Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Bioorg Med Chem Lett ; 40: 127900, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33684442

ABSTRACT

Although benzbromarone (BBR) is a conventional, highly potent uricosuric drug, it is not a standard medicine because it causes rare but fatal fulminant hepatitis. We transformed the bis-aryl ketone structure of BBR to generate novel monocyclic amide-linked phenol derivatives that should possess uric acid excretion activity without adverse properties associated with BBR. The derivatives were synthesized and tested for uric acid uptake inhibition (UUI) in two assays using either urate transporter 1-expressing cells or primary human renal proximal tubule epithelial cells. We also evaluated their inhibitory activity against mitochondrial respiration as a critical mitochondrial toxicity parameter. Some derivatives with UUI activity had no mitochondrial toxicity, including compound 3f, which effectively lowered the plasma uric acid level in Cebus apella. Thus, 3f is a promising candidate for further development as a uricosuric agent.


Subject(s)
Amides/chemistry , Phenol/chemical synthesis , Uric Acid/metabolism , Uricosuric Agents/chemical synthesis , Animals , Benzbromarone/chemistry , Benzbromarone/pharmacology , Drug Evaluation, Preclinical , Glucose Transport Proteins, Facilitative/metabolism , Humans , Mitochondria/drug effects , Phenol/adverse effects , Phenol/pharmacology , Pyrroles/chemistry , Sapajus apella , Signal Transduction , Structure-Activity Relationship , Uric Acid/blood , Uricosuric Agents/adverse effects , Uricosuric Agents/pharmacokinetics
2.
Clin Exp Nephrol ; 24(Suppl 1): 25-35, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31760530

ABSTRACT

BACKGROUND: Dotinurad is a novel, selective urate reabsorption inhibitor, which reduces serum uric acid levels by inhibiting the urate transporter 1 (URAT1). We compared the pharmacokinetics (PK), pharmacodynamics (PD), and safety of dotinurad in subjects with hepatic impairment and normal hepatic function. METHODS: This was a multicenter, open-label, single dose study. A total of 24 subjects were divided into four groups: the normal hepatic function group and the mild, moderate, and severe hepatic impairment groups. The primary endpoints were changes in plasma dotinurad levels and PK parameters. RESULTS: The geometric mean ratio of the maximum plasma concentration (Cmax) [two-sided 90% confidence interval (CI)] of dotinurad in in the mild, moderate, and severe hepatic impairment groups relative to that in the normal hepatic function group was 0.840 (0.674-1.047), 0.798 (0.653-0.976), and 0.747 (0.570-0.979), respectively, showing a lower Cmax in the moderate and severe hepatic impairment groups. Following adjustment for body weight, only the moderate hepatic impairment group had a lower Cmax than the normal hepatic function group. No meaningful differences in other PK parameters were observed between the groups. Regarding the PD of dotinurad, the changes in serum uric acid levels after dosing were similar in all groups. As for safety, no noteworthy concerns were raised in relation to any group. CONCLUSION: The study revealed no clinically meaningful influence of hepatic impairment on the PK, PD, or safety of dotinurad. These findings indicate possibility that dotinurad can be used without dose adjustment in patients with hepatic impairment.


Subject(s)
Benzothiazoles/pharmacokinetics , Liver Diseases/physiopathology , Uric Acid/blood , Uricosuric Agents/pharmacokinetics , Adult , Aged , Benzothiazoles/adverse effects , Benzothiazoles/pharmacology , Female , Humans , Japan , Male , Middle Aged
3.
J Pharmacol Exp Ther ; 371(1): 162-170, 2019 10.
Article in English | MEDLINE | ID: mdl-31371478

ABSTRACT

The effect of dotinurad [(3,5-dichloro-4-hydroxyphenyl)(1,1-dioxo-1,2-dihydro-3H-1λ 6-1,3-benzothiazol-3-yl)methanone] was compared with that of commercially available uricosuric agents-namely, benzbromarone, lesinurad, and probenecid. Its effect on urate secretion transporters was evaluated using probe substrates for respective transporters. Dotinurad, benzbromarone, lesinurad, and probenecid inhibited urate transporter 1 (URAT1) with IC50 values of 0.0372, 0.190, 30.0, and 165 µM, respectively. Dotinurad weakly inhibited ATP-binding cassette subfamily G member 2 (ABCG2), organic anion transporter 1 (OAT1), and OAT3, with IC50 values of 4.16, 4.08, and 1.32 µM, respectively, indicating higher selectivity for URAT1. The hypouricemic effects of dotinurad and benzbromarone were evaluated in Cebus monkeys. Dotinurad, at doses of 1-30 mg/kg, concomitantly decreased plasma urate levels and increased fractional excretion of urate (FEUA) in a dose-dependent manner. On the contrary, benzbromarone, at a dose of 30 mg/kg, showed a modest effect on plasma urate levels. The inhibitory effect of dotinurad on urate secretion transporters was evaluated in Sprague-Dawley rats, with sulfasalazine and adefovir as probe substrates of ABCG2 and OAT1, respectively. Drugs, including febuxostat as a reference ABCG2 inhibitor, were administered orally before sulfasalazine or adefovir administration. Dotinurad had no effect on urate secretion transporters in vivo, whereas benzbromarone, lesinurad, probenecid, and febuxostat increased the plasma concentrations of probe substrates. These results suggested dotinurad is characterized as a selective urate reabsorption inhibitor (SURI), which is defined as a potent URAT1 inhibitor with minimal effect on urate secretion transporters, including ABCG2 and OAT1/3, because of its high efficacy in decreasing plasma urate levels compared with that of other uricosuric agents. SIGNIFICANCE STATEMENT: Our study on the inhibitory effects on urate transport showed that dotinurad had higher selectivity for urate transporter 1 (URAT1) versus ATP-binding cassette subfamily G member 2 (ABCG2) and organic anion transporter (OAT) 1/3 compared to other uricosuric agents. In Cebus monkeys, dotinurad decreased plasma urate levels and increased fractional excretion of urate in a dose-dependent manner. To determine the inhibitory effect of dotinurad on urate secretion transporters, we studied the movement of substrates of ABCG2 and OAT1 in rats. Dotinurad had no effect on these transporters, whereas the other uricosuric agents increased the plasma concentrations of the substrates. These results suggested dotinurad as a potent and selective urate reabsorption inhibitor is characterized by increased efficacy with decreasing plasma urate levels.


