Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Nitric Oxide ; 104-105: 1-10, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32771473

ABSTRACT

Nitric oxide (NO) deficiency is often associated with several acute and chronic diseases. NO donors and especially S-nitrosothiols such as S-nitrosoglutathione (GSNO) have been identified as promising therapeutic agents. Although their permeability through the intestinal barrier have recently be proved, suitable drug delivery systems have to be designed for their oral administration. This is especially challenging due to the physico-chemical features of these drugs: high hydrophilicity and high lability. In this paper, three types of particles were prepared with an Eudragit® polymer: nanoparticles and microparticles obtained with a water-in-oil-in-water emulsion/evaporation process versus microparticles obtained with a solid-in-oil-in-water emulsion/evaporation process. They had a similar encapsulation efficiency (around 30%), and could be freeze-dried then be stored at least one month without modification of their critical attributes (size and GSNO content). However, microparticles had a slightly slower in vitro release of GSNO than nanoparticles, and were able to boost by a factor of two the drug intestinal permeability (Caco-2 model). Altogether, this study brings new data about GSNO intestinal permeability and three ready-to-use formulations suitable for further preclinical studies with oral administration.


Subject(s)
Drug Carriers/chemistry , Nanoparticles/chemistry , Nitric Oxide Donors/pharmacology , S-Nitrosoglutathione/pharmacology , Caco-2 Cells , Drug Carriers/toxicity , Drug Compounding , Drug Liberation , Drug Stability , Freeze Drying , Humans , Intestinal Mucosa/metabolism , Nanoparticles/toxicity , Nitric Oxide/metabolism , Nitric Oxide Donors/chemistry , Nitric Oxide Donors/toxicity , Particle Size , S-Nitrosoglutathione/chemistry , S-Nitrosoglutathione/toxicity
2.
Rapid Commun Mass Spectrom ; 33(1): 1-11, 2019 Jan 15.
Article in English | MEDLINE | ID: mdl-30248720

ABSTRACT

RATIONALE: The potency of S-nitrosoglutathione (GSNO) as a nitric oxide (NO) donor to treat cardiovascular diseases (CVDs) has been highlighted in numerous studies. In order to study its bioavailability after oral administration, which represents the most convenient route for the chronic treatment of CVDs, it is essential to develop an analytical method permitting (i) the simultaneous measurement of GSNO metabolites, i.e. nitrite, S-nitrosothiols (RSNOs) and nitrate and (ii) to distinguish them from other sources (endogenous synthesis and diet). METHODS: Exogenous GSNO was labeled with 15 N, and the GS15 NO metabolites after conversion into the nitrite ion were derivatized with 2,3-diaminonaphthalene. The resulting 2,3-naphthotriazole was quantified by liquid chromatography/tandem ion trap mass spectrometry (LC/ITMS/MS) in multiple reaction monitoring mode after Higher-energy Collision-induced Dissociation (HCD). Finally, the validated method was applied to an in vitro model of the intestinal barrier (monolayer of Caco-2 cells) to study GS15 NO intestinal permeability. RESULTS: A LC/ITMS/MS method based on an original transition (m/z 171 to 156) for sodium 15 N-nitrite, GS15 NO and sodium 15 N-nitrate measurements was validated, with recoveries of 100.8 ± 3.8, 98.0 ± 2.7 and 104.1 ± 3.3%, respectively. Intra- and inter-day variabilities were below 13.4 and 12.6%, and the limit of quantification reached 5 nM (signal over blank = 4). The permeability of labeled GS15 NO (10-100 µM) was evaluated by calculating its apparent permeability coefficient (Papp ). CONCLUSIONS: A quantitative LC/ITMS/MS method using HCD was developed for the first time to selectively monitor GS15 NO metabolites. The assay allowed evaluation of GS15 NO intestinal permeability and situated this drug candidate within the middle permeability class according to FDA guidelines. In addition, the present method has opened the perspective of a more fundamental work aiming at studying the fragmentation mechanism leading to the ion at m/z 156 in HCD tandem mass spectrometry in the presence of acetonitrile.


Subject(s)
Chromatography, Liquid/methods , Nitric Oxide/analysis , Nitric Oxide/metabolism , S-Nitrosoglutathione/pharmacokinetics , Tandem Mass Spectrometry/methods , 2-Naphthylamine/analogs & derivatives , 2-Naphthylamine/chemistry , Caco-2 Cells , Humans , Intestinal Absorption/drug effects , Limit of Detection , Nitrites/chemistry , Reproducibility of Results , S-Nitrosoglutathione/metabolism , Tandem Mass Spectrometry/instrumentation
3.
Drug Deliv ; 23(8): 2927-2935, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26666370

ABSTRACT

S-nitrosothiols are a class of NO-donors currently under investigation for the treatment of various diseases. In this study, we developed a novel NO-donor (S-nitrosoglutathione-alginate, SNA) by cross-linking alginate with S-nitrosothiols, which can deliver NO in a sustained manner. This compound can be further evaluated for oral delivery to treat Crohn's disease. This new compound was prepared using a two-step procedure involving (I) linkage of reduced glutathione to alginate and (II) post-nitrosation with sodium nitrite (NaNO2). The amount of linked thiol moieties for the possible nitrosation was calculated using Ellman's method, and the amount of NO abducted on the polymer was calculated using the Griess-Saville method. An ex vivo model (i.e. Ussing chamber) was used to investigate the permeation of this new NO-donor across the rat intestinal barrier. We obtained polymers with different numbers of abducted NOs (174 ± 21 µmol/g for SNA F1 and 468 ± 23 µmol/g for SNA F2) depending on the procedure used for nitrosation. In the ex vivo studies in the Ussing chamber, SNA F2 exhibited a sustained release for at least 10 h. The effect of pH on the stability of the new compound was also investigated, and the new compound was more stable at a mildly basic pH of 8.4 where 73% remained after 1 week. However, only 50% remained after 1 week at an acidic pH of 1.2. In the cytotoxicity studies (Caco2), this compound was nontoxic at concentrations of less than 200 µM.


Subject(s)
Alginates/chemistry , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/chemistry , Intestinal Mucosa/metabolism , Nitric Oxide/administration & dosage , Nitric Oxide/chemistry , S-Nitrosoglutathione/chemistry , Animals , Caco-2 Cells , Cell Line, Tumor , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Humans , Male , Nitrosation , Polymers/chemistry , Rats , Rats, Wistar , S-Nitrosothiols/chemistry , Sulfhydryl Compounds/chemistry
4.
Int J Pharm ; 495(1): 354-361, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26319632

ABSTRACT

S-nitrosoglutathione (GSNO) is a nitric oxide (NO) donor with therapeutic potential for cardiovascular disease treatment. Chronic oral treatment with GSNO is limited by high drug sensitivity to the environment and limited oral bioavailability, requiring the development of delivery systems able to sustain NO release. The present work describes new platforms based on polymer nanocomposite particles for the delivery of GSNO. Five types of optimized nanocomposite particles have been developed (three based on chitosan, two based on alginate sodium). Those nanocomposite particles encapsulate GSNO with high efficiency from 64% to 70% and an average size of 13 to 61 µm compatible with oral delivery. Sustained release of GSNO in vitro was achieved. Indeed, chitosan nanocomposites discharged their payload within 24h; whereas alginate nanocomposites released GSNO more slowly (10% of GSNO was still remaining in the dosage form after 24h). Their cytocompatibility toward intestinal Caco-2 cells (MTT assay) was acceptable (IC50: 6.07 ± 0.07-9.46 ± 0.08 mg/mL), demonstrating their suitability as oral delivery systems for GSNO. These delivery systems presented efficient GSNO loading and sustained release as well as cytocompatibility, showing their promise as a means of improving the oral bioavailability of GSNO and as a potential new treatment.


Subject(s)
Alginates/chemistry , Chitosan/chemistry , Nanocomposites/chemistry , Nitric Oxide Donors/administration & dosage , S-Nitrosoglutathione/administration & dosage , Administration, Oral , Caco-2 Cells , Chemistry, Pharmaceutical/methods , Delayed-Action Preparations , Drug Carriers/chemistry , Drug Liberation , Drug Stability , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Humans , Particle Size
5.
J Biomed Mater Res A ; 103(10): 3303-11, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25809572

ABSTRACT

Fabrication of scaffolds loaded with nitric oxide (NO) donors (S-nitrosoglutathione, GSNO, and isosorbide mononitrate, ISMN) with suitable cell compatibility and optimized properties for tissue-engineering applications is reported using "in situ" technique. Based on FDA-approved polymer, solvent and dosage forms, this gentle process allowed the incorporation of the GSNO labile drug into scaffolds made of either poly(lactide-co-glycolide) (PLGA) or PLGA/poly(ɛ-caprolactone) (PCL) blend. During scaffolds manufacturing process including washing cycles, NO donors were leached from scaffolds. However, GSNO and ISMN concentrations in the last washing medium (10(-7) M and 10(-4) M, respectively) were in the range of cell suitability for tissue engineering. Further morphological analyses indicated that smoother surfaces with fewer but bigger pores (compatible with cell penetration and ingrowth) were obtained with PLGA in comparison with PLGA/PCL scaffolds. Among all tested matrices, only unloaded PLGA and GSNO-loaded PLGA/PCL exhibited intermediate cell anchorage, with mitochondrial activity close to the control and an increase in protein content, a prognostic for scaffold cell colonization, defining them as promising candidates. Deeper analyses of these two scaffolds looking at intracellular redox balance through reactive oxygen species production, glutathione, S-nitrosothiols, and nitrite ions content exhibited GSNO-loaded PLGA/PCL as the best of all tested 3D scaffolds for tissue engineering.


Subject(s)
Drug Implants , Myocytes, Smooth Muscle/metabolism , Nitric Oxide , S-Nitrosoglutathione , Tissue Scaffolds/chemistry , Animals , Drug Implants/chemistry , Drug Implants/pharmacokinetics , Drug Implants/pharmacology , Lactic Acid/chemistry , Lactic Acid/pharmacokinetics , Lactic Acid/pharmacology , Myocytes, Smooth Muscle/cytology , Nitric Oxide/chemistry , Nitric Oxide/pharmacokinetics , Nitric Oxide/pharmacology , Polyesters/chemistry , Polyesters/pharmacokinetics , Polyesters/pharmacology , Polyglycolic Acid/chemistry , Polyglycolic Acid/pharmacokinetics , Polyglycolic Acid/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , S-Nitrosoglutathione/chemistry , S-Nitrosoglutathione/pharmacokinetics , S-Nitrosoglutathione/pharmacology
6.
Eur J Pharm Biopharm ; 89: 1-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25448077

ABSTRACT

Physiological S-nitrosothiols (RSNO), such as S-nitrosoglutathione (GSNO), can be used as nitric oxide (NO) donor for the treatment of vascular diseases. However, despite a half-life measured in hours, the stability of RSNO, limited by enzymatic and non-enzymatic degradations, is too low for clinical application. So, to provide a long-lasting effect and to deliver appropriate NO concentrations to target tissues, RSNO have to be protected. RSNO encapsulation is an interesting response to overcome degradation and provide protection. However, RSNO such as GSNO raise difficulties for encapsulation due to its hydrophilic nature and the instability of the S-NO bound during the formulation process. To our knowledge, the present study is the first description of the direct encapsulation of GSNO within polymeric nanoparticles (NP). The GSNO-loaded NP (GSNO-NP) formulated by a double emulsion process, presented a mean diameter of 289 ± 7 nm. They were positively charged (+40 mV) due to the methacrylic acid and ethylacrylate polymer (Eudragit® RL) used and encapsulated GSNO with a satisfactory efficiency (i.e. 54% or 40 mM GSNO loaded in the NP). In phosphate buffer (37 °C; pH 7.4), GSNO-NP released 100% of encapsulated GSNO within 3h and remained stable still 6h. However, in contact with smooth muscle cells, maximum protein nitrosation (a marker of NO bioavailability) was delayed from 1h for free GSNO to 18h for GSNO-NP. Therefore, protection and sustained release of NO were achieved by the association of a NO donor with a drug delivery system (such as polymeric NP), providing opportunities for vascular diseases treatment.


Subject(s)
Nanoparticles/administration & dosage , Nitrosation/drug effects , Polymers/pharmacology , Protein S/metabolism , S-Nitrosoglutathione/pharmacology , Animals , Cell Line , Drug Delivery Systems/methods , Half-Life , Nanoparticles/chemistry , Nitric Oxide/metabolism , Nitric Oxide Donors/chemistry , Nitric Oxide Donors/pharmacology , Polymers/chemistry , Rats , S-Nitrosoglutathione/chemistry , S-Nitrosothiols/metabolism
7.
Anesthesiology ; 106(5): 977-83, 2007 May.
Article in English | MEDLINE | ID: mdl-17457129

ABSTRACT

BACKGROUND: Epinephrine and more recently arginine vasopressin (AVP) alone or in combination have been proposed in patients with anaphylactic shock, but few experimental data exist. The authors investigated the effects of epinephrine only, AVP only, or epinephrine followed by AVP in a model of anaphylactic shock. METHODS: Ovalbumin-sensitized Brown Norway rats were anesthetized, intubated, and shock induced with ovalbumin. Rats (n = 6/group) were randomly allocated to receive 5 min after shock onset: (1) saline (no-treatment group); (2) two boluses of epinephrine followed by continuous infusion (epinephrine group); (3) AVP bolus followed by continuous infusion (AVP group); (4) epinephrine bolus followed by AVP continuous infusion (epinephrine + AVP group). Mean arterial pressure (MAP) and skeletal muscle oxygen pressure (PtiO2) were measured. Continuous infusion rates were titrated to reach MAP values of 60 mmHg. Survival was analyzed. RESULTS: Without treatment, MAP and PtiO2 decreased rapidly with 0% survival. In the epinephrine group, MAP and PtiO2 recovered after an initial decrease, with 84% survival. In the AVP group, MAP was partially restored and subsequently decreased; PtiO2 values decreased to values similar to those in the no-treatment group; survival was 0%. In the epinephrine + AVP group, MAP and PtiO2 values increased more slowly as compared with the epinephrine group; survival was 100%. CONCLUSIONS: In this model of anaphylactic shock, early treatment with epinephrine followed by continuous epinephrine or vasopressin infusion resulted in an excellent survival rate, whereas vasopressin only resulted in a 100% death rate. These experimental results suggest that epinephrine must still be considered as the first-line drug to treat anaphylactic shock.


Subject(s)
Anaphylaxis/drug therapy , Arginine Vasopressin/therapeutic use , Epinephrine/therapeutic use , Anaphylaxis/mortality , Animals , Arginine Vasopressin/administration & dosage , Blood Pressure/drug effects , Drug Therapy, Combination , Epinephrine/administration & dosage , Infusions, Intravenous , Rats , Rats, Inbred BN , Survival Rate , Weight Loss
8.
Int J Pharm ; 306(1-2): 107-21, 2005 Dec 08.
Article in English | MEDLINE | ID: mdl-16253447

ABSTRACT

Melarsoprol, a water-insoluble drug, is mainly used in the treatment of trypanosomiasis and has demonstrated an in vitro activity on myeloid and lymphoid leukemia derived cell lines. It is marketed as a very poorly tolerated non-aqueous solution (Arsobal). The aim of our work was to develop melarsoprol-cyclodextrin complexes in order to improve the tolerability and the bioavailability of melarsoprol. Phase-solubility analysis showed A(L)-type diagrams with beta-cyclodextrin (betaCD), randomly methylated beta-cyclodextrin (RAMEbetaCD) and hydroxypropyl-beta-cyclodextrin (HPbetaCD), which suggested the formation of 1:1 inclusion complexes. The solubility enhancement factor of melarsoprol (solubility in 250 mM of cyclodextrin/solubility in water) was about 7.2x10(3) with both beta-cyclodextrin derivatives. The 1:1 stoichiometry was confirmed in the aqueous solutions by the UV spectrophotometer using Job's plot method. The apparent stability constants K(1:1), calculated from mole-ratio titration plots, were 57 143+/-4 425M(-1) for RAMEbetaCD and 50 761+/-5 070 M(-1) for HPbetaCD. Data from 1H-NMR and ROESY experiments provided a clear evidence of inclusion complexation of melarsoprol with its dithiaarsane extremity inserted into the wide rim of the cyclodextrin torus. Moreover, RAMEbetaCD had a pronounced effect on the drug hydrolysis and the dissolution rate of melarsoprol. However, the cytotoxic properties of melarsoprol on K562 and U937 human leukemia cell lines was not modified by complexation.


Subject(s)
Antineoplastic Agents/chemistry , Cyclodextrins/chemistry , Melarsoprol/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cyclodextrins/pharmacology , Drug Compounding , Drug Stability , Humans , Kinetics , Magnetic Resonance Spectroscopy , Melarsoprol/pharmacology , Molecular Structure , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL
...