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:

Cellular and Molecular Biology

Metabolic stress induces GD2+ cancer stem cell-like phenotype in triple-negative breast cancer

Abstract

Background

Metabolic stress resulting from nutrient deficiency is one of the hallmarks of a growing tumour. Here, we tested the hypothesis that metabolic stress induces breast cancer stem-like cell (BCSC) phenotype in triple-negative breast cancer (TNBC).

Methods

Flow cytometry for GD2 expression, mass spectrometry and Ingenuity Pathway Analysis for metabolomics, bioinformatics, in vitro tumorigenesis and in vivo models were used.

Results

Serum/glucose deprivation not only increased stress markers but also enhanced GD2+ BCSC phenotype and function in TNBC cells. Global metabolomics profiling identified upregulation of glutathione biosynthesis in GD2high cells, suggesting a role of glutamine in the BCSC phenotype. Cueing from the upregulation of the glutamine transporters in primary breast tumours, inhibition of glutamine uptake using small-molecule inhibitor V9302 reduced GD2+ cells by 70–80% and BCSC characteristics in TNBC cells. Mechanistic studies revealed inhibition of the mTOR pathway and induction of ferroptosis by V9302 in TNBC cells. Finally, inhibition of glutamine uptake significantly reduced in vivo tumour growth in a TNBC patient-derived xenograft model using NSG (non-obese diabetic/severe combined immunodeficiency with a complete null allele of the IL-2 receptor common gamma chain) mice.

Conclusion

Here, we show metabolic stress results in GD2+ BCSC phenotype in TNBC and glutamine contributes to GD2+ phenotype, and targeting the glutamine transporters could complement conventional chemotherapy in TNBC.

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

Fig. 1: Serum and glucose deprivation induces GD2 expression in TNBC.
Fig. 2: Metabolomic profiling of breast cancer stem cells.
Fig. 3: METABRIC dataset show higher expression of SLC1A5 in breast cancer patients and is associated with poor survivability.
Fig. 4: V9302 inhibits GD2 expression, BCSC function and the glutathione (GSH) pathway in TNBC cells.
Fig. 5: V9302 induces synergistic killing and ferroptosis and inhibits the mTOR pathway in TNBC cells.
Fig. 6: Inhibition of glutamine metabolism reduces in vivo tumorigenesis.

Similar content being viewed by others

References

  1. Bauer KR, Brown M, Cress RD, Parise CA, Caggiano V. Descriptive analysis of estrogen receptor (ER)-negative, progesterone receptor (PR)-negative, and HER2-negative invasive breast cancer, the so-called triple-negative phenotype: a population-based study from the California cancer Registry. Cancer. 2007;109:1721–8.

    Article  PubMed  Google Scholar 

  2. Sharma P. Biology and management of patients with triple-negative breast cancer. Oncologist. 2016;21:1050–62.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Cluntun AA, Lukey MJ, Cerione RA, Locasale JW. Glutamine metabolism in cancer: understanding the heterogeneity trends. Cancer. 2017;3:169–80.

    CAS  Google Scholar 

  4. Ahmadiankia N, Bagheri M, Fazli M. Nutrient deprivation modulates the metastatic potential of breast cancer cells. Rep Biochem Mol Biol. 2019;8:139–46.

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Sancho P, Barneda D, Heeschen C. Hallmarks of cancer stem cell metabolism. Br J Cancer. 2016;114:1305–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Liu P, Liao J, Tang Z, Wu W, Yang J, Zeng Z, et al. Metabolic regulation of cancer cell side population by glucose through activation of the Akt pathway. Cell Death Differ. 2014;21:124–35.

    Article  PubMed  Google Scholar 

  7. Munir R, Lisec J, Swinnen JV, Zaidi N. Lipid metabolism in cancer cells under metabolic stress. Br J Cancer. 2019;120:1090–8.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Aykin-Burns N, Ahmad IM, Zhu Y, Oberley LW, Spitz DR. Increased levels of superoxide and H2O2 mediate the differential susceptibility of cancer cells versus normal cells to glucose deprivation. Biochem J. 2009;418:29–37.

    Article  CAS  PubMed  Google Scholar 

  9. Simons AL, Mattson DM, Dornfeld K, Spitz DR. Glucose deprivation-induced metabolic oxidative stress and cancer therapy. J Cancer Res Ther. 2009;5:S2–6. Suppl 1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Altman BJ, Stine ZE, Dang CV. From Krebs to clinic: glutamine metabolism to cancer therapy. Nat Rev Cancer. 2016;16:619–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Sun X, Wang M, Wang M, Yu X, Guo J, Sun T, et al. Metabolic reprogramming in triple-negative breast cancer. Front Oncol. 2020;10:428.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Qie S, Yoshida A, Parnham S, Oleinik N, Beeson GC, Beeson CC, et al. Targeting glutamine-addiction and overcoming CDK4/6 inhibitor resistance in human esophageal squamous cell carcinoma. Nat Commun. 2019;10:1296.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Wang JB, Erickson JW, Fuji R, Ramachandran S, Gao P, Dinavahi R, et al. Targeting mitochondrial glutaminase activity inhibits oncogenic transformation. Cancer Cell. 2010;18:207–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Yoo HC, Park SJ, Nam M, Kang J, Kim K, Yeo JH, et al. A variant of SLC1A5 is a mitochondrial glutamine transporter for metabolic reprogramming in cancer cells. Cell Metab. 2020;31:267.

    Article  CAS  PubMed  Google Scholar 

  15. Qie S, Liang D, Yin C, Gu W, Meng M, Wang C, et al. Glutamine depletion and glucose depletion trigger growth inhibition via distinctive gene expression reprogramming. Cell Cycle. 2012;11:3679–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Welbourne TC. Ammonia production and glutamine incorporation into glutathione in the functioning rat kidney. Can J Biochem. 1979;57:233–7.

    Article  CAS  PubMed  Google Scholar 

  17. Lampa M, Arlt H, He T, Ospina B, Reeves J, Zhang B, et al. Glutaminase is essential for the growth of triple-negative breast cancer cells with a deregulated glutamine metabolism pathway and its suppression synergizes with mTOR inhibition. PLoS ONE. 2017;12:e0185092.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Gross MI, Demo SD, Dennison JB, Chen L, Chernov-Rogan T, Goyal B. et al. Antitumor activity of the glutaminase inhibitor CB-839 in triple-negative breast cancer. Mol Cancer Ther. 2014;13:890–901.

    Article  CAS  PubMed  Google Scholar 

  19. van Geldermalsen M, Wang Q, Nagarajah R, Marshall AD, Thoeng A, Gao D. et al. ASCT2/SLC1A5 controls glutamine uptake and tumour growth in triple-negative basal-like breast cancer. Oncogene. 2016;35:3201–8.

    Article  PubMed  Google Scholar 

  20. Park S-Y, Choi J-H, Nam J-S. Targeting cancer stem cells in triple-negative breast cancer. Cancers. 2019;11:965

    Article  CAS  PubMed Central  Google Scholar 

  21. De Francesco EM, Sotgia F, Lisanti MP. Cancer stem cells (CSCs): metabolic strategies for their identification and eradication. Biochem J. 2018;475:1611–34.

    Article  PubMed  Google Scholar 

  22. Battula VL, Shi Y, Evans KW, Wang R-Y, Spaeth EL, Jacamo RO, et al. Ganglioside GD2 identifies breast cancer stem cells and promotes tumorigenesis. J Clin Investig. 2012;122:2066–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Ly S, Anand V, El-Dana F, Nguyen K, Cai Y, Cai S, et al. Anti-GD2 antibody dinutuximab inhibits triple-negative breast tumor growth by targeting GD2+ breast cancer stem-like cells. J Immunother. Cancer. 2021;9:e001197.

  24. Shao C, Anand V, Andreeff M, Battula VL. Ganglioside GD2: a novel therapeutic target in triple-negative breast cancer. Ann NY Acad Sci. 2021:1–19. Epub ahead of print.

  25. Nguyen K, Yan Y, Yuan B, Dasgupta A, Sun J, Mu H, et al. ST8SIA1 regulates tumor growth and metastasis in TNBC by activating the FAK–AKT–mTOR signaling pathway. Mol Cancer Ther. 2018;17:2689–701.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Yadav UP, Singh T, Kumar P, Sharma P, Kaur H, Sharma S, et al. Metabolic adaptations in cancer stem cells. Front Oncol. 2020;10:1010.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Amara CS, Ambati CR, Vantaku V, Badrajee Piyarathna DW, Donepudi SR, Ravi SS, et al. Serum metabolic profiling identified a distinct metabolic signature in bladder cancer smokers: a key metabolic enzyme associated with patient survival. Cancer Epidemiol Biomark Prev. 2019;28:770–81.

    Article  CAS  Google Scholar 

  28. Arnold JM, Gu F, Ambati CR, Rasaily U, Ramirez-Pena E, Joseph R. et al. UDP-glucose 6-dehydrogenase regulates hyaluronic acid production and promotes breast cancer progression. Oncogene. 2020;39:3089–101.

    Article  CAS  PubMed  Google Scholar 

  29. Martinez AM, Kim A, Yang WS. Detection of ferroptosis by BODIPY 581/591 C11. Methods Mol Biol. 2020;2108:125–30.

    Article  CAS  PubMed  Google Scholar 

  30. Rey O, Young SH, Jacamo R, Moyer MP, Rozengurt E. Extracellular calcium sensing receptor stimulation in human colonic epithelial cells induces intracellular calcium oscillations and proliferation inhibition. J Cell Physiol. 2010;225:73–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Echeverria GV, Ge Z, Seth S, Zhang X, Jeter-Jones S, Zhou X, et al. Resistance to neoadjuvant chemotherapy in triple-negative breast cancer mediated by a reversible drug-tolerant state. Sci Transl Med. 2019;11:eaav0936.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Schulte ML, Fu A, Zhao P, Li J, Geng L, Smith ST, et al. Pharmacological blockade of ASCT2-dependent glutamine transport leads to antitumor efficacy in preclinical models. Nat Med. 2018;24:194–202.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Broer A, Fairweather S, Broer S. Disruption of amino acid homeostasis by novel ASCT2 inhibitors involves multiple targets. Front Pharmacol. 2018;9:785.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Edwards DN, Ngwa VM, Raybuck AL, Wang S, Hwang Y, Kim LC, et al. Selective glutamine metabolism inhibition in tumor cells improves antitumor T lymphocyte activity in triple-negative breast cancer. J Clin Invest. 2021;131.

  35. Ueng SH, Chen SC, Chang YS, Hsueh S, Lin YC, Chien HP, et al. Phosphorylated mTOR expression correlates with poor outcome in early-stage triple negative breast carcinomas. Int J Clin Exp Pathol. 2012;5:806–13.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Lee E, Yang J, Ku M, Kim NH, Park Y, Park CB, et al. Metabolic stress induces a Wnt-dependent cancer stem cell-like state transition. Cell Death Dis. 2015;6:e1805.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Schug ZT, Peck B, Jones DT, Zhang QF, Grosskurth S, Alam IS, et al. Acetyl-CoA Synthetase 2 promotes acetate utilization and maintains cancer cell growth under metabolic stress. Cancer Cell. 2015;27:57–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. White E. Role of the metabolic stress responses of apoptosis and autophagy in tumor suppression. Ernst Scher Found Symp Proc. 2007;4:23–34.

    Google Scholar 

  39. Caino MC, Chae YC, Vaira V, Ferrero S, Nosotti M, Martin NM, et al. Metabolic stress regulates cytoskeletal dynamics and metastasis of cancer cells. J Clin Invest. 2013;123:2907–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. van den Bijgaart RJE, Kroesen M, Wassink M, Brok IC, Kers-Rebel ED, Boon L, et al. Combined sialic acid and histone deacetylase (HDAC) inhibitor treatment up-regulates the neuroblastoma antigen GD2. J Biol Chem. 2019;294:4437–49.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Moons SJ, Adema GJ, Derks MT, Boltje TJ, Bull C. Sialic acid glycoengineering using N-acetylmannosamine and sialic acid analogs. Glycobiology. 2019;29:433–45.

    CAS  PubMed  Google Scholar 

  42. Schnaar RL, Gerardy-Schahn R, Hildebrandt H. Sialic acids in the brain: gangliosides and polysialic acid in nervous system development, stability, disease, and regeneration. Physiol Rev. 2014;94:461–518.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Teoh ST, Ogrodzinski MP, Ross C, Hunter KW, Lunt SY. Sialic acid metabolism: a key player in breast cancer metastasis revealed by metabolomics. Front Oncol. 2018;8:174.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Schomel N, Hancock SE, Gruber L, Olzomer EM, Byrne FL, Shah D, et al. UGCG influences glutamine metabolism of breast cancer cells. Sci Rep. 2019;9:15665.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Luo Z, Xu J, Sun J, Huang H, Zhang Z, Ma W, et al. Co-delivery of 2-deoxyglucose and a glutamine metabolism inhibitor V9302 via a prodrug micellar formulation for synergistic targeting of metabolism in cancer. Acta Biomater. 2020;105:239–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Esaki N, Ohkawa Y, Hashimoto N, Tsuda Y, Ohmi Y, Bhuiyan RH, et al. ASC amino acid transporter 2, defined by enzyme-mediated activation of radical sources, enhances malignancy of GD2-positive small-cell lung cancer. Cancer Sci. 2018;109:141–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Desideri E, Vegliante R, Cardaci S, Nepravishta R, Paci M, Ciriolo MR. MAPK14/p38alpha-dependent modulation of glucose metabolism affects ROS levels and autophagy during starvation. Autophagy. 2014;10:1652–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Lettieri-Barbato D, Aquilano K. Pushing the limits of cancer therapy: the nutrient game. Front Oncol. 2018;8:148.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Verma N, Vinik Y, Saroha A, Nair NU, Ruppin E, Mills G, et al. Synthetic lethal combination targeting BET uncovered intrinsic susceptibility of TNBC to ferroptosis. Sci Adv. 2020;6(34):eaba8968.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Seibt TM, Proneth B, Conrad M. Role of GPX4 in ferroptosis and its pharmacological implication. Free Radic Biol Med. 2019;133:144–52.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We are grateful to the patients who provided tumour biopsies for PDX model establishment. PDX models and derivatives were obtained from the Cazalot Breast Cancer Model Resource at The University of Texas MD Anderson Cancer Center. This resource was established through a gift from the Cazalot family and from funds from the MD Anderson Cancer Center Breast Cancer Moon Shot Programme. Editorial support was provided by Sunita Patterson and Bryan Tutt in Editing Services, Research Medical Library, MD Anderson Cancer Center. This study was supported by the Department of Defense (DOD), Grant #BC181493 (to VLB) and a Breast Cancer Research Foundation (BCRF) grant (to MA). Metabolomics and data analysis were performed by Metabolomics core facility, Baylor College of Medicine. This project was supported by CPRIT Proteomics and Metabolomics Core Facility (to NP), (RP170005), National Institute of Health (NIH)/National Cancer Institute (NCI) grant (P30 CA125123), Dan L. Duncan Cancer Center and NIH/NCI R01CA220297 (to NP) and NIH/NCI R01CA216426 (to NP). Additional funding sources that supported this work include Cancer Prevention and Research Institute of Texas grants RP150148 (to HP-W).

Author information

Authors and Affiliations

Authors

Contributions

AJ and VLB conceived the ideas, designed experiments, interpreted data and wrote the manuscript with inputs from all authors. AJ, SL and KN performed a majority of the experiments. BY assisted with Western blot and animal experiments. VA performed flow cytometry, qPCR and mammosphere assays. FE-D and ZA assisted with qPCR and mammosphere assays. DWBP and NP performed experiments and analyses pertaining to metabolomics. YY performed bioinformatics and combination index analysis. HP-W contributed to TNBC PDX models. HCM contributed V9302. MA and VLB supervised and directed all aspects of the study.

Corresponding author

Correspondence to V. Lokesh Battula.

Ethics declarations

Competing interests

The authors declare no competing interests.

Ethics approval and consent to participate

This study was performed in accordance with the Declaration of Helsinki. All experiments involving animals were approved by and conducted in accordance with the policies of the Institutional Animal Care and Use Committee (IACUC) of The University of Texas MD Anderson Cancer Center.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jaggupilli, A., Ly, S., Nguyen, K. et al. Metabolic stress induces GD2+ cancer stem cell-like phenotype in triple-negative breast cancer. Br J Cancer 126, 615–627 (2022). https://doi.org/10.1038/s41416-021-01636-y

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41416-021-01636-y

This article is cited by

Search

Quick links