Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Eur J Pharm Biopharm ; 108: 220-225, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27648957

ABSTRACT

Formulating poorly water soluble drugs using ordered mesoporous silica materials is an emerging approach to tackle solubility-related bioavailability problems. The current study was conducted to assess the bioavailability-enhancing potential of ordered mesoporous silica in man. In this open-label, randomized, two-way cross-over study, 12 overnight fasted healthy volunteers received a single dose of fenofibrate formulated with ordered mesoporous silica or a marketed product based on micronized fenofibrate. Plasma concentrations of fenofibric acid, the pharmacologically active metabolite of fenofibrate, were monitored up to 96h post-dose. The rate (Cmax/dose increased by 77%; tmax reduced by 0.75h) and extent of absorption (AUC0-24h/dose increased by 54%) of fenofibrate were significantly enhanced following administration of the ordered mesoporous silica based formulation. The results of this study serve as a proof of concept in man for this novel formulation approach.


Subject(s)
Fenofibrate/pharmacokinetics , Silicon Dioxide/chemistry , Water/chemistry , Administration, Oral , Adult , Area Under Curve , Biological Availability , Cross-Over Studies , Female , Fenofibrate/analogs & derivatives , Fenofibrate/chemistry , Healthy Volunteers , Humans , Limit of Detection , Male , Middle Aged , Porosity , Solubility
2.
PLoS One ; 7(3): e33418, 2012.
Article in English | MEDLINE | ID: mdl-22428047

ABSTRACT

Switching to a glycolytic metabolism is a rapid adaptation of tumor cells to hypoxia. Although this metabolic conversion may primarily represent a rescue pathway to meet the bioenergetic and biosynthetic demands of proliferating tumor cells, it also creates a gradient of lactate that mirrors the gradient of oxygen in tumors. More than a metabolic waste, the lactate anion is known to participate to cancer aggressiveness, in part through activation of the hypoxia-inducible factor-1 (HIF-1) pathway in tumor cells. Whether lactate may also directly favor HIF-1 activation in endothelial cells (ECs) thereby offering a new druggable option to block angiogenesis is however an unanswered question. In this study, we therefore focused on the role in ECs of monocarboxylate transporter 1 (MCT1) that we previously identified to be the main facilitator of lactate uptake in cancer cells. We found that blockade of lactate influx into ECs led to inhibition of HIF-1-dependent angiogenesis. Our demonstration is based on the unprecedented characterization of lactate-induced HIF-1 activation in normoxic ECs and the consecutive increase in vascular endothelial growth factor receptor 2 (VEGFR2) and basic fibroblast growth factor (bFGF) expression. Furthermore, using a variety of functional assays including endothelial cell migration and tubulogenesis together with in vivo imaging of tumor angiogenesis through intravital microscopy and immunohistochemistry, we documented that MCT1 blockers could act as bona fide HIF-1 inhibitors leading to anti-angiogenic effects. Together with the previous demonstration of MCT1 being a key regulator of lactate exchange between tumor cells, the current study identifies MCT1 inhibition as a therapeutic modality combining antimetabolic and anti-angiogenic activities.


Subject(s)
Endothelial Cells/metabolism , Hypoxia-Inducible Factor 1/metabolism , Lactic Acid/metabolism , Monocarboxylic Acid Transporters/metabolism , Neoplasms/blood supply , Neovascularization, Pathologic/metabolism , Symporters/metabolism , Analysis of Variance , Animals , Blotting, Western , Cell Movement/physiology , Endothelial Cells/physiology , Enzyme-Linked Immunosorbent Assay , Fibroblast Growth Factors/metabolism , Humans , Immunohistochemistry , Lactic Acid/pharmacology , Luciferases , Mice , Nuclear Magnetic Resonance, Biomolecular , Polymerase Chain Reaction , RNA Interference , RNA, Small Interfering/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
3.
Mol Cancer Res ; 7(7): 1056-63, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19567781

ABSTRACT

The biological status of nitrite recently evolved from an inactive end product of nitric oxide (NO) metabolism to a major intravascular and tissue storage of NO. Several enzymes and proteins may indeed work as nitrite reductases. The endothelial NO synthase (eNOS) is proposed to be one of them, particularly when oxygen is lacking. Here, we examined whether the lack of caveolin, a scaffold protein known to limit eNOS activity under basal conditions and to be down-regulated in tumor vessels, could favor the reconversion of nitrite into NO and thereby promote angiogenesis. We found that nitrite-rich serum from caveolin-deficient mice and exogenous nitrite exert proangiogenic effects on aortic explants cultured in a three-dimensional collagen matrix. We identified a higher intrinsic capacity of caveolin-deficient vessels and endothelial cells to convert nitrite into bioactive NO. These effects did occur under moderate hypoxia and were abolished on exposure to a NO scavenger. Evidence for eNOS acting as a nitrite reductase derived from the failure to reproduce the proangiogenic effects of nitrite on eNOS-deficient aorta rings and endothelial cells. Finally, in a mouse tumor model, we documented the higher nitrite content in hypoxic tumors and identified inducible NO synthase as the major source of nitrite. Altogether, these data identify the lack of caveolin observed in the tumor vasculature as a favorable ground for nitrite-driven formation of endothelial tubes in the hypoxic tumor microenvironment. This work also strengthens the therapeutic value of the modulation of caveolin expression to interfere with tumor angiogenesis.


Subject(s)
Caveolin 1/deficiency , Neoplasms, Experimental/blood supply , Nitric Oxide/metabolism , Nitrites/pharmacology , Analysis of Variance , Animals , Aorta/growth & development , Aorta/metabolism , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Hypoxia/physiology , Disease Models, Animal , Down-Regulation , Female , Male , Mice , Mice, Transgenic , Neoplasm Transplantation , Neoplasms, Experimental/metabolism , Neovascularization, Pathologic/blood , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type III/metabolism , Nitrites/blood , Nitrites/metabolism , RNA, Small Interfering/metabolism
4.
Clin Cancer Res ; 14(9): 2768-74, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18451244

ABSTRACT

PURPOSE: The biological status of nitrite recently evolved from an inactive end product of nitric oxide catabolism to the largest intravascular and tissue storage of nitric oxide (NO). Although low partial O(2) pressure favors enzymatic reconversion of nitrite into NO, low pH supports a nonenzymatic pathway. Because hypoxia and acidity are characteristics of the tumor microenvironment, we examined whether nitrite injection could preferentially lead to NO production in tumors and influence response to treatments. EXPERIMENTAL DESIGN: The effects of nitrite were evaluated on arteriole vasorelaxation, tumor cell respiration and tumor blood flow, oxygenation, and response to radiotherapy. RESULTS: We first showed that a small drop in pH (-0.6 pH unit) favored the production of bioactive NO from nitrite by documenting a higher cyclic guanosine 3',5'-monophosphate-dependent arteriole vasorelaxation. We then documented that an i.v. bolus injection of nitrite to tumor-bearing mice led to a transient increase in partial O(2) pressure in tumor but not in healthy tissues. Blood flow measurements failed to reveal an effect of nitrite on tumor perfusion, but we found that O(2) consumption by nitrite-exposed tumor cells was decreased at acidic pH. Finally, we showed that low dose of nitrite could sensitize tumors to radiotherapy, leading to a significant growth delay and an increase in mouse survival (versus irradiation alone). CONCLUSIONS: This study identified low pH condition (encountered in many tumors) as an exquisite environment that favors tumor-selective production of NO in response to nitrite systemic injection. This work opens new perspectives for the use of nitrite as a safe and clinically applicable radiosensitizing modality.


Subject(s)
Liver Neoplasms/metabolism , Nitric Oxide/metabolism , Nitrites/metabolism , Radiation-Sensitizing Agents , Animals , Blood Vessels/drug effects , Hydrogen-Ion Concentration , Liver Neoplasms/blood supply , Liver Neoplasms/radiotherapy , Male , Mice , Mice, Inbred Strains , Nitrites/administration & dosage , Nitrites/pharmacology , Oxygen Consumption/drug effects , Vasodilation/drug effects
5.
Am J Pathol ; 171(5): 1619-28, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17916598

ABSTRACT

In the normal microvasculature, caveolin-1, the structural protein of caveolae, modulates transcytosis and paracellular permeability. Here, we used caveolin-1-deficient mice (Cav(-/-)) to track the potential active roles of caveolin-1 down-modulation in the regulation of vascular permeability and morphogenesis in tumors. In B16 melanoma-bearing Cav(-/-) mice, we found that fibrinogen accumulated in early-stage tumors to a larger extent than in wild-type animals. These results were confirmed by the observations of a net elevation of the interstitial fluid pressure and a relative deficit in albumin extravasation in Cav(-/-) tumors (versus healthy tissues). Immunostaining analyses of Cav(-/-) tumor sections further revealed a higher density of CD31-positive vascular structures and a dramatic deficit in alpha-smooth muscle actin-stained mural cells. The increase in blood plasma volume in Cav(-/-) tumors was confirmed by dynamic contrast enhanced-magnetic resonance imaging and found to be associated with a more rapid tumor growth. Finally, an in vitro wound test and the aorta ring assay revealed that silencing caveolin expression could directly impair the migration and the outgrowth of smooth muscle cells/pericytes, particularly in response to platelet-derived growth factor. In conclusion, a decrease in caveolin abundance, by promoting angiogenesis and preventing its termination by mural cell recruitment, appears as an important control point for the formation of new tumor blood vessels. Caveolin-1 therefore has the potential to be a marker of tumor vasculature maturity that may help adjusting anticancer therapies.


Subject(s)
Capillary Permeability/physiology , Caveolin 1/physiology , Endothelial Cells/physiology , Endothelium, Vascular/pathology , Melanoma, Experimental/blood supply , Neovascularization, Pathologic/metabolism , Pericytes/physiology , Animals , Caveolin 1/genetics , Cell Line, Tumor , Cell Movement/physiology , Fibrinogen/metabolism , Mice , Mice, Knockout , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Platelet-Derived Growth Factor/physiology , Transplantation, Heterologous
6.
Int J Radiat Oncol Biol Phys ; 67(4): 1155-62, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17276618

ABSTRACT

PURPOSE: To determine whether radiation-induced increases in nitric oxide (NO) production can influence tumor blood flow and improve delivery of Akt-targeting therapeutic DNA lipocomplexes to the tumor. METHODS AND MATERIALS: The contribution of NO to the endothelial response to radiation was identified using NO synthase (NOS) inhibitors and endothelial NOS (eNOS)-deficient mice. Reporter-encoding plasmids complexed with cationic lipids were used to document the tumor vascular specificity and the efficacy of in vivo lipofection after irradiation. A dominant-negative Akt gene construct was used to evaluate the facilitating effects of radiotherapy on the therapeutic transgene delivery. RESULTS: The abundance of eNOS protein was increased in both irradiated tumor microvessels and endothelial cells, leading to a stimulation of NO release and an associated increase in tumor blood flow. Transgene expression was subsequently improved in the irradiated vs. nonirradiated tumor vasculature. This effect was not apparent in eNOS-deficient mice and could not be reproduced in irradiated cultured endothelial cells. Finally, we combined low-dose radiotherapy with a dominant-negative Akt gene construct and documented synergistic antitumor effects. CONCLUSIONS: This study offers a new rationale to combine radiotherapy with gene therapy, by directly exploiting the stimulatory effects of radiation on NO production by tumor endothelial cells. The preferential expression of the transgene in the tumor microvasculature underscores the potential of such an adjuvant strategy to limit the angiogenic response of irradiated tumors.


Subject(s)
Endothelium, Vascular/radiation effects , Genetic Therapy/methods , Neoplasms/blood supply , Neoplasms/therapy , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Proto-Oncogene Proteins c-akt/genetics , Animals , Combined Modality Therapy , Endothelial Cells/metabolism , Endothelial Cells/radiation effects , Endothelium, Vascular/physiology , Enzyme Inhibitors/pharmacology , Gene Transfer Techniques , Green Fluorescent Proteins/genetics , Liver Neoplasms, Experimental/blood supply , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/therapy , Male , Mice , NG-Nitroarginine Methyl Ester/pharmacology , Neoplasms/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type III/deficiency , Proto-Oncogene Proteins c-akt/metabolism , Radiotherapy Dosage , Vasodilation
7.
Cancer Res ; 66(10): 5234-41, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16707448

ABSTRACT

Among matrix metalloproteinases (MMP), MMP-19 displays unique structural features and tissue distribution. In contrast to most MMPs, MMP-19 is expressed in normal human epidermis and down-regulated during malignant transformation and dedifferentiation. The contribution of MMP-19 during tumor angiogenesis is presently unknown. In an attempt to give new insights into MMP-19 in vivo functions, angiogenic response of mutant mice lacking MMP-19 was analyzed after transplantation of murine malignant PDVA keratinocytes and after injection of Matrigel supplemented with basic fibroblast growth factor. In situ hybridization and immunohistochemical analysis revealed that MMP-19 is produced by host mesenchymal cells but not by endothelial capillary cells or CD11b-positive inflammatory cells. Based on a new computer-assisted method of quantification, we provide evidence that host MMP-19 deficiency was associated with an increased early angiogenic response. In addition, increased tumor invasion was observed in MMP-19-/- mice. We conclude that, in contrast to most MMPs that promote tumor progression, MMP-19 is a negative regulator of early steps of tumor angiogenesis and invasion. These data highlight the requirement to understand the individual functions of each MMP to improve anticancer strategies.


Subject(s)
Metalloendopeptidases/deficiency , Skin Neoplasms/blood supply , Animals , Collagen , Drug Combinations , Female , Laminin , Male , Matrix Metalloproteinases, Secreted , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/pathology , Proteoglycans , Skin Neoplasms/enzymology , Skin Neoplasms/pathology , Stromal Cells/enzymology
8.
Circ Res ; 98(9): 1219-27, 2006 May 12.
Article in English | MEDLINE | ID: mdl-16601228

ABSTRACT

When neovascularization is triggered in ischemic tissues, angiogenesis but also (postnatal) vasculogenesis is induced, the latter requiring the mobilization of endothelial progenitor cells (EPC) from the bone marrow. Caveolin, the structural protein of caveolae, was recently reported to directly influence the angiogenic process through the regulation of the vascular endothelial growth factor (VEGF)/nitric oxide pathway. In this study, using caveolin-1 null mice (Cav(-/-)), we examined whether caveolin was also involved in the EPC recruitment in a model of ischemic hindlimb. Intravenous infusion of Sca-1(+) Lin(-) progenitor cells, but not bone marrow transplantation, rescued the defective neovascularization in Cav(-/-) mice, suggesting a defect in progenitor mobilization. The adhesion of Cav(-/-) EPC to bone marrow stromal cells indeed appeared to be resistant to the otherwise mobilizing SDF-1 (Stromal cell-Derived Factor-1) exposure because of a defect in the internalization of the SDF-1 cognate receptor CXCR4. Symmetrically, the attachment of Cav(-/-) EPC to SDF-1-presenting endothelial cells was significantly increased. Finally, EPC transduction with caveolin small interfering RNA reproduced this advantage in vitro and, importantly, led to a more extensive rescue of the ischemic hindlimb after intravenous infusion (versus sham-transfected EPC). These results underline the critical role of caveolin in ensuring the caveolae-mediated endocytosis of CXCR4, regulating both the SDF-1-mediated mobilization and peripheral homing of progenitor cells in response to ischemia. In particular, a transient reduction in caveolin expression was shown to therapeutically increase the engraftment of progenitor cells.


Subject(s)
Caveolin 1/metabolism , Cell Movement , Chemokines, CXC/metabolism , Endothelial Cells , Hindlimb/blood supply , Ischemia/physiopathology , Neovascularization, Physiologic , Stem Cells , Animals , Bone Marrow Cells , Bone Marrow Transplantation , Caveolin 1/deficiency , Cell Adhesion , Chemokine CXCL12 , Chemokines, CXC/blood , Ischemia/metabolism , Mice , Mice, Knockout , Regional Blood Flow , Stromal Cells
9.
Trends Cardiovasc Med ; 15(5): 157-62, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16165011

ABSTRACT

Caveolae are plasmalemmal invaginations formed by the sequestration of cholesterol and glycosphingolipids with self-associating molecules named caveolins, resulting in a platform for the assembly of signaling complexes at the surface of the cell. The enrichment of the endothelial nitric oxide synthase in caveolae and its direct interaction with caveolin both account for the exquisite regulation of nitric oxide production in cardiovascular tissues. Dissection of the angiogenic signaling cascade downstream vascular endothelial growth factor recently led to recognition that although the former enables the compartmentation of endothelial nitric oxide synthase and optimizes the process leading to its activation, the latter maintains the enzyme in its inactivated state in the absence of stimulation. Alteration in caveolin abundance or subcellular location may lead endothelial cells or cardiac myocytes to favor one mode of regulation over the other and thereby alter the subtle equilibrium governing nitric oxide production in these cells.


Subject(s)
Caveolae/metabolism , Caveolins/metabolism , Neovascularization, Physiologic , Nitric Oxide Synthase Type III/metabolism , Animals , Endothelial Cells/metabolism , Humans , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Smooth Muscle/metabolism , Neovascularization, Physiologic/physiology
10.
Circulation ; 112(8): 1198-205, 2005 Aug 23.
Article in English | MEDLINE | ID: mdl-16116070

ABSTRACT

BACKGROUND: The therapeutic effects of nonspecific beta-blockers are limited by vasoconstriction, thus justifying the interest in molecules with ancillary vasodilating properties. Nebivolol is a selective beta1-adrenoreceptor antagonist that releases nitric oxide (NO) through incompletely characterized mechanisms. We identified endothelial beta3-adrenoreceptors in human coronary microarteries that mediate endothelium- and NO-dependent relaxation and hypothesized that nebivolol activates these beta3-adrenoreceptors. METHODS AND RESULTS: Nebivolol dose-dependently relaxed rodent coronary resistance microarteries studied by videomicroscopy (10 micromol/L, -86+/-6% of prostaglandin F2alpha contraction); this was sensitive to NO synthase (NOS) inhibition, unaffected by the beta(1-2)-blocker nadolol, and prevented by the beta(1-2-3)-blocker bupranolol (P<0.05; n=3 to 8). Importantly, nebivolol failed to relax microarteries from beta3-adrenoreceptor-deficient mice. Nebivolol (10 micromol/L) also relaxed human coronary microvessels (-71+/-5% of KCl contraction); this was dependent on a functional endothelium and NO synthase but insensitive to beta(1-2)-blockade (all P<0.05). In a mouse aortic ring assay of neoangiogenesis, nebivolol induced neocapillary tube formation in rings from wild-type but not beta3-adrenoreceptor- or endothelial NOS-deficient mice. In cultured endothelial cells, 10 micromol/L nebivolol increased NO release by 200% as measured by electron paramagnetic spin trapping, which was also reversed by NOS inhibition. In parallel, endothelial NOS was dephosphorylated on threonine(495), and fura-2 calcium fluorescence increased by 91.8+/-23.7%; this effect was unaffected by beta(1-2)-blockade but abrogated by beta(1-2-3)-blockade (all P<0.05). CONCLUSIONS: Nebivolol dilates human and rodent coronary resistance microarteries through an agonist effect on endothelial beta3-adrenoreceptors to release NO and promote neoangiogenesis. These properties may prove particularly beneficial for the treatment of ischemic and cardiac failure diseases through preservation of coronary reserve.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Benzopyrans/pharmacology , Coronary Circulation/drug effects , Ethanolamines/pharmacology , Nitric Oxide/metabolism , Receptors, Adrenergic, beta-3/metabolism , Vasodilation/drug effects , Animals , Calcium/metabolism , Coronary Circulation/physiology , Endothelium, Vascular/metabolism , Humans , Male , Mice , Microcirculation/drug effects , Microcirculation/physiology , Nebivolol , Nitrates/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitrites/metabolism , Phosphorylation , Rats , Rats, Wistar , Vasodilation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...