Structure activity relationships of benzylproline-derived inhibitors of the glutamine transporter ASCT2

https://doi.org/10.1016/j.bmcl.2016.12.063Get rights and content

Abstract

The glutamine transporter ASCT2 has been identified as a promising target to inhibit rapid growth of cancer cells. However, ASCT2 pharmacology is not well established. In this report, we performed a systematic structure activity analysis of a series of substituted benzylproline derivatives. Substitutions on the phenyl ring resulted in compounds with characteristics of ASCT2 inhibitors. Apparent binding affinity increased with increasing hydrophobicity of the side chain. In contrast, interaction of the ASCT2 binding site with specific positions on the phenyl ring was not observed. The most potent compound inhibits the ASCT2 anion conductance with a Ki of 3 μM, which is in the same range as that of more bulky and higher molecular weight inhibitors recently reported by others. The experimental results are consistent with computational analysis based on docking of the inhibitors against an ASCT2 homology model. The benzylproline scaffold provides a valuable tool for further improving binding potency of future ASCT2 inhibitors.

Section snippets

Acknowledgements

This study was supported by a grant from the National Institutes of Health (http://www.nih.gov) (R01 GM108911) to AS, CC and CG.

References (22)

  • B.P. Bode

    J Nutr

    (2001)
  • O. Bussolati et al.

    J Biol Chem

    (1992)
  • B.C. Fuchs et al.

    Semin Cancer Biol

    (2005)
  • P. Nicklin et al.

    Cell

    (2009)
  • V. Ganapathy et al.

    Pharmacol Ther

    (2009)
  • C.S. Esslinger et al.

    Bioorg Med Chem

    (2005)
  • M.L. Schulte et al.

    Bioorg Med Chem Lett

    (2015)
  • M.L. Schulte et al.

    Bioorg Med Chem Lett

    (2016)
  • C.B. Zander et al.

    Amino acid transporters and glutamine

  • Y. Kanai et al.

    Pflugers Arch – Eur J Physiol

    (2004)
  • A. Broer et al.

    Biochem J

    (2000)
  • Cited by (24)

    • Diseases & disorders: Therapies targeting glutamine addiction in cancer

      2021, Encyclopedia of Biological Chemistry: Third Edition
    • Target the human Alanine/Serine/Cysteine Transporter 2(ASCT2): Achievement and Future for Novel Cancer Therapy

      2020, Pharmacological Research
      Citation Excerpt :

      These molecules which are designed based on rat ASCT2, are effective on inhibition of ASCT2 overexpressed even in human cell. In brief, it was suggested that benzylproline scaffold can provide a valuable tool to the understanding of the molecular parameters and the design of new and more efficient inhibitors against human ASCT2 [115]. In search of compounds with the ability to inhibit ASCT1/2, Alan C. Foster and co-workers screened a set of amino acid analogs and found that PG analogs with the ability to selectively and effectively inhibit ASCT1 and ASCT2.

    • Glutamine transporters as pharmacological targets: From function to drug design

      2020, Asian Journal of Pharmaceutical Sciences
      Citation Excerpt :

      These compounds, even if designed on rat ASCT2, showed cytotoxic effects also on human melanoma cell line [45]. Moving from this indication, the benzyl proline derivatives constituted the scaffold for designing more efficient inhibitors specific for the human ASCT2 and a Ki in the micromolar range has been measured in human cells for these compounds [46]. In another study, a competitive inhibitor of human ASCT2 has been identified starting from a previous screening conducted on 2-amino-4-bis (aryloxybenzyl)aminobutanoic acid derivatives [47].

    • Advances and Challenges in Rational Drug Design for SLCs

      2019, Trends in Pharmacological Sciences
      Citation Excerpt :

      Further, virtual screening of large compound libraries from the ZINC databaseiii, combined with cellular uptake and electrophysiology assays, identified new ASCT2 ligands, including five substrate-like compounds and two ASCT2 inhibitors (e.g., γ-2-fluorobenzyl proline) [44]. This study highlighted the utility of virtual screening to capture unexplored chemical spaces of transporter ligands, which have been further optimized to obtain higher-affinity inhibitors [51]. Subsequent ASCT2 models based on the human EAAT1 structure [27] (sequence identity of 46%) provided a refined conformation of TM8 and hairpin 2 (HP2) and overall a more accurate model, as measured by various measures such as its ability to enrich known ligands compared with decoy molecules [52].

    View all citing articles on Scopus
    1

    Current address: Department of Chemistry and Physics and Vascular Biology Center, Augusta University, Augusta, GA 30912, United States.

    View full text