Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Mol Cell Endocrinol ; 460: 24-35, 2018 01 15.
Article in English | MEDLINE | ID: mdl-28652169

ABSTRACT

Here we describe a conditional doxycycline-dependent mouse model of RET/PTC3 (NCOA4-RET) oncogene-induced thyroid tumorigenesis. In these mice, after 10 days of doxycycline (dox) administration, RET/PTC3 expression induced mitogen activated protein kinase (MAPK) stimulation and a proliferative response which resulted in the formation of hyperplastic thyroid lesions. This was followed, after 2 months, by growth arrest accompanied by typical features of oncogene-induced senescence (OIS), including upregulation of p16INK4A and p21CIP, positivity at the Sudan black B, activation of the DNA damage response (DDR) markers γH2AX and pChk2 T68, and induction of p53 and p19ARF. After 5 months, about half of thyroid lesions escaped OIS and formed tumors that remained dependent on RET/PTC3 expression. This progression was accompanied by activation of AKT-FOXO1/3a pathway and increased serum TSH levels.


Subject(s)
Cellular Senescence , Oncogenes , Thyroid Neoplasms/pathology , Animals , Apoptosis , Cattle , DNA Damage , Disease Models, Animal , Enzyme Activation , Female , Forkhead Transcription Factors/metabolism , Hyperplasia , Male , Mice, Transgenic , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Thyroid Epithelial Cells/pathology , Thyroid Gland/pathology , Thyrotropin/metabolism , Thyroxine/metabolism
2.
Clin Cancer Res ; 21(20): 4686-97, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26124204

ABSTRACT

PURPOSE: Resistance to tyrosine kinase inhibitors (TKI) of EGF receptor (EGFR) is often related to activation of other signaling pathways and evolution through a mesenchymal phenotype. EXPERIMENTAL DESIGN: Because the Hedgehog (Hh) pathway has emerged as an important mediator of epithelial-to-mesenchymal transition (EMT), we studied the activation of Hh signaling in models of EGFR-TKIs intrinsic or acquired resistance from both EGFR-mutated and wild-type (WT) non-small cell lung cancer (NSCLC) cell lines. RESULTS: Activation of the Hh pathway was found in both models of EGFR-mutated and EGFR-WT NSCLC cell line resistant to EGFR-TKIs. In EGFR-mutated HCC827-GR cells, we found SMO (the Hh receptor) gene amplification, MET activation, and the functional interaction of these two signaling pathways. In HCC827-GR cells, inhibition of SMO or downregulation of GLI1 (the most important Hh-induced transcription factor) expression in combination with MET inhibition exerted significant antitumor activity.In EGFR-WT NSCLC cell lines resistant to EGFR inhibitors, the combined inhibition of SMO and EGFR exerted a strong antiproliferative activity with a complete inhibition of PI3K/Akt and MAPK phosphorylation. In addition, the inhibition of SMO by the use of LDE225 sensitizes EGFR-WT NSCLC cells to standard chemotherapy. CONCLUSIONS: This result supports the role of the Hh pathway in mediating resistance to anti-EGFR-TKIs through the induction of EMT and suggests new opportunities to design new treatment strategies in lung cancer.


Subject(s)
Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , Gene Amplification/genetics , Hedgehog Proteins/genetics , Lung Neoplasms/genetics , Receptors, G-Protein-Coupled/genetics , Animals , Biphenyl Compounds/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Down-Regulation/drug effects , Down-Regulation/genetics , Drug Resistance, Neoplasm/drug effects , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Gene Amplification/drug effects , Humans , Lung Neoplasms/drug therapy , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphatidylinositol 3-Kinases/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-met/genetics , Pyridines/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/genetics , Smoothened Receptor
3.
Oncotarget ; 6(27): 23281-96, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-25966280

ABSTRACT

AXL is a tyrosine kinase receptor activated by GAS6 and regulates cancer cell proliferation migration and angiogenesis. We studied AXL as new therapeutic target in colorectal cancer (CRC). Expression and activation of AXL and GAS6 were evaluated in a panel of human CRC cell lines. AXL gene silencing or pharmacologic inhibition with foretinib suppressed proliferation, migration and survival in CRC cells. In an orthotopic colon model of human HCT116 CRC cells overexpressing AXL, foretinib treatment caused significant inhibition of tumour growth and peritoneal metastatic spreading. AXL and GAS6 overexpression by immunohistochemistry (IHC) were found in 76,7% and 73.5%, respectively, of 223 human CRC specimens, correlating with less differentiated histological grading. GAS6 overexpression was associated with nodes involvement and tumour stage. AXL gene was found amplified by Fluorescence in situ hybridization (FISH) in 8/146 cases (5,4%) of CRC samples. Taken together, AXL inhibition could represent a novel therapeutic approach in CRC.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Proto-Oncogene Proteins/chemistry , Receptor Protein-Tyrosine Kinases/chemistry , Adult , Aged , Aged, 80 and over , Anilides/chemistry , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Female , Gene Silencing , HCT116 Cells , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Transplantation , Neovascularization, Pathologic , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins/metabolism , Quinolines/chemistry , RNA Interference , Receptor Protein-Tyrosine Kinases/metabolism , Axl Receptor Tyrosine Kinase
4.
Clin Cancer Res ; 21(18): 4153-64, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26019172

