European Review for Medical and Pharmacological Sciences 2012; 16: 2162-2170 The effect of on the pharmacokinetics of enalaprilat in nephrotic rats

B.-T. BI, Y.-B. YANG*, A.-D. MA**, T. LIN***, H.-B. LIN, X.-M. YANG**, J.-P. XU

Department of Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China *Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Southern Medical Universi- ty, Guangzhou, China **Hygiene Detection Center of Southern Medical University, Guangzhou, China ***Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China

Abstract. – BACKGROUND AND OBJECTIVES: Introduction The adverse reactions in combination of an- giotensin-converting enzyme inhibitors (ACEIs) The - system (RAS) plays an and Ang II receptor blockers (ARBs) were severer than that in monotherapy for patients with important role in the progress of renal disease. nephropathy. The effect of candesartan on phar- The principal bioactive peptide in RAS is an- macokinetics of enalaprilat in nephrotic rats was giotensin II (Ang II), which may contribute to investigated to make references for the clinical vascular endothelial dysfunction, atherosclerosis, therapy in patients with nephropathy to avoid relat- remodeling and hypertrophy of renal tissue, and ed adverse effects. cell death. These changes represent a pathophysi- MATERIALS AND METHODS: Nephrotic rats 1,2 were prepared by adriamycin injection. Control ologic basis of renal diseases . group and one nephrotic group received Evidences have pointed out that blockade of alone, another nephrotic group received RAS can delay the progress of renal diseases3,4. The enalapril and candesartan simultaneously. Blood general RAS blockers in clinical application in- samples were drawn at time points after a single clude angiotensin-converting enzyme inhibitors oral administration. The concentration of enalaprilat was determined using LC-MS/MS. (ACEIs) and Ang II receptor blockers (ARBs). RESULTS: Compared with control group and ACEIs are able to reduce Ang II production by in- nephrotic group received enalapril alone respec- hibiting angiotensin-converting enzyme (ACE), and tively, Tmax of enalaprilat in nephrotic group re- ARBs can bind the specific receptor of Ang II, ar- ceived both enalapril and candesartan cilexetil resting its biological effects5. Because of the emerg- prolonged about 21.43% and 6.224%, respectively; ing “angiotensin escape phenomenon” in the use of AUC increased by 185.3% and 60.63%, respec- (0-t) ACEIs alone4, the combination of ACEIs and tively; Cmax increased by 219.4% and 56.64%, re- spectively; t1/2 incerased by 163.7% and 30.05%, ARBs has been proposed for clinical application. respectively; CL/F reduced by 65.12% and 40.78%, Studies found that the combination of ACEIs and respectively. There were no significant differences ARBs was more effective than the single entity of the V1/F of enalaprilat between three groups. alone in depressing systemic blood pressure and The CL/F and t1/2 of enalaprilat showed significant 6,7 correlations with serum creatinine (Scr) respec- treating albuminuria in diabetic nephropathy . The tively (r = –0.7502; r = 0.5626). joint use of them would lead to a dual RAS block- DISCUSSION: The combination with candesar- ade and a greater clinical benefit theoretically3,5-7. tan in nephrotic rats significantly changed the However, clinical observations revealed that ad- pharmacokinetics of enalaprilat, showing in- verse reactions in the combined therapy were sever- creased accumulation and decreased elimination. er than that in monotherapy, including hypotension, In view of these findings, we should lower dosage and prolong dosing interval for nephrotic patients hyperkalemia, and renal function deterioration, 8-11 in the combination of enalapril and candesartan. even if consciously reduced the dosages . Phar- macodynamic studies indicated that these adverse Key Words: reactions were related to the excessive inhibition of Angiotensin II, Candesartan, Enalaprilat, Nephrosis, Pharmacokinetics. RAS. However, the pharmacokinetic changes in the combined use of both drugs remain unclear.

Corresponding Authors: Jiang-Ping Xu, MD, Ph.D.; e-mail: [email protected]; and 2162 Ying-Bao Yang, MD, Ph.D.; e-mail: [email protected] The effect of candesartan on the pharmacokinetics of enalaprilat in nephrotic rats

Enalapril is a widely-used ACEI. Its active tology, the normal rats and nephrotic rats were metabolite enalaprilat is mainly excreted by the treated as the following: control group (n=6) and kidneys. Studies found elevated concentrations of one model group (n=6) received enalapril (15 enalaprilat in nephrotic patients with convention- mg/kg, p.o.) alone, and another model group al dosages12. Candesartan is a commonly used (n=6) received simultaneously enalapril (15 ARB. It is cleared via kidneys, biliary and in- mg/kg, p.o.) and candesartan cilexetil (3 mg/kg, testinal route. Whether the physiological disposi- p.o.). Blood samples were drawn under ether tion of enalaprilat and candesartan would be anesthesia from orbital venous plexus before ad- changed in their combined use for nephrotic pa- ministration and 1, 2, 3, 4, 6, 8, 12, 24, and 48 h tients is unknown. Compared with ARBs, ACEIs after single dosing. Blood samples were collect- have been used more generally, but can result in ed using heparinized tubes, centrifuged for 20 more adverse effects13. Accordingly, in this study min at 450 × g at 4°C. Plasma samples were we chose enalaprilat as the monitoring object, stored at -20°C before analysis. and investigate the effect of candesartan on phar- macokinetics of enalaprilat in nephrotic rats to Determination of Serum Creatinine make references to the clinical application for pa- Before injection with ADR or saline and 7 tients with nephropathy for avoiding related ad- weeks after injection, blood and serum collection verse effects. was carried out. Avoiding hemolysis during the collection process, the Scr was measured using corresponding kit with enzymatic method. Material and Methods Sample Preparation Chemicals The preparation of samples was carried out as Adriamycin was purchased from Main Luck described17 with adjusted. A 200 µL of plasma Pharmaceuticals Inc, ShenZhen, China. Kit for sample was mixed with 20 µL internal standard measuring creatinine was purchased from Shang- (3 µg/ml ) and 10 µL formic acid. The hai Mind Bioengineering Co. Ltd., Shanghai, mixture was vortexed with 370 µL methanol by China. Enalapril and candesartan cilexetil were oscillator followed by centrifuging for 15 min at purchased from YiBang Pharmaceutical Technol- 10,000 × g at 4°C. The supernatant was collected ogy Development Co. Ltd., GuangZhou, China. and filtered. 3-µL aliquot of supernatant was in- Enalaprilat was purchased from USPC Inc., jected to LC-MS/MS system for analysis. Rockville, MD, USA. Benazepril was supplied by the National Institute for the Control of Phar- LC-MS/MS maceutical and Biological Products. All chemi- LC-MS/MS was carried out as described18 cals were at least analytical grade. with adjusted. The mobile phase composed of methanol-water-formic acid (70:30:0.03, v/v/v) Animals and Treatment at a flow rate of 0.3 mL/min. An API 4000 Q Male and female Sprague-Dawley rats TRAP LC-MS/MS system (Applied Biosystems, (250~280 g) in this study were obtained from the Foster City, CA, USA) was used for analyses. Experimental Animal Center, Southern Medical The mass spectrometer was operated in positive University, Guangzhou, China. The animals were ion detection mode. Nitrogen was used as the ion housed in at 25 ± 2°C with water and standard source gas at 55 psi, the collision gas at medium, diet accessible ad libitum. All procedures were and the curtain gas at 25 psi. The ion spray volt- performed in accordance with the NIH Guide for age was adjusted to 5,500 V and the temperature the Care and Use of Laboratory Animals. After of vaporizer was at 350°C. The declustering po- one week of acclimatization, the rats were divid- tentials were set at 63 V and 100 V for enalapri- ed into control group (n=8) and model group lat and benazepril, respectively. (n=18). As adriamycin (ADR) has been reported to be able to induce renal impairment14-16, rats in Pharmacokinetic Analysis model group received injection of ADR (3 Non-compartmental pharmacokinetic parame- mg/kg, once every two days, thrice in total) ters were calculated from the plasma concentra- through the tail vein. Control rats received injec- tions of enalaprilat in each time point of each rat tion of saline (2 mL/kg). After confirming the using DAS 2.0 Program registrated from Mathe- nephropathy rat model in biochemistry and his- matical Pharmacology Professional Committee

2163 B.-T. Bi, Y.-B. Yang, A.-D. Ma, T. Lin, H.-B. Lin, X.-M. Yang, J.-P. Xu

Table I. Precision, recovery and accuracy of enalaprilat (n = 3 days, five replicates per day).

Concentration (ng/ml) R.S.D. (%) R.E. Extraction recovery Added Found Intra-day Inter-day (%) (%)

50 59.63 ± 3.05 11.44 2.03 19.26 85.80 500 599.9 ± 12.23 5.00 1.12 19.98 84.26 5000 5277 ± 65.74 2.79 0.80 5.54 74.53 of China. This program has been applicated in capsule, and nephric tubule dilatation in compari- other pharmacokinetic studies19-21. The non-com- son to the rats in control group (Figure 2). These partmental pharmacokinetic parameters includ- biochemical and histological changes indicated ing area under the concentration-time curve from that ADR administration successfully induced a zero to last quantifiable time (AUC(0-t)), half-life nephropathy model. of elimination (t1/2), total clearance (CL/F), vol- ume of distribution (V1/F) were calculated using Enalaprilat Detection and statistical moments22-24. The maximum plasma Method Validation concentration (Cmax) and the time to maximum The detection of enalaprilat was carried out concentration (Tmax) were measured according to by LC-MS/MS. Figure 3 shows the mass spec- the actual levels of enalaprilat. trums of enalaprilat and the internal standard be- nazepril of rat plasma. Quantification was per- Statistical Analysis formed using multiple reaction monitoring of the Statistical analyses were performed using transitions of m/z 349.2 → m/z 206.3 for SPSS for Windows 13.0 (SPSS Inc., Chicago, enalaprilat and m/z 425.4 → m/z 351.3 for be- IL, USA). Values were expressed as mean ± nazepril. The peak area ratios of enalaprilat to SEM. The comparison of Scr between control benazepril varied linearly over the concentration 2 group and model group was performed by un- range tested. Equation for calibration curve (1/χ paired t-test. One-way Analysis of Variance fol- weighting) was y = 0.000675 × + 0.0145 (r = lowed by LSD multiple comparison or Dunnett’s 0.9978) for the extent of 50~5000 ng/mL. The multiple comparison tests was performed for the lower limit of quantification was 0.785 ng/mL. comparison of pharmacokinetic parameters. As shown in Table I, the intra-day and inter-day Correlation analysis was used to investigate the precisions at concentrations of 50, 500, and relationships of factors. Significant difference was set at p < 0.05.

Results

Confirmation of Nephropathy Rat Model To confirm the nephropathy rat model with ADR induction, we determined Scr and exam- ined nephrons under optical microscopy. Before ADR administration, it was no difference of the Scr level between the two groups. 7 weeks after ADR administration, the concentration of Scr in- creased by 25.69% (p < 0.05). When compared to control group in which rats received saline in- Figure 1. Concentrations of serum creatinine before and 7 jection, Scr increased by 41.24% (p < 0.05) (Fig- weeks after injection of ADR. Rats in control group received ure 1). The kidneys of rats were examined for saline injection (n=8, 2 mL/kg) and in model group received ADR injection (n=18, 3 mg/kg) once every two days, thrice their histological changes with Hematoxylin- totally through the tail vein. Serum creatinine was deter- Eosin staining. 7 weeks after ADR administra- mined by commercial kit. Values are expressed as mean ± tion, the kidneys of model group showed SE. *p < 0.05 vs. model group before injection, #p < 0.05 vs. glomerular distention, glomerular syncretism to control group after injection.

2164 The effect of candesartan on the pharmacokinetics of enalaprilat in nephrotic rats

Figure 2. The histology of kidneys in rats received saline or ADR. Compared to control group (A) and (C), ADR administra- tion (B) and (D) induced glomerulus distention, glomerular syncretism to capsule, tubule dilatation, tubule epithelial cell vac- uolization, and nephric tubule cast. Magnification: (A) 60×; (B) 60×; (C) 150×; (D) 150×.

A B

Figure 3. Mass spectra of enalaprilat and internal standard benazepril in rat plasma. The spectra showed multiply charged ions of enalaprilat (A) at m/z 206.3, 303.3, 349.2 and benazepril (B) at m/z 190.3, 351.3, 425.4. Quantification was performed using multiple reaction monitoring of the transitions of m/z 349.2 → m/z 206.3 for enalaprilat and m/z 425.4 → m/z 351.3 for benazepril.

2165 B.-T. Bi, Y.-B. Yang, A.-D. Ma, T. Lin, H.-B. Lin, X.-M. Yang, J.-P. Xu

5,000 ng/mL expressed in terms of relative stan- dard deviation (R.S.D.) were within 15%; the relative error (R.E.) of accuracies were within 20% (n=3 days, five replicates/d). The extraction recoveries at concentrations of 50, 500, and 5,000 ng/mL were 85.80%, 84.26%, and 74.53%, respectively.

Pharmacokinetics of Enalaprilat in Nephrotic Rats Concentration-time curves of enalaprilat were shown in Figure 4, and corresponding non-com- partmental pharmacokinetic parameters were list- ed in Table II. As revealed in Figure 4, the con- centrations of enalaprilat increased rapidly after a single oral administration. The non-compartmen- Figure 4. Mean plasma concentrations of enalaprilat over tal pharmacokinetic analysis shown that com- time after single dosing. Control rats and one group of pared with the rats in control group, the time to nephrotic rats received enalapril (ENA) (15 mg/kg, p.o.) maximum concentration (Tmax) of enalaprilat in alone, another group of nephrotic rats received enalapril nephrotic rats received enalapril alone prolonged (ENA) (15 mg/kg, p.o.) and candesartan cilexetil (CAN) (3 about 14.32% (p = 0.251). The area under con- mg/kg, p.o.) simultaneously. Values are expressed as mean ± SE (n = 6/group). centration-time curve (AUC(0-t)) increased by 77.62% (p = 0.006), and the maximum plasma concentration (Cmax) increased by 103.9% (p = 0.031). Moreover, the elimination half-life (t1/2) pharmacokinetic parameters were listed in Table in nephrotic rats prolonged by 102.8% (p = II. As shown in Table II, although it was not sig- 0.024), and enalaprilat clearance (CL/F) reduced nificant differences for all of pharmacokinetic by 41.10% (p = 0.003). parameters between the nephrotic rats with single or combined administration, we found that the Effect of Candesartan on enalaprilat accumulation and its slower elimina- the Pharmacoinetics of tion were obvious in the combination group. In Enalaprilat in Nephrotic Rats contrast to control group and nephrotic group re- The concentration-time curve of enalaprilat in ceived enalapril alone, the time to maximum con- nephrotic rats received the combination of centration (Tmax) in nephrotic rats received the enalapril and candesartan cilexetil was shown in combination of enalapril and candesartan cilex- Figure 4, and corresponding non-compartmental etil prolonged about 21.43% (p = 0.093) and

Table II. Non-compartmental pharmacokinetic parameters of enalaprilat in control and nephrotic rats with or without com- bined use of candesartan cilexetil.

Group Control Nephrosis Nephrosis (Enalapril) (+) (+) (+) (Candesartan) (–) (–) (+)

a AUC(0-t) (ng/ml·h) 7458 ± 329.4 13247 ± 1683* 21278 ± 1388*† b Cmax (ng/ml) 1329 ± 331.4 2710± 342.5* 4245 ± 530.4*† c Tmax (h) 2.333 ± 0.2108 2.667 ± 0.2108 2.833 ± 0.1667 d t1/2 (h) 4.501 ± 0.3835 9.127 ± 1.726* 11.87 ± 1.680* CL/F (L/h/kg)e 2.029 ± 0.08942 1.195 ± 0.1412* 0.7077 ± 0.05499*† V1/F (L/kg)f 13.20 ± 1.281 15.74 ± 3.485 16.01 ± 1.718 a b AUC(0-t): area under the concentration-time curve from zero to last quantifiable time; Cmax: maximum plasma concentration; c d f Tmax: time to maximum concentration; t1/2: half-life of elimination; e CL/F: total clearance; V1/F: volume of distribution of enalaprilat. Values are expressed as mean ± SE (n = 6/guoup); *p < 0.05 vs. control group, †p < 0.05 vs. model group without the combined use of candesartan cilexetil.

2166 The effect of candesartan on the pharmacokinetics of enalaprilat in nephrotic rats

8-11 6.224% (p = 0.559), respectively; AUC(0-t) in- (GFR) . Therefore, the present study need to creased by 185.3% (p = 0.001) and 60.63% (p = establish a nephropathy model in rats to aim di-

0.001), respecitively; and Cmax increased by rectly at simulating this clinical phenomena. 219.4% (p = 0.001) and 56.64% (p = 0.019), re- ADR has been reported to be able to induce renal 14-16 spectively; t1/2 incerased by 163.7% (p = 0.003) impairment . The early pathological changes and 30.05% (p = 0.198), respectively; CL/F re- induced by ADR are mainly podocyte injury and duced by 65.12% (p = 0.001) and 40.78% (p = tubulointerstitial lesions with the concomitant 0.044), respectively. There were no significant symptom of proteinuria and electrolyte distur- differences of the volume of distribution (V1/F) bances, but it is not reflect the abnormal GFR in of enalaprilat between three groups in the study this stage14-16. GFR, a measurement of renal func- (p > 0.05). tion, will decrease after the further progression of renal impairment. Measuring Scr is a simple test The Relationship Between the Scr and and it is the most commonly used indicator of 25-27 the Cl/F and T1/2 of Enalaprilat GFR . A rise of Scr is observed only with To investigate the impact of renal function on marked damage to functioning nephrons. There- the CL/F of enalaprilat, we analyzed the relation- fore, this study carried out Scr test which is rela- ship between the Scr and the CL/F of enalaprilat. tive suitable for detecting dysfunction-stage of As shown in Figure 5(A), the enalaprilat CL/F kidney disease. Meanwhile, histological observa- showed an obvious negative correlation with Scr tion revealed that ADR induced glomerular dis- (r = –0.7502, p = 0.0003). Meanwhile, the rela- tention, glomerular syncretism to capsule, and tionship between the Scr and the t1/2 of enalapri- nephric tubule dilatation. The biochemical and lat was analyzed. As shown in Figure 5(B), the histological changes observed in the present enalaprilat t1/2 showed a positive correlation with study indicated that ADR administration success- Scr (r = 0.5626, p = 0.0151). fully induced a nephropathy model. Several studies on diabetic and non-diabetic nephropathy demonstrated the beneficial effects of ACEIs with a number of adverse reactions, in- Discussion cluding hypotension and deterioration of renal function at low or moderate doses28,29. These phe- Clinical observations revealed that the adverse nomenon have raised interest in studying the re- reactions from the combination of ACEIs and lationship between adverse effects and in vivo ac- ARBs usually happen to the patients with mild to cumulation of ACEIs. It has been reported that severe decrease in glomerular filtration rate patients with chronic renal failure given small or

A B

Figure 5. Correlation of serum creatinine levels with enalaprilat clearance and half-life of elimination. (A) Enalaprilat clear- ance was negatively correlated with serum creatinine levels (r = –0.7502, p = 0.0003). (B) The elimination half-life of enalapri- lat was positively correlated with serum creatinine levels (r = 0.5626, p = 0.0151). Each point represents one experimental rat.

2167 B.-T. Bi, Y.-B. Yang, A.-D. Ma, T. Lin, H.-B. Lin, X.-M. Yang, J.-P. Xu moderately high doses of enalapril may have sulted from the two drug competition for markedly elevated enalaprilat in vivo30. The phar- tubular excretion in the impaireds kidney. Given macokinetic analytic results in the present study that its metabolite enalaprilat is more difficult to also showed an significantly increased concentra- be excreted, the dosage of enalapril for nephrotic tion of enalaprilat and decreased clearance of patients combined with candesartan cilexetil enalaprilat in nephrotic rats with a single oral should be further reduced. Based on the findings dose of enalapril, and the CL/F and t1/2 of of this study that AUC(0-t) and Cmax increased by enalaprilat was significantly correlated with Scr 60.63% and 56.64% respectively, we recommend which reflects the deteriorated degree of renal at least for the nephrotic patients that the reduc- function. Given previous study observed that the tion of the dosage of enalapril should be no less decline of renal function in high-concentration than 50%, when combining with candesartan enalaprilat group was faster than that in low-con- cilexetil in conventional dosage. Further studies centration group during a 12-month medication are needed to verify the dosage adjustment of of enalapril for renal failure patients12, the above candesartan cilexetil for the nephrotic patients findings suggest that the dosage of enalapril in when combining with enalapril. clinical application for patients with nephrosis In fact, the dosage of ACEIs have often been re- should be carefully adjusted according to their duced in the therapy for nephrotic patients, but renal function because of the easier accumulation this approach is still difficult to prevent the ad- of enalaprilat which might induce the deteriora- verse reactions, especially hypotension and renal tion of renal function. function deterioration10. In this study, we observed

The pharmacodynamics of ACEIs and ARBs that the t1/2 of enalaprilat in nephrotic rats received have been extensively studied in patients with enalapril alone prolonged by 102.8% compared to nephropathy3,8-11,13, but there is a paucity of data control group, suggesting that the dosing interval on the pharmacokinetic interactions of these two of enalapril for nephrotic patients should be pro- types of drugs. In this study, we found no signifi- longed based on the individual t1/2 of enalaprilat. cant differences of the V1/F of enalaprilat be- Actually, monitoring plasma drug concentration is tween three groups in the study, indicating no an effective strategy to establish the individual t1/2 differences of the free fraction of enalaprilat in to obtain the optimal dosing interval, but it is diffi- plasma between the groups. However, it was con- cult to be carried out extensively in clinical prac- tradicted that the Tmax of enalaprilat did not sig- tice. Interestingly, we found an significant correla- nificantly change in three groups of this study, tion between t1/2 of enalaprilat and Scr, suggesting given that Tmax should rather be increased with that Scr of nephrotic patients should be considered decreased clearance and unchanged absorption in in the establishment of dosing interval to prevent pharmacokinetic theory. There is no obvious ex- adverse reactions from enalapril administration. planation for this finding at present. Notably, the Notably, compared to nephrotic group with

AUC(0-t) and Cmax of plasma enalaprilat increased enalapril alone, we found that the t1/2 of enalaprilat and the CL/F decreased when used together with in nephrotic rats received enalapril and candesar- enalapril and candesartan cilexetil, compared to tan prolonged by 30.05%. This exact t1/2 of administration of enalapril alone in nephrotic enalaprilat in the combination of enalapril and rats, suggesting that combining with candesartan candesartan, which has never been reported, is a cilexetil aggravated the accumulation of enalapri- valuable reference for the enalapril-candesartan lat, which probably related to the lower total combination regimen in patients with renal dis- clearance of enalaprilat. Similar to enalaprilat, ease, indicating the necessity of prolonging dosing candesartan in the systemic circulation is mainly interval. Further studies are needed to quantify the cleared by kidney. It was reported that patients relationships between t1/2 of enalaprilat, Scr and with decreased creatinine clearance would have a enalapril-candesartan combination regimen. decreased clearance of candesartan31. The results In conclusion, we found in the present study in recent studies supported the important role of that the pharmacokinetic changes of enalaprilat tubular transporters in the renal excretion of depended on the renal function and drug regi- many drugs, for example, organic anion trans- mens, especially combined with candesartan porters display a role in enalaprilat and candesar- cilexetil administration. In view of these find- tan excretion32. In this study, the nephric tubule ings, we should lower dosage and prolong dosing impairment of nephropathy rat model is obvious, interval for nephrotic patients in the combination thus the accumulation of enalaprilat may be re- of enalapril and candesartan cilexetil.

2168 The effect of candesartan on the pharmacokinetics of enalaprilat in nephrotic rats

–––––––––––––––––––– Acknowledgements 12) E LUNG -J ENSEN T,H EISTERBERG J,S ONNE J, STRANDGAARD S,KAMPER AL. Enalapril dosage in This work was supported by research grants from progressive chronic nephropathy: a randomised, Guangdong Hospital Pharmacy Foundation [grant controlled trial. Eur J Clin Pharmacol 2005; 61: number 2008A015]. 87-96. 13) TANG SC, LEUNG JC, CHAN LY, EDDY AA, LAI KN. An- giotensin converting enzyme inhibitor but not an- giotensin receptor blockade or statin ameliorates murine adriamycin nephropathy. Kidney Int 2008; References 73: 288-299. 14) VENKATESAN N,PUNITHAVATHI D,ARUMUGAM V. Cur- 1) NICKENIG G,HARRISON DG. The AT(1)-type an- cumin prevents adriamycin nephrotoxicity in rats. giotensin receptor in oxidative stress and athero- Br J Pharmacol 2000; 129: 231-234. genesis: part I: oxidative stress and atherogene- 15) EL-SHITANY NA, EL-HAGGAR S, EL-DESOKY K. Silymarin sis. Circulation 2002; 105: 393-396. prevents adriamycin-induced cardiotoxicity and 2) NICKENIG G,HARRISON DG. The AT(1)-type an- nephrotoxicity in rats. Food Chem Toxicol 2008; giotensin receptor in oxidative stress and athero- 46: 2422-2428. genesis: Part II: AT(1) receptor regulation. Circu- 16) HE L, RONG X, JIANG JM, LIU PQ, LI Y. Amelioration lation 2002; 105: 530-536. of anti-cancer agent adriamycin-induced nephrot- 3) TAAL MW, BRENNER BM. Renoprotective benefits of ic syndrome in rats by Wulingsan (Gorei-San), a RAS inhibition: from ACEI to angiotensin II antag- blended traditional Chinese herbal medicine. onists. Kidney Int 2000; 57: 1803-1817. Food Chem Toxicol 2008; 46: 1452-1460. 4) WERNER C,BAUMHAKEL M,TEO KK,SCHMIEDER R, 17) LU S, JIANG K, QIN F, LU X, LI F. Simultaneous quan- MANN J, UNGER T, YUSUF S, BOHM M. RAS blockade tification of enalapril and enalaprilat in human with ARB and ACE inhibitors: current perspective plasma by high-performance liquid chromatogra- on rationale and patient selection. Clin Res Cardi- phy-tandem mass spectrometry and its applica- ol 2008; 97: 418-431. tion in a pharmacokinetic study. J Pharm Biomed Anal 2009; 49: 163-167. 5) SONKODI S,MOGYOROSI A. Treatment of diabetic nephropathy with angiotensin II blockers. 18) GU Q,CHEN X,ZHONG D,WANG Y. Simultaneous Nephrol Dial Transplant 2003; 18(Suppl 5): v21- determination of enalapril and enalaprilat in hu- 23. man plasma by liquid chromatography-tandem mass spectrometry. J Chromatogr B Analyt 6) PARVING HH,ANDERSEN S,JACOBSEN P, CHRISTENSEN Technol Biomed Life Sci 2004; 813(1-2): 337- PK, ROSSING K, HOVIND P, ROSSING P, TARNOW L. An- giotensin receptor blockers in diabetic nephropa- 342. thy: renal and cardiovascular end points. Semin 19) XU H, HE L, NIE S, GUAN J, ZHANG X, YANG X, PAN Nephrol 2004; 24: 147-157. W. Optimized preparation of vinpocetine prolipo- somes by a novel method and in vivo evaluation 7) FRIED LF, DUCKWORTH W, ZHANG JH,O'CONNOR T, of its pharmacokinetics in New Zealand rabbits. J BROPHY M,EMANUELE N,HUANG GD,MCCULLOUGH Control Release 2009; 140: 61-68. PA, PALEVSKY PM,SELIGER S,WARREN SR,PEDUZZI P. Design of combination angiotensin receptor 20) YAN Z,ZHU ZL,WANG HQ,LI W, MI YX,LIU CX. blocker and angiotensin-converting enzyme in- Pharmacokinetics of panaxatrol disuccinate sodi- hibitor for treatment of diabetic nephropathy (VA um, a novel anti-cancer drug from Panax notogin- NEPHRON-D). Clin J Am Soc Nephrol 2009; 4: seng, in healthy volunteers and patients with ad- 361-368. vanced solid tumors. Acta Pharmacol Sin 2010; 31: 1515-1522. 8) FERRARI P, MARTI HP, PFISTER M, FREY FJ. Additive an- tiproteinuric effect of combined ACE inhibition 21) LIANG YG, LIU P, GAO HZ, LI HY, HAO GT, WANG XF, and angiotensin II receptor blockade. J Hypertens ZHANG XG,MENG QY, LIU ZY. Pharmacokinetics 2002; 20: 125-130. and safety of a new bioengineered thrombolytic agent, human tissue urokinase type plasminogen 9) KIM HJ,RYU JH,HAN SW, PARK IK,PAIK SS,PARK activator in Chinese healthy volunteers. Eur J MH,PAIK DJ,CHUNG HS,KIM SW, LEE JU. Com- bined therapy of and has no Drug Metab Pharmacokinet 2011; 35: 97-102. additive effects in ameliorating adriamycin-in- 22) YAMAOKA K,NAKAGAWA T, UNO T. Statistical mo- duced glomerulopathy. Nephron Physiol 2004; ments in pharmacokinetics. J Pharmacokinet Bio- 97: 58-65. pharm 1978; 6: 547-558. 10) MANGRUM AJ,BAKRIS GL. Angiotensin-converting 23) PIOTROVSKII VK. The method of statistical moments enzyme inhibitors and angiotensin receptor block- and the integral model-independent parameters ers in chronic renal disease: safety issues. Semin of pharmacokinetics. Farmakol Toksikol 1986; 49: Nephrol 2004; 24: 168-175. 118-127. 11) TAKAICHI K, TAKEMOTO F, UBARA Y, MORI Y. Analysis of 24) DUNNE A. Statistical moments in pharmacokinet- factors causing hyperkalemia. Intern Med 2007; ics: models and assumptions. J Pharm Pharma- 46: 823-829. col 1993; 45: 871-875.

2169 B.-T. Bi, Y.-B. Yang, A.-D. Ma, T. Lin, H.-B. Lin, X.-M. Yang, J.-P. Xu

25) MOGENSEN CE,HEILSKOV NS. Prediction of GFR 29) GISENGROUP (GRUPPO ITALIANO DI STUDI EPIDEMIO- from serum creatinine. Acta Endocrinol Suppl LOGICI IN NEFROLOGIA). Randomised placebo-con- (Copenh) 1980; 238: 109. trolled trial of effect of on decline in 26) LEWIS J, GREENE T, APPEL L, CONTRERAS G, DOUGLAS J, glomerular filtration rate and risk of terminal LASH J, TOTO R, VAN LENTE F, WANG X, WRIGHT JT.A renal failure in proteinuric, non-diabetic comparison of iothalamate-GFR and serum crea- nephropathy. Lancet 1997; 349(9069): 1857- tinine-based outcomes: acceleration in the rate of 1863. GFR decline in the African American Study of 30) ELUNG-JENSEN T, HEISTERBERG J, KAMPER AL, SONNE J, Kidney Disease and Hypertension. J Am Soc STRANDGAARD S, LARSEN NE. High serum enalaprilat Nephrol 2004; 15: 3175-3183. in chronic renal failure. J Renin Angiotensin Al- 27) JAFAR TH, SCHMID CH, LEVEY AS. Serum creatinine dosterone Syst 2001; 2: 240-245. as marker of kidney function in South Asians: a 31) GLEITER CH AND MORIKE KE. Clinical pharmacokinet- study of reduced GFR in adults in Pakistan. J Am ics of candesartan. Clin Pharmacokinet 2002; 41: Soc Nephrol 2005; 16: 1413-1419. 7-17. 28) MASCHIO G, ALBERTI D, JANIN G, LOCATELLI F, MANN JF, 32) YAMASHITA F, OHTANI H, KOYABU N, USHIGOME F, SATOH MOTOLESE M, PONTICELLI C, RITZ E, ZUCCHELLI P. Effect H, MURAKAMI H, UCHIUMI T, NAKAMURA T, KUWANO M, of the angiotensin-converting-enzyme inhibitor TSUJIMOTO M,SAWADA Y. Inhibitory effects of an- benazepril on the progression of chronic renal in- giotensin II receptor antagonists and leukotriene sufficiency. The Angiotensin-Converting-Enzyme receptor antagonists on the transport of human Inhibition in Progressive Renal Insufficiency organic anion transporter 4. J Pharm Pharmacol Study Group. N Engl J Med 1996; 334: 939-945. 2006; 58: 1499-1505.

2170