Skip to main content
Log in

A Novel Double-Tracer Technique to Characterize Absorption, Distribution, Metabolism and Excretion (ADME) of [14C]Tofogliflozin After Oral Administration and Concomitant Intravenous Microdose Administration of [13C]Tofogliflozin in Humans

  • Original Research Article
  • Published:
Clinical Pharmacokinetics Aims and scope Submit manuscript

Abstract

Background

Human mass balance studies and the assessment of absolute oral bioavailability (F) are usually assessed in separate studies. Intravenous microdose administration of an isotope tracer concomitant to an unlabeled oral dose is an emerging technique to assess F. We report a novel double-tracer approach implemented for tofogliflozin combining oral administration of a radiolabel tracer with concomitant intravenous administration of a stable isotope tracer. Tofogliflozin is a potent and selective sodium/glucose cotransporter 2 inhibitor for the treatment of type 2 diabetes mellitus currently in clinical development.

Objectives

The objectives of the present study were to assess the systemic exposure of major circulating metabolites, excretion balance, F and contribution of renal clearance (CLR) to total clearance (CL) of tofogliflozin in healthy subjects within one study applying a novel double-tracer technique.

Methods

Six healthy male subjects received 20 mg [12C/14C]tofogliflozin (3.73 MBq) orally and a concomitant microdose of 0.1 mg [13C]tofogliflozin intravenously. Pharmacokinetics of tofogliflozin were determined for the oral and intravenous route; the pharmacokinetics of the metabolites M1 and M5 were determined for the oral route. Quantification of [12C]tofogliflozin in plasma and urine and [13C]tofogliflozin in plasma was performed by selective LC-MS/MS methods. For the pre-selected metabolites of tofogliflozin, M1 and M5, a validated liquid chromatography–tandem mass spectrometry (LC-MS/MS) was applied to plasma and urine samples. Total radioactivity was assessed in plasma, urine and feces. Pharmacokinetic analysis was conducted by non-compartmental methods.

Results

The pharmacokinetics of tofogliflozin in healthy subjects were characterized by an F of 97.5 ± 12.3 %, CL of 10.0 ± 1.3 l/h and volume of distribution at steady-state (V ss) of 50.6 ± 6.7 l. The main route of elimination of total drug-related material was by excretion into urine (77.0 ± 4.1 % of the dose). The observed CLR of 25.7 ± 5.0 ml/min was higher than the product of the estimated glomerular filtration rate (eGFR) and fraction unbound in plasma (f u) (eGFR × f u 15 ml/min), indicating the presence of net active tubular secretion in the renal elimination of tofogliflozin. However, CLR contributed only 15.5 % to the CL of tofogliflozin, suggesting that reductions in CLR by renal impairment won’t significantly affect systemic exposure to tofogliflozin. Tofogliflozin and its metabolite M1 were the only major circulating entities accounting for 46 ± 8.6 and 50 ± 8.2 %, respectively, of total circulating drug-related material, while the metabolite M5 was a minor circulating metabolite accounting for 3.0 ± 0.3 % of total circulating drug-related material. Both the M1 and M5 metabolites were excreted into urine and the major metabolite M1 did not exhibit active tubular secretion.

Conclusions

These results demonstrate the utility of the double-tracer approach to provide essential pharmacokinetic data and excretion data for drug-related material in one study at the same dosing occasion. The data obtained allowed the characterization of absorption, distribution, metabolism and excretion of tofogliflozin. Tofogliflozin exhibited highly favorable pharmacokinetic properties as demonstrated by its high F, low CL and a low V ss. The presence of only one major circulating metabolite of tofogliflozin was unambiguously demonstrated. As a drug targeting the kidney, luminal exposure of the kidney is achieved by renal filtration and active tubular secretion.

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.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  1. Suzuki M, Honda K, Fukazawa M, Ozawa K, Hagita H, Kawai T, et al. Tofogliflozin, a potent and highly specific sodium/glucose cotransporter 2 inhibitor, improves glycemic control in diabetic rats and mice. J Pharmacol Exp Ther. 2012;341(3):692–701.

    Article  PubMed  CAS  Google Scholar 

  2. Ikeda S, Takano Y, Cynshi O, Christ AD, Boerlin V, Beyer U, Beck A. A novel and selective SGLT2 inhibitor, tofogliflozin improves glycaemic control and lowers body weight in patients with type 2 diabetes mellitus. In: European Association for the Study of Diabetes, annual meeting 2012, Berlin [abstract no. 768].

  3. Schwab D, Portron A, Fukushima Y, Backholer Z, Kuhlmann O, Saito T, Ikeda S. Tofogliflozin a selective SGLT2 inhibitor exhibits highly favourable drug properties for use in patients with renal impairment and for combination with other medicines. In: European Association for the Study of Diabetes, annual meeting 2012, Berlin [abstract no. 767].

  4. Hummel CS, Lu C, Loo DD, Hirayama BA, Voss AA, Wright EM. Glucose transport by human renal Na+/d-glucose cotransporters SGLT1 and SGLT2. Am J Physiol Cell Physiol. 2011;300(1):C14–21.

    Article  PubMed  CAS  Google Scholar 

  5. Bakris GL, Fonseca VA, Sharma K, Wright EM. Renal sodium-glucose transport: role in diabetes mellitus and potential clinical implications. Kidney Int. 2009;75(12):1272–7.

    Article  PubMed  CAS  Google Scholar 

  6. Abdul-Ghani MA, DeFronzo RA. Inhibition of renal glucose reabsorption: a novel strategy for achieving glucose control in type 2 diabetes mellitus. Endocr Pract. 2008;14(6):782–90.

    Article  PubMed  Google Scholar 

  7. Chao EC, Henry RR. SGLT2 inhibition—a novel strategy for diabetes treatment. Nat Rev Drug Discov. 2010;9(7):551–9.

    Article  PubMed  CAS  Google Scholar 

  8. ICH. International conference on harmonization of technical requirements for registration of pharmaceuticals for human use. ICH harmonized tripartite guideline. Guidance on nonclinical safety studies for the conduct of human clinical trials and marketing authorization for pharmaceuticals. M3(R2) Current Step 4 version dated 11 June 2009.

  9. Lappin G, Garner RC. The utility of microdosing over the past 5 years. Expert Opin Drug Metab Toxicol. 2008;4(12):1499–506.

    Article  PubMed  CAS  Google Scholar 

  10. Lappin G, Kuhnz W, Jochemsen R, Kneer J, Chaudhary A, Oosterhuis B, et al. Use of microdosing to predict pharmacokinetics at the therapeutic dose: experience with 5 drugs. Clin Pharmacol Ther. 2006;80(3):203–15.

    Article  PubMed  CAS  Google Scholar 

  11. Lappin G, Rowland M, Garner RC. The use of isotopes in the determination of absolute bioavailability of drugs in humans. Expert Opin Drug Metab Toxicol. 2006;2(3):419–27.

    Article  PubMed  CAS  Google Scholar 

  12. Lappin G, Shishikura Y, Jochemsen R, Weaver RJ, Gesson C, Houston B, et al. Pharmacokinetics of fexofenadine: evaluation of a microdose and assessment of absolute oral bioavailability. Eur J Pharm Sci. 2010;40(2):125–31.

    Article  PubMed  CAS  Google Scholar 

  13. Ni J, Ouyang H, Aiello M, Seto C, Borbridge L, Sakuma T, et al. Microdosing assessment to evaluate pharmacokinetics and drug metabolism in rats using liquid chromatography–tandem mass spectrometry. Pharm Res. 2008;25(7):1572–82.

    Article  PubMed  CAS  Google Scholar 

  14. Sparreboom A. Unexplored pharmacokinetic opportunities with microdosing in oncology. Clin Cancer Res. 2007;13(14):4033–4.

    Article  PubMed  Google Scholar 

  15. Sugiyama Y. Effective use of microdosing and positron emission tomography (PET) studies on new drug discovery and development. Drug Metab Pharmacokinet. 2009;24(2):127–9.

    Article  PubMed  CAS  Google Scholar 

  16. Tozuka Z, Kusuhara H, Nozawa K, Hamabe Y, Ikushima I, Ikeda T, et al. Microdose study of 14C-acetaminophen with accelerator mass spectrometry to examine pharmacokinetics of parent drug and metabolites in healthy subjects. Clin Pharmacol Ther. 2010;88(6):824–30.

    Article  PubMed  CAS  Google Scholar 

  17. Sarapa N, Hsyu PH, Lappin G, Garner RC. The application of accelerator mass spectrometry to absolute bioavailability studies in humans: simultaneous administration of an intravenous microdose of 14C-nelfinavir mesylate solution and oral nelfinavir to healthy volunteers. J Clin Pharmacol. 2005;45(10):1198–205.

    Article  PubMed  CAS  Google Scholar 

  18. Outcomes from EUMAPP—a study comparing in vitro, in silico, microdose and pharmacological dose pharmacokinetics. In: European Microdosing AMS Partnership Programme (EUMAPP); 2009. http://www.eumapp.com/pdfs/EUMAPP%20SUMMARY.pdf. Accessed 9 Nov 2012.

  19. Rubin GM, Waschek JA, Pond SM, Effeney DJ, Tozer TN. Concurrent intravenous administration of a labeled tracer to determine the oral bioavailability of a drug exhibiting Michaelis–Menten metabolism. J Pharmacokinet Biopharm. 1987;15(6):615–31.

    Article  PubMed  CAS  Google Scholar 

  20. Maeda K, Sugiyama Y. Novel strategies for microdose studies using non-radiolabeled compounds. Adv Drug Deliv Rev. 2011;63(7):532–8.

    Article  PubMed  CAS  Google Scholar 

  21. Gu H, Wang J, Aubry AF, Jiang H, Zeng J, Easter J, et al. Calculation and mitigation of isotopic interferences in liquid chromatography–mass spectrometry/mass spectrometry assays and its application in supporting microdose absolute bioavailability studies. Anal Chem. 2012;84(11):4844–50.

    Article  PubMed  CAS  Google Scholar 

  22. Gibaldi M, Perrier D. Pharmacokinetics. Noncompartmental analysis based on statistical moment theory. 2nd ed., revised and expanded ed. New York: Marcel Dekker; 1982.

  23. Stevens LA, Coresh J, Greene T, Levey AS. Assessing kidney function—measured and estimated glomerular filtration rate. N Engl J Med. 2006;354:2473–83.

    Article  PubMed  CAS  Google Scholar 

  24. Du Bois D, Du Bois EF. A formula to estimate the approximate surface area if height and weight be known. Arch Intern Med. 1916;17:863–71.

    Article  Google Scholar 

  25. Penner N, Klunk LJ, Prakash C. Human radiolabeled mass balance studies: objectives, utilities and limitations. Biopharm Drug Dispos. 2009;30(4):185–203.

    Article  PubMed  CAS  Google Scholar 

  26. Roffey SJ, Obach RS, Gedge JI, Smith DA. What is the objective of the mass balance study? A retrospective analysis of data in animal and human excretion studies employing radiolabeled drugs. Drug Metab Rev. 2007;39(1):17–43.

    Article  PubMed  CAS  Google Scholar 

  27. Zhang Y, Zhang L, Abraham S, Apparaju S, Wu T-C, Strong JM, Xiao S, Atkinson AJ Jr, Thummel KE, Leeder JS, Lee C, Burckart GJ, Lesko LJ, Huang S-M. Assessment of the impact of renal impairment on systemic exposure of new molecular entities: evaluation of recent new drug applications. Clin Pharm Ther. 2009;85(3):305–11.

    Article  CAS  Google Scholar 

  28. Rowland Yeo K, Aarabi M, Jamei M, Rostami-Hodjegan A. Modeling and predicting drug pharmacokinetics in patients with renal impairment. Expert Rev Clin Pharmacol. 2011;4(2):261–74.

    Article  PubMed  Google Scholar 

Download references

Acknowledgments

Dietmar Schwab designed the study, approved the protocol, supervised the analysis and drafted the manuscript. Agnes Portron contributed to the protocol, analysis and interpretation of data. Zoe Backholer contributed to the protocol and acquisition of data. Berthold Lausecker (present address: CRS Clinical Research Services Mannheim GmbH, Germany) contributed to the design of the stable isotopes and supervised the LC-MS analysis. Kosuke Kawashima provided structural identification data of metabolites required for the pre-specified quantitative measurements in this study. All authors read and approved the final manuscript.

The authors want to acknowledge the many people involved in the conduct of the clinical trial, in particular the responsible investigator involved in the study, Dr. Stuart Mair (Quotient Clinical, Edinburgh, EH14 4AP, UK). Furthermore, the authors want to acknowledge Guy Fischer and Dr. Andreas Gloge (F. Hoffmann-LaRoche AG, pRED Non-clinical safety, Basel, Switzerland) for their bioanalytical support, Dr. Thomas Hartung (F. Hoffmann-LaRoche AG, Process Research & Synthesis, Basel, Switzerland) for the synthesis of the tofogliflozin isotopes, and Mizuki Yamane, Keiichi Morita, Tsutomu Sato and Yoshihito Ohtake (Chugai Pharmaceutical Co. Ltd., Research Division, Gotemba, Japan) for metabolite identification and reference material supply. The authors want to acknowledge the valuable discussions and input from Bruno Reigner (F. Hoffmann-LaRoche AG, pRED Clinical Pharmacology, Basel, Switzerland).

This study was funded by F. Hoffmann-La Roche AG. The authors are or were employed by F. Hoffmann-La Roche AG (Dietmar Schwab, Agnes Portron, Berthold Lausecker, Zoe Backholer) or by Chugai Pharmaceutical Co., Ltd. (Kosuke Kawashima).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Dietmar Schwab.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Schwab, D., Portron, A., Backholer, Z. et al. A Novel Double-Tracer Technique to Characterize Absorption, Distribution, Metabolism and Excretion (ADME) of [14C]Tofogliflozin After Oral Administration and Concomitant Intravenous Microdose Administration of [13C]Tofogliflozin in Humans. Clin Pharmacokinet 52, 463–473 (2013). https://doi.org/10.1007/s40262-013-0051-z

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40262-013-0051-z

Keywords

Navigation