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.
Stem Cell Res Ther ; 8(1): 119, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28545562

ABSTRACT

BACKGROUND: Malignant pleural mesothelioma is an aggressive cancer, characterized by rapid progression and high mortality. Persistence of tumor-initiating cells (TICs, or cancer stem cells) after cytotoxic drug treatment is responsible for tumor relapse, and represents one of the main reasons for the poor prognosis of mesothelioma. In fact, identification of the molecules affecting TIC viability is still a significant challenge. METHODS: TIC-enriched cultures were obtained from 10 human malignant pleural mesotheliomas and cultured in vitro. Three fully characterized tumorigenic cultures, named MM1, MM3, and MM4, were selected and used to assess antiproliferative effects of the multi-kinase inhibitor sorafenib. Cell viability was investigated by MTT assay, and cell cycle analysis as well as induction of apoptosis were determined by flow cytometry. Western blotting was performed to reveal the modulation of protein expression and the phosphorylation status of pathways associated with sorafenib treatment. RESULTS: We analyzed the molecular mechanisms of the antiproliferative effects of sorafenib in mesothelioma TIC cultures. Sorafenib inhibited cell cycle progression in all cultures, but only in MM3 and MM4 cells was this effect associated with Mcl-1-dependent apoptosis. To investigate the mechanisms of sorafenib-mediated antiproliferative activity, TICs were treated with epidermal growth factor (EGF) or basic fibroblast growth factor (bFGF) causing, in MM3 and MM4 cells, MEK, ERK1/2, Akt, and STAT3 phosphorylation. These effects were abolished by sorafenib only in bFGF-treated cells, while a modest inhibition occurred after EGF stimulation, suggesting that sorafenib effects are mainly due to FGF receptor (FGFR) inhibition. Indeed, FGFR1 phosphorylation was inhibited by sorafenib. Moreover, in MM1 cells, which release high levels of bFGF and showed autocrine activation of FGFR1 and constitutive phosphorylation/activation of MEK-ERK1/2, sorafenib induced a more effective antiproliferative response, confirming that the main target of the drug is the inhibition of FGFR1 activity. CONCLUSIONS: These results suggest that, in malignant pleural mesothelioma TICs, bFGF signaling is the main target of the antiproliferative response of sorafenib, acting directly on the FGFR1 activation. Patients with constitutive FGFR1 activation via an autocrine loop may be more sensitive to sorafenib treatment and the analysis of this possibility warrants further clinical investigation.


Subject(s)
Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mesothelioma/drug therapy , Mesothelioma/pathology , Neoplastic Stem Cells/pathology , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Pleural Neoplasms/drug therapy , Pleural Neoplasms/pathology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Separation , Cell Survival/drug effects , Down-Regulation/drug effects , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Fibroblast Growth Factor 2/pharmacology , Humans , Lung Neoplasms/metabolism , Mesothelioma/metabolism , Mesothelioma, Malignant , Mice, Inbred NOD , Mice, SCID , Models, Biological , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Niacinamide/pharmacology , Niacinamide/therapeutic use , Phenylurea Compounds/pharmacology , Pleural Neoplasms/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction/drug effects , Sorafenib , Time Factors
2.
Cell Cycle ; 14(21): 3418-29, 2015.
Article in English | MEDLINE | ID: mdl-26566863

ABSTRACT

Notwithstanding current multimodal treatment, including surgery, radiotherapy and chemotherapy with temozolomide (TMZ), median survival of glioblastoma (GBM) patients is about 14 months, due to the rapid emergence of cell clones resistant to treatment. Therefore, understanding the mechanisms underlying chemoresistance is mandatory to improve treatments' outcome. We generated TMZ resistant cells (TMZ-R) from a GBM cell line and from cancer stem cell-enriched cultures isolated from human GBMs. We demonstrated that TMZ resistance is partially reverted by "drug wash-out" suggesting the contribution of epigenetic mechanisms in drug resistance and supporting the possibility of TMZ rechallenge in GBM patients after prior drug exposure. The expression of histone lysine demethylase genes (KDMs) was increased in TMZ-R cells compared to parental cells, and TMZ resistance or restored sensitivity was mimicked by over-expressing or inactivating KDM5A. Methylation and expression of O6-methylguanine-DNA methyltransferase (MGMT) and drug efflux mechanisms were not altered in TMZ-R cells compared to parental TMZ sensitive cells. TMZ-R cells transiently acquired morphologic and molecular characteristics of differentiated tumor cells, features that were lost after drug wash-out. In conclusion, we demonstrated that treatment-induced TMZ resistance in GBM involves epigenetic mechanisms in a subset of slow-cycling and transiently partially differentiated cells that escape drug cytotoxicity, overcome G2 checkpoint and sustain clonal growth. We found that TMZ-R cells are sensitive to histone deacethylase inhibitors (HDACi) that synergize with TMZ. This strong synergism could be exploited to develop novel combined adjuvant therapies for this rapidly progressing and invariably lethal cancer.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm , Glioblastoma/drug therapy , Neoplastic Stem Cells/drug effects , Retinoblastoma-Binding Protein 2/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Shape/drug effects , DNA Methylation/drug effects , Dacarbazine/pharmacology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , Drug Synergism , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genotype , Glioblastoma/enzymology , Glioblastoma/genetics , Glioblastoma/pathology , Histone Deacetylase Inhibitors/pharmacology , Humans , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Phenotype , RNA Interference , Retinoblastoma-Binding Protein 2/antagonists & inhibitors , Retinoblastoma-Binding Protein 2/genetics , Temozolomide , Time Factors , Transfection
3.
BMC Cancer ; 15: 228, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25884842

ABSTRACT

BACKGROUND: Cancer stem cells (CSCs) are considered the cell subpopulation responsible for breast cancer (BC) initiation, growth, and relapse. CSCs are identified as self-renewing and tumor-initiating cells, conferring resistance to chemo- and radio-therapy to several neoplasias. Nowadays, th (about 10mM)e pharmacological targeting of CSCs is considered an ineludible therapeutic goal. The antidiabetic drug metformin was reported to suppress in vitro and in vivo CSC survival in different tumors and, in particular, in BC preclinical models. However, few studies are available on primary CSC cultures derived from human postsurgical BC samples, likely because of the limited amount of tissue available after surgery. In this context, comparative oncology is acquiring a relevant role in cancer research, allowing the analysis of larger samples from spontaneous pet tumors that represent optimal models for human cancer. METHODS: Isolation of primary canine mammary carcinoma (CMC) cells and enrichment in stem-like cell was carried out from fresh tumor specimens by culturing cells in stem-permissive conditions. Phenotypic and functional characterization of CMC-derived stem cells was performed in vitro, by assessment of self-renewal, long-lasting proliferation, marker expression, and drug sensitivity, and in vivo, by tumorigenicity experiments. Corresponding cultures of differentiated CMC cells were used as internal reference. Metformin efficacy on CMC stem cell viability was analyzed both in vitro and in vivo. RESULTS: We identified a subpopulation of CMC cells showing human breast CSC features, including expression of specific markers (i.e. CD44, CXCR4), growth as mammospheres, and tumor-initiation in mice. These cells show resistance to doxorubicin but were highly sensitive to metformin in vitro. Finally, in vivo metformin administration significantly impaired CMC growth in NOD-SCID mice, associated with a significant depletion of CSCs. CONCLUSIONS: Similarly to the human counterpart, CMCs contain stem-like subpopulations representing, in a comparative oncology context, a valuable translational model for human BC, and, in particular, to predict the efficacy of antitumor drugs. Moreover, metformin represents a potential CSC-selective drug for BC, as effective (neo-)adjuvant therapy to eradicate CSC in mammary carcinomas of humans and animals.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Mammary Neoplasms, Animal , Metformin/pharmacology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Dogs , Drug Resistance, Neoplasm , ErbB Receptors/metabolism , Estrogen Receptor alpha/metabolism , Female , Humans , Hyaluronan Receptors/metabolism , Ki-67 Antigen/metabolism , Metformin/pharmacokinetics , Mice , Phenotype , Xenograft Model Antitumor Assays
4.
Int J Cancer ; 136(6): 1445-57, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25091220

ABSTRACT

We have recently reported that glioblastoma (GB)-initiating cells (GIC) with low expression and/or mutation of TP53 and high expression of PI3K ("responder" genetic profile) can be effectively and safely radiosensitized by the ATM inhibitor KU60019. We report here on drug's diffusion and elimination from the animal body and brain, its effects on orthotopic GB and efficacy toward pediatric GIC. Healthy mice were infused by convection enhanced delivery (CED) with KU60019 and the drug kinetics followed by high performance liquid chromatography-mass spectrometry. Already at the end of CED, KU60019 had diffused from the injection site to the ipsilateral and, to a lower extent, controlateral hemisphere. After 24 hr, no drug could be detected all over the brain or in other organs, indicating rapid draining and excretion. After intraperitoneal injection, traces only of KU60019 could be detected in the brain, indicating inability to cross the brain-blood barrier. Consistent with the induction of cell cycle progression previously observed in vitro, KU60019 stimulated proliferation of orthotopic GB cells with the highest effect observed 96 hr after drug delivery. Adult GIC with high expression of TP53 and low expression of PI3K could be radiosensitized by KU60019, although less promptly than GIC bearing the "responder" profile. Consistent with the kinetics of proliferation induction, the highest radiosensitizing effect was observed 96 hr after delivery of KU60019 to GIC. Pediatric GIC could be similarly radiosensitized after exposure to KU60019. The results indicate that ATM inhibition may allow to radiosensitize a wide range of adult and pediatric high-grade gliomas.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Brain Neoplasms/drug therapy , Glioma/drug therapy , Morpholines/pharmacokinetics , Radiation-Sensitizing Agents/pharmacokinetics , Thioxanthenes/pharmacokinetics , Adult , Animals , Brain Neoplasms/pathology , Child , Glioma/pathology , Humans , Ki-67 Antigen/analysis , Mice , Morpholines/pharmacology , Morpholines/toxicity , Thioxanthenes/pharmacology , Thioxanthenes/toxicity
5.
Oncotarget ; 5(22): 11252-68, 2014 Nov 30.
Article in English | MEDLINE | ID: mdl-25361004

ABSTRACT

Epidemiological and preclinical studies propose that metformin, a first-line drug for type-2 diabetes, exerts direct antitumor activity. Although several clinical trials are ongoing, the molecular mechanisms of this effect are unknown. Here we show that chloride intracellular channel-1 (CLIC1) is a direct target of metformin in human glioblastoma cells. Metformin exposure induces antiproliferative effects in cancer stem cell-enriched cultures, isolated from three individual WHO grade IV human glioblastomas. These effects phenocopy metformin-mediated inhibition of a chloride current specifically dependent on CLIC1 functional activity. CLIC1 ion channel is preferentially active during the G1-S transition via transient membrane insertion. Metformin inhibition of CLIC1 activity induces G1 arrest of glioblastoma stem cells. This effect was time-dependent, and prolonged treatments caused antiproliferative effects also for low, clinically significant, metformin concentrations. Furthermore, substitution of Arg29 in the putative CLIC1 pore region impairs metformin modulation of channel activity. The lack of drugs affecting cancer stem cell viability is the main cause of therapy failure and tumor relapse. We identified CLIC1 not only as a modulator of cell cycle progression in human glioblastoma stem cells but also as the main target of metformin's antiproliferative activity, paving the way for novel and needed pharmacological approaches to glioblastoma treatment.


Subject(s)
Chloride Channels/antagonists & inhibitors , Glioblastoma/drug therapy , Metformin/pharmacology , Neoplastic Stem Cells/drug effects , Aged , Animals , Antineoplastic Agents/pharmacology , CHO Cells , Chloride Channels/metabolism , Cricetulus , Drug Repositioning , Female , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Hypoglycemic Agents/pharmacology , Male , Middle Aged , Models, Molecular , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology
6.
PLoS One ; 9(2): e90085, 2014.
Article in English | MEDLINE | ID: mdl-24587218

ABSTRACT

Reactive oxygen species (ROS) are metabolism by-products that may act as signaling molecules to sustain tumor growth. Antioxidants have been used to impair cancer cell survival. Our goal was to determine the mechanisms involved in the response to antioxidants of a human cell culture (PT4) containing glioblastoma (GBM) tumorigenic initiating cells (TICs). ROS production in the absence or presence of N-acetyl-L-cysteine (NAC), tiron, and trolox was evaluated by flow cytometry (FCM). The effects of these antioxidants on cell survival and apoptosis were evaluated by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay (MTT) and FCM. The biological processes modulated by these drugs were determined by oligonucleotide microarray gene expression profiling. Our results showed that NAC, tiron and trolox impaired PT4 cell survival, had minor effects on ROS levels and caused wide deregulation of cell cycle genes. Furthermore, tiron and trolox caused inhibition of cell survival in two additional cell cultures containing TICs, FO-1 and MM1, established from a melanoma and a mesothelioma patient, respectively. NAC, instead, impaired survival of the MM1 cells but not of the FO-1 cells. However, when used in combination, NAC enhanced the inhibitory effect of PLX4032 (BRAF V600E inhibitor) and Gefitinib (EGFR inhibitor), on FO-1 and PT4 cell survival. Collectively, NAC, tiron and trolox modulated gene expression and impaired the growth of cultures containing TICs primarily by inhibiting cell cycle progression.


Subject(s)
1,2-Dihydroxybenzene-3,5-Disulfonic Acid Disodium Salt/pharmacology , Acetylcysteine/pharmacology , Cell Cycle Proteins/genetics , Cell Cycle/drug effects , Chromans/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Cell Cycle/genetics , Cell Cycle Proteins/metabolism , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Profiling , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Reactive Oxygen Species/metabolism , Signal Transduction , Tumor Cells, Cultured
7.
Int J Cancer ; 135(2): 479-91, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24443327

ABSTRACT

We have previously shown that pharmacological inhibition of ataxia telangiectasia mutated (ATM) protein sensitizes glioblastoma-initiating cells (GICs) to ionizing radiation (IR). Herein, we report the experimental conditions to overcome GIC radioresistance in vitro using the specific ATM inhibitor KU-60019, two major determinants of the tumor response to this drug and the absence of toxicity of this treatment in vitro and in vivo. Repeated treatments with KU-60019 followed by IR substantially delayed GIC proliferation in vitro and even eradicated radioresistant cells, whereas GIC treated with vehicle plus radiation recovered early and expanded. The tumor response to the drug occurred under a cutoff level of expression of TP53 and over a cutoff level of expression of phosphatidylinositol 3-kinase (PI3K). No increased clastogenicity or point mutagenicity was induced by KU-60019 plus radiation when compared to vehicle plus radiation. No significant histological changes to the brain or other organs were observed after prolonged infusion into the brain of KU-60019 at millimolar concentrations. Taken together, these findings suggest that GIC-driven tumors with low expression of TP53 and high expression of PI3K might be effectively and safely radiosensitized by KU-60019.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Morpholines/pharmacology , Neoplastic Stem Cells/drug effects , Radiation-Sensitizing Agents/pharmacology , Thioxanthenes/pharmacology , Animals , Cell Line, Tumor , Humans , Mice , Radiation Tolerance/drug effects , Xenograft Model Antitumor Assays
8.
Cell Cycle ; 12(3): 491-500, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23324350

ABSTRACT

Glioblastomas are grade IV brain tumors characterized by high aggressiveness and invasiveness, giving patients a poor prognosis. We investigated the effects of the multi-kinase inhibitor sorafenib on six cultures isolated from human glioblastomas and maintained in tumor initiating cells-enriching conditions. These cell subpopulations are thought to be responsible for tumor recurrence and radio- and chemo-resistance, representing the perfect target for glioblastoma therapy. Sorafenib reduces proliferation of glioblastoma cultures, and this effect depends, at least in part, on the inhibition of PI3K/Akt and MAPK pathways, both involved in gliomagenesis. Sorafenib significantly induces apoptosis/cell death via downregulation of the survival factor Mcl-1. We provide evidence that sorafenib has a selective action on glioblastoma stem cells, causing enrichment of cultures in differentiated cells, downregulation of the expression of stemness markers required to maintain malignancy (nestin, Olig2 and Sox2) and reducing cell clonogenic ability in vitro and tumorigenic potential in vivo. The selectivity of sorafenib effects on glioblastoma stem cells is confirmed by the lower sensitivity of glioblastoma cultures after differentiation as compared with the undifferentiated counterpart. Since current GBM therapy enriches the tumor in cancer stem cells, the evidence of a selective action of sorafenib on these cells is therapeutically relevant, even if, so far, results from first phase II clinical trials did not demonstrate its efficacy.


Subject(s)
Glioblastoma/metabolism , Neoplastic Stem Cells/drug effects , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Apoptosis/drug effects , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Down-Regulation , Glioblastoma/drug therapy , Humans , Intermediate Filament Proteins/biosynthesis , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein , Neoplastic Stem Cells/metabolism , Nerve Tissue Proteins/biosynthesis , Nestin , Niacinamide/pharmacology , Oligodendrocyte Transcription Factor 2 , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , SOXB1 Transcription Factors/biosynthesis , STAT3 Transcription Factor/drug effects , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Sorafenib , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...