Abstract
AIM
To investigate the effect of quercetin on organic anion transporting polypeptide 1B1 (OATP1B1) activities in vitro and on the pharmacokinetics of pravastatin, a typical substrate for OATP1B1 in healthy Chinese-Han male subjects.
METHODS
Using human embryonic kidney 293 (HEK293) cells stably expressing OATP1B1, we observed the effect of quercetin on OATP1B1-mediated uptake of estrone-3-sulphate (E3S) and pravastatin. The influence of quercetin on the pharmacokinetics of pravastatin was measured in 16 healthy Chinese-Han male volunteers receiving a single dose of pravastatin (40 mg orally) after co-administration of placebo or 500 mg quercetin capsules (once daily orally for 14 days).
RESULTS
Quercetin competitively inhibited OATP1B1-mediated E3S uptake with a Ki value of 17.9 ± 4.6 µm and also inhibited OATP1B1-mediated pravastatin uptake in a concentration dependent manner (IC50, 15.9 ± 1.4 µm). In healthy Chinese-Han male subjects, quercetin increased the pravastatin area under the plasma concentration – time curve (AUC(0,10 h) and the peak plasma drug concentration (Cmax) to 24% (95% CI 15, 32%, P < 0.001) and 31% (95% CI 20, 42%, P < 0.001), respectively. After administration of quercetin, the elimination half-life (t1/2) of pravastatin was prolonged by 14% (95% CI 4, 24%, P = 0.027), with no change in the time to reach Cmax (tmax). Moreover, quercetin decreased the apparent clearance (CL/F) of pravastatin by 18% (95% CI 75, 89%, P < 0.001).
CONCLUSIONS
These findings suggest that quercetin inhibits the OATP1B1-mediated transport of E3S and pravastatin in vitro and also has a modest inhibitory influence on the pharmacokinetics of pravastatin in healthy Chinese-Han male volunteers. The effects of quercetin on other OATP1B1 substrate drugs deserve further investigation.
Keywords: drug–drug interaction, OATP1B1, pravastatin, quercetin
WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT
OATP1B1 is a liver-specific expression drug transporter and mediates the uptake of a broad range of compounds into hepatocytes. Modulation the activity of OATP1B1 may alter the pharmacokinetics of its substrate drugs, causing potential drug–drug interactions. As a popular dietary supplement in the United States, quercetin has been shown to interact with some drug transporters and efficiently influences their activity, but the effect of quercetin on OATP1B1 activity is not well known.
WHAT THIS STUDY ADDS
Quercetin inhibits the OATP1B1-mediated transport of E3S and pravastatin in vitro, and also has a modest inhibitory influence on the pharmacokinetics of pravastatin in healthy Chinese-Han male volunteers.
Introduction
Quercetin (3,3′,4′,5,7-pentahydroxylflavoine), is a naturally occurring flavonoid ubiquitously present in fruit and vegetables, such as onion, tea, apples and berries. It exhibits anti-oxidative, anti-inflammatory and vasodilating effects and has been proposed to be a potential anti-cancer agent [1], [2]. Recently, quercetin has been marketed in the United States primarily as a dietary supplement [3]. Accumulating evidence has demonstrated that quercetin interacts with drug transporters and efficiently influences their activity, highlighting its potential for natural product – drug interaction. For example, previous studies have suggested that quercetin is a potent inhibitor of P-glycoprotein, multidrug resistance-associated protein 1/2 (MRP1/2), breast cancer resistance protein (BCRP), organic anion transporting polypeptide 1A2 (OATP1A2), OATP2B1 and organic anion transporter 1/3 (OAT1/3), indicating that it may potentially affect in vivo drug disposition by inhibiting different transporter systems [1], [4]–[6].
OATP1B1, encoded by SLCO1B1, is a 691-amino acid glycoprotein with an apparent molecular mass of 84 KDa. OATP1B1 is restrictedly expressed in the basolateral membrane of human hepatocytes and transports a broad range of compounds such as bile acids, conjugated steroids, thyroid hormones, peptides, and drugs including rifampicin and HMG-CoA reductase inhibitors [7]–[9]. Its apparent liver specific expression and capacity of transporting a large number of structurally different compounds suggest that OATP1B1 plays an important role in hepatic drug uptake and elimination. Thus, modulation of OATP1B1 function can alter the pharmacokinetics of OATP1B1 substrate drugs and potentially lead to adverse effects [10].
Pravastatin, a potent inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, is widely used in the treatment of hypercholesterolaemia [11]. As a hydrophilic compound, pravastatin is not significantly metabolized by the cytochrome P450 (CYP450) enzymes. It is taken up by the liver via active transport and mostly excreted unchanged in the bile, with much less excretion in urine [12], [13]. OATP1B1 is thought to be the major transporter involved in the hepatic uptake of pravastatin, and OATP1B1-mediated uptake has been proved to be the rate-limiting step in the hepatic clearance of pravastatin [14]–[16]. Therefore, pravastatin can be used as a probe drug for evaluating the function of OATP1B1 [17].
In this study, we aim to investigate the potential influence of quercetin on the transport activity of OATP1B1 in vitro and assess the impact of quercetin on pravastatin pharmacokinetics in healthy Chinese-Han male volunteers. Considering the frequent consumption of quercetin-containing foods and supplements, the hypothesized interaction with OATP1B1 might be an important determinant of intra-individual variability of drug disposition.
Methods
Materials
Radiolabeled [3H]-estrone-3-sulphate (E3S) (54.3 Ci mmol−1, >97% purity) was purchased from PerkinElmer Life Sciences. Pravastatin sodium salt hydrate, rifampicin, quercetin (purity ≥98%, HPLC), sodium butyrate and Krebs-Henseleit buffer were purchased from Sigma-Aldrich. Polyclonal goat antibody against human OATP1B1 was purchased from Santa Cruz Biotechnology. Blasticidin S HCl and LipofectaminTM 2000 reagent were purchased from Invitrogen. Quercetin (purity ≥98%, HPLC) was purchased from Sigma-Aldrich Chemie GmbH. Quercetin capsules were purchased from GNC (USA; BN: 401922). Human OATP1B1 complementary DNA plasmid was kindly provided by Professor Richard Kim (Division of Clinical Pharmacology, Vanderbilt University, Nashville). Parental human embryonic kidney (HEK) 293 cell line was purchased from the Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (Beijing, P.R. China).
Cell culture and transfection
HEK293 cells were grown at 37°C in a humidified 5% CO2 atmosphere in Minimum Essential Medium supplemented with 10% FBS (Hyclone, Logan, UT), 100 U ml−1 penicillin and 100 µg ml−1 streptomycin. OATP1B1 cDNA was transfected into HEK293 cells with LipofectaminTM 2000 according to the manufacturer's instructions. Then, HEK293 cells were selected by 6 µg ml−1 blasticidin and colonies were picked up. The colonies which had the highest transport activities were used in the functional analysis (HEK293-OATP1B1). HEK293 cells transfected with vector alone were used to obtain the background activity (HEK293-MOCK). Sodium butyrate (10 mm) was added to the medium 24 h before use to induce the expression of OATP1B1.
Transport assays
HEK293-OATP1B1 and HEK293-MOCK cells were seeded in poly-L-lysine coated 24-well plates at an initial density of 2 × 105 cells/well. Cells were washed twice and pre-incubated with Krebs-Henseleit buffer at 37°C for 15 min, then incubated in 200 µl of Krebs-Henseleit buffer cells containing [3H]-E3S (50 nm) or pravastatin (50 µm) together with quercetin or the vehicle (0.2% dimethyl sulfoxide) at 37°C for 5 min (E3S) or 10 min (pravastatin). Rifampicin (100 µm) was used as a positive control inhibitor. The uptake reaction was terminated by adding ice-cold Krebs-Henseleit buffer after removal of the incubation buffer. For experiments with [3H]-E3S as substrate, cells were solubilized in 200 µl of 0.2 n NaOH and kept overnight at 4°C. After adding 50 µl 1 n HCl, aliquots (200 µl) were transferred to scintillation vials. The radioactivity was measured in a liquid scintillation counter (LS6500; Beckman Coulter, Inc., Fullerton, CA) after adding 2 ml of scintillation fluid. The remaining 50 µl of cell lysate was used to determine the protein concentrations by the method of Lowry et al. with bovine serum albumin (BSA) as a standard [18]. For experiments with pravastatin as substrate, cells were solubilized in 250 µl of 0.2% SDS and kept overnight. Aliquots (200 µl) were used to determine the amount of intracellular pravastatin by a LC/MS/MS method, and the remaining 50 µl aliquots of the cell lysis solution were used to determine the protein concentration by the method of Lowry et al. [18] with BSA as a standard. The uptake of [3H]-E3S or pravastatin was expressed as percentage of the control (uptake in HEK293-MOCK cells in the presence of 0.2% dimethyl sulphoxide).
LC/MS/MS assay for pravastatin
Concentrations of pravastatin accumulated in cells were determined using a validated liquid chromatography with tandem mass spectrometric detection (LC/MS/MS) method. Samples were prepared by adding 100 µl of internal standard solution (200 ng ml−1 simvastatin) to 100 µl of the cell lysates. LC/MS/MS analysis was performed by liquid chromatography-mass spectrometry with the Finnigan LCQ Deca XPplus (Finnigan, San Jose, CA). An Intersil ODS-3 column (5 µm, 2.1 mm × 50 mm) and a mobile phase [acetonitrile : 1 mm methanoic acid amine (including 0.1% formic acid) = 3:2] at a flow rate of 0.2 ml min−1 were applied. The lower limit of quantification was 0.98 ng ml−1. The ion transitions monitored were as follows: m/z 447 to 327 for pravastatin and m/z 441 to 325 for simvastatin. These transitions represent the product ions of the [M+H]+ ions. The lower limit of quantification for pravastatin was 0.957 ng ml−1. The calibration curves were linear over the range 0.957 to 500 ng ml−1 with mean correlation coefficients of 0.9992. The intra- and inter-day coefficients of variation were less than 10%.
Assessment of the effect of quercetin on the pharmacokinetic profile of pravastatin in vivo
Chinese-Han male volunteers were recruited for this study. All subjects underwent past medical history interviews and physical and laboratory examinations (haematology, serum biochemistry, urinalysis and electrocardiography) to confirm that they were healthy. Females were excluded to avoid possible effects of other factors (e.g. menstruation) on drug metabolism. Smokers were not included, nor were subjects deemed alcohol or drug dependent. Other exclusion criteria also included having participated in a clinical study during the past 4 months, taking any prescription or non-prescription drugs, grapefruit juice, herbal dietary supplements, coffee, green tea or any other quercetin-containing products within 2 weeks before study entry and until the study was completed. On the basis of our preliminary observation, we proposed that quercetin would reduce pravastatin AUC(0,10 h) after 14 days pretreatment by about 20%. Given this effect size, a probability of type 1 error of 0.05 (two-tailed), a power of 0.8, expected SD of pravastatin AUC(0,10 h) as 101 ng ml−1 h, 16 volunteers would be required by power analysis (GraphPad, StatMate).
Sixteen healthy Chinese-Han male volunteers (aged 19 to 24 years, weight range 60–80 kg, body mass index range 20–25 kg m−2) participated in the study after giving their individual written informed consent. The study was carried out in a two-phase, randomized and crossover manner with a 2 week washout period between phases. In each phase, 16 volunteers received 500 mg quercetin capsules or placebo (identical capsules filled with corn starch), once daily for 14 days. On day 14, after an overnight fast, all subjects received a single oral dose of 40 mg pravastatin (Mevalotin; Daiichi-Sankyo) 1 h after the placebo or quercetin administration. They were given a standardized meal at 4 h after the intake of pravastatin and standardized light meals after 10 h. The quercetin capsules contained quercetin (500 mg) and some other ingredients including cellulose, silicon dioxide, magnesium stearate and gelatin. This study protocol was approved by the Ethics Committee Board of Central South University, Changsha, Hunan, P.R. China (No. CTXY-100008). This clinical trial was registered with the Chinese Clinical Trial Registry (No. ChiCTR-TRC-10001277).
A series of venous blood samples (5 ml) was collected into EDTA-containing tubes before and at 0.25, 0.5, 0.75, 1, 2, 3, 4, 6, 8 and 10 h after pravastatin intake. Plasma pravastatin concentrations were quantified by use of LC/MS/MS analysis as described above. The peak concentration (Cmax) and time to Cmax (tmax) were obtained by inspection of the concentration – time data. The area under the concentration – time curve from 0–10h (AUC(0,10 h)) of pravastatin was calculated by the linear trapezoidal rule. λz is the elimination rate constant determined from the terminal slope of the log concentration – time plot and the elimination half-life (t1/2) = 0.693/λz. The estimate of oral clearance (CL/F) was calculated as CL/F = Dose (40 mg)/AUC(0,10 h) (ng ml−1 h).
Statistical analysis
All data were analyzed with SPSS software for Windows (version 18.0; SPSS, Chicago, IL). In vitro data were analyzed using anova followed by a Dunnett's post hoc test or by Student's t-test. The AUC(0,10 h), t1/2 and CL/F values of pravastatin after treatment with placebo and quercetin were analyzed using paired Student's t-test. The Wilcoxon signed-rank test was performed for the non-parametric paired two-group comparisons of Cmax and tmax. The results are expressed as mean ± SD in the text and tables and, for clarity, as mean ± SEM in the figures, unless otherwise stated. P < 0.05 indicated statistical significance.
Results
In vitro influence of quercetin on the OATP1B1-mediated uptake of E3S
In the present study, we established a HEK293 cell line stably expressing the human OATP1B1. According to a previous study, the low endogenous transporter expression in HEK293 cells supports its suitability as system for the overexpression of human transport proteins, especially SLC transporters [19]. The HEK293-OATP1B1 cells were characterized by a significantly higher expression of SLCO1B1 mRNA and OATP1B1 protein compared with HEK293-MOCK cells. The functional characterization showed about 5.5-fold higher uptake of E3S, a well established model substrate of OATP1B1 [20], into HEK293-OATP1B1 cells compared with HEK293-MOCK cells. Rifampicin (100 µm), a known OATP1B1 inhibitor [21], significantly decreased the OATP1B1-mediated [3H]-E3S uptake (Figure 1). Quercetin also substantially decreased OATP1B1 activity and the kinetic study revealed that quercetin inhibited OATP1B1-mediated [3H]E3S uptake in a competitive manner with a Ki value of 17.9 ± 4.6 µm (determined from Dixon plots) (Figure 2).
Figure 1.

Characterization of the HEK293-OATP1B1 cell line overexpression human OATP1B1. HEK293-OATP1B1 cells were characterized by real-time PCR (A) and immunoblot analysis (B). Na+/K+-ATPase, a membrane protein naturally expressed in all HEK293 cells, was used as loading control of surface proteins. The HEK293-OATP1B1 cells showed a significantly higher uptake (about 5.5-fold) of [3H]-E3S compared with HEK293-MOCK cells, rifampicin (100 µm) significantly decreased the OATP1B1 activity (C). **P < 0.01 compared with HEK293-MOCK cells. Each value is the mean value ± SEM (n = 6)
Figure 2.

Dixon plot analysis of inhibition of OATP1B1-mediated [3H]-E3S uptake by quercetin in HEK293 cells. Transport of E3S at three different concentrations (solid circles 50 nm, open circles 100 nm, solid triangles 200 nm) was measured for 5 min in the absence or presence of increasing concentrations of quercetin (0–100 µm). The data are expressed as percentage (mean values ± SEM, n = 3) relative to the initial radioactivity. The Ki values were determined graphically from a Dixon plot
In vitro influence of quercetin on the OATP1B1-mediated uptake of pravastatin
To determine whether quercetin alters the OATP1B1-mediated uptake of pravastatin, we performed a pravastatin uptake and inhibition experiment. We confirmed that pravastatin was transported by OATP1B1 with a substantially higher uptake in HEK293-OATP1B1 cells compared with HEK293-MOCK cells (about 4.1-fold), which was significantly decreased by rifampicin (100 µm). Quercetin markedly inhibited the OATP1B1-mediated uptake of pravastatin in a concentration-dependent manner with an IC50 value of 15.9 ± 1.4 µm (Figure 3).
Figure 3.

Inhibition of OATP1B1-mediated pravastatin transport by quercetin. Intracellular pravastatin accumulation (50 µm) in HEK293-OATP1B1 cells was about 4.1-fold higher than in HEK293-MOCK cells. Rifampicin (100 µm) was used as a positive control (A). Quercetin inhibited the OATP1B1-mediated uptake of pravastatin (50 µm) in a concentration-dependent manner (B). The data are expressed as percentage of control (uptake without quercetin). All data are presented as means ± SEM (n = 6). **P < 0.01 compared with HEK293-MOCK cells
In vivo influence of quercetin on the pharmacokinetics of pravastatin
The main pharmacokinetic parameters of pravastatin after 14 days of pretreatment by placebo or quercetin are summarized in Table 1. Mean plasma concentration profiles for pravastatin administered in combination with placebo or quercetin are shown in Figure 4. Repeated treatment with quercetin significantly increased Cmax of pravastatin by 31% (95% CI 20, 42%, P < 0.001) and AUC(0,10 h) by 24% (95% CI 15, 32%, P < 0.001). After quercetin treatment, CL/F of pravastatin significantly decreased by 18% (95% CI 11, 25%, P < 0.001) compared with the placebo phase. Meanwhile, quercetin prolonged t1/2 by 14% (95% CI 4, 24%, P = 0.027) with no significant influence on the tmax of pravastatin in comparison with the placebo phase. Intrasubject changes in the AUC(0,10 h) of pravastatin are depicted in Figure 5. It can be seen that only one subject had lower plasma exposure to pravastatin after concomitant administration with quercetin.
Table 1.
Pharmacokinetic parameters of pravastatin in 16 healthy subjects after a single dose of pravastatin (40 mg) on day 14 after pretreatment with placebo or quercetin for 14 days
| Parameter | Placebo phase (control) | Quercetin phase | Percentage of control (95% CI) | P value |
|---|---|---|---|---|
| AUC(0,10 h) (ng ml−1 h) | 354.6 ± 89.6 | 437.8 ± 118.6 | 124% (115, 132%) | <0.001 |
| Cmax (ng ml−1) | 123.2 ± 48.3 | 162.0 ± 76.0 | 131% (120, 142%) | <0.001 |
| tmax (h) | 0.9 ± 0.4 | 0.9 ± 0.3 | NA | 0.065 |
| t1/2 (h) | 1.6 ± 0.3 | 1.8 ± 0.3 | 114% (104, 124%) | 0.027 |
| CL/F (l h−1) | 119.5 ± 29.2 | 97.3 ± 24.7 | 82% (75, 89%) | <0.001 |
Data are presented as mean ± SD. tmax data are given as medians with range. AUC(0,10 h), area under the plasma drug concentration – time curve from 0 to 10 h; Cmax, peak plasma drug concentration; tmax, time to reach Cmax; t1/2, elimination half-life; CL/F, apparent clearance; NA not applicable; CI, confidence interval.
Figure 4.

Plasma concentration–time profiles of pravastatin in 16 healthy subjects after a single oral dose of 40 mg pravastatin on day 14 after pretreatment with placebo (○) or quercetin (●) for 14 days. Inset depicts the same data on a semilogarithmic scale. Each value is the mean value ± SEM
Figure 5.

Individual values for AUC(0,10 h) of pravastatin in 16 healthy Chinese-Han male subjects after a single oral dose of 40 mg pravastatin on day 14 after pretreatment with placebo or quercetin for 14 days. Grey circles indicate the mean
Discussion
In this study, we focused on the interactions of quercetin with the human hepatocellular uptake transporter OATP1B1. Firstly, using newly established HEK293 cells with stable expression of human OATP1B1, we examined the effect of quercetin on OATP1B1-mediated [3H]-E3S transport in vitro. OATP1B1-mediated uptake of E3S is known to be biphasic with Km values for high and low affinity components of 67.5 nm and 7 µm, respectively [22]. Because pravastatin mainly shares the high affinity site with E3S on OATP1B1 [23], so we set the concentration of [3H]-E3S at 50 nm (a value lower than 67.6 nm) for estimating the effect of quercetin on the high affinity binding site. Our data showed that quercetin inhibited the OATP1B1-mediated [3H]-E3S uptake in a competitive manner with a Ki value of 17.9 ± 4.6 µm. However, Wang et al. reported that quercetin did not exhibit a statistically significant influence on OATP1B1-mediated [3H]-DHEAS transport, which is not consistent with our present study [10]. This discrepancy may be possibly explained by differences in the substrates used, because different substrates may interact with different contact points to trigger the subsequent transport process, and this may explain the broad substrate specificity that characterizes many transporters. Then, we investigated the influence of quercetin on OATP1B1-mediated pravastatin transport in vitro. Our preliminary study had shown that OATP1B1-mediated pravastatin transport was linear for approximately 10 min, and uptake was saturable with an apparent Km of 83.9 µm (data not shown), which was consistent with the results of Kameyama et al. [24]. Therefore we set the concentration of pravastatin at 50 µm (a value lower than 83.9 µm). The results suggested that quercetin inhibited OATP1B1-mediated pravastatin transport in a dose-dependent fashion.
In order to verify our in vitro results, an in vivo study was performed to determine whether plasma concentrations of pravastatin and quercetin might be sufficient to cause a drug–drug interaction. Our in vivo results showed that co-administration of quercetin had a modest inhibitory effect on pravastatin systemic exposures (AUC).
Pravastatin is a substrate of several drug transporters in vivo. Except for OATP1B1, multidrug resistance-associated protein 2 (MRP2, ABCC2) is considered to be the major transporter involved in the biliary excretion of pravastatin [25]. Previous studies indicated that variations in the MRP2 activity mainly alter the liver concentration of pravastatin and have only a minimal effect on the plasma concentration [26]. Other transporters that may play a limited role in the disposition of pravastatin in vitro include OATP2B1, OAT3, bile salt export pump (BSEP) and BCRP [27]–[31], but their activities are shown not to be associated with differences in pravastatin pharmacokinetics in healthy subjects [32], [33]. Therefore, we concluded that the changes in pravastatin pharmacokinetics in this study were mainly caused by quercetion-mediated inhibition of OATP1B1 activity.
Although quercetin is ubiquitously present in fruit and vegetables, it has been marketed in the United States primarily as a popular dietary supplement due to its beneficial health effects [3]. The recommended daily dosages of supplemental quercetin range from 200 mg to 1200 mg [34]. Therefore, we used a dose of 500 mg daily of quercetin according to the package insert in our study. In this study, we did not examine the pharmacokinetics of quercetin. Human pharmacokinetic studies have demonstrated that after a non-toxic i.v. dose of quercetin, its serum concentration ranges from 1 to 400 µm and after oral administration of 300 mg quercetin supplement, its maximum plasma concentration is 9.72 µm[35], [36]. Furthermore, quercetin has a relatively long plasma half-life of 11–28 h in humans, indicating that repeated dietary intake of quercetin will lead to accumulation in plasma [37]. Comparing these data with the in vitro and in vivo results obtained in this study, we speculate that our results may have some clinical relevance. Recently, several in vitro studies revealed that many metabolites of quercetin, such as 7-O-glucuronosyl quercetin and 3′-O-glucuronide quercetin, also had an inhibitory influence on the function of some drug transporters including MRP1, MRP2 and OAT1 [6], [38]. Further investigation is needed to determine the effects of metabolites of quercetin on OATP1B1 activity.
SLCO1B1 polymorphism, especially c.521T > C, can significantly influence the pharmacokinetics of pravastatin in humans. A number of recent studies suggested that the c.521T > C SNP was strongly associated with decreased activity of OATP1B1 resulting in higher oral bioavailability of pravastatin in different ethnic groups [39]. The SNP c.388A > G may be also associated with increased activity of OATP1B1 resulting in lower oral bioavailability of pravastatin [40]. The c.521T > C SNP frequency among Chinese individuals (13%) was slightly lower than the frequency observed in Caucasians (14.3%), and the c.388A > G SNP frequency in the Chinese population (79.5%) is much more higher compared with the Caucasian population (30.6%) [41]. Therefore, our results in this study had similar clinical values to Caucasians. However, pravastatin is never given clinically as a single dose and this is a limitation of the present study. Although previous studies had suggested that alteration in OATP1B1 function seemed to have a similar impact on the pharmacokinetics of single and multiple dose pravastatin [16], the effect of quercetion on the pharmacokinetics of multiple dose pravastatin requires further examination.
In conclusion, we showed for the first time that quercetin can inhibit OATP1B1-mediated transport of E3S and pravastatin in vitro, and also has a modest inhibitory influence on the pharmacokinetics of pravastatin in healthy Chinese-Han male subjects. The results of this study suggest a new mechanism to account for certain natural product – drug interactions, and may be clinically significant for patients taking quercetin and OATP1B1 substrate drugs simultaneously.
Acknowledgments
This work was supported by the National Scientific Foundation of China (No. 30801421, 30901834, 30873089, 81001445), Huge Project to Boost Chinese Drug Development (No.2009ZX09501-032, 2009ZX09304-003), 863 Project (No. 2009AA022710, 2009AA022703, 2009AA022704) and Program for NCET.
Competing Interests
There are no competing interests to declare.
REFERENCES
- 1.Chen C, Zhou J, Ji C. Quercetin: a potential drug to reverse multidrug resistance. Life Sci. 2010;87:333–8. doi: 10.1016/j.lfs.2010.07.004. [DOI] [PubMed] [Google Scholar]
- 2.Erlund I. Review of the flavonoids quercetin, hesperetin, and naringenin. Dietary sources, bioactivities, and epidemiology. Nut Res. 2004;24:851–74. [Google Scholar]
- 3.Theoharides TC, Bielory L. Mast cells and mast cell mediators as targets of dietary supplements. Ann Allergy Asthma Immunol. 2004;93:S24–34. doi: 10.1016/s1081-1206(10)61484-6. [DOI] [PubMed] [Google Scholar]
- 4.van Zanden JJ, Wortelboer HM, Bijlsma S, Punt A, Usta M, Bladeren PJ, Rietjens IM, Cnubben NH. Quantitative structure activity relationship studies on the flavonoid mediated inhibition of multidrug resistance proteins 1 and 2. Biochem Pharmacol. 2005;69:699–708. doi: 10.1016/j.bcp.2004.11.002. [DOI] [PubMed] [Google Scholar]
- 5.Mandery K, Bujok K, Schmidt I, Keiser M, Siegmund W, Balk B, König J, Fromm MF, Glaeser H. Influence of the flavonoids apigenin, kaempferol and quercetin on the function of organic anion transporting polypeptides 1A2 and 2B1. Biochem Pharmacol. 2010;80:1746–53. doi: 10.1016/j.bcp.2010.08.008. [DOI] [PubMed] [Google Scholar]
- 6.Wong CC, Botting NP, Orfila C, Al-Maharik N, Williamson G. Flavonoid conjugates interact with organic anion transporters (OATs) and attenuate cytotoxicity of adefovir mediated by organic anion transporter 1 (OAT1/SLC22A6) Biochem Pharmacol. 2011;81:942–9. doi: 10.1016/j.bcp.2011.01.004. [DOI] [PubMed] [Google Scholar]
- 7.Konig J, Cui Y, Nies AT, Keppler D. A novel human organic anion transporting polypeptide localized to the basolateral hepatocyte membrane. Am J Physiol. 2000;278:G156–64. doi: 10.1152/ajpgi.2000.278.1.G156. [DOI] [PubMed] [Google Scholar]
- 8.Abe T, Kakyo M, Tokui T, Nakagomi R, Nishio T, Nakai D, Nomura H, Unno M, Suzuki M, Naitoh T, Matsuno S, Yawo H. Identification of a novel gene family encoding human liver-specific organic anion transporter LST-1. J Biol Chem. 1999;274:17159–63. doi: 10.1074/jbc.274.24.17159. [DOI] [PubMed] [Google Scholar]
- 9.Kim RB. Organic anion-transporting polypeptide (OATP) transporter family and drug disposition. Eur J. Clin Investig. 2003;33(Suppl. 2):1–5. doi: 10.1046/j.1365-2362.33.s2.5.x. [DOI] [PubMed] [Google Scholar]
- 10.Wang X, Wolkoff AW, Morris ME. Flavonoids as a novel class of human organic anion-transporting polypeptide OATP1B1 (OATP-C) modulators. Drug Metab Dispos. 2005;33:1666–72. doi: 10.1124/dmd.105.005926. [DOI] [PubMed] [Google Scholar]
- 11.Grundy SM. HMG-CoA reductase inhibitors for treatment of hypercholesterolemia. N Engl J Med. 1988;319:24–33. doi: 10.1056/NEJM198807073190105. [DOI] [PubMed] [Google Scholar]
- 12.Hatanaka T. Clinical pharmacokinetics of pravastatin: mechanisms of pharmacokinetic events. Clin Pharmacokinet. 2000;39:397–412. doi: 10.2165/00003088-200039060-00002. [DOI] [PubMed] [Google Scholar]
- 13.Williams D, Feely J. Pharmacokinetic-pharmacodynamic drug interactions with HMG-CoA reductase inhibitors. Clin Pharmacokinet. 2002;41:343–70. doi: 10.2165/00003088-200241050-00003. [DOI] [PubMed] [Google Scholar]
- 14.Nakai D, Nakagomi R, Furuta Y, Tokui T, Abe T, Ikeda T, Nishimura K. Human liver-specific organic anion transporter, LST-1, mediates uptake of pravastatin by human hepatocytes. J Pharmacol Exp Ther. 2001;297:861–7. [PubMed] [Google Scholar]
- 15.Yamazaki M, Akiyama S, Nishigaki R, Sugiyama Y. Uptake is the rate-limiting step in the overall hepatic elimination of pravastatin at steady-state in rats. Pharm Res. 1996;13:1559–64. doi: 10.1023/a:1016044032571. [DOI] [PubMed] [Google Scholar]
- 16.Kivistö KT, Niemi M. Influence of drug transporter polymorphisms on pravastatin pharmacokinetics in humans. Pharm Res. 2007;24:239–47. doi: 10.1007/s11095-006-9159-2. [DOI] [PubMed] [Google Scholar]
- 17.Ieiri I, Takane H, Hirota T, Otsubo K, Higuchi S. Genetic polymorphisms of drug transporters: pharmacokinetic and pharmacodynamic consequences in pharmacotherapy. Expert Opin Drug Metab Toxicol. 2006;2:651–74. doi: 10.1517/17425255.2.5.651. [DOI] [PubMed] [Google Scholar]
- 18.Lowry OH, Rosebrough NJ, Farr AL, Randall RJ. Protein measurement with the Folin phenol reagent. J Biol Chem. 1951;193:265–75. [PubMed] [Google Scholar]
- 19.Ahlin G, Hilgendorf C, Karlsson J, Szigyarto CA, Uhlén M, Artursson P. Endogenous gene and protein expression of drug-transporting proteins in cell lines routinely used in drug discovery programs. Drug Metab Dispos. 2009;37:2275–83. doi: 10.1124/dmd.109.028654. [DOI] [PubMed] [Google Scholar]
- 20.Choi MK, Shin HJ, Choi YL, Deng JW, Shin JG, Song IS. Differential effect of genetic variants of Na(+)-taurocholate co-transporting polypeptide (NTCP) and organic anion-transporting polypeptide 1B1 (OATP1B1) on the uptake of HMG-CoA reductase inhibitors. Xenobiotica. 2011;41:24–34. doi: 10.3109/00498254.2010.523736. [DOI] [PubMed] [Google Scholar]
- 21.Vavricka SR, Van Montfoort J, Ha HR, Meier PJ, Fattinger K. Interactions of rifamycin SV and rifampicin with organic anion uptake systems of human liver. Hepatology. 2002;36:164–72. doi: 10.1053/jhep.2002.34133. [DOI] [PubMed] [Google Scholar]
- 22.Tamai I, Nozawa T, Koshida M, Nezu J, Sai Y, Tsuji A. Functional characterization of human organic anion transporting polypeptide B (OATP-B) in comparison with liver-specific OATP-C. Pharm Res. 2001;18:1263–9. doi: 10.1023/a:1013077609227. [DOI] [PubMed] [Google Scholar]
- 23.Kitamura A, Imai S, Yabuki M, Komuro S. The new acyl-CoA cholesterol acyltransferase inhibitor SMP-797 does not interact with statins via OATP1B1 in human cryopreserved hepatocytes and oocytes expressing systems. Biopharm Drug Dispos. 2007;28:517–25. doi: 10.1002/bdd.581. [DOI] [PubMed] [Google Scholar]
- 24.Kameyama Y, Yamashita K, Kobayashi K, Hosokawa M, Chiba K. Functional characterization of SLCO1B1 (OATP-C) variants, SLCO1B1*5, SLCO1B1*15 and SLCO1B1*15+C1007G, by using transient expression systems of HeLa and HEK293 cells. Pharmacogenet Genomics. 2005;15:513–22. doi: 10.1097/01.fpc.0000170913.73780.5f. [DOI] [PubMed] [Google Scholar]
- 25.Ieiri I, Higuchi S, Sugiyama Y. Genetic polymorphisms of uptake (OATP1B1, 1B3) and efflux (MRP2, BCRP) transporters: implications for inter-individual differences in the pharmacokinetics and pharmacodynamics of statins and other clinically relevant drugs. Expert Opin Drug Metab Toxicol. 2009;5:703–29. doi: 10.1517/17425250902976854. [DOI] [PubMed] [Google Scholar]
- 26.Ide T, Sasaki T, Maeda K, Higuchi S, Sugiyama Y, Ieiri I. Quantitative population pharmacokinetic analysis of pravastatin using an enterohepatic circulation model combined with pharmacogenomic Information on SLCO1B1 and ABCC2 polymorphisms. J Clin Pharmacol. 2009;49:1309–17. doi: 10.1177/0091270009341960. [DOI] [PubMed] [Google Scholar]
- 27.Shirasaka Y, Suzuki K, Nakanishi T, Tamai I. Intestinal absorption of HMG-CoA reductase inhibitor pravastatin mediated by organic anion transporting polypeptide. Pharm Res. 2010;27:2141–9. doi: 10.1007/s11095-010-0216-5. [DOI] [PubMed] [Google Scholar]
- 28.Kobayashi D, Nozawa T, Imai K, Nezu J, Tsuji A, Tamai I. Involvement of human organic anion transporting polypeptide OATP-B (SLC21A9) in pH-dependent transport across intestinal apical membrane. J Pharmacol Exp Ther. 2003;306:703–8. doi: 10.1124/jpet.103.051300. [DOI] [PubMed] [Google Scholar]
- 29.Takeda M, Noshiro R, Onozato ML, Tojo A, Hasannejad H, Huang XL, Narikawa S, Endou H. Evidence for a role of human organic anion transporters in the muscular side effects of HMG-CoA reductase inhibitors. Eur J Pharmacol. 2004;483:133–8. doi: 10.1016/j.ejphar.2003.10.017. [DOI] [PubMed] [Google Scholar]
- 30.Hirano M, Maeda K, Hayashi H, Kusuhara H, Sugiyama Y. Bile salt export pump (BSEP/ABCB11) can transport a nonbile acid substrate, pravastatin. J Pharmacol Exp Ther. 2005;314:876–82. doi: 10.1124/jpet.105.084830. [DOI] [PubMed] [Google Scholar]
- 31.Matsushima S, Maeda K, Kondo C, Hirano M, Sasaki M, Suzuki H, Sugiyama Y. Identification of the hepatic efflux transporters of organic anions using double-transfected Madin-Darby canine kidney II cells expressing human organic anion-transporting polypeptide 1B1 (OATP1B1)/multidrug resistance-associated protein 2, OATP1B1/multidrug resistance 1, and OATP1B1/breast cancer resistance protein. J Pharmacol Exp Ther. 2005;314:1059–67. doi: 10.1124/jpet.105.085589. [DOI] [PubMed] [Google Scholar]
- 32.Niemi M, Schaeffeler E, Lang T, Fromm MF, Neuvonen M, Kyrklund C, Backman JT, Kerb R, Schwab M, Neuvonen PJ, Eichelbaum M, Kivistö KT. High plasma pravastatin concentrations are associated with single nucleotide polymorphisms and haplotypes of organic anion transporting polypeptide-C (OATP-C, SLCO1B1) Pharmacogenetics. 2004;14:429–40. doi: 10.1097/01.fpc.0000114750.08559.32. [DOI] [PubMed] [Google Scholar]
- 33.Ho RH, Choi L, Lee W, Mayo G, Schwarz UI, Tirona RG, Bailey DG, Michael Stein C, Kim RB. Effect of drug transporter genotypes on pravastatin disposition in European- and African-American participants. Pharmacogenet Genomics. 2007;17:647–56. doi: 10.1097/FPC.0b013e3280ef698f. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Egert S, Wolffram S, Bosy-Westphal A, Boesch-Saadatmandi C, Wagner AE, Frank J, Rimbach G, Mueller MJ. Daily quercetin supplementation dose-dependently increases plasma quercetin concentrations in healthy humans. J Nutr. 2008;138:1615–21. doi: 10.1093/jn/138.9.1615. [DOI] [PubMed] [Google Scholar]
- 35.Ferry DR, Smith A, Malkhandi J, Fyfe DW, deTakats PG, Anderson D, Baker J, Kerr DJ. Phase I clinical trial of the flavonoid quercetin: pharmacokinetics and evidence for in vivo tyrosine kinase inhibition. Clin Cancer Res. 1996;2:659–68. [PubMed] [Google Scholar]
- 36.Hubbard GP, Wolffram S, Lovegrove JA, Gibbins JM. Ingestion of quercetin inhibits platelet aggregation and essential components of the collagen-stimulated platelet activation pathway in humans. J Thromb Haemost. 2004;2:2138–45. doi: 10.1111/j.1538-7836.2004.01067.x. [DOI] [PubMed] [Google Scholar]
- 37.Williamson G, Manach C. Bioavailability and bioefficacy of polyphenols in humans. II. Review of 93 intervention studies. Am J Clin Nutr. 2005;81:243S–55S. doi: 10.1093/ajcn/81.1.243S. [DOI] [PubMed] [Google Scholar]
- 38.van Zanden JJ, van der Woude H, Vaessen J, Usta M, Wortelboer HM, Cnubben NH, Rietjens IM. The effect of quercetin phase II metabolism on its MRP1 and MRP2 inhibiting potential. Biochem Pharmacol. 2007;74:345–51. doi: 10.1016/j.bcp.2007.04.002. [DOI] [PubMed] [Google Scholar]
- 39.Romaine SP, Bailey KM, Hall AS, Balmforth AJ. The influence of SLCO1B1 (OATP1B1) gene polymorphisms on response to statin therapy. Pharmacogenomics J. 2010;10:1–11. doi: 10.1038/tpj.2009.54. [DOI] [PubMed] [Google Scholar]
- 40.Rodrigues AC, Perin PMS, Purim SG, Silbiger VN, Genvigir FDV, Willrich MAV, Arazi SS, Luchessi AD, Hirata MH, Bernik MMS, Dorea EL, Santos C, Faludi AA, Bertolami MC, Salas A, Freire A, Lareu MV, Phillips C, Porras-Hurtado L, Fondevila M, Carracedo A, Hirata RDC. Pharmacogenetics of OATP transporters reveals that SLCO1B1 c.388A > G variant is determinant of increased atorvastatin response. Int J Mol Sci. 2011;12:5818–27. doi: 10.3390/ijms12095815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Mwinyi J, Köpke K, Schaefer M, Roots I, Gerloff T. Comparison of SLCO1B1 sequence variability among German, Turkish and African populations. Eur J Clin Pharmacol. 2008;64:257–66. doi: 10.1007/s00228-007-0409-y. [DOI] [PubMed] [Google Scholar]
