Skip to main content

Advertisement

Log in

Effects of adrenolytic mitotane on drug elimination pathways assessed in vitro

  • Original Article
  • Published:
Endocrine Aims and scope Submit manuscript

Abstract

Mitotane (1,1-dichloro-2-(o-chlorophenyl)-2-(p-chlorophenyl)ethane, o,p′-DDD) represents one of the most active drugs for the treatment of adrenocortical carcinoma. Its metabolites 1,1-(o,p′-dichlorodiphenyl) acetic acid (=o,p′-DDA) and 1,1-(o,p′-dichlorodiphenyl)-2,2 dichloroethene (=o,p′-DDE) partly contribute to its pharmacological effects. Because mitotane has a narrow therapeutic index and causes pharmacokinetic drug–drug interactions, knowledge about these compounds’ effects on drug metabolizing and transporting proteins is crucial. Using quantitative real-time polymerase chain reaction, our study confirmed the strong inducing effects of o,p′-DDD on mRNA expression of cytochrome P450 3A4 (CYP3A4, 30-fold) and demonstrated that other enzymes and transporters are also induced (e.g., CYP1A2, 8.4-fold; ABCG2 (encoding breast resistance cancer protein, BCRP), 4.2-fold; ABCB1 (encoding P-glycoprotein, P-gp) 3.4-fold). P-gp induction was confirmed at the protein level. o,p′-DDE revealed a similar induction profile, however, with less potency and o,p′-DDA had only minor effects. Reporter gene assays clearly confirmed o,p′-DDD to be a PXR activator and for the first time demonstrated that o,p′-DDE and o,p′-DDA also activate PXR albeit with lower potency. Using isolated, recombinant CYP enzymes, o,p′-DDD and o,p′-DDE were shown to strongly inhibit CYP2C19 (IC50 = 0.05 and 0.09 µM). o,p′-DDA exhibited only minor inhibitory effects. In addition, o,p′-DDD, o,p′-DDE, and o,p′-DDA are demonstrated to be neither substrates nor inhibitors of BCRP or P-gp function. In summary, o,p′-DDD and o,p′-DDE might be potential perpetrators in pharmacokinetic drug–drug interactions through induction of drug-metabolizing enzymes or drug transporters and by potent inhibition of CYP2C19. In tumors over-expressing BCRP or P-gp, o,p′-DDD and its metabolites should retain their efficacy due to a lack of substrate characteristics.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  1. A.P. Fay, A. Elfiky, G.H. Teló, R.R. McKay, M. Kaymakcalan, P.L. Nguyen, A. Vaidya, D.T. Ruan, J. Bellmunt, T.K. Choueiri, Adrenocortical carcinoma: the management of metastatic disease. Crit. Rev. Oncol. Hematol. 92, 123–132 (2014)

  2. M. Terzolo, A. Ardito, B. Zaggia, F. Laino, A. Germano, S. De Francia, F. Daffara, A. Berruti, Management of adjuvant mitotane therapy following resection of adrenal cancer. Endocrine 42, 521–525 (2012)

    Article  CAS  PubMed  Google Scholar 

  3. A. Colao, M. Boscaro, D. Ferone, F.F. Casanueva, Managing Cushing’s disease: the state of the art. Endocrine 47, 9–20 (2014)

    Article  CAS  PubMed  Google Scholar 

  4. D. Ferone, C. Pivonello, G. Vitale, M.C. Zatelli, A. Colao, R. Pivonello, Molecular basis of pharmacological therapy in Cushing’s disease. Endocrine 46, 181–198 (2014)

    Article  CAS  PubMed  Google Scholar 

  5. I. Veytsman, L. Nieman, T. Fojo, Management of endocrine manifestations and the use of mitotane as a chemotherapeutic agent for adrenocortical carcinoma. J. Clin. Oncol. 27, 4619–4629 (2009)

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  6. C.W. Lin, Y.H. Chang, H.F. Pu, Mitotane exhibits dual effects on steroidogenic enzymes gene transcription under basal and cAMP-stimulating microenvironments in NCI-H295 cells. Toxicology 298, 14–23 (2012)

    Article  CAS  PubMed  Google Scholar 

  7. T.P. Lehmann, T. Wrzesiński, P.P. Jagodziński, The effect of mitotane on viability, steroidogenesis and gene expression in NCI-H295R adrenocortical cells. Mol. Med. Rep. 7, 893–900 (2013)

    CAS  PubMed  Google Scholar 

  8. D.E. Schteingart, Adjuvant mitotane therapy of adrenal cancer—use and controversy. N. Eng. J. Med. 356, 2415–2418 (2007)

    Article  CAS  Google Scholar 

  9. I.G. Hermsen, M. Fassnacht, M. Terzolo, S. Houterman, J. den Hartigh, S. Leboulleux, F. Daffara, A. Berruti, R. Chadarevian, M. Schlumberger, B. Allolio, H.R. Haak, E. Baudin, Plasma concentrations of o, p’DDD, o, p’DDA, and o, p’-DDE as predictors of tumor response to mitotane in adrenocortical carcinoma: results of a retrospective ENS@T multicenter study. J. Clin. Endocrinol. Metabol. 96, 1844–1851 (2011)

    Article  CAS  Google Scholar 

  10. F. Martz, J.A. Straw, The in vitro metabolism of 1-(o-chlorophenyl)-1-(p-chlorophenyl)-2,2-dichloroethane (o, p’-DDD) by dog adrenal mitochondria and metabolite covalent binding to mitochondrial macromolecules: a possible mechanism for the adrenocorticolytic effect. Drug Metab. Dispos. 5, 482–486 (1977)

    CAS  PubMed  Google Scholar 

  11. C.L. Ronchi, S. Sbiera, M. Volante, S. Steinhauer, V. Scott-Wild, B. Altieri, M. Kroiss, M. Bala, M. Papotti, T. Deutschbein, M. Terzolo, M. Fassnacht, B. Allolio, CYP2W1 is highly expressed in adrenal glands and is positively associated with the response to mitotane in adrenocortical carcinoma. PLoS One 9, e105855 (2014)

    Article  PubMed Central  PubMed  Google Scholar 

  12. H.R. Haak, J. Hermans, C.J. van de Velde, E.G. Lentjes, B.M. Goslings, G.J. Fleuren, H.M. Krans, Optimal treatment of adrenocortical carcinoma with mitotane: results in a consecutive series of 96 patients. Br. J. Cancer 69, 947–951 (1994)

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  13. M. Terzolo, A.E. Baudin, A. Ardito, M. Kroiss, S. Leboulleux, F. Daffara, P. Perotti, R.A. Feelders, J.H. deVries, B. Zaggia, S. De Francia, M. Volante, H.R. Haak, B. Allolio, A. Ghuzlan, M. Fassnacht, A. Berruti, Mitotane levels predict the outcome of patients with adrenocortical carcinoma treated adjuvantly following radical resection. Eur. J. Endocrinol. 169, 263–270 (2013)

    Article  CAS  PubMed  Google Scholar 

  14. E. Baudin, G. Pellegriti, M. Bonnay, A. Penfornis, A. Laplanche, G. Vassal, M. Schlumberger, Impact of monitoring plasma 1,1-dichlorodiphenildichloroethane (o, p’DDD) levels on the treatment of patients with adrenocortical carcinoma. Cancer 92, 1385–1392 (2001)

    Article  CAS  PubMed  Google Scholar 

  15. M. Terzolo, A. Pia, A. Berruti, G. Osella, A. Alì, V. Carbone, E. Testa, L. Dogliotti, A. Angeli, Low-dose monitored mitotane treatment achieves the therapeutic range with manageable side effects in patients with adrenocortical cancer. J. Clin. Endocrinol. Metabol. 85, 2234–2238 (2000)

    CAS  Google Scholar 

  16. M. Fassnacht, M. Terzolo, B. Allolio, E. Baudin, H. Haak, A. Berruti, S. Welin, C. Schade-Brittinger, A. Lacroix, B. Jarzab, H. Sorbye, D.J. Torpy, V. Stepan, D.E. Schteingart, W. Arlt, M. Kroiss, S. Leboulleux, P. Sperone, A. Sundin, I. Hermsen, S. Hahner, H.S. Willenberg, A. Tabarin, M. Quinkler, C. de la Fouchardière, M. Schlumberger, F. Mantero, D. Weismann, F. Beuschlein, H. Gelderblom, H. Wilmink, M. Sender, M. Edgerly, W. Kenn, T. Fojo, H.H. Müller, B. Skogseid, FIRM-ACT Study Group. Combination chemotherapy in advanced adrenocortical carcinoma. N. Engl. J. Med. 366, 2189–2197 (2012)

    Article  CAS  PubMed  Google Scholar 

  17. V. Chortis, A.E. Taylor, P. Schneider, J.W. Tomlinson, B.A. Hughes, D.M. O’Neil, R. Libé, B. Allolio, X. Bertagna, J. Bertherat, F. Beuschlein, M. Fassnacht, N. Karavitaki, M. Mannelli, F. Mantero, G. Opocher, E. Porfiri, M. Quinkler, M. Sherlock, M. Terzolo, P. Nightingale, C.H. Shackleton, P.M. Stewart, S. Hahner, W. Arlt, Mitotane therapy in adrenocortical cancer induces CYP3A4 and inhibits 5α-reductase, explaining the need for personalized glucocorticoid and androgen replacement. J. Clin. Endocrinol. Metabol. 98, 161–171 (2013)

    Article  CAS  Google Scholar 

  18. N.P. van Erp, H.J. Guchelaar, B.A. Ploeger, J.A. Romijn, J.D. Hartigh, H. Gelderblom, Mitotane has a strong and a durable inducing effect on CYP3A4 activity. Eur. J. Endocrinol. 164, 621–626 (2011)

    Article  PubMed  Google Scholar 

  19. A. Takeshita, J. Igarashi-Migitaka, N. Koibuchi, Y. Takeuchi, Mitotane induces CYP3A4 expression via activation of the steroid and xenobiotic receptor. J. Endocrinol. 216, 297–305 (2013)

    Article  CAS  PubMed  Google Scholar 

  20. P.G. Cuddy, L.S. Loftus, Influence of mitotane on the hypoprothrombinemic effect of warfarin. South. Med. J. 79, 387–388 (1986)

    Article  CAS  PubMed  Google Scholar 

  21. T. Peters, H. Lindenmaier, W.E. Haefeli, J. Weiss, Interaction of the mitotic kinesin Eg5 inhibitor monastrol with P-glycoprotein. Naunyn Schmiedebergs Arch. Pharmacol. 372, 291–299 (2006)

    Article  CAS  PubMed  Google Scholar 

  22. S. Harmsen, A.S. Koster, J.H. Beijnen, J.H. Schellens, I. Meijerman, Comparison of two immortalized human cell lines to study nuclear receptor-mediated CYP3A4 induction. Drug Metab. Dispos. 36, 1166–1171 (2008)

    Article  CAS  PubMed  Google Scholar 

  23. A. Gupta, G.M. Mugundu, P.B. Desai, K.E. Thummel, J.D. Unadkat, Intestinal human colon adenocarcinoma cell line LS180 is an excellent model to study pregnane X receptor, but not constitutive androstane receptor, mediated CYP3A4 and multidrug resistance transporter 1 induction: studies with anti-human immunodeficiency virus protease inhibitors. Drug Metab. Dispos. 36, 1172–1180 (2008)

    Article  CAS  PubMed  Google Scholar 

  24. H. Brandin, E. Viitanen, O. Myrberg, A.K. Arvidsson, Effects of herbal medicinal products and food supplements on induction of CYP1A2, CYP3A4 and MDR1 in the human colon carcinoma cell line LS180. Phytother. Res. 21, 239–244 (2007)

    Article  PubMed  Google Scholar 

  25. W. Li, P.A. Harper, B.K. Tang, A.B. Okey, Regulation of cytochrome P450 enzymes by aryl hydrocarbon receptor in human cells: CYP1A2 expression in the LS180 colon carcinoma cell line after treatment with 2,3,7,8-tetrachlorodibenzo-p-dioxin or 3-methylcholanthrene. Biochem. Pharmacol. 56, 599–612 (1998)

    Article  CAS  PubMed  Google Scholar 

  26. J. Weiss, M. Herzog, W.E. Haefeli, Differential modulation of the expression of important drug metabolising enzymes and transporters by endothelin-1 receptor antagonists ambrisentan and bosentan in vitro. Eur. J. Pharmacol. 660, 298–304 (2011)

    Article  CAS  PubMed  Google Scholar 

  27. N. Albermann, F.H. Schmitz-Winnenthal, K. Z’graggen, C. Volk, M.M. Hoffmann, W.E. Haefeli, J. Weiss, Expression of the drug transporters MDR1/ABCB1, MRP1/ABCC1, MRP2/ABCC2, BCRP/ABCG2, and PXR in peripheral blood mononuclear cells and their relationship with the expression in intestine and liver. Biochem. Pharmacol. 70, 949–958 (2005)

    Article  CAS  PubMed  Google Scholar 

  28. S.J. König, M. Herzog, D. Theile, N. Zembruski, W.E. Haefeli, J. Weiss, Impact of drug transporters for the cellular resistance towards saquinavir and darunavir. J. Antimic. Chemother. 65, 2319–2328 (2010)

    Article  Google Scholar 

  29. Z. Dvorak, R. Vrzal, P. Henklova, P. Jancova, E. Anzenbacherova, P. Maurel, L. Svecova, P. Pavek, J. Ehrmann, R. Havlik, P. Bednar, K. Lemr, J. Ulrichova, JNK inhibitor SP600125 is a partial agonist of human aryl hydrocarbon receptor and induces CYP1A1 and CYP1A2 genes in primary human hepatocytes. Biochem. Pharmacol. 75, 580–588 (2008)

    Article  CAS  PubMed  Google Scholar 

  30. I. Ayed-Boussema, J.M. Pascussi, P. Maurel, H. Bacha, W. Hassen, Zearalenone activates pregnane X receptor, constitutive androstane receptor and aryl hydrocarbon receptor and corresponding phase I target genes mRNA in primary cultures of human hepatocytes. Environ. Toxicol. Pharmacol. 31, 79–87 (2011)

    Article  CAS  PubMed  Google Scholar 

  31. J. Vandesompele, K. De Preter, F. Pattyn, B. Poppe, N. Van Roy, A. De Paepe, F. Speleman, Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol. 3, Research0034 (2002)

    Article  PubMed Central  PubMed  Google Scholar 

  32. J. Weiss, D. Theile, A. Spalwisz, J. Burhenne, K.D. Riedel, W.E. Haefeli, Influence of sildenafil and tadalafil on the enzyme- and transporter-inducing effects of bosentan and ambrisentan in LS180 cells. Biochem. Pharmacol. 85, 265–273 (2013)

    Article  CAS  PubMed  Google Scholar 

  33. A. Novotna, P. Pavek, Z. Dvorak, Novel stably transfected gene reporter human hepatoma cell line for assessment of aryl hydrocarbon receptor transcriptional activity: construction and characterization. Environ. Sci. Technol. 45, 10133–10139 (2011)

    Article  CAS  PubMed  Google Scholar 

  34. A.H. Schinkel, E. Wagenaar, C.A. Mol, L. van Deemter, P-glycoprotein in the blood-brain barrier of mice influences the brain penetration and pharmacological activity of many drugs. J. Clin. Invest. 97, 2517–2524 (1996)

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  35. D. Boesch, C. Gavériaux, B. Jachez, A. Pourtier-Manzanedo, P. Bollinger, F. Loor, In vivo circumvention of P-glycoprotein -mediated multidrug resistance of tumor cells with SDZ PSC 833. Cancer Res. 51, 4226–4233 (1991)

    CAS  PubMed  Google Scholar 

  36. J. Weiss, S.M. Dormann, M. Martin-Facklam, C.J. Kerpen, N. Ketabi-Kiyanvash, W.E. Haefeli, Inhibition of P-glycoprotein by newer antidepressants. J. Pharmacol. Exp. Ther. 305, 197–204 (2003)

    Article  CAS  PubMed  Google Scholar 

  37. M. Fröhlich, N. Albermann, A. Sauer, I. Walter-Sack, W.E. Haefeli, J. Weiss, In vitro and ex vivo evidence for modulation of P-glycoprotein activity by progestins. Biochem. Pharmacol. 68, 2409–2416 (2004)

    Article  PubMed  Google Scholar 

  38. J. Weiss, J. Rose, C.H. Storch, N. Ketabi-Kiyanvash, A. Sauer, W.E. Haefeli, T. Efferth, Modulation of human BCRP (ABCG2) activity by anti-HIV drugs. J. Antimicrob. Chemother. 59, 238–245 (2007)

    Article  CAS  PubMed  Google Scholar 

  39. J. König, Y. Cui, A.T. Nies, D. Keppler, Localization and genomic organization of a new hepatocellular organic anion transporting polypeptide. J. Biol. Chem. 275, 23161–23168 (2000)

    Article  PubMed  Google Scholar 

  40. J. König, Y. Cui, A.T. Nies, D. Keppler, A novel human organic anion transporting polypeptide localized to the basolateral hepatocyte membrane. Am. J. Physiol. Gastrointest. Liver Physiol. 278, G156–G164 (2000)

    PubMed  Google Scholar 

  41. A.H. Schinkel, E. Wagenaar, L. van Deemter, C.A. Mol, P. Borst, Absence of the mdr1a P-glycoprotein in mice affects tissue distribution and pharmacokinetics of dexamethasone, digoxin, and cyclosporin A. J. Clin. Invest. 96, 1698–1705 (1995)

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  42. P. Pavek, G. Merino, E. Wagenaar, E. Bolscher, M. Novotna, J.W. Jonker, A.H. Schinkel, Human breast cancer resistance protein: interactions with steroid drugs, hormones, the dietary carcinogen 2-amino-1-methyl-6-phenylimidazo(4,5-b)pyridine, and transport of cimetidine. J. Pharmacol. Exp. Ther. 312, 144–152 (2005)

    Article  CAS  PubMed  Google Scholar 

  43. N.C. Zembruski, G. Büchel, L. Jödicke, M. Herzog, W.E. Haefeli, J. Weiss, Potential of novel antiretrovirals to modulate expression and function of drug transporters in vitro. J. Antimicrob. Chemother. 66, 802–812 (2011)

    Article  CAS  PubMed  Google Scholar 

  44. M. Kroiss, M. Quinkler, W.K. Lutz, B. Allolio, M. Fassnacht, Drug interactions with mitotane by induction of CYP3A4 metabolism in the clinical management of adrenocortical carcinoma. Clin. Endocrinol. (Oxf) 75, 585–591 (2011)

    Article  CAS  Google Scholar 

  45. A. Geick, M. Eichelbaum, O. Burk, Nuclear receptor response elements mediate induction of intestinal MDR1 by rifampin. J. Biol. Chem. 276, 14581–14587 (2001)

    Article  CAS  PubMed  Google Scholar 

  46. W. Xie, M.F. Yeuh, A. Radominska-Pandya, S.P. Saini, Y. Negishi, B.S. Bottroff, G.Y. Cabrera, R.H. Tukey, R.M. Evans, Control of steroid, heme, and carcinogen metabolism by nuclear pregnane X receptor and constitutive androstane receptor. Proc. Natl. Acad. Sci. USA 100, 4150–4155 (2003)

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  47. D. Gardner-Stephen, J.M. Heydel, A. Goyal, Y. Lu, W. Xie, T. Lindblom, P. Mackenzie, A. Radominska-Pandya, Human PXR variants and their differential effects on the regulation of human UDP-glucuronosyltransferase gene expression. Drug Metab. Dispos. 32, 340–347 (2004)

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  48. H.R. Kast, B. Goodwin, P.T. Tarr, S.A. Jones, A.M. Anisfeld, C.M. Stoltz, P. Tontonoz, S. Kliewer, T.M. Willson, P.A. Edwards, Regulation of multidrug resistance-associated protein 2 (ABCC2) by the nuclear receptors pregnane X receptor, farnesoid X-activated receptor, and constitutive androstane receptor. J. Biol. Chem. 277, 2908–2915 (2002)

    Article  CAS  PubMed  Google Scholar 

  49. J. König, F. Müller, M.F. Fromm, Transporters and drug-drug interactions: important determinants of drug disposition and effects. Pharmacol. Rev. 65, 944–966 (2013)

    Article  PubMed  Google Scholar 

  50. B. Greiner, M. Eichelbaum, P. Fritz, H.-P. Kreichgauer, O. von Richter, J. Zundler, H.K. Kroemer, The role of intestinal P-glycoprotein in the interaction of digoxin and rifampin. J. Clin. Invest. 104, 147–153 (1999)

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  51. M. Ehle, C. Patel, R.P. Giugliano, Digoxin: clinical highlights: a review of digoxin and its use in contemporary medicine. Crit. Pathw. Cardiol. 10, 93–98 (2011)

    Article  PubMed  Google Scholar 

  52. A. Ohnishi, M. Kato, J. Kojima, H. Ushiama, M. Yoneko, H. Kawai, Differential pharmacokinetics of theophylline in elderly patients. Drugs Aging 20, 71–84 (2003)

    Article  CAS  PubMed  Google Scholar 

  53. L.S. Kaminsky, Z.Y. Zhang, Human P450 metabolism of warfarin. Pharmacol. Ther. 73, 67–74 (1997)

    Article  CAS  PubMed  Google Scholar 

  54. M. Ufer, Comparative pharmacokinetics of vitamin K antagonists: warfarin, phenprocoumon and acenocoumarol. Clin. Pharmacokinet. 44, 1227–1246 (2005)

    Article  CAS  PubMed  Google Scholar 

  55. K. Kim, O. Park, S. Hong, J.Y. Park, The effect of CYP2C19 polymorphism on the pharmacokinetics and pharmacodynamics of clopidogrel: a possible mechanism for clopidogrel resistance. Clin. Pharmacol. Ther. 84, 236–242 (2008)

    Article  CAS  PubMed  Google Scholar 

  56. T. Simon, C. Verstuyft, M. Mary-Krause, L. Quteineh, E. Drouet, N. Méneveau, P.G. Steg, J. Ferrières, N. Danchin, L. Becquemont, French registry of acute ST-elevation and non-ST-elevation myocardial infarction (FAST-MI) investigators. Genetic determinants of response to clopidogrel and cardiovascular events. N. Engl. J. Med. 360, 363–375 (2009)

    Article  CAS  PubMed  Google Scholar 

  57. S.E. Bates, C.Y. Shieh, L.A. Mickley, H.L. Dichek, A. Gazdar, D.L. Loriaux, A.T. Fojo, Mitotane enhances cytotoxicity of chemotherapy in cell lines expressing a multidrug resistance gene (mdr-1/P-glycoprotein) which is also expressed by adrenocortical carcinomas. J. Clin. Endocrinol. Metabol. 73, 18–29 (1991)

    Article  Google Scholar 

  58. T. Gagliano, E. Gentilin, K. Benfini, C. Di Pasquale, M. Tassinari, S. Falletta, C. Feo, F. Tagliati, E.D. Uberti, M.C. Zatelli, Mitotane enhances doxorubicin cytotoxic activity by inhibiting P-gp in human adrenocortical carcinoma cells. Endocrine 2014 (Epub ahead of print)

  59. M. Essodaigui, H.J. Broxterman, A. Garnier-Suillerot, Kinetic analysis of calcein and calcein-acetoxymethylester efflux mediated by the multidrug resistance protein and P-glycoprotein. Biochemistry 37, 2243–2250 (1998)

    Article  CAS  PubMed  Google Scholar 

  60. S.P. Cole, K.E. Sparks, K. Fraser, D.W. Loe, C.E. Grant, G.M. Wilson, R.G. Deeley, Pharmacological characterization of multidrug resistant MRP-transfected human tumor cells. Cancer Res. 54, 5902–5910 (1994)

    CAS  PubMed  Google Scholar 

  61. P. Matsson, J.M. Pedersen, U. Norinder, C.A. Bergström, P. Artursson, Identification of novel specific and general inhibitors of the three major human ATP-binding cassette transporters P-gp, BCRP and MRP2 among registered drugs. Pharm. Res. 26, 1816–1831 (2009)

    Article  CAS  PubMed  Google Scholar 

  62. J. Abraham, S. Bakke, A. Rutt, B. Meadows, M. Merino, R. Alexander, D. Schrump, D. Bartlett, P. Choyke, R. Robey, E. Hung, S.M. Steinberg, S. Bates, T. Fojo, A phase II trial of combination chemotherapy and surgical resection for the treatment of metastatic adrenocortical carcinoma: continuous infusion doxorubicin, vincristine, and etoposide with daily mitotane as a P-glycoprotein antagonist. Cancer 94, 2333–2343 (2002)

    Article  CAS  PubMed  Google Scholar 

  63. A. D’Avolio, S. De Francia, V. Basile, J. Cusato, F. De Martino, E. Pirro, F. Piccione, A. Ardito, B. Zaggia, M. Volante, G. Di Perri, M. Terzolo, Influence of the CYP2B6 polymorphism on the pharmacokinetics of mitotane. Pharmacogenet. Genomics 23, 293–300 (2013)

    Article  PubMed  Google Scholar 

  64. S.L. Plasschaert, E.S. de Bont, M. Boezen, D.M. vander Kolk, S.M. Daenen, K.N. Faber, W.A. Kamps, E.G. de Vries, E. Vellenga, Expression of multidrug resistance-associated proteins predicts prognosis in childhood and adult acute lymphoblastic leukemia. Clin. Cancer Res. 11, 8661–8668 (2005)

    Article  CAS  PubMed  Google Scholar 

  65. E. Jigorel, M. Le Vee, C. Boursier-Neyret, Y. Parmentier, O. Fardel, Differential regulation of sinusoidal and canalicular hepatic drug transporter expression by xenobiotics activating drug-sensing receptors in primary human hepatocytes. Drug Metab. Dispos. 34, 1756–1763 (2006)

    Article  CAS  PubMed  Google Scholar 

  66. N. Hariparsad, S.C. Nallani, R.S. Sane, D.J. Buckley, A.R. Buckley, P.B. Desai, Induction of CYP3A4 by efavirenz in primary human hepatocytes: comparison with rifampin and phenobarbital. J. Clin. Pharmacol. 44, 1273–1281 (2004)

    Article  CAS  PubMed  Google Scholar 

  67. V. Dixit, N. Hariparsad, F. Li, P. Desai, K.E. Thummel, J.D. Unadkat, Cytochrome P450 enzymes and transporters induced by anti-human immunodeficiency virus protease inhibitors in human hepatocytes: implications for predicting clinical drug interactions. Drug Metab. Dispos. 35, 1853–1859 (2007)

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

We would like to thank Dr. Johannes Pöschl for providing access to the flow cytometer. We also thank Corina Mueller, Jutta Kocher, and Stephanie Rosenzweig for excellent technical assistance and Dr. Andreas Meid for his help with statistics.

Conflict of interest

The authors declare that they have no conflict of interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Johanna Weiss.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (DOCX 45 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Theile, D., Haefeli, W.E. & Weiss, J. Effects of adrenolytic mitotane on drug elimination pathways assessed in vitro. Endocrine 49, 842–853 (2015). https://doi.org/10.1007/s12020-014-0517-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12020-014-0517-2

Keywords

Navigation