메뉴 건너뛰기




Volumn 24, Issue 1, 2009, Pages 37-52

In vitro-in vivo extrapolation of transporter-mediated clearance in the liver and kidney

Author keywords

Hepatic elimination; Hepatocytes; In vitro in vivo extrapolation; Kidney slices; Tubular secretion

Indexed keywords

ATORVASTATIN; BOSENTAN; BREAST CANCER RESISTANCE PROTEIN; CARNITINE; CERIVASTATIN; DIGOXIN; EBASTINE; FAMOTIDINE; FEXOFENADINE; FUROSEMIDE; MEMBRANE PROTEIN; MULTIDRUG RESISTANCE PROTEIN; MULTIDRUG RESISTANCE PROTEIN 1; NATEGLINIDE; OCTAMER TRANSCRIPTION FACTOR; OLMESARTAN; ORGANIC ANION TRANSPORTER; ORGANIC CATION TRANSPORTER; OUABAIN; PITAVASTATIN; PRAVASTATIN; QUINIDINE; RANITIDINE; REPAGLINIDE; ROSUVASTATIN; TELMISARTAN; THIONEINE; UNINDEXED DRUG; VALSARTAN; VERAPAMIL;

EID: 66449128354     PISSN: 13474367     EISSN: 18800920     Source Type: Journal    
DOI: 10.2133/dmpk.24.37     Document Type: Review
Times cited : (196)

References (113)
  • 1
    • 56149110125 scopus 로고    scopus 로고
    • Impact of genetic polymorphisms of transporters on the pharmacokinetic, pharmacodynamic and toxicological properties of anionic drugs
    • Maeda, K. and Sugiyama, Y.: Impact of genetic polymorphisms of transporters on the pharmacokinetic, pharmacodynamic and toxicological properties of anionic drugs. Drug Metab Pharmacokinet, 23, 223-235 (2008).
    • (2008) Drug Metab Pharmacokinet , vol.23 , pp. 223-235
    • Maeda, K.1    Sugiyama, Y.2
  • 2
    • 85031350436 scopus 로고    scopus 로고
    • Giacomini, K. M. and Sugiyama, Y.: Membrane transporters and drug response. In L. L. Brunton, J. S. Lazo, K. L. Parker, L. S. Goodman and A. G. Gilman (ed.). The Pharmacological Basis of Therapeutics 11th edition, Goodman & Gilman's 205, pp41-70.
    • Giacomini, K. M. and Sugiyama, Y.: Membrane transporters and drug response. In L. L. Brunton, J. S. Lazo, K. L. Parker, L. S. Goodman and A. G. Gilman (ed.). The Pharmacological Basis of Therapeutics 11th edition, Goodman & Gilman's 205, pp41-70.
  • 4
    • 0035209269 scopus 로고    scopus 로고
    • Mechanisms and clinical implications of renal drug excretion
    • Masereeuw, R. and Russel, F. G.: Mechanisms and clinical implications of renal drug excretion. Drug Metab. Rev., 33, 299-351 (2001).
    • (2001) Drug Metab. Rev , vol.33 , pp. 299-351
    • Masereeuw, R.1    Russel, F.G.2
  • 5
    • 13844314148 scopus 로고    scopus 로고
    • Evaluation of drug-drug interaction in the hepatobiliary and renal transport of drugs
    • Shitara, Y., Sato, H. and Sugiyama, Y.: Evaluation of drug-drug interaction in the hepatobiliary and renal transport of drugs. Annu. Rev. Pharmacol. Toxicol., 45, 689-723 (2005).
    • (2005) Annu. Rev. Pharmacol. Toxicol , vol.45 , pp. 689-723
    • Shitara, Y.1    Sato, H.2    Sugiyama, Y.3
  • 6
    • 0032733974 scopus 로고    scopus 로고
    • Prediction of human clearance of twenty-nine drugs from hepatic microsomal intrinsic clearance data: An examination of in vitro half-life approach and nonspecific binding to microsomes
    • Obach, R. S.: Prediction of human clearance of twenty-nine drugs from hepatic microsomal intrinsic clearance data: An examination of in vitro half-life approach and nonspecific binding to microsomes. Drug Metab. Dispos., 27, 1350-1359 (1999).
    • (1999) Drug Metab. Dispos , vol.27 , pp. 1350-1359
    • Obach, R.S.1
  • 7
    • 85031358631 scopus 로고    scopus 로고
    • Physiologically based pharmacokinetic modeling to predict transportermediated clearance and distribution of pravastatin in humans
    • in press
    • Watanabe, T., Kusuhara, H., Maeda, K., et al.: Physiologically based pharmacokinetic modeling to predict transportermediated clearance and distribution of pravastatin in humans. J. Pharmacol. Exp. Ther., (in press).
    • J. Pharmacol. Exp. Ther
    • Watanabe, T.1    Kusuhara, H.2    Maeda, K.3
  • 8
    • 0035012079 scopus 로고    scopus 로고
    • Inhibition of biliary excretion of methotrexate by probenecid in rats: Quantitative prediction of interaction from in vitro data
    • Ueda, K., Kato, Y., Komatsu, K., et al.: Inhibition of biliary excretion of methotrexate by probenecid in rats: quantitative prediction of interaction from in vitro data. J. Pharmacol. Exp. Ther., 297, 1036-1043 (2001).
    • (2001) J. Pharmacol. Exp. Ther , vol.297 , pp. 1036-1043
    • Ueda, K.1    Kato, Y.2    Komatsu, K.3
  • 9
    • 54349088976 scopus 로고    scopus 로고
    • Increasing systemic exposure of methotrexate by active efflux mediated by multidrug resistance-associated protein 3 (Mrp3/Abcc3)
    • Kitamura, Y.,Hirouchi, M., Kusuhara, H., et al.: Increasing systemic exposure of methotrexate by active efflux mediated by multidrug resistance-associated protein 3 (Mrp3/Abcc3). J. Pharmacol. Exp. Ther., (2008).
    • (2008) J. Pharmacol. Exp. Ther
    • Kitamura, Y.1    Hirouchi, M.2    Kusuhara, H.3
  • 10
    • 33748042671 scopus 로고    scopus 로고
    • Pharmacokinetic and pharmacodynamic alterations of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors: Drug-drug interactions and interindividual differences in transporter and metabolic enzyme functions
    • Shitara, Y. and Sugiyama, Y.: Pharmacokinetic and pharmacodynamic alterations of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors: drug-drug interactions and interindividual differences in transporter and metabolic enzyme functions. Pharmacol. Ther., 112, 71-105 (2006).
    • (2006) Pharmacol. Ther , vol.112 , pp. 71-105
    • Shitara, Y.1    Sugiyama, Y.2
  • 11
    • 0030937636 scopus 로고    scopus 로고
    • Prediction of in vivo drug metabolism in the human liver from in vitro metabolism data
    • Iwatsubo, T., Hirota, N., Ooie, T., et al.: Prediction of in vivo drug metabolism in the human liver from in vitro metabolism data. Pharmacol. Ther., 73, 147-171 (1997).
    • (1997) Pharmacol. Ther , vol.73 , pp. 147-171
    • Iwatsubo, T.1    Hirota, N.2    Ooie, T.3
  • 12
    • 0027447887 scopus 로고
    • Comparison of the hepatic uptake clearances of fifteen drugs with a wide range of membrane permeabilities in isolated rat hepatocytes and perfused rat livers
    • Miyauchi, S., Sawada, Y., Iga, T., et al.: Comparison of the hepatic uptake clearances of fifteen drugs with a wide range of membrane permeabilities in isolated rat hepatocytes and perfused rat livers. Pharm Res, 10, 434-440 (1993).
    • (1993) Pharm Res , vol.10 , pp. 434-440
    • Miyauchi, S.1    Sawada, Y.2    Iga, T.3
  • 13
    • 0030862073 scopus 로고    scopus 로고
    • Biliary excretion of pravastatin in rats: Contribution of the excretion pathway mediated by canalicular multispecific organic anion transporter
    • Yamazaki, M., Akiyama, S., Ni'inuma, K., et al., Biliary excretion of pravastatin in rats: contribution of the excretion pathway mediated by canalicular multispecific organic anion transporter. Drug Metab. Dispos., 25, 1123-1129 (1997).
