Skip to main content
Erschienen in: Cancer Chemotherapy and Pharmacology 1/2014

01.01.2014 | Original Article

Midazolam as a phenotyping probe to predict sunitinib exposure in patients with cancer

verfasst von: D. de Wit, H. Gelderblom, A. Sparreboom, J. den Hartigh, M. den Hollander, J. M. C. König-Quartel, T. Hessing, H. J. Guchelaar, N. P. van Erp

Erschienen in: Cancer Chemotherapy and Pharmacology | Ausgabe 1/2014

Einloggen, um Zugang zu erhalten

Abstract

Purpose

Patients treated with sunitinib show substantial inter-patient variability in drug exposure (~30–40 %), which is largely unexplained. Since sunitinib is metabolized by cytochrome P450(CYP)3A4, variability in the activity of this enzyme may explain a considerable proportion of this inter-patient variability. Midazolam is widely used as a phenotyping probe to assess CYP3A4-activity. The objective of this study was to prospectively evaluate the relationship between midazolam and sunitinib exposure. Additionally, the correlation between sunitinib trough levels and exposure and the influence of sunitinib on midazolam exposure was determined.

Methods

Thirteen patients treated with sunitinib in a 4 weeks “on”—2 weeks “off” regimen received twice 7.5 mg midazolam; once with and once without sunitinib. Steady-state sunitinib, its active metabolite SU12662 and midazolam exposures were determined.

Results

A significant correlation between midazolam exposure (AUC0–7h) and steady-state sunitinib and sunitinib + SU12662 exposure (AUC0–24h) was found (p = 0.006 and p = 0.0018, respectively); midazolam exposure explained 51 and 41 % of the inter-patient variability in sunitinib and sunitinib + SU12622 exposure. Furthermore, C trough was highly correlated (r 2 = 0.94) with sunitinib AUC0–24h. Sunitinib decreased midazolam exposure with 24 % (p = 0.034).

Conclusion

Midazolam exposure is highly correlated with sunitinib exposure and explains a large proportion of the observed inter-patient variability in sunitinib pharmacokinetics. Consequently, midazolam could be used to identify patients that are at risk of under- or overtreatment, respectively, at the start of sunitinib therapy. Moreover, sunitinib and sunitinib + SU12662 trough levels are highly correlated with drug exposure and can thus be used in clinical practice to individualize sunitinib therapy. The decrease in midazolam exposure by sunitinib needs further investigation.
Literatur
1.
Zurück zum Zitat Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, Oudard S, Negrier S, Szczylik C, Kim ST, Chen I, Bycott PW, Baum CM, Figlin RA (2007) Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med 356:115–124PubMedCrossRef Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, Oudard S, Negrier S, Szczylik C, Kim ST, Chen I, Bycott PW, Baum CM, Figlin RA (2007) Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med 356:115–124PubMedCrossRef
2.
Zurück zum Zitat Demetri GD, van Oosterom AT, Garrett CR, Blackstein ME, Shah MH, Verweij J, McArthur G, Judson IR, Heinrich MC, Morgan JA, Desai J, Fletcher CD, George S, Bello CL, Huang X, Baum CM, Casali PG (2006) Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial. Lancet 368:1329–1338PubMedCrossRef Demetri GD, van Oosterom AT, Garrett CR, Blackstein ME, Shah MH, Verweij J, McArthur G, Judson IR, Heinrich MC, Morgan JA, Desai J, Fletcher CD, George S, Bello CL, Huang X, Baum CM, Casali PG (2006) Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial. Lancet 368:1329–1338PubMedCrossRef
3.
Zurück zum Zitat Goodman VL, Rock EP, Dagher R, Ramchandani RP, Abraham S, Gobburu JV, Booth BP, Verbois SL, Morse DE, Liang CY, Chidambaram N, Jiang JX, Tang S, Mahjoob K, Justice R, Pazdur R (2007) Approval summary: sunitinib for the treatment of imatinib refractory or intolerant gastrointestinal stromal tumors and advanced renal cell carcinoma. Clin Cancer Res 13:1367–1373PubMedCrossRef Goodman VL, Rock EP, Dagher R, Ramchandani RP, Abraham S, Gobburu JV, Booth BP, Verbois SL, Morse DE, Liang CY, Chidambaram N, Jiang JX, Tang S, Mahjoob K, Justice R, Pazdur R (2007) Approval summary: sunitinib for the treatment of imatinib refractory or intolerant gastrointestinal stromal tumors and advanced renal cell carcinoma. Clin Cancer Res 13:1367–1373PubMedCrossRef
4.
Zurück zum Zitat Raymond E, Dahan L, Raoul JL, Bang YJ, Borbath I, Lombard-Bohas C, Valle J, Metrakos P, Smith D, Vinik A, Chen JS, Horsch D, Hammel P, Wiedenmann B, Van CE, Patyna S, Lu DR, Blanckmeister C, Chao R, Ruszniewski P (2011) Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med 364:501–513PubMedCrossRef Raymond E, Dahan L, Raoul JL, Bang YJ, Borbath I, Lombard-Bohas C, Valle J, Metrakos P, Smith D, Vinik A, Chen JS, Horsch D, Hammel P, Wiedenmann B, Van CE, Patyna S, Lu DR, Blanckmeister C, Chao R, Ruszniewski P (2011) Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med 364:501–513PubMedCrossRef
5.
Zurück zum Zitat Adams VR, Leggas M (2007) Sunitinib malate for the treatment of metastatic renal cell carcinoma and gastrointestinal stromal tumors. Clin Ther 29:1338–1353PubMedCrossRef Adams VR, Leggas M (2007) Sunitinib malate for the treatment of metastatic renal cell carcinoma and gastrointestinal stromal tumors. Clin Ther 29:1338–1353PubMedCrossRef
7.
Zurück zum Zitat Houk BE, Bello CL, Kang D, Amantea M (2009) A population pharmacokinetic meta-analysis of sunitinib malate (SU11248) and its primary metabolite (SU12662) in healthy volunteers and oncology patients. Clin Cancer Res 15:2497–2506PubMedCrossRef Houk BE, Bello CL, Kang D, Amantea M (2009) A population pharmacokinetic meta-analysis of sunitinib malate (SU11248) and its primary metabolite (SU12662) in healthy volunteers and oncology patients. Clin Cancer Res 15:2497–2506PubMedCrossRef
8.
Zurück zum Zitat Speed B, Bu HZ, Pool WF, Peng GW, Wu EY, Patyna S, Bello C, Kang P (2012) Pharmacokinetics, distribution, and metabolism of [14C]sunitinib in rats, monkeys, and humans. Drug Metab Dispos 40:539–555PubMedCrossRef Speed B, Bu HZ, Pool WF, Peng GW, Wu EY, Patyna S, Bello C, Kang P (2012) Pharmacokinetics, distribution, and metabolism of [14C]sunitinib in rats, monkeys, and humans. Drug Metab Dispos 40:539–555PubMedCrossRef
9.
Zurück zum Zitat Houk BE, Bello CL, Poland B, Rosen LS, Demetri GD, Motzer RJ (2010) Relationship between exposure to sunitinib and efficacy and tolerability endpoints in patients with cancer: results of a pharmacokinetic/pharmacodynamic meta-analysis. Cancer Chemother Pharmacol 66:357–371PubMedCrossRef Houk BE, Bello CL, Poland B, Rosen LS, Demetri GD, Motzer RJ (2010) Relationship between exposure to sunitinib and efficacy and tolerability endpoints in patients with cancer: results of a pharmacokinetic/pharmacodynamic meta-analysis. Cancer Chemother Pharmacol 66:357–371PubMedCrossRef
10.
Zurück zum Zitat Puisset F, Dalenc F, Chatelut E, Cresteil T, Lochon I, Tisnes P, Roche H (2005) Dexamethasone as a probe for vinorelbine clearance. Br J Clin Pharmacol 60:45–53PubMedCrossRef Puisset F, Dalenc F, Chatelut E, Cresteil T, Lochon I, Tisnes P, Roche H (2005) Dexamethasone as a probe for vinorelbine clearance. Br J Clin Pharmacol 60:45–53PubMedCrossRef
11.
Zurück zum Zitat de Graan AJ, Teunissen SF, de Vos FY, Loos WJ, van Schaik RH, de Jongh FE, de Vos AI, van Alphen RJ, van der Holt B, Verweij J, Seynaeve C, Beijnen JH, Mathijssen RH (2011) Dextromethorphan as a phenotyping test to predict endoxifen exposure in patients on tamoxifen treatment. J Clin Oncol 29:3240–3246PubMedCrossRef de Graan AJ, Teunissen SF, de Vos FY, Loos WJ, van Schaik RH, de Jongh FE, de Vos AI, van Alphen RJ, van der Holt B, Verweij J, Seynaeve C, Beijnen JH, Mathijssen RH (2011) Dextromethorphan as a phenotyping test to predict endoxifen exposure in patients on tamoxifen treatment. J Clin Oncol 29:3240–3246PubMedCrossRef
12.
Zurück zum Zitat Goh BC, Lee SC, Wang LZ, Fan L, Guo JY, Lamba J, Schuetz E, Lim R, Lim HL, Ong AB, Lee HS (2002) Explaining interindividual variability of docetaxel pharmacokinetics and pharmacodynamics in Asians through phenotyping and genotyping strategies. J Clin Oncol 20:3683–3690PubMedCrossRef Goh BC, Lee SC, Wang LZ, Fan L, Guo JY, Lamba J, Schuetz E, Lim R, Lim HL, Ong AB, Lee HS (2002) Explaining interindividual variability of docetaxel pharmacokinetics and pharmacodynamics in Asians through phenotyping and genotyping strategies. J Clin Oncol 20:3683–3690PubMedCrossRef
13.
Zurück zum Zitat Mathijssen RH, de Jong FA, van Schaik RH, Lepper ER, Friberg LE, Rietveld T, de Bruijn P, Graveland WJ, Figg WD, Verweij J, Sparreboom A (2004) Prediction of irinotecan pharmacokinetics by use of cytochrome P450 3A4 phenotyping probes. J Natl Cancer Inst 96:1585–1592PubMedCrossRef Mathijssen RH, de Jong FA, van Schaik RH, Lepper ER, Friberg LE, Rietveld T, de Bruijn P, Graveland WJ, Figg WD, Verweij J, Sparreboom A (2004) Prediction of irinotecan pharmacokinetics by use of cytochrome P450 3A4 phenotyping probes. J Natl Cancer Inst 96:1585–1592PubMedCrossRef
14.
Zurück zum Zitat Li J, Karlsson MO, Brahmer J, Spitz A, Zhao M, Hidalgo M, Baker SD (2006) CYP3A phenotyping approach to predict systemic exposure to EGFR tyrosine kinase inhibitors. J Natl Cancer Inst 98:1714–1723PubMedCrossRef Li J, Karlsson MO, Brahmer J, Spitz A, Zhao M, Hidalgo M, Baker SD (2006) CYP3A phenotyping approach to predict systemic exposure to EGFR tyrosine kinase inhibitors. J Natl Cancer Inst 98:1714–1723PubMedCrossRef
15.
Zurück zum Zitat Swaisland HC, Cantarini MV, Fuhr R, Holt A (2006) Exploring the relationship between expression of cytochrome P450 enzymes and gefitinib pharmacokinetics. Clin Pharmacokinet 45:633–644PubMedCrossRef Swaisland HC, Cantarini MV, Fuhr R, Holt A (2006) Exploring the relationship between expression of cytochrome P450 enzymes and gefitinib pharmacokinetics. Clin Pharmacokinet 45:633–644PubMedCrossRef
16.
Zurück zum Zitat Opdam FL, Dezentje VO, den HJ, Modak AS, Vree R, Batman E, Smorenburg CH, Nortier JW, Gelderblom H, Guchelaar HJ (2013) The use of the 13C-dextromethorphan breath test for phenotyping CYP2D6 in breast cancer patients using tamoxifen: association with CYP2D6 genotype and serum endoxifen levels. Cancer Chemother Pharmacol 71(3):593–601 Opdam FL, Dezentje VO, den HJ, Modak AS, Vree R, Batman E, Smorenburg CH, Nortier JW, Gelderblom H, Guchelaar HJ (2013) The use of the 13C-dextromethorphan breath test for phenotyping CYP2D6 in breast cancer patients using tamoxifen: association with CYP2D6 genotype and serum endoxifen levels. Cancer Chemother Pharmacol 71(3):593–601
17.
Zurück zum Zitat Gurney H, Wong M, Balleine RL, Rivory LP, McLachlan AJ, Hoskins JM, Wilcken N, Clarke CL, Mann GJ, Collins M, Delforce SE, Lynch K, Schran H (2007) Imatinib disposition and ABCB1 (MDR1, P-glycoprotein) genotype. Clin Pharmacol Ther 82:33–40PubMedCrossRef Gurney H, Wong M, Balleine RL, Rivory LP, McLachlan AJ, Hoskins JM, Wilcken N, Clarke CL, Mann GJ, Collins M, Delforce SE, Lynch K, Schran H (2007) Imatinib disposition and ABCB1 (MDR1, P-glycoprotein) genotype. Clin Pharmacol Ther 82:33–40PubMedCrossRef
18.
Zurück zum Zitat Hirth J, Watkins PB, Strawderman M, Schott A, Bruno R, Baker LH (2000) The effect of an individual’s cytochrome CYP3A4 activity on docetaxel clearance. Clin Cancer Res 6:1255–1258PubMed Hirth J, Watkins PB, Strawderman M, Schott A, Bruno R, Baker LH (2000) The effect of an individual’s cytochrome CYP3A4 activity on docetaxel clearance. Clin Cancer Res 6:1255–1258PubMed
19.
Zurück zum Zitat Slaviero KA, Clarke SJ, McLachlan AJ, Blair EY, Rivory LP (2004) Population pharmacokinetics of weekly docetaxel in patients with advanced cancer. Br J Clin Pharmacol 57:44–53PubMedCrossRef Slaviero KA, Clarke SJ, McLachlan AJ, Blair EY, Rivory LP (2004) Population pharmacokinetics of weekly docetaxel in patients with advanced cancer. Br J Clin Pharmacol 57:44–53PubMedCrossRef
20.
Zurück zum Zitat Opdam FL, Gelderblom H, Guchelaar H-J (2012) Phenotyping drug disposition in oncology. Cancer Treat Rev 38:715–725PubMedCrossRef Opdam FL, Gelderblom H, Guchelaar H-J (2012) Phenotyping drug disposition in oncology. Cancer Treat Rev 38:715–725PubMedCrossRef
21.
Zurück zum Zitat van Erp NP, Gelderblom H, Guchelaar HJ (2009) Clinical pharmacokinetics of tyrosine kinase inhibitors. Cancer Treat Rev 35:692–706PubMedCrossRef van Erp NP, Gelderblom H, Guchelaar HJ (2009) Clinical pharmacokinetics of tyrosine kinase inhibitors. Cancer Treat Rev 35:692–706PubMedCrossRef
22.
Zurück zum Zitat van Erp NP, de WD, Guchelaar HJ, Gelderblom H, Hessing TJ, Hartigh JD (2013) A validated assay for the simultaneous quantification of six tyrosine kinase inhibitors and two active metabolites in human serum using liquid chromatography coupled with tandem mass spectrometry. J. Chromatogr.B Analyt. Technol Biomed Life Sci 937C:33–43CrossRef van Erp NP, de WD, Guchelaar HJ, Gelderblom H, Hessing TJ, Hartigh JD (2013) A validated assay for the simultaneous quantification of six tyrosine kinase inhibitors and two active metabolites in human serum using liquid chromatography coupled with tandem mass spectrometry. J. Chromatogr.B Analyt. Technol Biomed Life Sci 937C:33–43CrossRef
23.
Zurück zum Zitat van Erp NP, Guchelaar HJ, Ploeger BA, Romijn JA, Hartigh J, Gelderblom H (2011) Mitotane has a strong and a durable inducing effect on CYP3A4 activity. Eur J Endocrinol 164:621–626PubMedCrossRef van Erp NP, Guchelaar HJ, Ploeger BA, Romijn JA, Hartigh J, Gelderblom H (2011) Mitotane has a strong and a durable inducing effect on CYP3A4 activity. Eur J Endocrinol 164:621–626PubMedCrossRef
24.
Zurück zum Zitat Takeshita A, Igarashi-Migitaka J, Koibuchi N, Takeuchi Y (2013) Mitotane induces CYP3A4 expression via activation of the steroid and xenobiotic receptor. J Endocrinol 216:297–305PubMedCrossRef Takeshita A, Igarashi-Migitaka J, Koibuchi N, Takeuchi Y (2013) Mitotane induces CYP3A4 expression via activation of the steroid and xenobiotic receptor. J Endocrinol 216:297–305PubMedCrossRef
25.
Zurück zum Zitat Chortis V, Taylor AE, Schneider P, Tomlinson JW, Hughes BA, O’Neil DM, Libe R, Allolio B, Bertagna X, Bertherat J, Beuschlein F, Fassnacht M, Karavitaki N, Mannelli M, Mantero F, Opocher G, Porfiri E, Quinkler M, Sherlock M, Terzolo M, Nightingale P, Shackleton CH, Stewart PM, Hahner S, Arlt W (2013) Mitotane therapy in adrenocortical cancer induces CYP3A4 and inhibits 5alpha-reductase, explaining the need for personalized glucocorticoid and androgen replacement. J Clin Endocrinol Metab 98:161–171PubMedCrossRef Chortis V, Taylor AE, Schneider P, Tomlinson JW, Hughes BA, O’Neil DM, Libe R, Allolio B, Bertagna X, Bertherat J, Beuschlein F, Fassnacht M, Karavitaki N, Mannelli M, Mantero F, Opocher G, Porfiri E, Quinkler M, Sherlock M, Terzolo M, Nightingale P, Shackleton CH, Stewart PM, Hahner S, Arlt W (2013) Mitotane therapy in adrenocortical cancer induces CYP3A4 and inhibits 5alpha-reductase, explaining the need for personalized glucocorticoid and androgen replacement. J Clin Endocrinol Metab 98:161–171PubMedCrossRef
26.
Zurück zum Zitat Kroiss M, Quinkler M, Lutz WK, Allolio B, Fassnacht M (2011) Drug interactions with mitotane by induction of CYP3A4 metabolism in the clinical management of adrenocortical carcinoma. Clin Endocrinol (Oxf) 75:585–591CrossRef Kroiss M, Quinkler M, Lutz WK, Allolio B, Fassnacht M (2011) Drug interactions with mitotane by induction of CYP3A4 metabolism in the clinical management of adrenocortical carcinoma. Clin Endocrinol (Oxf) 75:585–591CrossRef
27.
Zurück zum Zitat Faivre S, Delbaldo C, Vera K, Robert C, Lozahic S, Lassau N, Bello C, Deprimo S, Brega N, Massimini G, Armand JP, Scigalla P, Raymond E (2006) Safety, pharmacokinetic, and antitumor activity of SU11248, a novel oral multitarget tyrosine kinase inhibitor, in patients with cancer. J Clin Oncol 24:25–35PubMedCrossRef Faivre S, Delbaldo C, Vera K, Robert C, Lozahic S, Lassau N, Bello C, Deprimo S, Brega N, Massimini G, Armand JP, Scigalla P, Raymond E (2006) Safety, pharmacokinetic, and antitumor activity of SU11248, a novel oral multitarget tyrosine kinase inhibitor, in patients with cancer. J Clin Oncol 24:25–35PubMedCrossRef
28.
Zurück zum Zitat T. Mizuno, M. Fukudo, T. Terada, T. Kamba, E. Nakamura, O. Ogawa, KI. Inui, T. Katsura (2012) Impact of genetic variation in breast cancer resistance protein (BCRP/ABCG2) on sunitinib pharmacokinetics. Drug Metab Pharmacokinet 27(6):631–639PubMedCrossRef T. Mizuno, M. Fukudo, T. Terada, T. Kamba, E. Nakamura, O. Ogawa, KI. Inui, T. Katsura (2012) Impact of genetic variation in breast cancer resistance protein (BCRP/ABCG2) on sunitinib pharmacokinetics. Drug Metab Pharmacokinet 27(6):631–639PubMedCrossRef
29.
Zurück zum Zitat Mizuno T, Terada T, Kamba T, Fukudo M, Katsura T, Nakamura E, Ogawa O, Inui K (2010) ABCG2 421C > A polymorphism and high exposure of sunitinib in a patient with renal cell carcinoma. Ann Oncol 21:1382–1383PubMedCrossRef Mizuno T, Terada T, Kamba T, Fukudo M, Katsura T, Nakamura E, Ogawa O, Inui K (2010) ABCG2 421C > A polymorphism and high exposure of sunitinib in a patient with renal cell carcinoma. Ann Oncol 21:1382–1383PubMedCrossRef
30.
Zurück zum Zitat van Schaik RH, van der Heiden IP, van den Anker JN, Lindemans J (2002) CYP3A5 variant allele frequencies in Dutch Caucasians. Clin Chem 48:1668–1671PubMed van Schaik RH, van der Heiden IP, van den Anker JN, Lindemans J (2002) CYP3A5 variant allele frequencies in Dutch Caucasians. Clin Chem 48:1668–1671PubMed
31.
Zurück zum Zitat Mendel DB, Laird AD, Xin X, Louie SG, Christensen JG, Li G, Schreck RE, Abrams TJ, Ngai TJ, Lee LB, Murray LJ, Carver J, Chan E, Moss KG, Haznedar JO, Sukbuntherng J, Blake RA, Sun L, Tang C, Miller T, Shirazian S, McMahon G, Cherrington JM (2003) In vivo antitumor activity of SU11248, a novel tyrosine kinase inhibitor targeting vascular endothelial growth factor and platelet-derived growth factor receptors: determination of a pharmacokinetic/pharmacodynamic relationship. Clin Cancer Res 9:327–337PubMed Mendel DB, Laird AD, Xin X, Louie SG, Christensen JG, Li G, Schreck RE, Abrams TJ, Ngai TJ, Lee LB, Murray LJ, Carver J, Chan E, Moss KG, Haznedar JO, Sukbuntherng J, Blake RA, Sun L, Tang C, Miller T, Shirazian S, McMahon G, Cherrington JM (2003) In vivo antitumor activity of SU11248, a novel tyrosine kinase inhibitor targeting vascular endothelial growth factor and platelet-derived growth factor receptors: determination of a pharmacokinetic/pharmacodynamic relationship. Clin Cancer Res 9:327–337PubMed
32.
Zurück zum Zitat Kozloff M, Chuang E, Starr A, Gowland PA, Cataruozolo PE, Collier M, Verkh L, Huang X, Kern KA, Miller K (2010) An exploratory study of sunitinib plus paclitaxel as first-line treatment for patients with advanced breast cancer. Ann Oncol 21:1436–1441PubMedCrossRef Kozloff M, Chuang E, Starr A, Gowland PA, Cataruozolo PE, Collier M, Verkh L, Huang X, Kern KA, Miller K (2010) An exploratory study of sunitinib plus paclitaxel as first-line treatment for patients with advanced breast cancer. Ann Oncol 21:1436–1441PubMedCrossRef
33.
Zurück zum Zitat Robert F, Sandler A, Schiller JH, Liu G, Harper K, Verkh L, Huang X, Ilagan J, Tye L, Chao R, Traynor AM (2010) Sunitinib in combination with docetaxel in patients with advanced solid tumors: a phase I dose-escalation study. Cancer Chemother Pharmacol 66:669–680PubMedCentralPubMedCrossRef Robert F, Sandler A, Schiller JH, Liu G, Harper K, Verkh L, Huang X, Ilagan J, Tye L, Chao R, Traynor AM (2010) Sunitinib in combination with docetaxel in patients with advanced solid tumors: a phase I dose-escalation study. Cancer Chemother Pharmacol 66:669–680PubMedCentralPubMedCrossRef
34.
Zurück zum Zitat de Jonge MJ, Dumez H, Kitzen JJ, Beuselinck B, Verweij J, Courtney R, Battista A, Brega N, Schoffski P (2011) Phase I safety and pharmacokinetic study of SU-014813 in combination with docetaxel in patients with advanced solid tumours. Eur J Cancer 47:1328–1335PubMedCrossRef de Jonge MJ, Dumez H, Kitzen JJ, Beuselinck B, Verweij J, Courtney R, Battista A, Brega N, Schoffski P (2011) Phase I safety and pharmacokinetic study of SU-014813 in combination with docetaxel in patients with advanced solid tumours. Eur J Cancer 47:1328–1335PubMedCrossRef
35.
Zurück zum Zitat Boven E, Massard C, Armand JP, Tillier C, Hartog V, Brega NM, Countouriotis AM, Ruiz-Garcia A, Soria JC (2010) A phase I, dose-finding study of sunitinib in combination with irinotecan in patients with advanced solid tumours. Br J Cancer 103:993–1000PubMedCentralPubMedCrossRef Boven E, Massard C, Armand JP, Tillier C, Hartog V, Brega NM, Countouriotis AM, Ruiz-Garcia A, Soria JC (2010) A phase I, dose-finding study of sunitinib in combination with irinotecan in patients with advanced solid tumours. Br J Cancer 103:993–1000PubMedCentralPubMedCrossRef
36.
Zurück zum Zitat Sugiyama M, Fujita K, Murayama N, Akiyama Y, Yamazaki H, Sasaki Y (2011) Sorafenib and sunitinib, two anticancer drugs, inhibit CYP3A4-mediated and activate CY3A5-mediated midazolam 1′-hydroxylation. Drug Metab Dispos 39:757–762PubMedCrossRef Sugiyama M, Fujita K, Murayama N, Akiyama Y, Yamazaki H, Sasaki Y (2011) Sorafenib and sunitinib, two anticancer drugs, inhibit CYP3A4-mediated and activate CY3A5-mediated midazolam 1′-hydroxylation. Drug Metab Dispos 39:757–762PubMedCrossRef
Metadaten
Titel
Midazolam as a phenotyping probe to predict sunitinib exposure in patients with cancer
verfasst von
D. de Wit
H. Gelderblom
A. Sparreboom
J. den Hartigh
M. den Hollander
J. M. C. König-Quartel
T. Hessing
H. J. Guchelaar
N. P. van Erp
Publikationsdatum
01.01.2014
Verlag
Springer Berlin Heidelberg
Erschienen in
Cancer Chemotherapy and Pharmacology / Ausgabe 1/2014
Print ISSN: 0344-5704
Elektronische ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-013-2322-7

Weitere Artikel der Ausgabe 1/2014

Cancer Chemotherapy and Pharmacology 1/2014 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.