Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 142
Filter
1.
Int J Nanomedicine ; 14: 8589-8601, 2019.
Article in English | MEDLINE | ID: mdl-31802871

ABSTRACT

BACKGROUND: Saquinavir mesylate (SQR) tablets are widely used against human immunodeficiency virus. SQR has bioavailability issues owing to its poor aqueous solubility, extensive first-pass metabolism, and even low gastrointestinal tract permeability and absorption. OBJECTIVE: An in-depth optimization process was carried out using factorial design to improve the permeation parameters and thereby the bioavailability of SQR by formulating self-nanoemulsifying drug delivery system (SNEDDS)-loaded polymeric transdermal films. METHODS: The solubility of SQR in different nanoemulsion components was examined. Various combinations of selected components were prepared in an extreme vertices mixture design to identify the useful nanoemulsion zone and to develop SNEDDS with minimum globule size. The optimized SQR-SNEDDS was loaded in polyvinyl alcohol (PVA)-based transdermal films. The Box-Behnken design was used to optimize and evaluate SQR permeability. The prepared films were characterized for thickness, tensile strength, elongation, folding endurance, and accelerated stability studies. The optimized film was examined for ex vivo skin permeation and in vivo pharmacokinetic parameters. RESULTS: The optimized SQR-SNEDDS was prepared in proportions of 0.1, 0.55, and 0.35 of clove oil, labrasol, and Transcutol, respectively. The implemented Box-Behnken design indicated the optimized film consisted of 1.0% PVA, 0.25% propylene glycol, and clove oil as the oil phase. The tensile strength, thickness, percent elongation, and folding endurance of the optimized SQR-SNEDDS film were 0.93 ± 0.013 kg/cm2, 0.22 ± 0.006 mm, 43.1 ± 0.022%, and >200 times, respectively. A higher Cmax and double the AUC were observed for SQR-SNEDDS-loaded film in comparison to pure SQR-loaded films. CONCLUSION: Implementation of a two-step design to optimize and control experimental factors in the preparation of SQR-SNEDDS and its loading onto PVA-based transdermal films was achieved. The films indicated improved ex vivo skin permeation, enhanced bioavailability, and overcame the limitations of the oral dosage form.


Subject(s)
Emulsions/chemistry , Nanoparticles/chemistry , Saquinavir/pharmacology , Administration, Cutaneous , Administration, Oral , Animals , Drug Delivery Systems , Humans , Male , Permeability , Phase Transition , Rats, Wistar , Saquinavir/blood , Saquinavir/pharmacokinetics , Skin/drug effects , Solubility , Surface-Active Agents/chemistry , Tablets
2.
Eur J Drug Metab Pharmacokinet ; 42(1): 135-141, 2017 Feb.
Article in English | MEDLINE | ID: mdl-26846485

ABSTRACT

BACKGROUND AND OBJECTIVES: Although lipid-based drug delivery systems have gained much importance in recent years due to their ability to improve the solubility and bioavailability of poorly soluble drugs, compartmental pharmacokinetic analyses have not been extensively explored. The oral pharmacokinetics of commercial liquid formulation and a developed semisolid system containing saquinavir mesylate (SQVM) were compared in Beagle dogs. A compartmental analysis after intravenous bolus administration of this drug (1 mg/kg) was also performed. METHOD: Pharmacokinetic profiles were analyzed using both non-compartmental and compartmental approaches. Plasma concentration of the drug was determined by high-performance liquid chromatography/tandem mass spectrometry (LC/MS/MS). RESULTS: The disposition curve of SQVM given intravenously was better described by a three-compartment model. In contrast, plasma profiles obtained following the oral administration were fitted to a two-compartment model with lag time due to the fact that the distribution phase was masked by the absorption phase in these formulations. CONCLUSION: The proposed semisolid lipid system was found to be a promising formulation for commercial purposes given the similarity of SQVM absorption rate to that from the commercial liquid formulation.


Subject(s)
Drug Delivery Systems/methods , Saquinavir/administration & dosage , Saquinavir/pharmacokinetics , Administration, Intravenous , Administration, Oral , Animals , Biological Availability , Dogs , Emulsions , Lipids/chemistry , Lipids/pharmacokinetics , Male , Models, Biological , Saquinavir/blood , Saquinavir/chemistry
3.
Drug Res (Stuttg) ; 66(9): 484-488, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27409329

ABSTRACT

The study was aim to assess the impact of biochanin A on the oral bioavailability and pharmacokinetics (PK) of saquinavir (SQV), a substrate of P-glycoprotein (P-gp), in rats. 10 male rats were randomized into 2 groups of equal size, and administered orally 30 mg/kg SQV with or without 20 mg/kg biochanin A. The PK of SQV was assessed using non-compartmental analysis. Results revealed that the area under the plasma concentration-time curve of SQV from time zero to time infinity (AUC0-∞) was reduced by 51.39% by biochanin A (P=0.038); while the apparent systemic clearance (CL/F) was increased by 87.62% (P=0.028). Double peak phenomenon was observed in the plasma SQV profiles. Biochanin A increased the first peak, yet decreased the second peak of plasma SQV levels. Our study demonstrates that biochanin A can significantly reduce SQV oral bioavailability and alter SQV PK profiles in rats. Findings in this study suggest a precaution in the clinic when SQV is administered with dietary/herbal supplements that contain biochanin A.


Subject(s)
Biological Availability , Genistein/pharmacology , Saquinavir/pharmacokinetics , Administration, Oral , Animals , Drug Interactions , Male , Rats , Saquinavir/administration & dosage , Saquinavir/blood
4.
Pharm Res ; 33(8): 1998-2009, 2016 08.
Article in English | MEDLINE | ID: mdl-27154460

ABSTRACT

PURPOSE: Since the adoption of highly active antiretroviral therapy, HIV disease progression has slowed across the world; however, patients are often required to take multiple medications daily of poorly bioavailable drugs via the oral route, leading to gastrointestinal irritation. Recently, long acting antiretroviral injectables that deliver drug for months at a time have moved into late phase clinical trials. Unfortunately, these solid phase crystal formulations have inherent drawbacks in potential dose dumping and a greater likelihood for burst release of drug compared to polymeric formulations. METHODS: Using electrospinning, acetalated dextran scaffolds containing the protease inhibitor saquinavir were created. Grinding techniques were then used to process these scaffolds into injectables which are termed saquinavir microconfetti. Microconfetti was analyzed for in vitro and in vivo release kinetics. RESULTS: Highly saquinavir loaded acetalated dextran electrospun fibers were able to be formed and processed into saquinavir microconfetti while other polymers such as poly lactic-co-glycolic acid and polycaprolactone were unable to do so. Saquinavir microconfetti release kinetics were able to be tuned via drug loading and polymer degradation rates. In vivo, a single subcutaneous injection of saquinavir microconfetti released drug for greater than a week with large tissue retention. CONCLUSIONS: Microconfetti is a uniquely tunable long acting injectable that would reduce the formation of adherence related HIV resistance. Our findings suggest that the injectable microconfetti delivery system could be used for long acting controlled release of saquinavir and other hydrophobic small molecule drugs.


Subject(s)
Dextrans/administration & dosage , Drug Carriers/administration & dosage , Drug Liberation , HIV Protease Inhibitors/administration & dosage , Saquinavir/administration & dosage , Acetylation , Animals , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/metabolism , Dextrans/blood , Drug Carriers/metabolism , Female , HIV Protease Inhibitors/blood , Injections, Subcutaneous , Mice , Mice, Inbred ICR , Saquinavir/blood , Time Factors
5.
Acta Pharmacol Sin ; 36(9): 1151-60, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26256404

ABSTRACT

AIM: Saquinavir (SQV) is the first protease inhibitor for the treatment of HIV infection, but with poor solubility. The aim of this study was to prepare a colloidal nanocrystal suspension for improving the oral absorption of SQV. METHODS: SQV nanocrystals were prepared using anti-solvent precipitation-high pressure homogenization method. The nanocrystals were characterized by a Zetasizer and transmission electron microscopy (TEM). Their dissolution, cellular uptake and transport across the human colorectal adenocarcinoma cell line (Caco-2) monolayer were investigated. Bioimaging of ex vivo intestinal sections of rats was conducted with confocal laser scanning microscopy. Pharmacokinetic analysis was performed in rats administered nanocrystal SQV suspension (50 mg/kg, ig), and the plasma SQV concentrations were measured with HPLC. RESULTS: The SQV nanocrystals were approximately 200 nm in diameter, with a uniform size distribution. The nanocrystals had a rod-like shape under TEM. The dissolution, cellular uptake, and transport across a Caco-2 monolayer of the nanocrystal formulation were significantly improved compared to those of the coarse crystals. The ex vivo intestinal section study revealed that the fluorescently labeled nanocrystals were located in the lamina propria and the epithelium of the duodenum and jejunum. Pharmacokinetic study showed that the maximal plasma concentration (Cmax) was 2.16-fold of that for coarse crystalline SQV suspension, whereas the area under the curve (AUC) of nanocrystal SQV suspension was 1.95-fold of that for coarse crystalline SQV suspension. CONCLUSION: The nanocrystal drug delivery system significantly improves the oral absorption of saquinavir.


Subject(s)
HIV Protease Inhibitors/pharmacokinetics , Saquinavir/pharmacokinetics , Administration, Oral , Animals , Caco-2 Cells , Crystallization/methods , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/blood , HIV Protease Inhibitors/chemistry , Humans , Intestinal Absorption , Intestinal Mucosa/metabolism , Male , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Rats , Rats, Sprague-Dawley , Saquinavir/administration & dosage , Saquinavir/blood , Saquinavir/chemistry , Solubility
6.
Drug Metab Pharmacokinet ; 30(3): 221-6, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25882073

ABSTRACT

Modulation of CYP3A and/or P-gp function by several excipients has been reported. However, relatively few studies have investigated their effects in humans. Therefore, the aim of this clinical study was to clarify the effects of Cremophor EL on the inhibition of CYP3A and P-gp in the human small intestine. Eight healthy Japanese subjects received an oral dose of saquinavir (2 mg, substrate of P-gp/CYP3A) or fexofenadine (50 µg, substrate of P-gp) without or with Cremophor EL (720 mg and 1440 mg). Significant increases in Cmax (1.3-fold) and AUC0-24 (1.6-fold) were observed for fexofenadine when administered with 1440 mg of Cremophor EL. In contrast, a significant decrease was observed for saquinavir when administered with 720 mg of Cremophor EL. The equilibrium dialysis experiment was performed to investigate the micellar interaction between Cremophor EL and drugs. The equilibrium dialysis study showed that saquinavir was far extensively entrapped into the micelles. The reduced concentration of free saquinavir by entrapping in micelles was considered to cause the reduction of systemic exposure for saquinavir. In conclusion, this clinical study suggests that Cremophor EL at least inhibits P-gp in the human small intestine.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Cytochrome P-450 CYP3A/metabolism , Excipients/pharmacology , Glycerol/analogs & derivatives , Saquinavir/blood , Terfenadine/analogs & derivatives , Absorption, Physiological , Administration, Oral , Cross-Over Studies , Excipients/chemistry , Glycerol/chemistry , Glycerol/pharmacology , Healthy Volunteers , Humans , Male , Saquinavir/administration & dosage , Saquinavir/chemistry , Substrate Specificity , Terfenadine/administration & dosage , Terfenadine/blood , Terfenadine/chemistry
7.
Biomed Chromatogr ; 28(12): 1874-80, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24944096

ABSTRACT

An ionic liquid-based dispersive liquid-liquid microextraction followed by RP-HPLC determination of the most commonly prescribed protease inhibitor, saquinavir, in rat plasma was developed and validated. The effects of different ionic liquids, dispersive solvents, extractant/disperser ratio and salt concentration on sample recovery and enrichment were studied. Among the ionic liquids investigated, 1-butyl-3-methylimidazolium hexafluorophosphate was found to be most effective for extraction of saquinavir from rat serum. The recovery was found to be 95% at an extractant/disperser ratio of 0.43 using 1-butyl-3-methylimidazolium hexafluorophosphate and methanol as extraction and dispersive solvents. The recovery was further enhanced to 99.5% by addition of 5.0% NaCl. A threefold enhancement in detection and quantification limits was achieved, at 0.01 and 0.03 µg/mL, compared with the conventional protein precipitation method. A linear relationship was observed in the range of 0.035-10.0 µg/mL with a correlation coefficient (r(2) ) of 0.9996. The method was validated and applied to study pharmacokinetics of saquinavir in rat serum.


Subject(s)
Chromatography, Reverse-Phase/methods , Liquid Phase Microextraction/methods , Saquinavir/blood , Saquinavir/isolation & purification , Animals , Chromatography, High Pressure Liquid/methods , Imidazoles/chemistry , Ionic Liquids/chemistry , Limit of Detection , Linear Models , Male , Methanol/chemistry , Rats , Rats, Wistar , Reproducibility of Results , Saquinavir/chemistry , Saquinavir/pharmacokinetics , Sodium Chloride
8.
J Am Soc Mass Spectrom ; 25(6): 1083-6, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24711229

ABSTRACT

Drug monitoring is usually performed by liquid chromatography coupled with optical detection or electrospray ionization mass spectrometry. More recently, matrix-assisted laser desorption/ionization (MALDI) in combination with triple quadrupole or Fourier-transform (FT) mass analyzers has also been reported to allow accurate quantification. Here, we present a strategy that employs standard MALDI time-of-flight (TOF) mass spectrometry (MS) for the sensitive and accurate quantification of saquinavir from an extract of blood peripheral mononuclear cells. Unambiguous identification of saquinavir in the mass spectra was possible because of using internal mass calibration and by an overall low chemical noise in the low mass range. Exact mass determination of the constant background peaks of the cell extract, which were used for recalibration, was performed by an initial MALDI-FT-MS analysis. Fast and multiplexed sample analysis was enabled by microarray technology, which provided 10 replicates in the lower nL range for each sample in parallel lanes on a chip. In order to validate the method, we employed various statistical tests, such as confidence intervals for linear regressions, three quality control samples, and inverse confidence limits of the estimated concentration ratios.


Subject(s)
Anti-Retroviral Agents/blood , Leukocytes, Mononuclear/chemistry , Microarray Analysis/methods , Saquinavir/blood , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Humans
9.
Eur J Pharm Sci ; 49(5): 800-4, 2013 Aug 16.
Article in English | MEDLINE | ID: mdl-23748001

ABSTRACT

The present study aimed to evaluate the effect of curcumin-loaded solid dispersion on the pharmacokinetics of saquinavir in rats. Solid dispersion (SD) formulation was prepared with Solutol® HS15 to improve the solubility and bioavailability of curcumin. Subsequently, its inhibition effect on P-gp mediated cellular efflux was examined by using NCI/ADR-RES cells overexpressing P-gp. Compared to the untreated curcumin, SD formulation enhanced the cellular uptake of rhodamine-123, a P-gp substrate by approximately 3 folds in NCI/ADR-RES cells. The oral and intravenous pharmacokinetics of saquinavir were also determined in rats with/without curcumin in the different formulations. Compared to the control given saquinavir alone, curcumin-loaded solid dispersion significantly (p<0.05) increased the oral exposure of saquinavir in rats, while it did not affect the intravenous pharmacokinetics of saquinavir. The AUC and Cmax of oral saquinavir increased by 3.8- and 2.7-folds, respectively in the presence of curcumin-loaded solid dispersion. In contrast, the untreated curcumin did not affect the oral pharmacokinetics of saquinavir. These results suggest that SD formulation of curcumin should be effective to improve the in vivo effectiveness of curcumin as an absorption enhancer, leading to the improved oral exposure of saquinavir.


Subject(s)
Curcumin/administration & dosage , HIV Protease Inhibitors/pharmacokinetics , Saquinavir/pharmacokinetics , Administration, Oral , Animals , Biological Availability , Cell Line, Tumor , Curcumin/chemistry , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/blood , Humans , Male , Polyethylene Glycols/chemistry , Rats , Rats, Sprague-Dawley , Saquinavir/administration & dosage , Saquinavir/blood , Stearic Acids/chemistry , Suspensions
10.
Bioanalysis ; 4(10): 1227-34, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22651566

ABSTRACT

Ionization matrix effects are one of the most difficult issues in LC-MS bioanalysis that are without a good and universal solution. Most people in the field are aware of it, but some are not sure how to deal with it. Many laboratories do not routinely assess matrix effects in method validation or substitute it with a different experiment that only indirectly and partially test matrix effects. Others, when matrix effects are mentioned, immediately link them with phospholipids and try to use means to remove them from the samples (e.g., SPE), without understanding if the phospholipids can really impact the analytes. In this article, key issues related to matrix effects will be examined, and methods to address matrix effect problems will be evaluated for effectiveness and practicality.


Subject(s)
Chromatography, High Pressure Liquid/methods , Mass Spectrometry/methods , Pharmaceutical Preparations/analysis , Animals , Excipients/analysis , Excipients/chemistry , Humans , Models, Chemical , Phospholipids/analysis , Phospholipids/chemistry , Rats , Saquinavir/blood , Sensitivity and Specificity
11.
Ther Drug Monit ; 33(6): 772-7, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22105596

ABSTRACT

Antiretroviral therapy during pregnancy is critical to preventing human immunodeficiency virus vertical transmission. Physiological changes during pregnancy can alter drug kinetics. The aim of this study was to assess the pharmacokinetics (PK) of saquinavir (SQV) boosted with ritonavir during pregnancy and postpartum. Fourteen human immunodeficiency virus-positive pregnant women started SQV 500 mg new tablet formulation plus ritonavir at a dose of 1000/100 mg twice a day + 2 nucleoside retrotranscriptase inhibitors during pregnancy. At weeks 24 and 34 of pregnancy and 6 weeks postpartum, a 12-hour PK study was conducted. PK parameters were calculated using Win Nolin software version 4.1. At week 24, the geometric mean values for SQV area under the plasma concentration-time curve from 0-12 hours (AUC0₋12), the maximum observed plasma concentration (C(max)), trough plasma concentration (C(min)), and the elimination half-life (t(1/2)) were 24.80 mg·h⁻¹·mL⁻¹, 4.66 mg/mL, 0.93 mg/mL, and 4.31 hours, respectively. At week 34, AUC0₋12, C(max), C(min), and t(1/2) were 12.71 mg·h⁻¹·mL⁻¹, 3.23 mg/mL, 0.26 mg/mL, and 4.06 hours, respectively. Finally, at 6 weeks postpartum, mean values for SQV AUC0₋12, C(max), C(min), and t(1/2) were 28.94 mg·h⁻¹·mL⁻¹, 3.92 mg/mL, 0.86 mg/mL, and 3.60 hours, respectively. Although PK parameters in week 24 and postpartum were very similar, those for week 34 showed an important reduction: -71.20%, -30.61%, -48.73%, and -5.81% in C(min), C(max), AUC0₋12, and t(1/2), respectively, compared with week 24, but no statistically significant differences were shown between patients. No vertical transmissions were reported. Therapeutic drug monitoring of SQV during pregnancy should be considered, mainly during the third trimester, to ensure adequate drug exposure throughout the entire pregnancy.


Subject(s)
HIV Protease Inhibitors/pharmacokinetics , HIV Seropositivity/blood , HIV Seropositivity/drug therapy , Pregnancy Complications, Infectious/blood , Pregnancy Complications, Infectious/drug therapy , Ritonavir/pharmacokinetics , Saquinavir/pharmacokinetics , Adult , Antiretroviral Therapy, Highly Active/adverse effects , Body Mass Index , Cohort Studies , Female , HIV Protease Inhibitors/adverse effects , HIV Protease Inhibitors/blood , HIV Protease Inhibitors/therapeutic use , HIV Seropositivity/complications , HIV Seropositivity/metabolism , Half-Life , Humans , Infectious Disease Transmission, Vertical/prevention & control , Overweight/complications , Pilot Projects , Postpartum Period , Pregnancy , Pregnancy Complications, Infectious/metabolism , Pregnancy Trimester, Second , Pregnancy Trimester, Third , Prospective Studies , Ritonavir/adverse effects , Ritonavir/blood , Ritonavir/therapeutic use , Saquinavir/adverse effects , Saquinavir/blood , Saquinavir/therapeutic use
12.
J Pharmacol Exp Ther ; 334(1): 21-32, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20348205

ABSTRACT

It is currently unknown whether gestational diabetes mellitus (GDM), a prevalent obstetrical complication, compounds the changes in drug disposition that occur naturally in pregnancy. Hyperlipidemia occurs in GDM. Using a rat model of GDM, we determined whether excess lipids compete with drugs for plasma protein binding. Because lipids activate nuclear receptors that regulate drug transporters and metabolic enzymes, we used proteome analysis to determine whether hyperlipidemia indirectly leads to the dysregulation of these proteins in the liver. GDM was induced on gestational day 6 (GD6) via streptozotocin injection. Controls received either vehicle alone or streptozotocin with subsequent insulin treatment. Liver and plasma were collected on GD20. Glyburide and saquinavir protein binding was determined by ultrafiltration, and an established solvent method was used for plasma delipidation. Proteomics analysis was performed by using isobaric tags for relative and absolute quantitation methodology with membrane-enriched hepatic protein samples. Relative to controls, GDM rat plasma contained more cholesterol and triglycerides. Plasma protein binding of glyburide and saquinavir was decreased in GDM. Delipidation normalized protein binding in GDM plasma. Proteins linked to lipid metabolism were strongly affected in the GDM proteomics data set, with prohyperlipidemic and antihyperlipidemic changes observed, and formed networks that implicated several nuclear receptors. Up-regulation of drug transporters and metabolic enzymes was observed (e.g., multidrug resistance 1/2, CYP2A1, CYP2B9, and CYP2D3). In this study, GDM-induced hyperlipidemia decreased protein binding and was associated with drug transporter and metabolic enzyme up-regulation in the liver. Both of these findings could change drug disposition in affected pregnancies, compounding changes associated with pregnancy itself.


Subject(s)
Blood Proteins/metabolism , Carrier Proteins/biosynthesis , Cytochrome P-450 Enzyme System/biosynthesis , Diabetes Mellitus, Experimental/blood , Diabetes, Gestational/blood , Hyperlipidemias/blood , Liver , Animals , Binding, Competitive , Biological Transport , Chromatography, High Pressure Liquid , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/metabolism , Diabetes, Gestational/drug therapy , Diabetes, Gestational/enzymology , Diabetes, Gestational/metabolism , Down-Regulation , Female , Gestational Age , Glyburide/blood , Glyburide/pharmacokinetics , Glyburide/therapeutic use , Hyperlipidemias/complications , Hyperlipidemias/enzymology , Hyperlipidemias/metabolism , Insulin/therapeutic use , Lipid Metabolism/drug effects , Lipids/blood , Liver/enzymology , Liver/metabolism , Pregnancy , Protein Binding , Proteomics , Rats , Rats, Sprague-Dawley , Saquinavir/blood , Saquinavir/pharmacokinetics , Saquinavir/therapeutic use , Streptozocin , Tandem Mass Spectrometry , Up-Regulation
13.
J Clin Pharmacol ; 50(4): 392-400, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20097936

ABSTRACT

The relationship between plasma protease inhibitor (PI) trough concentrations and hyperlipidemic effects were evaluated retrospectively using data from 2 pilot clinical trials of a double-boosted PI regimen (saquinavir/lopinavir/ritonavir) in 25 HIV patients. The patients' median age was 39 years (range, 25-60). At baseline, PI-naive patients had a median viral load of 53 500 copies/mL and median CD4 of 296 cells/mm(3), while PI-experienced patients had 37 750 copies/mL and 214 cells/mm(3). Plasma PI trough concentrations of saquinavir, lopinavir, and ritonavir at week 12 were 520, 4482, and 153 ng/mL, respectively. At week 12, median fasting lipids increased significantly from baseline: total cholesterol increased from 165 to 189 mg/dL (P = .0005) and the triglyceride increased from 113 to 159 mg/dL (P = .001). There were no associations between PI trough concentrations at week 12 and the percent total cholesterol change at week 12. No associations were found between PI trough concentrations and lipid changes in HIV patients on a double-boosted PI regimen (saquinavir/lopinavir/ritonavir). Factors other than systemic exposure to PIs (such as host or genetic factors) may modulate the hyperlipidemic effect of PIs.


Subject(s)
HIV Infections/blood , HIV Protease Inhibitors/blood , Lipids/biosynthesis , Adult , Clinical Trials as Topic/methods , Drug Therapy, Combination , Female , HIV Infections/drug therapy , HIV Infections/enzymology , HIV Protease Inhibitors/administration & dosage , Humans , Hyperlipidemias/blood , Hyperlipidemias/diagnosis , Lopinavir , Male , Middle Aged , Pilot Projects , Prospective Studies , Pyrimidinones/administration & dosage , Pyrimidinones/blood , Retrospective Studies , Ritonavir/administration & dosage , Ritonavir/blood , Saquinavir/administration & dosage , Saquinavir/blood
14.
AIDS ; 23(9): 1176-9, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19451794

ABSTRACT

Currently, the optimal boosting dose for saquinavir is unknown. Therefore, we evaluated the pharmacokinetics profiles in a cross over setting comparing saquinavir/ritonavir 1500/50 mg (plus NRTI backbone) to saquinavir/ritonavir 1500/100 mg in the same HIV-infected, Thai individuals. The 50% reduction of ritonavir boosting did not result in a change in the pharmacokinetics of saquinavir, whereas the ritonavir exposure was significantly lower when a dose of 50 mg was administered.


Subject(s)
Anti-HIV Agents/administration & dosage , HIV Infections/drug therapy , HIV-1/drug effects , Ritonavir/administration & dosage , Saquinavir/administration & dosage , Adult , Anti-HIV Agents/blood , Anti-HIV Agents/pharmacokinetics , Cross-Over Studies , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , HIV Infections/blood , Humans , Male , Middle Aged , Ritonavir/blood , Ritonavir/pharmacokinetics , Saquinavir/blood , Saquinavir/pharmacokinetics
15.
J Pharm Biomed Anal ; 49(4): 1034-9, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19269771

ABSTRACT

The aim of this work is to present a new concept, called on-line desorption of dried blood spots (on-line DBS), allowing the direct analysis of a dried blood spot coupled to liquid chromatography mass spectrometry device (LC/MS). The system is based on an inox cell which can receive a blood sample (10 microL) previously spotted on a filter paper. The cell is then integrated into LC/MS system where the analytes are desorbed out of the paper towards a column switching system ensuring the purification and separation of the compounds before their detection on a single quadrupole MS coupled to atmospheric pressure chemical ionisation (APCI) source. The described procedure implies that no pretreatment is necessary in spite the analysis is based on whole blood sample. To ensure the applicability of the concept, saquinavir, imipramine, and verapamil were chosen. Despite the use of a small sampling volume and a single quadrupole detector, on-line DBS allowed the analyses of these three compounds over their therapeutic concentrations from 50 to 500 ng/mL for imipramine and verapamil and from 100 to 1000 ng/mL for saquinavir. Moreover, the method showed good repeatability with relative standard deviation (RSD) lower than 15% based on two levels of concentration (low and high). Function responses were found to be linear over the therapeutic concentration for each compound and were used to determine the concentrations of real patient samples for saquinavir. Comparison of the founded values with those of a validated method used routinely in a reference laboratory showed a good correlation between the two methods. Moreover, good selectivity was observed ensuring that no endogenous or chemical components interfered with the quantitation of the analytes. This work demonstrates the feasibility and applicability of the on-line DBS procedure for bioanalysis.


Subject(s)
Blood Chemical Analysis/methods , Adsorption , Anti-HIV Agents/blood , Calcium Channel Blockers/blood , Chromatography, High Pressure Liquid , Humans , Mass Spectrometry , Online Systems , Reference Standards , Reproducibility of Results , Saquinavir/blood , Solvents , Verapamil/blood
16.
Clin Chem ; 55(1): 170-4, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19028822

ABSTRACT

BACKGROUND: Most HPLC-UV methods for therapeutic drug monitoring of anti-HIV drugs have long run times, which reduce their applicability for high-throughput analysis. We developed an ultra-performance liquid chromatography (UPLC)-diode array detection method for the simultaneous quantification of the HIV-protease inhibitors (PIs) amprenavir, atazanavir, indinavir, lopinavir, nelfinavir, ritonavir, saquinavir, and tipranavir (TPV), and the nonnucleoside reverse transcriptase inhibitors (NNRTIs) efavirenz and nevirapine. METHODS: Solid-phase extraction of 1 mL plasma was performed with Waters HLB cartridges. After 3 wash steps, we eluted the drugs with methanol, evaporated the alcohol, and reconstituted the residue with 50 microL methanol. We injected a 4-microL volume into the UPLC system (Waters ACQUITY UPLC BEH C8 column maintained at 60 degrees C) and used a linear gradient of 50 mmol/L ammonium acetate and 50 mmol/L formic acid in water versus acetonitrile to achieve chromatographic separation of the drugs and internal standard (A-86093). Three wavelengths (215, 240, and 260 nm) were monitored. RESULTS: All drugs were eluted within 15 min. Calibration curves with concentrations of 0.025-10 mg/L (1.875-75 mg/L for TPV) showed coefficients of determination (r(2)) between 0.993 and 0.999. The lower limits of quantification were well below the trough concentrations reported in the literature. Inter- and intraassay CVs and the deviations between the nominal and measured concentrations were <15%. The method was validated by successful participation in an international interlaboratory QC program. CONCLUSIONS: This method allows fast and simultaneous quantification of all commercially available PIs and NNRTIs for therapeutic drug monitoring.


Subject(s)
HIV Protease Inhibitors/blood , Reverse Transcriptase Inhibitors/blood , Alkynes , Atazanavir Sulfate , Benzoxazines/blood , Carbamates/blood , Chromatography, High Pressure Liquid , Cyclopropanes , Furans , Humans , Indinavir/blood , Lopinavir , Nelfinavir/blood , Nevirapine/blood , Oligopeptides/blood , Pyridines/blood , Pyrimidinones/blood , Pyrones/blood , Reproducibility of Results , Ritonavir/blood , Saquinavir/blood , Sensitivity and Specificity , Solid Phase Extraction , Sulfonamides/blood
18.
J Chromatogr B Analyt Technol Biomed Life Sci ; 872(1-2): 68-76, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18657486

ABSTRACT

We present herein an ultra-fast quantitative assay for the quantitation of saquinavir in human plasma, without prior chromatographic separation, with matrix-assisted laser desorption/ionization using the selected reaction monitoring quantitation mode (MALDI-SRM/MS). The method was found to be linear from 5 to 10,000 ng/ml using pentadeuterated saquinavir (SQV-d5) as an internal standard, and from 5 to 1000 ng/ml using reserpine as internal standard (IS). Accuracy and precision were in the range of 101-108%, 3.9-11% with SQV-d5 and in the range 93-108%, 3.5-15% with reserpine. Plasma samples (250 microl) were extracted with a mixture of ethyl acetate/hexane. MALDI spotting of the extract was automated using electrodeposition and the dried droplet method using alpha-cyano-4-hydroxycinnamic acid (CHCA) as matrix. A 96 spots MALDI plate was prepared within 20 min in a fully unattended manner. Each sample was spotted four times and quantitation was based on the average of their analyte/IS area ratio. Samples were analyzed on a triple quadrupole linear ion trap (QqQ(LIT)) equipped with a high repetition laser source (1000 Hz). The analysis time of one sample was approximately 6 s, therefore 96 samples could be analyzed in less than 10 min. With liquid-liquid extraction sample preparation no significant matrix effects were observed. Moreover, the assay showed sufficient selectivity for samples to be analyzed at the lower limit of quantification (LLOQ) in the presence of other antiretroviral drugs, without prior chromatographic steps. In parallel, to assess the selectivity of the assay with real samples, a liquid chromatography (LC)-SRM/MS method was developed and a cross validation with clinical samples was successfully performed.


Subject(s)
HIV Protease Inhibitors/blood , Saquinavir/blood , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Automation , Humans , Sensitivity and Specificity
19.
Acta Pol Pharm ; 65(1): 93-100, 2008.
Article in English | MEDLINE | ID: mdl-18536180

ABSTRACT

Combination therapy with protease inhibitors (PIs) is used for the treatment of patients infected with the human immunodeficiency virus (HIV). To achieve optimal drug concentrations for viral suppression and avoidance of drug toxicity, therapeutic drug monitoring of PIs levels has been considered essential. In this study an analytical procedure for simultaneous monitoring the plasma concentrations of a total four protease inhibitors: indinavir, lopinavir, nelfinavir and saquinavir was presented. The plasma samples were liquid/liquid extracted and subjected to high performance liquid chromatography (HPLC) analysis. The lopinavir and saquinavir in the patient plasma samples were monitored by ultraviolet detection at 215 nm. Extraction procedure using methyl tert-butyl ether and the mobile phase consisted from acetonitrile with phosphate buffer on a Symmetry C18 column were used to monitor these two compounds. Linearity of the method was obtained in the concentration range of 0.1 - 15.0 mg/mL for all four protease inhibitors. Under steady state conditions, the plasma concentrations of protein inhibitors in 23 patients were determined and area under the plasma concentration-time curve was estimated to maintain optimal viral suppression. We developed a simple and specific method for the simultaneous determination of lopinavir and saquinavir.


Subject(s)
Chromatography, High Pressure Liquid/methods , Drug Monitoring/methods , HIV Infections/drug therapy , HIV Protease Inhibitors/blood , Adult , Area Under Curve , Drug Combinations , HIV Protease Inhibitors/pharmacokinetics , Humans , Indinavir/blood , Indinavir/pharmacokinetics , Lopinavir , Middle Aged , Nelfinavir/blood , Nelfinavir/pharmacokinetics , Pyrimidinones/blood , Pyrimidinones/pharmacokinetics , Saquinavir/blood , Saquinavir/pharmacokinetics
20.
Drug Metab Dispos ; 36(5): 863-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18256206

ABSTRACT

The objective of this investigation was to differentiate the roles of P-glycoprotein (Pgp), multidrug resistance-associated protein 2 (Mrp2), and CYP3A on saquinavir (SQV) oral absorption. With use of single-pass jejunal perfusion (in situ) and portal vein-cannulated rats (in vivo), SQV absorption was studied under chemical inhibition of Pgp [N-(4-[2-(1,2,3,4-tetrahydro-6,7-dimethoxy-2 isoquinolinyl)-ethyl]-phenyl)-9,10-dihydro-5-methoxy-9-oxo-4-acridine carboxamide (GF120918)], Mrp2 [(3-(((3-(2-(7-chloro-2-quinolinyl)-(E)-ethenyl)phenyl) ((3-(dimethylamino-3-oxopropyl)thio)methyl)-thio) propanoic acid (MK571)], and/or CYP3A (midazolam). Plasma concentrations of SQV and related metabolites were analyzed by liquid chromatography-tandem mass spectrometry. When given alone, SQV absorption was extremely low both in situ (F(a) = 0.07%) and in vivo [C(max) = 0.068 microg/ml; area under the curve (AUC) = 6.8 microg x min/ml]. Coadministration of GF120918 boosted SQV absorption by more than 20-fold with decreased variation in AUCs (percent coefficient of variation = 30% versus 100%). In contrast, coadministration of MK571 or midazolam increased SQV absorption only 2- to 3-fold without improving the variation in AUCs. SQV oral absorption was not further improved when it was given with GF120918 and midazolam or with GF120918 and MK571. The current results provide, for the first time, direct and explicit evidence that the low oral absorption of SQV is controlled by a secretory transporter, Pgp, and not by limited passive diffusion owing to its poor physicochemical properties. Pgp-mediated transport is also responsible for the highly variable oral bioavailability of SQV. In contrast, intestinal Mrp2 and intestinal CYP3A appear to play minor roles in SQV oral bioavailability. Given the differential and complex roles of Pgp and CYP3A in SQV oral absorption, the optimization of AIDS boosting regimens requires careful consideration to avoid therapy-limiting drug-drug transporter and enzyme interactions.


Subject(s)
Cytochrome P-450 CYP3A/metabolism , HIV Protease Inhibitors/pharmacokinetics , Multidrug Resistance-Associated Proteins/metabolism , Saquinavir/pharmacokinetics , Administration, Oral , Animals , Biological Availability , HIV Protease Inhibitors/blood , HIV Protease Inhibitors/metabolism , Intestinal Absorption , Male , Multidrug Resistance-Associated Protein 2 , Rats , Rats, Sprague-Dawley , Saquinavir/blood , Saquinavir/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...