Commonly Used Surfactant, Tween 80, Improves Absorption of P-Glycoprotein Substrate, Digoxin, in Rats

  • Zhang, Hongjian (Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb Pharmaceutical Research Institute) ;
  • Yao, Ming (Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb Pharmaceutical Research Institute) ;
  • Morrison, Richard-A. (Schering-Plough Research Institute) ;
  • Chong, Sae-Ho (Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb Pharmaceutical Research Institute)
  • Published : 2003.09.01

Abstract

Tween 80 (Polysorbate 80) is a hydrophilic nonionic surfactant commonly used as an ingredient in dosing vehicles for pre-clinical in vivo studies (e.g., pharmacokinetic studies, etc.). Tween 80 increased apical to basolateral permeability of digoxin in Caco-2 cells suggesting that Tween 80 is an in vitro inhibitor of P-gp. The overall objective of the present study was to investigate whether an inhibition of P-gp by Tween 80 can potentially influence in vivo absorption of P-gp substrates by evaluating the effect of Tween 80 on the disposition of digoxin (a model P-gp substrate with minimum metabolism) after oral administration in rats. Rats were dosed orally with digoxin (0.2 mg/kg) formulated in ethanol (40%, v/v) and saline mixture with and without Tween 80 (1 or 10%, v/v). Digoxin oral AUC increased 30 and 61% when dosed in 1 % and 10% Tween 80, respectively, compared to control (P<0.05). To further examine whether the increase in digoxin AUC after oral administration of Tween 80 is due, in part, to a systemic inhibition of digoxin excretion in addition to an inhibition of P-gp in the GI tract, a separate group of rats received digoxin intravenously (0.2 mg/kg) and Tween 80 (10% v/v) orally. No significant changes in digoxin IV AUC was noted when Tween 80 was administered orally. In conclusion, Tween 80 significantly increased digoxin AUC and Cmax after oral administration, and the increased AUC is likely to be due to an inhibition of P-gp in the gut (i.e., improved absorption). Therefore, Tween 80 is likely to improve systemic exposure of P-gp substrates after oral administration. Comparing AUC after oral administration with and without Tween 80 may be a viable strategy in evaluating whether oral absorption of P-gp substrates is potentially limited by P-gp in the gut.

Keywords

References

  1. Batrakova, E. V., Han, H. Y., Alakhov, V. Y., Miller, D. W., and Kabanov, A. V., Effects of pluronic block copolymers on drug absorption in Caco-2 cell monolayers. Pharm. Res., 15, 850-855 (1998) https://doi.org/10.1023/A:1011964213024
  2. Borst, P., Evers, R., Kool, M., and Wijnholds, J., The multidrug resistance protein family. Biophys. Acta, 1461, 347-357 (1999) https://doi.org/10.1016/S0005-2736(99)00167-4
  3. Cornaire, G., Woodley, J. F., Saivin, S., Legendre, J.Y., Decourt, S., Cloarec, A., and Houin, G., Effect of polyoxyl 35 castor oil and Polysorbate 80 on the intestinal absorption of digoxin in vitro. Arzneimittel-forschung., 50, 576-579 (2000)
  4. Fromm, M. F., Kim, R. B., Stein, C. M., Wilkinson, G. R., and Roden, D. M., Inhibition of P-glycoprotein-mediated drug transport: A unifying mechanism to explain the interaction between digoxin and quinidine. Circulation, 99, 552-557 (1999) https://doi.org/10.1161/01.CIR.99.4.552
  5. Gottesman, M. M. and Pastan, I., Biochemistry of multidrug resistance mediated by the multidrug transporter. Annu. Rev. Biochem., 62, 385-427 (1993) https://doi.org/10.1146/annurev.bi.62.070193.002125
  6. Hedman, A., Angelin, B., Arvidsson, A., Dahlqvist, R., and Nilsson, B.. Interactions in the renal and biliary elimination of digoxin: stereoselective difference between quinine and quinidine. Clin. Pharmacol. Ther., 47, 20-26 (1990) https://doi.org/10.1038/clpt.1990.3
  7. Hinderling, P. H. and Hartmann, D., Pharmacokinetics of digoxin and main metabolites/ derivatives in healthy humans. Ther. Drug Monit., 13, 381-401 (1991) https://doi.org/10.1097/00007691-199109000-00001
  8. Hugger, E. D., Novak, B. L., Burton, P. S., Audus, K. L., and Borchardt, R. T., A comparison of commonly used polyehtoxylated pharmaceutical excipients on their ability to inhibit P-glycoprotein activity in vitro. J. Pharm. Sci., 91, 1991-2002 (2002) https://doi.org/10.1002/jps.10176
  9. Iisalo, E., Clinical pharmacokinetics of digoxin. Clin. Pharmacokinet., 2, 1-16 (1977) https://doi.org/10.2165/00003088-197702010-00001
  10. Knutsen, T., Mickley, L. A., Ried, T., Green, E. D., du Manoir, S., Schrock, E., Macville, M., Ning, Y., Robey, R., Polymeropoulos, M., Torres, R., and Fojo, T., Cytogenetic and molecular characterization of random chromosomal rearrangements activating the drug resistance gene, MDR1/P-glycoprotein, in drug-selected cell lines and patients with drug refractory ALL. Genes Chromosomes Cancer, 23, 44-54 (1998) https://doi.org/10.1002/(SICI)1098-2264(199809)23:1<44::AID-GCC7>3.0.CO;2-6
  11. Litman, T., Druley, T. E., Stein, W. D., and Bates, S. E., From MDR to MXR: new understanding of multidrug resistance systems, their properties and clinical significance. Cell. Mol. Life Sci. 58, 931-959 (2001) https://doi.org/10.1007/PL00000912
  12. Malingre, M M., Schellens, J. H., Van Tellingen, O., Ouwehand, M., Bardelmeijer, H. A., Rosing, H., Koopman, F. J., Schot, M. E., Ten Bokkel Huinink, W. W., and Beijnen, J. H., The cosolvent Cremophor EL limits absorption of orally administered paclitaxel in cancer patients. Br. J. Cancer, 85, 1472-1477 (2001) https://doi.org/10.1054/bjoc.2001.2118
  13. Nerurkar, M. M., Ho, N. F., Burton, P. S., Vidmar, T. J., and Borchardt, R. T., Mechanistic roles of neutral surfactants on concurrent polarized and passive membrane transport a model peptide in Caco-2 cells. J. Pharm. Sci., 86, 813-821 (1997) https://doi.org/10.1021/js960483y
  14. Salphati, L. and Benet, L. Z., Effects of ketoconazole on digoxin absorption and disposition in rat. Pharmacology, 56, 308-313 (1998) https://doi.org/10.1159/000028214
  15. Salphati, L. and Benet, L. Z., Metabolism of digoxin and digoxigenin digitoxosides in rat liver microsomes: involvement of cytochrome P4503A. Xenobiotica, 29, 171-85 (1999) https://doi.org/10.1080/004982599238722
  16. Silverman, J. A., Multidrug-resistance transporters. Pharm. Biotechnol., 12, 353-86 (1999)
  17. Song, S., Suzuki, H., Kawai, R., and Sugiyama, Y., Effect of PSC 833, a P-glycoprotein modulator, on the disposition of vincristine and digoxin in rats. Drug Metab. Disp., 27, 689-694 (1999)
  18. Stephens, R. H., O'Neill, C. A., Warhurst, A., Carlson, G. L., Rowland, M., and Warhurst, G., Kinetic profiling of P-glycoprotein-mediated drug efflux in rat and human intestinal epithelia. J. Pharmacol. Exp. Ther., 296, 584-591 (2001)
  19. Tanigawara, Y., Role of P-glycoprotein in drug disposition. Ther. Drug Monit., 22, 137-140 (2000) https://doi.org/10.1097/00007691-200002000-00029
  20. Troutman, M. D., Luo, G., Gan, L. S., and Thakker, D. R., The role of P-glycoprotein in drug disposition: significance to drug development. In Rodrigues, A. D. (ed.). Drug-drug interactions. Marcel Dekker, New York. pp. 295-357 (2001)
  21. van Zuylen, L., Verweij, J., and Sparreboom, A., Role of formulation vehicles in taxane pharmacology. Invest. New Drugs, 19,125-141 (2001) https://doi.org/10.1023/A:1010618632738
  22. Verschraagen, M., Koks, C. H., Schellens, J. H., and Beijnen, J. H., P-glycoprotein system as a determinant of drug interactions: the case of digoxin-verapamil. Pharmacol. Res., 40, 301-306 (1999) https://doi.org/10.1006/phrs.1999.0535
  23. Yamazaki, M., Neway, W. E., Ohe, T., Chen, I., Rowe, J. F., Hochman, J. H., Chiba, M., and Lin, J. H., In vitro substrate identification studies for p-glycoprotein-mediated transport: species difference and predictability of in vivo results. J. Pharmacol. Exp. Ther., 296, 723-735 (2001)
  24. Yao, M., Zhang, H., Chong, S., Zhu, M., and Morrison, R. A., A rapid and sensitive LC/MS/MS assay for quantitative determination of digoxin in rat plasma. J. Pharmaceu. Biomed. Anal., 32, 1189-1197 (2003) https://doi.org/10.1016/S0731-7085(03)00050-5