Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Transl Oncol ; 8(1): 1-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25749171

ABSTRACT

Our group has previously reported that the majority of human melanomas (>60%) express the metabotropic glutamate receptor 1 (GRM1) and that the glutamate release inhibitor riluzole, a drug currently used to treat amyotrophic lateral sclerosis, can induce apoptosis in GRM1-expressing melanoma cells. Our group previously reported that in vitro riluzole treatment reduces cell growth in three-dimensional (3D) soft agar colony assays by 80% in cells with wildtype phosphoinositide 3-kinase (PI3K) pathway activation. However, melanoma cell lines harboring constitutive activating mutations of the PI3K pathway (PTEN and NRAS mutations) showed only a 35% to 40% decrease in colony formation in soft agar in the presence of riluzole. In this study, we have continued our preclinical studies of riluzole and its effect on melanoma cells alone and in combination with inhibitors of the PI3 kinase pathway: the AKT inhibitor, API-2, and the mammalian target of rapamycin (mTOR) inhibitor, rapamycin. We modeled these combinatorial therapies on various melanoma cell lines in 3D and 2D systems and in vivo. Riluzole combined with mTOR inhibition is more effective at halting melanoma anchorage-independent growth and xenograft tumor progression than either agent alone. PI3K signaling changes associated with this combinatorial treatment shows that 3D (nanoculture) modeling of cell signaling more closely resembles in vivo signaling than monolayer models. Riluzole combined with mTOR inhibition is effective at halting tumor cell progression independent of BRAF mutational status. This makes this combinatorial therapy a potentially viable alternative for metastatic melanoma patients who are BRAF WT and are therefore ineligible for vemurafenib therapy.

2.
Clin Cancer Res ; 17(22): 7080-92, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21844014

ABSTRACT

PURPOSE: Melanoma is a heterogeneous disease where monotherapies are likely to fail due to variations in genomic signatures. B-RAF inhibitors have been clinically inadequate but response might be augmented with combination therapies targeting multiple signaling pathways. We investigate the preclinical efficacy of combining the multikinase inhibitor sorafenib or the mutated B-RAF inhibitor PLX4720 with riluzole, an inhibitor of glutamate release that antagonizes metabotropic glutamate receptor 1 (GRM1) signaling in melanoma cells. EXPERIMENTAL DESIGN: Melanoma cell lines that express GRM1 and either wild-type B-RAF or mutated B-RAF were treated with riluzole, sorafenib, PLX4720, or the combination of riluzole either with sorafenib or with PLX4720. Extracellular glutamate levels were determined by glutamate release assays. MTT assays and cell-cycle analysis show effects of the compounds on proliferation, viability, and cell-cycle profiles. Western immunoblotting and immunohistochemical staining showed apoptotic markers. Consequences on mitogen-activated protein kinase pathway were assessed by Western immunoblotting. Xenograft tumor models were used to determine the efficacy of the compounds in vivo. RESULTS: The combination of riluzole with sorafenib exhibited enhanced antitumor activities in GRM1-expressing melanoma cells harboring either wild-type or mutated B-RAF. The combination of riluzole with PLX4720 showed lessened efficacy compared with the combination of riluzole and sorafenib in suppressing the growth of GRM1-expressing cells harboring the B-RAF(V600E) mutation. CONCLUSIONS: The combination of riluzole with sorafenib seems potent in suppressing tumor proliferation in vitro and in vivo in GRM1-expressing melanoma cells regardless of B-RAF genotype and may be a viable therapeutic clinical combination.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Glutamic Acid/metabolism , Melanoma/drug therapy , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction/drug effects , Animals , Benzenesulfonates/administration & dosage , Benzenesulfonates/therapeutic use , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Indoles/administration & dosage , Indoles/therapeutic use , Melanoma/metabolism , Mice , Mice, Nude , Molecular Targeted Therapy , Mutation , Niacinamide/analogs & derivatives , Phenylurea Compounds , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , Pyridines/administration & dosage , Pyridines/therapeutic use , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Riluzole/administration & dosage , Riluzole/therapeutic use , Sorafenib , Sulfonamides/administration & dosage , Sulfonamides/therapeutic use , Xenograft Model Antitumor Assays
3.
Pigment Cell Melanoma Res ; 24(3): 512-24, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21477078

ABSTRACT

Melanoma cells are resistant to transforming growth factor-ß (TGFß)-induced cell-cycle arrest. In this study, we investigated a mechanism of resistance involving a regulatory domain, called linker region, in Smad2 and Smad3, main downstream effectors of TGFß. Melanoma cells in culture and tumor samples exhibited constitutive Smad2 and Smad3 linker phosphorylation. Treatment of melanoma cells with the MEK1/2 inhibitor, U0126, or the two pan-CDK and GSK3 inhibitors, Flavopiridol and R547, resulted in decreased linker phosphorylation of Smad2 and Smad3. Overexpression of the linker phosphorylation-resistant Smad3 EPSM mutant in melanoma cells resulted in an increase in expression of p15(INK4B) and p21(WAF1) , as compared with cells transfected with wild-type (WT) Smad3. In addition, the cell numbers of EPSM Smad3-expressing melanoma cells were significantly reduced compared with WT Smad3-expressing cells. These results suggest that the linker phosphorylation of Smad3 contributes to the resistance of melanoma cells to TGFß-mediated growth inhibition.


Subject(s)
Melanoma/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/pharmacology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p15/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Drug Resistance, Neoplasm , Flavonoids/pharmacology , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 2/genetics , MAP Kinase Kinase 2/metabolism , Melanoma/genetics , Melanoma/pathology , Mutation , Phosphorylation/drug effects , Phosphorylation/genetics , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Pyrimidines/pharmacology , Smad2 Protein/genetics , Smad3 Protein/genetics
4.
J Invest Dermatol ; 130(9): 2240-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20505744

ABSTRACT

The goal of this study was to examine the effects of metabotropic glutamate receptor-1 (GRM1) blockade on melanoma anchorage-independent growth and invasion. We performed colony and invasion assays using GRM1-expressing melanoma lines and the GRM1-negative UACC930 line. Using the glutamate-release inhibitor Riluzole or the non-competitive GRM1 antagonist BAY 36-7620 we were able to induce considerable inhibition of colony formation and invasion in GRM1-expressing melanoma lines. Neither pharmacological agent induced significant reduction in colony formation or invasion in the GRM1-negative melanoma line, UACC930. Additionally we assessed the efficacy of these inhibitors to inhibit the growth of fresh melanoma tumor samples cultured on a 74-mum nylon mesh. Both Riluzole and BAY 36-7620 significantly inhibited tumor cell growth into the interstitial spaces of the mesh. When repeated with normal mole samples both inhibitors were much less effective in preventing the outgrowth of cells. These experiments show that a specific antagonist of GRM1 (BAY 36-7620) or an inhibitor of glutamate release (Riluzole) can significantly suppress melanoma migration, invasion and colony formation as well as inhibit the proliferation of fresh melanoma cells. These findings, added to our previous work, strengthen the case that GRM1 is a valid therapeutic target in patients with melanoma.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/metabolism , Melanoma/drug therapy , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Riluzole/pharmacology , Skin Neoplasms/drug therapy , Cell Division/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Melanoma/metabolism , Melanoma/pathology , Naphthalenes/pharmacology , Neoplasm Invasiveness , Organ Culture Techniques , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction/drug effects , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
5.
Gynecol Oncol ; 114(3): 516-22, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19576622

ABSTRACT

OBJECTIVE: The current standard treatment for ovarian carcinoma, consisting of surgery followed by chemotherapy with carboplatin and paclitaxel, is fraught with a high rate of recurrences. We hypothesized that targeted inhibition of specific signaling pathways in combination with conventional drugs may increase chemotherapeutic efficacy. METHODS: We analyzed the expression and activation profiles of various signaling pathways in nine established ovarian cancer cell lines (CAOV-3, ES2, PA-1, SKOV-3, NIHOVCAR3, OV90, TOV112D, A1847, A2780) and 24 freshly procured human ovarian tumors. The PI3 kinase pathway component Akt was frequently overexpressed and/or activated in tumor cells. The effect of several PI3K pathway inhibitors (rapamycin, LY294002, SH-6) and rapamycin in combination with carboplatin on various tumor cell growth characteristics was tested in cell lines and fresh tumor-derived transient monolayer and organ cultures. RESULTS: Rapamycin by itself and additively with carboplatin inhibited the growth and invasion, and increased the sensitivity to anoikis of most of the ovarian cancer cell lines and fresh tumors. The additive inhibitory effect may be due to enhanced apoptosis as demonstrated by Poly-ADP-Ribose Polymerase (PARP) cleavage and Annexin V staining in cells treated with both rapamycin and carboplatin. CONCLUSIONS: Rapamycin in combination with standard chemotherapeutic agents may improve the efficiency of ovarian cancer treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Ovarian Neoplasms/drug therapy , Sirolimus/pharmacology , Apoptosis/drug effects , Carboplatin/administration & dosage , Cell Growth Processes/drug effects , Cell Line, Tumor , Chromones/administration & dosage , Chromones/pharmacology , Drug Synergism , Female , Humans , Mechanistic Target of Rapamycin Complex 1 , Morpholines/administration & dosage , Morpholines/pharmacology , Multiprotein Complexes , Oncogene Protein v-akt/antagonists & inhibitors , Oncogene Protein v-akt/biosynthesis , Oncogene Protein v-akt/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositols/administration & dosage , Phosphatidylinositols/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Proteins , Signal Transduction/drug effects , Sirolimus/administration & dosage , TOR Serine-Threonine Kinases , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism
6.
Clin Cancer Res ; 15(11): 3896-902, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19458050

ABSTRACT

PURPOSE: Ectopic expression of GRM1 in murine melanocytes results in transformation into a form of melanoma, and more than 60% of human melanoma samples tested ectopically express GRM1. Stimulation of this receptor in vitro results in up-regulation of activated extracellular signal-regulated kinase (ERK). Furthermore, a xenograft model of melanoma treated with riluzole, an oral GRM1 blocking agent, showed decreased tumor growth compared with the untreated controls. We have now completed a phase 0 trial of riluzole in patients with melanoma. EXPERIMENTAL DESIGN: Patients enrolled on this trial underwent a pretreatment biopsy, took 200 mg of oral riluzole per day for 14 days, and then underwent resection of their remaining tumor. We compared the levels of pERK and pAKT in the pretreatment and post-treatment samples and assessed the metabolic activity of pretreatment and post-treatment tumors using fluorodeoxyglucose positron emission tomography (FDG-PET) scanning. RESULTS: We accrued 12 patients and all expressed GRM1. We found a significant decrease in pAKT and/or pERK in post-treatment tumor samples as compared with pretreatment samples in 4 (34%) patients. These four patients had a significant decrease in FDG-PET intensity post-treatment as well. Two other patients had a clinical response with no corresponding metabolic response; five patients had similar pretreatment and post-treatment FDG-PET scan findings; and one patient had progressive disease. CONCLUSIONS: Our data show that glutamate blockade with riluzole can inhibit signaling through the mitogen-activated protein kinase and phosphatidylinositol 3-kinase/AKT pathways and suppress the metabolic activity of melanoma. The ectopic expression of metabotropic glutamate receptors may be important in the pathogenesis of human melanoma, and targeting this pathway may be an effective therapy.


Subject(s)
Melanoma/drug therapy , Riluzole/therapeutic use , Blotting, Western , Caspase 3/metabolism , Enzyme Activation/drug effects , Excitatory Amino Acid Antagonists/therapeutic use , Extracellular Signal-Regulated MAP Kinases/metabolism , Fluorodeoxyglucose F18 , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Melanoma/genetics , Melanoma/metabolism , Mutation , Neoplasm Staging , Oncogene Protein v-akt/metabolism , Positron-Emission Tomography , Proto-Oncogene Proteins B-raf/genetics , Receptors, Metabotropic Glutamate/metabolism , ras Proteins/genetics
7.
Int J Cancer ; 121(1): 157-64, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17304506

ABSTRACT

The objective of this study was to assess the anti-tumor efficacy of rapamycin alone or in combination with herceptin in breast cancer. A total of 20 human breast cancer lines were examined for expression of various receptor tyrosine kinases and activation of their down stream signaling molecules, as well as for their invasion and colony forming ability. The ErbB2 and PI3 kinase pathway inhibitors were tested for the inhibition on breast cancer cell growth and tumor development. Seven of the 20 lines displayed an elevated level of ErbB2, others had varying level of EGF, IGF-1 or insulin receptor. Over 30% of the lines also had constitutive activation of Akt and MAP kinase. The lines displayed a wide range of colony forming and invasion ability. The PI3 kinase pathway inhibitors LY294002 and rapamycin inhibited the colony forming ability of all of the lines with the ErbB2 overexpressing lines having a higher sensitivity. A similar trend was observed for inhibition of invasion by LY294002. Rapamycin alone and additively together with herceptin inhibited the breast cancer cell growth especially in ErbB2 overexpressing cells. Rapamycin and herceptin synergistically inhibited tumor growth and endpoint tumor load in a xenograft model using a MCF-7 subline and in a MMTV-ErbB2 transgenic model. Rapamycin and herceptin significantly reduced the level of cyclin D1 and D3 and increased the cleavage of caspase 3 suggesting an increased apoptosis. Our results suggest that rapamycin together with herceptin has an enhanced anti-cancer effect and could be developed as an improved therapeutic regimen for breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Receptor, ErbB-2/metabolism , Sirolimus/pharmacology , Animals , Breast Neoplasms/pathology , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/metabolism , Cyclin D3 , Cyclins/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Invasiveness/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
8.
J Biol Chem ; 277(13): 11107-15, 2002 Mar 29.
Article in English | MEDLINE | ID: mdl-11799110

ABSTRACT

Using loss-of-function mutants of Ros and inducible epidermal growth factor receptor-Ros chimeras we investigated the role of various signaling pathways in Ros-induced cell transformation. Inhibition of the mitogen-activated protein kinase (MAPK) pathway with the MEK (MAP/extracellular signal-regulated kinase kinase) inhibitor PD98059 had little effect on the Ros-induced monolayer and anchorage-independent growth of chicken embryo fibroblasts and NIH3T3 cells even though more than 70% of the MAPK was inhibited. In contrast, inhibiting the phosphatidylinositol 3-kinase (PI3K) pathway with the drug LY294002, a dominant negative mutant of PI3K, Deltap85, or the phosphatidylinositol phosphatase PTEN (phosphatase and tensin homologue deleted in chromosome ten) resulted in a dramatic reduction of v-Ros- and epidermal growth factor receptor-Ros-promoted anchorage-independent growth of chicken embryo fibroblasts and NIH3T3 cells, respectively. Parallel and downstream components of PI3K signaling such as the Rho family GTPases (Rac, Rho, Cdc42) and the survival factor Akt were all shown to contribute to Ros-induced anchorage-independent growth, although Rac appeared to be less important for Ros-induced colony formation in NIH3T3 cells. Furthermore, the transformation-attenuated v-Ros mutants F419 and DI could be complemented by constitutively active mutants of PI3K and Akt. Finally, we found that overexpressing a constitutively active mutant of STAT3 (STAT3C) conferred a resistance to the inhibition of Ros-induced anchorage-independent growth by LY294002, suggesting a possible overlap of functions between PI3K and STAT3 signaling in mediating Ros-induced anchorage-independent growth.


Subject(s)
Cell Transformation, Neoplastic , DNA-Binding Proteins/physiology , GTP Phosphohydrolases/physiology , Phosphatidylinositol 3-Kinases/physiology , Repressor Proteins/physiology , Trans-Activators/physiology , Animals , Cell Adhesion , Cell Division , Cell Line , Chick Embryo , Chromones/pharmacology , Fibroblasts , Flavonoids/pharmacology , Genes, Dominant , MAP Kinase Signaling System , Mice , Morpholines/pharmacology , Mutation , Phosphatidylinositol 3-Kinases/genetics , STAT3 Transcription Factor , Sirolimus/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...