Skip to main content

Advertisement

Log in

The small-molecule tyrosine kinase inhibitor nilotinib is a potent noncompetitive inhibitor of the SN-38 glucuronidation by human UGT1A1

  • Short Communication
  • Published:
Cancer Chemotherapy and Pharmacology Aims and scope Submit manuscript

Abstract

Purpose

Inhibition of the UDP-glucuronosyltransferase (UGT) 1A1 by nilotinib was examined in vitro with SN-38 as a substrate, to estimate the possibility of drug–drug interaction of nilotinib with other medicines predominantly detoxified by UGT1A1.

Methods

Inhibition of UGT1A1-catalyzed SN-38 glucuronidation by nilotinib was examined with human liver microsomes (HLM) and recombinant human UGT1A1 as enzyme sources. Inhibition constants (K i) were estimated with kinetic analysis.

Results

Nilotinib potently inhibited the SN-38 glucuronidation by human liver microsomal UGT1A1 and recombinant UGT1A1 in a noncompetitive manner, with K i values of 0.286 ± 0.0094 and 0.079 ± 0.0029 μM, respectively. If a drug that serves as a substrate of UGT1A1 is administered with nilotinib, the area under the plasma concentration–time curve of a drug estimated by using these K i values would be two times or higher than that without nilotinib, suggesting drug–drug interactions involving UGT1A1. These in vitro data and the prediction of drug–drug interaction are helpful for the clinical management of the nilotinib use.

Conclusion

We found that nilotinib is a potent noncompetitive inhibitor of human UGT1A1 activity.

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

References

  1. Zhang L, Reynolds KS, Zhao P, Huang SM (2010) Drug interactions evaluation: an integrated part of risk assessment of therapeutics. Toxicol Appl Pharmacol 243:134–145

    Article  CAS  PubMed  Google Scholar 

  2. Manley PW, Cowan-Jacob SW, Mestan J (2005) Advances in the structural biology, design and clinical development of Bcr-Abl kinase inhibitors for the treatment of chronic myeloid leukaemia. Biochim Biophys Acta 1754:3–13

    CAS  PubMed  Google Scholar 

  3. Weisberg E, Manley P, Mestan J, Cowan-Jacob S, Ray A, Griffin JD (2006) AMN107 (nilotinib): a novel and selective inhibitor of BCR-ABL. Br J Cancer 94:1765–1769

    Article  CAS  PubMed  Google Scholar 

  4. Hazarika M, Jiang X, Liu Q, Lee SL, Ramchandani R, Garnett C, Orr MS, Sridhara R, Booth B, Leighton JK, Timmer W, Harapanhalli R, Dagher R, Justice R, Pazdur R (2008) Tasigna for chronic and accelerated phase Philadelphia chromosome–positive chronic myelogenous leukemia resistant to or intolerant of imatinib. Clin Cancer Res 14:5325–5331

    Article  CAS  PubMed  Google Scholar 

  5. Singer JB, Shou Y, Giles F, Kantarjian HM, Hsu Y, Robeva AS, Rae P, Weitzman A, Meyer JM, Dugan M, Ottmann OG (2007) UGT1A1 promoter polymorphism increases risk of nilotinib-induced hyperbilirubinemia. Leukemia 21:2311–2315

    Article  CAS  PubMed  Google Scholar 

  6. Araki K, Fujita K, Ando Y, Nagashima F, Yamamoto W, Endo H, Miya T, Kodama K, Narabayashi M, Sasaki Y (2006) Pharmacogenetic impact of polymorphisms in the coding region of the UGT1A1 gene on SN-38 glucuronidation in Japanese patients with cancer. Cancer Sci 97:1255–1259

    Article  CAS  PubMed  Google Scholar 

  7. Copeland RA (2000) Enzymes: a practical introduction to structure, mechanism, and data analysis. Wiley, New York

    Google Scholar 

  8. Ito K, Brown HS, Houston JB (2004) Database analyses for the prediction of in vivo drug-drug interactions from in vitro data. Br J Clin Pharmacol 57:473–486

    Article  CAS  PubMed  Google Scholar 

  9. Tanaka C, Yin OQ, Sethuraman V, Smith T, Wang X, Grouss K, Kantarjian H, Giles F, Ottmann OG, Galitz L, Schran H (2010) Clinical pharmacokinetics of the BCR-ABL tyrosine kinase inhibitor nilotinib. Clin Pharmacol Ther 87:197–203

    Article  CAS  PubMed  Google Scholar 

  10. Gagne JF, Montminy V, Belanger P, Journault K, Gaucher G, Guillemette C (2002) Common human UGT1A polymorphisms and the altered metabolism of irinotecan active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38). Mol Pharmacol 62:608–617

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

We sincerely thank Drs. Yu, Sunakawa, Hiroo Ishida, Keishi Yamashita, Keiko Mizuno, Taro Yokoyama, Ken Shiozawa, Keisuke Miwa, Shigehira Saji, and Takashi Hirose for the valuable discussion on the present analyses. This study was supported in part by the Grant-in-Aid for Cancer Research 21S-8-1 from the Ministry of Health, Labour and Welfare of Japan, and in part by a grant-in-aid for “Support Project of Strategic Research Center in Private Universities” from the Ministry of Education, Culture, Sports, Science and Technology (MEXT) to Saitama Medical University Research Center for Genomic Medicine.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ken-ichi Fujita.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Fujita, Ki., Sugiyama, M., Akiyama, Y. et al. The small-molecule tyrosine kinase inhibitor nilotinib is a potent noncompetitive inhibitor of the SN-38 glucuronidation by human UGT1A1. Cancer Chemother Pharmacol 67, 237–241 (2011). https://doi.org/10.1007/s00280-010-1445-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00280-010-1445-3

Keywords

Navigation