Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Cancer Res ; 21(1): 49-59, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25320355

ABSTRACT

PURPOSE: Mammalian target of rapamycin (mTOR) inhibition activates compensatory insulin-like growth factor receptor (IGFR) signaling. We evaluated the ridaforolimus (mTOR inhibitor) and dalotuzumab (anti-IGF1R antibody) combination. EXPERIMENTAL DESIGN: In vitro and in vivo models, and a phase I study in which patients with advanced cancer received ridaforolimus (10-40 mg/day every day × 5/week) and dalotuzumab (10 mg/kg/week or 7.5 mg/kg/every other week) were explored. RESULTS: Preclinical studies demonstrated enhanced pathway inhibition with ridaforolimus and dalotuzumab. With 87 patients treated in the phase I study, main dose-limiting toxicities (DLT) of the combination were primarily mTOR-related stomatitis and asthenia at doses of ridaforolimus lower than expected, suggesting blockade of compensatory pathways in normal tissues. Six confirmed partial responses were reported (3 patients with breast cancer); 10 of 23 patients with breast cancer and 6 of 11 patients with ER(+)/high-proliferative breast cancer showed antitumor activity. CONCLUSIONS: Our study provides proof-of-concept that inhibiting the IGF1R compensatory response to mTOR inhibition is feasible with promising clinical activity in heavily pretreated advanced cancer, particularly in ER(+)/high-proliferative breast cancer (ClinicalTrials.gov identifier: NCT00730379).


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/drug therapy , Sirolimus/analogs & derivatives , Adult , Aged , Animals , Antibodies, Monoclonal, Humanized , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Humans , Middle Aged , Receptor, IGF Type 1 , Receptors, Somatomedin/antagonists & inhibitors , Receptors, Somatomedin/immunology , Receptors, Somatomedin/metabolism , Signal Transduction/drug effects , Sirolimus/administration & dosage , TOR Serine-Threonine Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays
2.
Cancer Cell ; 19(2): 192-205, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21295524

ABSTRACT

Despite evidence supporting an oncogenic role in breast cancer, the Notch pathway's contribution to metastasis remains unknown. Here, we report that the Notch ligand Jagged1 is a clinically and functionally important mediator of bone metastasis by activating the Notch pathway in bone cells. Jagged1 promotes tumor growth by stimulating IL-6 release from osteoblasts and directly activates osteoclast differentiation. Furthermore, Jagged1 is a potent downstream mediator of the bone metastasis cytokine TGFß that is released during bone destruction. Importantly, γ-secretase inhibitor treatment reduces Jagged1-mediated bone metastasis by disrupting the Notch pathway in stromal bone cells. These findings elucidate a stroma-dependent mechanism for Notch signaling in breast cancer and provide rationale for using γ-secretase inhibitors for the treatment of bone metastasis.


Subject(s)
Bone Neoplasms/secondary , Calcium-Binding Proteins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Mammary Neoplasms, Experimental/pathology , Membrane Proteins/physiology , Osteoblasts/metabolism , Receptors, Notch/metabolism , Signal Transduction , 3T3 Cells , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Cell Differentiation , Enzyme Inhibitors/pharmacology , Interleukin-6/metabolism , Jagged-1 Protein , Ligands , Mice , Osteoblasts/pathology , Serrate-Jagged Proteins , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism
3.
Blood ; 115(9): 1735-45, 2010 Mar 04.
Article in English | MEDLINE | ID: mdl-20007543

ABSTRACT

To identify dysregulated pathways in distinct phases of NOTCH1-mediated T-cell leukemogenesis, as well as small-molecule inhibitors that could synergize with or substitute for gamma-secretase inhibitors (GSIs) in T-cell acute lymphoblastic leukemia (T-ALL) therapy, we compared gene expression profiles in a Notch1-induced mouse model of T-ALL with those in human T-ALL. The overall patterns of NOTCH1-mediated gene expression in human and mouse T-ALLs were remarkably similar, as defined early in transformation in the mouse by the regulation of MYC and its target genes and activation of nuclear factor-kappaB and PI3K/AKT pathways. Later events in murine Notch1-mediated leukemogenesis included down-regulation of genes encoding tumor suppressors and negative cell cycle regulators. Gene set enrichment analysis and connectivity map algorithm predicted that small-molecule inhibitors, including heat-shock protein 90, histone deacetylase, PI3K/AKT, and proteasome inhibitors, could reverse the gene expression changes induced by NOTCH1. When tested in vitro, histone deacetylase, PI3K and proteasome inhibitors synergized with GSI in suppressing T-ALL cell growth in GSI-sensitive cells. Interestingly, alvespimycin, a potent inhibitor of the heat-shock protein 90 molecular chaperone, markedly inhibited the growth of both GSI-sensitive and -resistant T-ALL cells, suggesting that its loss disrupts signal transduction pathways crucial for the growth and survival of T-ALL cells.


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cell Transformation, Neoplastic/genetics , Down-Regulation , Enzyme Inhibitors/pharmacology , Gene Expression Profiling , Genes, myc , Humans , In Vitro Techniques , Leukemia, Experimental/drug therapy , Leukemia, Experimental/etiology , Leukemia, Experimental/genetics , Leukemia, Experimental/metabolism , Mice , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Receptor, Notch1/genetics , Signal Transduction , Species Specificity
4.
Cancer Res ; 69(7): 3060-8, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19318552

ABSTRACT

NOTCH signaling is deregulated in the majority of T-cell acute lymphoblastic leukemias (T-ALL) as a result of activating mutations in NOTCH1. Gamma secretase inhibitors (GSI) block proteolytic activation of NOTCH receptors and may provide a targeted therapy for T-ALL. We have investigated the mechanisms of GSI sensitivity across a panel of T-ALL cell lines, yielding an approach for patient stratification based on pathway activity and also providing a rational combination strategy for enhanced response to GSI. Whereas the NOTCH1 mutation status does not serve as a predictor of GSI sensitivity, a gene expression signature of NOTCH pathway activity does correlate with response, and may be useful in the selection of patients more likely to respond to GSI. Furthermore, inhibition of the NOTCH pathway activity signature correlates with the induction of the cyclin-dependent kinase inhibitors CDKN2D (p19(INK4d)) and CDKN1B (p27(Kip1)), leading to derepression of RB and subsequent exit from the cell cycle. Consistent with this evidence of cell cycle exit, short-term exposure of GSI resulted in sustained molecular and phenotypic effects after withdrawal of the compound. Combination treatment with GSI and a small molecule inhibitor of CDK4 produced synergistic growth inhibition, providing evidence that GSI engagement of the CDK4/RB pathway is an important mechanism of GSI action and supports further investigation of this combination for improved efficacy in treating T-ALL.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Cyclic S-Oxides/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protease Inhibitors/pharmacology , Receptor, Notch1/antagonists & inhibitors , Retinoblastoma Protein/metabolism , Thiadiazoles/pharmacology , Cell Line, Tumor , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p19/biosynthesis , Cyclin-Dependent Kinase Inhibitor p27 , G1 Phase/drug effects , G1 Phase/genetics , Gene Expression Profiling , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Phosphorylation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , S Phase/drug effects , S Phase/genetics , Signal Transduction/drug effects , Transcription, Genetic , Transfection
5.
Genetics ; 172(4): 2309-24, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16415372

ABSTRACT

Presenilin is the enzymatic component of gamma-secretase, a multisubunit intramembrane protease that processes several transmembrane receptors, such as the amyloid precursor protein (APP). Mutations in human Presenilins lead to altered APP cleavage and early-onset Alzheimer's disease. Presenilins also play an essential role in Notch receptor cleavage and signaling. The Notch pathway is a highly conserved signaling pathway that functions during the development of multicellular organisms, including vertebrates, Drosophila, and C. elegans. Recent studies have shown that Notch signaling is sensitive to perturbations in subcellular trafficking, although the specific mechanisms are largely unknown. To identify genes that regulate Notch pathway function, we have performed two genetic screens in Drosophila for modifiers of Presenilin-dependent Notch phenotypes. We describe here the cloning and identification of 19 modifiers, including nicastrin and several genes with previously undescribed involvement in Notch biology. The predicted functions of these newly identified genes are consistent with extracellular matrix and vesicular trafficking mechanisms in Presenilin and Notch pathway regulation and suggest a novel role for gamma-tubulin in the pathway.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Membrane Proteins/genetics , Receptors, Notch/genetics , Alleles , Amyloid beta-Protein Precursor/genetics , Animals , Crosses, Genetic , Enhancer Elements, Genetic , Extracellular Matrix , Female , Male , Mutation , Presenilin-1 , Receptors, Notch/metabolism , Signal Transduction , Tubulin/metabolism
6.
Genetics ; 170(1): 161-71, 2005 May.
Article in English | MEDLINE | ID: mdl-15744054

ABSTRACT

Mutations that inactivate the retinoblastoma (Rb) pathway are common in human tumors. Such mutations promote tumor growth by deregulating the G1 cell cycle checkpoint. However, uncontrolled cell cycle progression can also produce new liabilities for cell survival. To uncover such liabilities in Rb mutant cells, we performed a clonal screen in the Drosophila eye to identify second-site mutations that eliminate Rbf(-) cells, but allow Rbf(+) cells to survive. Here we report the identification of a mutation in a novel highly conserved peptidyl prolyl isomerase (PPIase) that selectively eliminates Rbf(-) cells from the Drosophila eye.


Subject(s)
Drosophila melanogaster/embryology , Eye/embryology , Peptidylprolyl Isomerase/genetics , Retinoblastoma Protein/genetics , Amino Acid Sequence , Animals , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Eye/enzymology , Molecular Sequence Data , Mutation
7.
Nat Genet ; 36(3): 288-92, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14981519

ABSTRACT

In fruit fly research, chromosomal deletions are indispensable tools for mapping mutations, characterizing alleles and identifying interacting loci. Most widely used deletions were generated by irradiation or chemical mutagenesis. These methods are labor-intensive, generate random breakpoints and result in unwanted secondary mutations that can confound phenotypic analyses. Most of the existing deletions are large, have molecularly undefined endpoints and are maintained in genetically complex stocks. Furthermore, the existence of haplolethal or haplosterile loci makes the recovery of deletions of certain regions exceedingly difficult by traditional methods, resulting in gaps in coverage. Here we describe two methods that address these problems by providing for the systematic isolation of targeted deletions in the D. melanogaster genome. The first strategy used a P element-based technique to generate deletions that closely flank haploinsufficient genes and minimize undeleted regions. This deletion set has increased overall genomic coverage by 5-7%. The second strategy used FLP recombinase and the large array of FRT-bearing insertions described in the accompanying paper to generate 519 isogenic deletions with molecularly defined endpoints. This second deletion collection provides 56% genome coverage so far. The latter methodology enables the generation of small custom deletions with predictable endpoints throughout the genome and should make their isolation a simple and routine task.


Subject(s)
DNA Transposable Elements , Drosophila melanogaster/genetics , Sequence Deletion , Animals , Genome , Mutagenesis, Insertional
SELECTION OF CITATIONS
SEARCH DETAIL
...