Novobiocin

Usefulness of novobiocin as a selective inhibitor of intestinal breast cancer resistance protein (Bcrp) in rats

Kei Suzuki1,*, Kazuhiro Taniyama1, Takao Aoyama2 and Yoshiaki Watanabe1

1Exploratory Research Laboratories, Drug Research department, TOA EIYO LTD., Fukushima, Japan

2Faculty of Pharmaceutical Science, Tokyo University of Science, Chiba, Japan

*Corresponding author: Kei Suzuki, Exploratory Research Section Ⅲ, Exploratory Research Laboratories, Drug Research Department, TOA EIYO LTD., 1, Yuno-tanaka, Iizaka-machi, Fukushima-shi, Fukushima 960-0280, Japan. Tel.: +81-24-542-3143. Fax: +81-24-542-8641. E-mail: [email protected]

Acknowledgements

The authors thank Takashi Sato (TOA EIYO LTD.) for valuable discussions. The authors also would like to thank Enago (www.enago.jp) for the English language review.

Usefulness of novobiocin as a selective inhibitor of intestinal breast cancer resistance protein (Bcrp) in rats

1.We investigated whether novobiocin is useful for elucidating the contribution of breast cancer resistance protein (Bcrp) to intestinal absorption without affecting the activities of P-glycoprotein (P-gp), cytochrome P450 (CYP) 3A and hepatic organic anion transporting polypeptide (Oatp) in rats.
2.To determine the effects of novobiocin on Bcrp, P-gp, CYP3A and Oatp activities, we used sulfasalazine, fexofenadine, bosentan and midazolam, respectively, as probe substrates. Each substrate was orally or intravenously administered to rats 15 min after oral novobiocin administration at a dose of 3 mg/kg.
3.Pre-treatment with novobiocin significantly increased the area under the plasma concentration–time curve and the peak plasma concentration of sulfasalazine after oral administration by 3.2- and 5.9-fold, respectively, in rats, whereas its systemic clearance following intravenous dosing was not influenced. These results indicate that
novobiocin selectively inhibits intestinal Bcrp-mediated efflux with limited effects on extra-intestinal Bcrp activity.
4.In addition, novobiocin pre-treatment did not significantly alter the pharmacokinetic parameters of orally administered fexofenadine and midazolam or intravenously administered bosentan, suggesting that the effects of novobiocin on other processes were negligible.
5.These findings demonstrate that novobiocin permits estimating the net contribution of Bcrp to intestinal absorption of drug candidates.

Keywords: novobiocin, Bcrp, P-gp, CYP3A, Oatp, rats, intestine

Word count: 4326

Introduction
Breast cancer resistant protein (BCRP/ABCG2) is an efflux transporter expressed in the apical membrane of epithelial cells in various normal tissues. It controls the absorption and biliary and urinary excretion of its substrates in the small intestine, liver and kidneys, respectively. Indeed, the area under the plasma concentration–time curve (AUC) of oral sulfasalazine and rosuvastatin, which are typical BCRP substrates, increased by 150- and 3.8- fold, respectively, in Bcrp-knockout mice compared with wild-type mice. (Gertz et al., 2010).
Several single nucleotide polymorphisms in the human ABCG2 gene reduce BCRP- mediated transport activity (Bedada et al., 2014; Dahan and Amidon 2009; Lau et al., 2004; Peng et al., 2014); among its variants, the ABCG2 421C>A variant is one of the most common one. Studies have reported that mutation in this variant significantly increases the AUC of orally administered sulfasalazine by 2.4–3.5-fold (Tachibana et al., 2012; Duan and You 2009; Gotanda et al., 2015) and rosuvastatin by 1.6–2.6-fold (Zhou et al., 2013; Wan et al., 2015; Zhang et al., 2006; Keskitalo et al., 2009; Lee et al., 2013) in humans. The allele frequency of ABCG2 421C>A varies by ethnicity as follows: 48%–57% in East Asians, 17% in Caucasians and 0%–5% in Africans (Li and Barton 2018; Soko et al., 2019); therefore, this polymorphism contributes to the inter-individual variability of drugs that are BCRP
substrates. Interestingly, an increase in the oral exposure of BCRP substrates is not associated with changes in tmax, the time to reach the peak plasma concentration (Cmax), and the apparent elimination half-life (t1/2). The ABCG2 421C>A variant might influence the gastrointestinal absorption process rather than the elimination of drugs. Therefore, it is important to estimate the net contribution of BCRP-mediated efflux to the intestinal absorption of drug candidates in the non-clinical drug development stage.
Studies have demonstrated that several compounds inhibit intestinal Bcrp in vivo in rats (Wang and Morris 2007; Kunimatsu et al., 2013; Matsuda et al., 2013; Su et al., 2007;

Zhou et al., 2017); however, little information is available about the in vivo specificity for intestinal Bcrp in rats. Among Bcrp inhibitors, curcumin is one of the most studied compounds, and it is reported that the compound significantly increases the AUC of orally administered rosuvastatin in rats (Zhou et al., 2017). However, the authors observed the in vitro inhibitory effects of curcumin and curcumin-O-glucuronide (a major curcumin metabolite) on human organic anion transporting polypeptide (OATP) 1B1 and OATP1B3 activities and suggested that inhibition of hepatic Oatp may be a part of their action mechanisms in rats. Furthermore, another study has reported that curcumin increases the AUC of oral docetaxel, an Oatp substrate, by approximately 2-fold and decreases its systemic clearance by approximately 50% in rats (Pade and Stavchansky 1998). The mechanism of
this interaction might involve the inhibition of hepatic Oatp. Considering the findings of these previous studies, curcumin could potentially inhibit hepatic Oatp in rats. Moreover, it has an inhibitory effect on cytochrome P450 (CYP) 3A4 with a 50% inhibitory concentration (IC50) of 2.7 μmol/L, and it significantly increases the AUC and Cmax of co-administered
etoposide, a substrate of CYP3A4, without affecting its systemic clearance in rats (Kadono et al., 2014). These authors have also stated that the increased oral exposure of etoposide might be attributable to the inhibition of intestinal CYP3A by curcumin. Based on these observations, even if the increased exposure of a compound by co-administration of curcumin is observed in rats, it is difficult to determine the contribution of Bcrp to the intestinal absorption. Therefore, a selective intestinal Bcrp inhibitor could be used to elucidate the net contribution of ABCG2 421C>A to the oral absorption of drug candidates.
Novobiocin exerts potent inhibitory effects on the efflux transport activity of human BCRP but not P-glycoprotein (P-gp) (Su et al., 2007). A study has demonstrated that the inhibitor at a dose of 50 mg/kg significantly increased the AUC and Cmax of orally co- administered topotecan, a typical Bcrp substrate (Su et al., 2007), in rats; however, this

novobiocin dose decreased the systemic clearance of topotecan following its intravenous administration, thereby making it difficult to estimate the contribution of Bcrp to intestinal absorption without an intravenous study. We hypothesised that reduced novobiocin doses exert few effects on the systemic clearance of a Bcrp substrate in rats because the inhibitor concentration in the intestine is assumed to be high compared with that in the systemic circulation. Therefore, the present study investigated the usefulness of reduced doses of novobiocin in the selective inhibition of intestinal Bcrp activity without affecting extra- intestinal Bcrp, other transporters (P-gp and hepatic Oatp) and CYP3A in rats.

Materials and Methods
Chemicals

Sulfasalazine, diclofenac sodium and flecainide acetate were purchased from Sigma-Aldrich (St Louis, MO, USA). Fexofenadine hydrochloride was purchased from Toronto Research Chemicals (Toronto, ON, Canada). Midazolam was purchased from FUJIFILM Wako Pure Chemical (Osaka, Japan). Bosentan monohydrate was purchased from Tokyo Chemical Industry. 1′-Hydroxymidazolam-d4 was purchased from Cerilliant Corporation (Austin, TX, USA). Dormicum (5 mg/mL midazolam solution) for intravenous injection was purchased from Astellas Pharma Inc. (Tokyo, Japan). Rat cDNA-expressed CYP3A2, NADPH Regenerating System and 1′-hydroxymidazolam were purchased from Corning (Corning, NY, USA). All other reagents and solutions were commercial products of analytical grade.

Animals

Male Sprague–Dawley rats (Japan SLC, Hamamatsu, Japan) aged 8 weeks (body weight, approximately 250 g) were used in this study. Animals were housed under a 12-h light/dark cycle with free access to a standard diet and tap water. The protocols for the animal

experiments were approved by the Animal Care and Use Committee of TOA EIYO LTD.

 

In vivo pharmacokinetic studies

All animals were fasted for approximately 18 h before dosing. Each group comprised 3–6 rats. Novobiocin was dissolved in distilled water, and sulfasalazine and fexofenadine were suspended in aqueous 0.5% methylcellulose. For the midazolam dosing solution, dormicam (5 mg/mL midazolam solution for intravenous injection) was diluted with distilled water. For the oral administration studies, sulfasalazine (5 mg/kg), fexofenadine (5 mg/kg) and midazolam (1 mg/kg) were administered 15 min after oral administration of vehicle or novobiocin (3 mg/kg) to rats. For intravenous administration studies, sulfasalazine and bosentan were dissolved in dimethyl sulfoxide (DMSO), diluted with phosphate-buffered saline (DMSO final concentration: 5%) and administered to rats at a dose of 1 mg/kg via the tail vein 15 min after the oral administration of vehicle or novobiocin (3 mg/kg). Animals were fed 4 h after dosing, and blood samples were obtained from the jugular vein at 0.083, 0.17 (for midazolam only), 0.25, 0.5, 1, 2, 4, 6 and 8 h following dosing. Plasma samples were separated by centrifugation at 12000 rpm for 10 min at 4°C and stored at -30°C until use. To measure all compounds, excluding novobiocin, a 50-μL aliquot of plasma was mixed with 50 μL of water, 50 μL of the internal standard (IS, 100 ng/mL) and 250 μL of acetonitrile. For novobiocin, a 20-μL aliquot of the plasma was mixed with 20 μL of water, 20 μL of 100 ng/mL IS and 200 μL of acetonitrile. These samples were then centrifuged at 3000 rpm for 20 min at 4°C. The resultant supernatants were diluted with suitable solvents and injected into a liquid chromatography tandem mass spectrometry (LC–MS/MS) system.

CYP3A2 inhibition assays using a rat cDNA-expressed system

Preliminary experiments indicated that 1′-hydroxymidazolam production was linear for

incubation up to 5 min at 1 pmol/mL in rat cDNA-expressed CYP3A2. Novobiocin (1, 3, 10,

30 and 100 μmol/L) was added to an incubation mixture containing rat cDNA-expressed CYP3A2 (1 pmol/mL), midazolam (2 μmol/L) and 100 mmol/L phosphate buffer (pH 7.4). The final methanol concentration was less than 0.1%. The incubation mixtures were pre- warmed for 5 min, and then the reaction was initiated by adding the NADPH Regenerating System (glucose 6-phosphate [3.3 mmol/L], NADP [1.3 mmol/L], glucose-6-phosphate dehydrogenase [0.4 units/mL] and MgCl2 [3.3 mmol/L]) in a shaking water bath at 37°C. After 5 min of incubation, the reaction was terminated by adding 200 μL of ice-cold acetonitrile containing 10 ng/mL IS. Assays were performed in triplicate. These samples were centrifuged at 3000 rpm for 15 min at 4°C, and the resultant supernatants were diluted with 0.1% formic acid in water, followed by injection into an LC–MS/MS system.

Determination of the unbound fraction of novobiocin in rat plasma protein (fu,p)

The fu,p was determined using a 96-well rapid equilibrium dialysis (RED) plate with an 8-kDa molecular weight cut-off dialysis membrane (Thermo Fisher Scientific, Waltham, MA,
USA). Rat plasma was spiked with novobiocin solution in phosphate buffer (pH 7.4) to obtain a final concentration of 1 μmol/L. A 300-μL aliquot of the plasma mixture was added to the donor side of the RED plate, 500 μL of 100 mmol/L phosphate buffer (pH 7.4) was added to the acceptor side and the plate was placed on a shaker for 16 h at 37°C. The assays were performed in triplicate. After incubation, a 50-μL aliquot from the donor side was mixed with 50 μL of 100 mmol/L phosphate buffer, and a 50-μL aliquot from the acceptor side was mixed with 50 μL of rat plasma. These samples were mixed with 50 μL of 100
ng/mL IS and 250 μL of acetonitrile and then centrifuged at 3000 rpm for 20 min at 4°C. The resultant supernatants were diluted with water and injected into an LC–MS/MS system.

LC–MS/MS analysis

An LC–MS/MS system comprising a Triple Quad 5500 (AB Sciex, Tokyo, Japan) combined with an Agilent 1290 Infinity LC system (Agilent, Santa Clara, CA, USA) was used for analysis. Chromatography was performed using an ACQUITY UPLC BEH C18 Column (130Å, 1.7 µm, 2.1 mm × 50 mm; Waters, Milford, MA, USA) warmed to 40°C and run at a flow rate of 0.4 mL/min. The mobile phases comprised a two-solvent pair: 1) A (distilled water) and B (0.1% formic acid in acetonitrile) and 2) C (15 mmol/L ammonium acetate) and D (acetonitrile). The elution gradients were established as follows: sulfasalazine, 0 min, 20% D; 2 min, 50% D; 2.5 min, 50% D; 2.51 min, 20% D; 3 min, 20% D; all other compounds,
0 min, 0% B; 0.5 min, 0% B; 1.5 min, 100% B; 2 min, 100% B; 2.01 min, 0% B; 2.5 min, 0% B. A multiple reaction monitoring experiment was conducted in the positive ion mode (excluding sulfasalazine, which required the negative ion mode) by monitoring the selected ions (precursor ion/product ion) for each compound as follows: sulfasalazine (397/197), fexofenadine (502/466), midazolam (326/291), bosentan (552/202), novobiocin (613/189), 1′- hydroxymidazolam (342/324), diclofenac (294/214), flecainide (415/301) and 1′- hydroxymidazolam-d4 (346/328). Flecainide was used as the IS for all experiments except for measuring sulfasalazine (IS: diclofenac) and 1′-hydroxymidazolam (IS: 1′- hydroxymidazolam-d4). The calibration range for each compound was as follows: sulfasalazine (1‒500 ng/mL); fexofenadine (0.1‒20 ng/mL); midazolam (0.02‒20 ng/mL); bosentan (2‒500 ng/mL); novobiocin (0.1‒10 ng/mL). Sample concentrations were calculated by linear regression of a standard curve of the ratio of the analyte peak area to the IS peak
area using Analyst ver. 1.6.3 (AB Sciex).

Data analysis

Plasma concentration data were analysed individually, and the pharmacokinetic parameters were obtained by non-compartmental analysis of plasma concentration versus time data (WinNonlin 2.1, Pharsight, Mountain View, CA, USA). Cmax and tmax were recorded directly from experimental observations. The AUC from time zero to the last quantifiable time (AUC0–last) was calculated using the linear trapezoidal method. The AUC from the time of the last quantifiable concentration (Clast) to infinity (AUClast–inf) was calculated by dividing the Clast by the first-order elimination rate constant (ke). Finally, the AUC from time zero to infinity (AUC0–inf) was obtained by adding AUC0–last and AUClast–inf. The total plasma clearance (CLtot), steady-state volume of distribution (Vdss) and t1/2 were estimated after intravenous administration. ke and t1/2 were estimated using at least three time points with quantifiable concentrations. Data for each pharmacokinetic parameter from individual animals were averaged and presented as the mean ± SD. Differences were assessed using a two-tailed unpaired Student’s t-test. In all cases, a probability level of p < 0.05 was considered statistically significant.

Results
Effects of novobiocin pre-treatment on the pharmacokinetics of probe substrates in rats

Figure 1 presents the plasma concentration–time profiles of orally (5 mg/kg) and intravenously (1 mg/kg) administered sulfasalazine 15 min after the oral administration of novobiocin (3 mg/kg) or vehicle in rats. The pharmacokinetic parameters of sulfasalazine are summarised in Table 1. Novobiocin pre-treatment significantly increased the AUC0–inf and Cmax of oral sulfasalazine by 3.2- and 5.9-fold, respectively, whereas no significant difference was observed in the pharmacokinetic parameters of intravenous sulfasalazine. As shown in

Figure 2 and Table 2, novobiocin pre-treatment did not affect the plasma concentrations of orally administered fexofenadine (5 mg/kg). Similarly, novobiocin pre-treatment did not influence the plasma concentrations of orally administered midazolam (1 mg/kg) significantly (Figure 3 and Table 2). The tmax and t1/2 of oral sulfasalazine, fexofenadine and midazolam remained unchanged regardless of novobiocin pre-treatment. Novobiocin pre- treatment had no significant effect on the pharmacokinetic parameters of intravenously administered bosentan compared with the control values (Figure 4 and Table 2).
We analysed plasma samples from the pre-treatment study with oral sulfasalazine to obtain the unbound plasma concentration–time profile of novobiocin in rats (Figure 5). The unbound Cmax of novobiocin was 19.7 nmol/L (total Cmax = 1.3 μmol/L; fu,p = 0.0146).

Inhibitory effect of novobiocin on rat CYP3A2 activity

Novobiocin did not exert inhibitory effects on midazolam 1′-hydroxylation activity up to 100 μmol/L using rat cDNA-expressed CYP3A2, whereas ketoconazole, a CYP3A2 positive control inhibitor, strongly inhibited CYP3A2 activity by 93% at 10 μmol/L.

Discussion
The present study investigated the usefulness of novobiocin for evaluating the contribution of Bcrp to the intestinal absorption of drugs in vivo in rats. We observed that 3 mg/kg novobiocin significantly increased the AUC0–inf of orally administered sulfasalazine, but it
did not affect the pharmacokinetic parameters of sulfasalazine after its intravenous administration. Furthermore, no significant difference was observed in the plasma concentrations of oral fexofenadine and midazolam and intravenous bosentan between the novobiocin pre-treatment and control group. These findings indicate that novobiocin at a dose of 3 mg/kg selectively inhibits intestinal Bcrp-mediated efflux without affecting P-gp,
CYP3A and hepatic Oatp in rats.

To evaluate the in vivo effects of novobiocin on Bcrp-mediated efflux in rats, we used sulfasalazine as a probe substrate. A previous study has demonstrated that the AUC of sulfasalazine after oral and intravenous administration in Bcrp knockout rats was markedly increased by 23.1- and 3.7-fold, respectively, compared with that observed in the wild-type (Mitschke et al., 2008), indicating that sulfasalazine is an ideal in vivo probe for studying intestinal and systemic Bcrp-mediated efflux transport in rats. In the present study, 3 mg/kg novobiocin drastically increased the exposure of oral sulfasalazine without affecting the systemic clearance of the substrate, indicating that it selectively inhibited intestinal Bcrp in rats in vivo. Although novobiocin was well absorbed after oral administration, the unbound Cmax was low owing to the high protein binding in the plasma, resulting in limited effects on systemic Bcrp. Considering that the ABCG2 421C>A variant mainly affects the intestinal absorption of BCRP substrates, rat pharmacokinetic studies, using novobiocin as an intestinal Bcrp inhibitor, could estimate the effects of the ABCG2 421C>A variant on the intestinal absorption of drug candidates in humans. In fact, the increased AUC of oral sulfasalazine
(3.2-fold) observed in the present study was comparable with that observed in humans carrying the ABCG2 421C>A variant.
To investigate the effects of novobiocin on P-gp-mediated efflux, fexofenadine was used as a probe of P-gp. Fexofenadine is a well-known substrate of P-gp, and a majority of the administrated dose is recovered unchanged in urine and faeces after its intravenous administration in rats (Cvetkovic et al., 1999; Kamath et al., 2005); therefore, it is a desirable probe for studying P-gp-mediated transport in vivo. The AUC of orally administered fexofenadine reportedly increases by approximately 7-fold upon co-administration of zosuquidar, a P-gp inhibitor, in rats (Matsuda et al., 2013). In the present, the plasma concentrations of fexofenadine following oral administration was not altered by novobiocin

pre-treatment, confirming that novobiocin has little in vivo inhibitory effect on P-gp activity in rats.
The inhibitory effect of curcumin on intestinal CYP3A activity in vivo has been reported in rats (Kadono et al., 2014). To compare novobiocin with curcumin, we elucidated the in vitro inhibitory effectiveness of novobiocin on rat CYP3A2 activity using a cDNA- expressed system. The result confirmed that novobiocin has little inhibitory effect on rat CYP3A2 activity. We investigated the actual in vivo effect of novobiocin on the pharmacokinetics of orally administered midazolam, a typical CYP3A substrate, in rats. The FaFg (Fa: fraction absorbed; Fg: intestinal availability) of midazolam was reported to be approximately 0.3 in rats (Yamamoto et al., 2018); midazolam can be a suitable probe of CYP3A-mediated intestinal metabolism, in addition to hepatic metabolism, in rats since the drug has sufficiently high membrane permeability to achieve complete absorption (Fa = 1.0). An in vivo pharmacokinetic study demonstrated that 3 mg/kg novobiocin did not significantly affect the plasma concentrations of orally administered midazolam in rats; the inhibitor exerts limited in vivo effects on CYP3A-mediated intestinal and hepatic metabolism, which is consistent with the result of the in vitro inhibition study.
Similar to curcumin, novobiocin is reported to weakly inhibit the in vitro activity of human OATP1B1 and OATP1B3 by approximately 60% and 85%, respectively, at 20 μmol/L in cells expressing the transporters (Bedada et al., 2016). Therefore, we investigated the in vivo effects of novobiocin on rat hepatic Oatp activity using bosentan as a probe substrate. Bosentan is a substrate of Oatp, and cyclosporin A, a well-known Oatp inhibitor,
dramatically increases the AUC of intravenously administered bosentan (Treiber et al., 2004); hence, the compound is an ideal probe substrate of hepatic Oatp-mediated uptake in rats. In the present study, no significant difference was observed in the plasma concentrations of bosentan after intravenous dosing between the two groups; therefore, the in vivo effects of

novobiocin on hepatic Oatp activity were considered negligible in rats owing to the low unbound Cmax of the inhibitor.
A previous study has demonstrated the inhibitory effects of novobiocin on human organic anion transporter (OAT) 1 and OAT3 activities with IC50 values of 34.76 and 4.99 μmol/L, respectively (Duan and You 2009), in addition to human OATP; however, the low unbound plasma concentration of oral novobiocin could hardly cause inhibition of Oat in rats in vivo.
In conclusion, reduced doses of novobiocin can selectively inhibit intestinal Bcrp without affecting the P-gp, CYP3A and hepatic Oatp activities in rats. Taken together, the findings of the present study suggest that novobiocin helps in estimating the net contribution of intestinal Bcrp to the pharmacokinetics of drug candidates.

 

 

Declaration of interest

The authors report no declarations of interest.

References

Bedada SK, Sudhakar YA, Neerati P. (2014). Resveratrol enhances the bioavailability of fexofenadine in healthy human male volunteers: involvement of P-glycoprotein inhibition. J Bioequivalence Bioavailab, 6, 158-63.

Bedada SK, Yellu NR, Neerati P. (2016). Effect of resveratrol on the pharmacokinetics of fexofenadine in rats: Involvement of P-glycoprotein inhibition. Pharmacol Rep, 68, 338-43.

Cvetkovic M, Leake B, Fromm MF et al. (1999). OATP and P-glycoprotein transporters mediate the cellular uptake and excretion of fexofenadine. Drug Metab Dispos, 27, 866-71.

Dahan A, Amidon GL. (2009). Grapefruit juice and its constituents augment colchicine intestinal absorption: potential hazardous interaction and the role of p-glycoprotein. Pharm Res, 26, 883-92.

Duan P, You G. (2009). Novobiocin is a potent inhibitor for human organic anion transporters. Drug Metab Dispos, 37, 1203-10.

Gertz M, Harrison A, Houston JB et al. (2010). Prediction of human intestinal first-pass metabolism of 25 CYP3A substrates from in vitro clearance and permeability data. Drug Metab Dispos, 38, 1147-58.

Gotanda K, Tokumoto T, Hirota T et al. (2015). Sulfasalazine disposition in a subject with 376C>T (nonsense mutation) and 421C>A variants in the ABCG2 gene. Br J Clin Pharmacol, 80, 1236-7.

Kadono K, Koakutsu A, Naritomi Y et al. (2014). Comparison of intestinal metabolism of CYP3A substrates between rats and humans: application of portal-systemic concentration difference method. Xenobiotica, 44, 511-21.

Kamath AV, Yao M, Zhang Y et al. (2005). Effect of fruit juices on the oral bioavailability of fexofenadine in rats. J Pharm Sci, 94, 233-9.

Keskitalo JE, Zolk O, Fromm MF et al. (2009). ABCG2 polymorphism markedly affects the pharmacokinetics of atorvastatin and rosuvastatin. Clin Pharmacol Ther, 86, 197-203.

Kunimatsu S, Mizuno T, Fukudo M et al. (2013). Effect of P-glycoprotein and breast cancer resistance protein inhibition on the pharmacokinetics of sunitinib in rats. Drug Metab Dispos, 41, 1592-7.

Lau YY, Chen YH, Liu TT et al. (2004). Evaluation of a novel in vitro Caco-2 hepatocyte hybrid system for predicting in vivo oral bioavailability. Drug Metab Dispos, 32, 937- 42.

Lee HK, Hu M, Lui SSh et al. (2013). Effects of polymorphisms in ABCG2, SLCO1B1, SLC10A1 and CYP2C9/19 on plasma concentrations of rosuvastatin and lipid response in Chinese patients. Pharmacogenomics, 14, 1283-94.

Li R, Barton HA. (2018). Explaining Ethnic Variability of Transporter Substrate Pharmacokinetics in Healthy Asian and Caucasian Subjects with Allele Frequencies of OATP1B1 and BCRP: A Mechanistic Modeling Analysis. Clin Pharmacokinet, 57, 491-503.

Matsuda Y, Konno Y, Hashimoto T et al. (2013). In vivo assessment of the impact of efflux transporter on oral drug absorption using portal vein-cannulated rats. Drug Metab Dispos, 41, 1514-21.

Mitschke D, Reichel A, Fricker G et al. (2008). Characterization of cytochrome P450 protein expression along the entire length of the intestine of male and female rats. Drug Metab Dispos, 36, 1039-45.

Pade V, Stavchansky S. (1998). Link between drug absorption solubility and permeability measurements in Caco-2 cells. J Pharm Sci, 87, 1604-7.

Peng Y, Yadava P, Heikkinen AT et al. (2014). Applications of a 7-day Caco-2 cell model in drug discovery and development. Eur J Pharm Sci, 56, 120-30.

Soko ND, Chimusa E, Masimirembwa C et al. (2019). An African-specific profile of pharmacogene variants for rosuvastatin plasma variability: limited role for SLCO1B1 c.521T>C and ABCG2 c.421A>C. Pharmacogenomics J, 19, 240-8.

Su Y, Hu P, Lee SH et al. (2007). Using novobiocin as a specific inhibitor of breast cancer resistant protein to assess the role of transporter in the absorption and disposition of topotecan. J Pharm Pharm Sci, 10, 519-36.

Tachibana T, Kato M, Sugiyama Y. (2012). Prediction of nonlinear intestinal absorption of CYP3A4 and P-glycoprotein substrates from their in vitro Km values. Pharm Res, 29, 651-68.

Treiber A, Schneiter R, Delahaye S et al. (2004). Inhibition of organic anion transporting polypeptide-mediated hepatic uptake is the major determinant in the pharmacokinetic interaction between bosentan and cyclosporin A in the rat. THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS, 308, 1121–9.

Wan Z, Wang G, Li T et al. (2015). Marked Alteration of Rosuvastatin Pharmacokinetics in Healthy Chinese with ABCG2 34G>A and 421C>A Homozygote or Compound Heterozygote. J Pharmacol Exp Ther, 354, 310-5.

Wang X, Morris ME. (2007). Effects of the flavonoid chrysin on nitrofurantoin pharmacokinetics in rats: potential involvement of ABCG2. Drug Metab Dispos, 35, 268-74.

Yamamoto S, Kosugi Y, Hirabayashi H et al. (2018). Impact of P-Glycoprotein on intestinal absorption of an Inhibitor of apoptosis protein antagonist in rats: mechanisms of nonlinear pharmacokinetics and food effects. Pharm Res, 35, 190.

Zhang W, Yu BN, He YJ et al. (2006). Role of BCRP 421C>A polymorphism on rosuvastatin pharmacokinetics in healthy Chinese males. Clin Chim Acta, 373, 99- 103.

Zhou Q, Ruan ZR, Yuan H et al. (2013). ABCB1 gene polymorphisms, ABCB1 haplotypes and ABCG2 c.421c > A are determinants of inter-subject variability in rosuvastatin pharmacokinetics. Pharmazie, 68, 129-34.

Zhou X, Zhang F, Chen C et al. (2017). Impact of curcumin on the pharmacokinetics of rosuvastatin in rats and dogs based on the conjugated metabolites. Xenobiotica, 47, 267-75.

