首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 453 毫秒
1.
Combined recirculation of the rat liver (L) and kidney (IPK) at 10 ml min–1 per organ (LK) was developed to examine the hepatorenal handling of the precursor-metabolite pair: [14C]-enalapril and [3H]enalaprilat. Loading doses followed by constant infusion of [14C]enalapril and preformed [3H]enalaprilat to the reservoirs of the IPK or the LK preparation was used to achieve steady stale conditions. In both organs, enalapril was mostly metabolized to its dicarboxylic acid metabolite, enalaprilat, which was excreted unchanged. At steady state, the fractional excretion for [14C]enalapril (FE=0.45 to 0.48) and preformed [3H]enalaprilat (FE{pmi}=1.1) were constant and similar for both the IPK and LK. The additivity of clearance was demonstrated in the LK preparation, namely, the total clearance of enalapril was the sum of its hepatic and renal clearances. However, the apparent fractional excretion for fanned [14C]enalaprilat, FE{mi} and the apparent urinary clearance were time-dependent and higher than the corresponding values for preformed [3H]enalaprilat in both the IPK and LK. The FE{mi} and urinary clearance values further differed between the IPK and LK. Biliary clearance of formed vs. preformed enalaprilat displayed the same discrepant trends as observed for FE{mi} vs. FE{pmi} for the LK. These observations on the time-dependent and variable excretory clearance (urinary or biliary) of the formed metabolite vs. the constant, and much reduced, excretory clearance of the preformed metabolite are due to dual contributions to formed metabolite excretion: the nascently formed, intracellular metabolite which immediately underwent excretion and the formed metabolite which reentered the circulation, behaved as a preformed species. When data for the IPK and LK preparations were modeled with a physiological model with parameters previously reported for the L and IPK, all data, including metabolite excretory clearances, were well predicted. Model simulations revealed that the apparent FE{mi} differed between the LK and IPK preparations when the liver was present as an additional metabolite formation organ; the apparent excretory (urinary orGlossary k0 infusion rate into the reservoir - CR reservoir concentration - COut,k and COut,L venous concentrations for the kidney and liver - Cp,k and cP,L concentrations in renal and hepatic plasma, respectively - Ck and CL concentrations in kidney and liver tissue, respectively - CU and CBile concentrations in urine and bile, respectively - CL b in andCL b ef influx and efflux clearances, respectively, at the basolateral membrane of the renal tubular cell - C l in and CL l ef influx and efflux clearances, respectively, at the luminal membrane of the renal tubular cell - CL int,K m renal metabolic intrinsic clearance of the drug - CL d in and CL d ef influx and efflux clearances, respectively, at the sinusoidal membrane - CL int m,L hepatic metabolic intrinsic clearance of the drug - CL int,L b biliary intrinsic clearance - VR plasma reservoir volume - VP,K and VP,L plasma volumes of the kidney and liver, respectively - VK and VL tissue volumes of the kidney and liver, respectively - VU and VBile volumes of urine and bile, respectively - QK and QL total renal and hepatic plasma flow rates, respectively - GFR glomerular filtration rate - QU and QBile urine and bile flow rates, respectively - fP, fK, and fL unbound fractions in plasma and kidney and liver tissue, respectively This work was supported by the Medical Research Council of Canada. I. A. M. de Lannoy was a recipient of the Ontario Graduate Scholarship from the Ontario Ministry of Health; K. S. Pang was a recipient of the Faculty Development Award, Medical Research Council.  相似文献   

2.
Studies in the once-through perfused rat liver with the simultaneous delivery of 14 C-enalapril and its polar diacid metabolite, 3H-enalaprilat, revealed different extents of elimination (exclusively by biliary excretion) for the generated (14C-enalaprilat) and preformed (3H-enalaprilat) metabolite (18 and 5% dose) [Pang, Cherry, Terrell, and Ulm: Drug Metab. Dispos. 12, 309-313 (1984)]. The present re-examination of data provided an explanation for these discrepant observations: enalaprilat, being a polar dicarboxylic acid, encounters more of a diffusional barrier than its precursor, enalapril, an ethyl ester of enalaprilat. Programs written in Fortran 77 on mass balance relationships were employed to simulate data upon varying the diffusional clearances for drug (CLd) and metabolite [CLd(mi)] from 1 to 5000 ml/min. The metabolic and biliary intrinsic clearances for drug and metabolite were found by trial and error such that the combinations of all clearance parameters yielded data similar to enalaprilat, and 3H-enalaprilat. Our finding indicated that the diffusional clearance for enalaprilat was low (2 ml/min) compared to that of enalapril (75 ml/min). The presence of a diffusional barrier for enalaprilat retards entry of the preformed metabolite into hepatocytes but prevents efflux of the intracellularly formed generated metabolite into sinusoidal blood, thereby enhancing generated metabolite elimination.  相似文献   

