Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 72
1.
J Sep Sci ; 44(11): 2301-2309, 2021 Jun.
Article En | MEDLINE | ID: mdl-33783965

Covalent organic nanospheres are new kind of nanospherical polymer with large specific surface area, uniform morphology, and excellent chemical and thermal stability. This material can be fabricated by a facile and rapid room temperature solution-phase strategy. In this work, magnetic nanoparticles were attached to the surface of covalent organic nanospheres, and the obtained composites were used for the extraction of blood lipid regulators such as clofibrate and fenofibrate. These composites were characterized with Fourier-transformed infrared spectroscopy, X-ray photoelectron spectroscopy, and transmission electron microscopy. Several parameters that might affect the extraction efficiency including acetonitrile content, pH value, extraction time, and sample volume were investigated. Under optimum conditions, the proposed analytical method showed high extraction efficiency toward clofibrate and fenofibrate with enrichment factors between 60 and 83. This method exhibited outstanding analytical performance with wide linear range and excellent reproducibility and had low limits of detection in the range of 0.02-0.03 ng/mL. This method was also applied to the detection of clofibrate and fenofibrate in lake water samples, and good recoveries in the range of 92.6-112.6% was obtained.


Clofibrate/isolation & purification , Fenofibrate/isolation & purification , Water Pollutants, Chemical/chemistry , Clofibrate/blood , Clofibrate/chemistry , Fenofibrate/blood , Fenofibrate/chemistry , Lakes , Magnetite Nanoparticles/chemistry , Nanospheres/chemistry
2.
Pharm Res ; 37(3): 47, 2020 Feb 03.
Article En | MEDLINE | ID: mdl-32016597

PURPOSE: To evaluate the role of supersaturation in the in vivo absorption of fenofibrate (FFB), after oral administration in a medium-chain lipid-based formulation (MCLBF). METHODS: FFB was loaded at 90% and 20% w/w of saturated solubility in MCLBF. The two formulations were pre-dispersed in purified water at 5% w/w (ME90% and 20%, respectively) and orally administered to rats to measure in vivo luminal drug concentrations. RESULTS: FFB precipitated in the stomach due to lipid digestion by gastric lipases and loss of solubilization capacity. This was most significant for ME90%. For ME90%, a high degree of supersaturation was also observed in the duodenum, however, precipitated FFB crystals rapidly re-dissolved. The combination of supersaturation and rapid re-dissolution appeared to drive effective absorption in the upper intestine. For ME20%, FFB precipitated in the stomach but not in the crystalline form and rapidly re-dissolved. Supersaturation in the duodenum again appeared to be the major driver of oral absorption. CONCLUSIONS: The data provide one of the first studies of in vivo luminal drug concentration, supersaturation and absorption from lipid based formulations and suggests that for FFB, whilst very high supersaturation may drive in vitro and in vivo precipitation, re-dissolution and drug absorption is rapid and efficient.


Drug Compounding/methods , Drug Delivery Systems/methods , Intestinal Absorption/drug effects , Lipids/chemistry , Administration, Oral , Animals , Duodenum/drug effects , Fenofibrate/blood , Fenofibrate/metabolism , Intestines/drug effects , Male , Rats , Rats, Sprague-Dawley , Solubility , Stomach/drug effects
3.
Int J Toxicol ; 38(3): 192-201, 2019.
Article En | MEDLINE | ID: mdl-31113311

Because dogs are widely used in drug development as nonrodent experimental animals, using a dog model for drug-induced adverse reactions is considered to be relevant for an evaluation and investigation of a mechanism and a biomarker of clinical drug-induced adverse reactions. Skeletal muscle injury occurs by various drugs, including statins and fibrates, during drug development. However, there is almost no report of a dog model for drug-induced skeletal muscle injury. In the present study, we induced skeletal muscle injury in dogs by oral coadministration of lovastatin (LV) and fenofibrate (FF) for 4 weeks. Increases in plasma levels of creatine phosphokinase, myoglobin, miR-1, and miR-133a and degeneration/necrosis of myofibers in skeletal muscles but not in the heart were observed in LV- and FF-coadministered dogs. Plasma levels of lovastatin lactone and lovastatin acid were higher in LV- and FF-coadministered dogs than LV-administered dogs. Taken together, FF coadministration is considered to affect LV metabolism and result in skeletal muscle injury.


Fenofibrate/toxicity , Hypolipidemic Agents/toxicity , Lovastatin/toxicity , Muscle, Skeletal/drug effects , Animals , Creatine Kinase/blood , Dogs , Drug Interactions , Female , Fenofibrate/blood , Fenofibrate/pharmacokinetics , Hypolipidemic Agents/blood , Hypolipidemic Agents/pharmacokinetics , Lovastatin/blood , Lovastatin/pharmacokinetics , Male , MicroRNAs/blood , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myoglobin/blood
4.
Int J Pharm ; 564: 39-47, 2019 Jun 10.
Article En | MEDLINE | ID: mdl-30981872

We investigated the effect of variation in the molecular weight of hypromellose (HPMC) on the oral absorption of fenofibrate (FFB) nanocrystal. Four types of HPMC with different molecular weights and sodium dodecyl sulfate (SDS) were used as dispersion stabilizers for FFB nanocrystal suspension. Wet-milling of FFB crystal with HPMC and SDS formed diamond-shaped FFB nanocrystals with approximately 150 nm diameter. HPMC was strongly adsorbed onto the FFB nanocrystal interface, and the amount of HPMC adsorbed was not dependent on the molecular weight of HPMC. However, the decrease in the molecular weight of adsorbed HPMC led to an improvement in the permeability of FFB nanocrystal through the mucin layer. The decrease in molecular weight of HPMC enhanced the flexibility of FFB nanocrystal interface and effectively inhibited its interaction with mucin. This led to faster diffusion of FFB nanocrystal through mucin. In vivo oral absorption studies showed rapid FFB absorption from FFB nanocrystal formulations using HPMC of low molecular weights. The present study revealed that the molecular weight of the dispersion stabilizer for drug nanocrystal formulation should be taken into consideration to achieve improved absorption of poorly water-soluble drugs after oral administration.


Fenofibrate/chemistry , Hypolipidemic Agents/chemistry , Hypromellose Derivatives/chemistry , Mucins/chemistry , Nanoparticles/chemistry , Administration, Oral , Animals , Diffusion , Fenofibrate/blood , Fenofibrate/pharmacokinetics , Hypolipidemic Agents/blood , Hypolipidemic Agents/pharmacokinetics , Hypromellose Derivatives/pharmacokinetics , Intestinal Absorption , Male , Molecular Weight , Permeability , Rats, Sprague-Dawley , Sodium Dodecyl Sulfate/chemistry
5.
Drug Des Devel Ther ; 13: 129-139, 2019.
Article En | MEDLINE | ID: mdl-30587933

PURPOSE: Fenofibrate (Fbt) is a prodrug that has been used to reduce low-density-lipoprotein cholesterol, triglycerides, and increase high-density-lipoprotein cholesterol. Simvastatin (Svt) is a classic lipid-lowering drug that is widely used in the treatment of hypercholesterolemia and hypertriglyceridemia, while berberine chloride (Bbr) is a novel hypolipidemic agent and its blood-lipid-reducing mechanism is distinct from traditional drugs. Currently, drug combination is the trend in treating hyperlipidemia to improve clinical efficacy. The purpose of this study was to evaluate drug interaction from the perspective of pharmacokinetics between Bbr and Fbt/Svt and the tolerability of combined administration in healthy Chinese subjects. METHODS: Healthy subjects (n=60) were randomly allocated to five treatment groups: Bbr alone, Fbt alone, Svt alone, Bbr plus Fbt, and Bbr plus Svt. The experiment was divided into two parts: single-dose administration and multiple-dose administration. Bbr, Fbt, and Svt were taken once every 8 hours, 24 hours, and 24 hours, respectively, over 7 days in the multidose group. Plasma samples were collected and liquid chromatography-mass spectrometry/mass spectrometry was used to detect drug concentrations. RESULTS: No serious adverse reactions or intolerance were observed throughout the trial. More importantly, the combined-administration groups did not show an increase in incidence of side effects. Coadministration of Fbt and Svt with Bbr had no significant effect on the pharmacokinetic parameters of Bbr, except time to maximum concentration, apparent volume of distribution, and apparent clearance. Concurrent coadministration of Bbr had no obvious impact on the pharmacokinetic behavior of Fbt or Svt. Additionally, there was no significant correlation between sex and pharmacokinetic results. CONCLUSION: All treatments were well tolerated. No clinically obvious pharmacokinetic interactions between Bbr and Fbt/Svt were observed with combined administration. The results demonstrated that Bbr can be coadministered safely with Fbt and Svt without dose adjustment.


Berberine/pharmacokinetics , Fenofibrate/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Hypolipidemic Agents/pharmacokinetics , Simvastatin/pharmacokinetics , Adult , Berberine/administration & dosage , Berberine/adverse effects , Berberine/blood , China , Chromatography, Liquid , Drug Administration Schedule , Drug Interactions , Female , Fenofibrate/administration & dosage , Fenofibrate/adverse effects , Fenofibrate/blood , Healthy Volunteers , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Hypolipidemic Agents/administration & dosage , Hypolipidemic Agents/adverse effects , Hypolipidemic Agents/blood , Male , Simvastatin/administration & dosage , Simvastatin/adverse effects , Simvastatin/blood , Tandem Mass Spectrometry , Young Adult
6.
J Control Release ; 255: 45-53, 2017 06 10.
Article En | MEDLINE | ID: mdl-28365295

This study investigates the effect of monoacyl phospholipid incorporation on the in vitro and in vivo performance of self-emulsifying drug delivery systems (SEDDS). Monoacyl phosphatidylcholine (Lipoid S LPC 80 (LPC)) was incorporated into four different fenofibrate (FF)-loaded long-chain SEDDS to investigate the impact of LPC on the emulsion droplet size, extent of digestion, colloidal structure evolution and drug precipitation during in vitro lipolysis simulating human conditions and drug bioavailability in a rat model. The four investigated SEDDS containing long-chain glycerides, polyoxyl 35 castor oil or polyoxyl 8 caprylocaproyl glycerides with or without LPC. In situ synchrotron small/wide-angle X-ray scattering (SAXS/WAXS) was used to simultaneously real-time monitor the kinetics of lamellar phase structure development and FF crystalline precipitation. Adding LPC increased the particle size and polydispersity of the dispersed SEDDS. The two LPC-free SEDDS generated lamellar phase structures (Lα) with d-spacing=4.76nm during digestion. Incorporating LPC into these systems inhibited the formation of lamellar phase structures. The amount of precipitated crystalline FF from the four SEDDS was similar during the first 15min but differed during the last 45min of in vitro digestion. The kinetics of colloidal structure development and FF precipitation was related to the digestion kinetics. The in vivo bioavailability data showed no significant differences between the four SEDDS, which correlates with the in vitro FF precipitation during the first 15min of lipolysis. Thus, the presence of LPC, different emulsion droplet sizes and concentration of lamellar phase structures observed in vitro did not correlate with the FF absorption in rats. The study suggests that later time points of the in vitro lipolysis overestimated FF precipitation in rats because of the high enzyme activity, the lack of gastric and absorption steps, and the low bile salts and phospholipid concentrations of the in vitro model.


Drug Delivery Systems , Phosphatidylcholines , Animals , Biological Availability , Castor Oil/chemistry , Chemical Precipitation , Emulsions , Fenofibrate/administration & dosage , Fenofibrate/analogs & derivatives , Fenofibrate/blood , Fenofibrate/chemistry , Fenofibrate/pharmacokinetics , Gastric Mucosa/metabolism , Glycerides/chemistry , Intestinal Absorption , Male , Particle Size , Phosphatidylcholines/administration & dosage , Phosphatidylcholines/chemistry , Phosphatidylcholines/pharmacokinetics , Rats, Sprague-Dawley
7.
Biomed Chromatogr ; 31(4)2017 Apr.
Article En | MEDLINE | ID: mdl-27594083

Choline fenofibrate is the choline salt of fenofibric acid, which releases free fenofibric acid in the gastrointestinal tract. To estimate the absolute oral bioavailability of fenofibric acid and choline fenofibrate, a novel and sensitive UPLC-MS/MS method with liquid-liquid extraction procedure was developed for the determination of fenofibric acid in rat plasma. The separation was achieved on a Phenomenex Kinetex C18 column (50 × 2.1 mm, 2.6 µm) containing 2 mm ammonium acetate-methanol with a gradient elution program. Validations of this method including specificity, sensitivity (limit of quantification, 5 ng/mL), linearity (0.005-10 µg/mL), accuracy (within ±4.3%), precision (intra- and inter-day coefficient of variation <11.3%), recovery (94.9-105.2% for fenofibric acid), matrix effect, stability and dilution, were all within acceptable limits. This method successfully supported the determination of fenofibric acid and choline fenofibrate. The absolute oral bioavailability was 93.4% for choline fenofibrate and 40.0% for fenofibric acid. These results suggested that choline fenofibrate and fenofibric acid had a better in vivo pharmacokinetic behavior than that of fenofibrate. The two new orally administrated pharmaceuticals, fenofibric acid and choline fenofibrate, can be developed as alternatives to fenofibrate.


Choline/pharmacokinetics , Chromatography, Liquid/methods , Fenofibrate/analogs & derivatives , Tandem Mass Spectrometry/methods , Administration, Intravenous , Administration, Oral , Animals , Biological Availability , Choline/administration & dosage , Fenofibrate/administration & dosage , Fenofibrate/blood , Fenofibrate/pharmacokinetics , Half-Life , Limit of Detection , Liquid-Liquid Extraction/methods , Rats, Sprague-Dawley , Reproducibility of Results , Sensitivity and Specificity
8.
Int J Nanomedicine ; 11: 2829-38, 2016.
Article En | MEDLINE | ID: mdl-27366063

BACKGROUND: Self-nanoemulsifying drug delivery systems (SNEDDS) have become a popular formulation option as nanocarriers for poorly water-soluble drugs. The objective of this study was to investigate the factor that can influence the design of successful lipid formulation classification system (LFCS) Type III SNEDDS formulation and improve the oral bioavailability (BA) of fenofibrate. MATERIALS AND METHODS: LFCS Type III SNEDDS were designed using various oils, water-soluble surfactants, and/or cosolvents (in considering the polarity of the lipids) for the model anticholesterol drug, fenofibrate. The developed SNEDDS were assessed visually and by measurement of the droplet size. Equilibrium solubility of fenofibrate in the SNEDDS was conducted to find out the maximum drug loading. Dynamic dispersion studies were carried out (1/100 dilution) in water to investigate how much drug stays in solution after aqueous dispersion of the formulation. The BA of SNEDDS formulation was evaluated in the rat. RESULTS: The results from the characterization and solubility studies showed that formulations containing mixed glycerides were highly efficient SNEDDS as they had higher solubility of the drug and produced nanosized droplets. The dispersion studies confirmed that SNEDDS (containing polar mixed glycerides) can retain >98% drug in solution for >24 hours in aqueous media. The in vivo pharmacokinetics parameters of SNEDDS formulation in comparison with pure drug showed significant increase in C max and AUC0- t , ~78% and 67%, respectively. The oral BA of fenofibrate from SNEDDS in rats was ~1.7-fold enhanced as compared with the BA from pure drug. CONCLUSION: Fenofibrate-loaded LFCS Type III SNEDDS formulations could be a potential oral pharmaceutical product for administering the poorly water-soluble drug, fenofibrate, with an enhanced oral BA.


Drug Delivery Systems/methods , Emulsions/chemistry , Fenofibrate/administration & dosage , Fenofibrate/pharmacology , Nanoparticles/chemistry , Water/chemistry , Absorption, Physicochemical , Administration, Oral , Animals , Biological Availability , Chemical Precipitation , Chemistry, Pharmaceutical , Emulsions/pharmacokinetics , Fenofibrate/blood , Fenofibrate/chemistry , Hydrogen-Ion Concentration , Lipid Droplets/chemistry , Male , Particle Size , Rats , Solubility , Surface-Active Agents/pharmacology
9.
Br J Clin Pharmacol ; 82(5): 1325-1332, 2016 11.
Article En | MEDLINE | ID: mdl-27367040

AIMS: Roux-en-Y gastric bypass (RYGB) alters the anatomical structure of the gastrointestinal tract, which can result in alterations in drug disposition. The aim of the present study was to evaluate the oral disposition of two compounds belonging to the Biopharmaceutical Classification System Class II - fenofibrate (bile salt-dependent solubility) and posaconazole (gastric pH-dependent dissolution) - before and after RYGB in the same individuals. METHODS: A single-dose pharmacokinetic study with two model compounds - namely, 67 mg fenofibrate (Lipanthyl®) and 400 mg posaconazole (Noxafil®) - was performed in 12 volunteers pre- and post-RYGB. After oral administration, blood samples were collected at different time points up to 48 h after administration. Plasma concentrations were determined by high-performance liquid chromatography in order to calculate the area under the concentration-time curve up to 48 h (AUC0-48 h ), the peak plasma concentration (Cmax) and the time to reach peak concentration (Tmax ). RESULTS: After administration of fenofibrate, no relevant differences in AUC0-48 h , Cmax and Tmax between the pre- and postoperative setting were observed. The geometric mean of the ratio of AUC0-48 h post/pre-RYGB for fenofibrate was 1.10 [95% confidence interval (CI) 0.87, 1.40; P = 0.40]. For posaconazole, an important decrease in AUC0-48 h and Cmax following RYGB was shown; the geometric mean of the AUC0-48 h post/pre-RYGB ratio was 0.68 (95% CI 0.48, 0.96; P = 0.03) and the geometric mean of the Cmax pre/post-RYGB ratio was 0.60 (95% CI 0.39, 0.94; P = 0.03). The decreased exposure of posaconazole could be explained by the increased gastric pH and accelerated gastric emptying of fluids post-RYGB. No difference for Tmax was observed. CONCLUSIONS: The disposition of fenofibrate was not altered after RYGB, whereas the oral disposition of posaconazole was significantly decreased following RYGB.


Area Under Curve , Fenofibrate/pharmacokinetics , Gastric Bypass , Triazoles/pharmacokinetics , Administration, Oral , Fenofibrate/administration & dosage , Fenofibrate/blood , Triazoles/administration & dosage , Triazoles/blood
10.
J Pharm Sci ; 105(8): 2381-5, 2016 08.
Article En | MEDLINE | ID: mdl-27364460

The present study aims to evaluate the in vitro and in vivo performance of ordered mesoporous silica (OMS) as a carrier for the poorly water-soluble compound fenofibrate. Fenofibrate was loaded into OMS via incipient wetness impregnation to obtain a 29% drug load and formulated into capsules. Two capsule dosage forms (containing 33.5 and 16.75 mg fenofibrate, respectively) were compared with the commercially available forms-Lipanthyl(®) (fenofibrate microcrystals) and Tricor(®) (fenofibrate nanocrystals). In vitro dissolution tests showed that the amount of fenofibrate released from Lipanthyl(®) and Tricor(®) was approximately 30%, whereas approximately 66% and 60% of the drug was released from OMS capsules containing 33.5 and 16.75 mg of fenofibrate, respectively. Storage of OMS capsules loaded with 33.5 mg of fenofibrate at 25°C/60% relative humidity (RH) or 40°C/75% RH did not alter the release kinetics, nor the physical state of the compound, pointing the stability of the present formulation. The in vivo study in dogs confirmed satisfying level of safety and tolerability of fenofibrate-OMS formulation (eq. 33.5 mg) with the potential to improve the absorption of fenofibrate. Though some variability in the data, this formulation is promising to be further investigated in a clinical trial setting.


Drug Carriers/chemistry , Fenofibrate/pharmacokinetics , Hypolipidemic Agents/pharmacokinetics , Silicon Dioxide/chemistry , Administration, Oral , Animals , Biological Availability , Dogs , Drug Liberation , Fenofibrate/blood , Fenofibrate/chemistry , Hypolipidemic Agents/blood , Hypolipidemic Agents/chemistry , Male , Porosity , Solubility
11.
Biomed Chromatogr ; 30(12): 2003-2008, 2016 Dec.
Article En | MEDLINE | ID: mdl-27270950

Nitrofibriate, a new compound of hypolipidemic, is modified based on fenofibrate. Both of them are used for prevention and treatment of cardiovascular diseases. In this study, an accurate and sensitive analytical method of reversed-phase high-performance liquid chromatography was developed to determine fenofibric acid, which is an active metabolite of both nitrofibriate and fenofibrate in rat plasma. This method was validated and successfully applied to pharmacokinetic study of nitrofibriate and fenofibrate after oral administration. The results suggested that the pharmacokinetic behavior of nitrofibriate followed a nonlinear process, while fenofibrate was linear, demonstrating that the two drugs were different in pharmacokinetic behaviors. Moreover, the effect of fenofibrate and nitrofibriate on releasing NO in rat serum was explored. This study showed that nitrofibriate, as a nitric oxide donor, could slowly release nitric oxide in vivo. This study provided a biopharmaceutical basis for further study of nitrofibriate.


Fenofibrate/analogs & derivatives , Fenofibrate/pharmacokinetics , Nitric Oxide/blood , Nitro Compounds/pharmacokinetics , Administration, Oral , Animals , Female , Fenofibrate/administration & dosage , Fenofibrate/blood , Limit of Detection , Male , Nitro Compounds/administration & dosage , Nitro Compounds/blood , Rats , Rats, Sprague-Dawley , Reproducibility of Results
12.
Int J Nanomedicine ; 11: 1067-76, 2016.
Article En | MEDLINE | ID: mdl-27042061

This study aimed to prepare the aminoclay-lipid hybrid composite to enhance the drug release and improve the oral bioavailability of poorly water-soluble fenofibrate. Antisolvent precipitation coupled with an immediate freeze-drying method was adopted to incorporate fenofibrate into aminoclay-lipid hybrid composite (ALC). The optimal composition of the ALC formulation was determined as the ratios of aminoclay to krill oil of 3:1 (w/w), krill oil to fenofibrate of 2:1 (w/w), and antisolvent to solvent of 6:4 (v/v). The morphological characteristics of ALC formulation were determined using scanning electron microscopy, differential scanning calorimetry, and X-ray powder diffraction, which indicated microcrystalline state of fenofibrate in ALC formulation. The ALC formulation achieved almost complete dissolution within 30 minutes, whereas the untreated powder and physical mixture exhibited less than 15% drug release. Furthermore, ALC formulation effectively increased the peak plasma concentration (C max) and area under the curve (AUC) of fenofibric acid (an active metabolite) in rats by approximately 13- and seven-fold, respectively. Furthermore, ALC formulation exhibited much lower moisture sorption behavior than the lyophilized formulation using sucrose as a cryoprotectant. Taken together, the present findings suggest that ALC formulation is promising for improving the oral absorption of poorly soluble fenofibrate.


Drug Carriers/administration & dosage , Fenofibrate/administration & dosage , Fenofibrate/pharmacokinetics , Animals , Area Under Curve , Biological Availability , Calorimetry, Differential Scanning , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Liberation , Fenofibrate/analogs & derivatives , Fenofibrate/blood , Fenofibrate/chemistry , Freeze Drying , Lipids/chemistry , Male , Microscopy, Electron, Scanning , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Rats, Sprague-Dawley , Solubility , X-Ray Diffraction
13.
Arch Pharm Res ; 39(4): 531-538, 2016 Apr.
Article En | MEDLINE | ID: mdl-26992922

To investigate the possibility of developing a novel oral pharmaceutical product using fenofibric acid instead of choline fenofibrate, the powder properties, solubility, dissolution and pharmacokinetics in rats of fenofibrate, choline fenofibrate and fenofibric acid were compared. Furthermore, the effect of magnesium carbonate, an alkalising agent on the solubility, dissolution and oral bioavailability of fenofibric acid was assessed, a mixture of fenofibric acid and magnesium carbonate being prepared by simple blending at a weight ratio of 2/1. The three fenofibrate derivatives showed different particle sizes and melting points with similar crystalline shape. Fenofibric acid had a significantly higher aqueous solubility and dissolution than fenofibrate, but significantly lower solubility and dissolution than choline fenofibrate. However, the fenofibric acid/magnesium carbonate mixture greatly improved the solubility and dissolution of fenofibric acid with an enhancement to levels similar with those for choline fenofibrate. Fenofibric acid gave lower plasma concentrations, AUC and Cmax values compared to choline fenofibrate in rats. However, the mixture resulted in plasma concentrations, AUC and Cmax values levels not significantly different from those for choline fenofibrate. Specifically, magnesium carbonate increased the aqueous solubility, dissolution and bioavailability of fenofibric acid by about 7.5-, 4- and 1.6-fold, respectively. Thus, the mixture of fenofibric acid and magnesium carbonate at the weight ratio of 2/1 might be a candidate for an oral pharmaceutical product with improved oral bioavailability.


Excipients/chemistry , Fenofibrate/analogs & derivatives , Hypolipidemic Agents/chemistry , Magnesium/chemistry , Technology, Pharmaceutical/methods , Administration, Oral , Animals , Biological Availability , Chromatography, High Pressure Liquid , Fenofibrate/administration & dosage , Fenofibrate/blood , Fenofibrate/chemistry , Fenofibrate/pharmacokinetics , Hypolipidemic Agents/administration & dosage , Hypolipidemic Agents/blood , Hypolipidemic Agents/pharmacokinetics , Male , Particle Size , Powders , Rats, Sprague-Dawley , Solubility , Spectroscopy, Fourier Transform Infrared , Surface Properties
14.
Pharmacotherapy ; 36(1): 49-56, 2016 Jan.
Article En | MEDLINE | ID: mdl-26799348

STUDY OBJECTIVE: Because we previously observed a significant 41% reduction in gemfibrozil exposure after 2 weeks of lopinavir-ritonavir administration, we sought to determine the influence of lopinavir-ritonavir and ritonavir alone on the pharmacokinetics of fenofibric acid, an alternative to gemfibrozil for the treatment of elevated triglyceride levels. DESIGN: Open-label, single-sequence pharmacokinetic study. SETTING: Clinical Research Center at the National Institutes of Health. SUBJECTS: Thirteen healthy adult volunteers. INTERVENTION: Subjects received a single oral dose of fenofibrate 145 mg during three study phases: before ritonavir administration, after 2 weeks of administration of ritonavir 100 mg twice/day, and after 2 weeks of administration of lopinavir 400 mg-ritonavir 100 mg twice/day. MEASUREMENTS AND MAIN RESULTS: Serial blood samples were collected over 120 hours for determination of fenofibric acid concentrations. Fenofibric acid pharmacokinetic parameter values were compared before and after concomitant ritonavir or lopinavir-ritonavir administration. The geometric mean ratios (90% confidence intervals) for fenofibric acid area under the plasma concentration-time curve were 0.89 (0.77-1.01) after 14 days of ritonavir alone compared with baseline (p>0.05) and 0.87 (0.69-1.05) after 14 days of lopinavir-ritonavir compared with baseline (p>0.05). Study drugs were generally well tolerated; all adverse events were mild or moderate, transient, and resolved without intervention. CONCLUSION: In contrast to a significant interaction between gemfibrozil and lopinavir-ritonavir, neither lopinavir-ritonavir nor ritonavir alone altered the pharmacokinetics of fenofibric acid in healthy volunteers. These data suggest that fenofibrate remains an important option in human immunodeficiency virus-infected patients receiving common ritonavir-boosted therapy.


Fenofibrate/analogs & derivatives , Hypolipidemic Agents/pharmacokinetics , Lopinavir/pharmacology , Ritonavir/pharmacology , Adult , Drug Administration Schedule , Drug Interactions , Drug Therapy, Combination , Female , Fenofibrate/blood , Fenofibrate/pharmacokinetics , Humans , Hypolipidemic Agents/blood , Lopinavir/administration & dosage , Male , Middle Aged , Ritonavir/administration & dosage , Young Adult
15.
Biomed Chromatogr ; 30(7): 1075-1082, 2016 Jul.
Article En | MEDLINE | ID: mdl-26577601

With the purpose of carrying out pharmacokinetic interaction studies ofnberberine (BBR) and fenofibrate (FBT), an UPLC-MS/MS method has been developed and validated. The analytes, BBR and fenofibric acid (FBA, metabolite of FBT) and the internal standard, tetrahydropalmatine, were extracted with dichloromethane-diethyl ether (3:2, v/v) and separated on an Agilent Eclipse XDB C18 column using a mobile phase composed of acetonitrile and water. With positive ion electrospray ionization, the analytes were monitored on a triple quadrupole mass spectrometer in multiple reaction monitoring mode. Linear calibration curves were obtained over the concentration ranges of 0.1-100.0 ng/mL for BBR and 10.0-50,000.0 ng/mL for FBA. For BBR and FBA, the intra- and inter-day precisions were <11.5 and 11.9%, respectively. The accuracy was within 11.7% and 11.3%. The mean recoveries of BBR at three concentrations of 0.2, 20.0, 80.0 ng/mL were >85.6%, and those of FBA at three concentrations of 20.0, 2500.0, 40,000.0 ng/mL were >87.9%. Consequently, the proposed method was applied to the pharmacokinetic interaction study of FBT combined with BBR after oral administration in rats and was proved to be sensitive, specific and reliable to analyze BBR and FBA in biological samples simultaneously. Copyright © 2016 John Wiley & Sons, Ltd.


Berberine/blood , Chromatography, Liquid/methods , Fenofibrate/blood , Tandem Mass Spectrometry/methods , Administration, Oral , Animals , Berberine/administration & dosage , Berberine/pharmacokinetics , Drug Combinations , Fenofibrate/administration & dosage , Fenofibrate/pharmacokinetics , Male , Rats , Rats, Wistar , Reproducibility of Results
16.
Pharm Res ; 33(4): 970-82, 2016 Apr.
Article En | MEDLINE | ID: mdl-26703975

PURPOSE: In vitro lipid digestion models are commonly used to screen lipid-based formulations (LBF), but in vitro-in vivo correlations are in some cases unsuccessful. Here we enhance the scope of the lipid digestion test by incorporating an absorption 'sink' into the experimental model. METHODS: An in vitro model of lipid digestion was coupled directly to a single pass in situ intestinal perfusion experiment in an anaesthetised rat. The model allowed simultaneous real-time analysis of the digestion and absorption of LBFs of fenofibrate and was employed to evaluate the influence of formulation digestion, supersaturation and precipitation on drug absorption. RESULTS: Formulations containing higher quantities of co-solvent and surfactant resulted in higher supersaturation and more rapid drug precipitation in vitro when compared to those containing higher quantities of lipid. In contrast, when the same formulations were examined using the coupled in vitro lipid digestion - in vivo absorption model, drug flux into the mesenteric vein was similar regardless of in vitro formulation performance. CONCLUSION: For some drugs, simple in vitro lipid digestion models may underestimate the potential for absorption from LBFs. Consistent with recent in vivo studies, drug absorption for rapidly absorbed drugs such as fenofibrate may occur even when drug precipitation is apparent during in vitro digestion.


Drug Carriers/metabolism , Fenofibrate/administration & dosage , Fenofibrate/pharmacokinetics , Hypolipidemic Agents/administration & dosage , Hypolipidemic Agents/pharmacokinetics , Lipid Metabolism , Animals , Digestion , Drug Carriers/chemistry , Fenofibrate/blood , Fenofibrate/chemistry , Hypolipidemic Agents/blood , Hypolipidemic Agents/chemistry , Lipids/chemistry , Male , Rats, Sprague-Dawley , Solubility
17.
Int J Pharm ; 495(1): 9-18, 2015 Nov 10.
Article En | MEDLINE | ID: mdl-26325310

Oral drug delivery is the most preferred route for patients; however, the low solubility of drugs and the resultant poor absorption compromise the benefits of oral administration. On the other hand, for years, the overwhelmingly accepted mechanism for enhanced oral absorption using lipid nanocarriers was based on the process of lipid digestion and drug solubilization in the small intestine. Few reports indicated that other bypass pathways are involved in drug absorption in the gastrointestinal tract (GIT) for oral delivery of nanocarriers. Herein, we report a new nanoemulsion system with a denatured globular protein with a diameter of 30 nm, soybean protein isolates (SPI), and bile salt as emulsifiers, aiming to enhance the absorption of insoluble drugs and explore other pathways for absorption. A BCS class II drug, fenofibrate (FB), was used as the model drug. The SPI and bile salt-coated Ns with a diameter of approximately 150 nm were prepared via a high-pressure homogenizing procedure. Interestingly, the present Ns could be converted to solid dosage form using fluid-bed coating technology, maintaining a nanoscale size. Most importantly, in a model of in situ rat intestinal perfusion, Ns could penetrate across the intestinal epithelial barrier into the systemic circulation and then obtain biodistribution into other tissues. In addition, Ns significantly improved FB oral absorption, exhibited as a greater than 2- and 2.5-fold increase in Cmax and AUC0-t, respectively, compared to the suspension formulation. Overall, the present Ns are promising nanocarriers for the oral delivery of insoluble drugs, and the penetration of intact Ns across the GIT barrier into systemic circulation may be a new strategy for improved drug absorption with the use of nanocarriers.


Bile Acids and Salts/administration & dosage , Bile Acids and Salts/chemistry , Drug Carriers/chemistry , Fenofibrate/blood , Fenofibrate/pharmacokinetics , Intestinal Absorption , Nanoparticles/administration & dosage , Soybean Proteins/chemistry , Administration, Oral , Animals , Bile Acids and Salts/pharmacokinetics , Biological Availability , Caco-2 Cells , Cell Survival/drug effects , Chemistry, Pharmaceutical , Drug Carriers/administration & dosage , Drug Carriers/pharmacokinetics , Emulsions/administration & dosage , Emulsions/adverse effects , Emulsions/chemistry , Emulsions/pharmacokinetics , Fenofibrate/administration & dosage , Fenofibrate/chemistry , Humans , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Particle Size , Protein Denaturation , Rats , Solubility , Soybean Proteins/administration & dosage , Soybean Proteins/pharmacokinetics , Suspensions/pharmacokinetics , Tissue Distribution , Water/chemistry
18.
Eur J Pharm Biopharm ; 96: 207-16, 2015 Oct.
Article En | MEDLINE | ID: mdl-26215636

Novel formulations that overcome the solubility limitations of poorly water soluble drugs (PWSD) are becoming ever more critical to a drug development process inundated with these compounds. There is a clear need for developing bio-enabling formulation approaches to improve oral bioavailability for PWSD, but also to establish a range of predictive in vitro and in silico biopharmaceutics based tools for guiding formulation design and forecasting in vivo effects. The dual aim of this study was to examine the potential for a novel lipid based formulation, termed a lipidic dispersion, to enhance fasted state oral bioavailability of fenofibrate, while also assessing the predictive ability of biorelevant in vitro and in silico testing. Formulation as a lipidic dispersion improved both dissolution and solubilisation of fenofibrate through a combination of altered solid state characteristics and incorporation of solubilising lipidic excipients. These changes resulted in an increased rate of absorption and increased maximal plasma concentrations compared to a commercial, micronised product (Lipantil® Micro) in a pig model. Combination of biorelevant in vitro measurements with in silico physiologically based pharmacokinetic (PBPK) modelling resulted in an accurate prediction of formulation performance and forecasts a reduction in food effects on fenofibrate bioavailability through maximising its fasted state dissolution.


Fenofibrate/pharmacokinetics , Hypolipidemic Agents/pharmacokinetics , Olive Oil/chemistry , Pharmaceutical Vehicles/chemistry , Polyethylene Glycols/chemistry , Polysorbates/chemistry , Povidone/chemistry , Surface-Active Agents/chemistry , Animals , Biological Availability , Computational Biology , Cross-Over Studies , Drug Compounding , Drug Liberation , Excipients/chemistry , Expert Systems , Fenofibrate/blood , Fenofibrate/chemistry , Fenofibrate/metabolism , Food-Drug Interactions , Hypolipidemic Agents/blood , Hypolipidemic Agents/chemistry , Hypolipidemic Agents/metabolism , Intestinal Absorption , Male , Random Allocation , Solubility , Sus scrofa
19.
Int J Nanomedicine ; 10: 1691-701, 2015.
Article En | MEDLINE | ID: mdl-25784799

In this study, mesoporous silica nanoparticles (MSNs) were used to prepare an oral push-pull osmotic pump. Fenofibrate, the selected model drug, was firstly loaded into the MSNs, followed by a suspending agent consisting of a drug layer of push-pull osmotic pump. Fenofibrate-loaded MSNs were characterized by scanning electron microscopy (SEM), transmission electron microscopy (TEM), nitrogen adsorption/desorption analysis, differential scanning calorimetry (DSC), powder X-ray diffractometry (PXRD) analysis, and Fourier-transform infrared (FT-IR) spectroscopy. Polyethylene oxide of molecular weight (MW) 100,000 and polyethylene oxide of MW 6,000,000 were selected as the suspending agent and the expanding agent, respectively. Cellulose acetate was used as the semipermeable membrane, along with polyethylene glycol 6,000 to increase the flexibility and control the membrane permeability. The in vitro dissolution studies indicated that the osmotic pump tablet combined with MSNs was able to deliver fenofibrate in an approximately zero-order manner in 24 hours. A pharmacokinetic study showed that, although the maximum plasma concentration of the osmotic pump was lower than that of the reference formulation, the relative bioavailability was increased, indicating that the osmotic pump was more efficient than the reference tablets. Therefore, using MSNs as a carrier for poorly water-soluble drugs is an effective method for preparing osmotic pump tablets.


Drug Delivery Systems/methods , Fenofibrate , Nanoparticles/chemistry , Silicon Dioxide/chemistry , Animals , Dogs , Fenofibrate/administration & dosage , Fenofibrate/blood , Fenofibrate/chemistry , Fenofibrate/pharmacokinetics , Solubility
20.
Alcohol Clin Exp Res ; 39(1): 136-45, 2015 Jan.
Article En | MEDLINE | ID: mdl-25516156

BACKGROUND: Peroxisome proliferator-activated receptor (PPAR) agonists reduce voluntary ethanol (EtOH) consumption in rat models and are promising therapeutics in the treatment for drug addictions. We studied the effects of different classes of PPAR agonists on chronic EtOH intake and preference in mice with a genetic predisposition for high alcohol consumption and then examined human genomewide association data for polymorphisms in PPAR genes in alcohol-dependent subjects. METHODS: Two different behavioral tests were used to measure intake of 15% EtOH in C57BL/6J male mice: 24-hour 2-bottle choice and limited access (3-hour) 2-bottle choice, drinking in the dark. We measured the effects of pioglitazone (10 and 30 mg/kg), fenofibrate (50 and 150 mg/kg), GW0742 (10 mg/kg), tesaglitazar (1.5 mg/kg), and bezafibrate (25 and 75 mg/kg) on EtOH intake and preference. Fenofibric acid, the active metabolite of fenofibrate, was quantified in mouse plasma, liver, and brain by liquid chromatography tandem mass spectrometry. Data from a human genome-wide association study (GWAS) completed in the Collaborative Study on the Genetics of Alcoholism (COGA) were then used to analyze the association of single nucleotide polymorphisms (SNPs) in different PPAR genes (PPARA, PPARD, PPARG, and PPARGC1A) with 2 phenotypes: DSM-IV alcohol dependence (AD) and the DSM-IV criterion of withdrawal. RESULTS: Activation of 2 isoforms of PPARs, α and γ, reduced EtOH intake and preference in the 2 different consumption tests in mice. However, a selective PPARδ agonist or a pan agonist for all 3 PPAR isoforms did not decrease EtOH consumption. Fenofibric acid, the active metabolite of the PPARα agonist fenofibrate, was detected in liver, plasma, and brain after 1 or 8 days of oral treatment. The GWAS from COGA supported an association of SNPs in PPARA and PPARG with alcohol withdrawal and PPARGC1A with AD but found no association for PPARD with either phenotype. CONCLUSIONS: We provide convergent evidence using both mouse and human data for specific PPARs in alcohol action. Reduced EtOH intake in mice and the genetic association between AD or withdrawal in humans highlight the potential for repurposing FDA-approved PPARα or PPARγ agonists for the treatment of AD.


Alcohol Drinking/genetics , Alcoholism/genetics , PPAR alpha/genetics , PPAR gamma/genetics , Adult , Alcohol Drinking/drug therapy , Alcoholism/drug therapy , Alkanesulfonates/therapeutic use , Animals , Bezafibrate/therapeutic use , Brain/metabolism , Female , Fenofibrate/blood , Fenofibrate/pharmacokinetics , Fenofibrate/therapeutic use , Genome-Wide Association Study , Humans , Liver/metabolism , Male , Mice , PPAR alpha/agonists , PPAR gamma/agonists , Phenylpropionates/therapeutic use , Pioglitazone , Polymorphism, Single Nucleotide/genetics , Thiazoles/therapeutic use , Thiazolidinediones/therapeutic use
...