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.
Thyroid ; 31(5): 787-799, 2021 05.
Article in English | MEDLINE | ID: mdl-33012268

ABSTRACT

Background: Several mechanisms likely cooperate with the mitogen-activated protein (MAP)-kinase pathway to promote cancer progression in the thyroid. One putative pathway is NOTCH signaling, which is implicated in several other malignancies. In thyroid cancer, data regarding the role of the NOTCH pathway are insufficient and even contradictory. Methods: A BRAFV600E-driven papillary thyroid carcinoma (PTC) mouse model was subjected to NOTCH pathway genetic alterations, and the tumor burden was followed by ultrasound. Further analyses were performed on PTC cell lines or noncancerous cells transfected with NOTCHIC or BRAFV600E, which were then subjected to pharmacological treatment with MAP-kinase or NOTCH pathway inhibitors. Results: The presence of the BRAFV600E mutation coupled with overexpression of the NOTCH intracellular domain led to significantly bigger thyroid tumors in mice, to a more aggressive carcinoma, and decreased overall survival. Although more cystic, the tumors did not progress into anaplastic thyroid carcinomas. On the contrary, the deletion of RBP-jκ (a major cofactor involved in NOTCH signaling) did not alter the phenotype in mice. BRAFV600E-mutated PTC cell lines were resistant to pharmacological inhibition of the NOTCH pathway. Inhibition of MEK1/2 uncovered a predominant effect on Hes1/Hey1 transcription compared with NOTCH inhibition in BRAFV600E-mutated cell lines. Finally, γ-secretase activity and γ-secretase subunit transcription levels were dependent on ERK activation. Our findings suggest that MAP-kinase activity overrides the NOTCH pathway in the context of thyroid cancer. Conclusions: The interaction between the BRAF and NOTCH pathways demonstrates that the BRAFV600E mutation might bypass NOTCH and exert a strong positive effect on NOTCH downstream targets in thyroid carcinoma.


Subject(s)
Proto-Oncogene Proteins B-raf/genetics , Receptor, Notch1/genetics , Thyroid Cancer, Papillary/genetics , Thyroid Carcinoma, Anaplastic/genetics , Thyroid Neoplasms/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , MAP Kinase Signaling System/genetics , Mice , Mutation , Receptor, Notch1/antagonists & inhibitors , Repressor Proteins/metabolism , Thyroid Cancer, Papillary/pathology , Thyroid Carcinoma, Anaplastic/pathology , Thyroid Neoplasms/pathology , Transcription Factor HES-1/metabolism , Tumor Burden
2.
ChemMedChem ; 14(2): 224-236, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30520265

ABSTRACT

By screening a focused library of kinase inhibitor analogues in a phenotypic co-culture assay for angiogenesis inhibition, we identified an aminotriazine that acts as a cytostatic nanomolar inhibitor. However, this aminotriazine was found to be completely inactive in a whole-kinome profiling assay. To decipher its mechanism of action, we used the online target prediction tool PPB2 (http://ppb2.gdb.tools), which suggested lysophosphatidic acid acyltransferaseâ€…ß (LPAAT-ß) as a possible target for this aminotriazine as well as several analogues identified by structure-activity relationship profiling. LPAAT-ß inhibition (IC50 ≈15 nm) was confirmed in a biochemical assay and by its effects on cell proliferation in comparison with a known LPAAT-ß inhibitor. These experiments illustrate the value of target-prediction tools to guide target identification for phenotypic screening hits and significantly expand the rather limited pharmacology of LPAAT-ß inhibitors.


Subject(s)
Acyltransferases/antagonists & inhibitors , Angiogenesis Inducing Agents/metabolism , Enzyme Inhibitors/chemistry , Small Molecule Libraries/chemistry , Triazines/chemistry , Acyltransferases/genetics , Acyltransferases/isolation & purification , Biological Assay/methods , Cell Culture Techniques , Cell Line , Cell Proliferation/drug effects , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Inhibitory Concentration 50 , Models, Molecular , Molecular Structure , Phenotype , Protein Binding , Small Molecule Libraries/metabolism , Software , Structure-Activity Relationship , Triazines/metabolism
3.
J Exp Clin Cancer Res ; 37(1): 234, 2018 Sep 21.
Article in English | MEDLINE | ID: mdl-30241549

ABSTRACT

BACKGROUND: The L-type amino acid transporter 1 (LAT1/SLC7A5) transports essential amino acids across the plasma membrane. While LAT1 is overexpressed in a variety of human neoplasms, its expression and its role in thyroid cancer is currently unknown. Anaplastic thyroid carcinoma (ATC) is a highly aggressive malignancy for which no effective therapy exists. The purpose of this study was to explore whether the inhibition of LAT1 in ATC would affect tumor growth both in vitro and in vivo. METHODS: LAT1 was pharmacologically blocked by JPH203 in human ATC and papillary thyroid cancer (PTC) cell lines. The effects on proliferation and mTORC1 activity were addressed in vitro. A genetically engineered mouse model of ATC was used to address the effect of blocking LAT1 on tumor growth in vivo. SLC7A5 transcription was measured in patient-derived ATC samples to address the clinical relevance of the findings. RESULTS: LAT1 block by JPH203 reduced proliferation and mTORC1 signaling in human thyroid cancer cell lines. SLC7A5 transcription was upregulated in ATC tissues derived from a genetically engineered mouse model and in ATC samples recovered from patients. JPH203 treatment induced thyroid tumor growth arrest in vivo in a fully immunocompetent mouse model of thyroid cancer. Additionally, analysis of publicly available datasets of thyroid carcinomas revealed that high LAT1 expression is associated with potentially untreatable PTC presenting reduced NIS/SLC5A5 transcription and with ATC. CONCLUSIONS: These preclinical results show that LAT1 inhibition is a novel therapeutic approach in the context of thyroid cancers, and more interestingly in untreatable thyroid cancers.


Subject(s)
Cell Proliferation/drug effects , Large Neutral Amino Acid-Transporter 1/genetics , Thyroid Carcinoma, Anaplastic/drug therapy , Animals , Animals, Genetically Modified/genetics , Apoptosis/drug effects , Benzoxazoles/administration & dosage , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Humans , Large Neutral Amino Acid-Transporter 1/drug effects , Mice , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins B-raf/genetics , Signal Transduction/drug effects , Thyroid Carcinoma, Anaplastic/genetics , Thyroid Carcinoma, Anaplastic/pathology , Thyroid Gland/drug effects , Thyroid Gland/pathology , Tyrosine/administration & dosage , Tyrosine/analogs & derivatives
4.
Oncotarget ; 8(61): 103207-103222, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-29262556

ABSTRACT

Thyroid carcinomas are the most prevalent endocrine cancers. The BRAFV600E mutation is found in 40% of the papillary type and 25% of the anaplastic type. BRAFV600E inhibitors have shown great success in melanoma but, they have been, to date, less successful in thyroid cancer. About 50% of anaplastic thyroid carcinomas present mutations/amplification of the phosphatidylinositol 3' kinase. Here we propose to investigate if the hyper activation of that pathway could influence the response to BRAFV600E specific inhibitors. To test this, we used two mouse models of thyroid cancer. Single mutant (BRAFV600E) mice responded to BRAFV600E-specific inhibition (PLX-4720), while double mutant mice (BRAFV600E; PIK3CAH1047R) showed resistance and even signs of aggravation. This resistance was abrogated by combination with a phosphoinositide 3-kinase inhibitor. At the molecular level, we showed that this resistance was concomitant to a paradoxical activation of the MAP-Kinase pathway, which could be overturned by phosphoinositide 3-kinase inhibition in vivo in our mouse model and in vitro in human double mutant cell lines. In conclusion, we reveal a phosphoinositide 3-kinase driven, paradoxical MAP-Kinase pathway activation as mechanism for resistance to BRAFV600E specific inhibitors in a clinically relevant mouse model of thyroid cancer.

5.
Oncotarget ; 8(15): 24604-24620, 2017 Apr 11.
Article in English | MEDLINE | ID: mdl-28445948

ABSTRACT

Anaplastic thyroid cancers and radioiodine resistant thyroid cancer are posing a major treat since surgery combined with Iodine131 therapy is ineffective on them. Small-molecule inhibitors are presenting a new hope for patients, but often lead to drug resistance in many cancers. Based on the major mutations found in thyroid cancer, we propose the combination of a MEK inhibitor and a Pi3'-kinase inhibitor in pre-clinical models. We used human thyroid cancer cell lines and genetically engineered double mutant BRAFV600E PIK3CAH1047R mice to evaluate the effect of both inhibitors separately or in combination in terms of proliferation and signaling in vitro; tumor burden, histology, cell death induction and tumor markers expression in vivo. The combination of MEK and Pi'3-kinase inhibition shows a synergistic effect in term of proliferation and apoptosis induction through Survivin down-regulation in vitro. We show for the first time the effects of the combination of a MEK inhibitor and Pi3'-kinase inhibitor in a genetically engineered mouse model of aggressively lethal thyroid cancer. In fine, the two drugs cooperate to promote tumor shrinkage by inducing a proliferation arrest and an elevation of apoptosis in vivo. Moreover, a phenotypic reversion is also observed with a partial restoration of normal thyroid marker transcription, and thyroid cancer marker expression reduction.In conclusion, combination therapy of MEK and Pi3'-kinase inhibition synergizes to target double mutant thyroid cancer in vitro and in vivo. This multidrug approach could readily be translated into clinical practice and bring new perspectives for the treatment of incurable thyroid carcinoma.


Subject(s)
Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Drug Synergism , Female , Humans , Mice , Thyroid Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...