3.
We examined data from our previous studies in which we not only delivered perfusate containing tracer concentrations of [14C]phenacetin and its metabolite [3H]acetaminophen under constant perfusate flow (10 ml/ min/ liver) into the rat liver preparation just once, but also recirculated fresh reservoir perfusate containing a tracer dose of [14C]phenacetin through the same rat liver preparation. From the single-pass studies, estimates of fm, the fractional rate of conversion for [14C]phenacetin to form [14C]acetaminophen, and F(M.P), the apparent availability of [14C]acetaminophen, were obtained by determining the concentrations of [14C]acetaminophen in the perfusate before and after incubation with Glusulase. These estimates were fm=0.871±0.16 and F(M.P)=0.43±0.10. These and the steady-state clearance values of phenacetin (9.1±0.8ml/min) and acetaminophen (6.7±0.7ml/min) from the single-pass studies were used to predict the concentrations of [14C]acetaminophen in the reservoir perfusate on recirculation of [14C]phenacetin. We found that the sequential first-pass elimination of the metabolite must be considered when the metabolite is highly extracted by the liver. If we had neglected to take this into account, the fractional rate of conversion of a precursor to form a metabolite and the rate of formation of the metabolite would have been underestimated by the factor F(M.P).  相似文献   

4.
The interrelationship of the pharmacokinetics of a drug and the expiration of carbon dioxide formed as a metabolite have been studied. The pharmacokinetic characteristics of the drug that affect the usefulness of the carbon dioxide excretion as a measure of liver function were examined by means of computer simulations. The parent drug extraction ratio, fraction demethylated, volume of distribution, and absorption rate of an oral dosage form all contribute to the carbon dioxide breath test result. A drug that would be a useful substrate when the carbon dioxide breath test is used as a probe for changes in liver function should be at least 50% metabolized by demethylation, have a hepatic extraction ratio of 0.2–0.5, and be administered in a form that is rapidly absorbed.Appendix b. symbols CL c net clearance of formaldehyde to carbon dioxide - CL int,f intrinsic hepatic clearance of formation of formaldehyde from parent drug (bound and unbound to plasma proteins) - CL int,p intrinsic hepatic clearance of total parent drug (bound and unbound to plasma proteins) - CL sys,f systemic hepatic clearance of formation of formaldehyde from parent drug,Q H CL int,f /(Q H +CL int,p ) - CL sys,p systemic hepatic clearance of parent drug,Q H CL int,p /(Q H +CL int,p ) - E extraction ratio,CL int,p /(Q H +CL int,p - F I-E fraction escaping first-pass metabolism,Q H/(Q H +CL int,p - fm fraction of parent drug metabolized by demethylation to formaldehyde,CL int,f /CL int,p - HCHO amount of formaldehyde - [HCHO] concentration of formaldehyde - a absorption rate constant - M i metabolite of P formed by routes other than demethylation - M 1 metabolite of P formed by demethylation - P amount of parent drug in the body - [P] concentration of parent drug measured in arterial blood - P A amount of parent drug at absorption site - P L amount of parent drug in the liver - Q H hepatic blood flow - V F volume of distribution of formaldehyde - V p volume of distribution of parent drug  相似文献   

5.
A physiologically based kidney model was developed to describe the metabolism of enalapril and explain the observed discrepancies between generated and preformed enalaprilat (metabolite) elimination in the constant flow single-pass and recirculating isolated perfused rat kidney preparations (IPKs) as a result of the differing points of origin of the metabolite within the kidney, subsequent to the simultaneous delivery of14Cenalapril and3H-enalaprilat. The model incorporated clearances for diffusion/transport of drug and metabolite across the basolateral and luminal membranes of the renal cells, an intrinsic clearance for renal drug metabolism, in addition to physiological variables such as perfusate flow rate, glomerular filtration rate, and urine flow rate. Nonlinear curve fitting of single-pass and recirculating data was performed to estimate the rate-limiting step in the renal elimination of enalaprilat. Through fitting and simulation procedures, we were able to predict metabolic and excretory events for enalapril (renal extraction ratio 0.25–0.3; fractional excretion, FE, was less than unity) and the relatively constant pattern of urinary excretion of preformed enalaprilat (extraction ratio 0.07; FE1). The extraction ratio of the intrarenally formed enalaprilat in single-pass IPK was about twofold that for the preformed metabolite, whereas the FEs of generated enalaprilat in recirculating IPKs were >1, and tended to increase, then decrease with perfusion time. These observations were explained by the optimized parameters which indicated that efflux from cell to lumen was rate-controlling in the excretion of enalaprilat, and another small transport barrier also existed at the basolateral membrane; the lower extraction ratio of preformed enalaprilat was due to its poor transmembrane clearance at the basolateral membrane. The variable FEs for generated enalaprilat vs. the relatively constant FE for preformed metabolite in the recirculating IPK was explained by the changing contributions of both circulating and intrarenal metabolite to metabolite excretion.This work was supported by the Medical Research Council of Canada. I. A. M. de Lannoy was a recipient of the Ontario Graduate Scholarship, and K. S. Pang is a recipient of the Faculty Development Award from MRC, Canada M5S 2S2.  相似文献   

6.
1. Following oral administration of [U-14C]-(+)-catechin to the rat, the major biliary metabolite was shown to be the glucuronide of 3′-O-methyl-(+) catechin by chromatography and mass spectrometry.

2. [methyl-14C]-O-Methyl-(+)-catechin was formed by incubation of (+)-catechin with S-adenosyl-L-[methyl-14C]methionine in vitro in both liver homogenates and in the presence of purified catechol-O-methyl transferase.

3. Alkaline micro-fusion techniques have been used to determine the position of the O-methyl substituent in the B-ring of the isolated O-methyl-(+)-catechin glucuronide.  相似文献   

7.
Enalaprilat (MK-422), a new and potent angiotensin- converting enzyme inhibitor, and its monoethyl ester precursor, enalapril, were studied in a single pass perfused rat liver preparation under constant perfusate flow (10 ml/min) at concentrations of 0.29-0.41 microM for 14C-enalapril and 0.01-0.015 microM for 3H- enalaprilat . During their simultaneous delivery to the same rat liver preparation, the steady state hepatic extraction ratio of 14C-enalapril was high (0.861 +/- 0.02) and 14C- enalaprilat appeared rapidly in effluent perfusate plasma. Of the enalapril dose, 22.7 +/- 6.9% appeared in bile. 14C- Enalaprilat accounted for 79% of the total radioactivity in bile (18% of dose) whereas 14C-enalapril was present only as 10% of the total (2.3% of dose). By contrast, the steady state hepatic extraction of 3H- enalaprilat was very low (0.053) and the disappearance was virtually identical to the appearance of 3H- enalaprilat in bile. These findings suggest that diffusional barrier exists for enalaprilat as the preformed metabolite, which hinders penetration into hepatocytes, and therefore, elimination. The precursor, enalapril, effectively brings enalaprilat into hepatocytes were more extensive biliary excretion of the generated metabolite takes place. This account adds to our further understanding of metabolite kinetics; in addition to the uneven distribution of enzyme system and the intrinsic clearance for metabolite formation and elimination, the presence of a diffusional barrier is another important determinant which may cause deviations between the kinetics of a generated and performed metabolite.  相似文献   

8.
Dasatinib (SPRYCEL®) is a multiple kinase inhibitor approved for the treatment of chronic myelogenous leukemia and Philadelphia chromosome‐positive acute lymphoblastic leukemia in patients with resistance to prior therapy, including imatinib mesylate (Gleevec®). Radiolabeled dasatinib and its piperazine N‐dealkyl metabolite were synthesized to investigate absorption, distribution, metabolism, and elimination of the compounds in humans and animals. These compounds were prepared following a three‐step sequence, which included thiazole carboxamide formation via cyclization of labeled thiourea with a brominated oxyacrylamide precursor. In the final step a common intermediate was converted to either [14C]dasatinib or the radiolabeled piperazine N‐dealkyl metabolite with labeling in the aminothiazole ring. Syntheses of both compounds were achieved with radiochemical purities in excess of 98%. Stable‐labeled dasatinib and the piperazine N‐dealkyl metabolite were also needed for use as mass spectral internal standards in support of bioanalytical assays. By following the same route used for the carbon‐14 synthesis, [13C4, 15N2]dasatinib and the [13C4, 15N2]metabolite were prepared with labeling in both the dichloropyrimidine and thiazole ring systems. This convergent process introduced stable isotope labeling through (1, 2, 3‐13C3) diethyl malonate and [13C,15N2]thiourea. Copyright © 2008 John Wiley & Sons, Ltd.  相似文献   

9.
1. The roles of multidrug resistance-associated protein (Mrp) 2 deficiency and Mrp3 up-regulation were evaluated on the metabolism and disposition of gemfibrozil.

2. Results from in vitro studies in microsomes showed that the hepatic intrinsic clearance (CLint) for the oxidative metabolism of gemfibrozil was slightly higher (1.5-fold) in male TR? rats, which are deficient in Mrp2, than in wild-type Wistar rats, whereas CLint for glucuronidation was similar in both strains.

3. The biliary excretion of intravenously administered [14C]gemfibrozil was significantly impaired in TR? rats compared with Wistar rats (22 versus 93% of the dose excreted as the acyl glucuronides over 72?h). Additionally, the extent of urinary excretion of radioactivity was much higher in TR? than in Wistar rats (78 versus 2.6% of the dose).

4. There were complex time-dependent changes in the total radioactivity levels and metabolite profiles in plasma, liver and kidney, some of which appeared to be related to the up-regulation of Mrp3.

5. Overall, it was demonstrated that alterations in the expression of the transporters Mrp2 and Mrp3 significantly affected the excretion as well as the secondary metabolism and distribution of [14C]gemfibrozil.  相似文献   

10.
Purpose The twofold aim of this study was to characterize in vivo in rats the pharmacokinetics (PK) and pharmacodynamics (PD) of L6-OH, a metabolite of lerisetron with in vitro pharmacological activity, and evaluate the extent to which L6-OH contributes to the overall effect. Methods The PK of L6-OH was determined directly postmetabolite i.v. dose (PK-1), and also simultaneously for L (lerisetron concentration) and for generated L6-OH after lerisetron dose (200 μg kg−1, i.v.), using Nonlinear Mixed Effects Modeling with an integrated parent–metabolite PK model (PK-2). Surrogate effect was measured by inhibition of serotonin-induced bradycardia. Protein binding was assayed via ultrafiltration and all quantification was performed via liquid chromatography-electrospray ionization-mass spectrometry. Results L6-OH showed elevated plasma and renal clearances, and volume of distribution (PK-1). The in vivo potency (PD) of L6-OH was high (EC50 = 0.098 ng mL−1 and EC50unbound = 0.040 ng mL−1). Total clearance for L (PK-2) in the presence of generated L6-OH (CLL = CL→L6-OH + CLn) was 0.0139 L min−1. Most of this clearance was L6-OH formation (Fc = 99.6%), but only an 8.6% fraction of L6-OH was released into the bloodstream. The remainder undergoes biliar and fecal elimination. The parameters estimated from PK-2 were used to predict concentrations of L6-OH (CpL6) generated after a lerisetron therapeutic dose (10 μg kg−1) in the rat. These concentrations are needed for the PD model and are below the quantification limit. CpL6max was less than the EC50 of L6-OH. Conclusions We conclude that after lerisetron administration, L6-OH is extensively formed in the rat but it is quickly eliminated; therefore, besides being equipotent with the parent drug, the L6-OH metabolite does not influence the effect of lerisetron.  相似文献   

11.
Synthesis of multiple stable isotope‐labeled antibacterial agent RWJ‐416457, (N‐{3‐[3‐fluoro‐4‐(2‐methyl‐2,6‐dihydro‐4H‐pyrrolo[3,4‐c]pyrazol‐5‐yl)‐phenyl]‐2‐oxo‐oxazolidin‐5‐ylmethyl}‐acetamide), and its major metabolite, N‐{3‐[4‐(2,6‐dihydro‐4H‐pyrrolo[3,4‐c]pyrazol‐5‐yl)‐3‐fluoro‐phenyl]‐2‐oxo‐oxazolidin‐5‐ylmethyl}‐acetamide, is described. The stable isotope‐labeled [13CD3]RWJ‐416457 was prepared readily by acetylation of the precursor amine, 5‐aminomethyl‐3‐[3‐fluoro‐4‐(2‐methyl‐2,6‐dihydro‐4H‐pyrrolo[3,4‐c]pyrazol‐5‐yl)‐phenyl]‐oxazolidin‐2‐one with CD313COCl in pyridine. Synthesis of the stable isotope‐labeled metabolite involved a construction of multiple isotope‐labeled pyrazole ring. N,N‐dimethyl(formyl‐13C,D)amide dimethyl acetal was first prepared by treating N,N‐dimethyl(formyl‐13C,D)amide with dimethyl sulfate, followed by sodium methoxide. Then, N‐{3‐[3‐fluoro‐4‐(3‐oxo‐pyrrolidin‐1‐yl)‐phenyl]‐2‐oxo‐oxazolidin‐5‐ylmethyl}‐acetamide was condensed with N,N‐dimethyl(formyl‐13C,D)amide dimethyl acetal, and the resultant β‐ketoenamine intermediate underwent pyrazole ring formation with hydrazine‐15N2, to give the [13C15N2D]‐labeled metabolite. Copyright © 2012 John Wiley & Sons, Ltd.  相似文献   

12.
Stable isotope‐labeled ([13C2, D3]) and carbon 14‐labeled vasopressin receptor antagonist RWJ‐676070, a spirobenzazepine amide, were prepared in separate syntheses for use in drug metabolism studies. The stable isotopically labeled sample was prepared starting from [13C2, D6]dimethyl sulfate and [13C]copper (I) cyanide in nine steps with a 14% overall isotopic yield. The 14C label was introduced in five steps starting from [14C]potassium cyanide to provide material having a specific activity of 58 mCi/mmol (2.15 GBq/mmol). Selective hydrolysis of the metabolically labile amide bond in [13C2, D3]RWJ‐676070 gave the corresponding labeled metabolite in one step. Copyright © 2008 John Wiley & Sons, Ltd.  相似文献   

13.
The ratio of metabolite to parent drug concentration (Cm/Cp)of a medium or high extraction ratio (E>0.1)drug administered intravenously has been shown to depend on intrinsic clearance of drug by other metabolic routes (CLr,int)as well as on organ blood flow (Q).In contrast, for a low extraction ratio drug given intravenously or for any drug given by a portal route, this ratio is equal to the ratio of formation clearance (CLf,int)and metabolite clearance (CLm,int).The sensitivity of Cm/Cpto changes in CLr,int and CLf,int has been analyzed quantitatively. It was shown to be dependent on the fraction metabolized to that particular metabolite (fm).Supported in part by National Institutes of Health National Research Service Awards GM07348 and GM-07750 from the National Institute of General Medical Sciences and NINCDS Research Grant NS-04053 and NINCDS Research Contract NO l-NS-1-2282.  相似文献   

14.
The importance of metabolites as active and toxic entities in drug therapy evokes the need for an examination of metabolite kinetics after drug administration. In the present review, emphasis is placed on single-compartmental characteristics for a drug and its primary metabolites under linear kinetic conditions. The determination of the first-order elimination rate constants for drug and metabolite are also detailed. For any ithprimary metabolite miformed solely in liver, kinetic parameters with respect to primary metabolite formation under first-order conditions require a comparison of the areas under the metabolite concentration-time curve after drug and preformed metabolite administrations. These area ratios hold regardless of the number of noneliminating compartments for the drug and metabolite. These parameters include fmi and gmi,the fractions of total body clearance that respectively furnishes mito the general circulation and forms mi,and hmi,the fraction of hepatic clearance responsible for the formation of mi.Moreover, the fraction of dose dmi converted to form miis defined with respect to the route of drug administration. The inherent assumption of these estimates, however, requires that the extent of sequential elimination of the generated mibe identical to the extent of metabolism of preformed mi.Discrepancies have been found, and may be attributed mostly to the uneven distribution of drug-metabolizing activities as well as to the presence of diffusional barriers. Other linear systems that involve miformation from multiple organs are briefly described.  相似文献   

15.
The plasma clearance and metabolic rate characteristics of valproic acid (VPA) were studied using guinea-pigs placed on various (0.08-9 μmol ml?1 = 11–1303 μg ml?1) steady-state plasma concentrations (Css) by constant intravenous (i.v.) infusion. The total clearance (CL) was significantly decreased at plasma concentration of 0.61 μmol ml?1 (88 μg ml?1). The metabolic clearance of VPA was apparently biphasic. The maximum metabolic rate (Vmax) and the Michaelis-Menten constant (Km) for the primary (Vmaxl, Kml) and the secondary (Vmax2, Km2) pathways were Vmaxl = 1.52 μmol min ?1kg?1, Kml = 0.15 μmol ml?1, Vmax2 = 24.98 μmol min ?1 kg?1 and Km2 = 11.70 μmol ml?1, respectively. The Kml value was within clinical therapeutic concentration range. The formation of conjugated VPA (cjVPA) metabolite in liver was shown to be saturable. Plasma protein binding of VPA was also nonlinear. The dose-dependent decrease in metabolic clearance was counterbalanced by the increased unbound fraction (fu), resulting in a relatively constant apparent clearance of VPA over a wide concentration range. The hepatic concentration of VPA was not significantly different from the plasma unbound concentration, again over a wide concentration range. The biliary and hepatic concentrations of VPA were not significantly different; but the concentration ratio of cjVPA in bile compared with that of VPA in liver decreased against hepatic concentration of VPA, which suggests a saturable conjugation rate. The Km value estimated from hepatic cjVPA production as a function of plasma VPA concentration was comparable with the Kml value. These results implied that the primary metabolic parameters may describe the conjugation pathway which is nonlinear within the clinical therapeutic concentration range.  相似文献   

16.
Abstract

1.?The human mass balance of 14C-labelled ASP015K ([14C]ASP015K), an orally bioavailable Janus kinase (JAK) inhibitor, was characterized in six healthy male subjects after a single oral dose of [14C]ASP015K (100?mg, 3.7?MBq) in solution. [14C]ASP015K was rapidly absorbed with tmax of 1.6 and 1.8?h for ASP015K and total radioactivity in plasma, respectively. Mean recovery in urine and feces amounted to 36.8% and 56.6% of the administered dose, respectively. The main components of radioactivity in plasma and urine were ASP015K and M2 (5′-O-sulfo ASP015K). In feces, ASP015K and M4 (7-N-methyl ASP015K) were the main components.

2.?In vitro study of ASP015K metabolism showed that the major isozyme contributing to the formation of M2 was human sulfotransferase (SULT) 2A1 and of M4 was nicotinamide N-methyltransferase (NNMT).

3.?The in vitro intrinsic clearance (CLint_in?vitro) of M4 formation from ASP015K in human liver cytosol (HLC) was 11-fold higher than that of M2. The competitive inhibitory effect of nicotinamide on M4 formation in the human liver was considered the reason for high CLint_in vitro of M4 formation, while each metabolic pathway made a near equal contribution to the in vivo elimination of ASP015K. ASP015K was cleared by multiple mechanisms.  相似文献   

17.
Summary It is generally assumed that the renal clearance of drugs in patients with renal impairment are affected to a similar extent regardless of the type of renal disease (intact nephron hypothesis). We have studied the effect of underlying renal disease on the pharmacokinetics of cefotaxime and desacetylcefotaxime in two groups of children (ages 7 to 16 y) with varying degrees of renal dysfunction.Patients in group 1 (n=5) had intrinsic renal disease and those in group 2 (n=5) had extrinsic renal disease, as identified by the primary renal lesion. After a single intravenous dose of cefotaxime timed blood and urine samples were collected for 24 h; cefotaxime and desacetylcefotaxime were measured by HPLC.There were no significant differences between the groups in age, body surface area, urine output, creatinine clearance, total body clearance, nonrenal clearance, renal clearance, and volume of distribution at steady state of cefotaxime, and renal clearance of desacetylcefotaxime. However, the renal clearance: creatinine clearance (CLR:CLCR) ratios for both cefotaxime [1.34 in group 1 vs. 0.51 in group 2] and desacetylcefotaxime [1.58 in group 1 vs. 0.75 in group 2] were statistically significant between the two groups. Group 1 patients had an average CLR:CLCR ratio greater than 1 for both the parent compound and the metabolite, suggesting that net tubular secretion was still intact, despite a diminished glomerular filtration rate (CLCR=24 ml·min–1·1.73 m–2). In contrast, patients in group 2 (CLCR=49) ml·min–1·1.73 m–2) had an average CLR:CLCR ratio less than 1 for both cefotaxime and desacetylcefotaxime, suggesting that renal tubular transport mechanisms did not remain functional in these patients.Our findings suggest that the effect of renal insufficiency on the renal elimination of cefotaxime and its metabolite desacetylcefotaxime may depend on the cause of renal insufficiency.  相似文献   

18.
Aims The aim of this study was to determine the potential influence of renal impairment on the pharmacokinetics of temocapril and its pharmacologically active diacid metabolite, temocaprilat. Methods Non-compartmental pharmacokinetics were assessed in four groups of hypertensive patients (n=8 per group, four investigational centres) with normal (creatinine clearance determined via 24 h urine sampling, CLCR, ≥60 ml min−1 ) and impaired renal function (CLCR 40–59, 20–39, <20 ml min−1 ) after 14 once daily oral doses of 10 mg temocapril hydrochloride. Results For temocapril, there were no statistically significant differences in median tmax or mean Cmax, AUCSS, t½,Z, CL/F between the four groups. Renal clearance, CLR, for temocapril showed a linear decreasing trend with decreasing CLCR [ mean (s.d.): 32.2 (10.7) to 3.7 (3.0) ml min−1]. Steady-state for temocaprilat was reached on day 5. For temocaprilat, no statistically significant differences in mean Cmax or median tmax were detected. With decreasing mean CLCR, mean AUCSS for temocaprilat increased statistically significantly although only 2.4-fold [mean (s.d.): 2115 (565) to 4989 (2338) ng ml−1 h] and t½,Z was prolonged [mean (s.d.): 15.2 (1.2) to 20.0 (7.5) h]. CLR for temocaprilat showed a linear decreasing trend with decreasing CLCR [mean (s.d.): 20.2 (4.3) to 3.0 (1.8) ml min−1]. Conclusions These results indicate that impaired renal function has only a limited effect on the pharmacokinetics of temocapril and its active metabolite, temocaprilat. This may be attributed to the dual, i.e. renal and biliary, elimination pathway of the drug.  相似文献   

19.
(S)‐2‐[(R)‐7‐(3,5‐Dichlorophenyl)‐5‐methyl‐6‐oxo‐5‐(4‐trifluoromethoxybenzyl)‐6,7‐dihydro‐5H‐imidazo[1,2‐a]imidazole‐3‐sulfonylamino]‐proprionamide (1), a potent lymphocyte function‐associated antigen‐1 antagonist and its sulfonamide metabolite (2) labeled with stable isotopes and carbon‐14 were prepared for Drug Metabolism and PharmacoKinetics and other studies. A long linear route was used to prepare [13C2, 2H3]‐(1) using [3,3,3‐2H]‐D‐alanine and [13C2]‐glycine in 15 steps and 2.5% overall yield. With the availability of [13C6]‐3,5‐dichloroaniline, the sulfonamide [13C6]‐(2) was prepared in 12 steps and in 5.6% overall yield. For the carbon‐14 synthesis, a six‐step synthesis gave both compounds [14C]‐(1) and [14C]‐(2) from the common sulfonyl chloride intermediate [14C]‐(15) in 18% and 4% radiochemical yields and specific activities of 44 and 40.5 mCi/mmol, respectively. Copyright © 2011 John Wiley & Sons, Ltd.  相似文献   

20.
Naproxen, a well‐known non‐steroidal anti‐inflammatory drug, and its 6‐O‐desmethylated metabolite have been labelled with 2H. (R,S)‐Naproxen 7 labelled with 2H was obtained in five steps using the commercially available [2H3]iodomethane 5 as the stable labelled reagent. The demethylation of 7 using 48% HBr in 1‐butyl‐3‐methylimidazolium tetrafluoborate gave the corresponding 2H‐labelled 6‐O‐desmethylated metabolite 8. Copyright © 2008 John Wiley & Sons, Ltd.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号