Skip to main content

Advertisement

Log in

Human mass balance study of TAS-102 using 14C analyzed by accelerator mass spectrometry

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

Abstract

Background

TAS-102 is an oral fluoropyrimidine prodrug composed of trifluridine (FTD) and tipiracil hydrochloride (TPI) in a 1:0.5 ratio. FTD is a thymidine analog, and it is degraded by thymidine phosphorylase (TP) to the inactive trifluoromethyluracil (FTY) metabolite. TPI inhibits degradation of FTD by TP, increasing systemic exposure to FTD.

Methods

Patients with advanced solid tumors (6 M/2 F; median age 58 years; PS 0–1) were enrolled on this study. Patients in group A (N = 4) received 60 mg TAS-102 with 200 nCi [14C]-FTD, while patients in group B (N = 4) received 60 mg TAS-102 with 1000 nCi [14C]-TPI orally. Plasma, blood, urine, feces, and expired air (group A only) were collected up to 168 h and were analyzed for 14C by accelerator mass spectrometry and analytes by LC–MS/MS.

Results

FTD: 59.8 % of the 14C dose was recovered: 54.8 % in urine mostly as FTY and FTD glucuronide isomers. The extractable radioactivity in the pooled plasma consisted of 52.7 % FTD and 33.2 % FTY. TPI: 76.8 % of the 14C dose was recovered: 27.0 % in urine mostly as TPI and 49.7 % in feces. The extractable radioactivity in the pooled plasma consisted of 53.1 % TPI and 30.9 % 6-HMU, the major metabolite of TPI.

Conclusion

Absorbed 14C-FTD was metabolized and mostly excreted in urine. The majority of 14C-TPI was recovered in feces, and the majority of absorbed TPI was excreted in urine. The current data with the ongoing hepatic and renal dysfunction studies will provide an enhanced understanding of the TAS-102 elimination profile.

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
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Tanaka N et al (2014) Repeated oral dosing of TAS-102 confers high trifluridine incorporation into DNA and sustained antitumor activity in mouse models. Oncol Rep 32(6):2319–2326

    PubMed Central  CAS  PubMed  Google Scholar 

  2. Temmink OH et al (2007) Therapeutic potential of the dual-targeted TAS-102 formulation in the treatment of gastrointestinal malignancies. Cancer Sci 98(6):779–789

    Article  CAS  PubMed  Google Scholar 

  3. Emura T et al (2004) A novel antimetabolite, TAS-102 retains its effect on FU-related resistant cancer cells. Int J Mol Med 13(4):545–549

    CAS  PubMed  Google Scholar 

  4. Emura T et al (2004) A novel combination antimetabolite, TAS-102, exhibits antitumor activity in FU-resistant human cancer cells through a mechanism involving FTD incorporation in DNA. Int J Oncol 25(3):571–578

    CAS  PubMed  Google Scholar 

  5. Emura T et al (2005) Potentiation of the antitumor activity of alpha, alpha, alpha-trifluorothymidine by the co-administration of an inhibitor of thymidine phosphorylase at a suitable molar ratio in vivo. Int J Oncol 27(2):449–455

    CAS  PubMed  Google Scholar 

  6. Overman MJ et al (2008) Phase I clinical study of three times a day oral administration of TAS-102 in patients with solid tumors. Cancer Investig 26(8):794–799

    Article  CAS  Google Scholar 

  7. Overman MJ et al (2008) Phase 1 study of TAS-102 administered once daily on a 5-day-per-week schedule in patients with solid tumors. Investig New Drugs 26(5):445–454

    Article  CAS  Google Scholar 

  8. Hong DS et al (2006) Phase I study to determine the safety and pharmacokinetics of oral administration of TAS-102 in patients with solid tumors. Cancer 107(6):1383–1390

    Article  CAS  PubMed  Google Scholar 

  9. Yoshino T et al (2012) TAS-102 monotherapy for pretreated metastatic colorectal cancer: a double-blind, randomised, placebo-controlled phase 2 trial. Lancet Oncol 13(10):993–1001

    Article  CAS  PubMed  Google Scholar 

  10. Mayer RJ et al (2015) Randomized trial of TAS-102 for refractory metastatic colorectal cancer. N Engl J Med 372(20):1909–1919

    Article  PubMed  Google Scholar 

  11. Kotani D, Fukuoka S, Yoshino T (2015) Efficacy of TAS-102. Gan To Kagaku Ryoho 42(1):1–5

    PubMed  Google Scholar 

  12. Beumer JH et al (2007) Human mass balance study of the novel anticancer agent ixabepilone using accelerator mass spectrometry. Investig New Drugs 25(4):327–334

    Article  CAS  Google Scholar 

  13. Graham RA et al (2011) A single dose mass balance study of the Hedgehog pathway inhibitor vismodegib (GDC-0449) in humans using accelerator mass spectrometry. Drug Metab Dispos 39(8):1460–1467

    Article  CAS  PubMed  Google Scholar 

  14. U.S. Department of Health and Human Services Food and Drug Administration (2001) Guidance for industry-bioanalytical method validation. U.S. Department of Health and Human Services; Food and Drug Administration; Center for Drug Evaluation; and Research Center for Veterinary Medicine

  15. Hamilton RA, Garnett WR, Kline BJ (1981) Determination of mean valproic acid serum level by assay of a single pooled sample. Clin Pharmacol Ther 29(3):408–413

    Article  CAS  PubMed  Google Scholar 

  16. Doi T et al (2012) Phase I study of TAS-102 treatment in Japanese patients with advanced solid tumours. Br J Cancer 107(3):429–434

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  17. Khalili P et al (2002) Pharmacokinetics and metabolism of the novel synthetic C-nucleoside, 1-(2-deoxy-beta-d-ribofuranosyl)-2,4-difluoro-5-iodobenzene: a potential mimic of 5-iodo-2′-deoxyuridine. Biopharm Drug Dispos 23(3):105–113

    Article  CAS  PubMed  Google Scholar 

  18. Zhou L et al (2010) Disposition of [1′-(14)C]stavudine after oral administration to humans. Drug Metab Dispos 38(4):655–666

    Article  CAS  PubMed  Google Scholar 

  19. Desmoulin F et al (2007) A glucuronidation pathway of capecitabine occurs in rats but not in mice and humans. Drug Metab Lett 1(2):101–107

    Article  CAS  PubMed  Google Scholar 

  20. Desmoulin F et al (2002) Metabolism of capecitabine, an oral fluorouracil prodrug: (19)F NMR studies in animal models and human urine. Drug Metab Dispos 30(11):1221–1229

    Article  CAS  PubMed  Google Scholar 

  21. Nicolas F et al (1995) Comparative metabolism of 3′-azido-3′-deoxythymidine in cultured hepatocytes from rats, dogs, monkeys, and humans. Drug Metab Dispos 23(3):308–313

    CAS  PubMed  Google Scholar 

  22. Good SS et al (1990) Isolation and characterization of an ether glucuronide of zidovudine, a major metabolite in monkeys and humans. Drug Metab Dispos 18(3):321–326

    CAS  PubMed  Google Scholar 

  23. Dexter DL et al (1972) The clinical pharmacology of 5-trifluoromethyl-2′-deoxyuridine. Cancer Res 32(2):247–253

    CAS  PubMed  Google Scholar 

  24. Shields AF et al (1996) Analysis of 2-carbon-11-thymidine blood metabolites in PET imaging. J Nucl Med 37(2):290–296

    CAS  PubMed  Google Scholar 

  25. Beumer JH, Beijnen JH, Schellens JH (2006) Mass balance studies, with a focus on anticancer drugs. Clin Pharmacokinet 45(1):33–58

    Article  CAS  PubMed  Google Scholar 

  26. Ghoos Y et al (1988) Measurement of 13C-glucose oxidation rate using mass spectrometric determination of the CO2: Ar ratio and spirometry. Biomed Environ Mass Spectrom 15(8):447–451

    Article  CAS  PubMed  Google Scholar 

  27. Rogers WI et al (1969) The fate of 5-trifluoromethyl-2′-deoxyuridine in monkeys, dogs, mice, and tumor-bearing mice. Cancer Res 29(4):953–961

    CAS  PubMed  Google Scholar 

  28. Heidelberger C, Boohar J, Kampschroer B (1965) Fluorinated pyrimidines. Xxiv. In vivo metabolism of 5-trifluoromethyluracil-2-C-14 and 5-trifluoromethyl-2′-deoxyuridine-2-C-14. Cancer Res 25:377–381

    CAS  PubMed  Google Scholar 

  29. Andersen JT et al (2014) Extending serum half-life of albumin by engineering neonatal Fc receptor (FcRn) binding. J Biol Chem 289(19):13492–13502

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  30. Hammond TG et al (2014) Mass spectrometric characterization of circulating covalent protein adducts derived from a drug acyl glucuronide metabolite: multiple albumin adductions in diclofenac patients. J Pharmacol Exp Ther 350(2):387–402

    Article  PubMed Central  PubMed  Google Scholar 

  31. Meng X et al (2013) Detection of drug bioactivation in vivo: mechanism of nevirapine–albumin conjugate formation in patients. Chem Res Toxicol 26(4):575–583

    Article  CAS  PubMed  Google Scholar 

  32. Ariza A et al (2012) Protein haptenation by amoxicillin: high resolution mass spectrometry analysis and identification of target proteins in serum. J Proteomics 77:504–520

    Article  CAS  PubMed  Google Scholar 

  33. Wang J et al (2010) Characterization of HKI-272 covalent binding to human serum albumin. Drug Metab Dispos 38(7):1083–1093

    Article  CAS  PubMed  Google Scholar 

  34. Shipkova M et al (2002) Pharmacokinetics and protein adduct formation of the pharmacologically active acyl glucuronide metabolite of mycophenolic acid in pediatric renal transplant recipients. Ther Drug Monit 24(3):390–399

    Article  CAS  PubMed  Google Scholar 

  35. Wang M, Dickinson RG (2000) Bile duct ligation promotes covalent drug-protein adduct formation in plasma but not in liver of rats given zomepirac. Life Sci 68(5):525–537

    Article  CAS  PubMed  Google Scholar 

  36. Georges H et al (1999) Glycation of human serum albumin by acylglucuronides of nonsteroidal anti-inflammatory drugs of the series of phenylpropionates. Life Sci 65(12):PL151-6

    Article  PubMed  Google Scholar 

  37. Benet LZ et al (1993) Predictability of the covalent binding of acidic drugs in man. Life Sci 53(8):PL141-6

    Article  PubMed  Google Scholar 

  38. Klaassen CD (ed) (2008) Casarett and Doull’s toxicology: the basic science of Poisons, 7th edn. McGraw-Hill, New York, p 1309

    Google Scholar 

Download references

Acknowledgments

We thank the patients and their family members for their dedication and commitment to this clinical study. We thank the nursing staff of the University of Pittsburgh Clinical Translational Research Center for their invaluable assistance. This work was supported by Taiho Oncology, Inc., and NIH/NCRR/CTSA Grant UL1 RR024153. This project used the UPCI Cancer Pharmacokinetics and Pharmacodynamics Facility (CPPF) and was supported in part by award P30CA047904. We would like to thank the following individuals and organizations for their input in the various stages of this project: Manuel Aivado, Brian Kiesel, Milind Narurkar, Mark Seymour, Dennis Swanson, Tracy Topp, Erin Sternberg, and JCL Bioassay USA Inc. The authors were responsible for all content and editorial decisions and received no honoraria related to the development of this publication. All authors contributed to the research, writing, and reviewing of all drafts of this publication, and all authors approved the final draft prior to submission.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jan Hendrik Beumer.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lee, J.J., Seraj, J., Yoshida, K. et al. Human mass balance study of TAS-102 using 14C analyzed by accelerator mass spectrometry. Cancer Chemother Pharmacol 77, 515–526 (2016). https://doi.org/10.1007/s00280-016-2965-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00280-016-2965-2

Keywords

Navigation