Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Medicine (Baltimore) ; 98(21): e15747, 2019 May.
Article in English | MEDLINE | ID: mdl-31124957

ABSTRACT

In chronic kidney disease (CKD), the design of the parenteral nutrition (PN) regimen becomes more challenging where only individualized PN is appropriate, coupled with the increased risk of unintended interactions with diuretic therapy. In an effort to ensure safe therapy in the home, we assessed the physical stability of bespoke PN formulations intended for use in CKD in the simultaneous presence of Y-site compatibility of furosemide and torasemide. The patient's daily needs were determined based on both metabolic demands as well as the demand for fluids.Complete admixtures were subjected to physical stability analysis consisting of visual inspection, a validated light microscope method, pH measurement, zeta potential measurement, and characterization of oily globule size distribution. Y-site compatibility of furosemide and torasemide with the formulated admixtures was also performed.The total parenteral admixture was stable over 7 days at +4°C and 24 h at +25°C and compatible via the Y-line together with furosemide and torasemide over 12 h at +25°C.The stability assessment guarantees the safety and efficiency of home PN with loop diuretics therapy in CKD patients. This means that these patients do not need long hospitalization and they can be safely treated at home. Furthermore, this study proved that torasemide is the same safety diuretic as furosemide, which has a great impact on clinical practice.


Subject(s)
Parenteral Nutrition, Home Total/methods , Renal Insufficiency, Chronic/therapy , Sodium Potassium Chloride Symporter Inhibitors/administration & dosage , Sodium Potassium Chloride Symporter Inhibitors/chemistry , Administration, Intravenous , Drug Incompatibility , Furosemide/administration & dosage , Furosemide/chemistry , Humans , Hydrogen-Ion Concentration , Particle Size , Torsemide/administration & dosage , Torsemide/chemistry
2.
Neuropharmacology ; 143: 186-204, 2018 12.
Article in English | MEDLINE | ID: mdl-30248303

ABSTRACT

Based on the potential role of Na-K-Cl cotransporters (NKCCs) in epileptic seizures, the loop diuretic bumetanide, which blocks the NKCC1 isoforms NKCC1 and NKCC2, has been tested as an adjunct with phenobarbital to suppress seizures. However, because of its physicochemical properties, bumetanide only poorly penetrates through the blood-brain barrier. Thus, concentrations needed to inhibit NKCC1 in hippocampal and neocortical neurons are not reached when using doses (0.1-0.5 mg/kg) in the range of those approved for use as a diuretic in humans. This prompted us to search for a bumetanide derivative that more easily penetrates into the brain. Here we show that bumepamine, a lipophilic benzylamine derivative of bumetanide, exhibits much higher brain penetration than bumetanide and is more potent than the parent drug to potentiate phenobarbital's anticonvulsant effect in two rodent models of chronic difficult-to-treat epilepsy, amygdala kindling in rats and the pilocarpine model in mice. However, bumepamine suppressed NKCC1-dependent giant depolarizing potentials (GDPs) in neonatal rat hippocampal slices much less effectively than bumetanide and did not inhibit GABA-induced Ca2+ transients in the slices, indicating that bumepamine does not inhibit NKCC1. This was substantiated by an oocyte assay, in which bumepamine did not block NKCC1a and NKCC1b after either extra- or intracellular application, whereas bumetanide potently blocked both variants of NKCC1. Experiments with equilibrium dialysis showed high unspecific tissue binding of bumetanide in the brain, which, in addition to its poor brain penetration, further reduces functionally relevant brain concentrations of this drug. These data show that CNS effects of bumetanide previously thought to be mediated by NKCC1 inhibition can also be achieved by a close derivative that does not share this mechanism. Bumepamine has several advantages over bumetanide for CNS targeting, including lower diuretic potency, much higher brain permeability, and higher efficacy to potentiate the anti-seizure effect of phenobarbital.


Subject(s)
Anticonvulsants/pharmacology , Benzylamines/pharmacology , Bumetanide/pharmacology , Phenobarbital/pharmacology , Animals , Anticonvulsants/chemical synthesis , Anticonvulsants/chemistry , Anticonvulsants/pharmacokinetics , Benzylamines/chemical synthesis , Benzylamines/chemistry , Benzylamines/pharmacokinetics , Brain/drug effects , Brain/metabolism , Bumetanide/analogs & derivatives , Bumetanide/chemistry , Bumetanide/pharmacokinetics , Drug Evaluation, Preclinical , Drug Synergism , Epilepsy/drug therapy , Epilepsy/metabolism , Female , Mice , Oocytes , Phenobarbital/pharmacokinetics , Rats, Wistar , Seizures/drug therapy , Seizures/metabolism , Sodium Potassium Chloride Symporter Inhibitors/chemistry , Sodium Potassium Chloride Symporter Inhibitors/pharmacokinetics , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Solute Carrier Family 12, Member 2/metabolism , Tissue Culture Techniques , Xenopus laevis
3.
Anal Chim Acta ; 934: 88-97, 2016 Aug 31.
Article in English | MEDLINE | ID: mdl-27506348

ABSTRACT

A magnetic nanocomposite consisting of nanoparticles-polybutylene terephthalate (MNPs-PBT) was electrospun and used as an extracting medium for an on-line µ-solid phase extraction (µ-SPE)-high performance liquid chromatography (HPLC) set-up with an ultraviolet (UV) detection system. Due to the magnetic property of the prepared nanofibers, the whole extraction procedure was implemented under an external magnetic field to enhance the extraction efficiencies. The developed method along with the synthesized nanocomposite were found to be appropriate for the determination of trace levels of selected drugs including furosemide, naproxen, diclofenac and clobetasol propionate in the urine sample. The prepared MNPs-PBT electrospun nanocomposite was characterized using the scanning electron microscopy (SEM), energy dispersive spectroscopy (EDX) and Fourier transform infrared (FT-IR) spectroscopy. The prepared magnetic fibers showed high porosity, which was another driving force for the extraction efficiency enhancement. Major parameters affecting the extraction efficiency of the selected drugs were optimized. The limits of detections (LOD) of the studied drugs were in the range of 0.4-1.6 µg L(-1) and the limits of quantification (LOQ) were 1-4 µg L(-1) under the optimized conditions. Relative standard deviation (RSD%) for three replicates at three concentration levels of 6, 100 and 400 µg L(-1) were 5.9-8.0% while acceptable linear range with two orders of magnitude was obtained (R(2) = 0.99). The method was validated by the determination of the selected drugs in urine samples and the results indicated that this method has sufficient potential for enrichment and determination of the desired drugs in the urine sample. The relative recovery values were found to be in the range of 78-91%. Implementing the developed on-line µ-SPE method under the external magnetic field induction, led to higher extraction efficiencies for the selected drugs with various diamagnetic properties.


Subject(s)
Anti-Inflammatory Agents/isolation & purification , Nanofibers/chemistry , Polyesters/chemistry , Sodium Potassium Chloride Symporter Inhibitors/isolation & purification , Solid Phase Extraction , Anti-Inflammatory Agents/chemistry , Magnetic Fields , Particle Size , Sodium Potassium Chloride Symporter Inhibitors/chemistry , Solid Phase Extraction/instrumentation , Surface Properties
4.
J Pharm Sci ; 104(12): 4207-4216, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26413799

ABSTRACT

A 1:1 monohydrate salt containing gefitinib, an orally administrated chemotherapy treatment for lung and breast cancers and furosemide, a loop diuretic drug, commonly used in the treatment of hypertension and edema, has been prepared. The molecular salt crystallized in triclinic P-1 space group. The C-O bond lengths (~1.26 Å) in the COOH group show that proton transfer has occurred from furosemide to morpholine moiety of the gefitinib suggesting cocrystal to be ionic. The morpholine moiety of the gefitinib showed significant conformational change because of its involvement in conformation dictating the strong N-H···O hydrogen bonding interaction. The strong hydrogen bonding interaction between gefitinib and furosemide places their benzene rings in stacking mode to facilitate the generation of π-stack dimers. The neighboring dimers are bridged to each other via water molecule through N-H···O, C-H···O, O-H···N, and O-H···O interactions. The remarkable stability of the salt hydrate could be attributed to the strong hydrogen bonding interactions in the crystal structure. Interestingly, release of water from the lattice at 140°C produced new anhydrous salt that has better solubility and dissolution rate than salt hydrate. The drug-drug molecular salt may have some bearing on the treatment of patient suffering from anticancer and hypertension.


Subject(s)
Antineoplastic Agents/chemistry , Furosemide/chemistry , Quinazolines/chemistry , Sodium Chloride/chemistry , Sodium Potassium Chloride Symporter Inhibitors/chemistry , Water/chemistry , Gefitinib , Hydrogen Bonding , Molecular Structure , Solubility
5.
Drug Des Devel Ther ; 9: 4291-302, 2015.
Article in English | MEDLINE | ID: mdl-26273191

ABSTRACT

PURPOSE: Diuretics are the primary treatment for the management of chronic heart failure (HF) symptoms and for the improvement of acute HF symptoms. The rate of delivery to the site of action has been suggested to affect diuretic pharmacodynamics. The main objective of this clinical trial was to explore whether a prolonged release tablet formulation of torasemide (torasemide-PR) was more natriuretically efficient in patients with chronic HF compared to immediate-release furosemide (furosemide-IR) after a single-dose administration. Moreover, the pharmacokinetics of torasemide-PR, furosemide-IR, and torasemide-IR were assessed in chronic HF patients as well as urine pharmacodynamics. METHODS: Randomized, open-label, blinded-endpoint, crossover, and single-dose Phase I clinical trial with three experimental periods. Torasemide-PR and furosemide-IR were administered as a single dose in a crossover fashion for the first two periods, and torasemide-IR 10 mg was administered for the third period. Blood and urine samples were collected at fixed timepoints. The primary endpoint was the natriuretic efficiency after administration of torasemide-PR and furosemide-IR, defined as the ratio between the average drug-induced natriuresis and the average drug recovered in urine over 24 hours. RESULTS: Ten patients were included and nine completed the study. Here, we present the results from nine patients. Torasemide-PR was more natriuretically efficient than furosemide-IR (0.096 ± 0.03 mmol/µg vs 0.015 ± 0.0007 mmol/µg; P < 0.0001). Mictional urgency was lower and more delayed with torasemide-PR than with furosemide-IR. CONCLUSION: In a study with a limited sample size, our results suggest that 10 mg of torasemide-PR is more natriuretically efficient than 40 mg of furosemide-IR after single-dose administration in patients with chronic HF over a 24-hour collection period. Further studies are necessary to evaluate potential pharmacodynamic differences between torasemide formulations and to assess its impact on clinical therapeutics.


Subject(s)
Furosemide/administration & dosage , Heart Failure/drug therapy , Sodium Potassium Chloride Symporter Inhibitors/administration & dosage , Sulfonamides/administration & dosage , Aged , Aged, 80 and over , Chemistry, Pharmaceutical , Chronic Disease , Cross-Over Studies , Delayed-Action Preparations , Furosemide/adverse effects , Furosemide/chemistry , Furosemide/pharmacokinetics , Heart Failure/diagnosis , Heart Failure/physiopathology , Humans , Middle Aged , Natriuresis/drug effects , Sodium Potassium Chloride Symporter Inhibitors/adverse effects , Sodium Potassium Chloride Symporter Inhibitors/chemistry , Sodium Potassium Chloride Symporter Inhibitors/pharmacokinetics , Spain , Sulfonamides/adverse effects , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Time Factors , Torsemide , Treatment Outcome , Urination/drug effects
6.
Pediatr Res ; 77(4): 554-62, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25585037

ABSTRACT

BACKGROUND: Periventricular leukomalacia (PVL) is a major form of preterm brain injury. Na(+)-K(+)-Cl(-) 1 cotransporter (NKCC1) expression on neurons and astrocytes is developmentally regulated and mediates Cl(-) reversal potential. We hypothesized that NKCC1 is highly expressed on oligodendrocytes (OLs) and increases vulnerability to hypoxia-ischemia (HI) mediated white matter injury, and that the NKCC1 inhibitor bumetanide would be protective in a rodent PVL model. METHODS: Immunohistochemistry in Long-Evans rats and PLP-EGFP transgenic mice was used to establish cell-specific expression of NKCC1 in the immature rodent brain. HI was induced on postnatal day 6 (P6) in rats and the protective efficacy of bumetanide (0.3 mg/kg/i.p. q12h × 60 h) established. RESULTS: NKCC1 was expressed on OLs and subplate neurons through the first 2 postnatal weeks, peaking in white matter and the subplate between P3-7. Following HI, NKCC1 is expressed on OLs and neurons. Bumetanide treatment significantly attenuates myelin basic protein loss and neuronal degeneration 7 d post-HI. CONCLUSION: Presence and relative overexpression of NKCC1 in rodent cerebral cortex coincides with a period of developmental vulnerability to HI white matter injury in the immature prenatal brain. The protective efficacy of bumetanide in this model of preterm brain injury suggests that Cl(-) transport is a factor in PVL and that its inhibition may have clinical application in premature human infants.


Subject(s)
Bumetanide/chemistry , Cerebral Cortex/growth & development , Leukomalacia, Periventricular/drug therapy , Sodium Potassium Chloride Symporter Inhibitors/chemistry , Solute Carrier Family 12, Member 2/metabolism , White Matter/drug effects , Animals , Cerebral Cortex/metabolism , Disease Models, Animal , Gene Expression Regulation , Hypoxia/pathology , Ischemia/pathology , Leukomalacia, Periventricular/prevention & control , Male , Mice , Mice, Transgenic , Neurons/metabolism , Oligodendroglia/metabolism , Rats , Rats, Long-Evans
7.
Ann Neurol ; 75(4): 550-62, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24615913

ABSTRACT

OBJECTIVE: There is considerable interest in using bumetanide, a chloride importer Na-K-Cl cotransporter antagonist, for treatment of neurological diseases, such as epilepsy or ischemic and traumatic brain injury, that may involve deranged cellular chloride homeostasis. However, bumetanide is heavily bound to plasma proteins (~98%) and highly ionized at physiological pH, so that it only poorly penetrates into the brain, and chronic treatment with bumetanide is compromised by its potent diuretic effect. METHODS: To overcome these problems, we designed lipophilic and uncharged prodrugs of bumetanide that should penetrate the blood-brain barrier more easily than the parent drug and are converted into bumetanide in the brain. The feasibility of this strategy was evaluated in mice and rats. RESULTS: Analysis of bumetanide levels in plasma and brain showed that administration of 2 ester prodrugs of bumetanide, the pivaloyloxymethyl (BUM1) and N,N-dimethylaminoethylester (BUM5), resulted in significantly higher brain levels of bumetanide than administration of the parent drug. BUM5, but not BUM1, was less diuretic than bumetanide, so that BUM5 was further evaluated in chronic models of epilepsy in mice and rats. In the pilocarpine model in mice, BUM5, but not bumetanide, counteracted the alteration in seizure threshold during the latent period. In the kindling model in rats, BUM5 was more efficacious than bumetanide in potentiating the anticonvulsant effect of phenobarbital. INTERPRETATION: Our data demonstrate that the goal of designing bumetanide prodrugs that specifically target the brain is feasible and that such drugs may resolve the problems associated with using bumetanide for treatment of neurological disorders.


Subject(s)
Brain/drug effects , Bumetanide/therapeutic use , Epilepsy/drug therapy , Serum/drug effects , Sodium Potassium Chloride Symporter Inhibitors/therapeutic use , Action Potentials/drug effects , Animals , Animals, Newborn , Brain/cytology , Brain/metabolism , Bumetanide/chemistry , Bumetanide/pharmacology , Convulsants/toxicity , Disease Models, Animal , Diuretics/pharmacology , Dogs , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Epilepsy/chemically induced , Humans , In Vitro Techniques , Mice , Neurons/drug effects , Pentylenetetrazole/toxicity , Phenobarbital/therapeutic use , Rats , Rats, Sprague-Dawley , Rats, Wistar , Sodium Potassium Chloride Symporter Inhibitors/chemistry , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Species Specificity , Time Factors
8.
Br J Pharmacol ; 171(1): 265-78, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24117047

ABSTRACT

BACKGROUND AND PURPOSE: Loop diuretics are widely used to inhibit the Na(+), K(+), 2Cl(-) co-transporter, but they also inhibit the cystic fibrosis transmembrane conductance regulator (CFTR) Cl(-) channel. Here, we investigated the mechanism of CFTR inhibition by loop diuretics and explored the effects of chemical structure on channel blockade. EXPERIMENTAL APPROACH: Using the patch-clamp technique, we tested the effects of bumetanide, furosemide, piretanide and xipamide on recombinant wild-type human CFTR. KEY RESULTS: When added to the intracellular solution, loop diuretics inhibited CFTR Cl(-) currents with potency approaching that of glibenclamide, a widely used CFTR blocker with some structural similarity to loop diuretics. To begin to study the kinetics of channel blockade, we examined the time dependence of macroscopic current inhibition following a hyperpolarizing voltage step. Like glibenclamide, piretanide blockade of CFTR was time and voltage dependent. By contrast, furosemide blockade was voltage dependent, but time independent. Consistent with these data, furosemide blocked individual CFTR Cl(-) channels with 'very fast' speed and drug-induced blocking events overlapped brief channel closures, whereas piretanide inhibited individual channels with 'intermediate' speed and drug-induced blocking events were distinct from channel closures. CONCLUSIONS AND IMPLICATIONS: Structure-activity analysis of the loop diuretics suggests that the phenoxy group present in bumetanide and piretanide, but absent in furosemide and xipamide, might account for the different kinetics of channel block by locking loop diuretics within the intracellular vestibule of the CFTR pore. We conclude that loop diuretics are open-channel blockers of CFTR with distinct kinetics, affected by molecular dimensions and lipophilicity.


Subject(s)
Chlorides/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Animals , Bumetanide/pharmacology , Cricetinae , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Dose-Response Relationship, Drug , Furosemide/pharmacology , Humans , Kinetics , Membrane Potentials , Mice , Molecular Structure , Rats , Sodium Potassium Chloride Symporter Inhibitors/chemistry , Structure-Activity Relationship , Sulfonamides/pharmacology , Xipamide/pharmacology
9.
ACS Appl Mater Interfaces ; 5(22): 12127-34, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24175755

ABSTRACT

A major challenge for a drug-delivery system is to engineer stable drug carriers with excellent biocompatibility, monodisperse size, and controllable release profiles. In this study, we used a microfluidic technique to encapsulate thermally hydrocarbonized porous silicon (THCPSi) microparticles within solid lipid microparticles (SLMs) to overcome the drawbacks accompanied by THCPSi microparticles. Formulation and process factors, such as lipid matrixes, organic solvents, emulsifiers, and methods to evaporate the organic solvents, were all evaluated and optimized to prepare monodisperse stable SLMs. FTIR analysis together with confocal images showed the clear deposition of THCPSi microparticles inside the monodisperse SLM matrix. The formation of monodisperse THCPSi-solid lipid microcomposites (THCPSi-SLMCs) not only altered the surface hydrophobicity and morphology of THCPSi microparticles but also remarkably enhanced their cytocompatibility with intestinal (Caco-2 and HT-29) cancer cells. Regardless of the solubility of the loaded therapeutics (aqueous insoluble, fenofibrate and furosemide; aqueous soluble, methotrexate and ranitidine) and the pH values of the release media (1.2, 5.0, and 7.4), the time for the release of 50% of the payloads from THCPSi-SLMC was at least 1.3 times longer than that from the THCPSi microparticles. The sustained release of both water-soluble and -insoluble drugs together with a reduced burst-release effect from monodisperse THCPSi-SLMC was achieved, indicating the successful encapsulation of THCPSi microparticles into the SLM matrix. The fabricated THCPSi-SLMCs exhibited monodisperse spherical morphology, enhanced cytocompatibility, and prolonged both water-soluble and -insoluble drug release, which makes it an attractive controllable drug-delivery platform.


Subject(s)
Drug Delivery Systems , Fenofibrate/chemistry , Furosemide/chemistry , Hypolipidemic Agents/chemistry , Lipids/chemistry , Microfluidic Analytical Techniques , Nanocomposites/chemistry , Silicon/chemistry , Sodium Potassium Chloride Symporter Inhibitors/chemistry , Caco-2 Cells , Fenofibrate/pharmacology , Furosemide/pharmacology , Humans , Hypolipidemic Agents/pharmacology , Lipids/pharmacology , Porosity , Silicon/pharmacology , Sodium Potassium Chloride Symporter Inhibitors/pharmacology
10.
J Control Release ; 169(1-2): 150-61, 2013 Jul 10.
Article in English | MEDLINE | ID: mdl-23570985

ABSTRACT

The aim of the present study was to develop and evaluate a novel drug solubilization platform (so-called solid nanodispersion) prepared by a simple co-grinding and solvent-free process. Using structurally diverse model compounds from the Pfizer drug library, including ingliforib, furosemide and celecoxib, we successfully prepared stable solid nanodispersions (SNDs) without the use of solvent or heat. Stable colloidal particles (<350 nm) containing drug, polyvinylpyrrolidone (PVP) K12 and sodium dodecyl sulfate (SDS) in 1:2.75:0.25 ratio were produced after 2 h of co-grinding. The composition and particle size of SNDs were optimized by varying the grinding media size, powder-to-grinding media ratio, milling speed and milling time. The resulting formulations contained crystalline drug and were stable at room temperature for over one month. Greater than 80% of the drug was released from the SND in less than 30 min, with sustained supersaturation over 4 h. Using furosemide (BCS class IV compound) as a model compound, we conducted transport studies with Madin-Darby canine kidney cells transfected with human MDR1 gene (MDCK/MDR1), followed by pharmacokinetics studies in rats. Results showed that the SND formulation enhanced the absorptive flux of furosemide by more than 3-fold. In the pharmacokinetics studies, the SND formulation increased C(max) and AUC of furosemide by 36.6 and 43.2 fold respectively, relative to Methocel formulation. Interestingly, physical mixture containing furosemide, PVP K12 and SDS produced a similar level of oral exposure as the SNDs, albeit with a longer T(max) than the SND formulation. The results suggest that PVP K12 and SDS were able to increase the furosemide free fraction available for oral absorption. Low solubility, poor permeability, and high first-pass effect of furosemide may also have produced the effect that small improvements in solubilization resulted in significant potentiation of the oral exposure of the physical mixture. However the use of a physical mixture of drug, polymer and surfactant, to increase drug bioavailability cannot be generalized to all drugs. There are only a few reported cases of such phenomenon. While SNDs may not be the only option to solubilize compounds in every case, SNDs are expected to be applicable to a broader chemical space of pharmaceutical compounds compared to a physical mixture. Ultimately, the formulation scientist will have to exercise judgment in choosing the appropriate formulation strategy for the compound of interest. SNDs represent a significant improvement over current enabling technologies such as nanocrystal and spray-dried dispersion technologies, in that SNDs are simple, do not require solvent or heat, are applicable to a structurally diverse chemical space, and are readily amenable to the development of solid dosage forms.


Subject(s)
Drug Compounding/instrumentation , Furosemide/pharmacokinetics , Sodium Potassium Chloride Symporter Inhibitors/pharmacokinetics , Administration, Oral , Animals , Cell Line , Dogs , Equipment Design , Furosemide/administration & dosage , Furosemide/chemistry , Humans , Male , Particle Size , Rats , Rats, Sprague-Dawley , Sodium Potassium Chloride Symporter Inhibitors/administration & dosage , Sodium Potassium Chloride Symporter Inhibitors/chemistry , Solubility
11.
Pak J Pharm Sci ; 25(4): 815-22, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23009999

ABSTRACT

To determine the effect of chitosan, starch powder, polyvinylpyrrolidone (PVP), Avicel PH 101 powder, Avicel PH 102 granules as a function of different concentrations on the solubility, disintegration and hence dissolution of furosemide from immediate release tablet dosage forms. The tablets were prepared by the wet granulation method and evaluated for hardness, friability, disintegration and in vitro dissolution. Chitosan 7% w/w showed the fastest disintegration of furosemide tablets among the other disintegrants studied. This was attributed to its highest swelling properties and velocity constant of water uptake. The step of adding chitosan during tablet preparation had a great effect on the physical properties and dissolution profiles of the prepared tablets with external addition of chitosan showed best results compared to best results comparing to internal-external or internal addition. The most appropriate force of compression was 4ton/cm(2). The selected formula F15 containing 7% w/w chitosan was successful and showed a high significant (p<0.001) enhancement in disintegration and dissolution behaviors of furosemide tablets in comparison with the commercially available Furosemide ® tablets. These results were supported by the simulated data where F15 formula showed the highest plasma concentration C-max 1.89mcg/mL after 0.5 hr compared to C-max 1.05mcg/mL after 1hr for the reference. The present study demonstrated that chitosan is a very good candidate to be used as a tablet disintegrant and was able to enhance the dissolution of poorly absorbable drugs.


Subject(s)
Chitosan/chemistry , Computer Simulation , Excipients/chemistry , Furosemide/chemistry , Furosemide/pharmacokinetics , Models, Biological , Sodium Potassium Chloride Symporter Inhibitors/chemistry , Sodium Potassium Chloride Symporter Inhibitors/pharmacokinetics , Administration, Oral , Biological Availability , Cellulose/chemistry , Chemistry, Pharmaceutical , Furosemide/administration & dosage , Furosemide/blood , Hardness , Hardness Tests , Povidone/chemistry , Powders , Pressure , Sodium Potassium Chloride Symporter Inhibitors/administration & dosage , Sodium Potassium Chloride Symporter Inhibitors/blood , Solubility , Starch/chemistry , Tablets , Technology, Pharmaceutical/methods
12.
Org Lett ; 14(5): 1258-61, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22360311

ABSTRACT

Tripartilactam, a structurally unprecedented cyclobutane-bearing tricyclic lactam metabolite, was discovered from Streptomyces sp. isolated from a brood ball of the dung beetle, Copris tripartitus. The structure of this compound was elucidated by the combination of NMR, MS, UV, and IR spectroscopy and multistep chemical derivatization. Tripartilactam was evaluated as a Na(+)/K(+) ATPase inhibitor (IC(50) = 16.6 µg/mL).


Subject(s)
Coleoptera/microbiology , Cyclobutanes/chemistry , Feces/microbiology , Lactams/chemistry , Sodium Potassium Chloride Symporter Inhibitors/chemistry , Streptomyces/chemistry , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cyclobutanes/isolation & purification , Cyclobutanes/pharmacology , Humans , Lactams/isolation & purification , Lactams/pharmacology , Molecular Structure , Sodium Potassium Chloride Symporter Inhibitors/isolation & purification , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Streptomyces/isolation & purification
13.
Nucleosides Nucleotides Nucleic Acids ; 30(12): 1302-11, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22132991

ABSTRACT

Uric acid (urate) is the end product of purine metabolism in humans. Human kidneys reabsorb a large proportion of filtered urate. This extensive renal reabsorption, together with the fact that humans do not possess uricase that catalyzes the biotransformation of urate into allantoin, results in a higher plasma urate concentration in humans compared to other mammals. A major determinant of plasma urate concentration is renal excretion as a function of the balance between reabsorption and secretion. We previously identified that renal urate absorption in proximal tubular epithelial cells occurs mainly via apical urate/anion exchanger, URAT1/SLC22A12, and by facilitated diffusion along the trans-membrane potential gradient by the basolateral voltage-driven urate efflux transporter, URATv1/SLC2A9/GLUT9. In contrast, the molecular mechanism by which renal urate secretion occurs remains elusive. Recently, we reported a newly characterized human voltage-driven drug efflux transporter, hNPT4/SLC17A3, which functions as a urate exit pathway located at the apical side of renal proximal tubules. This transporter protein has been hypothesized to play an important role with regard to net urate efflux. An in vivo role of hNPT4 is supported by the fact that missense mutations in SLC17A3 present in hyperuricemia patients with urate underexcretion abolished urate efflux capacity in vitro. Herein, we report data demonstrating that loop diuretics and thiazide diuretics substantially interact with hNPT4. These data provide molecular evidence for loop and thiazide-diuretics-induced hyperuricemia. Thus, we propose that hNPT4 is an important transepithelial proximal tubular transporter that transports diuretic drugs and operates functionally with basolateral organic anion transporters 1/3 (OAT1/OAT3).


Subject(s)
Cell Polarity , Electricity , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Organic Anion Transporters/metabolism , Animals , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Organic Anion Transporters/genetics , Sodium Potassium Chloride Symporter Inhibitors/chemistry
14.
J Pharm Pharmacol ; 58(11): 1475-82, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17132210

ABSTRACT

The biopharmaceutical properties of an in-house developed new crystal modification of torasemide (Torasemide N) were investigated in comparison with the most well known crystal modification form of torasemide (Torasemide I) in order to classify the drug according to the Biopharmaceutics Classification System (BCS), and to evaluate the data in line with current US Food and Drug Administration (FDA) guidance (with biowaiver provision for Class I drugs) to determine if the biowaiver provision could be improved. The solubility profiles of Torasemide I and Torasemide N were determined, and tablets prepared from both forms of the drug were studied for in-vitro release characteristics in media recommended by the current FDA guidance for biowaiver of generic products, and in other media considered more appropriate for the purpose than the ones recommended by the FDA. Two separate bioequivalence studies in healthy humans (following oral administration) were performed with two test products (both prepared from Torasemide I) against a single reference product (prepared from Torasemide N). The absorption profiles of the drug from the tablets were determined by deconvolution for comparison with the in-vitro release profiles to determine the appropriateness of some dissolution media for predicting in-vivo performance and to determine the comparative rate and extent of absorption. The drug was absorbed from the tested products quickly and almost completely (about 95% within 3.5 h of administration). However, one test product failed to meet the bioequivalence criteria and had a significant initial lower absorption rate profile compared with the reference product (P< or =0.05), whereas the other product was bioequivalent and had a similar absorption profile to the reference product. A dissolution medium at pH 5.0, in which torasemide has minimum solubility, was found to be more discriminatory than the media recommended by the FDA. Torasemide has been classified as a Class I drug according to the BCS up to a maximum dose of 40 mg and the data suggest that the current FDA guidance could be improved by giving more emphasis to selection of appropriate dissolution media than is given in its current form for approving biowaiver to generic products of Class I drugs.


Subject(s)
Drugs, Generic/classification , Sulfonamides/classification , United States Food and Drug Administration/standards , Area Under Curve , Biological Availability , Crystallization , Drug Approval , Drugs, Generic/chemistry , Drugs, Generic/pharmacokinetics , Guidelines as Topic/standards , Humans , Hydrogen-Ion Concentration , Molecular Structure , Reference Standards , Sodium Potassium Chloride Symporter Inhibitors/chemistry , Sodium Potassium Chloride Symporter Inhibitors/classification , Sodium Potassium Chloride Symporter Inhibitors/pharmacokinetics , Solubility , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Tablets , Therapeutic Equivalency , Torsemide , United States , United States Food and Drug Administration/legislation & jurisprudence
SELECTION OF CITATIONS
SEARCH DETAIL
...