Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Lett ; 414: 88-98, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29111348

ABSTRACT

Neuropilin-1 (NRP-1) is an extra-cellular receptor for the main Vascular Endothelial Growth Factor over-expressed in tumour tissues, VEGF-A165. Consequently, NRP-1 is involved in angiogenesis and in tumour growth, and its over-expression is related to a clinical poor prognosis. NRP-1 appears as a major target in oncology, which remains poorly exploited. Herein, we report a new series of 18 small-sized fully organic VEGF-A165/NRP-1 antagonists (NRPas). These compounds share an original scaffold, including two linkers (sulphonamide and amide) and three aromatic cores. Among them, 2a (renamed NRPa-308) emerges as a promising "hit". In vitro,2a exerts not only potent anti-angiogenic activity, but also significant effects on cell viability of large panel of human solid and haematological cancer cell lines. Importantly, 2a is less cytotoxic on healthy tissues than the marketed anti-angiogenic drug sunitinib. Lastly, in a mouse xenograft model (human MDA-MB-231 breast cancer cells), 2a improves the median survival and reduces the tumour growth, but does not exert visible acute toxicity. Altogether, these results highlight its huge potential for a further "hit-to-lead" optimization, leading to new anti-cancer drugs.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cell Proliferation/drug effects , Neoplasms/drug therapy , Neuropilin-1/antagonists & inhibitors , Xenograft Model Antitumor Assays , Angiogenesis Inhibitors/chemistry , Animals , Cell Line, Tumor , Cells, Cultured , Humans , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Molecular Structure , Neoplasms/metabolism , Neoplasms/pathology , Neuropilin-1/metabolism , Survival Analysis , Tumor Burden/drug effects
2.
Bioorg Med Chem Lett ; 24(17): 4254-9, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25091928

ABSTRACT

Neuropilins (NRPs) are VEGF-A165 co-receptors over-expressed in tumor cells, and considered as targets in angiogenic-related pathologies. We previously identified compound 1, the first non-peptidic antagonist of the VEGF-A165/NRP binding, which exhibits in vivo anti-angiogenic and anti-tumor activities. We report here the synthesis and biological evaluations of new antagonists structurally-related to compound 1. Among these molecules, 4a, 4c and 4d show cytotoxic effects on HUVEC and MDA-MB-31 cells, and antagonize VEGF-A165/NRP-1 binding. This study confirmed our key structure-activity relationships hypothesis and paved the way to compound 1 'hit to lead' optimization.


Subject(s)
Neuropilin-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Models, Molecular , Molecular Structure , Neuropilin-1/metabolism , Structure-Activity Relationship , Vascular Endothelial Growth Factor Receptor-1/metabolism
3.
Cancer Lett ; 349(2): 120-7, 2014 Jul 28.
Article in English | MEDLINE | ID: mdl-24752068

ABSTRACT

Neuropilin-1/-2 (+33 NRPs), VEGF-A165 co-receptors, are over-expressed during cancer progression. Thus, NRPs targeted drug development is challenged using a multistep in silico/in vitro screening procedure. The first fully non-peptidic VEGF-A165/NRPs protein-protein interaction antagonist (IC50=34 µM) without effect on pro-angiogenic kinases has been identified (compound-1). This hit showed breast cancer cells anti-proliferative activity (IC50=0.60 µM). Compound-1 treated NOG-xenografted mice significantly exerted tumor growth inhibition, which is correlated with Ki-67(low) expression and apoptosis. Furthermore, CD31(+)/CD34(+) vessels are reduced in accordance with HUVEC-tube formation inhibition (IC50=0.20 µM). Taking together, compound-1 is the first fully organic inhibitor targeting NRPs.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Neuropilins/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Drug Evaluation, Preclinical , Female , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Ligands , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Models, Molecular , Molecular Docking Simulation , Neuropilins/chemistry , Neuropilins/metabolism , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
4.
Cell Signal ; 24(1): 214-23, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21939755

ABSTRACT

Recently, we identified a new Vascular Endothelial Growth Factor (VEGF)-A(165)-induced gene Phactr-1, (Phosphatase Actin Regulator-1). We reported that Phactr-1 gene silencing inhibited tube formation in human umbilical endothelial cells (HUVECs) indicating a key role for Phactr-1 in tubulogenesis in vitro. In this study, we investigated the role of Phactr-1 in several cellular processes related to angiogenesis. We found that neuropilin-1 (NRP-1) and VEGF-R1 depletion inhibited Phactr-1 mRNA expression while NRP-2 and VEGF-R2 depletion had no effect. We described a new interaction site of VEGF-A(165) to VEGF-R1 in peptides encoded by exons 7 and 8 of VEGF-A(165). The specific inhibition of VEGF-A(165) binding on NRP-1 and VEGF-R1 by ERTCRC and CDKPRR peptides decreased the Phactr-1 mRNA levels in HUVECs indicating that VEGF-A(165)-dependent regulation of Phactr-1 expression required both NRP-1 and VEGF-R1 receptors. In addition, upon VEGFA(165)-stimulation Phactr-1 promotes formation and maintenance of cellular tubes through NRP-1 and VEGFR1. Phactr-1 was previously identified as protein phosphatase 1 (PP1) α-interacting protein that possesses actin-binding domains. We showed that Phactr-1 depletion decreased PP1 activity, disrupted the fine-tuning of actin polymerization and impaired lamellipodial dynamics. Taken together our results strongly suggest that Phactr-1 is a key component in the angiogenic process.


Subject(s)
Endothelial Cells/physiology , Microfilament Proteins/genetics , Microtubules/metabolism , Neuropilin-1/metabolism , Pseudopodia/metabolism , Vascular Endothelial Growth Factor A/physiology , Amino Acid Sequence , Binding, Competitive , Cell Culture Techniques , Cell Movement , Cells, Cultured , Endothelial Cells/metabolism , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Microfilament Proteins/metabolism , Neovascularization, Pathologic/metabolism , Neuropilin-1/genetics , Peptide Fragments/chemistry , Protein Binding , RNA Interference , Time-Lapse Imaging , Transcription, Genetic , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-1/chemistry , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
5.
Blood ; 112(6): 2575-8, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18617637

ABSTRACT

Mechanisms of protection against autoimmune diseases by transplantation of autologous hematopoietic progenitors remain poorly defined. We recently demonstrated that, unlike medullary hematopoietic stem cells (HSCs), mobilized hematopoietic progenitors (HPCs) stimulate peripheral Foxp3(+) regulatory T cell (Treg)-expansion through cell-contact activation of Notch signaling and through as yet undetermined soluble factor(s), distinct from TGF-beta1. Herein we identified one such soluble factor as granulocyte macrophage-colony stimulating factor (GM-CSF), which is produced at higher levels by HPCs than HSCs and whose neutralization significantly reduces the growth-promoting effect of HPCs on Treg. Treg express a functional GM-CSF receptor alpha-chain CD116 and proliferate in response to this cytokine independently from IL2. GM-CSF-expanded Treg-like HPC-expanded Treg-display enhanced suppressive capacity relative to control Treg. Hence, mobilized progenitors stimulate Treg expansion both by cell-contact dependent mechanisms and by their production of GM-CSF.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Hematopoietic Stem Cells/immunology , Immune Tolerance , Animals , Cell Communication , Cell Proliferation , Granulocyte-Macrophage Colony-Stimulating Factor/analysis , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Mice , Mice, Inbred NOD , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor , T-Lymphocytes, Regulatory/cytology
6.
Proc Natl Acad Sci U S A ; 104(15): 6335-40, 2007 Apr 10.
Article in English | MEDLINE | ID: mdl-17389382

ABSTRACT

Previous results have shown that CD4(+)CD25(+) regulatory T cells (Tregs) control autoimmunity in a spontaneous model of type 1 diabetes, the nonobese diabetic (NOD) mouse. Moreover, anti-CD3 reverses diabetes in this setting by promoting Tregs that function in a TGF-beta-dependent manner. This finding contrasts with a large body of work suggesting that CD4(+)CD25(high) Tregs act in a cytokine-independent manner, thus suggesting that another type of Treg is operational in this setting. We sought to determine the basis of suppression both in untreated NOD mice and in those treated with anti-CD3. Our present results show that a subset of foxP3(+) cells present within a CD4(+)CD25(low) lymphocyte subset suppresses T cell immunity in spontaneously diabetic NOD mice in a TGF-beta-dependent manner, a functional property typical of "adaptive" regulatory T cells. This distinct Treg subset is evident in NOD, but not normal, mice, suggesting that the NOD mice may generate these adaptive Tregs in an attempt to regulate ongoing autoimmunity. Importantly, in two distinct in vivo models, these TGF-beta-dependent adaptive CD4(+)CD25(low) T cells can be induced from peripheral CD4(+)CD25(-) T lymphocytes by anti-CD3 immunotherapy which correlates with the restoration of self-tolerance.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Forkhead Transcription Factors/immunology , Immunotherapy/methods , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Antibodies/immunology , CD28 Antigens/genetics , CD28 Antigens/immunology , CD3 Complex/immunology , Diabetes Mellitus, Type 1/therapy , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Forkhead Transcription Factors/metabolism , Immunoglobulin Fab Fragments/immunology , Mice , Mice, Inbred NOD , Mice, Knockout , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...