    • (1997) Drug Metab. Dispos , vol.25 , pp. 1123-1129
    • Yamazaki, M.1    Akiyama, S.2    Ni'inuma, K.3
  • 14
    • 0032589267 scopus 로고    scopus 로고
    • Use of Ca2+ modulation to evaluate biliary excretion in sandwich-cultured rat hepatocytes
    • Liu, X., LeCluyse, E. L., Brouwer, K. R., et al., Use of Ca2+ modulation to evaluate biliary excretion in sandwich-cultured rat hepatocytes. J. Pharmacol. Exp. Ther., 289, 1592-1599 (1999).
    • (1999) J. Pharmacol. Exp. Ther , vol.289 , pp. 1592-1599
    • Liu, X.1    LeCluyse, E.L.2    Brouwer, K.R.3
  • 15
    • 53849092394 scopus 로고    scopus 로고
    • In vitro biliary clearance of angiotensin II receptor blockers and 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors in sandwich-cultured rat hepatocytes: Comparison with in vivo biliary clearance
    • Abe, K., Bridges, A. S., Yue, W., et al.: In vitro biliary clearance of angiotensin II receptor blockers and 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors in sandwich-cultured rat hepatocytes: comparison with in vivo biliary clearance. J. Pharmacol. Exp. Ther., 326, 983-990 (2008).
    • (2008) J. Pharmacol. Exp. Ther , vol.326 , pp. 983-990
    • Abe, K.1    Bridges, A.S.2    Yue, W.3
  • 16
    • 46449095993 scopus 로고    scopus 로고
    • Effect of plasma protein binding on in vitro-in vivo correlation of biliary excretion of drugs evaluated by sandwich-cultured rat hepatocytes
    • Fukuda, H., Ohashi, R., Tsuda-Tsukimoto, M., et al.] Effect of plasma protein binding on in vitro-in vivo correlation of biliary excretion of drugs evaluated by sandwich-cultured rat hepatocytes. Drug Metab. Dispos., 36, 1275-1282 (2008).
    • (2008) Drug Metab. Dispos , vol.36 , pp. 1275-1282
    • Fukuda, H.1    Ohashi, R.2    Tsuda-Tsukimoto, M.3
  • 17
    • 0030849120 scopus 로고    scopus 로고
    • An evaluation of the integration of pharmacokinetic and pharmacodynamic principles in clinical drug development. Experience within Hoffmann La Roche
    • Reigner, B. G., Williams, P. E., Patel, I. H., et al.: An evaluation of the integration of pharmacokinetic and pharmacodynamic principles in clinical drug development. Experience within Hoffmann La Roche. Clin. Pharmacokinet., 33, 142-152 (1997).
    • (1997) Clin. Pharmacokinet , vol.33 , pp. 142-152
    • Reigner, B.G.1    Williams, P.E.2    Patel, I.H.3
  • 18
    • 0028818229 scopus 로고
    • Species similarities and differences in pharmacokinetics
    • Lin, J. H.: Species similarities and differences in pharmacokinetics. Drug Metab. Dispos., 23, 1008-1021 (1995).
    • (1995) Drug Metab. Dispos , vol.23 , pp. 1008-1021
    • Lin, J.H.1
  • 19
    • 0032553486 scopus 로고    scopus 로고
    • Interspecies scaling of renally secreted drugs
    • Mahmood, I.: Interspecies scaling of renally secreted drugs. Life Sci., 63, 2365-2371 (1998).
    • (1998) Life Sci , vol.63 , pp. 2365-2371
    • Mahmood, I.1
  • 20
    • 33947426344 scopus 로고    scopus 로고
    • Characterization of the uptake of organic anion transporter (OAT) 1 and OAT3 substrates by human kidney slices
    • Nozaki, Y., Kusuhara, H., Kondo, T., et al.: Characterization of the uptake of organic anion transporter (OAT) 1 and OAT3 substrates by human kidney slices. J. Pharmacol. Exp. Ther., 321, 362-369 (2007).
    • (2007) J. Pharmacol. Exp. Ther , vol.321 , pp. 362-369
    • Nozaki, Y.1    Kusuhara, H.2    Kondo, T.3
  • 21
    • 34548103547 scopus 로고    scopus 로고
    • Species difference in the inhibitory effect of nonsteroidal anti-inflammatory drugs on the uptake of methotrexate by human kidney slices
    • Nozaki, Y., Kusuhara, H., Kondo, T., et al.: Species difference in the inhibitory effect of nonsteroidal anti-inflammatory drugs on the uptake of methotrexate by human kidney slices. J. Pharmacol. Exp. Ther., 322, 1162-1170 (2007).
    • (2007) J. Pharmacol. Exp. Ther , vol.322 , pp. 1162-1170
    • Nozaki, Y.1    Kusuhara, H.2    Kondo, T.3
  • 22
    • 0036175127 scopus 로고    scopus 로고
    • Functional involvement of rat organic anion transporter 3 (rOat3; Slc22a8) in the renal uptake of organic anions
    • Hasegawa, M., Kusuhara, H., Sugiyama, D., et al.: Functional involvement of rat organic anion transporter 3 (rOat3; Slc22a8) in the renal uptake of organic anions. J. Pharmacol. Exp. Ther., 300, 746-753 (2002).
    • (2002) J. Pharmacol. Exp. Ther , vol.300 , pp. 746-753
    • Hasegawa, M.1    Kusuhara, H.2    Sugiyama, D.3
  • 23
    • 0038137530 scopus 로고    scopus 로고
    • Contribution of organic anion transporters to the renal uptake of anionic compounds and nucleoside derivatives in rat
    • Hasegawa, M., Kusuhara, H., Endou, H., et al.: Contribution of organic anion transporters to the renal uptake of anionic compounds and nucleoside derivatives in rat. J. Pharmacol. Exp. Ther., 305, 1087-1097 (2003).
    • (2003) J. Pharmacol. Exp. Ther , vol.305 , pp. 1087-1097
    • Hasegawa, M.1    Kusuhara, H.2    Endou, H.3
  • 24
    • 0023700224 scopus 로고
    • Determination of brush border membrane vesicle orientation using monoclonal antibodies recognizing extracytoplasmic and cytoplasmic domains of neutral endopeptidase-24.11
    • Venien, C., Aubry M., Crine P., et al.: Determination of brush border membrane vesicle orientation using monoclonal antibodies recognizing extracytoplasmic and cytoplasmic domains of neutral endopeptidase-24.11. Anal. Biochem., 174, 325-330 (1988).
    • (1988) Anal. Biochem , vol.174 , pp. 325-330
    • Venien, C.1    Aubry, M.2    Crine, P.3
  • 25
    • 0017819206 scopus 로고
    • Studies on the orientation of brush-border membrane vesicles
    • Haase, W., Schafer, A., Murer, H., et al.: Studies on the orientation of brush-border membrane vesicles. Biochem. J., 172, 57-62 (1978).
    • (1978) Biochem. J , vol.172 , pp. 57-62
    • Haase, W.1    Schafer, A.2    Murer, H.3
  • 26
    • 0035006408 scopus 로고    scopus 로고
    • LST-2, a human liver-specific organic anion transporter, determines methotrexate sensitivity in gastrointestinal cancers
    • Abe, T., Unno, M., Onogawa, T., et al.: LST-2, a human liver-specific organic anion transporter, determines methotrexate sensitivity in gastrointestinal cancers. Gastroenterology, 120, 1689-1699 (2001).
    • (2001) Gastroenterology , vol.120 , pp. 1689-1699
    • Abe, T.1    Unno, M.2    Onogawa, T.3
  • 27
    • 4644364576 scopus 로고    scopus 로고
    • Contribution of OATP2 (OATP1B1) and OATP8 (OATP1B3) to the hepatic uptake of pitavastatin in humans
    • Hirano, M., Maeda, K., Shitara, Y., et al.: Contribution of OATP2 (OATP1B1) and OATP8 (OATP1B3) to the hepatic uptake of pitavastatin in humans. J. Pharmacol. Exp. Ther., 311, 139-146 (2004).
    • (2004) J. Pharmacol. Exp. Ther , vol.311 , pp. 139-146
    • Hirano, M.1    Maeda, K.2    Shitara, Y.3
  • 28
    • 33745253947 scopus 로고    scopus 로고
    • Predominant contribution of OATP1B3 to the hepatic uptake of telmisartan, an angiotensin II receptor antagonist, in humans
    • Ishiguro, N., Maeda, K., Kishimoto, W., et al.: Predominant contribution of OATP1B3 to the hepatic uptake of telmisartan, an angiotensin II receptor antagonist, in humans. Drug Metab. Dispos., 34, 1109-1115 (2006).
    • (2006) Drug Metab. Dispos , vol.34 , pp. 1109-1115
    • Ishiguro, N.1    Maeda, K.2    Kishimoto, W.3
  • 29
    • 33751096467 scopus 로고    scopus 로고
    • SLCO1B1 polymorphism markedly affects the pharmacokinetics of simvastatin acid
    • Pasanen, M. K., Neuvonen, M., Neuvonen, P. J. et al.: SLCO1B1 polymorphism markedly affects the pharmacokinetics of simvastatin acid. Pharmacogenet. Genomics., 16, 873-879 (2006).
    • (2006) Pharmacogenet. Genomics , vol.16 , pp. 873-879
    • Pasanen, M.K.1    Neuvonen, M.2    Neuvonen, P.J.3
  • 30
    • 20444466590 scopus 로고    scopus 로고
    • Polymorphic organic anion transporting polypeptide 1B1 is a major determinant of repaglinide pharmacokinetics
    • Niemi, M., Backman, J. T., Kajosaari, L. I., et al.: Polymorphic organic anion transporting polypeptide 1B1 is a major determinant of repaglinide pharmacokinetics. Clin. Pharmacol. Ther., 77, 468-478 (2005).
    • (2005) Clin. Pharmacol. Ther , vol.77 , pp. 468-478
    • Niemi, M.1    Backman, J.T.2    Kajosaari, L.I.3
  • 31
    • 39149108768 scopus 로고    scopus 로고
    • Different effects of SLCO1B1 polymorphism on the pharmacokinetics and pharmacodynamics of repaglinide and nateglinide
    • Kalliokoski, A., Neuvonen, M., Neuvonen, P. J., et al.: Different effects of SLCO1B1 polymorphism on the pharmacokinetics and pharmacodynamics of repaglinide and nateglinide. J. Clin. Pharmacol., 48, 311-321 (2008).
    • (2008) J. Clin. Pharmacol , vol.48 , pp. 311-321
    • Kalliokoski, A.1    Neuvonen, M.2    Neuvonen, P.J.3
  • 32
    • 36148953896 scopus 로고    scopus 로고
    • Different effects of SLCO1B1 polymorphism on the pharmacokinetics of atorvastatin and rosuvastatin
    • Pasanen, M. K., Fredrikson, H., Neuvonen, P. J., et al.: Different effects of SLCO1B1 polymorphism on the pharmacokinetics of atorvastatin and rosuvastatin. Clin. Pharmacol. Ther., 82, 726-733 (2007).
    • (2007) Clin. Pharmacol. Ther , vol.82 , pp. 726-733
    • Pasanen, M.K.1    Fredrikson, H.2    Neuvonen, P.J.3
  • 33
    • 0037310871 scopus 로고    scopus 로고
    • Inhibition of transportermediated hepatic uptake as a mechanism for drug-drug interaction between cerivastatin and cyclosporin A
    • Shitara, Y., Itoh, T., Sato. H., et al.: Inhibition of transportermediated hepatic uptake as a mechanism for drug-drug interaction between cerivastatin and cyclosporin A. J. Pharmacol. Exp. Ther., 304, 610-616 (2003).
    • (2003) J. Pharmacol. Exp. Ther , vol.304 , pp. 610-616
    • Shitara, Y.1    Itoh, T.2    Sato, H.3
  • 34
    • 33846571368 scopus 로고    scopus 로고
    • effect of OATP1B transporter inhibition on the pharmacokinetics of atorvastatin in healthy volunteers
    • Lau, Y. Y., Huang, Y., Frassetto, L., et al.: effect of OATP1B transporter inhibition on the pharmacokinetics of atorvastatin in healthy volunteers. Clin. Pharmacol. Ther., 81, 194-204 (2007).
    • (2007) Clin. Pharmacol. Ther , vol.81 , pp. 194-204
    • Lau, Y.Y.1    Huang, Y.2    Frassetto, L.3
  • 35
    • 35448946484 scopus 로고    scopus 로고
    • SLCO1B1 (OATP1B1, an Uptake Transporter) and ABCG2 (BCRP, an Efflux Transporter) Variant Alleles and Pharmacokinetics of Pitavastatin in Healthy Volunteers
    • Ieiri, I., Suwannakul, S., Maeda, K., et al.: SLCO1B1 (OATP1B1, an Uptake Transporter) and ABCG2 (BCRP, an Efflux Transporter) Variant Alleles and Pharmacokinetics of Pitavastatin in Healthy Volunteers. Clin. Pharmacol. Ther., 82, 541-547 (2007).
    • (2007) Clin. Pharmacol. Ther , vol.82 , pp. 541-547
    • Ieiri, I.1    Suwannakul, S.2    Maeda, K.3
  • 36
    • 0038209381 scopus 로고    scopus 로고
    • Polymorphisms of OATP-C (SLC21A6) and OAT3 (SLC22A8) genes: Consequences for pravastatin pharmacokinetics
    • Nishizato, Y., Ieiri, I., Suzuki, H., et al.: Polymorphisms of OATP-C (SLC21A6) and OAT3 (SLC22A8) genes: consequences for pravastatin pharmacokinetics. Clin. Pharmacol. Ther., 73, 554-565 (2003).
    • (2003) Clin. Pharmacol. Ther , vol.73 , pp. 554-565
    • Nishizato, Y.1    Ieiri, I.2    Suzuki, H.3
  • 37
    • 33646205286 scopus 로고    scopus 로고
    • Effects of organic anion transporting polypeptide 1B1 haplotype on pharmacokinetics of pravastatin, valsartan, and temocapril
    • Maeda, K., Ieiri, I., Yasuda, K., et al.: Effects of organic anion transporting polypeptide 1B1 haplotype on pharmacokinetics of pravastatin, valsartan, and temocapril. Clin. Pharmacol. Ther., 79, 427-439 (2006).
    • (2006) Clin. Pharmacol. Ther , vol.79 , pp. 427-439
    • Maeda, K.1    Ieiri, I.2    Yasuda, K.3
  • 38
    • 49949104757 scopus 로고    scopus 로고
    • SLCO1B1 variants and statin-induced myopathy--a genomewide study
    • Link, E., Parish, S., Armitage, J., et al.: SLCO1B1 variants and statin-induced myopathy--a genomewide study. N. Engl. J. Med., 359, 789-799 (2008).
    • (2008) N. Engl. J. Med , vol.359 , pp. 789-799
    • Link, E.1    Parish, S.2    Armitage, J.3
  • 39
    • 18744388590 scopus 로고    scopus 로고
    • Acute effect of pravastatin on cholesterol synthesis are associated with SLCO1B1 (encoding OATP1B1) haplotype *17
    • Niemi, M., Neuvonen, P. J., Hofmann, U., et al.: Acute effect of pravastatin on cholesterol synthesis are associated with SLCO1B1 (encoding OATP1B1) haplotype *17. Pharmacogenet. Genomics., 15, 303-309 (2005).
    • (2005) Pharmacogenet. Genomics , vol.15 , pp. 303-309
    • Niemi, M.1    Neuvonen, P.J.2    Hofmann, U.3
  • 40
    • 33748645938 scopus 로고    scopus 로고
    • Pharmacogenetic determinants of variability in lipid-lowering response to pravastatin therapy
    • Takane, H., Miyata, M., Burioka, N., et al.: Pharmacogenetic determinants of variability in lipid-lowering response to pravastatin therapy. J. Hum. Genet., 51, 822-826 (2006).
    • (2006) J. Hum. Genet , vol.51 , pp. 822-826
    • Takane, H.1    Miyata, M.2    Burioka, N.3
  • 41
    • 33646198187 scopus 로고    scopus 로고
    • Impact of the SLCO1B1 polymorphism on the pharmacokinetics and lipid-lowering efficacy of multiple-dose pravastatin
    • Igel, M., Arnold, K. A., Niemi, M., et al.: Impact of the SLCO1B1 polymorphism on the pharmacokinetics and lipid-lowering efficacy of multiple-dose pravastatin. Clin. Pharmacol. Ther., 79, 419-426 (2006).
