growth. Western blot analysis revealed degradation of cyclin D1 and an increase in cyclin-dependent kinases p21 and p27 with VPA treatment. Flow cytometry confirmed that the mechanism of VPA-induced growth inhibition is G(1) phase cell cycle arrest. Furthermore, VPA suppressed expression of the neuroendocrine tumor marker chromogranin A. In addition to these effects, VPA also increased levels of full-length Notch-1 and the active Notch-1 intracellular domain. Luciferase reporter assays incorporating the centromere-binding factor 1 (CBF-1) binding site and the achaete-scute complex-like 1 (ASCL-1) promoter confirmed the functional activity of VPA-induced Notch-1. Transfection of Notch-1 small-interfering RNA into carcinoid tumor cells blocked the effects of VPA on Notch-1 activation, ASCL-1 suppression, p21 induction, and cell growth inhibition. VPA also suppressed growth of carcinoid tumors in vivo in a mouse tumor xenograft experiment. These findings confirm the important role of Notch-1 in regulating the growth and neuroendocrine phenotype of carcinoid tumor cells. On the basis of this study, a clinical trial of VPA for patients with advanced carcinoid cancer will be conducted. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoid Tumor/metabolism , Gastrointestinal Neoplasms/metabolism , Lung Neoplasms/metabolism , Receptor, Notch1/metabolism , Valproic Acid/pharmacology , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation , Chromogranin A/antagonists & inhibitors , Chromogranin A/metabolism , Cyclin D1/metabolism , Genes, Reporter , Humans , Luciferases/analysis , Luciferases/genetics , Male , Mice , Mice, Nude , Phenotype , RNA Interference , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
4.
Mol Cancer Ther ; 6(3): 1151-8, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17363508

ABSTRACT

Glycogen synthase kinase-3beta (GSK-3beta) is an important regulator of cell proliferation and survival. Conflicting observations have been reported regarding the regulation of GSK-3beta and extracellular signal-regulated kinase (ERK1/2) in cancer cells. In this study, we found that raf-1 activation in human medullary thyroid cancer cells, TT cells, resulted in phosphorylation of GSK-3beta. Inactivation of GSK-3beta in TT cells with well-known GSK-3beta inhibitors such as lithium chloride (LiCl) and SB216763 is associated with both growth suppression and a significant decrease in neuroendocrine markers such as human achaete-scute complex-like 1 and chromogranin A. Growth inhibition by GSK-3beta inactivation was found to be associated with cell cycle arrest due to an increase in the levels of cyclin-dependent kinase inhibitors such as p21, p27, and p15. Additionally, LiCl-treated TT xenograft mice had a significant reduction in tumor volume compared with those treated with control. For the first time, we show that GSK-3beta is a key downstream target of the raf-1 pathway in TT cells. Also, our results show that inactivation of GSK-3beta alone is sufficient to inhibit the growth of TT cells both in vitro and in vivo.


Subject(s)
Carcinoma, Medullary/prevention & control , Glycogen Synthase Kinase 3/antagonists & inhibitors , Indoles/pharmacology , Maleimides/pharmacology , Proto-Oncogene Proteins c-raf/metabolism , Thyroid Neoplasms/prevention & control , Adjuvants, Immunologic/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Carcinoma, Medullary/enzymology , Carcinoma, Medullary/pathology , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cell Proliferation/drug effects , Chromogranin A/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p15/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Lithium Chloride/pharmacology , Mice , Mice, Nude , NIH 3T3 Cells/drug effects , Phosphorylation , Proto-Oncogene Proteins c-raf/genetics , Signal Transduction/drug effects , Thyroid Neoplasms/enzymology , Thyroid Neoplasms/pathology , Xenograft Model Antitumor Assays
5.
J Biol Chem ; 281(52): 39819-30, 2006 Dec 29.
Article in English | MEDLINE | ID: mdl-17090547

ABSTRACT

The role of NOTCH1 as an oncogene or tumor suppressor appears to be cell type-specific. Medullary thyroid cancer (MTC) cells characteristically express the transcription factor ASCL1 (achaete-scute complex-like 1) as well as high levels of the neuroendocrine (NE) markers calcitonin and chromogranin A (CgA). In this study, we show that the active NOTCH1 intracellular domain is absent in human MTC tumor tissue samples and MTC-TT cells. To determine the effects of NOTCH1 expression, we created a doxycycline-inducible NOTCH1 intracellular domain in MTC cells (TT-NOTCH cells). Treatment of TT-NOTCH cells with doxycycline led to dose-dependent induction of NOTCH1 protein with corresponding decreases in ASCL1 protein and NE hormones. ASCL1 promoter-reporter assay and Northern analysis revealed that ASCL1 reduction by NOTCH1 activation is predominantly via silencing of ASCL1 gene transcription. Overexpression of ASCL1 in MTC cells indicated that CgA expression is highly dependent on the levels of ASCL1. This was further confirmed by experiments using small interfering RNA against ASCL1, in which reduction in ASCL1 led to reduction in both CgA and calcitonin. Furthermore, we demonstrate that NOTCH1 signaling activation leads to ERK1/2 phosphorylation, but that reduction in NE markers is independent of ERK1/2 activation. Activation of NOTCH1 resulted in significant MTC cell growth inhibition. Notably, reduction in MTC cell growth was dependent on the level of NOTCH1 protein present. Moreover, no increase in growth upon expression of ASCL1 in NOTCH1-activated cells was observed, indicating that the growth suppression observed upon NOTCH1 activation is independent of ASCL1 reduction. Mechanistically, we show that MTC cell growth inhibition by NOTCH1 is mediated by cell cycle arrest associated with up-regulation of p21.


Subject(s)
Carcinoma, Medullary/genetics , Cell Proliferation , Growth Inhibitors/genetics , Intracellular Space/physiology , Neurosecretory Systems/physiology , Receptor, Notch1/biosynthesis , Receptor, Notch1/genetics , Thyroid Neoplasms/genetics , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/physiology , Carcinoma, Medullary/metabolism , Cell Cycle/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/physiology , Growth Inhibitors/physiology , Humans , Intracellular Space/chemistry , Phenotype , Protein Structure, Tertiary/genetics , Receptor, Notch1/physiology , Thyroid Neoplasms/metabolism , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...