Skip to main content
Log in

Liver Safety of Fasiglifam (TAK-875) in Patients with Type 2 Diabetes: Review of the Global Clinical Trial Experience

  • Original Research Article
  • Published:
Drug Safety Aims and scope Submit manuscript

A Correction to this article was published on 17 October 2018

This article has been updated

Abstract

Introduction

Fasiglifam (TAK-875) is a G protein-coupled receptor 40 agonist that was being investigated for treatment of type 2 diabetes mellitus (T2DM). A development program was terminated late in phase III clinical trials due to liver safety concerns.

Methods

The liver safety of fasiglifam was assessed from data based on six phase II and nine phase III double-blind studies and two open-label studies with emphasis on pooled data from 15 double-blind studies from both global and Japanese development programs. Taking into consideration different daily doses of fasiglifam administered in clinical studies, the primary comparisons were between all patients exposed to fasiglifam (any dose) versus placebo, and, where applicable, versus the two active comparators, sitagliptin or glimepiride. A Liver Safety Evaluation Committee consisting of hepatologists blinded to treatment assignments evaluated hepatic adverse events (AEs) and serious AEs (SAEs) for causal relationship to study drug.

Results

The analysis included data from 9139 patients with T2DM in 15 double-blind controlled studies who received either fasiglifam (n = 5359, fasiglifam group), fasiglifam and sitagliptin (n = 123), or a comparator agent (n = 3657, non-exposed group consisting of placebo and other antidiabetic agents). Exposure to treatment for more than 1 year ranged from 249 patients in the placebo arm, to 370 patients in the glimepiride arm and 617 patients in the fasiglifam 50 mg arm. The primary focus of the analysis was on the hepatic safety of fasiglifam. The overall safety profile based on treatment-emergent AEs (TEAEs), SAEs, deaths, and withdrawal due to AEs was similar between fasiglifam and placebo (excluding liver test abnormalities). However, there was an increased incidence rate of serum alanine aminotransferase (ALT) elevations > 3 × upper limit of normal (ULN), 5 × ULN, and 10 × ULN in fasiglifam-treated patients compared with those treated with placebo or active comparators. ALT elevations > 3 × ULN for fasiglifam were 2.7% compared with 0.8 and 0.5% for the active comparators and placebo. There did not appear to be a clear dose response in incidence of ALT elevations between patients receiving 25 or 50 mg daily. The cumulative incidence of elevations in serum ALT > 3 × ULN was higher in the first 6 months of treatment with fasiglifam compared with both placebo and the active comparators, but the rate of new ALT elevations appeared to be similar across all treatment groups thereafter. No demographic or baseline patient characteristics were identified to predict elevations exceeding ALT > 3 × ULN in fasiglifam-treated patients. The pattern of liver injury with fasiglifam was hepatocellular, and there were no reports of liver-related deaths, liver failure or life-threatening liver injury. Most fasiglifam-associated ALT elevations were asymptomatic and resolved promptly upon discontinuing treatment, but in two patients the recovery was prolonged. Importantly, three important serious liver injury cases were identified among fasiglifam-treated patients; one case was adjudicated to be a clear Hy’s Law case and the two remaining cases were considered to closely approximate Hy’s Law cases.

Conclusions

Although the incidence of overall AEs, SAEs, and deaths was similar between fasiglifam and placebo, a liver signal was identified based primarily on the difference in liver chemistry values in the fasiglifam group compared with the placebo and active comparator groups. Three serious liver injuries were attributed to fasiglifam treatment. Clinical development of fasiglifam was halted due to these liver safety concerns.

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

Similar content being viewed by others

Change history

  • 17 October 2018

    In the original publication of the article, the ALT and AST values in Fig. 5a–e were capped at 10× ULN, which did not accurately reflect the narrative provided for each case. In this correction, the original Fig. 5a–e (Fig. 1a–e) and the correct Fig. 5a–5e (Fig. 2a–e) are published.

References

  1. Itoh Y, Kawamata Y, Harada M, et al. Free fatty acids regulate insulin secretion from pancreatic beta cells through GPR40. Nature. 2003;422:173–6.

    Article  CAS  PubMed  Google Scholar 

  2. Edfalk S, Steneberg P, Edlund H. Gpr40 is expressed in enteroendocrine cells and mediates free fatty acid stimulation of incretin secretion. Diabetes. 2008;57:2280–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Briscoe CP, Tadayyon M, Andrews JL, et al. The orphan G protein-coupled receptor GPR40 is activated by medium and long chain fatty acids. J Biol Chem. 2003;278:11303–11.

    Article  CAS  PubMed  Google Scholar 

  4. Tsujihata Y, Ito R, Suzuki M, et al. TAK-875, an orally available G protein-coupled receptor 40/free fatty acid receptor 1 agonist, enhances glucose-dependent insulin secretion and improves both postprandial and fasting hyperglycemia in type 2 diabetic rats. J Pharmacol Exp Ther. 2011;339:228–37.

    Article  CAS  PubMed  Google Scholar 

  5. Yashiro H, Tsujihata Y, Takeuchi K, Hazama M, Johnson PR, Rorsman P. The effects of TAK-875, a selective G protein-coupled receptor 40/free fatty acid 1 agonist, on insulin and glucagon secretion in isolated rat and human islets. J Pharmacol Exp Ther. 2012;340:483–9.

    Article  CAS  PubMed  Google Scholar 

  6. Naik H, Vakilynejad M, Wu J, et al. Safety, tolerability, pharmacokinetics, and pharmacodynamic properties of the GPR40 agonist TAK-875: results from a double-blind, placebo-controlled single oral dose rising study in healthy volunteers. J Clin Pharmacol. 2012;52:1007–16.

    Article  CAS  PubMed  Google Scholar 

  7. Leifke E, Naik H, Wu J, et al. A multiple-ascending-dose study to evaluate safety, pharmacokinetics, and pharmacodynamics of a novel GPR40 agonist, TAK-875, in subjects with type 2 diabetes. Clin Pharm Ther. 2012;92:29–39.

    Article  CAS  Google Scholar 

  8. Burant CF, Viswanathan P, Marcinak J, et al. TAK-875 versus placebo or glimepiride in type 2 diabetes mellitus: a phase 2, randomised, double-blind, placebo-controlled trial. Lancet. 2012;379:1403–11.

    Article  CAS  PubMed  Google Scholar 

  9. Kaku K, Enya K, Nakaya R, Ohira T, Matsuno R. Efficacy and safety of fasiglifam (TAK-875), a G protein-coupled receptor 40 agonist, in Japanese patients with type 2 diabetes inadequately controlled by diet and exercise: a randomized, double-blind, placebo-controlled, phase III trial. Diabetes Obes Metab. 2015;17:675–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Kaku K, Enya K, Nakaya R, Ohira T, Matsuno R. Long-term safety and efficacy of fasiglifam (TAK-875), a G-protein-coupled receptor 40 agonist, as monotherapy and combination therapy in Japanese patients with type 2 diabetes: a 52-week open-label phase III study. Diabetes Obes Metab. 2016;18:925–9.

    Article  CAS  PubMed  Google Scholar 

  11. Wolenski FS, Zhu AZX, Johnson M, et al. Fasiglifam (TAK-875) alters bile acid homeostasis in rats and dogs: a potential cause of drug induced liver injury. Toxicol Sci. 2017;157:50–61.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Araki T, Hirayama M, Hiroi S, Kaku K. GPR40-induced insulin secretion by the novel agonist TAK-875: first clinical findings in patients with type 2 diabetes. Diabetes Obes Metab. 2012;14:271–8.

    Article  CAS  PubMed  Google Scholar 

  13. Kaku K, Araki T, Yoshinaka R. Randomized, double-blind, dose-ranging study of TAK-875, a novel GPR40 agonist, in Japanese patients with inadequately controlled type 2 diabetes. Diabetes Care. 2013;36:245–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. US Food and Drug Administration. Guidance for industry drug-induced liver injury: premarketing clinical evaluation. Drug Safety. 2009. https://www.fda.gov/downloads/Drugs/../guidances/UCM174090.pdf. Accessed 12 Apr 2017.

  15. Rockey DC, Seeff LB, Rochon J, et al. Causality assessment in drug-induced liver injury using a structured expert opinion process: comparison to the Roussel–Uclaf causality assessment method. Hepatology. 2010;51:2117–26.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Aithal GP, Watkins PB, Andrade RJ, et al. Case definition and phenotype standardization in drug-induced liver injury. Clin Pharmacol Ther. 2011;89:806–15.

    Article  CAS  PubMed  Google Scholar 

  17. Merz M, Lee KR, Kullak-Ublick GA, Brueckner A, Watkins PB. Methodology to assess clinical liver safety data. Drug Saf. 2014;37(Suppl 1):S33–45.

    Article  CAS  PubMed  Google Scholar 

  18. Regev A. How to avoid being surprised by hepatotoxicity at the final stages of drug development and approval. Clin Liver Dis. 2013;17:749–67.

    Article  PubMed  Google Scholar 

  19. Mosedale M, Watkins PB. Drug-induced liver injury: advances in mechanistic understanding that will inform risk management. Clin Pharmacol Ther. 2017;101:469–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Otieno MA, Snoeys J, Lam W, et al. Fasiglifam (TAK 875): Mechanistic investigation and retrospective identification of hazards for drug induced liver injury. Toxicol Sci. 2017. https://doi.org/10.1093/toxsci/kfx040 (Epub 16 Feb 2017).

    Article  PubMed  Google Scholar 

Download references

Funding

The studies included in the analyses were funded by Takeda Development Center Americas, Inc. and Takeda Pharmaceutical Company Limited.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to John F. Marcinak.

Ethics declarations

Conflicts of interest

John F. Marcinak and Neila Smith are employed by Takeda Development Center Americas, Inc. (Deerfield, IL, USA). Melvin S. Munsaka is a former employee of Takeda Development Center Americas, Inc. Paul B. Watkins has served as a compensated consultant for Takeda regarding the liver safety of TAK-875 and other drugs. He received no compensation for the preparation of this article. Takashi Ohira is employed by Takeda Pharmaceutical Company Limited (Chuo-ku, Osaka, Japan).

Ethical approval

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards.

Informed consent

Informed consent was obtained from all individual participants included in the study.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplemental material 1 (PDF 616 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Marcinak, J.F., Munsaka, M.S., Watkins, P.B. et al. Liver Safety of Fasiglifam (TAK-875) in Patients with Type 2 Diabetes: Review of the Global Clinical Trial Experience. Drug Saf 41, 625–640 (2018). https://doi.org/10.1007/s40264-018-0642-6

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40264-018-0642-6

Navigation