Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters











Publication year range
1.
Sci Rep ; 10(1): 16883, 2020 10 09.
Article in English | MEDLINE | ID: mdl-33037252

ABSTRACT

Vitamin C (Vit C) benefits to human skin physiology notably by stimulating the biosynthesis of collagen. The main cutaneous collagens are types I and III, which are less synthesized with aging. Vit C is one of the main promotors of collagen formation but it poorly bypasses the epidermis stratum corneum barrier. To address this challenge, we developed a lipophilic version of Vit C for improving skin diffusion and delivery. Vit C was covalently conjugated to squalene (SQ), a natural lipid of the skin, forming a novel Vit C-SQ derivative suitable for cream formulation. Its biological activity was investigated on human whole skin explants in an ex vivo model, through histology and protein and gene expression analyses. Results were compared to Vit C coupled to the reference lipophilic compound palmitic acid, (Vit C-Palmitate). It was observed that Vit C-SQ significantly increased epidermal thickness and preferentially favored collagen III production in human skin after application for 10 days. It also promoted glycosaminoglycans production in a higher extent comparatively to Vit C-Palmitate and free Vit C. Microdissection of the explants to separate dermis and epidermis allowed to measure higher transcriptional effects either in epidermis or in dermis. Among the formulations studied, the strongest effects were observed with Vit C-SQ.


Subject(s)
Ascorbic Acid/pharmacology , Collagen/biosynthesis , Drug Delivery Systems , Epidermis/drug effects , Skin/drug effects , Skin/metabolism , Squalene , Adult , Ascorbic Acid/metabolism , Drug Compounding , Epidermis/metabolism , Female , Humans , In Vitro Techniques
2.
Eur J Pharm Biopharm ; 144: 165-173, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31546021

ABSTRACT

Among anticancer nanomedicines, squalenoyl nanocomposites have obtained encouraging outcomes in a great variety of tumors. The prodrug squalenoyl-gemcitabine has been chosen in this study to construct a novel multidrug nanosystem in combination with edelfosine, an alkyl-lysophopholipid with proven anticancer activity. Given their amphiphilic nature, it was hypothesized that both anticancer compounds, with complementary molecular targets, could lead to the formation of a new multitherapy nanomedicine. Nanoassemblies were formulated by the nanoprecipitation method and characterized by dynamic light scattering, transmission electron microscopy and X-ray photoelectron spectroscopy. Because free edelfosine is highly hemolytic, hemolysis experiments were performed using human blood erythrocytes and nanoassemblies efficacy was evaluated in a patient-derived metastatic pediatric osteosarcoma cell line. It was observed that these molecules spontaneously self-assembled as stable and monodisperse nanoassemblies of 51 ±â€¯1 nm in a surfactant/polymer free-aqueous suspension. Compared to squalenoyl-gemcitabine nanoassemblies, the combination of squalenoyl-gemcitabine with edelfosine resulted in smaller particle size and a new supramolecular conformation, with higher stability and drug content, and ameliorated antitumor profile.


Subject(s)
Deoxycytidine/analogs & derivatives , Lysophospholipids/chemistry , Phospholipid Ethers/chemistry , Prodrugs/chemistry , Squalene/chemistry , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Deoxycytidine/chemistry , Humans , Microscopy, Electron, Transmission/methods , Nanomedicine/methods , Particle Size , Gemcitabine
3.
Article in English | MEDLINE | ID: mdl-23747388

ABSTRACT

The photophysical properties of the antiviral 7-nicotinoyl-styrylquinoline (MB96) were investigated by means of UV-Vis linear dichroism (LD) spectroscopy on molecular samples aligned in stretched polyvinylalcohol (PVA), supported by time dependent density functional theory (TD-DFT) calculations. Experimentally, the directions of the transitions moments with respect to the long axis of the molecule were deduced from the orientation K factors, determined by means of "trial-and-error" procedure. The absorption spectrum presents two parts. The main transition in the lowest energy part, observed around 365 nm and showing the highest K value 0.8, is longitudinally in-plane polarized. The highest energy part which is extended between 230 and 320 nm, large, diffuse, and of weak intensity, shows estimated K values between 0.2 and 0.5. This complex structure is transversally polarized with some contamination by the longitudinal character of the first strong band. The TD-DFT results agree fairly well with the LD measurements.