Subject(s)
Benzothiazoles/pharmacology , Uricosuric Agents/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Animals , Benzothiazoles/adverse effects , Benzothiazoles/pharmacokinetics , Drug Evaluation, Preclinical , HEK293 Cells , Haplorhini , Humans , Male , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Organic Anion Transport Protein 1/antagonists & inhibitors , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters/antagonists & inhibitors , Organic Anion Transporters/metabolism , Organic Anion Transporters, Sodium-Independent/antagonists & inhibitors , Organic Anion Transporters, Sodium-Independent/metabolism , Organic Cation Transport Proteins/antagonists & inhibitors , Organic Cation Transport Proteins/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley , Uric Acid/blood , Uric Acid/urine , Uricosuric Agents/adverse effects
4.
Rheumatology (Oxford) ; 57(9): 1602-1610, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29868853

ABSTRACT

Objectives: Verinurad (RDEA3170) is a high-affinity inhibitor of the URAT1 transporter in clinical development for treating gout and asymptomatic hyperuricaemia. The aim of this Phase 2a, randomized, open-label study was to investigate the multiple-dose pharmacodynamics, pharmacokinetics and safety of oral verinurad combined with febuxostat vs febuxostat alone and verinurad alone. Methods: Japanese male subjects aged 21-65 years with gout (n = 37) or asymptomatic hyperuricaemia (n = 35) and serum urate (sUA) ⩾8 mg/dl were randomized to febuxostat (10, 20, 40 mg) in combination with verinurad (2.5-10 mg), verinurad alone (2.5-15 mg), febuxostat alone (10, 20, 40 mg) or benzbromarone alone (50 mg). There were four treatment periods per cohort and each treatment period was 7 days. Study drugs were administered once-daily after breakfast. Plasma, serum and urine samples were measured at pre-set intervals on days -1, 7, 14, 21 and 28. Results: Verinurad combined with febuxostat decreased sUA in dose-dependent manner, providing greater sUA lowering than febuxostat alone at the same dose (P < 0.001). Urinary uric acid excretion rate was increased by verinurad, reduced by febuxostat and comparable to baseline for verinurad combined with febuxostat. Verinurad from 2.5 mg to 15 mg was well tolerated, with no withdrawals due to adverse events. Laboratory assessments showed no clinically meaningful changes during combination treatment. Conclusion: Verinurad combined with febuxostat decreased sUA dose-dependently while maintaining uric acid excretion similar to baseline. All dose combinations of verinurad and febuxostat were generally well tolerated. These data support continued investigation of oral verinurad in patients with gout. Trial registration: ClinicalTrials.gov, https://clinicaltrials.gov, NCT02317861.


Subject(s)
Benzbromarone/administration & dosage , Febuxostat/administration & dosage , Gout/drug therapy , Hyperuricemia/drug therapy , Thioglycolates/administration & dosage , Triazoles/administration & dosage , Administration, Oral , Adult , Aged , Benzbromarone/pharmacokinetics , Dose-Response Relationship, Drug , Drug Therapy, Combination , Febuxostat/pharmacokinetics , Female , Follow-Up Studies , Gout/blood , Gout/epidemiology , Gout Suppressants/administration & dosage , Gout Suppressants/pharmacokinetics , Humans , Hyperuricemia/blood , Hyperuricemia/epidemiology , Male , Middle Aged , Organic Anion Transporters/antagonists & inhibitors , Organic Cation Transport Proteins/antagonists & inhibitors , Thioglycolates/pharmacokinetics , Time Factors , Treatment Outcome , Triazoles/pharmacokinetics , Uric Acid/blood , Uricosuric Agents/administration & dosage , Uricosuric Agents/pharmacokinetics , Young Adult
5.
Biol Pharm Bull ; 40(9): 1463-1467, 2017.
Article in English | MEDLINE | ID: mdl-28867729

ABSTRACT

To determine the response of hemodialysis (HD) patients to topiroxostat after a switch from febuxostat, we evaluated the efficacy, tolerability, and serum concentration of topiroxostat in HD patients after the switch. In this 16-month prospective observational study, we assessed the serum uric acid (UA) levels, other laboratory data, and serum topiroxostat concentrations of 10 HD patients who had been receiving febuxostat at a dose of 10 mg/d for over 1 year. No statistical difference was observed between the tolerability index at baseline and 16 months after the switch to topiroxostat. Serum UA after the switch in all patients (attained serum UA levels of ≤6 mg/dL) was 5.6±1.7 mg/dL (60%) at baseline, 4.9±0.5 mg/dL (100%) at 6 months and 5.7±0.4 mg/dL (50%) at 16 months (p=0.25), respectively. In patients with baseline serum UA levels >6 mg/dL, serum UA was significantly reduced at 6 and 16 months compared with baseline. Minimum serum concentrations of serum topiroxostat were lower than the limit of quantification (<25 ng/mL). Our results indicate that a switch from febuxostat 10 mg/d to topiroxostat 40 mg/d might reduce serum UA levels, with no change in other clinical laboratory data over the long term. These effects were more frequent in patients with high serum UA levels. Furthermore, topiroxostat therapy was more cost effective than febuxostat therapy. Thus, topiroxostat therapy could be a better treatment option for HD patients who develop high serum UA levels after febuxostat 10 mg/d administration.


Subject(s)
Febuxostat/therapeutic use , Nitriles/therapeutic use , Pyridines/therapeutic use , Renal Dialysis , Uricosuric Agents/therapeutic use , Adult , Aged , Febuxostat/adverse effects , Febuxostat/pharmacokinetics , Female , Follow-Up Studies , Humans , Hyperuricemia/drug therapy , Male , Middle Aged , Nitriles/adverse effects , Nitriles/pharmacokinetics , Prospective Studies , Pyridines/adverse effects , Pyridines/pharmacokinetics , Uric Acid/blood , Uricosuric Agents/adverse effects , Uricosuric Agents/pharmacokinetics
6.
Drug Metab Dispos ; 42(11): 1955-63, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25187484

ABSTRACT

Tienilic acid (TA) is selectively oxidized at the C-5 position of the thiophene ring by the human liver enzyme cytochrome P450 2C9 (CYP2C9). This oxidation is mediated by the proximal positioning of the thiophene over the heme iron, which is proposed to be coordinated by an interaction of the TA carboxylic acid to a cationic binding pocket in the enzyme active site. In this study, we investigated how chemical modification of TA influences the bioactivation by CYP2C9. For this investigation, nine analogs of TA were chosen with substitutions on either side of the molecule. We tested three parameters, including CYP2C9 inhibition, metabolic profiling, and in silico docking. Of the 10 compounds tested, only two (TA and a noncarboxyl analog) resulted in competitive and time-dependent inhibition of CYP2C9. Metabolic profiling revealed a trend in which substitution of the carboxylate with nonionizable functional groups resulted in metabolic switching from oxidation of the aromatic ring to dealkylation reactions at the opposite side of the structure. The in silico modeling predicted an opposite binding orientation to that of TA for many analogs, including the 3-thenoyl regio-isomer analog, which contradicts previous models. Together these data show that disrupting interactions with the cationic binding pocket of CYP2C9 will impact the sites of metabolism and inhibition of the enzyme.


Subject(s)
Cytochrome P-450 CYP2C9/metabolism , Ticrynafen/metabolism , Cations , Computer Simulation , Diuretics/metabolism , Diuretics/pharmacokinetics , Humans , In Vitro Techniques , Substrate Specificity , Ticrynafen/pharmacokinetics , Uricosuric Agents/metabolism , Uricosuric Agents/pharmacokinetics
7.
Clin Pharmacol Drug Dev ; 12(4): 392-396, 2023 04.
Article in English | MEDLINE | ID: mdl-36317751

ABSTRACT

SHR4640, also named as ruzinurad, is a selective human urate transporter 1 (URAT1) inhibitor developed for the treatment of hyperuricemia and gout. This study evaluated the high-fat, high-calorie food effect on the pharmacokinetics and pharmacodynamics of SHR4640 in healthy Chinese male volunteers. In this open-label, randomized, 2-period crossover phase 1 trial, 14 healthy male subjects were randomized to receive a single 10-mg dose of SHR4640 under both fasted and fed conditions. The washout period was 7 days. Blood samples were collected for pharmacokinetic and pharmacodynamic analysis. Pharmacokinetic parameters were analyzed by a noncompartmental method. The safety of the drug was also evaluated in the trial. A total of 14 healthy male volunteers were enrolled in the study, and finally 13 healthy volunteers completed the study. A single 10-mg dose of SHR4640 was safe and well tolerated in healthy Chinese male volunteers. After single-dose administration of SHR4640, the 90%CIs of the geometric mean ratios of the area under the plasma concentration-time curve from time 0 to the last quantifiable concentration and the area under the plasma concentration-time curve from time 0 to infinity were within the equivalence criteria of 0.80-1.25. The 90%CIs of maximum plasma concentration was slightly outside the lower limit of bioequivalent criteria, with about 13.40% decrease in the fed versus fasted condition. The time to maximum concentration was slightly delayed under the fasted condition. A single 10-mg dose of SHR4640 was safe and well tolerated in this trial. The main pharmacokinetic parameters and serum uric acid lowering of SHR4640 were not affected by food effect; thus, SHR4640 can be recommended to be administered with or without food.


Subject(s)
East Asian People , Uric Acid , Humans , Male , Healthy Volunteers , Area Under Curve , Uricosuric Agents/pharmacokinetics
8.
Eur J Med Chem ; 242: 114682, 2022 Nov 15.
Article in English | MEDLINE | ID: mdl-36001935

ABSTRACT

Benzbromarone (BM) is a potent URAT1 inhibitor approved for the treatment of gout. However, the low URAT1-selectivity and hepatotoxcity limit its clinical use. To solve these problems, we rationally designed and synthesized a series of BM derivatives by chemotype hybridization and bioisosteric replacement. Most compounds exhibited potent inhibitory activities against URAT1 with IC50 values ranging from 5.83 µM to 0.80 µM. Among them, JNS4 exhibited the highest URAT1 inhibitory activity with an IC50 of 0.80 µM, comparable to that of BM (IC50 = 0.53 µM). Molecular dynamic simulations showed that JNS4 formed π-cation interaction with R477, the same as BM. Different from BM, JNS4 bound to W357 and H245 via π-π interactions and formed a hydrogen bond with S35, which might contribute to the high URAT1 binding affinity of JNS4. JNS4 hardly inhibited GLUT9 (IC50 > 20 µM), another urate reabsorption transporter. In addition, JNS4 showed little inhibitory effects against OAT1 and ABCG2 with IC50 of 4.04 µM and 10.16 µM, respectively. Importantly, JNS4 displayed higher in vivo urate-lowering effects at doses of 1-4 mg/kg in a mouse model of hyperuricemia, as compared to BM and lesinurad. Furthermore, JNS4 possessed favorable pharmacokinetic properties with an oral bioavailability of 55.28%, significantly higher than that of BM (36.11%). Moreover, JNS4 demonstrated benign toxicity profiles (no cytotoxicities against HepG2 and HK2 cells; no hepatic and renal toxicities observed in vivo). Collectively, these results suggest that JNS4 represents a novel, safe and selective URAT1 inhibitor with excellent druggabilities and is worthy of further investigation as an anti-hyperuricemic agent.


