Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
BMC Pharmacol Toxicol ; 25(1): 57, 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39187897

ABSTRACT

Febuxostat is commonly used in clinic for the treatment of hyperuricemia. Multiple-peak phenomenon has been observed in human plasma concentration-time profiles of febuxostat, but has not been paid enough attention in previous research. This study takes a pivotal step forward by conducting a comprehensive population pharmacokinetic (PopPK) analysis of febuxostat in a healthy Chinese cohort, with a central focus on delineating its absorption profile under contrasting fasting and fed conditions, while concurrently assessing the influence of food alongside other potential covariates on febuxostat's PK profile. The plasma concentration data used for modeling was obtained from two bioequivalence (BE) studies. Subjects were administered febuxostat 20 mg or 80 mg under fasting or fed condition. Goodness-of-fit plots, visual predict check (VPC), and normalized prediction distribution error (NPDE) were used for model evaluation. Based on the established model, PK profiles in healthy Caucasian subjects were simulated with parameter adjustment for race difference on clearance and bioavailability. Data from 128 subjects were used in the PopPK analysis. Febuxostat concentration-time curves were described by a two-compartment model with two deposit absorption compartments and lag times (Tlag). Prandial states (Food) showed significant impact on absorption rate ka1 and ka2, as well as Tlag1, and body weight was identified as a significant covariate on the apparent distribution volume. The PopPK analysis of febuxostat in healthy Chinese volunteers, under both fasted and fed conditions, successfully characterized its PK profile and underscored the significant influence of food on absorption. The potential difference of absorption between Chinese population and Caucasian population indicated from the simulations needs further investigation.


Subject(s)
Fasting , Febuxostat , Food-Drug Interactions , Gout Suppressants , Models, Biological , Febuxostat/pharmacokinetics , Febuxostat/blood , Humans , Male , Adult , Gout Suppressants/pharmacokinetics , Gout Suppressants/blood , Gout Suppressants/administration & dosage , Female , Young Adult , Middle Aged , Asian People , Therapeutic Equivalency , Biological Availability , Cross-Over Studies , White People
2.
Biopharm Drug Dispos ; 42(2-3): 78-84, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33400825

ABSTRACT

The present study was aimed to develop a novel sustained-release formulation for allopurinol (ALP/SR) with the use of a pH-sensitive polymer, hydroxypropyl methylcellulose acetate succinate, to reduce nephrotoxicity. ALP/SR was evaluated in terms of crystallinity, the dissolution profile, pharmacokinetic behavior, and nephrotoxicity in a rat model of nephropathy. Under acidic conditions (pH1.2), sustained release behavior was seen for ALP/SR, although both crystalline ALP and ALP/SR exhibited rapid dissolution at neutral condition. After multiple oral administrations of ALP samples (10 mg-ALP/kg) for 4 days in a rat model of nephropathy, ALP/SR led to a low and sustained plasma concentration of ALP, as evidenced by half the maximum concentration of ALP and a 2.5-fold increase in the half-life of ALP compared with crystalline ALP, possibly due to suppressed dissolution behavior under acidic conditions. Repeated-dosing of ALP/SR resulted in significant reductions in plasma creatinine and blood urea nitrogen levels by 73% and 69%, respectively, in comparison with crystalline ALP, suggesting the low nephrotoxic risk of ALP/SR. From these findings, a strategic SR formulation approach might be an efficacious dosage option for ALP to avoid severe nephrotoxicity in patients with nephropathy.


Subject(s)
Allopurinol/pharmacokinetics , Gout Suppressants/pharmacokinetics , Methylcellulose/analogs & derivatives , Administration, Oral , Allopurinol/adverse effects , Allopurinol/blood , Allopurinol/chemistry , Animals , Antineoplastic Agents , Blood Urea Nitrogen , Cisplatin , Creatinine/blood , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Drug Liberation , Gout Suppressants/adverse effects , Gout Suppressants/blood , Gout Suppressants/chemistry , Half-Life , Kidney/drug effects , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Male , Methylcellulose/chemistry , Methylcellulose/pharmacokinetics , Rats, Sprague-Dawley
3.
J Clin Pharm Ther ; 46(2): 333-342, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33201513

ABSTRACT

WHAT IS KNOWN AND OBJECTIVE: Febuxostat is a well-known drug for treating hyperuricemia and gout. The published methods for determination of febuxostat in human plasma might be unsuitable for high-throughput determination and widespread application. We need to develop a highly selective, sensitive and rapid liquid chromatography-tandem mass spectrometry method. METHODS: The chromatographic separation was achieved on a Hypersil Gold-C18 (2.1 mm × 100 mm, 1.9 µm) column with mobile phase A (Water containing 0.1% formic acid) and mobile phase B (acetonitrile containing 0.1% formic acid). Multiple reaction monitoring (MRM) mode was used for quantification using target ions at m/z 315.3 â†’ m/z 271.3 for febuxostat and m/z 324.3 â†’ m/z 280.3 for Febuxostat-d9 (IS). A backpropagation artificial neural network (BPANN) pharmacokinetic model was constructed by the data of bioequivalence study. RESULTS AND DISCUSSION: After the LC-MS/MS method validated, it was successfully applied to the bioequivalence study of 30 human volunteers under fed condition. The predicted concentrations generated by BPANN model had a high correlation coefficient with experimental values. WHAT IS NEW AND CONCLUSION: A sensitive LC-MS/MS method had been developed and validated for determination of febuxostat in healthy subjects under fed condition, and a BPANN model was developed that can be used to predict the plasma concentration of febuxostat.