ABSTRACT

PURPOSE: The use of cetuximab in the treatment of metastatic colorectal cancer is limited by development of resistance. EXPERIMENTAL DESIGN: We have investigated in three models of highly epidermal growth factor receptor (EGFR)-dependent colorectal cancer xenografts, the effect of maintenance therapy with different kinase inhibitors alone or in combination with cetuximab, after cytotoxic treatment induction with irinotecan plus cetuximab. RESULTS: SW48, LIM 1215, and GEO colorectal cancer cell lines were engrafted into nude mice and treated for 3 weeks with irinotecan and/or cetuximab. The combined treatment induced a significant reduction of tumor size. A subsequent experiment was performed in all three xenograft models in which after an induction treatment with irinotecan plus cetuximab, mice were randomly assigned to one of the following treatments: control, cetuximab, regorafenib, a selective PIK3CA inhibitor (PIK3CAi), a selective MEK inhibitor (MEKi), and/or the combination of each inhibitor with cetuximab. The cetuximab plus MEKi treatment determined the best antitumor activity with suppression of tumor growth. This effect was prolonged for 13 to 15 weeks after cessation of therapy and was accompanied by prolonged survival. Antitumor activity was accompanied by inhibition of the MAPK and MEK pathways. Moreover, in the cetuximab plus MEKi-treated SW48 xenograft group, KRAS mutations as a mechanism of acquired resistance were detected in 25% of cases compared with 75% KRAS mutations in the MEKi-treated group. CONCLUSIONS: A possible strategy to prevent and/or overcome resistance to anti-EGFR inhibitors in metastatic colorectal cancer is a maintenance therapy with cetuximab plus MEKi after an initial treatment with irinotecan plus cetuximab.


Subject(s)
Camptothecin/analogs & derivatives , Cetuximab/administration & dosage , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Diphenylamine/analogs & derivatives , Sulfonamides/administration & dosage , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/administration & dosage , Cell Line, Tumor , Cell Survival , Class I Phosphatidylinositol 3-Kinases , Colorectal Neoplasms/genetics , Diphenylamine/administration & dosage , Drug Synergism , ErbB Receptors/metabolism , Female , Humans , Irinotecan , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Signaling System , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Xenograft Model Antitumor Assays
5.
Clin Cancer Res ; 21(13): 2975-83, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25838391

ABSTRACT

PURPOSE: In colorectal cancer, the activation of the intracellular RAS-RAF and PIK3CA-AKT pathways has been implicated in the resistance to anti-EGFR mAbs. We have investigated the role of regorafenib, an oral multikinase inhibitor, in combination with cetuximab, an anti-EGFR mAb, to overcome anti-EGFR resistance. EXPERIMENTAL DESIGN: We have tested, in vitro and in vivo, the effects of regorafenib in a panel of human colorectal cancer cell lines with a KRAS mutation (SW480, SW620, HCT116, LOVO, and HCT15) or with a BRAF mutation (HT29), as models of intrinsic resistance to cetuximab treatment, and in two human colorectal cancer cell lines (GEO and SW48) that are cetuximab-sensitive, as well as in their derived cells with acquired resistance to cetuximab (GEO-CR and SW48-CR). RESULTS: Treatment with regorafenib determined a dose-dependent growth inhibition in all colorectal cancer cell lines. The combined treatment with cetuximab and regorafenib induced synergistic antiproliferative and apoptotic effects in cetuximab-resistant cell lines by blocking MAPK and AKT pathways. Nude mice were injected s.c. with HCT116, HCT15, GEO-CR, and SW48-CR cells. The combined treatment caused significant tumor growth inhibition. Synergistic antitumor activity of regorafenib plus cetuximab was also observed in an orthotopic colorectal cancer model of HCT116 cells. In particular, the combined treatment induced a significant tumor growth inhibition in the primary tumor site (cecum) and completely prevented metastasis formation. CONCLUSIONS: The combined treatment with cetuximab and regorafenib could be a strategy to overcome resistance to anti-EGFR therapies in metastatic colorectal cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Cetuximab/administration & dosage , Drug Synergism , ErbB Receptors/antagonists & inhibitors , Female , HCT116 Cells , Humans , Mice, Inbred BALB C , Mice, Nude , Phenylurea Compounds/administration & dosage , Pyridines/administration & dosage , Xenograft Model Antitumor Assays
6.
Clin Cancer Res ; 20(14): 3775-86, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24812410

ABSTRACT

PURPOSE: The EGFR-independent activation of the RAS/RAF/MEK/MAPK pathway is one of the resistance mechanisms to cetuximab. EXPERIMENTAL DESIGN: We have evaluated, in vitro and in vivo, the effects of BAY 86-9766, a selective MEK1/2 inhibitor, in a panel of human colorectal cancer cell lines with primary or acquired resistance to cetuximab. RESULTS: Among the colorectal cancer cell lines, five with a KRAS mutation (LOVO, HCT116, HCT15, SW620, and SW480) and one with a BRAF mutation (HT29) were resistant to the antiproliferative effects of cetuximab, whereas two cells (GEO and SW48) were highly sensitive. Treatment with BAY 86-9766 determined dose-dependent growth inhibition in all cancer cells, including two human colorectal cancer cells with acquired resistance to cetuximab (GEO-CR and SW48-CR), with the exception of HCT15 cells. Combined treatment with cetuximab and BAY 86-9766 induced a synergistic antiproliferative and apoptotic effects with blockade in the MAPK and AKT pathway in cells with either primary or acquired resistance to cetuximab. The synergistic antiproliferative effects were confirmed using other two selective MEK1/2 inhibitors, selumetinib and pimasertib, in combination with cetuximab. Moreover, inhibition of MEK expression by siRNA restored cetuximab sensitivity in resistant cells. In nude mice bearing established human HCT15, HCT116, SW48-CR, and GEO-CR xenografts, the combined treatment with cetuximab and BAY 86-9766 caused significant tumor growth inhibition and increased mice survival. CONCLUSION: These results suggest that activation of MEK is involved in both primary and acquired resistance to cetuximab and the inhibition of EGFR and MEK could be a strategy for overcoming anti-EGFR resistance in patients with colorectal cancer.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Diphenylamine/analogs & derivatives , ErbB Receptors/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cetuximab , Diphenylamine/pharmacology , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/immunology , Female , Gene Knockdown Techniques , Humans , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Mice, Inbred BALB C , Mice, Nude , RNA Interference , Xenograft Model Antitumor Assays
7.
Clin Cancer Res ; 19(24): 6751-65, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24122793

ABSTRACT

PURPOSE: Although cetuximab, an anti-EGF receptor (EGFR) monoclonal antibody, is an effective treatment for patients with KRAS wild-type metastatic colorectal cancer (mCRC), its clinical use is limited by onset of resistance. EXPERIMENTAL DESIGN: We characterized two colorectal cancer models to study the mechanisms of acquired resistance to cetuximab. RESULTS: Following chronic treatment of nude mice bearing cetuximab-sensitive human GEO colon xenografts, cetuximab-resistant GEO (GEO-CR) cells were obtained. In GEO-CR cells, proliferation and survival signals were constitutively active despite EGFR inhibition by cetuximab treatment. Whole gene expression profiling identified a series of genes involved in the hepatocyte growth factor (HGF)-MET-dependent pathways, which were upregulated in GEO-CR cells. Furthermore, activated, phosphorylated MET was detected in GEO-CR cells. A second colorectal cancer cell line with acquired resistance to cetuximab was obtained (SW48-CR). Inhibition of MET expression by siRNA restored cetuximab sensitivity in GEO-CR and SW48-CR cells, whereas exogenous activation of MET by HGF stimulation in cetuximab-sensitive GEO and SW48 cells induced resistance to cetuximab. Treatment of GEO-CR and SW48-CR cells with PHA665752, a selective MET inhibitor, inhibited cell growth, proliferation, and survival signals and impaired cancer cell migration. Overexpression of TGF-α, a specific EGFR ligand, was involved in the acquisition of cetuximab resistance in GEO-CR and SW48-CR cells. In fact, TGF-α overexpression induced the EGFR-MET interaction, with subsequent MET phosphorylation and activation of MET downstream effectors in GEO-CR and SW48-CR cells. CONCLUSIONS: These results suggest that overexpression of TGF-α through induction of EGFR-MET interaction contributes to cetuximab resistance in colorectal cancer cells. The combined inhibition of EGFR and MET receptor could represent a strategy for preventing and/or overcoming cetuximab resistance in patients with colorectal cancer.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Colonic Neoplasms/drug therapy , ErbB Receptors/genetics , Proto-Oncogene Proteins c-met/genetics , Transforming Growth Factor alpha/biosynthesis , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cetuximab , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Hepatocyte Growth Factor/biosynthesis , Humans , Mice , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction/drug effects
8.
Int J Cancer ; 133(9): 2089-101, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23629727

ABSTRACT

The RAS/RAF/MEK/MAPK and the PTEN/PI3K/AKT/mTOR pathways are key regulators of proliferation and survival in human cancer cells. Selective inhibitors of different transducer molecules in these pathways have been developed as molecular targeted anti-cancer therapies. The in vitro and in vivo anti-tumor activity of pimasertib, a selective MEK 1/2 inhibitor, alone or in combination with a PI3K inhibitor (PI3Ki), a mTOR inhibitor (everolimus), or with multi-targeted kinase inhibitors (sorafenib and regorafenib), that block also BRAF and CRAF, were tested in a panel of eight human lung and colon cancer cell lines. Following pimasertib treatment, cancer cell lines were classified as pimasertib-sensitive (IC50 for cell growth inhibition of 0.001 µM) or pimasertib-resistant. Evaluation of basal gene expression profiles by microarrays identified several genes that were up-regulated in pimasertib-resistant cancer cells and that were involved in both RAS/RAF/MEK/MAPK and PTEN/PI3K/AKT/mTOR pathways. Therefore, a series of combination experiments with pimasertib and either PI3Ki, everolimus, sorafenib or regorafenib were conducted, demonstrating a synergistic effect in cell growth inhibition and induction of apoptosis with sustained blockade in MAPK- and AKT-dependent signaling pathways in pimasertib-resistant human colon carcinoma (HCT15) and lung adenocarcinoma (H1975) cells. Finally, in nude mice bearing established HCT15 and H1975 subcutaneous tumor xenografts, the combined treatment with pimasertib and BEZ235 (a dual PI3K/mTOR inhibitor) or with sorafenib caused significant tumor growth delays and increase in mice survival as compared to single agent treatment. These results suggest that dual blockade of MAPK and PI3K pathways could overcome intrinsic resistance to MEK inhibition.


Subject(s)
Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Lung Neoplasms/drug therapy , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Cycle/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Gene Expression Profiling , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , PTEN Phosphohydrolase/metabolism , Phenylurea Compounds/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyridines/pharmacology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Clin Cancer Res ; 19(13): 3508-19, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23695170

ABSTRACT

PURPOSE: EGF receptor (EGFR) tyrosine kinase inhibitors (TKI) have been found to be effective against lung cancer, but clinical resistance to these agents has developed as their usage has increased. Metformin is a widely used antidiabetic drug and also displays significant growth-inhibitory and proapoptotic effects in several cancer models, alone or in combination with chemotherapeutic drugs. EXPERIMENTAL DESIGN: The effects of gefitinib, a selective EGFR-TKI, and metformin on a panel of non-small cell lung cancer (NSCLC) cell lines were assessed by using MTT, bromide assay, flow cytometry, anchorage-independent growth, coimmunoprecipitation, and Western blot analysis. RESULTS: The combination of metformin with gefitinib induced a strong antiproliferative and proapoptotic effect in NSCLC cell lines that harbored wild-type LKB1 gene. Treatment with metformin as single agent, however, induced an activation and phosphorylation of mitogen-activated protein kinase (MAPK) through an increased C-RAF/B-RAF heterodimerization. The inhibition of EGFR phosphorylation and of downstream signaling by adding gefitinib to metformin treatment abrogated this phenomenon and induced a strong apoptotic effect in vitro and in vivo. CONCLUSIONS: Metformin and gefitinib are synergistic in LKB1 wild-type NSCLC cells. However, further studies are required to investigate better the effect of metformin action on the RAS/RAF/MAPK pathway and the best context in which to use metformin in combination with molecular targeted agents.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/metabolism , Metformin/pharmacology , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , AMP-Activated Protein Kinase Kinases , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , ErbB Receptors/metabolism , Female , Gefitinib , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Metformin/administration & dosage , Mice , Protein Kinase Inhibitors/administration & dosage , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Tumor Burden/drug effects , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
10.
Endocr Relat Cancer ; 19(3): 409-21, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22454401

ABSTRACT

For an identification of novel candidate genes in thyroid tumourigenesis, we have investigated gene copy number changes in a Trk-T1 transgenic mouse model of thyroid neoplasia. For this aim, 30 thyroid tumours from Trk-T1 transgenics were investigated by comparative genomic hybridisation. Recurrent gene copy number alterations were identified and genes located in the altered chromosomal regions were analysed by Gene Ontology term enrichment analysis in order to reveal gene functions potentially associated with thyroid tumourigenesis. In thyroid neoplasms from Trk-T1 mice, a recurrent gain on chromosomal bands 1C4-E2.3 (10.0% of cases), and losses on 3H1-H3 (13.3%), 4D2.3-E2 (43.3%) and 14E4-E5 (6.7%) were identified. The genes Twist2, Ptma, Pde6d, Bmpr1b, Pdlim5, Unc5c, Srm, Trp73, Ythdf2, Taf12 and Slitrk5 are located in these chromosomal bands. Copy number changes of these genes were studied by fluorescence in situ hybridisation on 30 human papillary thyroid carcinoma (PTC) samples and altered gene expression was studied by qRT-PCR analyses in 67 human PTC. Copy number gains were detected in 83% of cases for TWIST2 and in 100% of cases for PTMA and PDE6D. DNA losses of SLITRK1 and SLITRK5 were observed in 21% of cases and of SLITRK6 in 16% of cases. Gene expression was significantly up-regulated for UNC5C and TP73 and significantly down-regulated for SLITRK5 in tumours compared with normal tissue. In conclusion, a global genomic copy number analysis of thyroid tumours from Trk-T1 transgenic mice revealed a number of novel gene alterations in thyroid tumourigenesis that are also prevalent in human PTCs.


Subject(s)
Gene Expression Regulation, Neoplastic , Thyroid Neoplasms/genetics , Adolescent , Adult , Animals , Child , Comparative Genomic Hybridization , Female , Gene Dosage , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Receptor, trkA/genetics , Young Adult
11.
Endocr Relat Cancer ; 18(1): 1-11, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20943719

ABSTRACT

Oncogenic conversion of the RET tyrosine kinase is a frequent feature of medullary thyroid carcinoma (MTC). ZD6474 (vandetanib) is an ATP-competitive inhibitor of RET, epidermal growth factor receptor (EGFR), and vascular endothelial growth factor receptors kinases. In this study, we have studied ZD6474 mechanism of action in TT and MZ-CRC-1 human MTC cell lines, carrying cysteine 634 to tryptophan (C634W) and methionine 918 to threonine (M918T) RET mutation respectively. ZD6474 blunted MTC cell proliferation and RET, Shc and p44/p42 mitogen-activated protein kinase (MAPK) phosphorylation. Single receptor knockdown by RNA interference showed that MTC cells depended on RET for proliferation. Adoptive expression of the ZD6474-resistant V804M RET mutant rescued proliferation of TT cells under ZD6474 treatment, showing that RET is a key ZD6474 target in these MTC cells. Upon RET inhibition, adoptive stimulation of EGFR partially rescued TT cell proliferation, MAPK signaling, and expression of cell-cycle-related genes. This suggests that simultaneous inhibition of RET and EGFR by ZD6474 may overcome the risk of MTC cells to escape from RET blockade through compensatory over-activation of EGFR.


Subject(s)
Carcinoma, Medullary/drug therapy , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Quinazolines/pharmacology , Thyroid Neoplasms/drug therapy , Carcinoma, Medullary/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/physiology , Humans , Mutation , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/physiology , Signal Transduction/drug effects , Thyroid Neoplasms/pathology , Transforming Growth Factor alpha/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
12.
Clin Cancer Res ; 16(20): 4990-5001, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20810384

ABSTRACT

PURPOSE: Cancer cell survival, invasion, and metastasis depend on cancer cell proliferation and on tumor-induced angiogenesis. We evaluated the efficacy of the combination of sorafenib and erlotinib or cetuximab. EXPERIMENTAL DESIGN: Sorafenib, erlotinib, and cetuximab, alone or in combination, were tested in vitro in a panel of non-small cell lung cancer (NSCLC) and colorectal cancer cell lines and in vivo in H1299 tumor xenografts. RESULTS: Epidermal growth factor receptor (EGFR) ligand mRNAs were expressed in all NSCLC and colorectal cancer cell lines with variable levels ranging from 0.4- to 8.1-fold as compared with GEO colorectal cancer cells. Lung cancer cells had the highest levels of vascular endothelial growth factors (VEGF) A, B, and C, and of VEGF receptors as compared with colorectal cancer cells. Combined treatments of sorafenib with erlotinib or cetuximab produced combination index values between 0.02 and 0.5, suggesting a significant synergistic activity to inhibit soft agar colony formation in all cancer cell lines, which was accompanied by a marked blockade in mitogen-activated protein kinase and AKT signals. The in vitro migration of H1299 cells, which expressed high levels of both VEGF ligands and receptors, was inhibited by treatment with sorafenib, and this effect was significantly increased by the combination with anti-EGFR drugs. In nude mice bearing established human H1299 xenografts, treatment with the combination of sorafenib and erlotinib or cetuximab caused a significant tumor growth delay resulting in 70 to 90 days increase in mice median overall survival as compared with single-agent sorafenib treatment. CONCLUSIONS: Combination treatment with sorafenib and erlotinib or cetuximab has synergistic antitumor effects in human colorectal and lung cancer cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzenesulfonates/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Colorectal Neoplasms/drug therapy , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Benzenesulfonates/administration & dosage , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cetuximab , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Drug Synergism , ErbB Receptors/biosynthesis , Erlotinib Hydrochloride , Female , HCT116 Cells , HT29 Cells , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice , Mice, Nude , Niacinamide/analogs & derivatives , Phenylurea Compounds , Protein Kinase Inhibitors/administration & dosage , Pyridines/administration & dosage , Quinazolines/administration & dosage , Quinazolines/pharmacology , Receptors, Vascular Endothelial Growth Factor/biosynthesis , Signal Transduction/drug effects , Sorafenib , Xenograft Model Antitumor Assays
13.
Cancer Res ; 67(14): 6956-64, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17638907

ABSTRACT

The RET kinase has emerged as a promising target for the therapy of medullary thyroid cancers (MTC) and of a subset of papillary thyroid cancers. NVP-AST487, a N,N'-diphenyl urea with an IC(50) of 0.88 mumol/L on RET kinase, inhibited RET autophosphorylation and activation of downstream effectors, and potently inhibited the growth of human thyroid cancer cell lines with activating mutations of RET but not of lines without RET mutations. NVP-AST487 induced a dose-dependent growth inhibition of xenografts of NIH3T3 cells expressing oncogenic RET, and of the MTC cell line TT in nude mice. MTCs secrete calcitonin, a useful indicator of tumor burden. Human plasma calcitonin levels derived from the TT cell xenografts were inhibited shortly after treatment, when tumor volume was still unchanged, indicating that the effects of RET kinase inhibition on calcitonin secretion were temporally dissociated from its tumor-inhibitory properties. Accordingly, NVP-AST487 inhibited calcitonin gene expression in vitro in TT cells, in part, through decreased gene transcription. These data point to a previously unknown physiologic role of RET signaling on calcitonin gene expression. Indeed, the RET ligands persephin and GDNF robustly stimulated calcitonin mRNA, which was blocked by pretreatment with NVP-AST487. Antagonists of RET kinase activity in patients with MTC may result in effects on plasma calcitonin that are either disproportionate or dissociated from the effects on tumor burden, because RET kinase mediates a physiologic pathway controlling calcitonin secretion. The role of traditional tumor biomarkers may need to be reassessed as targeted therapies designed against oncoproteins with key roles in pathogenesis are implemented.


Subject(s)
Antineoplastic Agents/pharmacology , Calcitonin/antagonists & inhibitors , Carbanilides/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/enzymology , Animals , Calcitonin/metabolism , Cell Line, Tumor , Female , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Humans , Inhibitory Concentration 50 , Mice , Neoplasm Transplantation , Phosphorylation
14.
Cancer Res ; 64(11): 3823-9, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15172989

ABSTRACT

We show that treatment of a panel of thyroid carcinoma cell lines naturally harboring the RET/PTC1 oncogene, with the RET kinase inhibitors PP1 and ZD6474, results in reversible G(1) arrest. This is accompanied by interruption of Shc and mitogen-activated protein kinase (MAPK) phosphorylation, reduced levels of G(1) cyclins, and increased levels of the cyclin-dependent kinase inhibitor p27Kip1 because of a reduced protein turnover. MAP/extracellular signal-regulated kinase 1/2 inhibition by U0126 caused G(1) cyclins down-regulation and p27Kip1 up-regulation as well. Forced expression of RET/PTC in normal thyroid follicular cells caused a MAPK- and proteasome-dependent down-regulation of p27Kip1. Reduction of p27Kip1 protein levels by antisense oligonucleotides abrogated the G(1) arrest induced by RET/PTC blockade. Therefore, in thyroid cancer, RET/PTC-mediated MAPK activation contributes to p27Kip1 deregulation. This pathway is implicated in cell cycle progression and in response to small molecule kinase inhibitors.


Subject(s)
Cell Cycle Proteins/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Tumor Suppressor Proteins/metabolism , Cell Line, Tumor , Cyclin D , Cyclin E/metabolism , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/metabolism , Cysteine Endopeptidases/metabolism , Enzyme Inhibitors/pharmacology , Gene Rearrangement , Humans , MAP Kinase Signaling System , Multienzyme Complexes/metabolism , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/biosynthesis , Piperidines/pharmacology , Proteasome Endopeptidase Complex , Protein-Tyrosine Kinases , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ret , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Quinazolines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Thyroid Neoplasms/enzymology , Thyroid Neoplasms/genetics , Up-Regulation
15.
Oncogene ; 23(1): 109-21, 2004 Jan 08.
Article in English | MEDLINE | ID: mdl-14712216

ABSTRACT

Human thyroid papillary carcinomas are characterized by rearrangements of the RET protooncogene with a number of heterologous genes, which generate the RET/papillary thyroid carcinoma (PTC) oncogenes. One of the most frequent variants of these recombination events is the fusion of the intracellular kinase-encoding domain of RET to the first 101 amino acids of a gene named H4(D10S170). We have characterized the H4(D10S170) gene product, showing that it is a ubiquitously expressed 55 KDa nuclear and cytosolic protein that is phosphorylated following serum stimulation. This phosphorylation was found to depend on mitogen-activated protein kinase (MAPK) Erk1/2 activity and to be associated to the relocation of H4(D10S170) from the nucleus to the cytosol. Overexpression of the H4(D10S170) gene was able to induce apoptosis of thyroid follicular epithelial cells; conversely a carboxy-terminal truncated H4(D10S170) mutant H4(1-101), corresponding to the portion included in the RET/PTC1 oncoprotein, behaved as dominant negative on the proapoptotic function and nuclear localization of H4(D10S170). Furthermore, conditional expression of the H4(D10S170)-dominant negative truncated mutant protected cells from stress-induced apoptosis. The substitution of serine 244 with alanine abrogated the apoptotic function of H4(D10S170). These data suggest that loss of the H4(D10S170) gene function might have a role in thyroid carcinogenesis by impairing apoptosis.


Subject(s)
Apoptosis , Carcinoma, Papillary/genetics , Gene Rearrangement , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Thyroid Neoplasms/genetics , Animals , Carcinoma, Papillary/pathology , Humans , Mice , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/physiology , Phosphorylation , Protein-Tyrosine Kinases , Proto-Oncogene Proteins c-ret , Rabbits , Thyroid Neoplasms/pathology
16.
J Clin Endocrinol Metab ; 88(4): 1897-902, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12679489

ABSTRACT

Inappropriate activation of the RET receptor tyrosine kinase causes development of papillary and medullary thyroid cancer. We have previously shown that pyrazolopyrimidine is a potent inhibitor of the RET kinase. Here, we show that 4-amino-5-(4-chloro-phenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine) (PP2), another pyrazolopyrimidine, blocks the enzymatic activity of the isolated RET kinase and RET/PTC1 oncoprotein at IC(50) in the nanomolar range. PP2 blocked in vivo phosphorylation and signaling of the RET/PTC1 oncoprotein. PP2 prevented serum-independent growth of RET/PTC1-transformed NIH3T3 fibroblasts and of TPC1 and FB2, two human papillary thyroid carcinoma cell lines that carry spontaneous RET/PTC1 rearrangements. Finally, PP2 blocked invasion of type I collagen matrix by TPC1 cells. Thus, pyrazolopirimidines hold promise for the treatment of human cancers sustaining oncogenic activation of RET.


Subject(s)
Drosophila Proteins , Enzyme Inhibitors/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Pyrimidines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Thyroid Neoplasms/enzymology , 3T3 Cells , Animals , Carcinoma, Papillary/enzymology , Cell Line, Transformed , Enzyme Activation , Gene Rearrangement , Humans , Mice , Mitogen-Activated Protein Kinases/metabolism , Mutation , Neoplasm Invasiveness , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ret , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Thyroid Neoplasms/pathology , Transfection , Tumor Cells, Cultured
17.
Cancer Res ; 62(24): 7284-90, 2002 Dec 15.
Article in English | MEDLINE | ID: mdl-12499271

ABSTRACT

RET/papillary thyroid carcinoma (PTC) oncogenes, generated by recombination of the tyrosine kinase-encoding domain of RET with different heterologous genes, are prevalent in papillary carcinomas of the thyroid. Point mutations of RET cause multiple endocrine neoplasia type 2 (MEN2) familial cancer syndrome and are found in sporadic medullary thyroid carcinomas. Here, we show that ZD6474, a low molecular weight tyrosine kinase inhibitor, blocks the enzymatic activity of RET-derived oncoproteins at a one-half maximal inhibitory concentration of 100 nM. ZD6474 blocked in vivo phosphorylation and signaling of the RET/PTC3 and RET/MEN2B oncoproteins and of an epidermal growth factor (EGF)-activated EGF-receptor/RET chimeric receptor. RET/PTC3-transformed cells-treated ZD6474 lost proliferative autonomy and showed morphological reversion. ZD6474 prevented the growth of two human PTC cell lines that carry spontaneous RET/PTC1 rearrangements. Finally, it blocked anchorage-independent growth of RET/PTC3-transformed NIH3T3 fibroblasts and the formation of tumors after injection of NIH-RET/PTC3 cells into nude mice. Thus, targeting RET oncogenes with ZD6474 might offer a potential treatment strategy for carcinomas sustaining oncogenic activation of RET.


Subject(s)
Drosophila Proteins , Enzyme Inhibitors/pharmacology , Piperidines/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Quinazolines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , 3T3 Cells , Animals , Carcinoma, Papillary/genetics , Carcinoma, Papillary/pathology , Carcinoma, Papillary/prevention & control , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-ret , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/physiology , Signal Transduction/drug effects , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Thyroid Neoplasms/prevention & control , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
18.
J Clin Endocrinol Metab ; 87(6): 2525-31, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12050209

ABSTRACT

Being one of the most lethal human neoplasms and refractory to such conventional treatment as chemo- and radiotherapy, anaplastic thyroid carcinoma is a prime target for innovative therapy. p53 gene inactivation is a constant feature of this neoplasia. Therefore, we evaluated a therapeutic approach based on an E1B 55-kDa gene-defective adenovirus (ONYX-015) that replicates only in cells with impaired p53 function and leads to cell death. Here we report that the ONYX-015 virus induces cell death in three human thyroid anaplastic carcinoma cell lines (ARO, FRO, and KAT-4). In addition, we found that the growth of xenograft tumors induced in athymic mice by the injection of ARO cells was drastically reduced by ONYX-015 treatment. The ONYX-015 virus worked synergistically with two antineoplastic drugs (doxorubicin and paclitaxel) in inducing ARO and KAT-4 cell death. These results suggest that ONYX-015 may be a valid tool in the treatment of the human thyroid anaplastic carcinoma.


Subject(s)
Adenovirus E1B Proteins/genetics , Carcinoma/physiopathology , Carcinoma/virology , Defective Viruses/physiology , Thyroid Neoplasms/physiopathology , Thyroid Neoplasms/virology , Adenocarcinoma, Follicular/pathology , Adenocarcinoma, Follicular/physiopathology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Carcinogenicity Tests , Carcinoma/pathology , Carcinoma/prevention & control , Carcinoma, Papillary/pathology , Carcinoma, Papillary/physiopathology , Cell Death/drug effects , Cell Death/physiology , Defective Viruses/genetics , Doxorubicin/pharmacology , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Paclitaxel/pharmacology , Rats , Thyroid Neoplasms/pathology , Transplantation, Heterologous , Tumor Cells, Cultured/drug effects
19.
Cancer Res ; 62(4): 1077-82, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11861385

ABSTRACT

Oncogenic activation of the RET receptor tyrosine kinase is common in different human cancers. We found that the pyrazolo-pyrimidine PP1 inhibited RET-derived oncoproteins with a half maximal inhibitor concentration of 80 nM. Furthermore, RET/PTC3-transformed cells treated with 5 microM of PP1 lost proliferative autonomy and showed morphological reversion. PP1 prevented the growth of two human papillary thyroid carcinoma cell lines that carry spontaneous RET/PTC1 rearrangements and blocked anchorage-independent growth and tumorigenicity in nude mice of NIH3T3 fibroblasts expressing the RET/PTC3 oncogene. These findings suggest targeting RET oncogenes with PP1 or related compounds as a novel treatment strategy for RET-associated neoplasms.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Drosophila Proteins , Enzyme Inhibitors/pharmacology , Gonadotropin-Releasing Hormone/analogs & derivatives , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , 3T3 Cells/cytology , 3T3 Cells/drug effects , Animals , Cell Transformation, Neoplastic/metabolism , Gene Expression , Humans , Male , Mice , Mice, Inbred BALB C , Oncogenes , Phosphorylation/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ret , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/enzymology , Thyroid Neoplasms/genetics , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...