Subject(s)
Hyperuricemia , Organic Anion Transporters , Animals , Benzbromarone/pharmacology , Benzbromarone/therapeutic use , Hyperuricemia/drug therapy , Mice , Organic Cation Transport Proteins/metabolism , Organic Cation Transport Proteins/therapeutic use , Uric Acid/metabolism , Uricosuric Agents/pharmacokinetics , Uricosuric Agents/therapeutic use
9.
Ther Umsch ; 68(1): 11-8, 2011 Jan.
Article in German | MEDLINE | ID: mdl-21184389

ABSTRACT

Nephrotoxicity is a common and often clinically relevant adverse drug reaction. Mechanisms include vascular, tubulo-toxic, tubulo-obstructive, and immunological effects. Drug-drug interactions may occur at a pharmacokinetic or pharmacodynamic level. Such interactions can both increase (cisplatin and aminoglycoside) but also protect from nephrotoxicity (cidofovir and probenecid).Important measures for preventing nephrotoxicity are (1) consideration of potential pharmacokinetic and pharmacodynamic interactions when prescribing a drug, (2) prescription of nephrotoxic drugs for the shortest possible period, (3) detection of high-risk patients, and (4) consideration of hydration and prophylactic comedication.


Subject(s)
Drug Interactions , Drug-Related Side Effects and Adverse Reactions , Kidney/drug effects , Aminoglycosides/pharmacokinetics , Aminoglycosides/toxicity , Analgesics/pharmacokinetics , Analgesics/toxicity , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/toxicity , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/toxicity , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Antirheumatic Agents/pharmacokinetics , Antirheumatic Agents/toxicity , Cidofovir , Cisplatin/pharmacokinetics , Cisplatin/toxicity , Contrast Media/pharmacokinetics , Contrast Media/toxicity , Creatinine/blood , Cytosine/analogs & derivatives , Cytosine/pharmacokinetics , Cytosine/toxicity , Humans , Organophosphonates/pharmacokinetics , Organophosphonates/toxicity , Probenecid/pharmacokinetics , Probenecid/toxicity , Risk Factors , Uricosuric Agents/pharmacokinetics , Uricosuric Agents/toxicity
10.
Article in English | MEDLINE | ID: mdl-32905987

ABSTRACT

Urate anion exchanger 1 (URAT1) expressed in the proximal renal tubules is responsible for about 90% of the reabsorption of uric acid. URAT1 is identified as an important target of uricosuric drugs. Here we present an LC-MS/MS-based approach, combined with URAT1-transgenic MDCK cells, for the assessment of uric acid. Cell lysis was executed with 50 mM NaOH to release uric acid. 1,3-15N2 uric acid was employed as the internal standard. The harvested uric acid, along with the stable isotope-labeled uric acid, was analyzed by LC-MS/MS in multiple reactions monitoring and negative modes. Validation, i.e. determination of selectivity, precision, accuracy, extraction recovery, and matrix effect, and feasibility was evaluated by use of the approach developed. The linearity was observed in the range of 1.0-250 µM (r = 0.9960) with limit of detection of 50 nM and limit of quantitation of 200 nM. The precision and accuracy were found to be RSD ≤ 20% and 80-120% of the nominal value, respectively. Uric acid uptake showed concentration and time dependency in URAT1-transgenic cells. The observed inhibitory effects of three URAT1-targeted uricosuric drugs were consistent with those reported in literature. The stable isotope dilution-based approach was proven to be selective, sensitive, and convenient, which is a good in vitro model for URAT1-targeted drug candidate screening.


Subject(s)
Chromatography, Liquid/methods , Drug Evaluation, Preclinical/methods , Organic Anion Transporters , Organic Cation Transport Proteins , Tandem Mass Spectrometry/methods , Uricosuric Agents , Animals , Dogs , Humans , Limit of Detection , Linear Models , Madin Darby Canine Kidney Cells , Organic Anion Transporters/antagonists & inhibitors , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Organic Cation Transport Proteins/antagonists & inhibitors , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Reproducibility of Results , Uric Acid/analysis , Uric Acid/metabolism , Uricosuric Agents/analysis , Uricosuric Agents/pharmacokinetics
11.
Drug Metab Lett ; 13(2): 111-122, 2019.
Article in English | MEDLINE | ID: mdl-31613735

ABSTRACT

BACKGROUND: Clinical development of lesinurad, a selective uric acid reabsorption inhibitor, required analysis of lesinurad in plasma from special patient populations. METHODS: EMA and FDA bioanalytical method validation guidance have recommended studying matrix effects on quantitation if samples from special patient populations are to be analyzed. In addition to lesinurad (plasma protein binding 98.2%), the matrix effects from special population plasma on the quantitation of verapamil (PPB 89.6%), allopurinol and oxypurinol (PPB negligible) were also investigated. RESULTS: The plasma from special population patients had no matrix effects on the three quantification methods with stable isotope labeled internal standard, protein precipitation extraction, and LC-MS/MS detection. The validated lesinurad plasma quantification method was successfully applied for the pharmacokinetic evaluations to support the clinical studies in renal impaired patients. CONCLUSION: Special population plasma did not affect quantitation of drugs with a wide range of plasma protein binding levels in human plasma. With the confirmation that there is no impact on quantification from the matrix, the bioanalytical method can be used to support the pharmacokinetic evaluations for clinical studies in special populations.


Subject(s)
Hepatic Insufficiency/metabolism , Renal Insufficiency/metabolism , Thioglycolates/blood , Triazoles/blood , Uricosuric Agents/blood , Allopurinol/pharmacokinetics , Calibration , Chromatography, High Pressure Liquid/methods , Chromatography, High Pressure Liquid/standards , Clinical Trials as Topic , Hepatic Insufficiency/blood , Hepatic Insufficiency/physiopathology , Humans , Kidney/metabolism , Kidney/physiopathology , Liver/metabolism , Liver/physiopathology , Oxypurinol/blood , Oxypurinol/pharmacokinetics , Reference Standards , Renal Insufficiency/blood , Renal Insufficiency/physiopathology , Renal Reabsorption , Reproducibility of Results , Tandem Mass Spectrometry/methods , Tandem Mass Spectrometry/standards , Thioglycolates/pharmacokinetics , Triazoles/pharmacokinetics , Uricosuric Agents/pharmacokinetics , Verapamil/blood , Verapamil/pharmacokinetics
12.
Drug Saf ; 31(8): 643-65, 2008.
Article in English | MEDLINE | ID: mdl-18636784

ABSTRACT

Benzbromarone, a potent uricosuric drug, was introduced in the 1970s and was viewed as having few associated serious adverse reactions. It was registered in about 20 countries throughout Asia, South America and Europe. In 2003, the drug was withdrawn by Sanofi-Synthélabo, after reports of serious hepatotoxicity, although it is still marketed in several countries by other drug companies. The withdrawal has greatly limited its availability around the world, and increased difficulty in accessing it in other countries where it has never been available.The overall aim of this paper is to determine if the withdrawal of benzbromarone was in the best interests of gouty patients and to present a benefit-risk assessment of benzbromarone. To determine this, we examined (i) the clinical benefits associated with benzbromarone treatment and compared them with the success of alternative therapies such as allopurinol and probenecid, particularly in patients with renal impairment; (ii) the attribution of the reported cases of hepatotoxicity to treatment with benzbromarone; (iii) the incidence of hepatotoxicity possibly due to benzbromarone; (iv) adverse reactions to allopurinol and probenecid. From these analyses, we present recommendations on the use of benzbromarone.Large reductions in plasma urate concentrations in patients with hyperuricaemia are achieved with benzbromarone and most patients normalize their plasma urate. The half-life of benzbromarone is generally short (about 3 hours); however, a uricosuric metabolite, 6-hydroxybenzbromarone, has a much longer half-life (up to 30 hours) and is the major species responsible for the uricosuric activity of benzbromarone, although its metabolism by cytochrome P450 (CYP) 2C9 in the liver may vary between patients as a result of polymorphisms in this enzyme. It is effective in patients with moderate renal impairment. Standard dosages of benzbromarone (100 mg/day) tend to produce greater hypouricaemic effects than standard doses of allopourinol (300 mg/day) or probenecid (1000 mg/day).Adverse effects associated with benzbromarone are relatively infrequent, but potentially severe. Four cases of benzbromarone-induced hepatotoxicity were identified from the literature. Eleven cases have been reported by Sanofi-Synthélabo, but details are not available in the public domain. Only one of the four published cases demonstrated a clear relationship between the drug and liver injury as demonstrated by rechallenge. The other three cases lacked incontrovertible evidence to support a diagnosis of benzbromarone-induced hepatotoxicity. If all the reported cases are assumed to be due to benzbromarone, the estimated risk of hepatotoxicity in Europe was approximately 1 in 17 000 patients but may be higher in Japan.Benzbromarone is also an inhibitor of CYP2C9 and so may be involved in drug interactions with drugs dependent on this enzyme for clearance, such as warfarin. Alternative drugs to benzbromarone have significant adverse reactions. Allopurinol is associated with rare life-threatening hypersensitivity syndromes; the risk of these reactions is approximately 1 in 56 000. Rash occurs in approximately 2% of patients taking allopurinol and usually leads to cessation of prescription of the drug. Probenecid has also been associated with life-threatening reactions in a very small number of case reports, but it frequently interacts with many renally excreted drugs. Febuxostat is a new xanthine oxidoreductase inhibitor, which is still in clinical trials, but abnormal liver function is the most commonly reported adverse reaction.Even assuming a causal relationship between benzbromarone and hepatotoxicity in the identified cases, benefit-risk assessment based on total exposure to the drug does not support the decision by the drug company to withdraw benzbromarone from the market given the paucity of alternative options. It is likely that the risks of hepatotoxicity could be ameliorated by employing a graded dosage increase, together with regular monitoring of liver function. Determination of CYP2C9 status and consideration of potential interactions through inhibition of this enzyme should be considered. The case for wider and easier availability of benzbromarone for treating selected cases of gout is compelling, particularly for patients in whom allopurinol produces insufficient response or toxicity.We conclude that the withdrawal of benzbromarone was not in the best interest of patients with gout.


Subject(s)
Benzbromarone/therapeutic use , Gout/drug therapy , Uricosuric Agents/therapeutic use , Animals , Benzbromarone/adverse effects , Benzbromarone/pharmacokinetics , Chemical and Drug Induced Liver Injury , Clinical Trials as Topic , Drug Approval , Gout/epidemiology , Humans , Risk Factors , Uricosuric Agents/adverse effects , Uricosuric Agents/pharmacokinetics
13.
Drug Des Devel Ther ; 12: 1799-1807, 2018.
Article in English | MEDLINE | ID: mdl-29950814

ABSTRACT

PURPOSE: Verinurad (RDEA3170) is a selective uric acid reabsorption inhibitor in clinical development for treatment of gout and asymptomatic hyperuricemia. This study evaluated verinurad pharmacokinetics, pharmacodynamics, and tolerability in healthy Japanese and non-Asian adult male subjects. METHODS: This was a Phase I, randomized, single-blind, placebo-controlled study. Panels of 8 Japanese subjects were randomized to receive oral verinurad (2.5-15 mg) or placebo administered as a single dose in a fasted and fed state and as once-daily doses for 7 days in a fed state. Eight non-Asian subjects received verinurad 10 mg as a single dose (fasted and fed) and multiple doses in the fed state. Serial plasma/serum and urine samples were assayed for verinurad and uric acid. Safety was assessed by adverse events and laboratory data. RESULTS: Of 48 randomized subjects, 46 (Japanese, 39; non-Asian, 7) completed the study. Following single or multiple doses in Japanese subjects, maximum plasma concentration (Cmax) and area under the plasma concentration-time curve (AUC) increased in a near dose-proportional manner. Time to Cmax (Tmax) was ~1.25-2.0 hours with fasting. A moderate-fat meal delayed Tmax (range 3.0-5.0 hours) and had a variable effect on AUC (0%-97% increase) and Cmax (0%-26% increase) across the dose groups. Following multiple verinurad 10 mg doses, Cmax and AUC were 38% and 23% higher, respectively, in Japanese vs non-Asian subjects, largely due to body weight differences. Mean reduction of serum urate following multiple verinurad 10 mg doses was 46% and 44% after 24 hours in Japanese and non-Asian subjects, respectively. Verinurad was well tolerated at all doses. CONCLUSION: Verinurad monotherapy lowered serum urate and was well tolerated in both healthy Japanese and non-Asian males, while small differences in plasma pharmacokinetics were observed. These data support further evaluation of once-daily verinurad as a treatment for gout and asymptomatic hyperuricemia.


Subject(s)
Asian People , Kidney Tubules/drug effects , Naphthalenes , Propionates , Pyridines , Renal Elimination/drug effects , Renal Reabsorption/drug effects , Uric Acid/metabolism , Uricosuric Agents/pharmacokinetics , Administration, Oral , Adult , Area Under Curve , Drug Administration Schedule , Half-Life , Healthy Volunteers , Humans , Kidney Tubules/metabolism , Male , Metabolic Clearance Rate , Middle Aged , Models, Biological , Naphthalenes/administration & dosage , Naphthalenes/adverse effects , Naphthalenes/pharmacokinetics , Naphthalenes/pharmacology , Propionates/administration & dosage , Propionates/adverse effects , Propionates/pharmacokinetics , Propionates/pharmacology , Pyridines/administration & dosage , Pyridines/adverse effects , Pyridines/pharmacokinetics , Pyridines/pharmacology , Single-Blind Method , Uric Acid/blood , Uric Acid/urine , Uricosuric Agents/administration & dosage , Uricosuric Agents/blood , Young Adult
14.
Drug Des Devel Ther ; 11: 2077-2086, 2017.
Article in English | MEDLINE | ID: mdl-28744099

ABSTRACT

PURPOSE: Verinurad (RDEA3170) is a selective uric acid reabsorption inhibitor in clinical development for the treatment of gout and asymptomatic hyperuricemia. The aim of this study was to evaluate the pharmacokinetics, pharmacodynamics, and tolerability of verinurad in healthy adult males. SUBJECTS AND METHODS: This was a Phase I, randomized, double-blind, placebo-controlled, single and multiple ascending dose study. Panels of eight male subjects received a single oral dose of verinurad or placebo in either a fasted or fed state; panels of 10-12 male subjects received ascending doses of once-daily verinurad or placebo in a fasted state for 10 days. Serial blood and urine samples were assayed for verinurad and uric acid. Safety was assessed by adverse event (AE) reports, laboratory tests, vital signs, and electrocardiograms (ECGs). RESULTS: A total of 81 adult males completed the study. Following single doses of verinurad, maximum observed plasma concentration (Cmax) and area under the plasma concentration-time curve (AUC) increased in a dose-proportional manner; Cmax occurred at 0.5-0.75 hours and 1.25 hours in the fasted and fed states, respectively. Food decreased AUC by 23% and Cmax by 37%-53%. There was a modest accumulation of verinurad following multiple daily doses. Verinurad reduced serum urate levels by up to 62% (40 mg, single dose) and 61% (10 mg, multiple dose). The increase in urinary excretion of uric acid was greatest in the first 6 hours after dosing and was still evident ≥24 hours for verinurad doses ≥2 mg. Verinurad was well tolerated at all doses. No serious AEs, severe AEs, discontinuations due to AEs, or clinically significant laboratory or ECG abnormalities were reported. CONCLUSION: Single and multiple doses of verinurad were well tolerated, absorption was rapid, and exposure was dose proportional. Verinurad increased urinary uric acid elimination and resulted in sustained reductions in serum urate. These data support further clinical evaluation of once-daily verinurad as a treatment for gout.


Subject(s)
Uric Acid/metabolism , Uricosuric Agents/pharmacology , Adolescent , Adult , Dose-Response Relationship, Drug , Double-Blind Method , Female , Healthy Volunteers , Humans , Intestinal Absorption , Male , Metabolic Clearance Rate , Middle Aged , Uric Acid/urine , Uricosuric Agents/adverse effects , Uricosuric Agents/pharmacokinetics , Young Adult
15.
Clin Pharmacol Drug Dev ; 6(4): 377-387, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28074640

ABSTRACT

Lesinurad is a novel selective uric acid reabsorption inhibitor approved for treatment of hyperuricemia associated with gout in combination with xanthine oxidase inhibitors (XOIs). Open-label pharmacokinetic studies were performed in volunteers or subjects with hyperuricemia (serum uric acid ≥ 8 mg/dL) to investigate interactions of lesinurad (with and without concurrent XOIs) with colchicine and 2 nonsteroidal anti-inflammatory drugs: naproxen and indomethacin. Colchicine studies included consecutive 7-day treatment periods of (1) allopurinol 300 mg, allopurinol 300 mg plus lesinurad 400 or 600 mg, and continued lesinurad 400 or 600 mg; or (2) febuxostat 40 or 80 mg, febuxostat 40 or 80 mg plus lesinurad 400 mg, and continued febuxostat 40 or 80 mg plus lesinurad 600 mg. Naproxen and indomethacin studies included lesinurad 400 mg on day 1, naproxen 250 mg twice daily or indomethacin 25 mg twice daily on days 2-6, and lesinurad 400 mg plus continued naproxen or indomethacin on days 7-13 and the morning of day 14. Lesinurad did not alter the pharmacokinetics of naproxen and modestly altered exposure to colchicine (AUC decrease of ≤ 25%) and indomethacin (AUC increase of ∼35%). Indomethacin did not alter the pharmacokinetics of lesinurad, whereas naproxen modestly decreased the Cmax of lesinurad by ∼27%.


Subject(s)
Gout Suppressants/pharmacokinetics , Hyperuricemia/drug therapy , Thioglycolates/pharmacokinetics , Triazoles/pharmacokinetics , Uricosuric Agents/pharmacokinetics , Adult , Aged , Aged, 80 and over , Colchicine/administration & dosage , Colchicine/pharmacokinetics , Drug Administration Schedule , Drug Interactions , Female , Gout Suppressants/administration & dosage , Humans , Indomethacin/administration & dosage , Indomethacin/pharmacokinetics , Male , Middle Aged , Naproxen/administration & dosage , Naproxen/pharmacokinetics , Thioglycolates/administration & dosage , Treatment Outcome , Triazoles/administration & dosage , Uricosuric Agents/administration & dosage , Young Adult
16.
J Vet Med Sci ; 68(4): 361-5, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16679727

ABSTRACT

Pharmacokinetics (PK) of probenecid including plasma probenecid concentrations, in vitro plasma protein binding properties, and in vivo PK parameters were determined in dogs. Probenecid concentrations were best determined by HPLC, which showed good linearity and good recovery with simple plasma preparation. The quantification limit of probenecid was approximately 50 ng/ml at S/N ratio = 3, by simple procedure with HCl and methanol treatment. Probenecid showed two types of binding characteristics, i.e., high-affinity with low-capacity and low-affinity with high-capacity binding. This result indicated 80-88% of probenecid was bound to plasma protein(s) at observed concentrations (< 80 microg/ml) in vivo at an intravenous dose of 20 mg/kg. Plasma probenecid concentration-time profile following i.v. administration in dogs showed biphasic decline and well fitted a two-compartment open model. The total body clearance was 0.34 +/- 0.04 ml/min/kg, volume of distribution at steady-state was 0.46 +/- 0.07 l/kg, elimination half-life was 18 +/- 6 hr, and mean residence time (MRT) was 23 +/- 6 hr. Since probenecid has been known as a potent inhibitor of renal tubular excretion of acidic drugs and highly binds to plasma proteins, our observation in relation to plasma protein binding and PK parameters will serve as the basic information concerning drug-drug interactions in dogs and in other mammalian species.


Subject(s)
Blood Proteins/metabolism , Dogs/metabolism , Probenecid/metabolism , Probenecid/pharmacokinetics , Uricosuric Agents/metabolism , Uricosuric Agents/pharmacokinetics , Animals , Dogs/blood , Female , Half-Life , Protein Binding , Tissue Distribution
17.
Arthritis Res Ther ; 18(1): 214, 2016 10 03.
Article in English | MEDLINE | ID: mdl-27716403

ABSTRACT

BACKGROUND: Excess body burden of uric acid promotes gout. Diminished renal clearance of uric acid causes hyperuricemia in most patients with gout, and the renal urate transporter (URAT)1 is important for regulation of serum uric acid (sUA) levels. The URAT1 inhibitors probenecid and benzbromarone are used as gout therapies; however, their use is limited by drug-drug interactions and off-target toxicity, respectively. Here, we define the mechanism of action of lesinurad (Zurampic®; RDEA594), a novel URAT1 inhibitor, recently approved in the USA and Europe for treatment of chronic gout. METHODS: sUA levels, fractional excretion of uric acid (FEUA), lesinurad plasma levels, and urinary excretion of lesinurad were measured in healthy volunteers treated with lesinurad. In addition, lesinurad, probenecid, and benzbromarone were compared in vitro for effects on urate transporters and the organic anion transporters (OAT)1 and OAT3, changes in mitochondrial membrane potential, and human peroxisome proliferator-activated receptor gamma (PPARγ) activity. RESULTS: After 6 hours, a single 200-mg dose of lesinurad elevated FEUA 3.6-fold (p < 0.001) and reduced sUA levels by 33 % (p < 0.001). At concentrations achieved in the clinic, lesinurad inhibited activity of URAT1 and OAT4 in vitro, did not inhibit GLUT9, and had no effect on ABCG2. Lesinurad also showed a low risk for mitochondrial toxicity and PPARγ induction compared to benzbromarone. Unlike probenecid, lesinurad did not inhibit OAT1 or OAT3 in the clinical setting. CONCLUSION: The pharmacodynamic effects and in vitro activity of lesinurad are consistent with inhibition of URAT1 and OAT4, major apical transporters for uric acid. Lesinurad also has a favorable selectivity and safety profile, consistent with an important role in sUA-lowering therapy for patients with gout.


Subject(s)
Gout , Organic Anion Transporters/drug effects , Organic Cation Transport Proteins/drug effects , Thioglycolates/pharmacokinetics , Triazoles/pharmacokinetics , Uric Acid/blood , Uricosuric Agents/pharmacokinetics , Cell Line , Humans , Kidney/drug effects , Male
18.
Drug Des Devel Ther ; 9: 3423-34, 2015.
Article in English | MEDLINE | ID: mdl-26170627

ABSTRACT

Lesinurad is a selective uric acid reabsorption inhibitor under investigation for the treatment of gout. Single and multiple ascending dose studies were conducted to evaluate pharmacokinetics, pharmacodynamics, and safety of lesinurad in healthy males. Lesinurad was administered as an oral solution between 5 mg and 600 mg (single ascending dose; N=34) and as an oral solution or immediate-release capsules once daily (qday) between 100 mg and 400 mg for 10 days under fasted or fed condition (multiple ascending dose; N=32). Following single doses of lesinurad solution, absorption was rapid and exposure (maximum observed plasma concentration and area under the plasma concentration-time curve) increased in a dose-proportional manner. Following multiple qday doses, there was no apparent accumulation of lesinurad. Urinary excretion of unchanged lesinurad was generally between 30% and 40% of dose. Increases in urinary excretion of uric acid and reductions in serum uric acid correlated with dose. Following 400 mg qday dosing, serum uric acid reduction was 35% at 24 hours post-dose, supporting qday dosing. A relative bioavailability study in healthy males (N=8) indicated a nearly identical pharmacokinetic profile following dosing of tablets or capsules. Lesinurad was generally safe and well tolerated.


Subject(s)
Organic Anion Transporters/antagonists & inhibitors , Organic Cation Transport Proteins/antagonists & inhibitors , Renal Elimination/drug effects , Thioglycolates/pharmacokinetics , Triazoles/pharmacokinetics , Uric Acid/urine , Uricosuric Agents/pharmacokinetics , Administration, Oral , Adolescent , Adult , Biological Availability , Capsules , Cross-Over Studies , Double-Blind Method , Gastrointestinal Absorption , Healthy Volunteers , Humans , Male , Organic Anion Transporters/metabolism , Organic Cation Transport Proteins/metabolism , Pharmaceutical Solutions , Thioglycolates/administration & dosage , Thioglycolates/adverse effects , Thioglycolates/blood , Triazoles/administration & dosage , Triazoles/adverse effects , Triazoles/blood , Uric Acid/blood , Uricosuric Agents/administration & dosage , Uricosuric Agents/adverse effects , Uricosuric Agents/blood , Young Adult
19.
Clin Pharmacol Ther ; 66(6): 569-81, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10613612

ABSTRACT

OBJECTIVE: To clarify the mechanism(s) for the interaction between warfarin and benzbromarone, a uricosuric agent, and to predict changes in the in vivo pharmacokinetics of (S)-warfarin from in vitro data. METHODS: Warfarin enantiomers and benzbromarone in serum, 7-hydroxywarfarin in urine, and serum unbound fractions of warfarin enantiomers were measured in patients with heart disease given warfarin with (n = 13) or without (n = 18) oral benzbromarone (50 mg/d). In vitro inhibition constants (K(i)) of benzbromarone for (S)-warfarin 7-hydroxylation were determined with use of human CYP2C9 and liver microsomes. The magnitude of changes in the formation clearance for 7-hydroxylation (CLf), the unbound oral clearance (CL(oral,u)), and the oral clearance (CL(oral)) for (S)-warfarin were predicted by equations incorporating the in vitro Ki, the theoretical maximum unbound hepatic benzbromarone concentration, and the fractions of warfarin eliminated through metabolism and of CYP2C9-mediated metabolic reaction susceptible to inhibition by benzbromarone. RESULTS: The patients given warfarin with benzbromarone required a 36% less (P < .01) warfarin dose than those given warfarin alone (2.5 versus 3.9 mg/d) to attain similar international normalized ratios (2.1 and 2.2, respectively), and the former had 65%, 53%, and 54% lower (P < .05 or P < .01) CLf, CL(oral),u, and CL(oral) for (S)-warfarin than the latter, respectively. In contrast, no significant differences were observed for (R)-warfarin kinetics between the groups. Benzbromarone was found to be a potent competitive inhibitor (Ki < 0.01 micromol/L) for (S)-warfarin 7-hydroxylation mediated by CYP2C9. The average changes in the in vivo CLf, CL(oral),u, and CL(oral)values for (S)-warfarin induced by benzbromarone were largely predictable by the proposed equations. CONCLUSION: Benzbromarone would intensify anticoagulant response of warfarin through an enantioselective inhibition of CYP2C9-mediated metabolism of pharmacologically more potent (S)-warfarin. The magnitude of changes in the in vivo warfarin kinetics may be predicted by in vitro data.


Subject(s)
Anticoagulants/pharmacokinetics , Aryl Hydrocarbon Hydroxylases , Benzbromarone/pharmacokinetics , Steroid 16-alpha-Hydroxylase , Uricosuric Agents/pharmacokinetics , Warfarin/pharmacokinetics , Aged , Anticoagulants/blood , Anticoagulants/urine , Benzbromarone/blood , Benzbromarone/urine , Cytochrome P-450 CYP2C9 , Cytochrome P-450 Enzyme System/metabolism , Drug Synergism , Female , Humans , Male , Microsomes, Liver/enzymology , Middle Aged , Stereoisomerism , Steroid Hydroxylases/metabolism , Uricosuric Agents/blood , Uricosuric Agents/urine , Warfarin/blood , Warfarin/urine
20.
J Pharm Sci ; 81(9): 935-9, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1432643

ABSTRACT

5-Dimethylsulfamoyl-6,7-dichloro-2,3-dihydrobenzofuran-2-carboxyli c acid (DBCA), a promising uricosuric, diuretic, and antihypertensive agent, was administered intravenously to rats. The levels of DBCA in plasma and the areas under the curve of concentration versus time (AUC values) of the S(-)-enantiomer were higher than those of the R(+)-enantiomer. Total body clearance was significantly greater for the R(+)-enantiomer. This stereoselective elimination was due to a difference in the nonrenal clearance, which seemed to reflect hepatic metabolism or biliary excretion. Hepatic metabolism seemed more likely because AUC and the amount of urinary excretion of the N-monodemethylated metabolite of DBCA were greater for the R(+)-enantiomer. The plasma had higher free fractions of the S(-)-enantiomer, a result suggesting that this enantiomer is distributed more readily to the tissues, including the liver. This result indicates that protein binding was not responsible for the stereoselective metabolism of (R)-(+)-DBCA. Although there was no difference in the renal clearances of the enantiomers, the renal clearance of free (R)-(+)-DBCA exceeded that of the S(-)-enantiomer, a result indicating the preferential excretion of the R(+)-enantiomer into the urine. Comparison of the pharmacokinetics of individual enantiomers after intravenous administration of each enantiomer or its racemate showed that the enantiomers interact with one another; dosing with racemate delayed the elimination of each enantiomer because of mutual inhibition of hepatic metabolism and renal excretion for (R)-(+)-DBCA and of renal excretion for (S)-(-)-DBCA.


Subject(s)
Antihypertensive Agents/pharmacokinetics , Diuretics/pharmacokinetics , Sulfonamides/pharmacokinetics , Uricosuric Agents/pharmacokinetics , Animals , Drug Interactions , Male , Rats , Rats, Inbred Strains , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL