Skip to main content

Advertisement

Log in

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

  • Original Article
  • Published:
Cancer Chemotherapy and Pharmacology Aims and scope Submit manuscript

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.

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

Similar content being viewed by others

References

  1. 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 14:7272–7283

    Article  CAS  PubMed  Google Scholar 

  2. 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 53:491–504

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. 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 13:350–358

    Article  PubMed  Google Scholar 

  4. Terada T, Hira D (2015) Intestinal and hepatic drug transporters: pharmacokinetic, pathophysiological, and pharmacogenetic roles. J Gastroenterol 50:508–519

    Article  CAS  PubMed  Google Scholar 

  5. 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 1:611–616

    CAS  PubMed  Google Scholar 

  6. Kimchi-Sarfaty C, Oh JM, Kim IW et al (2007) A “silent” polymorphism in the MDR1 gene changes substrate specificity. Science 315:525–528

    Article  CAS  PubMed  Google Scholar 

  7. 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 27:631–639

    Article  CAS  Google Scholar 

  8. 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 13:615–624

    Article  CAS  PubMed  Google Scholar 

  9. 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 39:729–735

    Article  CAS  PubMed  Google Scholar 

  10. 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 68:645–655

    Article  CAS  PubMed  Google Scholar 

  11. 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 378:1931–1939

    Article  CAS  PubMed  Google Scholar 

  12. 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 11:107–114

    Article  PubMed  Google Scholar 

  13. 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 49:679–687

    Article  CAS  PubMed  Google Scholar 

  14. 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 12:6073–6078

    Article  CAS  PubMed  Google Scholar 

  15. 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 68:723–733

    Article  CAS  PubMed  Google Scholar 

  16. 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 66:357–371

    Article  CAS  PubMed  Google Scholar 

  17. 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 52:593–609

    Article  CAS  PubMed  Google Scholar 

  18. 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 16:274–281

    Article  CAS  PubMed  Google Scholar 

  19. 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 73:87–96

    Article  PubMed  Google Scholar 

  20. 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 3:132–143

    Article  CAS  PubMed  Google Scholar 

  21. 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 103:763–773

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. 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 14:552–562

    Article  CAS  PubMed  Google Scholar 

  23. Rini BI, Schiller JH, Fruehauf JP et al (2011) Diastolic blood pressure as a biomarker of axitinib efficacy in solid tumors. Clin Cancer Res 17:3841–3849

    Article  CAS  PubMed  Google Scholar 

  24. 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 26:1372–1377

    CAS  PubMed  Google Scholar 

Download references

Acknowledgments

This work was supported in part by JSPS KAKENHI Grant Number JP 16H00497.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Hiroshi Kato.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kato, H., Sassa, N., Miyazaki, M. et al. Association of axitinib plasma exposure and genetic polymorphisms of ABC transporters with axitinib-induced toxicities in patients with renal cell carcinoma. Cancer Chemother Pharmacol 78, 855–862 (2016). https://doi.org/10.1007/s00280-016-3145-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00280-016-3145-0

Keywords

Navigation