Skip to main content
Erschienen in: Cancer Chemotherapy and Pharmacology 4/2016

01.09.2016 | Original Article

Association of axitinib plasma exposure and genetic polymorphisms of ABC transporters with axitinib-induced toxicities in patients with renal cell carcinoma

verfasst von: Hiroshi Kato, Naoto Sassa, Masayuki Miyazaki, Mio Takeuchi, Miho Asai, Akane Iwai, Yukihiro Noda, Momokazu Gotoh, Kiyofumi Yamada

Erschienen in: Cancer Chemotherapy and Pharmacology | Ausgabe 4/2016

Einloggen, um Zugang zu erhalten

Abstract

Purpose

Axitinib is a selective tyrosine kinase inhibitor of VEGF receptors, approved for advanced renal cell carcinoma (RCC). Associations between axitinib plasma exposure, genetic polymorphisms of ABC transporters and axitinib-induced toxicities have not been adequately explored.

Methods

Twenty RCC patients treated with axitinib were enrolled in this study. Blood samples were collected 0, 0.5, 1, 2, 4, and 6 h after administration of axitinib on day 1 and at steady state. Plasma concentrations of axitinib were analyzed by UPLC–MS/MS. The ABCG2 (421C>A) and ABCB1 (1236C>T, 2677G>T/A, 3435C>T) genetic polymorphisms were determined by real-time PCR.

Results

ABCB1 haplotype was associated with increased dose-adjusted area under the plasma concentration–time curve (AUC) of axitinib at steady state. The incidence of fatigue during therapy was associated with high AUC0–6 of axitinib (P = 0.013). The treatment period without discontinuation or dose reduction due to adverse events in patients with high AUC0–6 of axitinib was significantly shorter than for those with low AUC0–6 (P = 0.024). No significant differences were found in the frequency of adverse events among the ABCG2 genotype and ABCB1 haplotype groups.

