Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Cancer Res Commun ; 2(11): 1504-1519, 2022 11.
Article in English | MEDLINE | ID: mdl-36970050

ABSTRACT

The receptor tyrosine kinase VEGFR-3 plays a crucial role in cancer-induced angiogenesis and lymphangiogenesis, promoting tumor development and metastasis. Here, we report the novel VEGFR-3 inhibitor EVT801 that presents a more selective and less toxic profile than two major inhibitors of VEGFRs (i.e., sorafenib and pazopanib). As monotherapy, EVT801 showed a potent antitumor effect in VEGFR-3-positive tumors, and in tumors with VEGFR-3-positive microenvironments. EVT801 suppressed VEGF-C-induced human endothelial cell proliferation in vitro and tumor (lymph)angiogenesis in different tumor mouse models. In addition to reduced tumor growth, EVT801 decreased tumor hypoxia, favored sustained tumor blood vessel homogenization (i.e., leaving fewer and overall larger vessels), and reduced important immunosuppressive cytokines (CCL4, CCL5) and myeloid-derived suppressor cells (MDSC) in circulation. Furthermore, in carcinoma mouse models, the combination of EVT801 with immune checkpoint therapy (ICT) yielded superior outcomes to either single treatment. Moreover, tumor growth inhibition was inversely correlated with levels of CCL4, CCL5, and MDSCs after treatment with EVT801, either alone or combined with ICT. Taken together, EVT801 represents a promising anti(lymph)angiogenic drug for improving ICT response rates in patients with VEGFR-3 positive tumors. Significance: The VEGFR-3 inhibitor EVT801 demonstrates superior selectivity and toxicity profile than other VEGFR-3 tyrosine kinase inhibitors. EVT801 showed potent antitumor effects in VEGFR-3-positive tumors, and tumors with VEGFR-3-positive microenvironments through blood vessel homogenization, and reduction of tumor hypoxia and limited immunosuppression. EVT801 increases immune checkpoint inhibitors' antitumor effects.


Subject(s)
Neoplasms , Vascular Endothelial Growth Factor Receptor-3 , Humans , Mice , Animals , Vascular Endothelial Growth Factor Receptor-3/therapeutic use , Neovascularization, Pathologic/drug therapy , Neoplasms/drug therapy , Angiogenesis Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Immunotherapy , Tumor Microenvironment
2.
Br J Cancer ; 123(10): 1481-1489, 2020 11.
Article in English | MEDLINE | ID: mdl-32839491

ABSTRACT

BACKGROUND: AsiDNA, a first-in-class oligonucleotide-mimicking double-stranded DNA breaks, acts as a decoy agonist to DNA damage response in tumour cells. It also activates DNA-dependent protein kinase and poly (adenosine diphosphate [ADP]-ribose) polymerase enzymes that induce phosphorylation of H2AX and protein PARylation. METHODS: The aim of this Phase 1 study was to determine dose-limiting toxicities (DLTs), maximum tolerated dose (MTD), safety and pharmacokinetics/pharmacodynamics of AsiDNA administered daily for 3 days in the first week then weekly thereafter. Twenty-two patients with advanced solid tumours were enrolled in 5 dose levels: 200, 400, 600, 900, and 1300 mg, using a 3 + 3 design. RESULTS: The MTD was not reached. IV AsiDNA was safe. Two DLTs (grade 4 and grade 3 hepatic enzymes increased at 900 and 1300 mg), and two related SAE at 900 mg (grade 3 hypotension and grade 4 hepatic enzymes increased) were reported. AsiDNA PK increased proportionally with dose. A robust activation of DNA-PK by a significant posttreatment increase of γH2AX was evidenced in tumour biopsies. CONCLUSION: The dose of 600 mg was identified as the optimal dose for further clinical development. CLINICAL TRIAL REGISTRATION: Clinical trial registration (NCT number): NCT03579628.


Subject(s)
Cholesterol/analogs & derivatives , DNA/administration & dosage , DNA/adverse effects , DNA/pharmacokinetics , Neoplasms/drug therapy , Administration, Intravenous , Adult , Aged , Belgium , Cholesterol/administration & dosage , Cholesterol/adverse effects , Cholesterol/pharmacokinetics , DNA Repair/drug effects , Disease Progression , Dose-Response Relationship, Drug , Female , France , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/metabolism , Neoplasms/pathology
4.
Front Oncol ; 9: 1097, 2019.
Article in English | MEDLINE | ID: mdl-31781480

ABSTRACT

Purpose: Carboplatin is used to treat many cancers, but occurrence of drug resistance and its high toxicity remain a clinical hurdle limiting its efficacy. We compared the efficacy and toxicity of DNA repair inhibitors olaparib or AsiDNA administered alone or in combination with carboplatin. Olaparib acts by inhibiting PARP-dependent repair pathways whereas AsiDNA inhibits double-strand break repair by preventing recruitment of enzymes involved in homologous recombination and non-homologous end joining. Experimental Design: Mice with MDA-MB-231 tumors were treated with carboplatin or/and olaparib or AsiDNA for three treatment cycles. Survival and tumor growth were monitored. Toxicities of treatments were assayed in C57BL/6 immunocompetent mice. Circulating blood hematocrits, bone marrow cells, and organs were analyzed 10 and 21 days after end of treatment using flow cytometry and microscopy analysis. Resistance occurrence was monitored after cycles of treatments with combination of AsiDNA and carboplatin in independent BC227 cell cultures. Results: Olaparib or AsiDNA monotherapies decreased tumor growth and increased mean survival of grafted animals. The combination with carboplatin further increased survival. Carboplatin toxicity resulted in a decrease of most blood cells, platelets, thymus, and spleen lymphocytes. Olaparib or AsiDNA monotherapies had no toxicity, and their combination with carboplatin did not increase toxicity in the bone marrow or thrombocytopenia. All animals receiving carboplatin combined with olaparib developed high liver toxicity with acute hepatitis at 21 days. In vitro, carboplatin resistance occurs after three cycles of treatment in all six tested cultures, whereas only one became resistant (1/5) after five cycles when carboplatin was associated to low doses of AsiDNA. All selected carboplatin-resistant clones retain sensitivity to AsiDNA. Conclusion: DNA repair inhibitor treatments are efficient in the platinum resistant model, MDA-MB-231. The combination with carboplatin improves survival. The association of carboplatin with olaparib is associated with high liver toxicity, which is not observed with AsiDNA. AsiDNA could delay resistance to carboplatin without increasing its toxicity.

5.
Neoplasia ; 21(9): 863-871, 2019 09.
Article in English | MEDLINE | ID: mdl-31362243

ABSTRACT

The Achilles heel of anticancer treatments is intrinsic or acquired resistance. Among many targeted therapies, the DNA repair inhibitors show limited efficacy due to rapid emergence of resistance. We examined evolution of cancer cells and tumors treated with AsiDNA, a new DNA repair inhibitor targeting all DNA break repair pathways. Effects of AsiDNA or Olaparib were analyzed in various cell lines. Frequency of AsiDNA- and olaparib-resistant clones was measured after 2 weeks of continuous treatment in KBM7 haploid cells. Cell survivals were also measured after one to six cycles of 1-week treatment and 1-week recovery in MDA-MB-231 and NCI-H446. Transcriptomes of cell populations recovering from cyclic treatments or mock treatment were compared. MDA-MB-231 xenografted models were treated with three cycles of AsiDNA to monitor the effects of treatment on tumor growth and transcriptional modifications. No resistant clones were selected after AsiDNA treatment (frequency < 3x10-8) in treatment conditions that generate resistance to olaparib at a frequency of 7.2x10-7 resistant clones per treated cell. Cyclic treatments promote cumulative sensitivity characterized by a higher mortality of cells having undergone previous treatment cycles. This sensitization was stable, and transcriptome analysis revealed a major gene downregulation with a specific overrepresentation of genes coding for targets of DNA-PK. Such changes were also detected in tumor models which showed impaired growth after cycles of AsiDNA treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , DNA Repair , DNA/administration & dosage , DNA/genetics , Drug Resistance, Neoplasm , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival , Dose-Response Relationship, Drug , Gene Expression Profiling , Haploidy , Heterografts , Humans , Mice , RNA Processing, Post-Transcriptional
6.
JCI Insight ; 2(4): e87489, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28239646

ABSTRACT

In breast cancer, a key feature of peritumoral adipocytes is their loss of lipid content observed both in vitro and in human tumors. The free fatty acids (FFAs), released by adipocytes after lipolysis induced by tumor secretions, are transferred and stored in tumor cells as triglycerides in lipid droplets. In tumor cell lines, we demonstrate that FFAs can be released over time from lipid droplets through an adipose triglyceride lipase-dependent (ATGL-dependent) lipolytic pathway. In vivo, ATGL is expressed in human tumors where its expression correlates with tumor aggressiveness and is upregulated by contact with adipocytes. The released FFAs are then used for fatty acid ß-oxidation (FAO), an active process in cancer but not normal breast epithelial cells, and regulated by coculture with adipocytes. However, in cocultivated cells, FAO is uncoupled from ATP production, leading to AMPK/acetyl-CoA carboxylase activation, a circle that maintains this state of metabolic remodeling. The increased invasive capacities of tumor cells induced by coculture are completely abrogated by inhibition of the coupled ATGL-dependent lipolysis/FAO pathways. These results show a complex metabolic symbiosis between tumor-surrounding adipocytes and cancer cells that stimulate their invasiveness, highlighting ATGL as a potential therapeutic target to impede breast cancer progression.


Subject(s)
Adipocytes/metabolism , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Lobular/metabolism , Fatty Acids, Nonesterified/metabolism , Lipase/metabolism , Lipolysis , Acetyl-CoA Carboxylase/metabolism , Adenosine Triphosphate/metabolism , Adenylate Kinase/metabolism , Aged , Animals , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Lobular/pathology , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Female , Humans , Mice , Middle Aged , Neoplasm Invasiveness , Oxidation-Reduction , Triglycerides/metabolism
8.
Angiogenesis ; 19(3): 389-406, 2016 07.
Article in English | MEDLINE | ID: mdl-27234973

ABSTRACT

Anti-vascular endothelial growth factor (VEGF) therapies have improved clinical outcomes for patients with cancers and retinal vascular diseases. Three anti-VEGF agents, pegaptanib, ranibizumab, and aflibercept, are approved for ophthalmic indications, while bevacizumab is approved to treat colorectal, lung, and renal cancers, but is also used off-label to treat ocular vascular diseases. The efficacy of bevacizumab relative to ranibizumab in treating neovascular age-related macular degeneration has been assessed in several trials. However, questions persist regarding its safety, as bevacizumab can form large complexes with dimeric VEGF165, resulting in multimerization of the Fc domain and platelet activation. Here, we compare binding stoichiometry, Fcγ receptor affinity, platelet activation, and binding to epithelial and endothelial cells in vitro for bevacizumab and aflibercept, in the absence or presence of VEGF. In contrast to bevacizumab, aflibercept forms a homogenous 1:1 complex with each VEGF dimer. Unlike multimeric bevacizumab:VEGF complexes, the monomeric aflibercept:VEGF complex does not exhibit increased affinity for low-affinity Fcγ receptors, does not activate platelets, nor does it bind to the surface of epithelial or endothelial cells to a greater degree than unbound aflibercept or control Fc. The latter finding reflects the fact that aflibercept binds VEGF in a unique manner, distinct from antibodies not only blocking the amino acids necessary for VEGFR1/R2 binding but also occluding the heparin-binding site on VEGF165.


Subject(s)
Bevacizumab/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Recombinant Fusion Proteins/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/therapeutic use , Animals , Antigen-Antibody Complex/chemistry , Antigen-Antibody Complex/metabolism , Bevacizumab/adverse effects , Bevacizumab/therapeutic use , Cell Line , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , Macular Degeneration/immunology , Macular Degeneration/metabolism , Macular Degeneration/therapy , Mice , Mice, Transgenic , Platelet Activation , Protein Binding , Protein Multimerization , Receptors, IgG/genetics , Receptors, IgG/metabolism , Receptors, Vascular Endothelial Growth Factor/therapeutic use , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/therapeutic use , Thrombocytopenia/etiology , Thrombosis/etiology , Vascular Endothelial Growth Factor A/immunology
9.
Brain ; 139(Pt 6): 1762-82, 2016 06.
Article in English | MEDLINE | ID: mdl-27084575

ABSTRACT

The p75 neurotrophin receptor is important in multiple physiological actions including neuronal survival and neurite outgrowth during development, and after central nervous system injury. We have discovered a novel piperazine-derived compound, EVT901, which interferes with p75 neurotrophin receptor oligomerization through direct interaction with the first cysteine-rich domain of the extracellular region. Using ligand binding assays with cysteine-rich domains-fused p75 neurotrophin receptor, we confirmed that EVT901 interferes with oligomerization of full-length p75 neurotrophin receptor in a dose-dependent manner. Here we report that EVT901 reduces binding of pro-nerve growth factor to p75 neurotrophin receptor, blocks pro-nerve growth factor induced apoptosis in cells expressing p75 neurotrophin receptor, and enhances neurite outgrowth in vitro Furthermore, we demonstrate that EVT901 abrogates p75 neurotrophin receptor signalling by other ligands, such as prion peptide and amyloid-ß. To test the efficacy of EVT901 in vivo, we evaluated the outcome in two models of traumatic brain injury. We generated controlled cortical impacts in adult rats. Using unbiased stereological analysis, we found that EVT901 delivered intravenously daily for 1 week after injury, reduced lesion size, protected cortical neurons and oligodendrocytes, and had a positive effect on neurological function. After lateral fluid percussion injury in adult rats, oral treatment with EVT901 reduced neuronal death in the hippocampus and thalamus, reduced long-term cognitive deficits, and reduced the occurrence of post-traumatic seizure activity. Together, these studies provide a new reagent for altering p75 neurotrophin receptor actions after injury and suggest that EVT901 may be useful in treatment of central nervous system trauma and other neurological disorders where p75 neurotrophin receptor signalling is affected.


Subject(s)
Oligodendroglia/drug effects , Piperazines/pharmacology , Receptor, Nerve Growth Factor/antagonists & inhibitors , Animals , Apoptosis/drug effects , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Cell Count , Cell Proliferation/drug effects , Cells, Cultured , Demyelinating Diseases/pathology , Dose-Response Relationship, Drug , Humans , Male , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oligodendroglia/metabolism , Phosphorylation/drug effects , Primary Cell Culture , Radioligand Assay , Rats , Receptor, Nerve Growth Factor/biosynthesis , Receptor, trkA/metabolism , Recovery of Function
10.
J Biol Chem ; 290(6): 3405-17, 2015 Feb 06.
Article in English | MEDLINE | ID: mdl-25384978

ABSTRACT

The formation of new vessels in the tumor, termed angiogenesis, is essential for primary tumor growth and facilitates tumor invasion and metastasis. Hypoxia has been described as one trigger of angiogenesis. Indeed, hypoxia, which is characterized by areas of low oxygen levels, is a hallmark of solid tumors arising from an imbalance between oxygen delivery and consumption. Hypoxic conditions have profound effects on the different components of the tumoral environment. For example, hypoxia is able to activate endothelial cells, leading to angiogenesis but also thereby initiating a cascade of reactions involving neutrophils, smooth muscle cells, and fibroblasts. In addition, hypoxia directly regulates the expression of many genes for which the role and the importance in the tumoral environment remain to be completely elucidated. In this study, we used a method to selectively label sialoglycoproteins to identify new membrane and secreted proteins involved in the adaptative process of endothelial cells by mass spectrometry-based proteomics. We used an in vitro assay under hypoxic condition to observe an increase of protein expression or modifications of glycosylation. Then the function of the identified proteins was assessed in a vasculogenesis assay in vivo by using a morpholino strategy in zebrafish. First, our approach was validated by the identification of sialoglycoproteins such as CD105, neuropilin-1, and CLEC14A, which have already been described as playing key roles in angiogenesis. Second, we identified several new proteins regulated by hypoxia and demonstrated for the first time the pivotal role of GLUT-1, TMEM16F, and SDF4 in angiogenesis.


Subject(s)
Neovascularization, Physiologic , Protein Processing, Post-Translational , Sialoglycoproteins/metabolism , Adaptation, Physiological , Animals , Anoctamins , Antigens, CD/genetics , Antigens, CD/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Hypoxia , Endoglin , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Glycosylation , Human Umbilical Vein Endothelial Cells , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Neuropilin-1/genetics , Neuropilin-1/metabolism , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/metabolism , Proteome/chemistry , Proteome/metabolism , Proteomics/methods , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Sialoglycoproteins/genetics , Zebrafish
11.
J Cell Physiol ; 230(1): 43-51, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24760775

ABSTRACT

Tumor angiogenesis is accompanied by vasculogenesis, which is involved in the differentiation and mobilization of human bone marrow cells. In order to further characterize the role of vasculogenesis in the tumor growth process, the effects of FGF2 on the differentiation of human bone marrow AC133(+) cells (BM-AC133(+)) into vascular precursors were studied in vitro. FGF2, like VEGFA, induced progenitor cell differentiation into cell types with endothelial cell characteristics. SSR128129E, a newly discovered specific FGFR antagonist acting by allosteric interaction with FGFR, abrogated FGF2-induced endothelial cell differentiation, showing that FGFR signaling is essential during this process. To assess the involvement of the FGF/FRGR signaling in vivo, the pre-clinical model of Lewis lung carcinoma (LL2) in mice was used. Subcutaneous injection of LL2 cells into mice induced an increase of circulating EPCs from peripheral blood associated with tumor growth and an increase of intra-tumoral vascular index. Treatment with the FGFR antagonist SSR128129E strongly decreased LL2 tumor growth as well as the intra-tumoral vascular index (41% and 50% decrease vs. vehicle-treated mice respectively, P < 0.01). Interestingly, SSR128129E treatment significantly decreased the number of circulating EPCs from the peripheral blood (53% inhibition vs. vehicle-treated mice, P < 0.01). These results demonstrate for the first time that the blockade of the FGF/FGFR pathway by SSR128129E reduces EPC recruitment during angiogenesis-dependent tumor growth. In this context, circulating EPCs could be a reliable surrogate marker for tumor growth and angiogenic activity.


Subject(s)
Carcinoma, Lewis Lung/blood supply , Hematopoietic Stem Cells/cytology , Indolizines/pharmacology , Neovascularization, Pathologic/pathology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , ortho-Aminobenzoates/pharmacology , AC133 Antigen , Animals , Antigens, CD/biosynthesis , Bone Marrow Cells/metabolism , Cell Adhesion , Cell Differentiation/drug effects , Cell Line , Cell Movement , Endothelial Cells/cytology , Fibroblast Growth Factor 2/metabolism , Glycoproteins/biosynthesis , Humans , Mice , Mice, Inbred C57BL , Peptides , RNA, Messenger/biosynthesis , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction
12.
Pharm Pat Anal ; 3(6): 585-612, 2014.
Article in English | MEDLINE | ID: mdl-25489913

ABSTRACT

Fibroblast growth factor receptors (FGFRs) and associated ligands (FGFs) are a family of well-validated targets for therapeutic interventions notably in cancer diseases in relation to their prominent roles in cell growth, survival, differentiation and angiogenesis. This patent review encompasses all different approaches (modulators of FGF or FGFR expression, anti-FGF antibodies, anti-FGFR antibodies, FGF traps, tyrosine-kinase (TK) inhibitors, allosteric modulators) used to block completely or partially the activities of the FGF-FGFR complexes resulting in clinical drug candidates or tool agents. Comparative analysis of biochemical, pharmacological or clinical data will be discussed for each class of molecules together with some perspectives.


Subject(s)
Fibroblast Growth Factors , Neoplasms/drug therapy , Receptors, Fibroblast Growth Factor , Animals , Antibodies/therapeutic use , Antineoplastic Agents/therapeutic use , Aptamers, Nucleotide/therapeutic use , Fibroblast Growth Factors/immunology , Fibroblast Growth Factors/metabolism , Humans , Patents as Topic , Peptides/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/immunology , Receptors, Fibroblast Growth Factor/metabolism
13.
PLoS One ; 9(11): e113215, 2014.
Article in English | MEDLINE | ID: mdl-25405900

ABSTRACT

RGS18 is a myeloerythroid lineage-specific regulator of G-protein signaling, highly expressed in megakaryocytes (MKs) and platelets. In the present study, we describe the first generation of a RGS18 knockout mouse model (RGS18-/-). Interesting phenotypic differences between RGS18-/- and wild-type (WT) mice were identified, and show that RGS18 plays a significant role in both platelet generation and function. RGS18 deficiency produced a gain of function phenotype in platelets. In resting platelets, the level of CD62P expression was increased in RGS18-/- mice. This increase correlated with a higher level of plasmatic serotonin concentration. RGS18-/- platelets displayed a higher sensitivity to activation in vitro. RGS18 deficiency markedly increased thrombus formation in vivo. In addition, RGS18-/- mice presented a mild thrombocytopenia, accompanied with a marked deficit in MK number in the bone marrow. Analysis of MK maturation in vitro and in vivo revealed a defective megakaryopoiesis in RGS18-/- mice, with a lower bone marrow content of only the most committed MK precursors. Finally, RGS18 deficiency was correlated to a defect of platelet recovery in vivo under acute conditions of thrombocytopenia. Thus, we highlight a role for RGS18 in platelet generation and function, and provide additional insights into the physiology of RGS18.


Subject(s)
Megakaryocytes/metabolism , Platelet Activation/physiology , RGS Proteins/genetics , RGS Proteins/metabolism , Signal Transduction/physiology , Analysis of Variance , Animals , Blood Cell Count , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Phylogeny , Platelet Activation/genetics , Promoter Regions, Genetic/genetics , Serotonin/blood , Signal Transduction/genetics , Thrombosis/metabolism
14.
Chem Biol ; 21(10): 1310-1317, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25200605

ABSTRACT

Angiogenesis contributes to the development of numerous disorders. Even though fibroblast growth factors (FGFs) were discovered as mediators of angiogenesis more than 30 years ago, their role in developmental angiogenesis still remains elusive. We use a recently described chemical probe, SSR128129E (SSR), that selectively inhibits the action of multiple FGF receptors (FGFRs), in combination with the zebrafish model to examine the role of FGF signaling in vascular development. We observe that while FGFR signaling is less important for vessel guidance, it affects vascular outgrowth and is especially required for the maintenance of blood vessel integrity by ensuring proper cell-cell junctions between endothelial cells. In conclusion, our work illustrates the power of a small molecule probe to reveal insights into blood vessel formation and stabilization and thus of broad interest to the vascular biology community.


Subject(s)
Blood Vessels/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Zebrafish Proteins/metabolism , Animal Fins/physiology , Animals , Blood Vessels/drug effects , Cadherins/metabolism , Embryo, Nonmammalian/metabolism , Indolizines/chemistry , Indolizines/metabolism , Indolizines/pharmacology , Intercellular Junctions/metabolism , Neovascularization, Physiologic/drug effects , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Regeneration , Signal Transduction/drug effects , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/antagonists & inhibitors , ortho-Aminobenzoates/chemistry , ortho-Aminobenzoates/metabolism , ortho-Aminobenzoates/pharmacology
15.
J Med Chem ; 57(17): 7293-316, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25075638

ABSTRACT

In the search of a potential backup for clopidogrel, we have initiated a HTS campaign designed to identify novel reversible P2Y12 antagonists. Starting from a hit with low micromolar binding activity, we report here the main steps of the optimization process leading to the identification of the preclinical candidate SAR216471. It is a potent, highly selective, and reversible P2Y12 receptor antagonist and by far the most potent inhibitor of ADP-induced platelet aggregation among the P2Y12 antagonists described in the literature. SAR216471 displays potent in vivo antiplatelet and antithrombotic activities and has the potential to differentiate from other antiplatelet agents.


Subject(s)
Indoles/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Purinergic P2 Receptor Antagonists/pharmacology , Pyridazines/pharmacology , Receptors, Purinergic P2Y12/metabolism , Acute Coronary Syndrome/prevention & control , Adenosine Diphosphate/pharmacology , Administration, Oral , Animals , Binding, Competitive , CHO Cells , Cricetinae , Cricetulus , Humans , Indoles/chemical synthesis , Indoles/metabolism , Injections, Intravenous , Male , Models, Chemical , Molecular Structure , Platelet Aggregation Inhibitors/chemical synthesis , Platelet Aggregation Inhibitors/metabolism , Purinergic P2 Receptor Antagonists/chemical synthesis , Purinergic P2 Receptor Antagonists/metabolism , Pyridazines/chemical synthesis , Pyridazines/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2Y12/genetics , Thrombosis/prevention & control
16.
Eur J Cancer ; 50(13): 2351-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24953334

ABSTRACT

Resistance of glioblastoma to radiotherapy is mainly due to tumour cell radioresistance, which is partially controlled by growth factors such as fibroblast growth factor (FGF). Because we have previously demonstrated the role of FGF-2 in tumour cell radioresistance, we investigate here whether inhibiting FGF-2 pathways by targeting fibroblast growth factor receptor (FGFR) may represent a new strategy to optimise the efficiency of radiotherapy in glioblastoma. Treating radioresistant U87 and SF763 glioblastoma cells with the FGFR inhibitor, SSR12819E, radiosensitises these cells while the survival after irradiation of the more radiosensitive U251 and SF767 cells was not affected. SSR128129E administration to U87 cells increases the radiation-induced mitotic cell death. It also decreased cell membrane availability of the FGFR-1 mainly expressed in these cells, increased this receptor's ubiquitylation, inhibited radiation-induced RhoB activation and modulated the level of hypoxia inducible factor, HIF-1α, a master regulator of hypoxia, thus suggesting a role of FGFR in the regulation of hypoxia pathways. Moreover, treating orthotopically U87 xenografted mice with SSR128129E before two subsequent local 2.5Gy irradiations significantly increased the animals neurological sign free survival (NSFS) compared to the other groups of treatment. These results strongly suggest that targeting FGFR with the FGFR blocker SSR128129E might represent an interesting strategy to improve the efficiency of radiotherapy in glioblastoma.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Indolizines/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , ortho-Aminobenzoates/pharmacology , Animals , Brain Neoplasms/pathology , Cell Hypoxia/physiology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Signal Transduction , Transfection , Xenograft Model Antitumor Assays
17.
Cancers (Basel) ; 6(1): 472-90, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24589997

ABSTRACT

Myeloid derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) represent prominent components in cancer progression. We previously showed that inhibition of the VEGFR-3 pathway by SAR131675 leads to reduction of TAM infiltration and tumor growth. Here, we found that treatment with SAR131675 prevents the accumulation of immunosuppressive blood and splenic MDSCs which express VEGFR-3, in 4T1 tumor bearing mice. Moreover we showed that soluble factors secreted by tumor cells promote MDSCs proliferation and differentiation into M2 polarized F4/80+ macrophages. In addition, cell sorting and transcriptomic analysis of tumor infiltrating myeloid cells revealed the presence of a heterogeneous population that could be divided into 3 subpopulations: (i) immature cells with a MDSC phenotype (GR1+/CD11b+/F4/80-); (ii) "immuno-incompetent" macrophages (F4/80high/CD86neg/MHCIILow) strongly expressing M2 markers such as Legumain, CD206 and Mgl1/2 and (iii) "immuno-competent"-M1 like macrophages (F4/80Low/CD86+/MHCIIHigh). SAR131675 treatment reduced MDSCs in lymphoid organs as well as F4/80High populations in tumors. Interestingly, in the tumor SAR131675 was able to increase the immunocompetent M1 like population (F4/80low). Altogether these results demonstrate that the specific VEGFR-3 inhibitor SAR131675 exerts its anti tumoral activity by acting on different players that orchestrate immunosuppression and cancer progression in a tumoral context: MDSCs in peripheral lymphoid organs and TAMs infiltrating the tumor.

18.
PLoS One ; 8(11): e80027, 2013.
Article in English | MEDLINE | ID: mdl-24224032

ABSTRACT

OBJECTIVE: The role of fibroblast growth factors (FGFs) in the development of vascular diseases remains incompletely understood. The objective of this study was to examine the effects of a new small-molecule multi-FGF receptor blocker with allosteric properties, SSR128129E, on neointimal proliferation after a vein graft procedure in mice and on the development of atherosclerosis in atherosclerosis-prone apolipoprotein E (apoE)-deficient mice. METHODS AND RESULTS: Vein grafts were performed in 3 month-old male C57BL6 mice. Segments of the vena cava were interposed at the level of the carotid artery. In SSR128129E (50 mg/kg/d)-treated animals, a dramatic decrease in neointimal proliferation was observed 2 and 8 weeks after the graft (72.5%, p<0.01, and 47.8%, p<0.05, respectively). Four-week old male apoE-deficient mice were treated with SSR128129E (50 mg/kg/d) for 3 and 5 months in comparison with a control group. SSR128129E treatment resulted in a reduction of lesion size in the aortic sinus (16.4% (ns) at 3 months and 42.9% (p<0.01) at 5 months, without any change in serum lipids. SSR128129 significantly reduced FGFR2 mRNA levels in the aortic sinus (p<0.05, n=5-6), but did not affect the mRNA expression levels of other FGF receptors or ligands. CONCLUSION: These studies indicate that FGFs have an important role in the development of vascular diseases like atherosclerosis and graft arteriosclerosis. These data suggest that inhibition of FGF receptors by compounds like SSR128129E might be useful as a new therapeutic approach for these vascular pathologies.


Subject(s)
Atherosclerosis/drug therapy , Atherosclerosis/surgery , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/metabolism , Veins/transplantation , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Veins/surgery
19.
J Med Chem ; 56(23): 9441-56, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24175584

ABSTRACT

Compound 15 (SAR107375), a novel potent dual thrombin and factor Xa inhibitor resulted from a rational optimization process. Starting from compound 14, with low factor Xa and modest anti-thrombin inhibitory activities (IC50's of 3.5 and 0.39 µM, respectively), both activities were considerably improved, notably through the incorporation of a neutral chlorothiophene P1 fragment and tuning of P2 and P3-P4 fragments. Final optimization of metabolic stability with microsomes led to the identification of 15, which displays strong activity in vitro vs factor Xa and thrombin (with Ki's of 1 and 8 nM, respectively). In addition 15 presents good selectivity versus related serine proteases (roughly 300-fold), including trypsin (1000-fold), and is very active (0.39 µM) in the thrombin generation time (TGT) coagulation assay in human platelet rich plasma (PRP). Potent in vivo activity in a rat model of venous thrombosis following iv and, more importantly, po administration was also observed (ED50 of 0.07 and 2.8 mg/kg, respectively). Bleeding liability was reduced in the rat wire coil model, more relevant to arterial thrombosis, with 15 (blood loss increase of 2-fold relative to the ED80 value) compared to rivaroxaban 2 and dabigatran etexilate 1a.


Subject(s)
Anticoagulants/chemical synthesis , Factor Xa Inhibitors , Fibrinolytic Agents/chemical synthesis , Piperazines/chemical synthesis , Sulfonamides/chemical synthesis , Thrombin/antagonists & inhibitors , Animals , Anticoagulants/pharmacology , Crystallography, X-Ray , Drug Design , Fibrinolytic Agents/pharmacology , Humans , Piperazines/pharmacology , Piperazines/therapeutic use , Rats , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Venous Thrombosis/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...