Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Br J Cancer ; 116(11): 1425-1435, 2017 May 23.
Article in English | MEDLINE | ID: mdl-28441382

ABSTRACT

BACKGROUND: Several evidences suggest a marked angiogenic dependency in triple-negative breast cancer (TNBC) tumorigenesis and a potential sensitivity to anti-angiogenic agents. Herein, the putative role of Hedgehog (Hh) pathway in regulating TNBC-dependent angiogenesis was investigated. METHODS: Expression and regulation of the Hh pathway transcription factor glioma-associated oncogene homolog1 protein (GLI1) were studied on the endothelial compartment and on TNBC-initiated angiogenesis. To evaluate the translational relevance of our findings, the combination of paclitaxel with the Smo inhibitor NVP-LDE225 was tested in TNBC xenografted mice. RESULTS: Tissue microarray analysis on 200 TNBC patients showed GLI1 overexpression paired with vascular endothelial growth factor receptor 2 (VEGFR2) expression. In vitro, Hh pathway promotes TNBC progression in an autocrine manner, regulating the VEGF/VEGFR2 loop on cancer cell surface, and in a paracrine manner, orchestrating tumour vascularisation. These effects were counteracted by Smo pharmacological inhibition. In TNBC xenografted mice, scheduling NVP-LDE225 rather than bevacizumab provided a better sustained inhibition of TNBC cells proliferation and endothelial cells organisation. CONCLUSIONS: This study identifies the Hh pathway as one of the main regulators of tumour angiogenesis in TNBC, thus suggesting Hh inhibition as a potential new anti-angiogenic therapeutic option to be clinically investigated in GLI1 overexpressing TNBC patients.


Subject(s)
Hedgehog Proteins/metabolism , Neovascularization, Pathologic/metabolism , Signal Transduction , Triple Negative Breast Neoplasms/metabolism , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab/pharmacology , Biphenyl Compounds/administration & dosage , Cell Proliferation/drug effects , Coculture Techniques , Endothelial Cells/drug effects , Female , Gene Silencing , Hedgehog Proteins/antagonists & inhibitors , Human Umbilical Vein Endothelial Cells , Humans , MCF-7 Cells , Membrane Proteins , Mice , Mice, Nude , Middle Aged , Neoplasm Transplantation , Neovascularization, Pathologic/drug therapy , Paclitaxel/administration & dosage , Pyridines/administration & dosage , RNA, Messenger/metabolism , Thrombospondin 1/genetics , Thrombospondin 1/metabolism , Tissue Array Analysis , Transfection , Triple Negative Breast Neoplasms/chemistry , Triple Negative Breast Neoplasms/drug therapy , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/analysis , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Young Adult , Zinc Finger Protein GLI1/analysis
2.
Clin Mol Allergy ; 14: 7, 2016.
Article in English | MEDLINE | ID: mdl-27499704

ABSTRACT

BACKGROUND: Airborne allergens can induce an immunological chronic disease characterized by airway hyper responsiveness and inflammation, mediated by exaggerated Th2 immune response. Allergen-specific immunotherapy (AIT) is effective for treating this condition because it is able to modify its natural course by opposing the underlying pathogenic mechanisms and determining immune suppression, immune deviation and tolerance. The rational for the present study was to investigate the possibility of improving allergoid-based IT in terms of efficacy and safety. Recently, 1α,25-dihydroxyvitamin D3 (VD3), the active metabolite of vitamin D3, was described to be a potent inducer of T regulatory cells and to be a good adjuvant in AIT settings. METHODS: We investigated whether the co-administration of VD3 could potentiate the effect of AIT even when added to a low dose of chemically-modified monomeric allergoid of Der p 2 (d2-OID), in a Derp p 2 (d2)-sensitized BALB/c mice model. Control groups where treated with sham, VD3 alone or d2-OID only. RESULTS: The d2-OID alone was not fully successful, as expected for a low dose. VD3 administration was associated with some valuable, although limited, changes in the immunological parameters in the lung. On the contrary, the VD3 adjuvated allergoid vaccine induced the most prominent reduction of airway eosinophilia and Th2 cytokines and concomitant increase of T regulatory cells and IL-10 in the lung and Der p 2-specific IgG2a in the serum. CONCLUSIONS: The addition of VD3 to a conventional AIT protocol would allow the reduction of allergoid dose needed and therefore, the production costs. Moreover, beneficial immunomodulatory effects have been achieved by the oral administration which might favour the management of the therapy by the patients and their adherence, possibly enhancing the efficacy of the treatment.

3.
Clin Mol Allergy ; 13(1): 13, 2015.
Article in English | MEDLINE | ID: mdl-26180517

ABSTRACT

Almost all people in developed countries are exposed to metal nanoparticles (MeNPs) that are used in a large number of applications including medical (for diagnostic and therapeutic purposes). Once inside the body, absorbed by inhalation, contact, ingestion and injection, MeNPs can translocate to tissues and, as any foreign substance, are likely to encounter the innate immunity system that represent a non-specific first line of defense against potential threats to the host. In this review, we will discuss the possible effects of MeNPs on various components of the innate immunity (both specific cells and barriers). Most important is that there are no reports of immune diseases induced by MeNPs exposure: we are operating in a safe area. However, in vitro assays show that MeNPs have some effects on innate immunity, the main being toxicity (both cyto- and genotoxicity) and interference with the activity of various cells through modification of membrane receptors, gene expression and cytokine production. Such effects can have both negative and positive relevant impacts on humans. On the one hand, people exposed to high levels of MeNPs, as workers of industries producing or applying MeNPs, should be monitored for possible health effects. On the other hand, understanding the modality of the effects on immune responses is essential to develop medical applications for MeNPs. Indeed, those MeNPs that are able to stimulate immune cells could be used to develop of new vaccines, promote immunity against tumors and suppress autoimmunity.

4.
Breast Cancer Res ; 16(3): R45, 2014 May 05.
Article in English | MEDLINE | ID: mdl-24887236

ABSTRACT

INTRODUCTION: Src tyrosine kinase overactivation has been correlated with a poor response to human epidermal growth factor receptor 2 (HER2) inhibitors in breast cancer. To identify the mechanism by which Src overexpression sustains this resistance, we tested a panel of breast cancer cell lines either sensitive or resistant to lapatinib. METHODS: To determine the role of Src in lapatinib resistance, we evaluated the effects of Src inhibition/silencing in vitro on survival, migration, and invasion of lapatinib-resistant cells. In vivo experiments were performed in JIMT-1 lapatinib-resistant cells orthotopically implanted in nude mice. We used artificial metastasis assays to evaluate the effect of Src inhibition on the invasiveness of lapatinib-resistant cells. Src-dependent signal transduction was investigated with Western blot and ELISA analyses. RESULTS: Src activation was higher in lapatinib-resistant than in lapatinib-sensitive cells. The selective small-molecule Src inhibitor saracatinib combined with lapatinib synergistically inhibited the proliferation, migration, and invasion of lapatinib-resistant cells. Saracatinib combined with lapatinib significantly prolonged survival of JIMT-1-xenografted mice compared with saracatinib alone, and impaired the formation of lung metastases. Unexpectedly, in lapatinib-resistant cells, Src preferentially interacted with epidermal growth factor receptor (EGFR) rather than with HER2. Moreover, EGFR targeting and lapatinib synergistically inhibited survival, migration, and invasion of resistant cells, thereby counteracting Src-mediated resistance. These findings demonstrate that Src activation in lapatinib-resistant cells depends on EGFR-dependent rather than on HER2-dependent signaling. CONCLUSIONS: Complete pharmacologic EGFR/HER2 inhibition is required to reverse Src-dependent resistance to lapatinib in breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , ErbB Receptors/antagonists & inhibitors , Quinazolines/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , src-Family Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzodioxoles/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Survival/genetics , Drug Resistance, Neoplasm , Drug Synergism , Enzyme Activation , Enzyme Inhibitors/pharmacology , ErbB Receptors/metabolism , Female , Humans , Lapatinib , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/genetics , Neoplasm Transplantation , Protein Kinase Inhibitors/pharmacology , RNA Interference , RNA, Small Interfering , Receptor, ErbB-2/metabolism , Signal Transduction/genetics , Transplantation, Heterologous , src-Family Kinases/genetics , src-Family Kinases/metabolism
5.
J Biol Chem ; 285(31): 24141-53, 2010 Jul 30.
Article in English | MEDLINE | ID: mdl-20495008

ABSTRACT

Nerve growth factor (NGF) induces terminal differentiation in PC12, a pheochromocytoma-derived cell line. NGF binds a specific receptor on the membrane and triggers the ERK1/2 cascade, which stimulates the transcription of neural genes. We report that NGF significantly affects mitochondrial metabolism by reducing mitochondrial-produced reactive oxygen species and stabilizing the electrochemical gradient. This is accomplished by stimulation of mitochondrial manganese superoxide dismutase (MnSOD) both transcriptionally and post-transcriptionally via Ki-Ras and ERK1/2. Activation of MnSOD is essential for completion of neuronal differentiation because 1) expression of MnSOD induces the transcription of a neuronal specific promoter and neurite outgrowth, 2) silencing of endogenous MnSOD by small interfering RNA significantly reduces transcription induced by NGF, and 3) a Ki-Ras mutant in the polylysine stretch at the COOH terminus, unable to stimulate MnSOD, fails to induce complete differentiation. Overexpression of MnSOD restores differentiation in cells expressing this mutant. ERK1/2 is also downstream of MnSOD, as a SOD mimetic drug stimulates ERK1/2 with the same kinetics of NGF and silencing of MnSOD reduces NGF-induced late ERK1/2. Long term activation of ERK1/2 by NGF requires SOD activation, low levels of hydrogen peroxide, and the integrity of the microtubular cytoskeleton. Confocal immunofluorescence shows that NGF stimulates the formation of a complex containing membrane-bound Ki-Ras, microtubules, and mitochondria. We propose that active NGF receptor induces association of mitochondria with plasma membrane. Local activation of ERK1/2 by Ki-Ras stimulates mitochondrial SOD, which reduces reactive oxygen species and produces H(2)O(2). Low and spatially restricted levels of H(2)O(2) induce and maintain long term ERK1/2 activity and ultimately differentiation of PC12 cells.


Subject(s)
Genes, ras/genetics , Nerve Growth Factor/metabolism , Reactive Oxygen Species , Superoxide Dismutase/metabolism , ras Proteins/metabolism , Animals , Cell Differentiation , Cytoskeleton/metabolism , Humans , MAP Kinase Signaling System , Mitochondria/enzymology , Mitogen-Activated Protein Kinase 3/metabolism , Mutation , PC12 Cells , Rats
6.
Crit Rev Oncol Hematol ; 147: 102886, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32014673

ABSTRACT

In several tumors the PI3K/AKT/mTOR pathway is frequently disrupted, an event that results in uncontrolled cell proliferation and tumor growth. Through the years, several compounds have been developed to inhibit the pathway at different steps: the mammalian target of rapamycin (mTOR) seemed to be the most qualified target. However, this kinase has such a key role in cell survival that mechanisms of resistance are rapidly developed. Nevertheless, clinical results obtained with mTOR inhibitors in breast cancer, renal cell carcinoma, neuroendocrine tumors and mantle cell lymphoma push oncologists to actively further develop these drugs, maybe by better selecting the population to which they are offered, through the research of predictive factors of responsiveness. In this review, we aim to describe mechanisms of resistance to mTOR inhibitors, from preclinical and clinical perspectives.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Lymphoma, Mantle-Cell , Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Adult , Cell Proliferation , Drug Resistance, Neoplasm , Humans , Protein Kinase Inhibitors
7.
Sci Rep ; 8(1): 2998, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29445216

ABSTRACT

HER2 transmembrane receptor is an important target in immunotherapy treatment of breast and gastroesophageal cancer. Molecular imaging of HER2 expression may provide essential prognostic and predictive information concerning disseminated cancer and aid in selection of an optimal therapy. Radiolabeled low molecular weight peptide ligands are particularly attractive as probes for molecular imaging, since they reach and bind to the target and clear from non-target organs and blood stream faster than bulky antibodies. In this study, we evaluated a potential HER2-imaging probe, an A9 nonapeptide, derived from the trastuzumab-Fab portion. Its cellular uptake was investigated by mass spectrometry analysis of the cytoplasmic cellular extracts. Moreover, based on in-silico modeling, DTPA chelator was conjugated to N-terminus of A9. 111In-labeled A9 demonstrated nanomolar affinity to HER2-expressing BT474 cells and favorable biodistribution profile in NMRI mice. This study suggests that the peptide A9 represents a good lead candidate for development of molecular probe, to be used for imaging purposes and for the delivery of cytotoxic agents.


Subject(s)
Breast Neoplasms/diagnosis , Molecular Probes/metabolism , Peptides/metabolism , Radioimmunodetection/methods , Receptor, ErbB-2/immunology , Animals , Animals, Outbred Strains , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Delivery Systems , Female , Humans , Immunoglobulin Fab Fragments/genetics , Mice , Molecular Probes/genetics , Peptides/genetics , Protein Binding , Trastuzumab/genetics
8.
Sci Rep ; 7(1): 9388, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28839232

ABSTRACT

The urokinase-type plasminogen activator receptor (uPAR) is a GPI-anchored cell membrane receptor that focuses urokinase (uPA) proteolytic activity on the cell surface. Its expression is increased in many human cancers, including non-small cell lung cancer (NSCLC) and colorectal cancer (CRC), and correlates with a poor prognosis and early invasion and metastasis. uPAR is able to control, through a cross-talk with tyrosine kinase receptors, the shift between tumor dormancy and proliferation, that usually precedes metastasis formation. Therefore, we investigated the role of uPAR expression in RAS mutated NSCLC and CRC cells. In this study we provided evidence, for the first time, that RAS mutational condition is functionally correlated to uPAR overexpression in NSCLC and CRC cancer cell lines and patient-derived tissue samples. Moreover, oncogenic features related to uPAR overexpression in RAS mutated NSCLC and CRC, such as adhesion, migration and metastatic process may be targeted, in vitro and in vivo, by new anti-uPAR small molecules, specific inhibitors of uPAR-vitronectin interaction. Therefore, anti-uPAR drugs could represent an effective pharmacological strategy for NSCLC and CRC patients carrying RAS mutations.


Subject(s)
Gene Expression Regulation, Neoplastic , Mutation , Neoplasms/genetics , Neoplasms/pathology , Receptors, Urokinase Plasminogen Activator/genetics , ras Proteins/genetics , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Adhesion/drug effects , Cell Movement/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/metabolism , Receptors, Urokinase Plasminogen Activator/antagonists & inhibitors , Receptors, Urokinase Plasminogen Activator/metabolism , Signal Transduction , Xenograft Model Antitumor Assays , ras Proteins/metabolism
9.
Expert Opin Drug Discov ; 11(12): 1201-1212, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27700193

ABSTRACT

INTRODUCTION: Several reports have suggested that a population of undifferentiated cells known as cancer stem cells (CSCs), is responsible for cancer formation and maintenance. In the last decade, the presence of CSCs in solid cancers have been reported. Areas covered: This review summarizes the main approaches for targeting CSCs drug resistance. It is indeed known that CSCs may contribute to resistance to conventional chemotherapy, radiotherapy and targeted agents. Among the mechanisms by which CSCs escape anticancer therapies, removal of therapeutic agents by drug efflux pumps, enhanced DNA damage repair, activation of mitogenic/anti-apoptotic pathways; the main features of CSCs, stemness and EMT, are involved, as well as the capability to evade immune response. Expert opinion: Different approaches are suitable to target CSCs mediated drug resistance. Some of them are currently under clinical evaluation in different cancer types. A better understanding of CSC biology, as well as more accurate study design, may maximize the therapeutic effects of these agents. In this respect, it is important to establish: (i) which molecules should be targeted; (ii) what drug combinations may be suitable; (iii) which patient settings will CSC targeting offer the highest clinical benefit; and (iv) how to integrate therapeutic approaches targeting CSCs with standard cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Neoplastic Stem Cells/metabolism , Animals , Antineoplastic Agents/administration & dosage , Drug Resistance, Neoplasm , Humans , Molecular Targeted Therapy , Neoplasms/pathology , Research Design
10.
Oncotarget ; 7(26): 40073-40084, 2016 Jun 28.
Article in English | MEDLINE | ID: mdl-27223077

ABSTRACT

Inhibition of the mechanistic target of rapamycin (mTOR) is a promising treatment strategy for several cancer types. Rapamycin derivatives such as everolimus are allosteric mTOR inhibitors acting through interaction with the intracellular immunophilin FKBP12, a prolyl isomerase with different cellular functions. Although mTOR inhibitors have significantly improved survival of different cancer patients, resistance and lack of predictive factors of response remain unsolved issues. To elucidate the mechanisms of resistance to everolimus, we evaluated Met activation in everolimus-sensitive/resistant human cancer cells, in vitro and in vivo. Biochemical and computational analyses were performed. Everolimus-resistant cells were xenografted into mice (10/group) and studied for their response to everolimus and Met inhibitors. The statistical significance of the in vitro results was evaluated by Student's t test.Everolimus reduced Met phosphorylation in everolimus-sensitive cells. This event was mediated by the formation of a Met-FKBP12 complex, which in turn is disrupted by everolimus. Aberrant Met activation in everolimus-resistant cells and overexpression of wild-type/mutant Met caused everolimus resistance. Pharmacological inhibition and RNA silencing of Met are effective in condition of everolimus resistance (P<0.01). In mice xenografted with everolimus-resistant cells, the combination of everolimus with the Met inhibitor PHA665752 reduced tumor growth and induced a statistically significant survival advantage (combination vs control P=0.0005).FKBP12 binding is required for full Met activation and everolimus can inhibit Met. Persistent Met activation might sustain everolimus resistance. These results identify a novel everolimus mechanism of action and suggest the development of clinical strategies based on Met inhibitors in everolimus-resistant cancers.


Subject(s)
Drug Resistance, Neoplasm , Everolimus/pharmacology , Gene Expression Regulation, Neoplastic , Receptor Protein-Tyrosine Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , Allosteric Site , Animals , Cell Line, Tumor , Female , HCT116 Cells , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Phosphorylation , RNA Interference
11.
Cancer Treat Rev ; 41(10): 877-83, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26276735

ABSTRACT

Targeted therapies have been approved for various malignancies but the acquisition of resistance remains a substantial challenge in the clinical management of advanced cancers. Twenty-five per cent of breast cancers overexpress ErbB2/HER2, which confers a more aggressive phenotype and is associated with a poor prognosis. HER2-targeting therapies (trastuzumab, pertuzumab, TDM1 and lapatinib) are available, but a significant fraction of HER2-positive breast cancers eventually relapse or progress. This suggests that acquired or intrinsic resistance enables escape from HER2 inhibition. This review focuses on mechanisms of intrinsic/acquired resistance to lapatinib identified in preclinical and clinical studies. A better understanding of these mechanisms could lead to novel predictive markers of lapatinib response and to novel therapeutic strategies for breast cancer patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Quinazolines/therapeutic use , Receptor, ErbB-2/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Female , Gene Amplification , Humans , Lapatinib , Mutation , Phosphotransferases/metabolism , Protein-Tyrosine Kinases/metabolism , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism
12.
Oncotarget ; 6(28): 26090-103, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26325669

ABSTRACT

Resistance to the EGFR tyrosine kinase inhibitors (TKIs) gefitinib and erlotinib, often related to Ras or secondary EGFR mutations, is a relevant clinical issue in Non-Small Cell Lung Cancer (NSCLC). Although Src TK has been involved in such resistance, clinical development of its inhibitors has been so far limited. To better define the molecular targets of the Src TKIs saracatinib, dasatinib and bosutinib, we used a variety of in vitro/in vivo studies. Kinase assays supported by docking analysis demonstrated that all the compounds directly inhibit EGFR TK variants. However, in live cells only saracatinib efficiently reduced EGFR activation, while dasatinib was the most effective agent in inhibiting Src TK. Consistently, a pronounced anti-proliferative effect was achieved with saracatinib, in EGFR mutant cells, or with dasatinib, in wt EGFR/Ras mutant cells, poorly dependent on EGFR and erlotinib-resistant. We then identified the most effective drug combinations to overcome resistance to EGFR inhibitors, both in vitro and in nude mice: in T790M EGFR erlotinib-resistant cells, saracatinib with the anti-EGFR mAb cetuximab; in Ras mutant erlotinib-resistant models, dasatinib with the MEK inhibitor selumetinib. Src inhibitors may act with different mechanisms in NSCLCs, depending on EGFR/Ras mutational profile, and may be integrated with EGFR or MEK inhibitors for different cohorts of NSCLCs.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/metabolism , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , ras Proteins/metabolism , src-Family Kinases/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzodioxoles/administration & dosage , Benzodioxoles/pharmacology , Blotting, Western , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cetuximab/administration & dosage , Cetuximab/pharmacology , Dasatinib/administration & dosage , Dasatinib/pharmacology , ErbB Receptors/genetics , Erlotinib Hydrochloride/administration & dosage , Erlotinib Hydrochloride/pharmacology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice, Inbred BALB C , Mice, Nude , Mutation , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/metabolism , Quinazolines/administration & dosage , Quinazolines/pharmacology , RNA Interference , Tumor Burden/drug effects , Tumor Burden/genetics , Xenograft Model Antitumor Assays , ras Proteins/genetics , src-Family Kinases/genetics , src-Family Kinases/metabolism
13.
Clin Cancer Res ; 19(1): 138-47, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23166225

ABSTRACT

PURPOSE: Although the anti-EGF receptor (EGFR) monoclonal antibody cetuximab is an effective strategy in colorectal cancer therapy, its clinical use is limited by intrinsic or acquired resistance. Alterations in the "sphingolipid rheostat"-the balance between the proapoptotic molecule ceramide and the mitogenic factor sphingosine-1-phosphate (S1P)-due to sphingosine kinase 1 (SphK1) overactivation have been involved in resistance to anticancer-targeted agents. Moreover, cross-talks between SphK1 and EGFR-dependent signaling pathways have been described. EXPERIMENTAL DESIGN: We investigated SphK1 contribution to cetuximab resistance in colorectal cancer, in preclinical in vitro/in vivo models, and in tumor specimens from patients. RESULTS: SphK1 was found overexpressed and overactivated in colorectal cancer cells with intrinsic or acquired resistance to cetuximab. SphK1 contribution to resistance was supported by the demonstration that SphK1 inhibition by N,N-dimethyl-sphingosine or silencing via siRNA in resistant cells restores sensitivity to cetuximab, whereas exogenous SphK1 overexpression in sensitive cells confers resistance to these agents. Moreover, treatment of resistant cells with fingolimod (FTY720), a S1P receptor (S1PR) antagonist, resulted in resensitization to cetuximab both in vitro and in vivo, with inhibition of tumor growth, interference with signal transduction, induction of cancer cells apoptosis, and prolongation of mice survival. Finally, a correlation between SphK1 expression and cetuximab response was found in colorectal cancer patients.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Colorectal Neoplasms/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cetuximab , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Fingolimod Hydrochloride , Gene Expression , Humans , Mice , Mice, Nude , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Propylene Glycols/pharmacology , Receptors, Lysosphingolipid/antagonists & inhibitors , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Sphingosine-1-Phosphate Receptors , Treatment Outcome , Xenograft Model Antitumor Assays
14.
Microsc Res Tech ; 74(4): 347-54, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20734413

ABSTRACT

The family of B1 Sox transcription factors plays critical roles in the early stages of development, including the central nervous system. It was demonstrated that Sox2 is expressed in repressed neural stem cells. Therefore, we decided to investigate the expression of Sox2 in the brain of zebrafish at different ages to identify potential neurogenic areas, and to establish the developmental changes they undergo. The brains were assessed by qRT-PCR, western blot, and immunohistochemistry. The maximal expression of Sox2 was found at 15 dpf progressively decreases up to 30 dpf, then increases up to 40 dpf and remains unchanged up to 180 dpf. By western blot three protein bands of 28 kDa, 34 kDa (main band), and 38 kDa were detected in the brain of 180 dpf animals. The immunolocalization of Sox2 revealed that by 15 dpf Sox2 was detected in cells of the olfactory bulb, the walls of the telencephalic and diencephalic ventricles, several nucleus in the diencephalons, and the tectum opticum; by 25-50 dpf the Sox2 positive areas were the same as above, and in the rhombencephalic ventricle and cerebellum. In adult animals Sox2 was restricted to the olfactory bulb and to cells of the telencephalic ventricle walls. Taken together present results demonstrate that the potential neurogenic areas in the brain of zebrafish are widespread than in mammals and change with development, but they are primarily concentrated around the ventricles and olfactory bulb in adults, following a similar localization as in mammals.


Subject(s)
Brain Chemistry , Brain/embryology , Gene Expression Profiling , Gene Expression Regulation, Developmental , SOX Transcription Factors/biosynthesis , Zebrafish Proteins/biosynthesis , Zebrafish/embryology , Animals , Blotting, Western , Immunohistochemistry , Mammals , Molecular Weight , Real-Time Polymerase Chain Reaction , Time Factors
15.
Ann Transl Med ; 3(21): 338, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26733003
SELECTION OF CITATIONS
SEARCH DETAIL