Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Mol Syst Biol ; 15(8): e8828, 2019 08.
Article in English | MEDLINE | ID: mdl-31464372

ABSTRACT

Endothelins (EDN) are peptide hormones that activate a GPCR signalling system and contribute to several diseases, including hypertension and cancer. Current knowledge about EDN signalling is fragmentary, and no systems level understanding is available. We investigated phosphoproteomic changes caused by endothelin B receptor (ENDRB) activation in the melanoma cell lines UACC257 and A2058 and built an integrated model of EDNRB signalling from the phosphoproteomics data. More than 5,000 unique phosphopeptides were quantified. EDN induced quantitative changes in more than 800 phosphopeptides, which were all strictly dependent on EDNRB. Activated kinases were identified based on high confidence EDN target sites and validated by Western blot. The data were combined with prior knowledge to construct the first comprehensive logic model of EDN signalling. Among the kinases predicted by the signalling model, AKT, JNK, PKC and AMP could be functionally linked to EDN-induced cell migration. The model contributes to the system-level understanding of the mechanisms underlying the pleiotropic effects of EDN signalling and supports the rational selection of kinase inhibitors for combination treatments with EDN receptor antagonists.


Subject(s)
Endothelins/pharmacology , Gene Expression Regulation, Neoplastic , Melanocytes/metabolism , Phosphoproteins/genetics , Protein Processing, Post-Translational , Signal Transduction , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Endothelins/genetics , Endothelins/metabolism , Gene Regulatory Networks , Humans , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Melanocytes/drug effects , Melanocytes/pathology , Phosphoproteins/metabolism , Phosphorylation , Protein Kinase C/genetics , Protein Kinase C/metabolism , Proteomics/methods , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Endothelin B/genetics , Receptor, Endothelin B/metabolism
2.
EMBO J ; 30(21): 4489-99, 2011 Sep 06.
Article in English | MEDLINE | ID: mdl-21897365

ABSTRACT

Acquiring resistance against transforming growth factor ß (TGFß)-induced growth inhibition at early stages of carcinogenesis and shifting to TGFß's tumour-promoting functions at later stages is a pre-requisite for malignant tumour progression and metastasis. We have identified the transcription factor distal-less homeobox 2 (Dlx2) to exert critical functions during this switch. Dlx2 counteracts TGFß-induced cell-cycle arrest and apoptosis in mammary epithelial cells by at least two molecular mechanisms: Dlx2 acts as a direct transcriptional repressor of TGFß receptor II (TGFßRII) gene expression and reduces canonical, Smad-dependent TGFß signalling and expression of the cell-cycle inhibitor p21(CIP1) and increases expression of the mitogenic transcription factor c-Myc. On the other hand, Dlx2 directly induces the expression of the epidermal growth factor (EGF) family member betacellulin, which promotes cell survival by stimulating EGF receptor signalling. Finally, Dlx2 expression supports experimental tumour growth and metastasis of B16 melanoma cells and correlates with tumour malignancy in a variety of human cancer types. These results establish Dlx2 as one critical player in shifting TGFß from its tumour suppressive to its tumour-promoting functions.


Subject(s)
Apoptosis/genetics , Cell Cycle Checkpoints/genetics , Homeodomain Proteins/physiology , Transcription Factors/physiology , Transforming Growth Factor beta/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Female , Genes, Tumor Suppressor/physiology , HEK293 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology
3.
Cancer Cell ; 9(4): 261-72, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16616332

ABSTRACT

The expression of podoplanin, a small mucin-like protein, is upregulated in the invasive front of a number of human carcinomas. We have investigated podoplanin function in cultured human breast cancer cells, in a mouse model of pancreatic beta cell carcinogenesis, and in human cancer biopsies. Our results indicate that podoplanin promotes tumor cell invasion in vitro and in vivo. Notably, the expression and subcellular localization of epithelial markers are unaltered, and mesenchymal markers are not induced in invasive podoplanin-expressing tumor cells. Rather, podoplanin induces collective cell migration by filopodia formation via the downregulation of the activities of small Rho family GTPases. In conclusion, podoplanin induces an alternative pathway of tumor cell invasion in the absence of epithelial-mesenchymal transition (EMT).


Subject(s)
Actins/metabolism , Cytoskeleton/metabolism , Epithelial Cells/pathology , Membrane Glycoproteins/metabolism , Mesoderm/pathology , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , DNA-Binding Proteins/metabolism , Disease Progression , Down-Regulation , Epithelial Cells/metabolism , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Leukemia, B-Cell/metabolism , Leukemia, B-Cell/pathology , Mesoderm/metabolism , Mice , Mice, Transgenic , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Organic Cation Transport Proteins/metabolism , Pseudopodia/metabolism , Transcription Factors/metabolism , cdc42 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
4.
Semin Cancer Biol ; 22(3): 261-71, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22484560

ABSTRACT

Disseminated tumor cells are present in many patients at diagnosis. At a time when the disseminated disease becomes prominent, patients have already been treated with many cycles of therapy to which their metastases were also exposed. These metastases have genetically evolved from primary tumors. Furthermore, their interaction with the tissue microenvironment plays an important role in all phases of disease development. These facts have only partially been taken into consideration when profiling anti-cancer compounds foreseen to treat patients with disseminated metastatic disease. In this perspective, we discuss the unique features of metastatic disease and review the model systems available for drug profiling. Based on an analysis of how compounds are profiled today in pre-clinical models of metastatic disease and what would be desirable and possible with the present know-how, we recommend a refined profiling process to validate drugs with potential to treat patients with overt metastatic disease.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Drug Resistance , Neoplasm Metastasis/drug therapy , Tumor Microenvironment/drug effects , Antineoplastic Agents/therapeutic use , Humans , Models, Biological
5.
ChemMedChem ; 19(2): e202300606, 2024 01 15.
Article in English | MEDLINE | ID: mdl-37983645

ABSTRACT

Prostaglandin E2 (PGE2) plays a key role in various stages of cancer. PGE2 signals through the EP2 and the EP4 receptors, promoting tumorigenesis, metastasis, and/or immune suppression. Dual inhibition of both the EP2 and the EP4 receptors has the potential to counteract the effect of PGE2 and to result in antitumor efficacy. We herein disclose for the first time the structure of dual EP2/EP4 antagonists. By merging the scaffolds of EP2 selective and EP4 selective inhibitors, we generated a new chemical series of compounds blocking both receptors with comparable potency. In vitro and in vivo profiling suggests that the newly identified compounds are promising lead structures for further development into dual EP2/EP4 antagonists for use in cancer therapy.


Subject(s)
Dinoprostone , Neoplasms , Humans , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP4 Subtype
6.
EMBO J ; 27(19): 2603-15, 2008 Oct 08.
Article in English | MEDLINE | ID: mdl-18772882

ABSTRACT

Loss of expression of the cell-cell adhesion molecule E-cadherin is a hallmark of epithelial-mesenchymal transition (EMT) in development and in the progression from epithelial tumours to invasive and metastatic cancers. Here, we demonstrate that the loss of E-cadherin function upregulates expression of the neuronal cell adhesion molecule (NCAM). Subsequently, a subset of NCAM translocates from fibroblast growth factor receptor (FGFR) complexes outside lipid rafts into lipid rafts where it stimulates the non-receptor tyrosine kinase p59(Fyn) leading to the phosphorylation and activation of focal adhesion kinase and the assembly of integrin-mediated focal adhesions. Ablation of NCAM expression during EMT inhibits focal adhesion assembly, cell spreading and EMT. Conversely, forced expression of NCAM induces epithelial cell delamination and migration, and high NCAM expression correlates with tumour invasion. These results establish a mechanistic link between the loss of E-cadherin expression, NCAM function, focal adhesion assembly and cell migration and invasion.