Subject(s)
Hydroxyquinolines/pharmacology , Models, Molecular , Optical Phenomena , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Quantum Theory , Hydroxyquinolines/chemistry , Molecular Conformation , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-pim-1/metabolism , Solutions , Spectrophotometry, Ultraviolet , Temperature
4.
Eur J Pharm Biopharm ; 79(3): 612-20, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21784150

ABSTRACT

We have designed an amphiphilic prodrug of the anticancer agent gemcitabine (dFdC), by covalent coupling to squalene. This bioconjugate, which self-assembled into nanoparticles (NPs) in water, was previously found to display an impressive anticancer activity both in vitro and in vivo. The present study aims to investigate the impact of SQdFdC nanoparticles on cellular membranes. MTT assays showed that, in the nanomolar range, squalenoyl gemcitabine (SQdFdC) was slightly less active than dFdC on a panel of human cancer cell lines, in vitro. However, above 10 µmol L(-1) SQdFdC was considerably more cytotoxic than dFdC. Contrarily to its parent drug, SQdFdC also induced cell lysis in a few hours, as evidenced by LDH release assays. Erythrocytes were used as an experimental model insensitive to the antimetabolic activity of dFdC to further investigate the putative membrane-related cytotoxic activity of SQdFdC. The bioconjugate also induced hemolysis in a time- and dose-dependent fashion, unlike squalene or dFdC, which clearly proved that SQdFdC could permeabilize cellular membranes. Structural X-ray diffraction and calorimetry studies were conducted in order to elucidate the mechanism accounting for these observations. They confirmed that SQdFdC could be transferred from NPs to phospholipid bilayers and that the insertion of the prodrug within model membranes resulted in the formation of nonlamellar structures, which are known to promote membrane leakage. As a whole, our results suggested that due to its amphiphilic nature, the cell uptake of SQdFdC resulted in its insertion into cellular membranes, which could lead to the formation of nonlamellar structures and to membrane permeation. Whether this mechanism could be the source of toxicity in vivo, however, remains to be established, since preclinical studies have clearly proven that squalenoyl gemcitabine displayed a good toxicity profile.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Cell Membrane/drug effects , Deoxycytidine/analogs & derivatives , Prodrugs/pharmacology , Squalene/analogs & derivatives , Surface-Active Agents/pharmacology , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/pharmacokinetics , Calorimetry, Differential Scanning , Cell Culture Techniques , Cell Line, Tumor , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Survival/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/chemistry , Deoxycytidine/pharmacokinetics , Deoxycytidine/pharmacology , Erythrocytes/drug effects , Female , Hemolysis/drug effects , Humans , Mice , Mice, Inbred Strains , Phospholipids/chemistry , Prodrugs/administration & dosage , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Squalene/administration & dosage , Squalene/chemistry , Squalene/pharmacokinetics , Squalene/pharmacology , Surface-Active Agents/administration & dosage , Surface-Active Agents/chemistry , Surface-Active Agents/pharmacokinetics , X-Ray Diffraction
5.
J Control Release ; 147(2): 163-70, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20691740

ABSTRACT

We have designed an amphiphilic prodrug of gemcitabine (dFdC) by its covalent coupling to a derivative of squalene, a natural lipid. The resulting bioconjugate self-assembled spontaneously in water as nanoparticles that displayed a promising in vivo anticancer activity. The aim of the present study was to provide further insight into the in vitro subcellular localization and on the metabolization pathway of the prodrug. Cells treated with radiolabelled squalenoyl gemcitabine (SQdFdC) were studied by differential detergent permeation, and microautography coupled to fluorescent immunolabeling and confocal microscopy. This revealed that the bioconjugate accumulated within cellular membranes, especially in those of the endoplasmic reticulum. Radio-chromatography analysis proved that SQdFdC delivered dFdC directly in the cell cytoplasm. Mass spectrometry studies confirmed that gemcitabine was then either converted into its biologically active triphosphate metabolite or exported from the cells through membrane transporters. To our knowledge, this is the first description of such an intracellular drug delivery pathway. In vitro cytotoxicity assays revealed that SQdFdC was more active than dFdC on a transporter-deficient human resistant leukemia model, which was explained by the subcellular distribution of the drugs and their metabolites. The squalenoylation drug delivery strategy might, therefore, dramatically improve the efficacy of gemcitabine on transporter-deficient resistant cancer in the clinical context.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Cell Membrane/metabolism , Deoxycytidine/analogs & derivatives , Drug Carriers/chemistry , Nanoparticles/chemistry , Prodrugs/pharmacokinetics , Squalene/analogs & derivatives , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacology , Autoradiography , Cell Line, Tumor , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacokinetics , Deoxycytidine/pharmacology , Drug Compounding , Humans , Particle Size , Prodrugs/administration & dosage , Prodrugs/pharmacology , Squalene/administration & dosage , Squalene/pharmacokinetics , Squalene/pharmacology , Subcellular Fractions/metabolism , Surface-Active Agents/chemistry , Tandem Mass Spectrometry , Tissue Distribution , Gemcitabine
6.
Int J Pharm ; 381(2): 140-5, 2009 Nov 03.
Article in English | MEDLINE | ID: mdl-19782881

ABSTRACT

Nucleoside analogues are potent anticancer or antiviral agents that however display some limitations (rapid metabolism, induction of resistance). In order to overcome these drawbacks, we recently proposed new prodrugs, in which nucleoside analogues were covalently coupled to squalene (SQ). The resulting amphiphilic compounds spontaneously formed nanoparticles (NPs) and displayed a promising efficacy both in vitro and in vivo. Since long-term stability is essential for further clinical development we needed to develop a laboratory-scale freeze-drying protocol in order to improve the colloidal stability of those NPs. Squalenoylated gemcitabine (SQdFdC) has been successfully freeze-dried with trehalose (10%, w/w) as a cryoprotectant. Concentrations of SQdFdC up to 4mg/mL after freeze-drying and rehydration have been obtained, which is necessary for in vivo studies. Stability measurements by dynamic light scattering showed that trehalose had a stabilizing effect on SQdFdC NPs, and that freeze-dried SQdFdC NPs could be stored up to four months at room temperature before rehydration, without loss of stability. In vitro cytotoxicity studies on three murine cell lines showed that SQdFdC NPs retained their cytotoxic activity after freeze-drying. We showed that this freeze-drying protocol could also be applied to squalenoylated didanosine (SQddI) and zalcitabine (SQddC). Overall, these results allow for the use of freeze-dried NPs in upcoming preclinical trials of the different squalenoylated compounds developed in our laboratory.


Subject(s)
Antimetabolites/chemistry , Cryoprotective Agents/chemistry , Freeze Drying , Nanoparticles/chemistry , Nucleosides/chemistry , Prodrugs/chemistry , Squalene/analogs & derivatives , Algorithms , Animals , Antimetabolites/pharmacology , Cell Line , Cell Survival/drug effects , Cold Temperature , Colloids , Deoxycytidine/analogs & derivatives , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Dideoxynucleosides/chemistry , Dideoxynucleosides/pharmacology , Drug Stability , Freeze Drying/methods , Inhibitory Concentration 50 , Mice , Nanoparticles/administration & dosage , Nephelometry and Turbidimetry , Nucleosides/pharmacology , Prodrugs/pharmacology , Squalene/chemistry , Squalene/pharmacology , Surface Properties , Time Factors , Trehalose/chemistry
7.
Int J Pharm ; 344(1-2): 62-70, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17616448

ABSTRACT

Didanosine is a polar drug with poor membrane absorption and high hepatic first pass metabolism. This study aimed at developing a lipidic formulation of a glycerolipidic prodrug of didanosine in order to improve its bioavailability. In the course of a preformulation study, the glycerolipidic prodrug of didanosine was characterized by microscopy, DSC and XRDT. In anhydrous conditions, the prodrug displayed a polymorphic behaviour similar to that of triglycerides. Then, we evaluated three types of lipidic formulations (emulsions, mixed micelles and liposomes) in order to encapsulate the prodrug. Solubilities in water - even in the presence of taurocholate micelles - but also in some oils were very low (max 244 microg/mL) as the prodrug was found to be amphiphilic (log P=2). On the contrary, the prodrug was found to be perfectly incorporated in dipalmitoylphosphatidylcholine (DPPC) multilamellar liposomes up to a ratio of 1:5 (mol:mol) prodrug:DPPC as suggested by HPLC-UV and DSC experiments. Moreover, these liposomes could be freeze-dried whereas the chemical integrity of the prodrug was preserved. Then, the freeze-dried liposomal preparation could be formulated as gastro-resistant capsules to prevent didanosine from acidic degradation. Further experiments are on the way to evaluate in vitro the absorption of prodrug incorporated in liposomes by enterocytes.


Subject(s)
Anti-HIV Agents , Didanosine , Prodrugs , 1,2-Dipalmitoylphosphatidylcholine , Administration, Oral , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/chemistry , Calorimetry, Differential Scanning , Chemistry, Pharmaceutical , Didanosine/administration & dosage , Didanosine/chemistry , Drug Stability , Emulsions , Freeze Drying , Liposomes , Lymphatic System , Micelles , Prodrugs/administration & dosage , Prodrugs/chemistry , Triglycerides/chemistry , X-Ray Diffraction
8.
J Phys Chem B ; 111(21): 6042-50, 2007 May 31.
Article in English | MEDLINE | ID: mdl-17488111

ABSTRACT

A new target in AIDS therapy development is HIV-1 integrase (IN). It was proven that HIV-1 IN required divalent metal cations to achieve phosphodiester bond cleavage of DNA. Accordingly, all newly investigated potent IN inhibitors contain chemical fragments possessing a high ability to chelate metal cations. One of the promising leads in the polyhydroxylated styrylquinolines (SQLs) series is (E)-8-hydroxy-2-[2-(4,5-dihydroxy-3-methoxyphenyl)-ethenyl]-7-quinoline carboxylic acid (1). The present study focuses on the quinoline-based progenitor (2), which is actually the most probable chelating part of SQLs. Conventional and synchrotron low-temperature X-ray crystallographic studies were used to investigate the chelating power of progenitor 2. Mg2+ and Cu2+ cations were selected for this purpose, and three types of metal complexes of 2 were obtained: Mg(II) complex (4), Cu(II) complex (5) and mixed Mg(II)-Cu(II) complexes (6 and 7). The analysis of the crystal structure of complex 4 indicates that two tridentate ligands coordinate two Mg2+ cations, both in octahedral geometry. The Mg-Mg distance was found equal to 3.221(1) A, in agreement with the metal-metal distance of 3.9 A encountered in the crystal structure of Escherichia coli DNA polymerase I. In 5, the complex is formed by two bidentate ligands coordinating one copper ion in tetrahedral geometry. Both mixed Mg(II)-Cu(II) complexes, 6 and 7 exhibit an original arrangement of four ligands linked to a central heterometallic cluster consisting of three octahedrally coordinated magnesium ions and one tetrahedrally coordinated copper ion. Quantum mechanics calculations were also carried out in order to display the electrostatic potential generated by the dianionic ligand 2 and complex 4 and to quantify the binding energy (BE) during the formation of the magnesium complex of progenitor 2. A comparison of the binding energies of two hypothetical monometallic Mg(II) complexes with that found in the bimetallic magnesium complex 4 was made.


Subject(s)
Copper/chemistry , HIV Integrase Inhibitors/chemistry , Magnesium/chemistry , Organometallic Compounds/chemistry , Quinolines/chemistry , Computer Simulation , Crystallization , Crystallography, X-Ray , Electrons , HIV Integrase Inhibitors/chemical synthesis , Hydrogen Bonding , Models, Chemical , Models, Molecular , Molecular Structure , Organometallic Compounds/chemical synthesis , Quantum Theory , Static Electricity
9.
Cell Mol Life Sci ; 64(3): 356-64, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17256088

ABSTRACT

Poly(methoxypolyethyleneglycol cyanoacrylate-co-hexadecylcyanoacrylate) (PEG-PHDCA) nanoparticles have demonstrated their capacity to diffuse through the blood-brain barrier after intravenous administration. However, the mechanism of transport of these nanoparticles into brain has not yet been clearly elucidated. The development of a model of rat brain endothelial cells (RBEC) in culture has allowed investigations into this mechanism. A study of the intracellular trafficking of nanoparticles by cell fractionation and confocal microscopy showed that nanoparticles are internalized by the endocytic pathway. Inhibition of the caveolae-mediated pathway by preincubation with filipin and nystatin did not modify the cellular uptake of the nanoparticles. In contrast, chlorpromazine and NaN(3) pretreatment, which interferes with clathrin and energy-dependent endocytosis, caused a significant decrease of nanoparticle internalization. Furthermore, cellular uptake experiments with nanoparticles preincubated with apolipoprotein E and blocking of low-density lipoprotein receptors (LDLR) clearly suggested that the LDLR-mediated pathway was involved in the endocytosis of PEGPHDCA nanoparticles by RBEC.


Subject(s)
Brain/cytology , Brain/metabolism , Cyanoacrylates/metabolism , Endocytosis , Endothelial Cells/metabolism , Nanoparticles , Polyethylene Glycols/metabolism , Receptors, LDL/metabolism , Animals , Apolipoproteins E/pharmacology , Biological Transport/drug effects , Brain/drug effects , Caveolae/metabolism , Cells, Cultured , Clathrin/metabolism , Endocytosis/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Membrane Transport Proteins/metabolism , Rats , Subcellular Fractions/drug effects , Time Factors
10.
Cell Mol Life Sci ; 62(12): 1400-8, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15905957

ABSTRACT

Poly(MePEG2000cyanoacrylate-co-hexadecylcyanoacrylate) (PEG-PHDCA) nanoparticles have demonstrated their capacity to reach the rat central nervous system after intravenous injection. For insight into the transport of colloidal systems across the blood-brain barrier (BBB), we developed a relevant in vitro rat BBB model consisting of a coculture of rat brain endothelial cells (RBECs) and rat astrocytes. The RBECs used in our model displayed and retained structural characteristics of brain endothelial cells, such as expression of P-glycoprotein, occludin and ZO-1, and immunofluorescence studies showed the specific localization of occludin and ZO1. The high values of transendothelial electrical resistance and low permeability coefficients of marker molecules demonstrated the functionality of this model. The comparative passage of polyhexadecylcyanoacrylate and PEG-PHDCA nanoparticles through this model was investigated, showing a higher passage of PEGylated nanoparticles, presumably by endocytosis. This result was confirmed by confocal microscopy. Thanks to a good in vitro/in vivo correlation, this rat BBB model will help in understanding the mechanisms of nanoparticle translocation and in designing new types of colloidal carriers as brain delivery systems.


Subject(s)
Biological Transport , Blood-Brain Barrier/metabolism , Brain/metabolism , Cyanoacrylates/pharmacokinetics , Models, Biological , Polyethylene Glycols/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Astrocytes/metabolism , Capillary Permeability , Drug Delivery Systems , Endothelial Cells/metabolism , Fluorescent Antibody Technique , In Vitro Techniques , Membrane Proteins/metabolism , Nanotechnology , Occludin , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL