Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Cell Rep ; 36(4): 109455, 2021 07 27.
Article in English | MEDLINE | ID: mdl-34320350

ABSTRACT

In glioblastoma (GBM), the most frequent and lethal brain tumor, therapies suppressing recurrently altered signaling pathways failed to extend survival. However, in patient subsets, specific genetic lesions can confer sensitivity to targeted agents. By exploiting an integrated model based on patient-derived stem-like cells, faithfully recapitulating the original GBMs in vitro and in vivo, here, we identify a human GBM subset (∼9% of all GBMs) characterized by ERBB3 overexpression and nuclear accumulation. ERBB3 overexpression is driven by inheritable promoter methylation or post-transcriptional silencing of the oncosuppressor miR-205 and sustains the malignant phenotype. Overexpressed ERBB3 behaves as a specific signaling platform for fibroblast growth factor receptor (FGFR), driving PI3K/AKT/mTOR pathway hyperactivation, and overall metabolic upregulation. As a result, ERBB3 inhibition by specific antibodies is lethal for GBM stem-like cells and xenotransplants. These findings highlight a subset of patients eligible for ERBB3-targeted therapy.


Subject(s)
Glioblastoma/genetics , MicroRNAs/metabolism , Receptor, ErbB-3/metabolism , Antibodies/metabolism , Apoptosis , Cell Line, Tumor , Fibroblast Growth Factor 2 , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , MicroRNAs/genetics , Oligodendroglia/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-3/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction , Spheroids, Cellular/pathology , TOR Serine-Threonine Kinases/metabolism
2.
Clin Cancer Res ; 24(4): 807-820, 2018 02 15.
Article in English | MEDLINE | ID: mdl-28974546

ABSTRACT

Purpose: Patient-derived xenografts ("xenopatients") of colorectal cancer metastases have been essential to identify genetic determinants of resistance to the anti-EGFR antibody cetuximab and to explore new therapeutic strategies. From xenopatients, a genetically annotated collection of stem-like cultures ("xenospheres") was generated and characterized for response to targeted therapies.Experimental Design: Xenospheres underwent exome-sequencing analysis, gene expression profile, and in vitro targeted treatments to assess genetic, biological, and pharmacologic correspondence with xenopatients, and to investigate nongenetic biomarkers of therapeutic resistance. The outcome of EGFR family inhibition was tested in an NRG1-expressing in vivo model.Results: Xenospheres faithfully retained the genetic make-up of their matched xenopatients over in vitro and in vivo passages. Frequent and rare genetic lesions triggering primary resistance to cetuximab through constitutive activation of the RAS signaling pathway were conserved, as well as the vulnerability to their respective targeted treatments. Xenospheres lacking such alterations (RASwt) were highly sensitive to cetuximab, but were protected by ligands activating the EGFR family, mostly NRG1. Upon reconstitution of NRG1 expression, xenospheres displayed increased tumorigenic potential in vivo and generated tumors completely resistant to cetuximab, and sensitive only to comprehensive EGFR family inhibition.Conclusions: Xenospheres are a reliable model to identify both genetic and nongenetic mechanisms of response and resistance to targeted therapies in colorectal cancer. In the absence of RAS pathway mutations, NRG1 and other EGFR ligands can play a major role in conferring primary cetuximab resistance, indicating that comprehensive inhibition of the EGFR family is required to achieve a significant therapeutic response. Clin Cancer Res; 24(4); 807-20. ©2017 AACRSee related commentary by Napolitano and Ciardiello, p. 727.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colonic Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , Neoplastic Stem Cells/drug effects , Xenograft Model Antitumor Assays/methods , Animals , Cetuximab/administration & dosage , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Profiling/methods , Humans , Mice, Inbred NOD , Mice, SCID , Molecular Targeted Therapy/methods , Neoplastic Stem Cells/metabolism , Tumor Burden/drug effects , Tumor Burden/genetics , Tumor Cells, Cultured , Exome Sequencing/methods
3.
Mol Oncol ; 9(2): 377-88, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25306394

ABSTRACT

The inflammatory cytokine Tumor Necrosis Factor Alpha (TNF-α) is known to trigger invasive growth, a physiological property for tissue healing, turning into a hallmark of progression in cancer. However, the invasive response to TNF-α relies on poorly understood molecular mechanisms. We thus investigated whether it involves the MET oncogene, which regulates the invasive growth program by encoding the tyrosine kinase receptor for Hepatocyte Growth Factor (HGF). Here we show that the TNF-α pro-invasive activity requires MET function, as it is fully inhibited by MET-specific inhibitors (small-molecules, antibodies, and siRNAs). Mechanistically, we show that TNF-α induces MET transcription via NF-κB, and exploits MET to sustain MEK/ERK activation and Snail accumulation, leading to E-cadherin downregulation. We then show that TNF-α not only induces MET expression in cancer cells, but also HGF secretion by fibroblasts. Consistently, we found that, in human colorectal cancer tissues, high levels of TNF-α correlates with increased expression of both MET and HGF. These findings suggest that TNF-α fosters a HGF/MET pro-invasive paracrine loop in tumors. Targeting this ligand/receptor pair would contribute to prevent cancer progression associated with inflammation.


Subject(s)
Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , MAP Kinase Signaling System , Proto-Oncogene Proteins c-met/biosynthesis , Tumor Necrosis Factor-alpha/metabolism , Cell Line, Tumor , Colorectal Neoplasms/genetics , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Humans , Neoplasm Invasiveness/genetics , Paracrine Communication/genetics , Proto-Oncogene Proteins c-met/genetics , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/genetics
4.
Cancer Res ; 74(6): 1857-69, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24448239

ABSTRACT

Metastatic colorectal cancer remains largely incurable, although in a subset of patients, survival is prolonged by new targeting agents such as anti-EGF receptor (anti-EGFR) antibodies. This disease is believed to be supported by a subpopulation of stem-like cells termed colon cancer-initiating cell (CCIC), which may also confer therapeutic resistance. However, how CCICs respond to EGFR inhibition has not been fully characterized. To explore this question, we systematically generated CCICs through spheroid cultures of patient-derived xenografts of metastatic colorectal cancer. These cultures, termed "xenospheres," were capable of long-term self-propagation in vitro and phenocopied the original patient tumors in vivo, thus operationally defining CCICs. Xenosphere CCICs retained the genetic determinants for EGFR therapeutic response in vitro and in xenografts; like the original tumors, xenospheres harboring a mutated KRAS gene were resistant to EGFR therapy, whereas those harboring wild-type RAS pathway genes (RAS(wt)) were sensitive. Notably, the effects of EGFR inhibition in sensitive CCICs could be counteracted by cytokines secreted by cancer-associated fibroblasts. In particular, we found that the MET receptor ligand hepatocyte growth factor (HGF) was especially active in supporting in vitro CCIC proliferation and resistance to EGFR inhibition. Ectopic production of human HGF in CCIC xenografts rendered the xenografts susceptible to MET inhibition, which sensitized the response to EGFR therapy. By showing that RAS(wt) CCICs rely on both EGFR and MET signaling, our results offer a strong preclinical proof-of-concept for concurrent targeting of these two pathways in the clinical setting.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Colonic Neoplasms/enzymology , ErbB Receptors/antagonists & inhibitors , Neoplastic Stem Cells/enzymology , Proto-Oncogene Proteins c-met/metabolism , Animals , Cell Proliferation , Cell Survival , Cetuximab , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Drug Resistance, Neoplasm , Drug Synergism , ErbB Receptors/metabolism , Female , Hepatocyte Growth Factor/physiology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Neoplastic Stem Cells/drug effects , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Signal Transduction , Spheroids, Cellular/enzymology , Tumor Burden/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL