Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Oncol Rep ; 34(6): 2925-34, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26397133

ABSTRACT

In tobacco-associated lung cancers, the protein kinase B/mammalian target of rapamycin (Akt/mTOR) pathway frequently is activated by nicotine and its metabolite 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK). The aim of the present study was to examine the effects of early or late intervention with rapamycin in NNK-induced lung adenoma and progression to adenocarcinoma in female A/J mice. At 7 weeks of age, 40 mice/each carcinogen group received one dose of 10 µmol NNK i.p. Three weeks later, the early intervention groups (25/group) were fed diets containing 0, 8 or 16 ppm rapamycin. The mice were sacrificed after 17 or 34 weeks of drug exposure and tumors were evaluated via histopathology. For late intervention (late adenoma and adenocarcinoma stage), groups of 15 mice were administered diets containing 8 or 16 ppm rapamycin starting 20 weeks after NNK treatment and continuing for 17 weeks before evaluation of tumor progression. Administration of 8 or 16 ppm rapamycin as an early or a late stage intervention significantly suppressed lung adenoma and adenocarcinoma formation (p<0.01-0.0001) after 17 or 34 weeks of exposure. The effect was more pronounced (>50­60% tumor inihibition; p<0.0001) at the early intervention and the size of NNK-induced tumors decreased from >2.10 to <~0.75 mm3 (p=0.0056). Lung tumors harvested from mice exposed to rapamycin showed a significant decrease in p-mTOR, p-S6K1, PCNA and Bcl-xL as compared with controls in the early and late stage intervention studies. These observations suggest that rapamycin is highly effective even with administration after dysplastic adenoma or early adenocarcinoma stages and is useful for high-risk lung cancer patients.


Subject(s)
Adenocarcinoma/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Lung Neoplasms/drug therapy , Nicotiana/adverse effects , Sirolimus/administration & dosage , Adenocarcinoma/chemically induced , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Carcinogens/toxicity , Disease Progression , Female , Humans , Lung Neoplasms/chemically induced , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Nicotine/adverse effects , Nitrosamines/toxicity , Proliferating Cell Nuclear Antigen/biosynthesis , Ribosomal Protein S6 Kinases, 90-kDa/biosynthesis , TOR Serine-Threonine Kinases/biosynthesis , bcl-X Protein/biosynthesis
2.
Oncotarget ; 6(17): 15524-39, 2015 Jun 20.
Article in English | MEDLINE | ID: mdl-25906749

ABSTRACT

Recent development of genetically engineered mouse models (GEMs) for pancreatic cancer (PC) that recapitulates human disease progression has helped to identify new strategies to delay/inhibit PC development. We first found that expression of the pancreatic tumor-initiating/cancer stem cells (CSC) marker DclK1 occurs in early stage PC and in both early and late pancreatic intraepithelial neoplasia (PanIN) and that it increases as disease progresses in GEM and also in human PC. Genome-wide next generation sequencing of pancreatic ductal adenocarcinoma (PDAC) from GEM mice revealed significantly increased DclK1 along with inflammatory genes. Genetic ablation of cyclo-oxygenase-2 (COX-2) decreased DclK1 in GEM. Induction of inflammation/pancreatitis with cerulein in GEM mice increased DclK1, and the novel dual COX/5-lipoxygenase (5-LOX) inhibitor licofelone reduced it. Dietary licofelone significantly inhibited the incidence of PDAC and carcinoma in situ with significant inhibition of pancreatic CSCs. Licofelone suppressed pancreatic tumor COX-2 and 5-LOX activities and modulated miRNAs characteristic of CSC and inflammation in correlation with PDAC inhibition. These results offer a preclinical proof of concept to target the inflammation initiation to inhibit cancer stem cells early for improving the treatment of pancreatic cancers, with immediate clinical implications for repositioning dual COX/5-LOX inhibitors in human trials for high risk patients.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/pathology , Pancreatitis/pathology , Protein Serine-Threonine Kinases/metabolism , Pyrroles/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Arachidonic Acid/metabolism , Carcinoma in Situ/pathology , Carcinoma in Situ/prevention & control , Carcinoma, Pancreatic Ductal/prevention & control , Cell Proliferation/drug effects , Ceruletide , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Disease Models, Animal , Disease Progression , Doublecortin-Like Kinases , Lipoxygenase Inhibitors/pharmacology , Mice , Mice, Knockout , MicroRNAs/antagonists & inhibitors , Pancreatic Neoplasms/prevention & control , Pancreatitis/chemically induced
3.
Cancer Prev Res (Phila) ; 7(12): 1198-209, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25248858

ABSTRACT

Ornithine decarboxylase (ODC) is the key rate-limiting enzyme in the polyamine synthesis pathway and it is overexpressed in a variety of cancers. We found that polyamine synthesis and modulation of ODC signaling occurs at early stages of pancreatic precursor lesions and increases as the tumor progresses in Kras-activated p48(Cre/+)-LSL-Kras(G12D/+) mice. Interest in use of the ODC inhibitor eflornithine (DFMO) as a cancer chemopreventive agent has increased in recent years since ODC was shown to be transactivated by the c-myc oncogene and to cooperate with the ras oncogene in malignant transformation of epithelial tissues. We tested the effects of DFMO on pancreatic intraepithelial neoplasias (PanIN) and their progression to pancreatic ductal adenocarcinoma (PDAC) in genetically engineered Kras mice. The Kras(G12D/+) mice fed DFMO at 0.1% and 0.2% in the diet showed a significant inhibition (P < 0.0001) of PDAC incidence compared with mice fed control diet. Pancreatic tumor weights were decreased by 31% to 43% (P < 0.03-0.001) with both doses of DFMO. DFMO at 0.1% and 0.2% caused a significant suppression (27% and 31%; P < 0.02-0.004) of PanIN 3 lesions (carcinoma in situ). DFMO-treated pancreas exhibited modulated ODC pathway components along with decreased proliferation and increased expression of p21/p27 as compared with pancreatic tissues derived from mice fed control diet. In summary, our preclinical data indicate that DFMO has potential for chemoprevention of pancreatic cancer and should be evaluated in other PDAC models and in combination with other drugs in anticipation of future clinical trials.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Eflornithine/pharmacology , Ornithine Decarboxylase/chemistry , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins p21(ras)/physiology , Signal Transduction/drug effects , Animals , Apoptosis , Blotting, Western , Carcinoma, Pancreatic Ductal/enzymology , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation , Disease Progression , Female , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Mutation/genetics , Ornithine Decarboxylase/metabolism , Ornithine Decarboxylase Inhibitors/pharmacology , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...