Subject(s)
Chromatography, Liquid/methods , Febuxostat/blood , Febuxostat/pharmacokinetics , Gout Suppressants/blood , Neural Networks, Computer , Tandem Mass Spectrometry/methods , Area Under Curve , Chromatography, Liquid/standards , Drug Stability , Half-Life , Healthy Volunteers , Humans , Metabolic Clearance Rate , Reproducibility of Results , Tandem Mass Spectrometry/standards
4.
Medicine (Baltimore) ; 98(11): e14850, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30882682

ABSTRACT

RATIONALE: A gouty tophus, arising from the deposition of monosodium urate crystals (MSU), rarely occurs in the nasal bridge. There have been only 7 documented cases of a gouty tophus in the nasal bridge from 1978 to 2018 in English-language literature. PATIENT CONCERNS: A 65-year-old male had a chief complaint of a lump in the nasal bridge that was slowly growing for over 1 year. DIAGNOSIS: MSU crystals were confirmed through ultrasonography (US) and pathological examinations. INTERVENTIONS: A cosmetically less destructive method, ultrasound-guided fine needle aspiration cytology (FNAC) was used to approach the mass lesion of nasal bridge. OUTCOMES: The diagnosis was confirmed as a gouty tophus without performing a nasal subdermal exploration. LESSONS: This case report is the first use of US with FNAC to approach and diagnosed a gouty tophus in the nasal bridge.


Subject(s)
Arthritis, Gouty/diagnosis , Nose/abnormalities , Aged , Arthritis, Gouty/complications , Biopsy, Fine-Needle/methods , C-Reactive Protein/analysis , Colchicine/analysis , Colchicine/blood , Febuxostat/analysis , Febuxostat/blood , Gout/drug therapy , Gout Suppressants/analysis , Gout Suppressants/blood , Humans , Male , Nose/physiopathology , Prevalence , Tomography, X-Ray Computed/methods , Uric Acid/analysis , Uric Acid/blood
5.
Clin Pharmacol Drug Dev ; 8(5): 647-656, 2019 07.
Article in English | MEDLINE | ID: mdl-30748125

ABSTRACT

Two clinical studies were performed in healthy volunteers to investigate food and antacid effects on lesinurad, a novel selective uric acid reabsorption inhibitor approved for treatment of hyperuricemia associated with gout in combination with xanthine oxidase inhibitors. Study 1 evaluated a high-fat, high-calorie meal or high doses of antacids (3000 mg calcium carbonate or 1600 mg magnesium hydroxide/1600 mg aluminum hydroxide) on the pharmacokinetics (PK) and pharmacodynamics (PD) of 400 mg oral lesinurad. Study 2 evaluated low doses of antacids (1250 mg calcium carbonate or 800 mg magnesium hydroxide/800 mg aluminum hydroxide) on the PK and PD of 400 mg lesinurad. Food did not alter the plasma AUC of lesinurad and only reduced its Cmax by 18%. In the fasted conditions, high-dose calcium carbonate reduced the Cmax and AUC of lesinurad by 54% and 38%, respectively, whereas high-dose magnesium hydroxide/aluminum hydroxide reduced Cmax and AUC by 36% and 31%, respectively. Food enhanced the maximum serum urate (sUA)-lowering effect of lesinurad by approximately 20% despite reducing the Cmax of lesinurad. High-dose calcium carbonate decreased the urate-lowering effect approximately 20% in the first 6 hours, whereas high-dose magnesium hydroxide/aluminum hydroxide reduced the effect by 26%. Low-dose calcium carbonate or magnesium hydroxide/aluminum hydroxide in the presence of food did not significantly affect plasma lesinurad Cmax and AUC or the sUA lowering and renal handling of uric acid. In summary, study results suggest food did not meaningfully alter lesinurad PK and PD. High doses of antacids reduced lesinurad AUC up to 40% and reduced the lesinurad uric acid-lowering effect.


Subject(s)
Aluminum Hydroxide/pharmacology , Antacids/pharmacology , Calcium Carbonate/pharmacology , Food-Drug Interactions , Gout Suppressants , Magnesium Hydroxide/pharmacology , Thioglycolates , Triazoles , Uric Acid/blood , Adolescent , Adult , Cross-Over Studies , Dietary Fats/administration & dosage , Drug Combinations , Gout Suppressants/blood , Gout Suppressants/pharmacokinetics , Gout Suppressants/pharmacology , Gout Suppressants/urine , Healthy Volunteers , Humans , Male , Middle Aged , Thioglycolates/blood , Thioglycolates/pharmacokinetics , Thioglycolates/pharmacology , Thioglycolates/urine , Triazoles/blood , Triazoles/pharmacokinetics , Triazoles/pharmacology , Triazoles/urine , Young Adult
7.
Talanta ; 176: 318-328, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-28917757

ABSTRACT

A newly developed thin-layer chromatographic (TLC) method coupled with fluorescence detection for specific determination of Febuxostat (FEB) was designed. The proposed method adopts exposure of FEB on a developed TLC plate to hydrochloric acid vapors, resulting in a large enhancement of its weak fluorescence, permitting its specific and sensitive determination in real human plasma and urine after excitation at 345nm on 60 F254 silica gel plates using toluene-ethyl acetate-methanol-glacial acetic acid; (30:10:5:0.1,v/v/v/v) as mobile phase. The retention factor (Rf) value for FEB was 0.33 ± 0.03 with a correlation coefficient of 0.9974 in the concentration range of 2.5-50ng/band. Upon using polynomial regression, the correlation coefficient was greatly improved (0.9999), with detection and quantification limits of 0.55 and 1.67 (ng/band), respectively. The proposed method was validated according to the International Conference of Harmonization and was successfully used for specific and selective determination of FEB in its commercial dosage form without excipient interference. Moreover, the proposed method was extended to efficient determination of the studied drug in real human plasma and urine samples in the presence of its metabolites without any interference, allowing clinical application of the proposed method for direct FEB determination in biological fluids as well as in pharmacokinetics studies and for quality control of the pharmaceutical dosage form without sample pretreatment or exhausting extraction steps.


Subject(s)
Chromatography, Thin Layer/methods , Febuxostat/blood , Febuxostat/urine , Gout Suppressants/blood , Gout Suppressants/urine , Febuxostat/pharmacokinetics , Fluorescence , Gout Suppressants/pharmacokinetics , Humans , Limit of Detection , Reproducibility of Results , Silica Gel/chemistry , Solvents/chemistry
8.
Eur J Clin Pharmacol ; 73(1): 71-78, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27683090

ABSTRACT

PURPOSE: The aims of this study were to characterise the population pharmacokinetics of oxypurinol in patients receiving haemodialysis and to compare oxypurinol exposure in dialysis and non-dialysis patients. METHODS: Oxypurinol plasma concentrations from 6 gout people receiving haemodialysis and 19 people with gout not receiving dialysis were used to develop a population pharmacokinetic model in NONMEM. Deterministic simulations were used to predict the steady-state area under the oxypurinol plasma concentration time curve over 1 week (AUC7days). RESULTS: The pharmacokinetics of oxypurinol were best described by a one-compartment model with a separate parameter for dialytic clearance. Allopurinol 100 mg daily produced an AUC7days of 279 µmol/L h in dialysis patients, a value 50-75 % lower than the AUC7days predicted for patients with normal renal function taking 200 to 400 mg daily (427-855 µmol/L h). Dosing pre-dialysis resulted in about a 25-35 % reduction in exposure compared to post-dialysis. CONCLUSIONS: Oxypurinol is efficiently removed by dialysis. The population dialytic and total (non-dialytic) clearance of oxypurinol were found to be 8.23 and 1.23 L/h, standardised to a fat-free mass of 70 kg and creatinine clearance of 6 L/h, respectively. Our results suggest that if the combination of low-dose allopurinol and haemodialysis does not result in sustained urate lowering below treatment targets (serum urate ≤0.36 mmol/L), then allopurinol doses may be increased to optimise oxypurinol exposure.


Subject(s)
Allopurinol/pharmacokinetics , Gout Suppressants/pharmacokinetics , Models, Biological , Oxypurinol/blood , Renal Dialysis , Adult , Aged , Aged, 80 and over , Allopurinol/blood , Female , Gout/blood , Gout/drug therapy , Gout/metabolism , Gout Suppressants/blood , Humans , Male , Middle Aged
9.
Pharmacogenomics J ; 17(2): 201-203, 2017 03.
Article in English | MEDLINE | ID: mdl-26810134

ABSTRACT

Many patients fail to achieve the recommended serum urate (SU) target (<6 mgdl-1) with allopurinol. The aim of our study was to examine the association of ABCG2 with SU target in response to standard doses of allopurinol using a cohort with confirmed adherence. Good response was defined as SU<6 mgdl-1 on allopurinol ⩽300 mgd-1 and poor response as SU⩾6 mgdl-1 despite allopurinol >300 mgd-1. Adherence was confirmed by oxypurinol concentrations. ABCG2 genotyping was performed using pre-designed single nucleotide polymorphism (SNP) TaqMan assays. Of 264 patients, 120 were good responders, 68 were poor responders and 76 were either non-adherent or could not be classified. The minor allele of ABCG2 SNP rs2231142 conferred a significantly increased risk of poor response to allopurinol (odds ratio=2.71 (1.70-4.48), P=6.0 × 10-5). This association remained significant after adjustment for age, sex, body mass index, ethnicity, estimated glomerular filtration rate, diuretic use and SU off urate-lowering therapy. ABCG2 rs2231142 predicts poor response to allopurinol, as defined by SU⩾6 mgdl-1 despite allopurinol >300 mgd-1.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Allopurinol/therapeutic use , Gout Suppressants/therapeutic use , Gout/drug therapy , Neoplasm Proteins/genetics , Pharmacogenomic Variants , Polymorphism, Single Nucleotide , Adult , Aged , Aged, 80 and over , Allopurinol/blood , Biomarkers/blood , Female , Gene Frequency , Genotype , Gout/blood , Gout/genetics , Gout Suppressants/blood , Humans , Logistic Models , Male , Middle Aged , Odds Ratio , Oxypurinol/blood , Pharmacogenetics , Phenotype , Risk Factors , Treatment Outcome , Uric Acid/blood , Young Adult
10.
Biol Pharm Bull ; 39(6): 1013-21, 2016.
Article in English | MEDLINE | ID: mdl-27251504

ABSTRACT

Whether renal dysfunction influences the hypouricemic effect of febuxostat, a xanthine oxidase (XO) inhibitor, in patients with hyperuricemia due to overproduction or underexcretion of uric acid (UA) remains unclear. We aimed to address this question with a modeling and simulation approach. The pharmacokinetics (PK) of febuxostat were analyzed using data from the literature. A kinetic model of UA was retrieved from a previous human study. Renal UA clearance was estimated as a function of creatinine clearance (CLcr) but non-renal UA clearance was assumed constant. A reversible inhibition model for bovine XO was adopted. Integrating these kinetic formulas, we developed a PK-pharmacodynamic (PK-PD) model for estimating the time course of the hypouricemic effect of febuxostat as a function of baseline UA level, febuxostat dose, treatment duration, body weight, and CLcr. Using the Monte Carlo simulation method, we examined the performance of the model by comparing predicted UA levels with those reported in the literature. We also modified the models for application to hyperuricemia due to UA overproduction or underexcretion. Thirty-nine data sets comprising 735 volunteers or patients were retrieved from the literature. A good correlation was observed between the hypouricemic effects of febuxostat estimated by our PK-PD model and those reported in the articles (observed) (r=0.89, p<0.001). The hypouricemic effect was estimated to be augmented in patients with renal dysfunction irrespective of the etiology of hyperuricemia. While validation in clinical studies is needed, the modeling and simulation approach may be useful for individualizing febuxostat doses in patients with various clinical characteristics.


Subject(s)
Febuxostat , Gout Suppressants , Hyperuricemia/blood , Models, Biological , Renal Insufficiency, Chronic/blood , Uric Acid/blood , Computer Simulation , Febuxostat/blood , Febuxostat/pharmacokinetics , Febuxostat/pharmacology , Febuxostat/therapeutic use , Gout Suppressants/blood , Gout Suppressants/pharmacokinetics , Gout Suppressants/pharmacology , Gout Suppressants/therapeutic use , Humans , Hyperuricemia/drug therapy , Xanthine/blood , Xanthine Oxidase/antagonists & inhibitors
11.
J Med Assoc Thai ; 98 Suppl 3: S46-50, 2015 Apr.
Article in English | MEDLINE | ID: mdl-26387388

ABSTRACT

BACKGROUND: Effective treatment in gouty arthritis can prevent joint and renal damage. Target serum uric acid levels of < 6 mg/dl and < 5 mg/dl are recommended in gouty arthritis and those with tophi, respectively. OBJECTIVE: To evaluate: (i) whether patients achieved recommended serum uric acid target and assess influencing factors and (ii) renal function between patients who achieved and not achieved the goal. MATERIAL AND METHOD: The medical records of gouty arthritis patients treated in outpatient department at Thammasat University Hospital between January 2013 and December 2013 were reviewed. Patients were divided into adequately (ATG) and inadequately treated groups (ITG) based on the ACR uric acid criteria after six months of treatment. Factors associated with inadequate treatment were explored and post treatment renal function compared between A and ITGs. RESULTS: Of 139 patients, 46 (33%) achieved target serum uric acid concentrations. Alcoholic consumption was the significant factor influencing the outcome. 75.5% of patients were followed-up > 1 month for second evaluation of uric acid and most of them not receiving dosage up-titration even though not achieving the target. Both groups had similar alterations of renal function after treatment (p = 0.68). CONCLUSION: Most patients failed to achieve recommended uric acid targets. Alcohol consumption was identified as a key risk factorfor a suboptimal outcome. The treat-to-target approach should be underlined. Other risk factors should be explored prospectively.


Subject(s)
Allopurinol/therapeutic use , Arthritis, Gouty/drug therapy , Benzbromarone/therapeutic use , Alcohol Drinking/blood , Allopurinol/blood , Arthritis, Gouty/blood , Benzbromarone/blood , Female , Follow-Up Studies , Gout Suppressants/blood , Gout Suppressants/therapeutic use , Humans , Kidney Function Tests , Male , Middle Aged , Retrospective Studies , Risk Factors , Treatment Outcome , Uric Acid/blood , Uricosuric Agents/blood , Uricosuric Agents/therapeutic use
12.
Pharmacology ; 96(1-2): 90-8, 2015.
Article in English | MEDLINE | ID: mdl-26183164

ABSTRACT

The aim of the present study was to determine the influence of severe renal dysfunction (estimated glomerular filtration rate <30 ml/min/1.73 m(2), including hemodialysis) on the pharmacokinetics and therapeutic effects of febuxostat using a population pharmacokinetic analysis. This study recruited patients with hyperuricemia who were initially treated with allopurinol, but were switched to febuxostat, and it consists of 2 sub-studies: a pharmacokinetic study (26 patients) and retrospective efficacy evaluation study (51 patients). The demographic and clinical data of patients were collected from electronic medical records. Plasma febuxostat concentrations were obtained at each hospital visit. Population pharmacokinetic modeling was performed with NONMEM version 7.2. A total of 128 plasma febuxostat concentrations from 26 patients were used in the population pharmacokinetic analysis. The data were best described by a 1-compartment model with first order absorption. Covariate analysis revealed that renal function did not influence the pharmacokinetics of febuxostat, whereas actual body weight significantly influenced apparent clearance and apparent volume of distribution. The retrospective efficacy analysis showed the favorable therapeutic response of febuxostat switched from allopurinol in patients with moderate to severe renal impairment. No serious adverse event associated with febuxostat was observed irrespective of renal function. The population pharmacokinetic analysis and therapeutic analysis of febuxostat revealed that severe renal dysfunction had no influence on the pharmacokinetic parameters of febuxostat. These results suggest that febuxostat is tolerated well by patients with severe renal impairment.


Subject(s)
Febuxostat/pharmacokinetics , Febuxostat/therapeutic use , Hyperuricemia/drug therapy , Renal Insufficiency/drug therapy , Adult , Aged , Aged, 80 and over , Febuxostat/adverse effects , Febuxostat/blood , Female , Gout Suppressants/adverse effects , Gout Suppressants/blood , Gout Suppressants/pharmacokinetics , Gout Suppressants/therapeutic use , Humans , Hyperuricemia/blood , Hyperuricemia/complications , Male , Middle Aged , Renal Insufficiency/blood , Renal Insufficiency/complications , Retrospective Studies
13.
Int J Clin Pharmacol Ther ; 53(8): 667-73, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25997544

ABSTRACT

OBJECTIVE: Febuxostat is a selective inhibitor of xanthine oxidase, which is used to manage hyperuricemia in patients with gout. The objective of the study was to compare the pharmacokinetics of two different strength of febuxostat formulations (80 mg and 40 mg). METHODS: A randomized, single-dose, open-label, two-period, two-sequence crossover study with a 7-day washout period was conducted in 30 healthy male subjects. Participants received either reference (1 — 80 mg) or test (2 — 40 mg) formulations during the first period and the alternative formulation during the second period. Plasma samples for the drug analysis were collected up to 24 hours after treatment. RESULTS: All pharmacokinetic parameters were comparable between the two formulations The observed mean Cmax, AUC(last), and AUC(∞) values for the reference formulation were 3,670 ng/mL, 12,086 ng x h/mL, and 12,880 ng x h/mL, respectively. Corresponding values for the test formulation were 4,108 ng/mL, 12,689 ng x h/mL, and 13,278 ng x h/mL, respectively. The geometric mean ratios (90% CI) between the two formulations were 1.1273 (1.0286 - 1.2355) for Cmax, 1.054 (1.0115 - 1.0980) for AUC(last), and 1.0395 (0.9959 - 1.0851) for AUC(∞). The changes of serum uric acid at 24 hours after reference and test formations were comparable (-1.36 mg/dL for reference and -1.37 mg/dL for test; p = 0.892). CONCLUSION: The results of the present study indicated that the reference and test formulations have comparable pharmacokinetics and that these two formulations meet the regulatory criteria for bioequivalence. In addition, the reduction of serum UA levels in the reference formulation was similar to that of the test formulation after a single dose.


Subject(s)
Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Gout Suppressants/administration & dosage , Gout Suppressants/pharmacokinetics , Thiazoles/administration & dosage , Thiazoles/pharmacokinetics , Uric Acid/blood , Adult , Area Under Curve , Biomarkers/blood , Cross-Over Studies , Down-Regulation , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/blood , Febuxostat , Gout Suppressants/adverse effects , Gout Suppressants/blood , Half-Life , Healthy Volunteers , Humans , Linear Models , Male , Metabolic Clearance Rate , Middle Aged , Models, Biological , Republic of Korea , Therapeutic Equivalency , Thiazoles/adverse effects , Thiazoles/blood , Treatment Outcome , Xanthine Oxidase/antagonists & inhibitors , Xanthine Oxidase/metabolism , Young Adult
14.
Clin Nephrol ; 83(5): 286-92, 2015 May.
Article in English | MEDLINE | ID: mdl-25816806

ABSTRACT

AIMS: Phase 3 trial data indicate that treatment of chronic tophaceous gout with pegloticase, a recombinant uricase conjugated to polyethylene glycol, does not reduce estimated glomerular filtration rate in chronic kidney disease (CKD) patients and that pegloticase therapeutics are independent of CKD stages 1 - 4. We determined the pharmacokinetics/pharmacodynamics of pegloticase after a single-dose in non-gout subjects with stage 5 CKD receiving hemodialysis. METHODS: In this open-label phase 1 study, 12 subjects received a single intravenous dose of pegloticase 8 mg 3 hours prior to hemodialysis. Blood samples for determination of serum pegloticase concentrations and serum uric acid (SUA) levels were collected immediately predose and at regular intervals before, during, and after hemodialysis. RESULTS: Mean serum pegloticase concentrations remained stable and were unaffected by dialysis sessions. Mean SUA fell to undetectable levels within 3 hours and remained undetected for up to 72 hours postdose. CONCLUSION: Our findings indicate no significant effect of hemodialysis on either the stability of serum pegloticase concentrations after a single dose or the capacity of pegloticase to lower SUA. No new safety signals were detected. Administration of pegloticase in patients with comorbid chronic tophaceous gout and endstage renal failure requiring hemodialysis appears feasible.


Subject(s)
Gout Suppressants/pharmacokinetics , Kidney Failure, Chronic/therapy , Polyethylene Glycols/pharmacokinetics , Renal Dialysis , Urate Oxidase/pharmacokinetics , Adult , Biomarkers/blood , Drug Monitoring , Female , Gout Suppressants/administration & dosage , Gout Suppressants/adverse effects , Gout Suppressants/blood , Humans , Infusions, Intravenous , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/diagnosis , Male , Middle Aged , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/adverse effects , Renal Dialysis/adverse effects , Risk Assessment , Urate Oxidase/administration & dosage , Urate Oxidase/adverse effects , Urate Oxidase/blood , Uric Acid/blood
15.
Acta Pharmacol Sin ; 35(11): 1447-52, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25283504

ABSTRACT

AIM: Pharmacodynamic analysis of intravenous recombinant urate oxidase produced by Escherichia coli was performed in healthy subjects using a pharmacokinetic/pharmacodynamic (PK/PD) model. METHODS: A randomized, single-blind, placebo-controlled study was performed in 40 healthy Chinese subjects (4 groups of 10 subjects each, placebo 4:1 ratio) who received infusions of uricase (single doses of 0.1, 0.2, and 0.3 mg/kg; multiple doses of 0.2 mg·kg(-1)·d(-1) for 7 d). PK profiles were determined through plasma uricase activity, and PD profiles were established using uric acid levels in plasma and urine. The plasma PD parameter was estimated as changes in plasma uric acid levels as the effect in the indirect response model. Adverse events were also monitored. RESULTS: A two-compartment PK model with constant iv input and first-order output was used to describe the kinetic process of plasma uricase. The low value (2.8 U/L) of drug concentration that achieved 50% of maximum effect (EC50) indicated that low plasma uricase concentrations were sufficient to produce pharmacological effects. A strong relationship (r(2)=0.9991) between the mean uric acid concentration in blood and the mean uric acid excretion rate in urine in the range of 11 to 30 h after single dosing was found. Infusions of uricase were well tolerated in all subjects. CONCLUSION: The PK/PD model predicted the effective dose to be 0.1 mg/kg in healthy subjects. The excretion rate of uric acid in urine may be used as a new index for pharmacological effects in further clinical trials.


Subject(s)
Gout Suppressants/administration & dosage , Gout Suppressants/pharmacokinetics , Models, Biological , Urate Oxidase/administration & dosage , Urate Oxidase/pharmacokinetics , China , Drug Dosage Calculations , Female , Gout Suppressants/blood , Healthy Volunteers , Humans , Infusions, Intravenous , Male , Recombinant Proteins/administration & dosage , Recombinant Proteins/blood , Recombinant Proteins/pharmacokinetics , Renal Elimination , Single-Blind Method , Urate Oxidase/blood , Uric Acid/blood , Uric Acid/urine , Young Adult
16.
Rheumatology (Oxford) ; 53(12): 2167-74, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24509406

ABSTRACT

OBJECTIVE: The aim of this study was to evaluate the pharmacodynamics (PDs), pharmacokinetics (PKs) and safety of lesinurad (selective uric acid reabsorption inhibitor) in combination with febuxostat (xanthine oxidase inhibitor) in patients with gout. METHODS: This study was a phase IB, multicentre, open-label, multiple-dose study of gout patients with serum uric acid (sUA) >8 mg/dl following washout of urate-lowering therapy with colchicine flare prophylaxis. Febuxostat 40 or 80 mg/day was administered on days 1-21, lesinurad 400 mg/day was added on days 8-14 and then lesinurad was increased to 600 mg/day on days 15-21. sUA, urine uric acid and PK profiles were evaluated at the end of each week. Safety was assessed by adverse events, laboratory tests and physical examinations. RESULTS: Initial treatment with febuxostat 40 or 80 mg/day monotherapy resulted in 67% and 56% of subjects, respectively, achieving a sUA level <6 mg/dl. Febuxostat 40 or 80 mg/day plus lesinurad 400 or 600 mg/day resulted in 100% of subjects achieving sUA <6 mg/dl and up to 100% achieving sUA <5 mg/dl. No clinically relevant changes in the PKs of either drug were noted. The combination was well tolerated. CONCLUSION: The clinically important targets of sUA <6 mg/dl and <5 mg/dl are achievable in 100% of patients when combining lesinurad and febuxostat.


Subject(s)
Gout Suppressants/administration & dosage , Gout/blood , Hyperuricemia/blood , Thiazoles/administration & dosage , Thioglycolates/administration & dosage , Triazoles/administration & dosage , Adult , Aged , Colchicine/blood , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Therapy, Combination , Febuxostat , Female , Gout/drug therapy , Gout Suppressants/adverse effects , Gout Suppressants/blood , Humans , Hyperuricemia/drug therapy , Male , Middle Aged , Thiazoles/adverse effects , Thiazoles/blood , Thioglycolates/adverse effects , Thioglycolates/blood , Triazoles/adverse effects , Triazoles/blood , Uric Acid/blood
17.
Rheumatology (Oxford) ; 53(6): 988-97, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24196384

ABSTRACT

Rapid and effective suppression of inflammation is a primary goal in the treatment of rheumatic diseases. However, the therapeutic effect of most medications may be slow to manifest, in the order of weeks or months in the case of DMARDs. Monitoring of drug concentrations allows the possibility of appropriate dose adjustment or changes in medication to achieve more rapid or better outcomes. We review the evidence for drug concentration monitoring. Despite the theoretical utility for monitoring of MTX polyglutamate concentrations in red blood cells in patients with RA, studies have not shown a clear association between concentrations and either efficacy or toxicity and routine measurement is not yet recommended. Small studies associating disease control with concentrations of anti-TNF therapies and anti-drug antibodies suggest that routine monitoring may be useful in the future. However, the data are not yet sufficient for this recommendation. With the use of allopurinol in gout, there is a putative therapeutic range for the active metabolite oxypurinol; however, adjusting the allopurinol dose to achieve a target urate concentration is likely to be most effective, and measuring oxypurinol may be best suited to assessing drug adherence. Although measuring thiopurine metabolite concentrations with AZA therapy has been shown to be useful in IBD, studies in rheumatic diseases have so far failed to confirm a useful association between concentrations and disease control or drug toxicity. Whole blood concentrations of HCQ have been associated with disease control in SLE and future studies may be able to determine a therapeutic range.


Subject(s)
Antirheumatic Agents/blood , Drug Monitoring/methods , Rheumatic Diseases/blood , Allopurinol/blood , Allopurinol/therapeutic use , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/drug therapy , Gout Suppressants/blood , Gout Suppressants/therapeutic use , Humans , Methotrexate/blood , Methotrexate/therapeutic use , Rheumatic Diseases/drug therapy , Tumor Necrosis Factor-alpha/antagonists & inhibitors
18.
Clin Pharmacol Drug Dev ; 3(5): 353-7, 2014 09.
Article in English | MEDLINE | ID: mdl-27129007

ABSTRACT

Allopurinol is the most commonly used urate-lowering therapy in gout. This study was undertaken to evaluate the pharmacokinetics and relative bioavailability of two brands of allopurinol tablets. The in vivo study was established according to a single-center, randomized, single-dose, laboratory-blinded, Two Way, Cross-Over Study with a washout period of 1 week. Under fasting conditions, 24 healthy male volunteers were randomly allocated to receive a single oral dose (200 mg) of either test and reference formulations. Plasma samples were obtained over a 6-hour interval and analyzed for allopurinol by reversed phase liquid chromatography with ultraviolet detection. The 90% confidence intervals for the ratio of log transformed values of Cmax , AUC0-t , and AUCt-∞ of the two treatments were within the acceptable range (0.8-1.25) for bioequivalence. From PK perspectives, the two allopurinol formulations were considered bioequivalent, based on the rate and extent of absorption. No adverse events occurred or were reported after a single 200-mg allopurinol and both formulations were well tolerated.