Conclusions

Our results have demonstrated that adverse events leading to discontinuation or dose reduction in axitinib were associated with increased axitinib plasma exposure, but not directly with genetic polymorphisms of ABC transporters. Therefore, measurement of steady state axitinib plasma concentrations may be useful in avoiding adverse events in axitinib therapy.
Literatur
1.
Zurück zum Zitat Hu-Lowe DD, Zou HY, Grazzini ML et al (2008) Nonclinical antiangiogenesis and antitumor activities of axitinib (AG-013736), an oral, potent, and selective inhibitor of vascular endothelial growth factor receptor tyrosine kinases 1, 2, 3. Clin Cancer Res 14:7272–7283CrossRefPubMed Hu-Lowe DD, Zou HY, Grazzini ML et al (2008) Nonclinical antiangiogenesis and antitumor activities of axitinib (AG-013736), an oral, potent, and selective inhibitor of vascular endothelial growth factor receptor tyrosine kinases 1, 2, 3. Clin Cancer Res 14:7272–7283CrossRefPubMed
2.
Zurück zum Zitat Rini BI, Garrett M, Poland B et al (2013) Axitinib in metastatic renal cell carcinoma: results of a pharmacokinetic and pharmacodynamic analysis. J Clin Pharmacol 53:491–504CrossRefPubMedPubMedCentral Rini BI, Garrett M, Poland B et al (2013) Axitinib in metastatic renal cell carcinoma: results of a pharmacokinetic and pharmacodynamic analysis. J Clin Pharmacol 53:491–504CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Noda S, Otsuji T, Baba M et al (2015) Assessment of sunitinib-induced toxicities and clinical outcomes based on therapeutic drug monitoring of sunitinib for patients with renal cell carcinoma. Clin Genitourin Cancer 13:350–358CrossRefPubMed Noda S, Otsuji T, Baba M et al (2015) Assessment of sunitinib-induced toxicities and clinical outcomes based on therapeutic drug monitoring of sunitinib for patients with renal cell carcinoma. Clin Genitourin Cancer 13:350–358CrossRefPubMed
4.
Zurück zum Zitat Terada T, Hira D (2015) Intestinal and hepatic drug transporters: pharmacokinetic, pathophysiological, and pharmacogenetic roles. J Gastroenterol 50:508–519CrossRefPubMed Terada T, Hira D (2015) Intestinal and hepatic drug transporters: pharmacokinetic, pathophysiological, and pharmacogenetic roles. J Gastroenterol 50:508–519CrossRefPubMed
5.
Zurück zum Zitat Imai Y, Nakane M, Kage K et al (2002) C421A polymorphism in the human breast cancer resistance protein gene is associated with low expression of Q141 K protein and low-level drug resistance. Mol Cancer Ther 1:611–616PubMed Imai Y, Nakane M, Kage K et al (2002) C421A polymorphism in the human breast cancer resistance protein gene is associated with low expression of Q141 K protein and low-level drug resistance. Mol Cancer Ther 1:611–616PubMed
6.
Zurück zum Zitat Kimchi-Sarfaty C, Oh JM, Kim IW et al (2007) A “silent” polymorphism in the MDR1 gene changes substrate specificity. Science 315:525–528CrossRefPubMed Kimchi-Sarfaty C, Oh JM, Kim IW et al (2007) A “silent” polymorphism in the MDR1 gene changes substrate specificity. Science 315:525–528CrossRefPubMed
7.
Zurück zum Zitat Mizuno T, Fukudo M, Terada T et al (2012) Impact of genetic variation in breast cancer resistance protein (BCRP/ABCG2) on sunitinib pharmacokinetics. Drug Metab Pahrmacokinet 27:631–639CrossRef Mizuno T, Fukudo M, Terada T et al (2012) Impact of genetic variation in breast cancer resistance protein (BCRP/ABCG2) on sunitinib pharmacokinetics. Drug Metab Pahrmacokinet 27:631–639CrossRef
8.
Zurück zum Zitat Hamada A, Sasaki J, Saeki S et al (2012) Association of ABCB1 polymorphisms with erlotinib pharmacokinetics and toxicity in Japanese patients with non-small-cell lung cancer. Pharmacogenomics 13:615–624CrossRefPubMed Hamada A, Sasaki J, Saeki S et al (2012) Association of ABCB1 polymorphisms with erlotinib pharmacokinetics and toxicity in Japanese patients with non-small-cell lung cancer. Pharmacogenomics 13:615–624CrossRefPubMed
9.
Zurück zum Zitat Poller B, Iusuf D, Sparidans RW et al (2011) Differential impact of P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) on axitinib brain accumulation and oral plasma pharmacokinetics. Drug Metab Dispos 39:729–735CrossRefPubMed Poller B, Iusuf D, Sparidans RW et al (2011) Differential impact of P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) on axitinib brain accumulation and oral plasma pharmacokinetics. Drug Metab Dispos 39:729–735CrossRefPubMed
10.
Zurück zum Zitat Brennan M, Williams JA, Chen Y et al (2012) Meta-analysis of contribution of genetic polymorphisms in drug-metabolizing enzymes or transporters to axitinib pharmacokinetics. Eur J Clin Pharmacol 68:645–655CrossRefPubMed Brennan M, Williams JA, Chen Y et al (2012) Meta-analysis of contribution of genetic polymorphisms in drug-metabolizing enzymes or transporters to axitinib pharmacokinetics. Eur J Clin Pharmacol 68:645–655CrossRefPubMed
11.
Zurück zum Zitat Rini BI, Escudier B, Tomczak P et al (2011) Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Lancet 378:1931–1939CrossRefPubMed Rini BI, Escudier B, Tomczak P et al (2011) Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Lancet 378:1931–1939CrossRefPubMed
12.
Zurück zum Zitat Rini BI, de La Motte RT, Harzstark AL et al (2013) Five-year survival in patients with cytokine-refractory metastatic renal cell carcinoma treated with axitinib. Clin Genitourin Cancer 11:107–114CrossRefPubMed Rini BI, de La Motte RT, Harzstark AL et al (2013) Five-year survival in patients with cytokine-refractory metastatic renal cell carcinoma treated with axitinib. Clin Genitourin Cancer 11:107–114CrossRefPubMed
13.
Zurück zum Zitat Chen Y, Jiang J, Zhang J et al (2011) A Phase I study to evaluate the pharmacokinetics of axitinib (AG-13736) in healthy Chinese volunteers. Int J Clin Pharmacol Ther 49:679–687CrossRefPubMed Chen Y, Jiang J, Zhang J et al (2011) A Phase I study to evaluate the pharmacokinetics of axitinib (AG-13736) in healthy Chinese volunteers. Int J Clin Pharmacol Ther 49:679–687CrossRefPubMed
14.
Zurück zum Zitat Delbaldo C, Chatelut E, Ré M et al (2006) Pharmacokinetic-pharmacodynamic relationships of imatinib and its main metabolite in patients with advanced gastrointestinal stromal tumors. Clin Cancer Res 12:6073–6078CrossRefPubMed Delbaldo C, Chatelut E, Ré M et al (2006) Pharmacokinetic-pharmacodynamic relationships of imatinib and its main metabolite in patients with advanced gastrointestinal stromal tumors. Clin Cancer Res 12:6073–6078CrossRefPubMed
15.
Zurück zum Zitat Larson RA, Yin OQ, Hochhaus A et al (2012) Population pharmacokinetic and exposure-response analysis of nilotinib in patients with newly diagnosed Ph + chronic myeloid leukemia in chronic phase. Eur J Clin Pharmacol 68:723–733CrossRefPubMed Larson RA, Yin OQ, Hochhaus A et al (2012) Population pharmacokinetic and exposure-response analysis of nilotinib in patients with newly diagnosed Ph + chronic myeloid leukemia in chronic phase. Eur J Clin Pharmacol 68:723–733CrossRefPubMed
16.
Zurück zum Zitat Houk BE, Bello CL, Poland B et al (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 66:357–371CrossRefPubMed Houk BE, Bello CL, Poland B et al (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 66:357–371CrossRefPubMed
17.
Zurück zum Zitat Fukudo M, Ikemi Y, Togashi Y et al (2013) Population pharmacokinetics/pharmacodynamics of erlotinib and pharmacogenomic analysis of plasma and cerebrospinal fluid drug concentrations in Japanese patients with non-small cell lung cancer. Clin Pharmacokinet 52:593–609CrossRefPubMed Fukudo M, Ikemi Y, Togashi Y et al (2013) Population pharmacokinetics/pharmacodynamics of erlotinib and pharmacogenomic analysis of plasma and cerebrospinal fluid drug concentrations in Japanese patients with non-small cell lung cancer. Clin Pharmacokinet 52:593–609CrossRefPubMed
18.
Zurück zum Zitat Kobayashi H, Sato K, Niioka T et al (2015) Relationship among gefitinib exposure, polymorphisms of its metabolizing enzymes and transporters, and side effects in Japanese patients with non-small-cell lung cancer. Clin Lung Cancer 16:274–281CrossRefPubMed Kobayashi H, Sato K, Niioka T et al (2015) Relationship among gefitinib exposure, polymorphisms of its metabolizing enzymes and transporters, and side effects in Japanese patients with non-small-cell lung cancer. Clin Lung Cancer 16:274–281CrossRefPubMed
19.
Zurück zum Zitat de Wit D, Gelderblom H, Sparreboom A et al (2014) Midazolam as a phenotyping probe to predict sunitinib exposure in patients with cancer. Cancer Chemother Pharmacol 73:87–96CrossRefPubMed de Wit D, Gelderblom H, Sparreboom A et al (2014) Midazolam as a phenotyping probe to predict sunitinib exposure in patients with cancer. Cancer Chemother Pharmacol 73:87–96CrossRefPubMed
20.
Zurück zum Zitat Roodhart JM, Langenberg MH, Witteveen E et al (2008) The molecular basis of class side effects due to treatment with inhibitors of the VEGF/VEGFR pathway. Curr Clin Pharmacol 3:132–143CrossRefPubMed Roodhart JM, Langenberg MH, Witteveen E et al (2008) The molecular basis of class side effects due to treatment with inhibitors of the VEGF/VEGFR pathway. Curr Clin Pharmacol 3:132–143CrossRefPubMed
21.
Zurück zum Zitat Rini BI, Cohen DP, Lu DR et al (2011) Hypertension as a biomarker of efficacy in patients with metastatic renal cell carcinoma treated with sunitinib. J Natl Cancer Inst 103:763–773CrossRefPubMedPubMedCentral Rini BI, Cohen DP, Lu DR et al (2011) Hypertension as a biomarker of efficacy in patients with metastatic renal cell carcinoma treated with sunitinib. J Natl Cancer Inst 103:763–773CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Motzer RJ, Escudier B, Tomczak P et al (2013) Axitinib versus sorafenib as second-line treatment for advanced renal cell carcinoma: overall survival analysis and updated results from a randomised phase 3 trial. Lancet Oncol 14:552–562CrossRefPubMed Motzer RJ, Escudier B, Tomczak P et al (2013) Axitinib versus sorafenib as second-line treatment for advanced renal cell carcinoma: overall survival analysis and updated results from a randomised phase 3 trial. Lancet Oncol 14:552–562CrossRefPubMed
23.
Zurück zum Zitat Rini BI, Schiller JH, Fruehauf JP et al (2011) Diastolic blood pressure as a biomarker of axitinib efficacy in solid tumors. Clin Cancer Res 17:3841–3849CrossRefPubMed Rini BI, Schiller JH, Fruehauf JP et al (2011) Diastolic blood pressure as a biomarker of axitinib efficacy in solid tumors. Clin Cancer Res 17:3841–3849CrossRefPubMed
24.
Zurück zum Zitat Rini BI, Melichar B, Fishman MN et al (2015) Axitinib dose titration: analyses of exposure, blood pressure and clinical response from a randomized phase II study in metastatic renal cell carcinoma. Ann Oncol 26:1372–1377PubMed Rini BI, Melichar B, Fishman MN et al (2015) Axitinib dose titration: analyses of exposure, blood pressure and clinical response from a randomized phase II study in metastatic renal cell carcinoma. Ann Oncol 26:1372–1377PubMed
Metadaten
Titel
Association of axitinib plasma exposure and genetic polymorphisms of ABC transporters with axitinib-induced toxicities in patients with renal cell carcinoma
verfasst von
Hiroshi Kato
Naoto Sassa
Masayuki Miyazaki
Mio Takeuchi
Miho Asai
Akane Iwai
Yukihiro Noda
Momokazu Gotoh
Kiyofumi Yamada
Publikationsdatum
01.09.2016
Verlag
Springer Berlin Heidelberg
Erschienen in
Cancer Chemotherapy and Pharmacology / Ausgabe 4/2016
Print ISSN: 0344-5704
Elektronische ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-016-3145-0

Weitere Artikel der Ausgabe 4/2016

Cancer Chemotherapy and Pharmacology 4/2016 Zur Ausgabe

Update Onkologie

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