Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Pharmacological blockade of ASCT2-dependent glutamine transport leads to antitumor efficacy in preclinical models

Abstract

The unique metabolic demands of cancer cells underscore potentially fruitful opportunities for drug discovery in the era of precision medicine. However, therapeutic targeting of cancer metabolism has led to surprisingly few new drugs to date. The neutral amino acid glutamine serves as a key intermediate in numerous metabolic processes leveraged by cancer cells, including biosynthesis, cell signaling, and oxidative protection. Herein we report the preclinical development of V-9302, a competitive small molecule antagonist of transmembrane glutamine flux that selectively and potently targets the amino acid transporter ASCT2. Pharmacological blockade of ASCT2 with V-9302 resulted in attenuated cancer cell growth and proliferation, increased cell death, and increased oxidative stress, which collectively contributed to antitumor responses in vitro and in vivo. This is the first study, to our knowledge, to demonstrate the utility of a pharmacological inhibitor of glutamine transport in oncology, representing a new class of targeted therapy and laying a framework for paradigm-shifting therapies targeting cancer cell metabolism.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: V-9302 is an inhibitor of glutamine transport.
Figure 2: In silico modeling of V-9302 interactions with hASCT2.
Figure 3: In vitro efficacy of V-9302.
Figure 4: Molecular determinants of ASCT2 antagonism in vitro.
Figure 5: Evaluation of V-9302 in vivo.
Figure 6: Summary of cancer cell programs modulated by V-9302.

Similar content being viewed by others

Accession codes

Accessions

Protein Data Bank

References

  1. Pochini, L., Scalise, M., Galluccio, M. & Indiveri, C. Membrane transporters for the special amino acid glutamine: structure/function relationships and relevance to human health. Front. Chem. 2, 61 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Jin, L., Alesi, G.N. & Kang, S. Glutaminolysis as a target for cancer therapy. Oncogene 35, 3619–3625 (2016).

    Article  CAS  PubMed  Google Scholar 

  3. Hassanein, M. et al. SLC1A5 mediates glutamine transport required for lung cancer cell growth and survival. Clin. Cancer Res. 19, 560–570 (2013).

    Article  CAS  PubMed  Google Scholar 

  4. van Geldermalsen, M. et al. ASCT2/SLC1A5 controls glutamine uptake and tumour growth in triple-negative basal-like breast cancer. Oncogene 35, 3201–3208 (2016).

    Article  CAS  PubMed  Google Scholar 

  5. Schulte, M.L. et al. Non-invasive glutamine PET reflects pharmacological inhibition of BRAFV600Ein vivo. Mol. Imaging Biol. 19, 421–428 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Gao, P. et al. c-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism. Nature 458, 762–765 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Watanabe, T. et al. Differential gene expression signatures between colorectal cancers with and without KRAS mutations: crosstalk between the KRAS pathway and other signalling pathways. Eur. J. Cancer 47, 1946–1954 (2011).

    Article  CAS  PubMed  Google Scholar 

  8. Romero, R. et al. Keap1 loss promotes Kras-driven lung cancer and results in dependence on glutaminolysis. Nat. Med. 23, 1362–1368 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Shukla, K. et al. Design, synthesis, and pharmacological evaluation of bis-2-(5-henylacetamido-1,2,4-thiadiazol-2-yl)ethyl sulfide 3 (BPTES) analogs as glutaminase inhibitors. J. Med. Chem. 55, 10551–10563 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Harding, J.J. et al. Safety and tolerability of increasing doses of CB-839, a first-in-class, orally administered small molecule inhibitor of glutaminase, in solid tumors. J. Clin. Oncol. 33, 2512 (2015).

    Article  Google Scholar 

  11. Rhoads, J.M. et al. Glutamine metabolism stimulates intestinal cell MAPKs by a cAMP-inhibitable, Raf-independent mechanism. Gastroenterology 118, 90–100 (2000).

    Article  CAS  PubMed  Google Scholar 

  12. Willems, L. et al. Inhibiting glutamine uptake represents an attractive new strategy for treating acute myeloid leukemia. Blood 122, 3521–3532 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Schulte, M.L., Khodadadi, A.B., Cuthbertson, M.L., Smith, J.A. & Manning, H.C. 2-Amino-4-bis(aryloxybenzyl)aminobutanoic acids: a novel scaffold for inhibition of ASCT2-mediated glutamine transport. Bioorg. Med. Chem. Lett. 26, 1044–1047 (2016).

    Article  CAS  PubMed  Google Scholar 

  14. Esslinger, C.S., Cybulski, K.A. & Rhoderick, J.F. Nγ-Aryl glutamine analogues as probes of the ASCT2 neutral amino acid transporter binding site. Bioorg. Med. Chem. 13, 1111–1118 (2005).

    Article  CAS  PubMed  Google Scholar 

  15. Lomenick, B. et al. Target identification using drug affinity responsive target stability (DARTS). Proc. Natl. Acad. Sci. USA 106, 21984–21989 (2009).

    Article  CAS  PubMed  Google Scholar 

  16. Canul-Tec, J.C. et al. Structure and allosteric inhibition of excitatory amino acid transporter 1. Nature 544, 446–451 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Fuchs, B.C. & Bode, B.P. Amino acid transporters ASCT2 and LAT1 in cancer: partners in crime? Semin. Cancer Biol. 15, 254–266 (2005).

    Article  CAS  PubMed  Google Scholar 

  18. Nicklin, P. et al. Bidirectional transport of amino acids regulates mTOR and autophagy. Cell 136, 521–534 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Vichai, V. & Kirtikara, K. Sulforhodamine B colorimetric assay for cytotoxicity screening. Nat. Protoc. 1, 1112–1116 (2006).

    Article  CAS  PubMed  Google Scholar 

  20. Rathmell, J.C. T cell Myc-tabolism. Immunity 35, 845–846 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Skala, M. & Ramanujam, N. Multiphoton redox ratio imaging for metabolic monitoring in vivo. Methods Mol. Biol. 594, 155–162 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Walsh, A.J. et al. Quantitative optical imaging of primary tumor organoid metabolism predicts drug response in breast cancer. Cancer Res. 74, 5184–5194 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. DeBerardinis, R.J., Sayed, N., Ditsworth, D. & Thompson, C.B. Brick by brick: metabolism and tumor cell growth. Curr. Opin. Genet. Dev. 18, 54–61 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Hanover, J.A., Krause, M.W. & Love, D.C. The hexosamine signaling pathway: O-GlcNAc cycling in feast or famine. Biochim. Biophys. Acta 1800, 80–95 (2010).

    Article  CAS  PubMed  Google Scholar 

  25. Obeid, L.M., Linardic, C.M., Karolak, L.A. & Hannun, Y.A. Programmed cell death induced by ceramide. Science 259, 1769–1771 (1993).

    Article  CAS  PubMed  Google Scholar 

  26. Tresse, E., Kosta, A., Giusti, C., Luciani, M.F. & Golstein, P. A UDP-glucose derivative is required for vacuolar autophagic cell death. Autophagy 4, 680–691 (2008).

    Article  CAS  PubMed  Google Scholar 

  27. Sentelle, R.D. et al. Ceramide targets autophagosomes to mitochondria and induces lethal mitophagy. Nat. Chem. Biol. 8, 831–838 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Dall'Armi, C., Devereaux, K.A. & Di Paolo, G. The role of lipids in the control of autophagy. Curr. Biol. 23, R33–R45 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Shatz, O., Holland, P., Elazar, Z. & Simonsen, A. Complex relations between phospholipids, autophagy, and neutral lipids. Trends Biochem. Sci. 41, 907–923 (2016).

    Article  CAS  PubMed  Google Scholar 

  30. Huang, F., Zhang, Q., Ma, H., Lv, Q. & Zhang, T. Expression of glutaminase is upregulated in colorectal cancer and of clinical significance. Int. J. Clin. Exp. Pathol. 7, 1093–1100 (2014).

    PubMed  PubMed Central  Google Scholar 

  31. Xiang, Y. et al. Targeted inhibition of tumor-specific glutaminase diminishes cell-autonomous tumorigenesis. J. Clin. Invest. 125, 2293–2306 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Seltzer, M.J. et al. Inhibition of glutaminase preferentially slows growth of glioma cells with mutant IDH1. Cancer Res. 70, 8981–8987 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Gross, M.I. et al. Antitumor activity of the glutaminase inhibitor CB-839 in triple-negative breast cancer. Mol. Cancer Ther. 13, 890–901 (2014).

    Article  CAS  PubMed  Google Scholar 

  34. Suzuki, S. et al. Phosphate-activated glutaminase (GLS2), a p53-inducible regulator of glutamine metabolism and reactive oxygen species. Proc. Natl. Acad. Sci. USA 107, 7461–7466 (2010).

    Article  CAS  PubMed  Google Scholar 

  35. Chiu, M. et al. GPNA inhibits the sodium-independent transport system L for neutral amino acids. Amino Acids 49, 1365–1372 (2017).

    Article  CAS  PubMed  Google Scholar 

  36. McKinley, E.T., Zhao, P., Coffey, R.J., Washington, M.K. & Manning, H.C. 3′-Deoxy-3′-[18F]-fluorothymidine PET imaging reflects PI3K–mTOR-mediated pro-survival response to targeted therapy in colorectal cancer. PLoS One 9, e108193 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Wiza, C., Nascimento, E.B. & Ouwens, D.M. Role of PRAS40 in Akt and mTOR signaling in health and disease. Am. J. Physiol. Endocrinol. Metab. 302, E1453–E1460 (2012).

    Article  CAS  PubMed  Google Scholar 

  38. Santio, N.M. et al. The PIM1 kinase promotes prostate cancer cell migration and adhesion via multiple signalling pathways. Exp. Cell Res. 342, 113–124 (2016).

    Article  CAS  PubMed  Google Scholar 

  39. Rhoads, J.M. et al. L-Glutamine stimulates intestinal cell proliferation and activates mitogen-activated protein kinases. Am. J. Physiol. 272, G943–G953 (1997).

    CAS  PubMed  Google Scholar 

  40. Meiler, J. & Baker, D. ROSETTALIGAND: protein–small molecule docking with full side-chain flexibility. Proteins 65, 538–548 (2006).

    Article  CAS  PubMed  Google Scholar 

  41. Kondo, J. et al. Retaining cell–cell contact enables preparation and culture of spheroids composed of pure primary cancer cells from colorectal cancer. Proc. Natl. Acad. Sci. USA 108, 6235–6240 (2011).

    Article  CAS  PubMed  Google Scholar 

  42. McKinley, E.T. et al. 18FDG-PET predicts pharmacodynamic response to OSI-906, a dual IGF-1R/IR inhibitor, in preclinical mouse models of lung cancer. Clin. Cancer Res. 17, 3332–3340 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors acknowledge M. Tantawy for assistance with PET imaging, F. Revetta for histology expertise, and A. Rosenberg and A. Cohen for helpful discussions and editorial assistance. The authors wish to acknowledge research support from the Vanderbilt Ingram Cancer Center Support Grant (National Institutes of Health (NIH) National Cancer Institute (NCI) P30CA068485, H.C.M.), which supports the Vanderbilt-Ingram Cancer Center (VICC) Chemical Synthesis Core, Vanderbilt University Medical Center (VUMC) Radiochemistry Core, and Center for Small Animal Imaging; the Kleberg Foundation (H.C.M.); a Vanderbilt Trans-Institutional Program (TIPS) Award to the Vanderbilt Center for Molecular Probes (H.C.M.); the Vanderbilt Specialized Program of Research Excellence (SPORE) in Gastrointestinal Cancer (NIH NCI P50CA095103, R.J.C. and H.C.M.); an Outstanding Investigator Award from the NCI (R35CA197570, R.J.C.); and the Vanderbilt Digestive Disease Research Center (NIH National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) P30DK058404, H.C.M.).

Author information

Authors and Affiliations

Authors

Contributions

M.L.S. performed or designed most of the experiments with assistance from A.F., L.G., P.Z., and J.L. S.T.S. and J.A.S. performed computational modeling. J.K. and R.J.C. performed organoid studies. J.B. enrolled individuals who provided tissues for organoids and PDXs. J.T.S. and M.C.S. performed optical redox ratio measurements. M.O.J. and J.C.R. performed T cell experiments. M.K.W. interpreted pathology samples. M.L.N. oversaw radiopharmaceutical production and provided technical assistance. H.C.M. designed and supervised the study. M.L.S., M.L.N., and H.C.M. wrote the manuscript. All authors edited and approved the manuscript.

Corresponding author

Correspondence to H Charles Manning.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–21 (PDF 4353 kb)

Life Sciences Reporting Summary (PDF 165 kb)

Supplementary Data 1

Full in vivo metabolomic data set (XLSX 627 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Schulte, M., Fu, A., Zhao, P. et al. Pharmacological blockade of ASCT2-dependent glutamine transport leads to antitumor efficacy in preclinical models. Nat Med 24, 194–202 (2018). https://doi.org/10.1038/nm.4464

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4464

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer