Skip to main content
Advertisement

Main menu

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET

User menu

  • My alerts
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Drug Metabolism & Disposition
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET
  • My alerts
  • Log in
  • Log out
  • My Cart
Drug Metabolism & Disposition

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Visit dmd on Facebook
  • Follow dmd on Twitter
  • Follow ASPET on LinkedIn
OtherShort Communication

Modest Effect of Impaired P-glycoprotein on the Plasma Concentrations of Fexofenadine, Quinidine, and Loperamide following Oral Administration in Collies

Yoshiaki Kitamura, Hisao Koto, Shinobu Matsuura, Takeshi Kawabata, Hiroshi Tsuchiya, Hiroyuki Kusuhara, Hajime Tsujimoto and Yuichi Sugiyama
Drug Metabolism and Disposition May 2008, 36 (5) 807-810; DOI: https://doi.org/10.1124/dmd.107.017624
Yoshiaki Kitamura
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Hisao Koto
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Shinobu Matsuura
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Takeshi Kawabata
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Hiroshi Tsuchiya
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Hiroyuki Kusuhara
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Hajime Tsujimoto
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Yuichi Sugiyama
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF
Loading

Abstract

P-glycoprotein (P-gp), encoded by the multidrug resistance 1 gene (MDR1/ABCB1), exhibits very broad substrate specificity and plays important roles in drug disposition. The purpose of the present study was to examine the effect of impaired P-gp activity on the plasma pharmacokinetics of P-gp substrates in collies with or without homozygous mutant alleles producing truncated P-gp. Three therapeutic agents, fexofenadine (0.1 mg/kg), quinidine (0.1 mg/kg), and loperamide (0.01 mg/kg), were simultaneously given orally, and their plasma concentration-time profiles were determined. The plasma concentrations of these drugs tended to be higher in dogs with the homozygous mutated allele. The Cmax was 53.9 ± 13.1 and 90.7 ± 23.1 ng/ml for fexofenadine, 16.5 ± 3.4 and 20.0 ± 7.9 ng/ml for quinidine, and 80.8 ± 9.0 and 101 ± 15 pg/ml for loperamide, and the AUC0–8 was 263 ± 62 and 435 ± 95 ng·h/ml for fexofenadine, 54.5 ± 11.5 and 75.7 ± 21.8 ng·h/ml for quinidine, and 467 ± 85 and 556 ± 91 pg·h/ml for loperamide in homozygous wild-type and homozygous mutated dogs, respectively. Only the plasma concentration differences of fexofenadine at 4 to 8 h after oral administration were statistically significant. This result suggests that P-gp limits the intestinal absorption of fexofenadine in dogs. Collies with the Mdr1 mutation will be useful for examining the effect of P-gp on the oral availability of drugs.

The multidrug resistance 1 gene (MDR1/ABCB1) encodes a 170-kDa transmembrane protein pump, called P-glycoprotein (P-gp), belonging to the ATP-binding cassette (ABC) superfamily of membrane transporters. An increasing number of studies have shown that P-gp exhibits broad substrate specificity, and a number of structurally unrelated drugs are substrates for P-gp (Choudhuri and Klaassen, 2006). P-gp is expressed in the apical membrane of the epithelial cells in the liver, intestine, and kidney and in brain capillary endothelial cells. A number of studies, particularly using Mdr1a-/- and Mdr1a/1b-/- mice, have demonstrated that P-gp limits oral availability and penetration into the brain and mediates biliary and urinary excretion by actively extruding xenobiotics into the adjacent luminal space (Chen et al., 2003; Mizuno et al., 2003). For instance, Mdr1a/1b-/- mice exhibited greater plasma concentrations than wild-type mice after oral administration of paclitaxel (Sparreboom et al., 1997), fexofenadine (Tahara et al., 2005), cyclosporin A (Lee et al., 2005), etoposide (Allen et al., 2003), and vinblastine (Ogihara et al., 2006). For daunomycin, loperamide, quinidine, ritonavir, and verapamil, the permeability-surface area product in the small intestine increased more than three times in Mdr1a/1b-/- mice (Adachi et al., 2003). In humans, P-gp has been suggested to limit the oral availability of some drugs. Coadministration of quinidine increased the plasma concentration of digoxin following oral administration (Pedersen et al., 1983; Fromm et al., 1999). Repeated rifampicin treatment decreased the plasma levels of digoxin and talinolol (Greiner et al., 1999; Westphal et al., 2000). A single dose of St. John's wort increased the plasma concentration of fexofenadine, whereas long-term treatment reversed the changes in fexofenadine disposition (Wang et al., 2002; Dresser et al., 2003). These changes have been accounted for by modulation of P-gp activity in the small intestine, inhibition, or induction. Apart from these drugs, the effect of P-gp on drug absorption in humans remains under question since some clinically important drugs have been developed as oral formulations even though they are P-gp substrates (Lin and Yamazaki, 2003).

A subpopulation of collies is extremely sensitive to ivermectin, which is used extensively in veterinary medicine to treat and control infections caused by nematode and arthropod parasites. Sequence analysis of Mdr1 cDNA from ivermectin-sensitive dogs identified a 4-base-pair deletion, which causes a frame-shift mutation generating several stop codons that result in a severely truncated, nonfunctional protein (Mealey et al., 2001). The frequency of the mutant allele is about as high as 60% in collies (Hugnet et al., 2004; Neff et al., 2004; Kawabata et al., 2005). Since dogs (Canis familiaris) have one Mdr1 gene corresponding to human MDR1, collies with homozygous mutant alleles would be a good animal model for examining the importance of P-gp in the pharmacokinetics of drugs in relatively large animals. In particular, in pharmaceutical companies, dogs are commonly used preclinically as a convenient animal species for testing oral dosage forms because of their anatomical similarity to humans (Lin, 1995), their ability to ingest human-scale dosage forms, and because they are easy to handle.

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

Structures of fexofenadine, quinidine, and loperamide.

The purpose of the present study was to examine the effect of impaired P-gp activity on the plasma pharmacokinetics of P-gp substrates in collies. Three therapeutic agents, fexofenadine, quinidine, and loperamide (Fig. 1), were selected. Fexofenadine exhibited a 6-fold increase in oral availability (Tahara et al., 2005), and quinidine and loperamide exhibited an 8- and 3-fold increase in the permeability-surface area product determined by in situ intestinal perfusion in Mdr1a/1b-/- mice (Adachi et al., 2003). The three drugs were simultaneously administered orally, and the plasma concentration-time profiles were determined.

Materials and Methods

Animals. Eleven collies were used in this study (Table 1). The animals were aged between 14 and 68 months and had a body weight of between 18 and 28 kg. Of the 11 dogs, 5 (1 male and 4 females) were homozygous for the wild-type Mdr1 allele, and 6 (2 males and 4 females) were homozygous for the mutant allele.

View this table:
  • View inline
  • View popup
TABLE 1

Information on collies used in this study

Study Design. After a 12-h fast, each animal received 0.5 mg/kg famotidine (Progogue for injection, 20 mg; Nichi-Iko Pharmaceutical Co., Ltd., Toyama, Japan) intramuscularly to reduce the interindividual variability of the gastrointestinal tract by controlling gastric acid secretion. One hour after famotidine administration, the dogs were orally given a capsule filled with a pulverized commercial preparation of fexofenadine (Allegra 60-mg tablets; sanofi-aventis, Bridgewater, NJ), quinidine sulfate (Quinidine Sulfate tablets; Merck Seiyaku Ltd., Osaka, Japan), and loperamide (Lopemin capsules, 1 mg; Janssen Pharmaceutical, Tokyo, Japan) at a dose of 0.1 mg/kg, 0.1 mg/kg, and 0.01 mg/kg, respectively. Venous blood samples (2 ml each) were collected in tubes containing EDTA-2K at 0.5, 1, 2, 4, 6, and 8 h after oral administration. Plasma samples were separated by centrifugation and stored at -30°C until analysis.

In this study, three drugs were simultaneously given orally to the dogs. If the concentration of these drugs in the enterocytes exceeds the Ki value for P-gp inhibition, a drug-drug interaction may reduce P-gp function. A prediction criterion for intestinal enzyme-mediated drug-drug interactions was proposed based on the fact that, if dose/Ki is below 2.76 liters, there have been no reports of an obvious drug-drug interaction in humans (Tachibana et al., 2006). Considering that the in vitro IC50 values for inhibition of digoxin transport in Caco-2 cells are above 100 μM for fexofenadine (Cvetkovic et al., 1999), 2.2 μM for quinidine (Choo et al., 2000), and 2.5 μM for loperamide (Wandel et al., 2002), the dose/Ki values were calculated to be below 0.04, 2.8, and 0.17 liters, respectively, taking the IC50 values as the Ki values. This suggests that the doses used in this study are low enough to avoid any P-gp based drug-drug interaction in the intestine, although species differences between dogs and humans cannot be excluded. Following the same logic, autosaturation of intestinal P-gp activity may not take place.

Quantification of Drug Concentrations in Plasma. For fexofenadine and quinidine quantification, a 10-μl plasma sample was mixed vigorously with 20 μl of acetonitrile and deproteinized by centrifugation. Then, 25 μl of supernatant was added to 100 μl of water and subjected to liquid chromatography-mass spectrometry. To determine the loperamide concentration, a 500-μl plasma sample was added to a mixture of 0.5 M Na2CO3 (50 μl) and ethyl acetate (1000 μl) and mixed vigorously. After centrifugation, 800 μl of the organic layer were collected, dried in a centrifugal concentrator (TOMY, Tokyo, Japan), and dissolved in 40 μl of dimethylsulfoxide. Then, 10-μl aliquots were subjected to liquid chromatography-mass spectrometry.

The high-performance liquid chromatography separation involved a Waters Alliance 2695 Separation Module (Waters, Milford, MA) with an L-column ODS (2.1 × 150 mm, 5 μm; Chemicals Evaluation and Research Institute, Tokyo, Japan). The composition of the mobile phase was acetonitrile/0.05% formic acid (26:74, 7:93, and 29:71 for fexofenadine, quinidine, and loperamide, respectively). The flow rate was 0.3 ml/min. Mass spectra were determined using a Micromass ZQ2000 mass spectrometer (Waters) with an electrospray ionization interface in the selected ion-monitoring mode using positive ions, m/z 502.5, 325.3, and 477.4, respectively.

Pharmacokinetic Analysis. Pharmacokinetic parameters were calculated using noncompartmental analysis. The peak plasma concentration (Cmax) was determined by inspection of individual plasma concentration-time curves. The elimination half-life (t1/2) was calculated using the equation t1/2 = ln(2)/ke, where ke is the elimination rate constant calculated from the slope of the terminal portion of the log-transformed plasma concentration-time curve. For fexofenadine and quinidine, the last 3 points, which gave a determination coefficient (r2) > 0.8, were considered the terminal phase. However, in the case of loperamide, we could not determine ke because of lack of clarity in its terminal phase.

The area under the concentration-time curve was calculated by the linear trapezoidal rule up to 8 h (AUC0–8) and then extrapolated to infinity (AUC0-∞) using the elimination rate constant. The area under the first-moment curve (AUMC0-∞) and mean residence time (MRT) were calculated using the following equations: MathMathMathMath where Cp, 8h is the plasma concentration 8 h after oral administration.

Statistical Analysis. All data represent five or six experiments and are expressed as the mean ± S.E. Any statistical significance in the differences of the means was assessed using a two-tailed Student's t test. P < 0.05 was considered statistically significant.

Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

Time profiles of the plasma concentrations of fexofenadine (A), quinidine (B), and loperamide (C) after a single oral administration in wild-type and Mdr1 mutated collies. Fexofenadine (0.1 mg/kg), quinidine (0.1 mg/kg), and loperamide (0.01 mg/kg) were given simultaneously to wild-type (closed symbols) and Mdr1 mutated (open symbols) collies by a single oral administration. Each point represents the mean ± S.E. (n = 5 or 6). *, P < 0.05, significantly different from wild-type dogs.

Results and Discussion

A dog has one Mdr1 gene, and the homozygous mutant alleles of the Mdr1 gene totally impair P-gp activity in the body. The present study examined the effect of impaired P-gp activity on the oral availability of P-gp substrates. The plasma concentration-time profiles of fexofenadine, quinidine, and loperamide following simultaneous oral administration are shown in Fig. 2. P-gp deficient mutant collies generally exhibited a higher plasma concentration than normal dogs. However, the difference was marginal for quinidine and loperamide, but fexofenadine showed a significant increase at 4 to 8 h following administration (Fig. 2), although such increase was not great enough to produce a statistically significant change in the pharmacokinetic parameters (Table 2). Since the contribution of P-gp to the elimination of fexofenadine from the systemic circulation is minimal in mice (Tahara et al., 2005), the increase in the plasma concentration of fexofenadine in the mutant collies may be ascribed to the impaired intestinal efflux. A similar elimination half-life in normal and mutant collies also supports the absence of the effect of functional impairment of P-gp on the systemic elimination of fexofenadine. Comparison of the plasma concentration-time profile following oral and intravenous administration will be required to confirm definitely that the higher plasma levels of fexofenadine in mutant dogs are strictly accounted for by increased intestinal absorption. On average, the change was at most 2-fold, which was smaller than that observed in mice (Tahara et al., 2005). It is possible that the effect of P-gp on drug absorption is not as important as that observed in mice.

View this table:
  • View inline
  • View popup
TABLE 2

Pharmacokinetic parameters of fexofenadine, quinidine, and loperamide after a single oral administration in wild-type and Mdr1-mutated collies The details of pharmacokinetic analyses are described in Materials and Methods. Each value represents the mean ± S.E. of five or six independent experiments. P values were obtained by a two-tailed Student's t test.

It should be noted that three drugs were simultaneously administered to dogs in this study. We selected the doses of the drugs tested to be much smaller than the clinical doses considering the fact that drug-drug interaction involving P-gp does not occur in clinical studies when the inhibitor dose is smaller than 50 mg/body weight (approximately 0.7 mg/kg) (Tachibana T, Kato M, Sugiyama Y, unpublished observations). If these doses are still enough to saturate P-gp mediated efflux, the effect of functional impairment of P-gp will be underestimated. Further studies are necessary to elucidate the importance of P-gp in limiting oral availability of drugs at linear dose ranges and clinical doses for elucidating the functional importance of P-gp in the small intestine and its clinical relevance.

The present study highlights the usefulness of collies with a hereditary deficiency in P-gp for examining the effect of P-gp on the oral absorption of drugs. Exhaustive comparison of transport activities of 3300 P-gp substrates by human and mouse P-gp exhibits a good linear correlation, suggesting that they exhibit similar substrate specificities (Feng et al., 2007). Considering closer sequence similarity between dog and human MDR1 (90% identity at the amino acid level), dog studies will provide clues for how to evaluate the impact of P-gp on the disposition of test compounds in humans. However, we must pay attention to the species difference when the results of dog studies are extrapolated to humans, since it is true that some P-gp substrates were outliers of the correlation of the transport activities by human and dog P-gp (Takeuchi et al., 2006).

In addition to the intestinal absorption and hepatobiliary and urinary excretion, P-gp plays an indispensable role in limiting brain penetration in the blood-brain barrier. Recently, the P-gp function at the blood-brain barrier was evaluated in monkeys using a specific P-gp inhibitor (PSC833) by a positron emission tomography technique (Lee et al., 2006). However, the magnitude of the increase observed in PSC833-treated monkeys was not as high as that observed in P-gp knockout mice. This may be explained by incomplete inhibition of P-gp activity by PSC833 (Kusuhara et al., 1997). Mdr1 mutated collies will be useful for in vivo evaluation of the role of P-gp in the brain penetration of test compounds in relatively large animals.

In conclusion, this result suggests that P-gp limits the intestinal absorption of fexofenadine in dogs. Collies with the Mdr1 mutation will be useful for examining the effect of P-gp on the oral availability of drugs.

Acknowledgments

We thank Chizuko Kawabata, Mitsuru Kawabata, Uiko Yoshino, and Toru Yoshino for allowing us to use their breeding or pet collies in this study.

Footnotes

  • This work was supported by a Grant-in-Aid for Scientific Research (A) (KAKENHI 17209005) (for Y.S.) and by a Grant-in-Aid for Scientific Research (B) (KAKENHI 18390046) (for H.K.) from Japan Society for the Promotion of Science.

  • Article, publication date, and citation information can be found at http://dmd.aspetjournals.org.

  • doi:10.1124/dmd.107.017624.

  • ABBREVIATIONS: ABC, ATP-binding cassette; MDR/Mdr, multidrug resistance; P-gp, P-glycoprotein; AUC, area under the concentration-time curve; AUMC, area under the first-moment curve; MRT, mean residence time.

    • Received July 11, 2007.
    • Accepted February 14, 2008.
  • The American Society for Pharmacology and Experimental Therapeutics

References

  1. ↵
    Adachi Y, Suzuki H, and Sugiyama Y (2003) Quantitative evaluation of the function of small intestinal P-glycoprotein: comparative studies between in situ and in vitro. Pharm Res 20: 1163-1169.
    OpenUrlCrossRefPubMed
  2. ↵
    Allen JD, Van Dort SC, Buitelaar M, van Tellingen O, and Schinkel AH (2003) Mouse breast cancer resistance protein (Bcrp1/Abcg2) mediates etoposide resistance and transport, but etoposide oral availability is limited primarily by P-glycoprotein. Cancer Res 63: 1339-1344.
    OpenUrlAbstract/FREE Full Text
  3. ↵
    Chen C, Liu X, and Smith BJ (2003) Utility of Mdr1-gene deficient mice in assessing the impact of P-glycoprotein on pharmacokinetics and pharmacodynamics in drug discovery and development. Curr Drug Metab 4: 272-291.
    OpenUrlCrossRefPubMed
  4. ↵
    Choo EF, Leake B, Wandel C, Imamura H, Wood AJ, Wilkinson GR, and Kim RB (2000) Pharmacological inhibition of P-glycoprotein transport enhances the distribution of HIV-1 protease inhibitors into brain and testes. Drug Metab Dispos 28: 655-660.
    OpenUrlAbstract/FREE Full Text
  5. ↵
    Choudhuri S and Klaassen CD (2006) Structure, function, expression, genomic organization, and single nucleotide polymorphisms of human ABCB1 (MDR1), ABCC (MRP), and ABCG2 (BCRP) efflux transporters. Int J Toxicol 25: 231-259.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    Cvetkovic M, Leake B, Fromm MF, Wilkinson GR, and Kim RB (1999) OATP and P-glycoprotein transporters mediate the cellular uptake and excretion of fexofenadine. Drug Metab Dispos 27: 866-871.
    OpenUrlAbstract/FREE Full Text
  7. ↵
    Dresser GK, Schwarz UI, Wilkinson GR, and Kim RB (2003) Coordinate induction of both cytochrome P4503A and MDR1 by St John's wort in healthy subjects. Clin Pharmacol Ther 73: 41-50.
    OpenUrlCrossRefPubMed
  8. Feng B, Mills J, Davidson R, Mireles R, Janiszewski J, Troutman M, and de Morais S (2008) In vitro P-glycoprotein assays to predict the in vivo interactions of P-glycoprotein with drugs in the central nervous system. Drug Metab Dispos 36: 268-275.
    OpenUrlAbstract/FREE Full Text
  9. ↵
    Fromm MF, Kim RB, Stein CM, Wilkinson GR, and Roden DM (1999) Inhibition of P-glycoprotein-mediated drug transport: a unifying mechanism to explain the interaction between digoxin and quinidine. Circulation 99: 552-557.
    OpenUrlAbstract/FREE Full Text
  10. ↵
    Greiner B, Eichelbaum M, Fritz P, Kreichgauer HP, von Richter O, Zundler J, and Kroemer HK (1999) The role of intestinal P-glycoprotein in the interaction of digoxin and rifampin. J Clin Invest 104: 147-153.
    OpenUrlCrossRefPubMed
  11. ↵
    Hugnet C, Bentjen SA, and Mealey KL (2004) Frequency of the mutant MDR1 allele associated with multidrug sensitivity in a sample of collies from France. J Vet Pharmacol Ther 27: 227-229.
    OpenUrlCrossRefPubMed
  12. ↵
    Kawabata A, Momoi Y, Inoue-Murayama M, and Iwasaki T (2005) Canine mdr1 gene mutation in Japan. J Vet Med Sci 67: 1103-1107.
    OpenUrlCrossRefPubMed
  13. ↵
    Kusuhara H, Suzuki H, Terasaki T, Kakee A, Lemaire M, Sugiyama Y (1997) P-glycoprotein mediates the efflux of quinidine across the blood-brain barrier. J Pharmacol Exp Ther 283: 574-580.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    Lee YJ, Chung SJ, and Shim CK (2005) Limited role of P-glycoprotein in the intestinal absorption of cyclosporin A. Biol Pharm Bull 28: 760-763.
    OpenUrlCrossRefPubMed
  15. ↵
    Lee YJ, Maeda J, Kusuhara H, Okauchi T, Inaji M, Nagai Y, Obayashi S, Nakao R, Suzuki K, Sugiyama Y, et al. (2006) In vivo evaluation of P-glycoprotein function at the blood-brain barrier in nonhuman primates using [11C]verapamil. J Pharmacol Exp Ther 316: 647-653.
    OpenUrlAbstract/FREE Full Text
  16. ↵
    Lin JH (1995) Species similarities and differences in pharmacokinetics. Drug Metab Dispos 23: 1008-1021.
    OpenUrlAbstract
  17. ↵
    Lin JH and Yamazaki M (2003) Clinical relevance of P-glycoprotein in drug therapy. Drug Metab Rev 35: 417-454.
    OpenUrlCrossRefPubMed
  18. ↵
    Mealey KL, Bentjen SA, Gay JM, and Cantor GH (2001) Ivermectin sensitivity in collies is associated with a deletion mutation of the mdr1 gene. Pharmacogenetics 11: 727-733.
    OpenUrlCrossRefPubMed
  19. ↵
    Mizuno N, Niwa T, Yotsumoto Y, and Sugiyama Y (2003) Impact of drug transporter studies on drug discovery and development. Pharmacol Rev 55: 425-461.
    OpenUrlAbstract/FREE Full Text
  20. ↵
    Neff MW, Robertson KR, Wong AK, Safra N, Broman KW, Slatkin M, Mealey KL, and Pedersen NC (2004) Breed distribution and history of canine mdr1–1Delta, a pharmacogenetic mutation that marks the emergence of breeds from the collie lineage. Proc Natl Acad Sci U S A 101: 11725-11730.
    OpenUrlAbstract/FREE Full Text
  21. ↵
    Ogihara T, Kamiya M, Ozawa M, Fujita T, Yamamoto A, Yamashita S, Ohnishi S, and Isomura Y (2006) What kinds of substrates show P-glycoprotein-dependent intestinal absorption? Comparison of verapamil with vinblastine. Drug Metab Pharmacokinet 21: 238-244.
    OpenUrlCrossRefPubMed
  22. ↵
    Pedersen KE, Christiansen BD, Klitgaard NA, and Nielsen-Kudsk F (1983) Effect of quinidine on digoxin bioavailability. Eur J Clin Pharmacol 24: 41-47.
    OpenUrlCrossRefPubMed
  23. ↵
    Sparreboom A, van Asperen J, Mayer U, Schinkel AH, Smit JW, Meijer DK, Borst P, Nooijen WJ, Beijnen JH, and van Tellingen O (1997) Limited oral bioavailability and active epithelial excretion of paclitaxel (Taxol) caused by P-glycoprotein in the intestine. Proc Natl Acad Sci U S A 94: 2031-2035.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    Tachibana T, Kato M, Watanabe T, Mitsui T, and Sugiyama Y (2006) Prediction of intestinal enzyme mediated drug-drug interaction and nonlinear intestinal first-pass metabolism, in 21st JSSX Annual Meeting; 2006 Nov 29–Dec 1; Tokyo, Japan.
  25. ↵
    Tahara H, Kusuhara H, Fuse E, and Sugiyama Y (2005) P-glycoprotein plays a major role in the efflux of fexofenadine in the small intestine and blood-brain barrier, but only a limited role in its biliary excretion. Drug Metab Dispos 33: 963-968.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    Takeuchi T, Yoshitomi S, Higuchi T, Ikemoto K, Niwa S, Ebihara T, Katoh M, Yokoi T, and Asahi S (2006) Establishment and characterization of the transformants stably-expressing MDR1 derived from various animal species in LLC-PK1. Pharm Res 23: 1460-1472.
    OpenUrlCrossRefPubMed
  27. ↵
    Wandel C, Kim R, Wood M, and Wood A (2002) Interaction of morphine, fentanyl, sufentanil, alfentanil, and loperamide with the efflux drug transporter P-glycoprotein. Anesthesiology 96: 913-920.
    OpenUrlCrossRefPubMed
  28. ↵
    Wang Z, Hamman MA, Huang SM, Lesko LJ, and Hall SD (2002) Effect of St John's wort on the pharmacokinetics of fexofenadine. Clin Pharmacol Ther 71: 414-420.
    OpenUrlCrossRefPubMed
  29. ↵
    Westphal K, Weinbrenner A, Zschiesche M, Franke G, Knoke M, Oertel R, Fritz P, von Richter O, Warzok R, Hachenberg T, et al. (2000) Induction of P-glycoprotein by rifampin increases intestinal secretion of talinolol in human beings: a new type of drug/drug interaction. Clin Pharmacol Ther 68: 345-355.
    OpenUrlCrossRefPubMed
View Abstract
PreviousNext
Back to top

In this issue

Drug Metabolism and Disposition: 36 (5)
Drug Metabolism and Disposition
Vol. 36, Issue 5
1 May 2008
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Back Matter (PDF)
  • Editorial Board (PDF)
  • Front Matter (PDF)
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Drug Metabolism & Disposition article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Modest Effect of Impaired P-glycoprotein on the Plasma Concentrations of Fexofenadine, Quinidine, and Loperamide following Oral Administration in Collies
(Your Name) has forwarded a page to you from Drug Metabolism & Disposition
(Your Name) thought you would be interested in this article in Drug Metabolism & Disposition.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
OtherShort Communication

Modest Effect of Impaired P-glycoprotein on the Plasma Concentrations of Fexofenadine, Quinidine, and Loperamide following Oral Administration in Collies

Yoshiaki Kitamura, Hisao Koto, Shinobu Matsuura, Takeshi Kawabata, Hiroshi Tsuchiya, Hiroyuki Kusuhara, Hajime Tsujimoto and Yuichi Sugiyama
Drug Metabolism and Disposition May 1, 2008, 36 (5) 807-810; DOI: https://doi.org/10.1124/dmd.107.017624

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
OtherShort Communication

Modest Effect of Impaired P-glycoprotein on the Plasma Concentrations of Fexofenadine, Quinidine, and Loperamide following Oral Administration in Collies

Yoshiaki Kitamura, Hisao Koto, Shinobu Matsuura, Takeshi Kawabata, Hiroshi Tsuchiya, Hiroyuki Kusuhara, Hajime Tsujimoto and Yuichi Sugiyama
Drug Metabolism and Disposition May 1, 2008, 36 (5) 807-810; DOI: https://doi.org/10.1124/dmd.107.017624
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results and Discussion
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Preincubation Effects on Inhibition of OCT1 by CsA
  • Carbamazepine Metabolite and Hypersensitivity Reactions
  • SULT4A1 Preserves Mitochondrial Function
Show more SHORT COMMUNICATIONS

Similar Articles

  • Home
  • Alerts
Facebook   Twitter   LinkedIn   RSS

Navigate

  • Current Issue
  • Fast Forward by date
  • Fast Forward by section
  • Latest Articles
  • Archive
  • Search for Articles
  • Feedback
  • ASPET

More Information

  • About DMD
  • Editorial Board
  • Instructions to Authors
  • Submit a Manuscript
  • Customized Alerts
  • RSS Feeds
  • Subscriptions
  • Permissions
  • Terms & Conditions of Use

ASPET's Other Journals

  • Journal of Pharmacology and Experimental Therapeutics
  • Molecular Pharmacology
  • Pharmacological Reviews
  • Pharmacology Research & Perspectives
ISSN 1521-009X (Online)

Copyright © 2021 by the American Society for Pharmacology and Experimental Therapeutics