    • (2006) Clin. Pharmacol. Ther , vol.79 , pp. 419-426
    • Igel, M.1    Arnold, K.A.2    Niemi, M.3
  • 42
    • 25144450046 scopus 로고    scopus 로고
    • Apical/basolateral surface expression of drug transporters and its role in vectorial drug transport
    • Ito, K., Suzuki, H., Horie, T., et al.: Apical/basolateral surface expression of drug transporters and its role in vectorial drug transport. Pharm. Res., 22, 1559-1577 (2005).
    • (2005) Pharm. Res , vol.22 , pp. 1559-1577
    • Ito, K.1    Suzuki, H.2    Horie, T.3
  • 43
    • 33644847684 scopus 로고    scopus 로고
    • Transporters as a determinant of drug clearance and tissue distribution
    • Shitara, Y., Horie, T. and Sugiyama, Y., Transporters as a determinant of drug clearance and tissue distribution. Eur. J. Pharm. Sci., 27, 425-446 (2006).
    • (2006) Eur. J. Pharm. Sci , vol.27 , pp. 425-446
    • Shitara, Y.1    Horie, T.2    Sugiyama, Y.3
  • 44
    • 0032434272 scopus 로고    scopus 로고
    • Excretion of GSSG and glutathione conjugates mediated by MRP1 and cMOAT/MRP2
    • Suzuki, H. and Sugiyama, Y.: Excretion of GSSG and glutathione conjugates mediated by MRP1 and cMOAT/MRP2. Semin. Liver. Dis., 18, 359-376 (1998).
    • (1998) Semin. Liver. Dis , vol.18 , pp. 359-376
    • Suzuki, H.1    Sugiyama, Y.2
  • 45
    • 34748877360 scopus 로고    scopus 로고
    • Involvement of Breast Cancer Resistance Protein (ABCG2) in the Biliary Excretion Mechanism of Fluoroquinolones
    • Ando, T., Kusuhara, H., Merino, G., et al.: Involvement of Breast Cancer Resistance Protein (ABCG2) in the Biliary Excretion Mechanism of Fluoroquinolones. Drug. Metab. Dispos., 35, 1873-1879 (2007).
    • (2007) Drug. Metab. Dispos , vol.35 , pp. 1873-1879
    • Ando, T.1    Kusuhara, H.2    Merino, G.3
  • 46
    • 23944475903 scopus 로고    scopus 로고
    • Involvement of BCRP (ABCG2) in the biliary excretion of pitavastatin
    • Hirano, M., Maeda, K., Matsushima, S., et al.: Involvement of BCRP (ABCG2) in the biliary excretion of pitavastatin. Mol. Pharmacol., 68, 800-807 (2005).
    • (2005) Mol. Pharmacol , vol.68 , pp. 800-807
    • Hirano, M.1    Maeda, K.2    Matsushima, S.3
  • 47
    • 4944225107 scopus 로고    scopus 로고
    • Prediction of in vivo biliary clearance from the in vitro transcellular transport of organic anions across a double-transfected Madin-Darby canine kidney II monolayer expressing both rat organic anion transporting polypeptide 4 and multidrug resistance associated protein 2
    • Sasaki, M., Suzuki, H., Aoki, J., et al.: Prediction of in vivo biliary clearance from the in vitro transcellular transport of organic anions across a double-transfected Madin-Darby canine kidney II monolayer expressing both rat organic anion transporting polypeptide 4 and multidrug resistance associated protein 2. Mol. Pharmacol., 66, 450-459 (2004).
    • (2004) Mol. Pharmacol , vol.66 , pp. 450-459
    • Sasaki, M.1    Suzuki, H.2    Aoki, J.3
  • 48
    • 0037155204 scopus 로고    scopus 로고
    • Transcellular transport of organic anions across a double-transfected Madin-Darby canine kidney II cell monolayer expressing both human organic anion-transporting polypeptide (OATP2/SLC21A6) and Multidrug resistance-associated protein 2 (MRP2/ABCC2)
    • Sasaki, M., Suzuki, H., Ito, K., et al.: Transcellular transport of organic anions across a double-transfected Madin-Darby canine kidney II cell monolayer expressing both human organic anion-transporting polypeptide (OATP2/SLC21A6) and Multidrug resistance-associated protein 2 (MRP2/ABCC2). J. Biol. Chem., 277, 6497-6503 (2002).
    • (2002) J. Biol. Chem , vol.277 , pp. 6497-6503
    • Sasaki, M.1    Suzuki, H.2    Ito, K.3
  • 49
    • 22944446686 scopus 로고    scopus 로고
    • 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
    • Matsushima, S., Maeda, K., Kondo, C., et al.: 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., 314, 1059-1067 (2005).
    • (2005) J. Pharmacol. Exp. Ther , vol.314 , pp. 1059-1067
    • Matsushima, S.1    Maeda, K.2    Kondo, C.3
  • 50
    • 0034753042 scopus 로고    scopus 로고
    • Vectorial transport by double-transfected cells expressing the human uptake transporter SLC21A8 and the apical export pump ABCC2
    • Cui, Y., Konig, J. and Keppler, D.: Vectorial transport by double-transfected cells expressing the human uptake transporter SLC21A8 and the apical export pump ABCC2. Mol. Pharmacol., 60, 934-943 (2001).
    • (2001) Mol. Pharmacol , vol.60 , pp. 934-943
    • Cui, Y.1    Konig, J.2    Keppler, D.3
  • 51
    • 29144523534 scopus 로고    scopus 로고
    • A human transporter protein that mediates the final excretion step for toxic organic cations
    • Otsuka, M., Matsumoto, T., Morimoto, R., et al.: A human transporter protein that mediates the final excretion step for toxic organic cations. Proc. Natl. Acad. Sci. USA, 102, 17923-17928 (2005).
    • (2005) Proc. Natl. Acad. Sci. USA , vol.102 , pp. 17923-17928
    • Otsuka, M.1    Matsumoto, T.2    Morimoto, R.3
  • 52
    • 38349085099 scopus 로고    scopus 로고
    • Effect of genetic variation in the organic cation transporter 1, OCT1, on metformin pharmacokinetics
    • Shu, Y., Brown, C., Castro, R. A., et al.: Effect of genetic variation in the organic cation transporter 1, OCT1, on metformin pharmacokinetics. Clin. Pharmacol. Ther., 83, 273-80 (2008).
    • (2008) Clin. Pharmacol. Ther , vol.83 , pp. 273-280
    • Shu, Y.1    Brown, C.2    Castro, R.A.3
  • 53
    • 21044435938 scopus 로고    scopus 로고
    • Mice lacking multidrug resistance protein 3 show altered morphine pharmacokinetics and morphine-6-glucuronide antinociception
    • Zelcer, N., van de Wetering, K., Hillebrand, M., et al.: Mice lacking multidrug resistance protein 3 show altered morphine pharmacokinetics and morphine-6-glucuronide antinociception. Proc. Natl. Acad. Sci. USA, 102, 7274-7279 (2005).
    • (2005) Proc. Natl. Acad. Sci. USA , vol.102 , pp. 7274-7279
    • Zelcer, N.1    van de Wetering, K.2    Hillebrand, M.3
  • 55
    • 42449122721 scopus 로고    scopus 로고
    • Impact of basolateral multidrug resistance-associated protein (Mrp) 3 and Mrp4 on the hepatobiliary disposition of fexofenadine in perfused mouse livers
    • Tian, X., Swift, B., Zamek-Gliszczynski, M. J., et al.: Impact of basolateral multidrug resistance-associated protein (Mrp) 3 and Mrp4 on the hepatobiliary disposition of fexofenadine in perfused mouse livers. Drug Metab. Dispos., 36, 911-915 (2008).
    • (2008) Drug Metab. Dispos , vol.36 , pp. 911-915
    • Tian, X.1    Swift, B.2    Zamek-Gliszczynski, M.J.3
  • 56
    • 0036205460 scopus 로고    scopus 로고
    • Gene expression levels and immunolocalization of organic ion transporters in the human kidney
    • Motohashi, H., Sakurai, Y., Saito, H., et al.: Gene expression levels and immunolocalization of organic ion transporters in the human kidney. J. Am. Soc. Nephrol., 13, 866-874 (2002).
    • (2002) J. Am. Soc. Nephrol , vol.13 , pp. 866-874
    • Motohashi, H.1    Sakurai, Y.2    Saito, H.3
  • 57
    • 12144288563 scopus 로고    scopus 로고
    • Isolation and characterization of a digoxin transporter and its rat homologue expressed in the kidney
    • Mikkaichi, T., Suzuki, T., Onogawa, T., et al.: Isolation and characterization of a digoxin transporter and its rat homologue expressed in the kidney. Proc. Natl. Acad, Sci, USA, 101, 3569-3574 (2004).
    • (2004) Proc. Natl. Acad, Sci, USA , vol.101 , pp. 3569-3574
    • Mikkaichi, T.1    Suzuki, T.2    Onogawa, T.3
  • 58
    • 33645798634 scopus 로고    scopus 로고
    • Inhibition of oat3-mediated renal uptake as a mechanism for drug-drug interaction between fexofenadine and probenecid
    • Tahara, H., Kusuhara, H., Maeda, K., et al.: Inhibition of oat3-mediated renal uptake as a mechanism for drug-drug interaction between fexofenadine and probenecid. Drug Metab. Dispos., 34, 743-747 (2006).
    • (2006) Drug Metab. Dispos , vol.34 , pp. 743-747
    • Tahara, H.1    Kusuhara, H.2    Maeda, K.3
  • 59
    • 25644452793 scopus 로고    scopus 로고
    • A species difference in the transport activities of H2 receptor antagonists by rat and human renal organic anion and cation transporters
    • Tahara, H., Kusuhara, H., Endou, H., et al.: A species difference in the transport activities of H2 receptor antagonists by rat and human renal organic anion and cation transporters. J. Pharmacol. Exp. Ther., 315, 337-345 (2005).
    • (2005) J. Pharmacol. Exp. Ther , vol.315 , pp. 337-345
    • Tahara, H.1    Kusuhara, H.2    Endou, H.3
  • 60
    • 33646165531 scopus 로고    scopus 로고
    • Decreased renal organic anion secretion and plasma accumulation of endogenous organic anions in OAT1 knock-out mice
    • Eraly, S. A., Vallon, V., Vaughn, D. A., et al.: Decreased renal organic anion secretion and plasma accumulation of endogenous organic anions in OAT1 knock-out mice. J. Biol. Chem., 281, 5072-5083 (2006).
    • (2006) J. Biol. Chem , vol.281 , pp. 5072-5083
    • Eraly, S.A.1    Vallon, V.2    Vaughn, D.A.3
  • 61
    • 44949088665 scopus 로고    scopus 로고
    • Overlapping in vitro and in vivo specificities of the organic anion transporters OAT1 and OAT3 for loop and thiazide diuretics
    • Vallon, V., Rieg, T., Ahn, S. Y., et al.: Overlapping in vitro and in vivo specificities of the organic anion transporters OAT1 and OAT3 for loop and thiazide diuretics. Am. J. Physiol. Renal. Physiol., 294, F867-873 (2008).
    • (2008) Am. J. Physiol. Renal. Physiol , vol.294
    • Vallon, V.1    Rieg, T.2    Ahn, S.Y.3
  • 62
    • 45749158039 scopus 로고    scopus 로고
    • Organic anion transporter 3 (oat3/slc22a8) interacts with carboxyfluoroquinolones, and deletion increases systemic exposure to ciprofloxacin
    • Vanwert, A. L., Srimaroeng, C. and Sweet, D. H.: Organic anion transporter 3 (oat3/slc22a8) interacts with carboxyfluoroquinolones, and deletion increases systemic exposure to ciprofloxacin. Mol. Pharmacol.,, 122-131 (2008).
    • (2008) Mol. Pharmacol , vol.122-131
    • Vanwert, A.L.1    Srimaroeng, C.2    Sweet, D.H.3
  • 63
    • 35348889668 scopus 로고    scopus 로고
    • Organic anion transporter 3 (Oat3/Slc22a8) knockout mice exhibit altered clearance and distribution of penicillin G
    • Vanwert, A. L., Bailey, R. M. and Sweet, D. H.: Organic anion transporter 3 (Oat3/Slc22a8) knockout mice exhibit altered clearance and distribution of penicillin G. Am. J. Physiol. Renal. Physiol., 293, F1332-1341 (2007).
    • (2007) Am. J. Physiol. Renal. Physiol , vol.293
    • Vanwert, A.L.1    Bailey, R.M.2    Sweet, D.H.3
  • 64
    • 39149106149 scopus 로고    scopus 로고
    • Impaired clearance of methotrexate in organic anion transporter 3 (Slc22a8) knockout mice: A gender specific impact of reduced folates
    • VanWert, A. L. and Sweet, D. H.: Impaired clearance of methotrexate in organic anion transporter 3 (Slc22a8) knockout mice: a gender specific impact of reduced folates. Pharm Res, 25, 453-462 (2008).
    • (2008) Pharm Res , vol.25 , pp. 453-462
    • VanWert, A.L.1    Sweet, D.H.2
  • 65
    • 0034595094 scopus 로고    scopus 로고
    • p-aminohippuric acid transport at renal apical membrane mediated by human inorganic phosphate transporter NPT1
    • Uchino, H., Tamai, I., Yamashita, K., et al.: p-aminohippuric acid transport at renal apical membrane mediated by human inorganic phosphate transporter NPT1. Biochem. Biophys. Res. Commun., 270, 254-359 (2000).
    • (2000) Biochem. Biophys. Res. Commun , vol.270 , pp. 254-359
    • Uchino, H.1    Tamai, I.2    Yamashita, K.3
  • 66
    • 3543124167 scopus 로고    scopus 로고
    • The renal-specific transporter mediates facilitative transport of organic anions at the brush border membrane of mouse renal tubules
    • Imaoka, T., Kusuhara, H., Adachi-Akahane, S., et al.: The renal-specific transporter mediates facilitative transport of organic anions at the brush border membrane of mouse renal tubules. J. Am. Soc. Nephrol., 15, 2012-2022 (2004).
    • (2004) J. Am. Soc. Nephrol , vol.15 , pp. 2012-2022
    • Imaoka, T.1    Kusuhara, H.2    Adachi-Akahane, S.3
  • 67
    • 37549068911 scopus 로고    scopus 로고
    • The breast cancer resistance protein transporter ABCG2 is expressed in the human kidney proximal tubule apical membrane
    • Huls, M., Brown, C. D., Windass, A. S., et al.: The breast cancer resistance protein transporter ABCG2 is expressed in the human kidney proximal tubule apical membrane. Kidney. Int., 73, 220-225 (2008).
    • (2008) Kidney. Int , vol.73 , pp. 220-225
    • Huls, M.1    Brown, C.D.2    Windass, A.S.3
  • 68
    • 0036186107 scopus 로고    scopus 로고
    • The MRP4/ ABCC4 gene encodes a novel apical organic anion transporter in human kidney proximal tubules: Putative efflux pump for urinary cAMP and cGMP
    • van Aubel, R. A., Smeets, P. H., Peters, J. G., et al.: The MRP4/ ABCC4 gene encodes a novel apical organic anion transporter in human kidney proximal tubules: putative efflux pump for urinary cAMP and cGMP. J. Am. Soc. Nephrol., 13, 595-603 (2002).
    • (2002) J. Am. Soc. Nephrol , vol.13 , pp. 595-603
    • van Aubel, R.A.1    Smeets, P.H.2    Peters, J.G.3
  • 69
    • 0030832802 scopus 로고    scopus 로고
    • Expression of the conjugate export pump encoded by the mrp2 gene in the apical membrane of kidney proximal tubules
    • Schaub, T. P., Kartenbeck, J., Konig, J., et al.: Expression of the conjugate export pump encoded by the mrp2 gene in the apical membrane of kidney proximal tubules. J. Am. Soc. Nephrol., 8, 1213-1221 (1997).
    • (1997) J. Am. Soc. Nephrol , vol.8 , pp. 1213-1221
    • Schaub, T.P.1    Kartenbeck, J.2    Konig, J.3
  • 70
    • 34347327217 scopus 로고    scopus 로고
    • Involvement of MRP4 (ABCC4) in the luminal efflux of ceftizoxime and cefazolin in the kidney
    • Ci, L., Kusuhara, H., Adachi, M., et al.: Involvement of MRP4 (ABCC4) in the luminal efflux of ceftizoxime and cefazolin in the kidney. Mol. Pharmacol., 71, 1591-1597 (2007).
    • (2007) Mol. Pharmacol , vol.71 , pp. 1591-1597
    • Ci, L.1    Kusuhara, H.2    Adachi, M.3
  • 71
    • 33846019659 scopus 로고    scopus 로고
    • Multidrug resistance-associated protein 4 is involved in the urinary excretion of hydrochlorothiazide and furosemide
    • Hasegawa, M., Kusuhara, H., Adachi, M., et al.: Multidrug resistance-associated protein 4 is involved in the urinary excretion of hydrochlorothiazide and furosemide. J. Am. Soc. Nephrol., 18, 37-45 (2007).
    • (2007) J. Am. Soc. Nephrol , vol.18 , pp. 37-45
    • Hasegawa, M.1    Kusuhara, H.2    Adachi, M.3
  • 72
    • 33846420624 scopus 로고    scopus 로고
    • Functional involvement of multidrug resistance-associated protein 4 (MRP4/ABCC4) in the renal elimination of the antiviral drugs adefovir and tenofovir
    • Imaoka, T., Kusuhara, H., Adachi, M., et al.: Functional involvement of multidrug resistance-associated protein 4 (MRP4/ABCC4) in the renal elimination of the antiviral drugs adefovir and tenofovir. Mol. Pharmacol., 71, 619-627 (2007).
    • (2007) Mol. Pharmacol , vol.71 , pp. 619-627
    • Imaoka, T.1    Kusuhara, H.2    Adachi, M.3
  • 73
    • 41849115973 scopus 로고    scopus 로고
    • Methotrexate (MTX) pathway gene polymorphisms and their effects on MTX toxicity in Caucasian and African American patients with rheumatoid arthritis
    • Ranganathan, P., Culverhouse, R., Marsh, S., et al.: Methotrexate (MTX) pathway gene polymorphisms and their effects on MTX toxicity in Caucasian and African American patients with rheumatoid arthritis. J. Rheumatol., 35, 572-579 (2008).
    • (2008) J. Rheumatol , vol.35 , pp. 572-579
    • Ranganathan, P.1    Culverhouse, R.2    Marsh, S.3
  • 74
    • 33750958330 scopus 로고    scopus 로고
    • High-dose methotrexate in pediatric acute lymphoblastic leukemia: Impact of ABCC2 polymorphisms on plasma concentrations
    • Rau, T., Erney, B., Gores, R., et al.: High-dose methotrexate in pediatric acute lymphoblastic leukemia: impact of ABCC2 polymorphisms on plasma concentrations. Clin. Pharmacol. Ther., 80, 468-476 (2006).
    • (2006) Clin. Pharmacol. Ther , vol.80 , pp. 468-476
    • Rau, T.1    Erney, B.2    Gores, R.3
  • 75
    • 34249943356 scopus 로고    scopus 로고
    • Polyspecific organic cation transporters: Structure, function, physiological roles, and biopharmaceutical implications
    • Koepsell, H., Lips, K. and Volk, C.: Polyspecific organic cation transporters: structure, function, physiological roles, and biopharmaceutical implications. Pharm. Res., 24, 1227-1251 (2007).
    • (2007) Pharm. Res , vol.24 , pp. 1227-1251
    • Koepsell, H.1    Lips, K.2    Volk, C.3
  • 76
    • 0021255131 scopus 로고
    • Carrier-mediated transport systems of tetraethylammonium in rat renal brush-border and basolateral membrane vesicles
    • Takano, M., Inui, K., Okano, T., et al.: Carrier-mediated transport systems of tetraethylammonium in rat renal brush-border and basolateral membrane vesicles. Biochim. Biophys. Acta., 773, 113-124 (1984).
    • (1984) Biochim. Biophys. Acta , vol.773 , pp. 113-124
    • Takano, M.1    Inui, K.2    Okano, T.3
  • 77
    • 0031434178 scopus 로고    scopus 로고
    • Cloning and characterization of a novel human pH-dependent organic cation transporter, OCTN1
    • Tamai, I., Yabuuchi, H., Nezu, J., et al.: Cloning and characterization of a novel human pH-dependent organic cation transporter, OCTN1. FEBS Lett., 419, 107-111 (1997).
    • (1997) FEBS Lett , vol.419 , pp. 107-111
    • Tamai, I.1    Yabuuchi, H.2    Nezu, J.3
  • 78
    • 0035132434 scopus 로고    scopus 로고
    • Molecular and physiological evidence for multifunctionality of carnitine/organic cation transporter OCTN2
    • Ohashi, R., Tamai, I., Nezu, Ji J., et al.: Molecular and physiological evidence for multifunctionality of carnitine/organic cation transporter OCTN2. Mol. Pharmacol., 59, 358-366 (2001).
    • (2001) Mol. Pharmacol , vol.59 , pp. 358-366
    • Ohashi, R.1    Tamai, I.2    Nezu, J.J.3
  • 79
    • 33746548445 scopus 로고    scopus 로고
    • Identification and functional characterization of a new human kidney-specific H +/organic cation antiporter, kidney-specific multidrug and toxin extrusion 2
    • Masuda, S., Terada, T., Yonezawa, A., et al.: Identification and functional characterization of a new human kidney-specific H +/organic cation antiporter, kidney-specific multidrug and toxin extrusion 2. J. Am. Soc. Nephrol., 17, 2127-2135 (2006).
    • (2006) J. Am. Soc. Nephrol , vol.17 , pp. 2127-2135
    • Masuda, S.1    Terada, T.2    Yonezawa, A.3
  • 80
    • 34250005960 scopus 로고    scopus 로고
    • Substrate specificity of MATE1 and MATE2-K, human multidrug and toxin extrusions\H, organic cation antiporters
    • Tanihara, Y., Masuda, S., Sato, T., et al.: Substrate specificity of MATE1 and MATE2-K, human multidrug and toxin extrusions\H(+)-organic cation antiporters. Biochem. Pharmacol., 74, 359-371 (2007).
    • (2007) Biochem. Pharmacol , vol.74 , pp. 359-371
    • Tanihara, Y.1    Masuda, S.2    Sato, T.3
  • 81
    • 0035703732 scopus 로고    scopus 로고
    • The roles of P-glycoprotein and MRP1 in the blood-brain and blood-cerebrospinal fluid barriers
    • Schinkel, A. H.: The roles of P-glycoprotein and MRP1 in the blood-brain and blood-cerebrospinal fluid barriers. Adv. Exp. Med. Biol., 500, 365-372 (2001).
    • (2001) Adv. Exp. Med. Biol , vol.500 , pp. 365-372
    • Schinkel, A.H.1
  • 82
    • 33748896303 scopus 로고    scopus 로고
    • Expression and function of multidrug resistance transporters at the blood-brain barriers
    • Scherrmann, J. M.: Expression and function of multidrug resistance transporters at the blood-brain barriers. Expert. Opin. Drug Metab. Toxicol., 1, 233-46 (2005).
    • (2005) Expert. Opin. Drug Metab. Toxicol , vol.1 , pp. 233-246
    • Scherrmann, J.M.1
  • 83
    • 0028229150 scopus 로고
    • Disruption of the mouse mdr1a P-glycoprotein gene leads to a deficiency in the blood-brain barrier and to increased sensitivity to drugs
    • Schinkel, A. H., Smit, J. J., van Tellingen, O., et al.: Disruption of the mouse mdr1a P-glycoprotein gene leads to a deficiency in the blood-brain barrier and to increased sensitivity to drugs. Cell, 77, 491-502 (1994).
    • (1994) Cell , vol.77 , pp. 491-502
    • Schinkel, A.H.1    Smit, J.J.2    van Tellingen, O.3
  • 84
    • 0032960014 scopus 로고    scopus 로고
    • Role of blood-brain barrier P-glycoprotein in limiting brain accumulation and sedative side-effects of asimadoline, a peripherally acting analgaesic drug
    • Jonker, J. W., Wagenaar, E. van Deemter, L., et al.: Role of blood-brain barrier P-glycoprotein in limiting brain accumulation and sedative side-effects of asimadoline, a peripherally acting analgaesic drug. Br. J. Pharmacol., 127, 43-50 (1999).
    • (1999) Br. J. Pharmacol , vol.127 , pp. 43-50
    • Jonker, J.W.1    Wagenaar, E.2    van Deemter, L.3
  • 85
    • 0035668339 scopus 로고    scopus 로고
    • Comparative studie on in vitro methods for evaluating in vivo function of MDR1 Pglycoprotein
    • Adachi, Y., Suzuki, H. and Sugiyama, Y.: Comparative studie on in vitro methods for evaluating in vivo function of MDR1 Pglycoprotein. Pharm. Res., 18, 1660-1668 (2001).
    • (2001) Pharm. Res , vol.18 , pp. 1660-1668
    • Adachi, Y.1    Suzuki, H.2    Sugiyama, Y.3
  • 86
    • 0035119989 scopus 로고    scopus 로고
    • In vitro substrate identification studies for p-glycoprotein-mediated transport: Species difference and predictability of in vivo results
    • Yamazaki, M., Neway, W. E., Ohe, T., et al.: In vitro substrate identification studies for p-glycoprotein-mediated transport: species difference and predictability of in vivo results. J. Pharmacol. Exp. Ther., 296, 723-35 (2001).
    • (2001) J. Pharmacol. Exp. Ther , vol.296 , pp. 723-735
    • Yamazaki, M.1    Neway, W.E.2    Ohe, T.3
  • 87
    • 0037180446 scopus 로고    scopus 로고
    • The breast cancer resistance protein protects against a major chlorophyllderived dietary phototoxin and protoporphyria
    • Jonker, J. W., Buitelaar, M., Wagenaar, E., et al.: The breast cancer resistance protein protects against a major chlorophyllderived dietary phototoxin and protoporphyria. Proc. Natl. Acad. Sci. USA, 99, 15649-15654 (2002).
    • (2002) Proc. Natl. Acad. Sci. USA , vol.99 , pp. 15649-15654
    • Jonker, J.W.1    Buitelaar, M.2    Wagenaar, E.3
  • 88
    • 4344632950 scopus 로고    scopus 로고
    • Mrp4 confers resistance to topotecan and protects the brain from chemotherapy
    • Leggas, M., Adachi, M., Scheffer, G. L., et al.: Mrp4 confers resistance to topotecan and protects the brain from chemotherapy. Mol. Cell. Biol., 24, 7612-7621 (2004).
    • (2004) Mol. Cell. Biol , vol.24 , pp. 7612-7621
    • Leggas, M.1    Adachi, M.2    Scheffer, G.L.3
  • 89
    • 34748926909 scopus 로고    scopus 로고
    • Effect of breast cancer resistance protein (Bcrp/Abcg2) on the disposition of phytoestrogens
    • Enokizono, J., Kusuhara, H. and Sugiyama, Y.: Effect of breast cancer resistance protein (Bcrp/Abcg2) on the disposition of phytoestrogens. Mol. Pharmacol., 72, 967-975 (2007).
    • (2007) Mol. Pharmacol , vol.72 , pp. 967-975
    • Enokizono, J.1    Kusuhara, H.2    Sugiyama, Y.3
  • 90
    • 44149086662 scopus 로고    scopus 로고
    • Quantitative investigation of the role of breast cancer resistance protein (Bcrp/ Abcg2) in limiting brain and testis penetration of xenobiotic compounds
    • Enokizono, J., Kusuhara, H., Ose, A., et al.: Quantitative investigation of the role of breast cancer resistance protein (Bcrp/ Abcg2) in limiting brain and testis penetration of xenobiotic compounds. Drug Metab. Dispos., 36, 995-1002 (2008).
    • (2008) Drug Metab. Dispos , vol.36 , pp. 995-1002
    • Enokizono, J.1    Kusuhara, H.2    Ose, A.3
  • 91
    • 67649397523 scopus 로고    scopus 로고
    • Limited brain distribution of Ro 64-0802, a pharmacologically active form of oseltamivir, by active efflux across the blood-brain barrier mediated by organic anion transporter 3 (Oat3/Slc22a8) and multidrug resistance-associated protein 4 (Mrp4/Abcc4)
    • Ose, A., Ito, M., Kusuhara, H., et al.: Limited brain distribution of Ro 64-0802, a pharmacologically active form of oseltamivir, by active efflux across the blood-brain barrier mediated by organic anion transporter 3 (Oat3/Slc22a8) and multidrug resistance-associated protein 4 (Mrp4/Abcc4). Drug Metab. Dispos., (2008).
    • (2008) Drug Metab. Dispos
    • Ose, A.1    Ito, M.2    Kusuhara, H.3
  • 92
    • 33846441251 scopus 로고    scopus 로고
    • Multidrug resistance protein 4 protects bone marrow, thymus, spleen, and intestine from nucleotide analogue-induced damage
    • Belinsky, M. G., Guo, P., Lee, K., et al.: Multidrug resistance protein 4 protects bone marrow, thymus, spleen, and intestine from nucleotide analogue-induced damage. Cancer Res., 67, 262-8 (2007).
    • (2007) Cancer Res , vol.67 , pp. 262-268
    • Belinsky, M.G.1    Guo, P.2    Lee, K.3
  • 94
    • 2342436512 scopus 로고    scopus 로고
    • PET Studies on P-glycoprotein function in the blood-brain barrier: How it affects uptake and binding of drugs within the CNS
    • Elsinga, P. H., Hendrikse, N. H., Bart, J., et al.: PET Studies on P-glycoprotein function in the blood-brain barrier: how it affects uptake and binding of drugs within the CNS. Curr. Pharm. Des., 10, 1493-1503 (2004).
    • (2004) Curr. Pharm. Des , vol.10 , pp. 1493-1503
    • Elsinga, P.H.1    Hendrikse, N.H.2    Bart, J.3
  • 95
    • 31144434780 scopus 로고    scopus 로고
    • In Vivo Evaluation of P-glycoprotein Function at the Blood-Brain Barrier in Nonhuman Primates Using [11C]Verapamil
    • Lee, Y. J., Maeda, J., Kusuhara, H., et al.: In Vivo Evaluation of P-glycoprotein Function at the Blood-Brain Barrier in Nonhuman Primates Using [11C]Verapamil. J. Pharmacol. Exp. Ther., 316, 647-553 (2006).
    • (2006) J. Pharmacol. Exp. Ther , vol.316 , pp. 647-553
    • Lee, Y.J.1    Maeda, J.2    Kusuhara, H.3
  • 96
    • 20444480695 scopus 로고    scopus 로고
    • Imaging Pglycoprotein transport activity at the human blood-brain barrier with positron emission tomography
    • Sasongko, L., Link, J. M., Muzi, M., et al.: Imaging Pglycoprotein transport activity at the human blood-brain barrier with positron emission tomography. Clin. Pharmacol. Ther., 77, 503-514 (2005).
    • (2005) Clin. Pharmacol. Ther , vol.77 , pp. 503-514
    • Sasongko, L.1    Link, J.M.2    Muzi, M.3
  • 97
    • 0036070666 scopus 로고    scopus 로고
    • Involvement of organic cation transporter 1 in hepatic and intestinal distribution of metformin
    • Wang, D. S., Jonker, J. W., Kato, Y., et al.: Involvement of organic cation transporter 1 in hepatic and intestinal distribution of metformin. J. Pharmacol. Exp. Ther., 302, 510-515 (2002).
    • (2002) J. Pharmacol. Exp. Ther , vol.302 , pp. 510-515
    • Wang, D.S.1    Jonker, J.W.2    Kato, Y.3
  • 98
    • 0037381942 scopus 로고    scopus 로고
    • Involvement of organic cation transporter 1 in the lactic acidosis caused by metformin
    • Wang, D. S., Kusuhara, H., Kato, Y., et al.: Involvement of organic cation transporter 1 in the lactic acidosis caused by metformin. Mol. Pharmacol., 63, 844-848 (2003).
    • (2003) Mol. Pharmacol , vol.63 , pp. 844-848
    • Wang, D.S.1    Kusuhara, H.2    Kato, Y.3
  • 99
    • 48549088791 scopus 로고    scopus 로고
    • Transporter-mediated protection against thiopurine-induced hematopoietic toxicity
    • Krishnamurthy, P., Schwab, M., Takenaka, K., et al.: Transporter-mediated protection against thiopurine-induced hematopoietic toxicity. Cancer. Res., 68, 4983-4989 (2008).
    • (2008) Cancer. Res , vol.68 , pp. 4983-4989
    • Krishnamurthy, P.1    Schwab, M.2    Takenaka, K.3
  • 100
    • 0033045409 scopus 로고    scopus 로고
    • Hepatic uptake of the magnetic resonance imaging contrast agent gadoxetate by the organic anion transporting polypeptide Oatp1
    • van Montfoort, J. E., Stieger, B., Meijer, D. K., et al.: Hepatic uptake of the magnetic resonance imaging contrast agent gadoxetate by the organic anion transporting polypeptide Oatp1. J. Pharmacol. Exp. Ther., 290, 153-157 (1999).
    • (1999) J. Pharmacol. Exp. Ther , vol.290 , pp. 153-157
    • van Montfoort, J.E.1    Stieger, B.2    Meijer, D.K.3
  • 101
    • 46749120546 scopus 로고    scopus 로고
    • Use of tc-99m mebrofenin as a clinical probe to assess altered hepatobiliary transport: Integration of in vitro, pharmacokinetic modeling, and simulation studies
    • Ghibellini, G., Leslie, E. M., Pollack, G. M., et al.: Use of tc-99m mebrofenin as a clinical probe to assess altered hepatobiliary transport: integration of in vitro, pharmacokinetic modeling, and simulation studies. Pharm. Res., 25, 1851-1860 (2008).
    • (2008) Pharm. Res , vol.25 , pp. 1851-1860
    • Ghibellini, G.1    Leslie, E.M.2    Pollack, G.M.3
  • 102
    • 0029044441 scopus 로고
    • Noninvasive assessment of hepatobiliary and renal elimination of cysteinyl leukotrienes by positron emission tomography
    • Guhlmann, A., Krauss, K., Oberdorfer, F., et al.: Noninvasive assessment of hepatobiliary and renal elimination of cysteinyl leukotrienes by positron emission tomography. Hepatology, 21, 1568-1575 (1995).
    • (1995) Hepatology , vol.21 , pp. 1568-1575
    • Guhlmann, A.1    Krauss, K.2    Oberdorfer, F.3
  • 103
    • 0033622160 scopus 로고    scopus 로고
    • Evaluation of 11C-colchicine for PET imaging of multiple drug resistance
    • Levchenko, A., Mehta, B. M., Lee, J. B., et al.: Evaluation of 11C-colchicine for PET imaging of multiple drug resistance. J. Nucl. Med., 41, 493-501 (2000).
    • (2000) J. Nucl. Med , vol.41 , pp. 493-501
    • Levchenko, A.1    Mehta, B.M.2    Lee, J.B.3
  • 104
    • 0033563120 scopus 로고    scopus 로고
    • A new in vivo method to study P-glycoprotein transport in tumors and the blood-brain barrier
    • Hendrikse, N. H., de Vries, E. G., Eriks-Fluks, L., et al.: A new in vivo method to study P-glycoprotein transport in tumors and the blood-brain barrier. Cancer Res., 59, 2411-2416 (1999).
    • (1999) Cancer Res , vol.59 , pp. 2411-2416
    • Hendrikse, N.H.1    de Vries, E.G.2    Eriks-Fluks, L.3
  • 105
    • 33644818018 scopus 로고    scopus 로고
    • Estimation of paclitaxel biodistribution and uptake in human-derived xenografts in vivo with (18)F-fluoropaclitaxel
    • Gangloff, A., Hsueh, W. A., Kesner, A. L., et al.: Estimation of paclitaxel biodistribution and uptake in human-derived xenografts in vivo with (18)F-fluoropaclitaxel. J. Nucl. Med., 46, 1866-71 (2005).
    • (2005) J. Nucl. Med , vol.46 , pp. 1866-1871
    • Gangloff, A.1    Hsueh, W.A.2    Kesner, A.L.3
  • 106
    • 0031874816 scopus 로고    scopus 로고
    • Complete in vivo reversal of P-glycoprotein pump function in the blood-brain barrier visualized with positron emission tomography
    • Hendrikse, N. H., Schinkel, A. H., de Vries, E. G., et al.: Complete in vivo reversal of P-glycoprotein pump function in the blood-brain barrier visualized with positron emission tomography. Br. J. Pharmacol., 124, 1413-8 (1998).
    • (1998) Br. J. Pharmacol , vol.124 , pp. 1413-1418
    • Hendrikse, N.H.1    Schinkel, A.H.2    de Vries, E.G.3
  • 107
    • 25144446459 scopus 로고    scopus 로고
    • Positron emission tomography studies on binding of central nervous system drugs and P-glycoprotein function in the rodent brain
    • Elsinga, P. H., Hendrikse, N. H., Bart, J., et al.: Positron emission tomography studies on binding of central nervous system drugs and P-glycoprotein function in the rodent brain. Mol. Imaging. Biol., 7, 37-44 (2005).
    • (2005) Mol. Imaging. Biol , vol.7 , pp. 37-44
    • Elsinga, P.H.1    Hendrikse, N.H.2    Bart, J.3
  • 108
    • 30544440661 scopus 로고    scopus 로고
    • New positron emission tomography tracer [(11)C]carvedilol reveals Pglycoprotein modulation kinetics
    • Bart, J., Dijkers, E. C., Wegman, T. D., et al.: New positron emission tomography tracer [(11)C]carvedilol reveals Pglycoprotein modulation kinetics. Br. J. Pharmacol., 145, 1045-1051 (2005).
    • (2005) Br. J. Pharmacol , vol.145 , pp. 1045-1051
    • Bart, J.1    Dijkers, E.C.2    Wegman, T.D.3
  • 109
    • 25444477513 scopus 로고    scopus 로고
    • Synthesis and evaluation of dopamine D3 receptor antagonist 11C-GR218231 as PET tracer for P-glycoprotein
    • de Vries, E. F., Kortekaas, R., vanWaarde, A., et al.: Synthesis and evaluation of dopamine D3 receptor antagonist 11C-GR218231 as PET tracer for P-glycoprotein. J. Nucl. Med., 46, 1384-1392 (2005).
    • (2005) J. Nucl. Med , vol.46 , pp. 1384-1392
    • de Vries, E.F.1    Kortekaas, R.2    vanWaarde, A.3
  • 110
    • 33847275232 scopus 로고    scopus 로고
    • In vivo evaluation of P-glycoprotein modulation of 8 PET radioligands used clinically
    • Ishiwata, K., Kawamura, K., Yanai, K., et al.: In vivo evaluation of P-glycoprotein modulation of 8 PET radioligands used clinically. J. Nucl. Med., 48, 81-87 (2007).
    • (2007) J. Nucl. Med , vol.48 , pp. 81-87
    • Ishiwata, K.1    Kawamura, K.2    Yanai, K.3
  • 111
    • 0027478443 scopus 로고
    • Functional imaging of multidrug-resistant P-glycoprotein with an organotechnetium complex
    • Piwnica-Worms, D., Chiu, M. L., Budding, M., et al.: Functional imaging of multidrug-resistant P-glycoprotein with an organotechnetium complex. Cancer. Res., 53, 977-84 (1993).
    • (1993) Cancer. Res , vol.53 , pp. 977-984
    • Piwnica-Worms, D.1    Chiu, M.L.2    Budding, M.3
  • 112
    • 3843081865 scopus 로고    scopus 로고
    • In vivo imaging of hepatobiliary transport function mediated by multidrug resistance associated protein and P-glycoprotein
    • Hendrikse, N. H., Kuipers, F., Meijer, C., et al.: In vivo imaging of hepatobiliary transport function mediated by multidrug resistance associated protein and P-glycoprotein. Cancer Chemother Pharmacol., 54, 131-138 (2004).
    • (2004) Cancer Chemother Pharmacol , vol.54 , pp. 131-138
    • Hendrikse, N.H.1    Kuipers, F.2    Meijer, C.3
  • 113
    • 0036290428 scopus 로고    scopus 로고
    • In vitro and in vivo hepatic transport of the magnetic resonance imaging contrast agent B22956/1: Role of MRP proteins
    • Lorusso, V., Pascolo, L., Fernetti, C., et al.: In vitro and in vivo hepatic transport of the magnetic resonance imaging contrast agent B22956/1: role of MRP proteins. Biochem. Biophys. Res. Commun., 293, 100-105 (2002).
    • (2002) Biochem. Biophys. Res. Commun , vol.293 , pp. 100-105
    • Lorusso, V.1    Pascolo, L.2    Fernetti, C.3


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.