Skip to main content

Advertisement

Log in

Contribution of UGT1A1 genetic polymorphisms related to axitinib pharmacokinetics to safety and efficacy in patients with renal cell carcinoma

  • Original Paper
  • Published:
Medical Oncology Aims and scope Submit manuscript

Abstract

Axitinib is a potent second-line molecular-targeted agent for metastatic renal cell carcinoma (mRCC). Axitinib pharmacokinetics and its relation with genetic polymorphisms were evaluated to predict the adverse events (AEs) and efficacy of axitinib. We analyzed 46 patients with mRCC who were treated with axitinib. The plasma axitinib level was measured at 0, 2, 4, 8, and 12 h after administration (C0, C2, C4, C8, and C12; ng/mL) on day 7 of the treatment. Genetic polymorphisms related to axitinib pharmacokinetics, including SLCO1B1, SLCO1B3, SLCO2B1, ABCB1, ABCG2, CYP2C19, CYP3A5, and UGT1A1, were analyzed. Axitinib C0 and AUC0–12 in patients with UGT1A1 poor metabolisers (*6/*6, *6/*28, and *28/*28; n = 10) were significantly higher than those in patients with UGT1A1 extensive metabolisers (*1/*1, *1/*6,*1/*28, and *27/*28; n = 36) (23.6 vs. 7.8 ng/mL, p = 0.030, and 441.3 vs. 217.1 ng h/mL, p = 0.007). The cutoff levels of C0 to predict ≥ G2 hypothyroidism and ≥ G2 anorexia were 6.6 and 7.1 ng/mL, respectively (p = 0.005 and p = 0.035). The overall survival (OS) in patients with C0 > 5 ng/mL was significantly better than that in patients with C0 < 5 ng/mL (p = 0.022). Genetic polymorphisms in UGT1A1 were significantly associated with the plasma axitinib level. The plasma axitinib level was significantly associated with the frequency of AEs and OS in patients with mRCC. No direct relationship was observed between UGT1A1 genotypes and the frequency of AEs or OS.

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

Similar content being viewed by others

References

  1. Gandaglia G, Ravi P, Abdollah F, et al. Contemporary incidence and mortality rates of kidney cancer in the United States. Can Urol Assoc J. 2014;8:247–52.

    Article  PubMed Central  PubMed  Google Scholar 

  2. Tsushima T. Epidemiology of renal cell carcinoma in Japan. Nippon Rinsho. 2017;75:23–6.

    Google Scholar 

  3. Sun M, Thuret R, Abdollah F, et al. Age-adjusted incidence, mortality, and survival rates of stage-specific renal cell carcinoma in North America: a trend analysis. Eur Urol. 2011;59:135–41.

    Article  PubMed  Google Scholar 

  4. Wahlgren T, Harmenberg U, Sandström P, et al. Treatment and overall survival in renal cell carcinoma: a Swedish population-based study (2000–2008). Br J Cancer. 2013;108:1541–9.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  5. Yu H, Steeghs N, Nijenhuis CM, Schellens JH, Beijnen JH, Huitema AD. Practical guidelines for therapeutic drug monitoring of anticancer tyrosine kinase inhibitors: focus on the pharmacokinetic targets. Clin Pharmacokinet. 2014;53:305–25.

    Article  CAS  PubMed  Google Scholar 

  6. Numakura K, Tsuchiya N, Kagaya H, et al. Clinical effects of single nucleotide polymorphisms on drug-related genes in Japanese metastatic renal cell carcinoma patients treated with sunitinib. Anticancer Drugs. 2017;28:97–103.

    Article  CAS  PubMed  Google Scholar 

  7. Noda S, Otsuji T, Baba M, et al. Assessment of sunitinib-induced toxicities and clinical outcomes based on therapeutic drug monitoring of sunitinib for patients with renal cell carcinoma. Clin Genitourin Cancer. 2015;13:350–8.

    Article  PubMed  Google Scholar 

  8. 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. 2016;78:855–62.

    Article  CAS  PubMed  Google Scholar 

  9. Brennan M, Williams JA, Chen Y, Tortorici M, Pithavala Y, Liu YC. Meta-analysis of contribution of genetic polymorphisms in drug-metabolizing enzymes or transporters to axitinib pharmacokinetics. Eur J Clin Pharmacol. 2012;68:645–55.

    Article  CAS  PubMed  Google Scholar 

  10. Rini BI, Escudier B, Tomczak P, et al. Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Lancet. 2011;378:1931–9.

    Article  CAS  PubMed  Google Scholar 

  11. Motzer RJ, Escudier B, Tomczak P, et al. 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. 2013;14:552–62.

    Article  CAS  PubMed  Google Scholar 

  12. Rini BI, Garrett M, Poland B, et al. Axitinib in metastatic renal cell carcinoma: results of a pharmacokinetic and pharmacodynamic analysis. J Clin Pharmacol. 2013;53:491–504.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  13. Rini BI, Melichar B, Fishman MN, et al. Axitinib dose titration: analyses of exposure, blood pressure and clinical response from a randomized phase II study in metastatic renal cell carcinoma. Ann Oncol. 2015;26:1372–7.

    Article  CAS  PubMed  Google Scholar 

  14. Escudier B, Rini BI, Motzer RJ, et al. Cenotype correlations with blood pressure and efficacy from randomized phase III trial of second-line axitinib versus sorafenib in metastatic renal cell carcinoma. Clin Genitourin Cancer. 2015;13:328–37.

    Article  PubMed  Google Scholar 

  15. Trotti A, Colevas AD, Setser A, et al. CTCAE v3.0: development of a comprehensive grading system for the adverse effects of cancer treatment. Semin Radiat Oncol. 2003;13:176–81.

    Article  PubMed  Google Scholar 

  16. Therasse P, Arbuck SG, Eisenhauer EA, et al. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst. 2000;92:205–16.

    Article  CAS  PubMed  Google Scholar 

  17. Chen Y, Tortorici MA, Garrett M, Hee B, Klamerus KJ, Pithavala YK. Clinical pharmacology of axitinib. Clin Pharmacokinet. 2013;52:713–25.

    Article  CAS  PubMed  Google Scholar 

  18. Ando Y, Saka H, Ando M, et al. Polymorphisms of UDP-glucuronosyltransferase gene and irinotecan toxicity: a pharmacogenetic analysis. Cancer Res. 2000;60:6921–6.

    CAS  PubMed  Google Scholar 

  19. Araki K, Fujita K, Ando Y, et al. Pharmacogenetic impact of polymorphisms in the coding region of the UGT1A1 gene on SN-38 glucuronidation in Japanese patients with cancer. Cancer Sci. 2006;97:1255–9.

    Article  CAS  PubMed  Google Scholar 

  20. Ramchandani RP, Wang Y, Booth BP, et al. The role of SN-38 exposure, UGT1A1*28 polymorphism, and baseline bilirubin level in predicting severe irinotecan toxicity. J Clin Pharmacol. 2007;47:78–86.

    Article  CAS  PubMed  Google Scholar 

  21. Minami H, Sai K, Saeki M, et al. Irinotecan pharmacokinetics/pharmacodynamics and UGT1A genetic polymorphisms in Japanese: roles of UGT1A1*6 and *28. Pharmacogenet Genomics. 2007;17:497–504.

    Article  CAS  PubMed  Google Scholar 

  22. Liu X, Cheng D, Kuang Q, Liu G, Xu W. Association of UGT1A1*28 polymorphisms with irinotecan-induced toxicities in colorectal cancer: a meta-analysis in Caucasians. Pharmacogenomics J. 2014;14:120–9.

    Article  CAS  PubMed  Google Scholar 

  23. Rini BI, Melichar B, Ueda T, et al. Axitinib with or without dose titration for first-line metastatic renal-cell carcinoma: a randomised double-blind phase 2 trial. Lancet Oncol. 2013;14:1233–42.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank Yoko Mitobe, Yukiko Sugiyama, and Ikuka Izumida for their contribution of clinical sample collections and preparations. This work was supported by the Grant Numbers 25293332, 16H02679, and 23590168 from the Japanese Society for the Promotion of Science and AMED-CREST, Japan Agency for Medical Research and Development (AMED).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Takamitsu Inoue.

Ethics declarations

Conflict of interest

The authors declare that they have no competing interests.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Figure 1

Progression-free survival (PFS) based on the plasma axitinib level. A: PFS in patients with C0 ≥5 ng/mL (n = 31) was not significantly better than that in patients with C0 < 5 ng/mL (n = 15). B: PFS in patients with AUC0–12 ≥300 ng·h/mL (n = 28) was not significantly better than that in patients with AUC0–12 < 300 ng·h/mL (n = 18). C: PFS in TKI-naïve patients with C0 ≥5 ng/mL (n = 22) was not significantly better than that in patients with C0 < 5 ng/mL (n = 11). B: PFS in TKI-naïve patients with AUC0–12 ≥300 ng·h/mL (n = 20) was not significantly better than that in patients with AUC0–12 < 300 ng·h/mL (n = 13)

Supplementary material 2 (PDF 132 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Igarashi, R., Inoue, T., Fujiyama, N. et al. Contribution of UGT1A1 genetic polymorphisms related to axitinib pharmacokinetics to safety and efficacy in patients with renal cell carcinoma. Med Oncol 35, 51 (2018). https://doi.org/10.1007/s12032-018-1113-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s12032-018-1113-8

Keywords

Navigation