 
Table 1. Pharmacokinetic parameters of orally (5 mg/kg) and intravenously (1 mg/kg) administered sulfasalazine with or without novobiocin pre-treatment (3 mg/kg) in rats.

Compound Control + Novobiocin

Sulfasalazine (5 mg/kg oral)
AUC0–inf (ng·h/mL) 269 ± 123 865 ± 199*
Cmax (ng/mL) 117 ± 30 688 ± 101***
tmax (h) 0.25 ± 0 0.25 ± 0
t1/2 (h) 0.95 ± 0.15 0.96 ± 0.39 Sulfasalazine (1 mg/kg intravenous)
AUC0–inf (ng·h/mL) 1728 ± 205 1735 ± 376
CLtot (mL/min/kg) 9.7 ± 1.1 9.9 ± 2.0
Vdss (L/kg) 0.24 ± 0.04 0.30 ± 0.04
t1/2 (h) 1.4 ± 0.1 1.1 ± 0.1
Data are presented as the mean ± SD for three rats. *p < 0.05, ***p < 0.001 versus the control group.

Table 2. Pharmacokinetic parameters of orally administered fexofenadine (5 mg/kg) and midazolam (1 mg/kg), and intravenously administered bosentan (1 mg/kg) with or without novobiocin pre-treatment (3 mg/kg) in rats.
Compound

Fexofenadine (5 mg/kg oral)
Control + Novobiocin

AUC0–inf (ng·h/mL) 10.5 ± 4.5 11.7 ± 2.6
Cmax (ng/mL) 3.4 ± 1.8 3.7 ± 0.7
tmax (h) 0.33 ± 0.14 0.33 ± 0.14
t1/2 (h) 2.6 ± 0.6 2.1 ± 0.7 Midazolam (1 mg/kg oral)
AUC0–inf (ng·h/mL) 4.6 ± 4.5 4.4 ± 3.4
Cmax (ng/mL) 3.4 ± 2.5 4.2 ± 4.4
tmax (h) 0.28 ± 0.11 0.21 ± 0.04
t1/2 (h) 0.86 ± 0.23 0.75 ± 0.12 Bosentan (1 mg/kg intravenous)
AUC0–inf (ng·h/mL) 1008 ± 165 1045 ± 295
CLtot (mL/min/kg) 16.9 ± 3.1 16.8 ± 4.8
Vdss (L/kg) 1.5 ± 0.4 1.5 ± 0.1
t1/2 (h) 1.9 ± 0.4 1.9 ± 0.3
Data are presented as the mean ± SD for three to six rats.
There were no significant differences between the control and novobiocin pre-treatment groups.

Figure Captions

 
Figure 1. Plasma concentration–time profiles of orally (5 mg/kg, A) and intravenously (1 mg/kg, B) administered sulfasalazine with or without novobiocin pre-treatment (3 mg/kg) in rats. Data are presented as the mean ± SD for three rats.

 
Figure 2. Plasma concentration–time profiles of orally administered fexofenadine (5 mg/kg) with or without novobiocin pre-treatment (3 mg/kg) in rats. Data are presented as the mean ± SD for three rats.

 
Figure 3. Plasma concentration–time profiles of orally administered midazolam (1 mg/kg) with or without novobiocin pre-treatment (3 mg/kg) in rats. Data are presented as the mean ± SD for six rats.
Figure 4. Plasma concentration–time profiles of intravenously administered bosentan (1 mg/kg) with or without novobiocin pre-treatment (3 mg/kg) in rats. Data are presented as the mean ± SD for three rats.
Figure 5. Unbound plasma concentration–time profiles of orally administered novobiocin (3 mg/kg) after administration of a substrate in rats. Data are presented as the mean ± SD for three rats.

Figure 1

 

 

 

 

 

 

 

 

 

 

 

 

Figure 2

 

 

 
Accepted
Manuscript

Figure 3

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Figure 4

 

 

Accepted
Manuscript

Figure 5

 

 

 

 

 

 
Manuscript
Accepted

Leave a Reply

Your email address will not be published. Required fields are marked *

*

You may use these HTML tags and attributes: <a href="" title=""> <abbr title=""> <acronym title=""> <b> <blockquote cite=""> <cite> <code> <del datetime=""> <em> <i> <q cite=""> <strike> <strong>