Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Mol Nutr Food Res ; 67(1): e2200162, 2023 01.
Article in English | MEDLINE | ID: mdl-36308034

ABSTRACT

SCOPE: This study aims to investigate the anti-hyperuricemic and nephroprotective effects and the potential mechanisms of the separated gastrointestinal hydrolysates of α-lactalbumin on hyperuricemic mice. METHODS AND RESULTS: The gastrointestinal hydrolysate of α-lactalbumin, the hydrolysate fraction with molecular weight (MW) < 3 kDa (LH-3k), and the fragments with smallest MW among LH-3K harvested through dextran gel chromatography (F5) are used. Hyperuricemia mice are induced via daily oral gavage of potassium oxonate and hypoxanthine. F5 displays the highest in vitro xanthine oxidase (XO) inhibition among all the fractions separated from LH-3k. Oral administration of F5 significantly reduces the levels of serum uric acid (UA), creatinine, and urea nitrogen. F5 treatment could ameliorate kidney injury through alleviating oxidative stress and inflammation. F5 alleviates hyperuricemia in mice by inhibiting hepatic XO activity and regulating the expression of renal urate transporters. Gut microbiota analysis illustrates that F5 administration increases the abundance of some SCFAs producers, and inhibits the growth of hyperuricemia and inflammation associated genera. LH-3k exhibits similar effects but does not show significance as those of the F5 fraction. CONCLUSION: The anti-hyperuricemia and nephroprotective functions of F5 are mediated by inhibiting hepatic XO activity, ameliorating oxidative stress and inflammation, regulating renal urate transporters, and modulating the gut microbiota in hyperuricemic mice.


Subject(s)
Gastrointestinal Microbiome , Hyperuricemia , Mice , Animals , Uric Acid , Lactalbumin/metabolism , Hyperuricemia/drug therapy , Kidney/metabolism , Oxonic Acid/metabolism , Oxonic Acid/pharmacology , Transcription Factors/metabolism , Inflammation/metabolism , Hypoxanthines/metabolism , Hypoxanthines/pharmacology
2.
Oxid Med Cell Longev ; 2020: 6943860, 2020.
Article in English | MEDLINE | ID: mdl-32695259

ABSTRACT

Previous studies have disclosed the antihyperuricemic effect of polydatin, a natural precursor of resveratrol; however, the mechanisms of action still remain elusive. The present study was undertaken to evaluate the therapeutic effects and the underlying mechanisms of polydatin on potassium oxonate-induced hyperuricemia in rats through metabonomic technology from a holistic view. Nuclear magnetic resonance (NMR) spectroscopy was applied to capture the metabolic changes in sera and urine collected from rats induced by hyperuricemia and polydatin treatment. With multivariate data analysis, significant metabolic perturbations were observed in hyperuricemic rats compared with the healthy controls. A total of eleven and six metabolites were identified as differential metabolites related to hyperuricemia in serum and urine of rats, respectively. The proposed pathways primarily included branched-chain amino acid (BCAA) metabolism, glycolysis, the tricarboxylic acid cycle, synthesis and degradation of ketone bodies, purine metabolism, and intestinal microflora metabolism. Additionally, some metabolites indicated the risk of renal injury induced by hyperuricemia. Polydatin significantly lowered the levels of serum uric acid, creatinine, and blood urea nitrogen and alleviated the abnormal metabolic status in hyperuricemic rats by partially restoring the balance of the perturbed metabolic pathways. Our findings shed light on the understanding of the pathophysiological process of hyperuricemia and provided a reference for revealing the metabolic mechanism produced by polydatin in the treatment of hyperuricemia.


Subject(s)
Drugs, Chinese Herbal/therapeutic use , Glucosides/therapeutic use , Hyperuricemia/drug therapy , Stilbenes/therapeutic use , Animals , Blood Urea Nitrogen , Creatinine/blood , Disease Models, Animal , Humans , Kidney/pathology , Male , Metabolomics , Oxonic Acid/metabolism , Rats , Rats, Sprague-Dawley , Uric Acid/blood
3.
Cancer Lett ; 400: 223-231, 2017 08 01.
Article in English | MEDLINE | ID: mdl-27838412

ABSTRACT

Metronomic chemotherapy is currently considered an emerging therapeutic option in clinical oncology. S-1, an oral formulation of Tegafur (TF), a prodrug of 5-fluorouracil (5-FU), is designed to improve the antitumor activity of 5-FU in tandem with reducing its toxicity. Clinically, metronomic S-1 dosing has been approved for the standard first- and second-line treatment of metastatic or advanced stage of colorectal (CRC). However, expression of intratumor thymidylate synthase (TS), a significant gene in cellular proliferation, is associated with poor outcome to 5-FU-based chemotherapeutic regimens. In this study, therefore, we examined the effect of a combination of TS silencing by an RNA interfering molecule, chemically synthesized short hairpin RNA against TS (shTS), and 5-FU on the growth of human colorectal cancer cell (DLD-1) both in vitro and in vivo. The combined treatment of both shTS with 5-FU substantially inhibited cell proliferation in vitro. For in vivo treatments, the combined treatment of metronomic S-1 dosing with intravenously injected polyethylene glycol (PEG)-coated shTS-lipoplex significantly suppressed tumor growth, compared to a single treatment of either S-1 or PEG-coated shTS-lipoplex. In addition, the combined treatment increased the proportion of apoptotic cells in the DLD-1 tumor tissue. Our results suggest that metronomic S-1 dosing combined with TS silencing might represent an emerging therapeutic strategy for the treatment of patients with advanced CRC.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Colorectal Neoplasms/therapy , Oxonic Acid/administration & dosage , RNA Interference , RNAi Therapeutics/methods , Tegafur/administration & dosage , Thymidylate Synthase/genetics , Administration, Metronomic , Animals , Antimetabolites, Antineoplastic/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Drug Combinations , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Oxonic Acid/metabolism , Tegafur/metabolism , Thymidylate Synthase/metabolism , Time Factors , Transfection , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
4.
Cancer Chemother Pharmacol ; 79(1): 69-79, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27913881

ABSTRACT

BACKGROUND: The first one-arm phase II trial aimed to evaluate and predict efficacy and safety of S-1 plus oral leucovorin (S-1/LV) as first-line chemotherapy for patients with advanced gastric cancer (AGC), using S-1 pharmacogenetic pathway approach. PATIENTS AND METHODS: A total of 39 patients orally took S-1 at conventional dose and LV simultaneously at a dose of 25 mg twice daily for a week, within a 2-week cycle. The primary endpoint was overall response rate (ORR), while the secondary endpoints were progression-free survival (PFS), time to failure (TTF), overall survival (OS), disease control rate (DCR), and adverse events (AEs). Peripheral blood was sampled prospectively for baseline expression of dihydropyrimidine dehydrogenase (DPD), orotate phosphoribosyltransferase (OPRT), thymidine phosphorylase (TP), and thymidylate synthase (TS), CYP2A6 gene polymorphisms, and 5-FU pharmacokinetics. RESULTS: The ORR and DCR were 41.0 and 76.9%. The median PFS, TTF, and OS were 4.13, 3.70, and 11.40 months. Grade 3-4 AEs occurred in only 13 patients, and grade 4 AEs occurred in only 1 of them. High OPRT/TS and peritoneal metastasis (vs. liver metastasis) independently predicted responding. High OPRT/DPD independently predicted grade 3-4 AEs. High AUC0-24h of 5-FU and metastatic/recurrent sites ≤2 (vs. >3) independently predicted prolonged PFS. Low baseline plasmic DPD independently predicted prolonged OS. CONCLUSIONS: Two-week, oral S-1/LV regimen demonstrated promising efficacy and safety as first-line chemotherapy for AGC. CLINICALTRIALS. GOV IDENTIFIER: NCT02090153.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Oxonic Acid/metabolism , Stomach Neoplasms/drug therapy , Tegafur/metabolism , Adult , Aged , Aged, 80 and over , Cytochrome P-450 CYP2A6/genetics , Disease-Free Survival , Drug Combinations , Female , Fluorouracil/pharmacokinetics , Humans , Leucovorin/administration & dosage , Leucovorin/adverse effects , Male , Middle Aged , Orotate Phosphoribosyltransferase/blood , Oxonic Acid/administration & dosage , Oxonic Acid/adverse effects , Pharmacogenomic Testing , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Tegafur/administration & dosage , Tegafur/adverse effects
5.
BMC Nephrol ; 16: 35, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25886588

ABSTRACT

BACKGROUND: Recent studies suggest a causal role for increased plasma uric acid in the progression of chronic renal insufficiency (CRI). However, uric acid also functions as an antioxidant with possible beneficial effects. METHODS: We investigated the influence of hyperuricemia on mesenteric arterial tone (main and second order branch) and morphology in experimental CRI. Forty-four Sprague-Dawley rats were 5/6 nephrectomized (NX) or Sham-operated and fed 2.0% oxonic acid or control diet for 9 weeks. RESULTS: Oxonic acid feeding elevated plasma uric acid levels 2.4 and 3.6-fold in the NX and Sham groups, respectively. Plasma creatinine and urea were elevated 2-fold and blood pressure increased by 10 mmHg in NX rats, while hyperuricemia did not significantly influence these variables. Right and left ventricular weight, and atrial and B-type natriuretic peptide mRNA content were increased in NX rats, but were not affected by hyperuricemia. In the mesenteric artery, hyperuricemia did not influence vasoconstrictor responses in vitro to norepinephrine or potassium chloride. The small arteries of NX rats featured hypertrophic remodeling independent of uric acid levels: wall to lumen ratio, wall thickness and cross-sectional area were increased without changes in lumen diameter. In the main branch, vasorelaxations to acetylcholine were impaired in NX rats, but were not affected by hyperuricemia. In contrast, relaxations to the large-conductance Ca(2+)-activated K(+)-channel (BKCa) opener NS-1619 were reduced by oxonic acid feeding, whereas responses to nitroprusside were not affected. CONCLUSIONS: Experimental hyperuricemia did not influence cardiac load or vascular remodeling, but impaired BKCa -mediated vasorelaxation in experimental CRI.


Subject(s)
Cardiac Output/physiology , Hyperuricemia/chemically induced , Mesenteric Arteries/drug effects , Oxonic Acid/pharmacology , Uric Acid/blood , Analysis of Variance , Animals , Cardiac Output/drug effects , Disease Models, Animal , Hyperuricemia/physiopathology , Male , Mesenteric Arteries/physiology , Nephrectomy , Oxonic Acid/metabolism , Random Allocation , Rats , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/physiopathology , Vasoconstriction/drug effects , Vasodilation/drug effects
6.
Curr Microbiol ; 69(6): 852-7, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25086583

ABSTRACT

The hypouricemic actions of exopolysaccharide produced by Cordyceps militaris (EPCM) in potassium oxonate-induced hyperuricemia in mice were examined. Hyperuricemic mice were administered intragastrically with EPCM (200, 400 and 800 mg/kg body weight) or allopurinol (5 mg/kg body weight) once daily. Serum uric acid, blood urea nitrogen and liver xanthine oxidase (XOD) activities of each treatment were measured after administration for 7 days. EPCM showed dose-dependent uric acid-lowering actions. EPCM at a dose of 400 mg/kg body weight and allopurinol showed the same effect in serum uric acid, blood urea nitrogen and liver XOD activities in hyperuricemic mice. An increase in liver XOD activities was observed in hyperuricemic mice due to administration of EPCM at a dose of 200 mg/kg body weight. EPCM at a dose of 800 mg/kg body weight did not show significant effects on serum uric acid and XOD activities. We conclude that EPCM has a hypouricemic effect caused by decreases in urate production and the inhibition of XOD activities in hyperuricemic mice, and this natural product exhibited more potential efficacy than allopurinol in renal protection.


Subject(s)
Antimetabolites/therapeutic use , Cordyceps/chemistry , Hyperuricemia/drug therapy , Oxonic Acid/metabolism , Polysaccharides/therapeutic use , Animals , Antimetabolites/isolation & purification , Cordyceps/drug effects , Disease Models, Animal , Hyperuricemia/chemically induced , Liver/enzymology , Mice , Polysaccharides/isolation & purification , Serum/chemistry , Treatment Outcome , Urine/chemistry , Xanthine Oxidase/analysis
7.
Gan To Kagaku Ryoho ; 39(11): 1603-7, 2012 Nov.
Article in Japanese | MEDLINE | ID: mdl-23152010

ABSTRACT

The elucidation in recent years of intracellular signaling mechanisms related to cancer cell growth has been accompanied by increases in both drug development and biomarker research. While treatment strategies using biomarkers have been established and put to clinical use for various types of cancers and medications, most are limited to drugs targeting specific molecules, and none have been established for traditional cytotoxic drugs. For fluoropyrimidines, the standard drugs used in chemotherapy for gastrointestinal cancer, biomarker research has been conducted on targets such as thymidylate synthase(TS), dihydropyrimidine dehydrogenase(DPD), and thymidine phosphorylase(TP). The results of research on these targets have recently been reported, albeit retrospectively, in a number of additional studies and large-scale clinical trials. While some studies suggested that there is future potential for these targets, in general, it appears that there are insufficient data for their clinical application as biomarkers at present. Given the advances made toward the realization of personalized medicine, the discovery of biomarkers for fluoropyrimidines is of great importance and warrants further study.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Deoxycytidine/analogs & derivatives , Fluorouracil/analogs & derivatives , Fluorouracil/therapeutic use , Neoplasms/drug therapy , Oxonic Acid/therapeutic use , Tegafur/therapeutic use , Antimetabolites, Antineoplastic/metabolism , Capecitabine , Deoxycytidine/metabolism , Deoxycytidine/therapeutic use , Drug Combinations , Fluorouracil/metabolism , Humans , Neoplasms/diagnosis , Neoplasms/metabolism , Oxonic Acid/metabolism , Prognosis , Tegafur/metabolism
9.
Expert Opin Investig Drugs ; 18(3): 335-48, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19243284

ABSTRACT

S-1 is an oral fluoropyrimidine that is designed to improve the antitumor activity of 5-fluorouracil (5-FU) concomitantly with an intent to reduce its toxicity. S-1 consists of tegafur, a prodrug of 5-FU combined with two 5-FU biochemical modulators:5-chloro-2,4-dihydroxypyridine (gimeracil or CDHP), a competitive inhibitor of dihydropyrimidine dehydrogenase and oteracil potassium which inhibits phosphorylation of 5-FU in the gastrointestinal tract decreasing serious gastrointestinal toxicities,including nausea, vomiting, stomatitis and diarrhea. Being an oral agent, S-1 offers convenience of administration and prevents complications of central venous access such as infection, thrombosis and bleeding. S-1 has shown efficacy in both gastrointestinal as well non-gastrointestinal malignancies. The authors review the current literature and provide their expert opinion on the incorporation of S-1 in the treatment of solid malignancies [corrected].


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Oxonic Acid/pharmacology , Tegafur/pharmacology , Administration, Oral , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Clinical Trials as Topic , Drug Combinations , Drug Evaluation, Preclinical , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Oxonic Acid/administration & dosage , Oxonic Acid/metabolism , Oxonic Acid/therapeutic use , Tegafur/administration & dosage , Tegafur/metabolism , Tegafur/therapeutic use
10.
J Hypertens ; 26(12): 2326-38, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19008712

ABSTRACT

BACKGROUND AND OBJECTIVES: It has been demonstrated that hyperuricemia induces reno-cardiovascular damage resulting in hypertension and renal injury because of vascular endothelial dysfunction. The pathogenesis of hyperuricemia, endothelial dysfunction, hypertension, and renal injury is progressive, and develops into a vicious cycle. It is reasonable to suggest that an antihypertensive drug with endothelial protection may block this vicious cycle. Iptakalim, a novel antihypertensive drug undergoing phase-three clinical trials, is a new ATP-sensitive potassium channel opener and can ameliorate endothelial dysfunction. We hypothesized that iptakalim could prevent hypertension and retard the pathogenesis of endothelial dysfunction and renal injury in hyperuricemic rats. METHODS AND RESULTS: In rats with hyperuricemia induced by 2% oxonic acid and 0.1 mmol/l uric acid, iptakalim prevented increases in systolic blood pressure, reduced the impairment of endothelial vasodilator function, and attenuated renal dysfunction and pathological changes in glomerular and renal interstitial tissue at 0.5, 1.5, and 4.5 mg/kg orally daily for 4 weeks. Serum levels of nitric oxide and prostacyclin, and gene expression of endothelial nitric oxide synthase in the aortic and intrarenal tissue, were increased, whereas the serum levels of endothelin-1 and gene expression of endothelin-1 in aortic and intrarenal tissue were decreased. However, serum levels of angiotensin II and renin remained unchanged in the hyperuricemic rats treated with iptakalim. In cultured rat aortic endothelial cells, amelioration of endothelial dysfunction by iptakalim was suggested by inhibition of the overexpression of intercellular adhesive molecule-1, vascular cell adhesive molecule-1, and monocyte chemoattractant protein-1 mRNA induced by uric acid, and reversal of the inhibitory effects of uric acid on nitric oxide release in a concentration-dependent manner, which could be abolished by pretreatment with glibenclamide, an ATP-sensitive potassium channel blocker. Iptakalim ameliorated hyperuricemia in this rat model by decreasing renal damage through its antihypertensive and endothelial protective properties, and it had no direct effects on anabolism, catabolism and excretion of uric acid. CONCLUSION: These findings suggest that the activation of ATP-sensitive potassium channels by iptakalim can protect endothelial function against hypertension and renal injury induced by hyperuricemia. Iptakalim is suitable for use in hypertensive individuals with hyperuricemia.


Subject(s)
Endothelium, Vascular/metabolism , Hypertension/prevention & control , Hyperuricemia/complications , KATP Channels/metabolism , Kidney Diseases/prevention & control , 6-Ketoprostaglandin F1 alpha/metabolism , Angiotensin II/metabolism , Angiotensins , Animals , Cells, Cultured , Disease Models, Animal , Endothelin-1/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Hypertension/drug therapy , Hypertension/metabolism , Hyperuricemia/chemically induced , Hyperuricemia/metabolism , KATP Channels/agonists , KATP Channels/drug effects , Kidney/blood supply , Kidney/pathology , Kidney Diseases/drug therapy , Kidney Diseases/metabolism , Male , Nitric Oxide/metabolism , Oxonic Acid/metabolism , Propylamines/pharmacology , Propylamines/therapeutic use , Rats , Rats, Sprague-Dawley , Urate Oxidase/metabolism , Uric Acid/metabolism , Xanthine Oxidase/metabolism
11.
Clin Cancer Res ; 10(15): 4913-21, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15297391

ABSTRACT

PURPOSE: The oral fluoropyrimidine S-1, which consists of a mixture of a 5-fluorouracil (5-FU) prodrug (tegafur), a dihydropyrimidine dehydrogenase inhibitor [5-chloro-2,4-dihydroxypyrimidine (CDHP)], and an inhibitor of orotate phosphoribosyltransferase [potassium oxonate (oxonic acid)], was developed to increase the feasibility and therapeutic index of 5-FU administered orally. The principal objective of this study was to assess the feasibility of administering S-1 on a once-daily-for-28-day schedule every 5 weeks, determine the maximum tolerated dose, characterize the pharmacokinetics of S-1, and seek evidence of anticancer activity. EXPERIMENTAL DESIGN: Patients with advanced solid malignancies were treated with escalating doses of S-1 on a once-daily oral schedule for 28 days every 5 weeks. The maximum tolerated dose was defined as the highest dose in which fewer than two of the first six new patients experienced dose-limiting toxicity. The pharmacokinetic profiles of the tegafur, CDHP, and oxonic acid constituents were characterized. RESULTS: Twenty patients were treated with 72 courses of S-1 at three dose levels ranging from 50 to 70 mg/m(2)/day. Diarrhea, which was often associated with abdominal discomfort and cramping, was the principal dose-limiting toxicity of S-1 on this protracted schedule. Nausea, vomiting, mucositis, fatigue, and cutaneous effects were also observed but were rarely severe. Myelosuppression was modest and uncommon. A partial response and a 49% reduction in tumor size were observed in patients with fluoropyrimidine- and irinotecan-resistant colorectal carcinoma. The pharmacokinetic data suggested potent inhibition of 5-FU clearance by CHDP, with resultant 5-FU exposure at least 10-fold higher than that reported from equitoxic doses of tegafur modulated by uracil in the oral fluoropyrimidine UFT. CONCLUSIONS: The recommended dose for Phase II studies of S-1 administered once daily for 28 consecutive days every 5 weeks is 50 mg/m(2)/day. The pharmacokinetic data indicate substantial modulation of 5-FU clearance by CDHP. Based on these pharmacokinetic data, the predictable toxicity profile of S-1, and the low incidence of severe adverse effects at the recommended Phase II dose, evaluations of S-1 on this schedule are warranted in malignancies that are sensitive to the fluoropyrimidines.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Oxonic Acid/administration & dosage , Oxonic Acid/pharmacokinetics , Pyridines/administration & dosage , Pyridines/pharmacokinetics , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics , Tegafur/administration & dosage , Tegafur/pharmacokinetics , Adult , Aged , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacokinetics , Antineoplastic Agents/pharmacology , Area Under Curve , Dose-Response Relationship, Drug , Drug Combinations , Enzyme Inhibitors/pharmacology , Female , Follow-Up Studies , Humans , Lung Neoplasms/drug therapy , Male , Maximum Tolerated Dose , Middle Aged , Orotate Phosphoribosyltransferase/antagonists & inhibitors , Oxonic Acid/metabolism , Pyridines/pharmacology , Stomach Neoplasms/drug therapy , Tegafur/pharmacology , Time Factors
12.
Anticancer Drugs ; 15(2): 85-106, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15075664

ABSTRACT

The fluoropyrimidine anticancer agent 5-fluorouracil (5-FU) is active in a wide range of solid tumors, particularly gastric, colorectal, and head and neck cancers. Whilst infusional 5-FU is associated with higher response rates and a favorable safety profile as compared to the classical i.v. bolus administration, prolonged infusions can be inconvenient for the patients and catheter-related problems are common complications. An oral 5-FU formulation would allow for sustained 5-FU plasma concentrations, mimicking the pharmacokinetics (PK) of a continuous infusion with the addition of convenience of administration. The oral administration of 5-FU itself is not feasible due to the high activity of dihydropyrimidine dehydrogenase in the gut wall, which causes rapid metabolism of the drug, and results in decreased and erratic absorption of 5-FU and non-linear PK. To bypass this problem, oral fluoropyrimidine derivatives were developed either in the form of 5-FU prodrugs (i.e. tegafur, doxifluridine or capecitabine), or as enzyme inhibitors (i.e. eniluracil) administered with 5-FU, or as both prodrugs and enzyme inhibitors (i.e. S-1, UFT or BOF-A2). This review will focus on the oral fluoropyrimidine S-1, which consists of the 5-FU prodrug tegafur (ftorafur, FT) and two enzyme inhibitors, i.e. CDHP (5-chloro-2,4-dihydroxypyridine) and OXO (potassium oxonate), in a molar ratio of 1(FT):0.4 (CDHP):1(OXO). Phase II trials have demonstrated that S-1, as a single agent, is active for the treatment of gastric, colorectal, head and neck, breast, non-small cell lung, and pancreatic cancers. Phase III trials are currently underway in gastric cancer and these results are awaited to confirm the phase II findings. Furthermore, the combination of S-1 with cisplatin (CDDP), irinotecan or docetaxel for the treatment of gastric cancer and with CDDP for non-small cell and pancreatic cancer is feasible and active. The activity observed with S-1 in the phase II studies is at least equivalent, if not better, than continuous i.v. and bolus 5-FU and the other oral fluoropyrimidines. Thus, we may finally be seeing the realization of oral treatments for the management of various solid tumors and could be on the brink of a new approach to treatment strategies.


Subject(s)
Administration, Oral , Enzyme Inhibitors/administration & dosage , Gastrointestinal Neoplasms/drug therapy , Oxonic Acid/administration & dosage , Oxonic Acid/therapeutic use , Prodrugs/therapeutic use , Pyridines/administration & dosage , Pyridines/therapeutic use , Pyrimidines/therapeutic use , Tegafur/administration & dosage , Tegafur/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Clinical Trials as Topic/methods , Dosage Forms , Drug Administration Schedule , Drug Combinations , Drug Delivery Systems , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacokinetics , Humans , Oxonic Acid/metabolism , Prodrugs/metabolism , Prodrugs/pharmacology , Pyridines/metabolism , Pyridines/pharmacokinetics , Pyrimidines/metabolism , Pyrimidines/pharmacokinetics , Tegafur/metabolism , Tegafur/pharmacokinetics
13.
Acta Crystallogr D Biol Crystallogr ; 60(Pt 3): 453-62, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14993669

ABSTRACT

High-resolution X-ray structures of the complexes of Aspergillus flavus urate oxidase (Uox) with three inhibitors, 8-azaxanthin (AZA), 9-methyl uric acid (MUA) and oxonic acid (OXC), were determined in an orthorhombic space group (I222). In addition, the ligand-free enzyme was also crystallized in a monoclinic form (P2(1)) and its structure determined. Higher accuracy in the three new enzyme-inhibitor complex structures (Uox-AZA, Uox-MUA and Uox-OXC) with respect to the previously determined structure of Uox-AZA (PDB code 1uox) leads to a reversed position of the inhibitor in the active site of the enzyme. The corrected anchoring of the substrate (uric acid) allows an improvement in the understanding of the enzymatic mechanism of urate oxidase.


Subject(s)
Aspergillus flavus/enzymology , Catalytic Domain , Fungal Proteins , Oxonic Acid , Urate Oxidase , Uric Acid , Xanthines , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Fungal Proteins/antagonists & inhibitors , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Humans , Mice , Molecular Conformation , Molecular Sequence Data , Oxonic Acid/chemistry , Oxonic Acid/metabolism , Protein Binding/physiology , Protein Structure, Tertiary , Sequence Alignment , Urate Oxidase/antagonists & inhibitors , Urate Oxidase/chemistry , Urate Oxidase/metabolism , Uric Acid/analogs & derivatives , Uric Acid/chemistry , Uric Acid/metabolism , Xanthines/chemistry , Xanthines/metabolism
14.
Gastric Cancer ; 6 Suppl 1: 13-8, 2003.
Article in English | MEDLINE | ID: mdl-12775014

ABSTRACT

BACKGROUND: Favorable results have been reported for the novel oral anticancer agent S-1 (TS-1) in clinical studies of advanced gastric cancer with peritoneal dissemination. In the present study we assessed its pharmacokinetics, inhibitory effects, and effect on survival time in an animal model. METHODS: A model of peritoneal dissemination was created by intraperitoneally implanting 4-week-old female BALBc nu/nu mice with the human gastric cancer cell line MKN-45 after transfection with a fluorescent protein-expressing vector. Pharmacokinetics were investigated by measuring intratumor, peritoneal lining, and blood concentrations after the administration of S-1 and fluorouracil (5-FU). The effect of S-1 on survival time was also assessed, by administration once daily to seven animals per group, starting on day 7 after implantation, and survival time was compared with that of an untreated control group. The inhibitory effect of S-1 on peritoneal dissemination was evaluated by killing mice at the start of administration, and 1 and 3 weeks after the start of administration, and examining them for the presence of peritoneal dissemination under a fluorescence stereomicroscope. RESULTS: Maintenance of high 5-FU concentrations in the intraperitoneal tumors was confirmed in the S-1 group, and survival time was prolonged without any decrease in oral food intake or body weight. CONCLUSION: Assessment in a model of peritoneal dissemination of gastric cancer showed that the novel oral anticancer agent S-1 was effective against peritoneal dissemination, and that it improved the survival rate.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Oxonic Acid/pharmacology , Peritoneal Neoplasms/secondary , Pyridines/pharmacology , Stomach Neoplasms/pathology , Tegafur/pharmacology , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/metabolism , Body Weight , Disease Models, Animal , Drug Combinations , Eating , Female , Fluorouracil/administration & dosage , Fluorouracil/metabolism , Fluorouracil/pharmacology , Japan , Maximum Tolerated Dose , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Oxonic Acid/administration & dosage , Oxonic Acid/metabolism , Pyridines/administration & dosage , Pyridines/metabolism , Stomach Neoplasms/drug therapy , Survival Analysis , Tegafur/administration & dosage , Tegafur/metabolism , Time Factors
15.
Pharmacogenetics ; 12(4): 299-306, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12042667

ABSTRACT

In a clinical study, a newly developed anticancer drug, TS-1 capsule, which contained tegafur (FT) and 5-chloro-2,4-dihydroxypyridine, an inhibitor of dihydropyrimidine dehydrogenase, was orally administered to five gastric cancer patients (patients 1-5). The total area under the plasma FT concentration-time curve in patient 1 was four-fold higher than in other patients. Since cytochrome P450 2A6 (CYP2A6) has been reported to metabolize FT to yield 5-fluorouracil (5-FU), it was postulated that the poor metabolic phenotype of patient 1 was caused by mutations of the CYP2A6 gene. Thus, alleles for the CYP2A6 genes derived from patient 1 were completely sequenced. It was found that one allele was CYP2A6*4C, which was a whole deleted allele for the human CYP2A6 gene. The other allele was a novel mutant allele (CYP2A6*11) in which thymine at nucleotide 670 was changed to cytosine. The nucleotide change caused an amino acid change from serine at residue 224 to proline. To examine whether or not the amino acid change affected CYP2A6 activity, we expressed an intact or mutant CYP2A6 together with NADPH-P450 oxidoreductase in Escherichia coli, and compared the capacity of the wild and mutant enzymes to metabolize FT to 5-FU. The Vmax value for FT metabolism by the mutant CYP2A6 was approximately one-half of the value of the intact CYP2A6, although the Km values were nearly the same. From these results, we conclude that the poor metabolic phenotype of patient 1 was caused by the existence of the two mutant alleles, CYP2A6*4C and the new variant CYP2A6*11.


Subject(s)
Antimetabolites, Antineoplastic/metabolism , Aryl Hydrocarbon Hydroxylases/genetics , Mixed Function Oxygenases/genetics , Oxonic Acid/metabolism , Pyridines/metabolism , Stomach Neoplasms/genetics , Tegafur/metabolism , Administration, Oral , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/blood , Area Under Curve , Aryl Hydrocarbon Hydroxylases/metabolism , Coumarins/metabolism , Cytochrome P-450 CYP2A6 , DNA Primers/chemistry , DNA, Neoplasm/blood , DNA, Neoplasm/metabolism , Drug Combinations , Escherichia coli , Genotype , Humans , Kinetics , Middle Aged , Mixed Function Oxygenases/metabolism , Mutagenesis, Site-Directed , Oxonic Acid/administration & dosage , Oxonic Acid/blood , Polymerase Chain Reaction , Polymorphism, Genetic , Pyridines/administration & dosage , Pyridines/blood , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Stomach Neoplasms/enzymology , Tegafur/administration & dosage , Tegafur/blood , Transfection
16.
Drug Metab Dispos ; 28(10): 1162-7, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10997934

ABSTRACT

S-1, a new oral 5-fluorouracil (5-FU)-derivative antitumor agent, is composed of tegafur, 5-chloro-2,4-dihydropyridine, and potassium oxonate (Oxo). Oxo, which inhibits the phosphorylation of 5-FU, is added to reduce the gastrointestinal (GI) toxicity of the agent. In this study, we investigated the tissue distribution and the metabolic fate of Oxo in rats after oral administration of S-1. Oxo was mainly distributed to the intracellular sites of the small intestines in a much higher concentration than 5-FU, but little distributed to other tissues, including tumorous ones in which 5-FU was observed after oral administration of S-1. Plasma concentration-time profiles of Oxo and its metabolites after i.v. and oral administration of S-1 revealed that Oxo was mainly converted to cyanuric acid in the GI tract. Furthermore, the analysis of drug-related radioactivity in GI contents and in vitro studies suggested that Oxo was converted to cyanuric acid by two routes, the first being direct conversion by the gut flora in the cecum, and the second, conversion by xanthine oxidase or perhaps by aldehyde oxidase after degradation to 5-azauracil (5-AZU) by the gastric acid. These results indicate that, although a part of the administered Oxo was degraded in the GI tract, Oxo was mainly distributed to the intracellular sites of the small intestines in a much higher concentration than 5-FU and that little was distributed to other tissues, including tumors. We conclude that this is the reason why Oxo suppresses the GI toxicity of 5-FU without affecting its antitumor activity.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Oxonic Acid/pharmacokinetics , Pyridines/pharmacokinetics , Tegafur/pharmacokinetics , Uracil/analogs & derivatives , Administration, Oral , Allopurinol/pharmacology , Animals , Antimetabolites, Antineoplastic/metabolism , Area Under Curve , Biotransformation , Carbon Radioisotopes , Chlorpromazine/pharmacology , Drug Combinations , Drugs, Chinese Herbal/pharmacology , Fluorouracil/blood , Fluorouracil/metabolism , Glycyrrhiza , Intestine, Small/metabolism , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Oxonic Acid/blood , Oxonic Acid/metabolism , Paeonia , Pyridines/blood , Pyridines/metabolism , Rats , Rats, Inbred Strains , Sarcoma, Yoshida/drug therapy , Sarcoma, Yoshida/metabolism , Tegafur/blood , Tegafur/metabolism , Tissue Distribution , Triazines/blood , Triazines/metabolism , Uracil/metabolism , Xanthine Oxidase/metabolism
18.
J Membr Biol ; 169(1): 13-27, 1999 May 01.
Article in English | MEDLINE | ID: mdl-10227848

ABSTRACT

Recombinant protein, designated UAT, prepared from a cloned rat renal cDNA library functions as a selective voltage-sensitive urate transporter/channel when fused with lipid bilayers. Since we previously suggested that UAT may represent the mammalian electrogenic urate transporter, UAT has been functionally characterized in the presence and absence of potential channel blockers, several of which are known to block mammalian electrogenic urate transport. Two substrates, oxonate (a competitive uricase inhibitor) and pyrazinoate, that inhibit renal electrogenic urate transport also block UAT activity. Of note, oxonate selectively blocks from the cytoplasmic side of the channel while pyrazinoate only blocks from the channel's extracellular face. Like oxonate, anti-uricase (an electrogenic transport inhibitor) also selectively blocks channel activity from the cytoplasmic side. Adenosine blocks from the extracellular side exclusively while xanthine blocks from both sides. These effects are consistent with newly identified regions of homology to uricase and the adenosine A1/A3 receptor in UAT and localize these homologous regions to the cytoplasmic and extracellular faces of UAT, respectively. Additionally, computer analyses identified four putative alpha-helical transmembrane domains, two beta sheets, and blocks of homology to the E and B loops of aquaporin-1 within UAT. The experimental observations substantiate our proposal that UAT is the molecular representation of the renal electrogenic urate transporter and, in conjunction with computer algorithms, suggest a possible molecular structure for this unique channel.


Subject(s)
Carrier Proteins/genetics , Organic Anion Transporters , Uric Acid/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Carrier Proteins/drug effects , Carrier Proteins/metabolism , Cattle , Cloning, Molecular , Lipid Bilayers/metabolism , Models, Molecular , Molecular Sequence Data , Oxonic Acid/metabolism , Pyrazinamide/analogs & derivatives , Pyrazinamide/pharmacology , Rats , Receptors, Purinergic P1/analysis , Sequence Homology, Amino Acid , Structure-Activity Relationship , Urate Oxidase/metabolism , Xanthine/metabolism
19.
Cancer Res ; 53(17): 4004-9, 1993 Sep 01.
Article in English | MEDLINE | ID: mdl-7689420

ABSTRACT

The possibility of decreasing the gastrointestinal (GI) toxic effects of 5-fluorouracil (5-FU) on the digestive tract such as its injury of cells and induction of diarrhea, without reducing its antitumor activity, was investigated in rats. Oxonic acid was found to inhibit the phosphorylation of 5-FU to 5-fluorouridine-5'-monophosphate catalyzed by pyrimidine phosphoribosyl-transferase in a different manner from allopurinol in cell-free extracts and intact cells in vitro. On p.o. administration of 5-FU (2 mg/kg) and a potent inhibitor of 5-FU degradation to Yoshida sarcoma-bearing rats, oxonic acid (10 mg/kg) was found to inhibit the formation of 5-fluorouridine-5'-monophosphate from 5-FU and its subsequent incorporation into the RNA fractions of small and large intestine but not of tumor and bone marrow tissues. This selective inhibition of 5-FU phosphorylation in the GI tract was due to the much higher concentrations of oxonic acid in GI tissues than in other tissues and the blood. On p.o. administration with the 5-FU derivative, UFT, which is a combined form of 1 M tegafur and 4 M uracil and usually administered p.o. to cancer patients in Japan, oxonic acid (10-50 mg/kg) markedly reduced injury of GI tissues and/or severe diarrhea without influencing the antitumor effect of UFT. These findings suggest that coadministration of oxonic acid suppresses the GI toxicity of 5-FU and its derivatives without affecting their antitumor activity and thus prolongs the life span of cancer-bearing rats.


Subject(s)
Digestive System/drug effects , Fluorouracil/antagonists & inhibitors , Oxonic Acid/pharmacology , Administration, Oral , Animals , Digestive System/metabolism , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Fluorouracil/metabolism , Infusions, Intravenous , Liver/metabolism , Male , Oxonic Acid/administration & dosage , Oxonic Acid/metabolism , Phosphorylation/drug effects , RNA/metabolism , Rats , Sarcoma, Yoshida/drug therapy , Sarcoma, Yoshida/metabolism , Uracil Nucleotides/metabolism
20.
Chem Biol Interact ; 73(2-3): 235-47, 1990.
Article in English | MEDLINE | ID: mdl-2155712

ABSTRACT

Uric acid is an end-product of purine metabolism in Man, and has been suggested to act as an antioxidant in vivo. Products of attack upon uric acid by various oxidants were measured by high performance liquid chromatography. Hypochlorous acid rapidly oxidized uric acid, forming allantoin, oxonic/oxaluric and parabanic acids, as well as several unidentified products. HOCl could oxidize all these products further. Hydrogen peroxide did not oxidize uric acid at detectable rates, although it rapidly oxidized oxonic acid and slowly oxidized allantoin and parabanic acids. Hydroxyl radicals generated by hypoxanthine/xanthine oxidase or Fe2(+)-EDTA/H2O2 systems also oxidized uric acid to allantoin, oxonic/oxaluric acid and traces of parabanic acid. Addition of ascorbic acid to the Fe2(+)-EDTA/H2O2 system did not increase formation of oxidation products from uric acid, possibly because ascorbic acid can 'repair' the radicals resulting from initial attack of hydroxyl radicals upon uric acid. Mixtures of methaemoglobin or metmyoglobin and H2O2 also oxidized uric acid: allantoin was the major product, but some parabanic and oxonic/oxaluric acids were also produced. Caeruloplasmin did not oxidize uric acid under physiological conditions, although simple copper (Cu2+) ions could, but this was prevented by albumin or histidine. The possibility of using oxidation products of uric acid, such as allantoin, as an index of oxidant generation in vivo in humans is discussed.


Subject(s)
Hydantoins , Uric Acid/metabolism , Allantoin/isolation & purification , Allantoin/metabolism , Ceruloplasmin/metabolism , Chromatography, High Pressure Liquid , Edetic Acid , Ferric Compounds/metabolism , Hemoglobins/metabolism , Hydrogen Peroxide/metabolism , Hydroxides/metabolism , Hydroxyl Radical , Hypochlorous Acid , Hypoxanthine , Hypoxanthines/metabolism , Imidazoles/isolation & purification , Imidazoles/metabolism , Kinetics , Molecular Structure , Myoglobin/metabolism , Oxidation-Reduction , Oxonic Acid/isolation & purification , Oxonic Acid/metabolism , Xanthine Oxidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...