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










Publication year range
1.
Clin Pharmacol Ther ; 110(2): 480-485, 2021 08.
Article in English | MEDLINE | ID: mdl-33880760

ABSTRACT

The objective of this study was to determine the effects of the OATP inhibitor rifampin on pharmacokinetic of Biopharmaceutics Drug Disposition Classification System Class 1 compound fluvastatin. A crossover study was carried out in 10 healthy subjects who were randomized to 2 phases to receive fluvastatin 20 mg orally alone and following a 30-minute 600 mg i.v. infusion of rifampin. The results demonstrated that i.v. rifampin increased the mean area under the plasma fluvastatin concentration-time curve (AUC0-∞ ) by 255%, mean peak plasma concentration (Cmax ) by 254%, decreased oral volume of distribution by 71%, whereas the mean elimination terminal half-life (T1/2 ), mean absorption time (MAT), and time to peak concentration (Tpeak ) of fluvastatin did not significantly change. The study demonstrated that rifampin exhibited a significant drug interaction with fluvastatin. The mechanism of the increased plasma concentrations is likely due to inhibition of OATP transporters in hepatocytes.


Subject(s)
Anti-Bacterial Agents/adverse effects , Fluvastatin/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Rifampin/adverse effects , Administration, Oral , Adult , Anti-Bacterial Agents/administration & dosage , Area Under Curve , Cross-Over Studies , Drug Interactions , Female , Fluvastatin/administration & dosage , Half-Life , Healthy Volunteers , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Infusions, Intravenous , Intestinal Absorption , Male , Middle Aged , Organic Anion Transporters , Prospective Studies , Rifampin/administration & dosage
2.
Drug Metab Dispos ; 48(5): 419-425, 2020 05.
Article in English | MEDLINE | ID: mdl-32114507

ABSTRACT

The organic anion transporting polypeptide (OATP)2B1 is localized on the basolateral membrane of hepatocytes and is expressed in enterocytes. Based on its distribution pattern and functional similarity to OATP1B-type transporters, OATP2B1 might have a role in the absorption and disposition of a range of xenobiotics. Although several prescription drugs, including hydroxymethylglutaryl-coenzyme A-CoA reductase inhibitors (statins) such as fluvastatin, are OATP2B1 substrates in vitro, evidence supporting the in vivo relevance of this transporter remains limited, and most has relied on substrate-inhibitor interactions resulting in altered pharmacokinetic properties of the victim drugs. To address this knowledge deficit, we developed and characterized an Oatp2b1-deficient mouse model and evaluated the impact of this transporter on the absorption and disposition of fluvastatin. Consistent with the intestinal localization of Oatp2b1, we found that the genetic deletion or pharmacological inhibition of Oatp2b1 was associated with decreased absorption of fluvastatin by 2- to 3-fold. The availability of a viable Oatp2b1-deficient mouse model provides an opportunity to unequivocally determine the contribution of this transporter to the absorption and drug-drug interaction potential of drugs. SIGNIFICANCE STATEMENT: The current investigation suggests that mice deficient in Oatp2b1 provide a valuable tool to study the in vivo importance of this transporter. In addition, our studies have identified novel potent inhibitors of OATP2B1 among the class of tyrosine kinase inhibitors, a rapidly expanding class of drugs used in various therapeutic areas that may cause drug-drug interactions with OATP2B1 substrates.


Subject(s)
Fluvastatin/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Organic Anion Transporters/metabolism , Administration, Oral , Animals , Drug Interactions , Female , Fluvastatin/administration & dosage , HEK293 Cells , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Intestinal Absorption , Intestinal Mucosa/metabolism , Male , Mice , Mice, Knockout , Organic Anion Transporters/genetics , RNA/genetics , RNA/metabolism
3.
J Pharm Biomed Anal ; 182: 113128, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32004772

ABSTRACT

Fluvastatin and atorvastatin are inhibitors of hydroxy-methylglutaryl-CoA (HMG-CoA) reductase, the enzyme that converts HMG-CoA to mevalonic acid (MVA). The present study reports for the first time the analysis of mevalonolactone (MVL) in plasma samples by UPLC-MS/MS as well as the use of MVA, analyzed as MVL, as a pharmacodynamics parameter of fluvastatin in multiple oral doses (20, 40 or 80 mg/day for 7 days) and atorvastatin in a single oral dose (20, 40 or 80 mg) in healthy female volunteers. this study presents the use of MVL exposure as a pharmacodynamics biomarker of fluvastatin in multiple oral doses (20, 40 or 80 mg/day for 7 days) or atorvastatin in a single oral dose (20, 40 or 80 mg) in healthy volunteers (n = 30). The administration of multiple doses of fluvastatin (n = 15) does not alter the values (geometric mean and 95 % CI) of AUC0-24 h of MVL [72.00 (57.49-90.18) vs 65.57 (51.73-83.12) ng∙h/mL], but reduces AUC0-6 h [15.33 (11.85-19.83) vs 8.15 (6.18-10.75) ng∙h/mL] by approximately 47 %, whereas single oral dose administration of atorvastatin (n = 15) reduces both AUC0-24 h [75.79 (65.10-88.24) vs 32.88 (27.05-39.96) ng∙h/mL] and AUC0-6 h [17.07 (13.87-21.01) vs 7.01 (5.99-8.22) ng∙h/mL] values by approximately 57 % and 59 %, respectively. In conclusion, the data show that the plasma exposure of MVL represents a reliable pharmacodynamic parameter for PK-PD (pharmacokinetic-pharmacodynamic) studies of fluvastatin in multiple doses and atorvastatin in a single dose.


Subject(s)
Atorvastatin/administration & dosage , Fluvastatin/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Mevalonic Acid/analogs & derivatives , Administration, Oral , Adult , Area Under Curve , Atorvastatin/pharmacokinetics , Atorvastatin/pharmacology , Chromatography, High Pressure Liquid/methods , Dose-Response Relationship, Drug , Female , Fluvastatin/pharmacokinetics , Fluvastatin/pharmacology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mevalonic Acid/analysis , Mevalonic Acid/blood , Reproducibility of Results , Tandem Mass Spectrometry/methods
4.
Nanoscale ; 12(5): 2966-2972, 2020 Feb 07.
Article in English | MEDLINE | ID: mdl-31971210

ABSTRACT

Lactate, the main contributor to the acidic tumor microenvironment, not only promotes the proliferation of tumor cells, but also closely relates to tumor invasion and metastasis. Here, a tumor targeting nanoplatform, designated as Me&Flu@MSN@MnO2-FA, was fabricated for effective tumor suppression and anti-metastasis by interfering with lactate metabolism of tumor cells. Metformin (Me) and fluvastatin sodium (Flu) were incorporated into MnO2-coated mesoporous silicon nanoparticles (MSNs), the synergism between Me and Flu can modulate the pyruvate metabolic pathway to produce more lactate, and concurrently inhibit lactate efflux to induce intracellular acidosis to kill tumor cells. As a result of the restricted lactate efflux, the extracellular lactate concentration is reduced, and the ability of the tumor cells to migrate is also weakened. This ingenious strategy based on Me&Flu@MSN@MnO2-FA showed an obvious inhibitory effect on tumor growth and resistance to metastasis.


Subject(s)
Fluvastatin , Lactates/metabolism , Manganese Compounds , Metformin , Nanoparticles , Neoplasms , Tumor Microenvironment/drug effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Fluvastatin/chemistry , Fluvastatin/pharmacokinetics , Fluvastatin/pharmacology , Folic Acid/metabolism , Humans , Manganese Compounds/chemistry , Manganese Compounds/pharmacokinetics , Manganese Compounds/pharmacology , Metformin/chemistry , Metformin/pharmacokinetics , Metformin/pharmacology , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasm Metastasis , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Porosity , Silicon/chemistry , Silicon/pharmacokinetics , Silicon/pharmacology
6.
Clin Pharmacol Ther ; 106(3): 668-680, 2019 09.
Article in English | MEDLINE | ID: mdl-30989645

ABSTRACT

The aim of this study was to investigate how variability in multiple genes related to pharmacokinetics affects fluvastatin exposure. We determined fluvastatin enantiomer pharmacokinetics and sequenced 379 pharmacokinetic genes in 200 healthy volunteers. CYP2C9*3 associated with significantly increased area under the plasma concentration-time curve (AUC) of both 3R,5S-fluvastatin and 3S,5R-fluvastatin (by 67% and 94% per variant allele copy, P = 3.77 × 10-9 and P = 3.19 × 10-12 ). In contrast, SLCO1B1 c.521T>C associated with increased AUC of active 3R,5S-fluvastatin only (by 34% per variant allele copy; P = 8.15 × 10-8 ). A candidate gene analysis suggested that CYP2C9*2 also affects the AUC of both fluvastatin enantiomers and that SLCO2B1 single-nucleotide variations may affect the AUC of 3S,5R-fluvastatin. Thus, SLCO transporters have enantiospecific effects on fluvastatin pharmacokinetics in humans. Genotyping of both CYP2C9 and SLCO1B1 may be useful in predicting fluvastatin efficacy and myotoxicity.


Subject(s)
Anticholesteremic Agents/chemistry , Anticholesteremic Agents/pharmacokinetics , Cytochrome P-450 CYP2C9/genetics , Fluvastatin/chemistry , Fluvastatin/pharmacokinetics , Liver-Specific Organic Anion Transporter 1/genetics , Area Under Curve , Half-Life , Humans , Pharmacogenetics , Polymorphism, Single Nucleotide
7.
Xenobiotica ; 49(5): 602-610, 2019 May.
Article in English | MEDLINE | ID: mdl-29768081

ABSTRACT

1. Oatp inhibitors have been shown to significantly increase the plasma exposure of statins. However, understanding alterations of liver concentration is also important. While modeling has simulated liver concentration changes, availability of experimental data is limited, especially when concerning drug-drug interactions (DDI). The objective of this work was to determine blood and liver concentrations of fluvastatin, lovastatin and pitavastatin, when blocking uptake transporters. 2. In wild-type mouse, rifampin pre-treatment decreased the unbound liver-to-plasma ratio (Kp,uu) of fluvastatin by 4.2-fold to 2.2, lovastatin by 4.9-fold to 0.81 and pitavastatin by 10-fold to 0.21. Changes in Kp,uu were driven by increases in systemic exposures as liver concentrations were not greatly altered. 3. In Oatp1a/1b knockout mouse (KO), rifampin exerted no additional effect on fluvastatin and lovastatin. Contrarily, rifampin further decreased pitavastatin Kp,uu by 3.4-fold, suggesting that the KO is inadequate to completely block liver uptake of pitavastatin as there are additional rifampin-sensitive uptake mechanism(s) not captured in the KO model. 4. This work provides experimental data showing that the plasma compartment is more sensitive to Oatp modulation than the liver compartment, even for rifampin-mediated DDI. Consistent with previous simulations, inhibiting or targeting Oatps may change Kp,uu, but exhibit only a minimal effect on absolute liver concentrations.


Subject(s)
Fluvastatin , Liver/metabolism , Lovastatin , Organic Cation Transport Proteins/antagonists & inhibitors , Quinolines , Animals , Fluvastatin/pharmacokinetics , Fluvastatin/pharmacology , Lovastatin/pharmacokinetics , Lovastatin/pharmacology , Mice , Mice, Knockout , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Quinolines/pharmacokinetics , Quinolines/pharmacology
8.
Biochim Biophys Acta Biomembr ; 1861(3): 584-593, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30578770

ABSTRACT

Statins are drugs that specifically inhibit the enzyme HMG-CoA reductase and thereby reduce the concentration of low-density lipoprotein cholesterol, which represents a well-established risk factor for the development of atherosclerosis. The results of several clinical trials have shown that there are important intermolecular differences responsible for the broader pharmacologic actions of statins, even beyond HMG-CoA reductase inhibition. According to one hypothesis, the biological effects exerted by these compounds depend on their localization in the cellular membrane. The aim of the current work was to study the interactions of different statins with phospholipid membranes and to investigate their influence on the membrane structure and dynamics using various solid-state NMR techniques. Using 1H NOESY MAS NMR, it was shown that atorvastatin, cerivastatin, fluvastatin, rosuvastatin, and some percentage of pravastatin intercalate the lipid-water interface of POPC membranes to different degrees. Based on cross-relaxation rates, the different average distribution of the individual statins in the bilayer was determined quantitatively. Investigation of the influence of the investigated statins on membrane structure revealed that lovastatin had the least effect on lipid packing and chain order, pravastatin significantly lowered lipid chain order, while the other statins slightly decreased lipid chain order parameters mostly in the middle segments of the phospholipid chains.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Lipid Bilayers/metabolism , Phospholipids/metabolism , Fluvastatin/chemistry , Fluvastatin/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Lipid Bilayers/chemistry , Lovastatin/chemistry , Lovastatin/pharmacokinetics , Magnetic Resonance Spectroscopy/methods , Phosphatidylcholines/chemistry , Phosphatidylcholines/metabolism , Phospholipids/chemistry , Pravastatin/chemistry , Pravastatin/pharmacokinetics , Simvastatin/chemistry , Simvastatin/pharmacokinetics
9.
Drug Des Devel Ther ; 12: 3491-3499, 2018.
Article in English | MEDLINE | ID: mdl-30425452

ABSTRACT

BACKGROUND: In this study, fluvastatin (FVT) nanosuspensions containing cyclodextrin were developed to improve oral bioavailability. METHODS: FVT nanosuspensions containing cyclodextrin were prepared by a high pressure homogenization technique. The nanosuspensions system was then characterized by transmission electron microscopy (TEM), particle size, differential scanning calorimetry (DSC) and powder X-ray diffractometry (PXRD). In addition, in vitro drug release properties, pharmacokinetics and pharmacodynamics were also investigated in detail. RESULTS: After lyophilization, the nanosuspensions could be redispersed gently and with a narrow particle size distribution, but the particle size has no obvious change. The powder X-ray diffraction and differential scanning calorimetry of FVT nanosuspensions showed that FVT existed in amorphous form in nanosuspensions. In vitro release, FVT nanosuspensions have sustained-release properties. Meanwhile, FVT nanosuspensions could significantly modify the pharmacokinetic profile and increase the bioavailability of FVT by more than 2.4-fold in comparison with the FVT capsules group. In vivo irritation test showed that there was almost no evidence of hemorrhagic mucosal erosion and intestinal villus destruction in rat gastric mucosa. CONCLUSION: The combination of nanocrystallization and cyclodextrin complexation techniques is a new attempt to formulate poorly water-soluble FVT.


Subject(s)
Cyclodextrins/pharmacokinetics , Fluvastatin/pharmacokinetics , Nanoparticles/chemistry , Animals , Biological Availability , Cyclodextrins/administration & dosage , Cyclodextrins/chemistry , Drug Stability , Drug Tolerance , Fluvastatin/administration & dosage , Fluvastatin/chemistry , Hydrogen-Ion Concentration , Nanoparticles/administration & dosage , Rats , Rats, Wistar , Suspensions/administration & dosage , Suspensions/chemistry , Suspensions/pharmacokinetics
10.
Curr Drug Deliv ; 15(9): 1245-1260, 2018.
Article in English | MEDLINE | ID: mdl-30033871

ABSTRACT

BACKGROUND: FR&D scientists continuously try to increase the in vivo performance of low soluble and bioavailable drugs. Solid SMEDDS and liquisolid formulations are relatively simple to develop and fall within the novel drug delivery approaches. Here, a comparison is made to know relative superiority. OBJECTIVE: The study aimed to conduct comparative pharmacokinetic (PK) and pharmacodynamic (PD) studies of developed Fluvastatin (FLU) solid SMEDDS (SSMED) and liquisolid formulation (LS) for their relative in vivo efficacy. METHOD: FLU liquid SMEDDS were optimized by central composite design (CCD). Components, oil, surfactant and co-surfactant were selected as variables; particle size, self-emulsifying time and % drug release in 15min were selected as responses. L-SMEDDS with positive charge inducer were adsorbed on to porous carriers and characterized. Liquisolid formulations were prepared with Avicel PH-102 and Neusilin US2 as carriers. RESULTS: Optimized L-SMEDDS contained 24.92 mg of oil, 45.18 mg of surfactant and 34.28 mg of cosurfactant. SSMEDs containing Syloid XDP (SSMED-XDP) as carrier was selected based on flow properties and liquid retention potential. The average particle size of SSMED-XDP was 154.30±1.10 nm, PDI was 0.311±0.03 and ZP was +19.57±1.34 mV after dilution. The drug release from SSMEDXDP and LS formulations was higher than FLU powder. The bioavailability of SSMEDs was increased by 3.00 fold and that of LS by 1.49 fold more than FLU-suspension. SSMEDs showed 12 h, while LS and suspension showed only 6 h lipid-lowering effect. CONCLUSION: The development of solid SMEDDS resulted in superior performance in both PK and PD effects over the LS formulation.


Subject(s)
Antihypertensive Agents/pharmacokinetics , Fluvastatin/pharmacokinetics , Administration, Oral , Animals , Antihypertensive Agents/administration & dosage , Biological Availability , Chemistry, Pharmaceutical , Emulsions/administration & dosage , Emulsions/pharmacokinetics , Fluvastatin/administration & dosage , Male , Particle Size , Rats , Rats, Wistar , Solubility , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...