Subject(s)
Allopurinol/administration & dosage , Allopurinol/pharmacokinetics , Gout Suppressants/administration & dosage , Gout Suppressants/pharmacokinetics , Administration, Oral , Adolescent , Adult , Allopurinol/blood , Allopurinol/chemistry , Area Under Curve , Biological Availability , Chromatography, High Pressure Liquid , Cross-Over Studies , Drug Compounding , Drug Monitoring/methods , Egypt , Gout Suppressants/blood , Gout Suppressants/chemistry , Healthy Volunteers , Humans , Male , Metabolic Clearance Rate , Tablets , Therapeutic Equivalency , Young Adult
19.
Article in English | MEDLINE | ID: mdl-24184830

ABSTRACT

Allopurinol is used widely for the treatment of gout, but its pharmacokinetics is complex and some patients show hypersensitivity, necessitating careful monitoring and improved detection methods. In this study, a sensitive and reliable liquid chromatography-tandem mass spectrometry method was developed to determine the concentrations of allopurinol and its active metabolite oxypurinol in human plasma and urine using 2,6-dichloropurine as the internal standard (IS). Analytes and the IS were extracted from 0.5ml aliquots of plasma or urine using ethyl acetate and separated on an Agilent Eclipse Plus C18 column using methanol and ammonium formate-formic acid buffer containing 5mM ammonium formate and 0.1% formic acid (95:5, v/v) as the mobile phase (A) for allopurinol or methanol plus 5mM ammonium formate aqueous solution (95:5, v/v) as the mobile phase (B) for oxypurinol. Allopurinol was detected in positive ion mode and the analysis time was about 7min. The calibration curve was linear from 0.05 to 5µg/mL allopurinol in plasma and 0.5-30µg/mL in urine. The lower limit of quantification (LLOQ) was 0.05µg/mL in plasma and 0.5µg/mL in urine. The intra- and inter-day precision and relative errors of quality control (QC) samples were ≤11.1% for plasma and ≤ 8.7% for urine. Oxypurinol was detected in negative mode with an analysis time of about 4min. The calibration curve was linear from 0.05 to 5µg/mL in plasma (LLOQ, 0.05µg/mL) and from 1 to 50µg/mL in urine (LLOQ, 1µg/mL). The intra- and inter-day precision and relative errors were ≤7.0% for plasma and ≤9.6% for urine. This method was then successfully applied to investigate the pharmacokinetics of allopurinol and oxypurinol in humans.


Subject(s)
Allopurinol/pharmacokinetics , Chromatography, Liquid/methods , Gout Suppressants/pharmacokinetics , Oxypurinol/pharmacokinetics , Tandem Mass Spectrometry/methods , Allopurinol/blood , Allopurinol/urine , Calibration , Gout Suppressants/blood , Gout Suppressants/urine , Humans , Limit of Detection , Oxypurinol/blood , Oxypurinol/urine , Reference Standards , Reproducibility of Results , Spectrometry, Mass, Electrospray Ionization
20.
Biomed Chromatogr ; 27(11): 1406-12, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23733262

ABSTRACT

An improved, simple and highly sensitive LC-MS/MS method has been developed and validated for quantification of febuxostat with 100 µL human plasma using febuxostat-d7 as an internal standard (IS) according to regulatory guidelines. The analyte and IS were extracted from human plasma via liquid-liquid extraction using diethyl ether. The chromatographic separation was achieved on a Zorbax C18 column using a mixture of acetonitrile and 5 mm ammonium formate (60:40, v/v) as the mobile phase at a flow rate of 0.5 mL/min. The total run time was 5.0 min and the elution of febuxostat and IS occurred at 1.0 and 1.5 min, respectively. A linear response function was established for the range of concentrations 1-6000 ng/mL (r > 0.99). The precursor to product ion transitions monitored for febuxostat and IS were m/z 317.1 → 261.1 and 324.2 → 262.1, respectively. The intra- and inter-day precisions (%RSD) were within 1.29-9.19 and 2.85-7.69%, respectively. The proposed method was successfully applied to pharmacokinetic studies in humans.


Subject(s)
Chromatography, High Pressure Liquid/methods , Gout Suppressants/blood , Thiazoles/blood , Febuxostat , Humans , Limit of Detection , Liquid-Liquid Extraction/methods , Male , Tandem Mass Spectrometry/methods , Xanthine Oxidase/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL