The MTH1 inhibitor TH588 demonstrates anti-tumoral effects alone and in combination with everolimus, 5-FU and gamma-irradiation in neuroendocrine tumor cells

PLoS One. 2017 May 25;12(5):e0178375. doi: 10.1371/journal.pone.0178375. eCollection 2017.

Abstract

Modulation of the redox system in cancer cells has been considered a promising target for anti-cancer therapy. The novel MTH1 inhibitor TH588 proved tremendous potential in terms of cancer cell eradication, yet its specificity has been questioned by recent reports, indicating that TH588 may also induce cancer cell death by alternative mechanisms than MTH1 inhibition. Here we used a panel of heterogeneous neuroendocrine tumor cells in order to assess cellular mechanisms and molecular signaling pathways implicated in the effects of TH588 alone as well as dual-targeting approaches combining TH588 with everolimus, cytotoxic 5-fluorouracil or γ-irradiation. Our results reflect that TH588 alone efficiently decreased the survival of neuroendocrine cancer cells by PI3K-Akt-mTOR axis downregulation, increased apoptosis and oxidative stress. However, in the dual-targeting approaches cell survival was further decreased due to an even stronger downregulation of the PI3K-Akt-mTOR axis and augmentation of apoptosis but not oxidative stress. Furthermore, we could attribute TH588 chemo- and radio-sensitizing properties. Collectively our data not only provide insights into how TH588 exactly kills cancer cells but also depict novel perspectives for combinatorial treatment approaches encompassing TH588.

MeSH terms

  • Antineoplastic Combined Chemotherapy Protocols / pharmacology*
  • Apoptosis / drug effects
  • Apoptosis / radiation effects
  • Cell Line, Tumor
  • Cell Proliferation / drug effects
  • Cell Proliferation / radiation effects
  • DNA Repair Enzymes / antagonists & inhibitors*
  • Down-Regulation / drug effects
  • Down-Regulation / radiation effects
  • Drug Resistance, Neoplasm / drug effects
  • Drug Resistance, Neoplasm / radiation effects
  • Everolimus / pharmacology*
  • Fluorouracil / pharmacology*
  • Gamma Rays / therapeutic use
  • Humans
  • Neuroendocrine Cells / drug effects
  • Neuroendocrine Cells / radiation effects
  • Neuroendocrine Tumors / drug therapy*
  • Neuroendocrine Tumors / metabolism
  • Neuroendocrine Tumors / radiotherapy*
  • Oxidative Stress / drug effects
  • Oxidative Stress / radiation effects
  • Phosphatidylinositol 3-Kinases / metabolism
  • Phosphoric Monoester Hydrolases / antagonists & inhibitors*
  • Proto-Oncogene Proteins c-akt / metabolism
  • Pyrimidines / pharmacology*
  • Radiotherapy, Adjuvant
  • Signal Transduction / drug effects
  • TOR Serine-Threonine Kinases / metabolism

Substances

  • Pyrimidines
  • TH588 compound
  • Everolimus
  • MTOR protein, human
  • Proto-Oncogene Proteins c-akt
  • TOR Serine-Threonine Kinases
  • Phosphoric Monoester Hydrolases
  • 8-oxodGTPase
  • DNA Repair Enzymes
  • Fluorouracil

Grants and funding

CJ Auernhammer has received research contracts (Ipsen, Novartis), lecture honorarium (Ipsen, Novartis, Pfizer, Amgen, Roche, Falk) and advisory board honorarium (Novartis) within other projects. The authors received no funding from commercial sources for this work. Commercial sources had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. We state adherence to PLOS ONE policies on sharing data and materials. The authors declare that there is no conflict of interest that would prejudice the impartiality of this scientific work. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.