Subject(s)
Cadherins/metabolism , Focal Adhesions/metabolism , Neural Cell Adhesion Molecules/metabolism , Animals , Cadherins/genetics , Cell Line , Cell Movement/physiology , Epithelium/physiology , Gene Expression Regulation , Humans , Membrane Microdomains/metabolism , Mesoderm/physiology , Mice , Mice, Knockout , Neoplasm Invasiveness , Neoplasms/metabolism , Neoplasms/pathology , Neural Cell Adhesion Molecules/genetics , Neurons/cytology , Neurons/metabolism , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/metabolism , Receptors, Fibroblast Growth Factor/metabolism
7.
Front Pharmacol ; 12: 748740, 2021.
Article in English | MEDLINE | ID: mdl-34803691

ABSTRACT

Loss of control in the trafficking of immune cells to the inflamed lung tissue contributes to the pathogenesis of life-threatening acute lung injury (ALI) and its more severe form, acute respiratory distress syndrome (ARDS). Targeting CXCR7 has been proposed as a potential therapeutic approach to reduce pulmonary inflammation; however, its role and its crosstalk with the two chemokine receptors CXCR3 and CXCR4 via their shared ligands CXCL11 and CXCL12 is not yet completely understood. The present paper aimed to characterize the pathological role of the CXCR3/CXCR4/CXCR7 axis in a murine model of ALI. Lipopolysaccharide (LPS) inhalation in mice resulted in the development of key pathologic features of ALI/ARDS, including breathing dysfunctions, alteration in the alveolar capillary barrier, and lung inflammation. LPS inhalation induced immune cell infiltration into the bronchoalveolar space, including CXCR3+ and CXCR4+ cells, and enhanced the expression of the ligands of these two chemokine receptors. The first-in-class CXCR7 antagonist, ACT-1004-1239, increased levels of CXCL11 and CXCL12 in the plasma without affecting their levels in inflamed lung tissue, and consequently reduced CXCR3+ and CXCR4+ immune cell infiltrates into the bronchoalveolar space. In the early phase of lung inflammation, characterized by a massive influx of neutrophils, treatment with ACT-1004-1239 significantly reduced the LPS-induced breathing pattern alteration. Both preventive and therapeutic treatment with ACT-1004-1239 reduced lung vascular permeability and decreased inflammatory cell infiltrates. In conclusion, these results demonstrate a key pathological role of CXCR7 in ALI/ARDS and highlight the clinical potential of ACT-1004-1239 in ALI/ARDS pathogenesis.

8.
Oncogene ; 40(9): 1659-1673, 2021 03.
Article in English | MEDLINE | ID: mdl-33500549

ABSTRACT

The clinical benefit of MAPK pathway inhibition in melanoma patients carrying BRAF mutations is temporal. After the initial response to treatment, the majority of tumors will develop resistance and patients will relapse. Here we demonstrate that the endothelin-endothelin receptor B (ETBR) signaling pathway confers resistance to MAPK pathway inhibitors in BRAF mutated melanoma. MAPK blockade, in addition to being anti-proliferative, induces a phenotypic change which is characterized by increased expression of melanocyte-specific genes including ETBR. In the presence of MAPK inhibitors, activation of ETBR by endothelin enables the sustained proliferation of melanoma cells. In mouse models of melanoma, including patient-derived xenograft models, concurrent inhibition of the MAPK pathway and ETBR signaling resulted in a more effective anti-tumor response compared to MAPK pathway inhibition alone. The combination treatment significantly reduced tumor growth and prolonged survival compared to therapies with MAPK pathway inhibitors alone. The phosphoproteomic analysis revealed that ETBR signaling did not induce resistance towards MAPK pathway inhibitors by restoring MAPK activity, but instead via multiple alternative signaling pathways downstream of the small G proteins GNAq/11. Together these data indicate that a combination of MAPK pathway inhibitors with ETBR antagonists could have a synergistically beneficial effect in melanoma patients with hyperactivated MAPK signaling pathways.


Subject(s)
Melanoma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Proto-Oncogene Proteins B-raf/genetics , Receptor, Endothelin B/genetics , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Endothelin B Receptor Antagonists/pharmacology , Humans , Melanoma/genetics , Melanoma/pathology , Mice , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mutation/genetics , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
9.
Dev Cell ; 9(6): 769-79, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16326389

ABSTRACT

Covalent modification by SUMO regulates a wide range of cellular processes, including transcription, cell cycle, and chromatin dynamics. To address the biological function of the SUMO pathway in mammals, we generated mice deficient for the SUMO E2-conjugating enzyme Ubc9. Ubc9-deficient embryos die at the early postimplantation stage. In culture, Ubc9 mutant blastocysts are viable, but fail to expand after 2 days and show apoptosis of the inner cell mass. Loss of Ubc9 leads to major chromosome condensation and segregation defects. Ubc9-deficient cells also show severe defects in nuclear organization, including nuclear envelope dysmorphy and disruption of nucleoli and PML nuclear bodies. Moreover, RanGAP1 fails to accumulate at the nuclear pore complex in mutant cells that show a collapse in Ran distribution. Together, these findings reveal a major role for Ubc9, and, by implication, for the SUMO pathway, in nuclear architecture and function, chromosome segregation, and embryonic viability in mammals.


Subject(s)
Cell Nucleus/metabolism , Chromosome Segregation , Embryo, Mammalian/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitin-Conjugating Enzymes/physiology , Animals , Apoptosis , Blastocyst/cytology , Blastocyst/metabolism , Cell Nucleus/genetics , Embryo Loss/genetics , Embryo, Mammalian/cytology , Female , Fluorescent Antibody Technique , GTPase-Activating Proteins/metabolism , Immunoblotting , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitosis , Signal Transduction , Small Ubiquitin-Related Modifier Proteins/genetics , Ubiquitins/metabolism , ran GTP-Binding Protein/metabolism
10.
J Med Chem ; 63(24): 15864-15882, 2020 12 24.
Article in English | MEDLINE | ID: mdl-33314938

ABSTRACT

The chemokine receptor CXCR7, also known as ACKR3, is a seven-transmembrane G-protein-coupled receptor (GPCR) involved in various pathologies such as neurological diseases, autoimmune diseases, and cancers. By binding and scavenging the chemokines CXCL11 and CXCL12, CXCR7 regulates their extracellular levels. From an original high-throughput screening campaign emerged hit 3 among others. The hit-to-lead optimization led to the discovery of a novel chemotype series exemplified by the trans racemic compound 11i. This series provided CXCR7 antagonists that block CXCL11- and CXCL12-induced ß-arrestin recruitment. Further structural modifications on the trisubstituted piperidine scaffold of 11i yielded compounds with high CXCR7 antagonistic activities and balanced ADMET properties. The effort described herein culminated in the discovery of ACT-1004-1239 (28f). Biological characterization of ACT-1004-1239 demonstrated that it is a potent, insurmountable antagonist. Oral administration of ACT-1004-1239 in mice up to 100 mg/kg led to a dose-dependent increase of plasma CXCL12 concentration.


Subject(s)
Piperidines/chemistry , Receptors, CXCR/antagonists & inhibitors , Administration, Oral , Amides/chemistry , Amines/chemistry , Animals , Chemokine CXCL12/blood , Crystallography, X-Ray , Dogs , Drug Evaluation, Preclinical , Half-Life , Humans , Inhibitory Concentration 50 , Mice , Molecular Conformation , Piperidines/metabolism , Piperidines/pharmacokinetics , Protein Binding , Rats , Receptors, CXCR/genetics , Receptors, CXCR/metabolism , Structure-Activity Relationship
11.
Trends Mol Med ; 13(12): 535-41, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17981506

ABSTRACT

Most cancer deaths are caused by metastasis rather than the primary tumor. Cancer cells invade normal tissue as epithelial sheets or single cells by inducing expression of programs characteristic of developmental processes. Depending on their tissue of origin, cancer cells subsequently spread to distinct target organs where they seed secondary tumors (metastasis). Recent experimental evidence suggests that metastasis requires changes not only in cancer cells but also in the tumor microenvironment and in the metastatic target site. For example, a premetastatic niche is formed in target organs that attract cancer cells. Understanding the distinct mechanisms used by cancer cells to form metastasis will enable better patient evaluation and the design of innovative therapeutic approaches.


Subject(s)
Neoplasm Invasiveness , Neoplasm Metastasis , Humans
12.
Neuro Oncol ; 18(4): 486-96, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26995790

ABSTRACT

BACKGROUND: We recently demonstrated that brain endothelial cells and astrocytes protect cancer cells from chemotherapy through an endothelin-dependent signaling mechanism. Here, we evaluated the efficacy of macitentan, a dual endothelin receptor (ETAR and ETBR) antagonist, in the treatment of experimental breast and lung cancer brain metastases. METHODS: The effect of macitentan on astrocyte- and brain endothelial cell-mediated chemoprotective properties was measured in cytotoxic assays. We compared survival of mice bearing established MDA-MB-231 breast cancer or PC-14 non-small cell lung cancer (NSCLC) brain metastases that were treated with vehicle, macitentan, paclitaxel, or macitentan plus paclitaxel. Cell division, apoptosis, tumor vasculature, and expression of survival-related proteins were assessed by immunofluorescent microscopy. RESULTS: Cancer cells and tumor-associated endothelial cells expressed activated forms of AKT and MAPK in vehicle- and paclitaxel-treated groups in both metastasis models, but these proteins were downregulated in metastases of mice that received macitentan. The survival-related proteins Bcl2L1, Gsta5, and Twist1 that localized to cancer cells and tumor-associated endothelial cells in vehicle- and paclitaxel-treated tumors were suppressed by macitentan. Macitentan or paclitaxel alone had no effect on survival. However, when macitentan was combined with paclitaxel, we noted a significant reduction in cancer cell division and marked apoptosis of both cancer cells and tumor-associated endothelial cells. Moreover, macitentan plus paclitaxel therapy significantly increased overall survival by producing complete responses in 35 of 35 mice harboring brain metastases. CONCLUSIONS: Dual antagonism of ETAR and ETBR signaling sensitizes experimental brain metastases to paclitaxel and may represent a new therapeutic option for patients with brain metastases.


Subject(s)
Breast Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Paclitaxel/pharmacology , Pyrimidines/pharmacology , Receptors, Endothelin/chemistry , Sulfonamides/pharmacology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Drug Therapy, Combination , Female , Humans , Immunoenzyme Techniques , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Mice, Nude , NIH 3T3 Cells , Receptors, Endothelin/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Oncotarget ; 7(18): 25983-6002, 2016 May 03.
Article in English | MEDLINE | ID: mdl-27036020

ABSTRACT

An epithelial to mesenchymal transition (EMT) enables epithelial tumor cells to break out of the primary tumor mass and to metastasize. Understanding the molecular mechanisms driving EMT in more detail will provide important tools to interfere with the metastatic process. To identify pharmacological modulators and druggable targets of EMT, we have established a novel multi-parameter, high-content, microscopy-based assay and screened chemical compounds with activities against known targets. Out of 3423 compounds, we have identified 19 drugs that block transforming growth factor beta (TGFß)-induced EMT in normal murine mammary gland epithelial cells (NMuMG). The active compounds include inhibitors against TGFß receptors (TGFBR), Rho-associated protein kinases (ROCK), myosin II, SRC kinase and uridine analogues. Among the EMT-repressing compounds, we identified a group of inhibitors targeting multiple receptor tyrosine kinases, and biochemical profiling of these multi-kinase inhibitors reveals TGFBR as a thus far unknown target of their inhibitory spectrum. These findings demonstrate the feasibility of a multi-parameter, high-content microscopy screen to identify modulators and druggable targets of EMT. Moreover, the newly discovered "off-target" effects of several receptor tyrosine kinase inhibitors have important consequences for in vitro and in vivo studies and might beneficially contribute to the therapeutic effects observed in vivo.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , High-Throughput Screening Assays/methods , Mammary Neoplasms, Animal/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Animals , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Cell Proliferation/drug effects , Female , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Mice , Receptor Protein-Tyrosine Kinases , Receptor, Transforming Growth Factor-beta Type II , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured
14.
Clin Cancer Res ; 21(20): 4630-41, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26106074

ABSTRACT

PURPOSE: The objective of the study was to determine whether astrocytes and brain endothelial cells protect glioma cells from temozolomide through an endothelin-dependent signaling mechanism and to examine the therapeutic efficacy of the dual endothelin receptor antagonist, macitentan, in orthotopic models of human glioblastoma. EXPERIMENTAL DESIGN: We evaluated several endothelin receptor antagonists for their ability to inhibit astrocyte- and brain endothelial cell-induced protection of glioma cells from temozolomide in chemoprotection assays. We compared survival in nude mice bearing orthotopically implanted LN-229 glioblastomas or temozolomide-resistant (LN-229(Res) and D54(Res)) glioblastomas that were treated with macitentan, temozolomide, or both. Tumor burden was monitored weekly with bioluminescence imaging. The effect of therapy on cell division, apoptosis, tumor-associated vasculature, and pathways associated with cell survival was assessed by immunofluorescent microscopy. RESULTS: Only dual endothelin receptor antagonism abolished astrocyte- and brain endothelial cell-mediated protection of glioma cells from temozolomide. In five independent survival studies, including temozolomide-resistant glioblastomas, 46 of 48 (96%) mice treated with macitentan plus temozolomide had no evidence of disease (P < 0.0001), whereas all mice in other groups died. In another analysis, macitentan plus temozolomide therapy was stopped in 16 mice after other groups had died. Only 3 of 16 mice eventually developed recurrent disease, 2 of which responded to additional cycles of macitentan plus temozolomide. Macitentan downregulated proteins associated with cell division and survival in glioma cells and associated endothelial cells, which enhanced their sensitivity to temozolomide. CONCLUSIONS: Macitentan plus temozolomide are well tolerated, produce durable responses, and warrant clinical evaluation in glioblastoma patients.


Subject(s)
Dacarbazine/analogs & derivatives , Endothelin Receptor Antagonists/pharmacology , Glioblastoma/drug therapy , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Animals , Cell Division/drug effects , Cell Line , Cell Line, Tumor , Dacarbazine/pharmacology , Down-Regulation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Glioblastoma/pathology , Glioma/drug therapy , Glioma/pathology , Humans , Mice , Mice, Nude , NIH 3T3 Cells , Temozolomide
15.
Transl Oncol ; 5(1): 39-47, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22348175

ABSTRACT

Endothelin receptors (ETRs) are often overexpressed in ovarian tumors, which can be resistant to conventional therapies. Thus, we investigated whether blockage of the ETR pathways using the dual ETR antagonist macitentan combined with taxol or cisplatinum can produce therapy for orthotopically growing multidrug-resistant (MDR) human ovarian carcinoma. In several studies, nude mice were injected in the peritoneal cavity with HeyA8-MDR human ovarian cancer cells. Ten days later, mice were randomized to receive vehicle (saline), macitentan (oral, daily), taxol (intraperitoneal, weekly), cisplatinum (intraperitoneal, weekly), macitentan plus taxol, or macitentan plus cisplatinum. Moribund mice were killed, and tumors were collected, weighed, and prepared for immunohistochemical analysis. The HeyA8-MDR tumors did not respond to taxol, cisplatinum, or macitentan administered as single agents. In contrast, combination therapy with macitentan and taxol or macitentan and cisplatinum significantly decreased the tumor incidence and weight and significantly increased the survival of mice and their general condition. Multiple immunohistochemical analyses revealed that treatment with macitentan and macitentan plus taxol or cisplatinum inhibited the phosphorylation of ETRs, decreased the levels of pVEGFR2, pAkt, and pMAPK in tumor cells after 2 weeks of treatment and induced a first wave of apoptosis in tumor-associated endothelial cells followed by apoptosis in surrounding tumor cells. Our study shows that ovarian cancer cells, which express the endothelin axis and are multidrug resistant, are exquisitely sensitive to treatment with a dual ET antagonist and can be resensitized to both taxol and cisplatinum. This combined therapy led to a significant reduction in tumor weight.

16.
J Biomol Screen ; 16(8): 805-17, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21807962

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and most aggressive type of primary brain tumor. Identification of new therapeutic regimens is urgently needed. A major challenge remains the development of a relevant in vitro model system with the necessary capacity and flexibility to profile compounds. The authors have developed and characterized a 3D culture system of brain cells (brain Hi-Spot) where GBM-derived cells can be incorporated (GBM/brain Hi-Spot). Immuno-fluorescence and electrophysiological recordings demonstrate that brain Hi-Spots recapitulate many features of brain tissue. Within this tissue, GBM-derived cell growth is monitored using a fluorescence assay. GBM-derived cells growing in Hi-Spots form tumor nodules that display properties of GBM such as 5-Ala positive staining, an acidic environment, and tumor-surrounding astrocyte activation. Temozolomide inhibits GBM growth in brain Hi-Spots, but it is not effective in 2D cultures. Other chemotherapeutics that have proven to be inefficient in GBM treatment display low activity against GBM-derived cells growing in brain Hi-Spots in comparison to their activity against GBM 2D cultures. These findings suggest that GBM/brain Hi-Spots represent a simple system to culture cells derived from brain tumors in an orthotopic environment in vitro and that the system is reliable to test GBM targeting compounds.


Subject(s)
Brain Neoplasms/drug therapy , Brain/drug effects , Cell Culture Techniques , Drug Screening Assays, Antitumor , Glioblastoma/drug therapy , Aminolevulinic Acid/analysis , Animals , Antineoplastic Agents/pharmacology , Astrocytes/cytology , Brain/pathology , Brain Neoplasms/pathology , Cell Proliferation , Diffusion Chambers, Culture , Electrophysiology , Fluorescent Antibody Technique , Glioblastoma/pathology , Histocytochemistry , Rats , Rats, Wistar , Tumor Cells, Cultured
17.
Neoplasia ; 13(2): 167-79, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21403842

ABSTRACT

Potential treatments for ovarian cancers that have become resistant to standard chemotherapies include modulators of tumor cell survival, such as endothelin receptor (ETR) antagonist. We investigated the therapeutic efficacy of the dual ETR antagonist, macitentan, on human ovarian cancer cells, SKOV3ip1 and IGROV1, growing orthotopically in nude mice. Mice with established disease were treated with vehicle (control), paclitaxel (weekly, intraperitoneal injections), macitentan (daily oral administrations), or a combination of paclitaxel and macitentan. Treatment with paclitaxel decreased tumor weight and volume of ascites. Combination therapy with macitentan and paclitaxel reduced tumor incidence and further reduced tumor weight and volume of ascites when compared with paclitaxel alone. Macitentan alone occasionally reduced tumor weight but alone had no effect on tumor incidence or ascites. Immunohistochemical analyses revealed that treatment with macitentan and macitentan plus paclitaxel inhibited the phosphorylation of ETRs and suppressed the survival pathways of tumor cells by decreasing the levels of pVEGFR2, pAkt, and pMAPK. The dose of macitentan necessary for inhibition of phosphorylation correlated with the dose required to increase antitumor efficacy of paclitaxel. Treatment with macitentan enhanced the cytotoxicity mediated by paclitaxel as measured by the degree of apoptosis in tumor cells and tumor-associated endothelial cells. Collectively, these results show that administration of macitentan in combination with paclitaxel prevents the progression of ovarian cancer in the peritoneal cavity of nude mice in part by inhibiting survival pathways of both tumor cells and tumor-associated endothelial cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Endothelin Receptor Antagonists , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Paclitaxel/therapeutic use , Pyrimidines/therapeutic use , Sulfonamides/therapeutic use , Animals , Female , Humans , Mice , Mice, Nude , Peritoneal Neoplasms/prevention & control , Peritoneal Neoplasms/secondary , Pyrimidines/administration & dosage , Sulfonamides/administration & dosage , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
EMBO J ; 26(12): 2832-42, 2007 Jun 20.
Article in English | MEDLINE | ID: mdl-17541405

ABSTRACT

Integrins are transmembrane receptors that bind extracellular matrix proteins and enable cell adhesion and cytoskeletal organization, as well as transduction of signals into cells, to promote various aspects of cellular behavior, such as proliferation or survival. Integrins participate in many aspects of tumor biology. Here, we have employed the Rip1Tag2 transgenic mouse model of pancreatic beta cell carcinogenesis to investigate the role of beta(1)-integrin in tumor progression. Specific ablation of beta(1)-integrin function in pancreatic beta cells resulted in a defect in sorting between insulin-expressing beta cells and glucagon-expressing alpha cells in islets of Langerhans. Ablation of beta(1)-integrin in beta tumor cells of Rip1Tag2 mice led to the dissemination of tumor cell emboli into lymphatic blood vessels in the absence of ongoing lymphangiogenesis. Yet, disseminating beta(1)-integrin-deficient beta tumor cells did not elicit metastasis. Rather, primary tumor growth was significantly impaired by reduced tumor cell proliferation and the acquisition of cellular senescence by beta(1)-integrin-deficient beta tumor cells. The results indicate a critical role of beta(1)-integrin function in mediating metastatic dissemination and preventing tumor cell senescence.


Subject(s)
Cellular Senescence , Integrin beta1/physiology , Neoplasm Metastasis , Pancreatic Neoplasms/pathology , Animals , Cell Separation , Flow Cytometry , Integrin beta1/metabolism , Islets of Langerhans/pathology , Mice , Mice, Transgenic , Pancreatic Neoplasms/physiopathology , Signal Transduction
19.
EMBO J ; 25(15): 3534-45, 2006 Aug 09.
Article in English | MEDLINE | ID: mdl-16858414

ABSTRACT

Hepatocyte growth factor/scatter factor (HGF) exerts several functions in physiological and pathological processes, among them the induction of epithelial cell scattering and motility. Its pivotal role in angiogenesis, tumor progression, and metastasis is evident; however, the underlying molecular mechanisms are still poorly understood. Here, we demonstrate that HGF induces scattering of epithelial cells by upregulating the expression of Snail, a transcriptional repressor involved in epithelial-mesenchymal transition (EMT). Snail is required for HGF-induced cell scattering, since shRNA-mediated ablation of Snail expression prevents this process. HGF-induced upregulation of Snail transcription involves activation of the mitogen-activated protein kinase (MAPK) pathway and requires the activity of early growth response factor-1 (Egr-1). Upon induction by Egr-1, Snail represses the expression of E-cadherin and claudin-3 genes. It also binds to the Egr-1 promoter and represses Egr-1 transcription, thereby establishing a negative regulatory feedback loop. These findings indicate that Snail upregulation by HGF is mediated via the MAPK/Egr-1 signaling pathway and that both Snail and Egr-1 play a critical role in HGF-induced cell scattering, migration, and invasion.


Subject(s)
Early Growth Response Protein 1/metabolism , Epithelial Cells/physiology , Hepatocyte Growth Factor/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction , Transcription Factors/genetics , Up-Regulation , Base Sequence , Cadherins/genetics , Cadherins/metabolism , Cell Movement , Cells, Cultured , Early Growth Response Protein 1/genetics , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Kinetics , Models, Biological , Molecular Sequence Data , Signal Transduction/genetics , Snail Family Transcription Factors , Transcription Factors/metabolism , Transfection , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL