Skip to main content
Log in

Identification and Validation of a Novel Glycolysis-Related Gene Signature for Predicting the Prognosis and Therapeutic Response in Triple-Negative Breast Cancer

  • Original Research
  • Published:
Advances in Therapy Aims and scope Submit manuscript

Abstract

Introduction

A high malignancy rate and poor prognosis are common problems with triple-negative breast cancer (TNBC). There is increasing evidence that glycolysis plays vital roles in tumorigenesis, tumor invasion, immune evasion, chemoresistance, and metastasis. However, a comprehensive analysis of the diagnostic and prognostic significance of glycolysis in TNBC is lacking.

Methods

Transcriptomic and clinical data of TNBC patients were obtained from The Cancer Genome Atlas (TCGA) and Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) databases, respectively. Glycolysis-related genes (GRGs) were collected from the Molecular Signatures Database (MSigDB). Differential comparative analysis was performed to obtain the differentially expressed (DE)-GRGs associated with TNBC. Based on the DE-GRGs, a glycolysis-related risk signature was established using Least Absolute Shrinkage and Selector Operation (LASSO) and multivariable Cox regression analyses. The prognostic value, tumor microenvironment, mutation status, and chemotherapy response of different risk groups were analyzed. An independent cohort from the METABRIC database was used for external validation. Furthermore, the expression patterns of five genes derived from the prognostic model were validated by quantitative real-time polymerase chain reaction (RT-qPCR).

Results

The glycolysis-related prognostic signature included five genes (IFNG, ACSS2, IRS2, GFUS, and GAL3ST1) and predicted the prognosis of TNBC patients independent of clinical factors (p < 0.05). Patients were divided into high- and low-risk groups based on the median risk score. Compared to low-risk TNBC patients, high-risk patients had significantly decreased overall survival (HR = 2.718, p < 0.001). Receiver operating characteristic and calibration curves demonstrated that the model had high performance in terms of predicting survival and risk stratification. The results remained consistent after external verification. Additionally, the tumor immune microenvironment significantly differed between the risk groups. Low-risk TNBC patients had a better immunotherapy response than high-risk patients. High-risk TNBC patients with a poor prognosis may benefit from targeted therapy.

Conclusions

This study developed a novel glycolysis and prognosis-related (GRP) signature based on GRGs to predict the prognosis of TNBC patients, and may aid clinical decision-making for these patients.

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
Fig. 6
Fig. 7
Fig. 8
Fig. 9
Fig. 10

Similar content being viewed by others

References

  1. Dent R, Trudeau M, Pritchard KI, Hanna WM, Kahn HK, Sawka CA, et al. Triple-negative breast cancer: clinical features and patterns of recurrence. Clin Cancer Res. 2007;13(15 Pt 1):4429–34.

    Article  Google Scholar 

  2. Bianchini G, Balko JM, Mayer IA, Sanders ME, Gianni L. Triple-negative breast cancer: challenges and opportunities of a heterogeneous disease. Nat Rev Clin Oncol. 2016;13(11):674–90.

    Article  CAS  Google Scholar 

  3. Emens LA. Breast cancer immunotherapy: facts and hopes. Clin Cancer Res. 2018;24(3):511–20.

    Article  CAS  Google Scholar 

  4. Waks AG, Winer EP. Breast cancer treatment: a review. JAMA. 2019;321(3):288–300.

    Article  CAS  Google Scholar 

  5. Lee JS, Yost SE, Yuan Y. Neoadjuvant treatment for triple negative breast cancer: recent progresses and challenges. Cancers. 2020;12(6):1404.

    Article  CAS  Google Scholar 

  6. André F, Zielinski CC. Optimal strategies for the treatment of metastatic triple-negative breast cancer with currently approved agents. Ann Oncol. 2012;23(Suppl 6):vi46-51.

    Article  Google Scholar 

  7. Lluch A, Barrios CH, Torrecillas L, Ruiz-Borrego M, Bines J, Segalla J, et al. Phase III trial of adjuvant capecitabine after standard neo-/adjuvant chemotherapy in patients with early triple-negative breast cancer (GEICAM/2003-11_CIBOMA/2004-01). J Clin Oncol. 2020;38(3):203–13.

    Article  CAS  Google Scholar 

  8. Ganapathy-Kanniappan S. Molecular intricacies of aerobic glycolysis in cancer: current insights into the classic metabolic phenotype. Crit Rev Biochem Mol Biol. 2018;53(6):667–82.

    Article  CAS  Google Scholar 

  9. Dias AS, Almeida CR, Helguero LA, Duarte IF. Metabolic crosstalk in the breast cancer microenvironment. Eur J Cancer (Oxford, England: 1990). 2019;121:154–71.

    Article  CAS  Google Scholar 

  10. Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science (New York, NY). 2009;324(5930):1029–33.

    Article  CAS  Google Scholar 

  11. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.

    Article  CAS  Google Scholar 

  12. Sansone P, Bromberg J. Targeting the interleukin-6/Jak/stat pathway in human malignancies. J Clin Oncol. 2012;30(9):1005–14.

    Article  CAS  Google Scholar 

  13. Yu H, Lee H, Herrmann A, Buettner R, Jove R. Revisiting STAT3 signalling in cancer: new and unexpected biological functions. Nat Rev Cancer. 2014;14(11):736–46.

    Article  CAS  Google Scholar 

  14. Meric-Bernstam F, Gonzalez-Angulo AM. Targeting the mTOR signaling network for cancer therapy. J Clin Oncol. 2009;27(13):2278–87.

    Article  CAS  Google Scholar 

  15. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490(7418):61–70.

  16. Alam H, Tang M, Maitituoheti M, Dhar SS, Kumar M, Han CY, et al. KMT2D deficiency impairs super-enhancers to confer a glycolytic vulnerability in lung cancer. Cancer Cell. 2020;37(4):599-617.e7.

    Article  CAS  Google Scholar 

  17. Kim HS, Xiao C, Wang RH, Lahusen T, Xu X, Vassilopoulos A, et al. Hepatic-specific disruption of SIRT6 in mice results in fatty liver formation due to enhanced glycolysis and triglyceride synthesis. Cell Metab. 2010;12(3):224–36.

    Article  CAS  Google Scholar 

  18. Warburg O. On the origin of cancer cells. Science (New York, NY). 1956;123(3191):309–14.

    Article  CAS  Google Scholar 

  19. Gatenby RA, Gillies RJ. Why do cancers have high aerobic glycolysis? Nat Rev Cancer. 2004;4(11):891–9.

    Article  CAS  Google Scholar 

  20. Fong MY, Zhou W, Liu L, Alontaga AY, Chandra M, Ashby J, et al. Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat Cell Biol. 2015;17(2):183–94.

    Article  CAS  Google Scholar 

  21. Jin J, Qiu S, Wang P, Liang X, Huang F, Wu H, et al. Cardamonin inhibits breast cancer growth by repressing HIF-1α-dependent metabolic reprogramming. J Exp Clin Cancer Res. 2019;38(1):377.

    Article  Google Scholar 

  22. Schug ZT, Peck B, Jones DT, Zhang Q, 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(1):57–71.

    Article  CAS  Google Scholar 

  23. Bulusu V, Tumanov S, Michalopoulou E, van den Broek NJ, MacKay G, Nixon C, et al. Acetate recapturing by nuclear acetyl-CoA synthetase 2 prevents loss of histone acetylation during oxygen and serum limitation. Cell Rep. 2017;18(3):647–58.

    Article  CAS  Google Scholar 

  24. Miller KD, Pniewski K, Perry CE, Papp SB, Shaffer JD, Velasco-Silva JN, et al. Targeting ACSS2 with a transition-state mimetic inhibits triple-negative breast cancer growth. Can Res. 2021;81(5):1252–64.

    Article  CAS  Google Scholar 

  25. Alderton GK. Metabolism: acetate nourishes stressed tumour cells. Nat Rev Cancer. 2015;15(2):67.

    Article  CAS  Google Scholar 

  26. Jackson JG, Zhang X, Yoneda T, Yee D. Regulation of breast cancer cell motility by insulin receptor substrate-2 (IRS-2) in metastatic variants of human breast cancer cell lines. Oncogene. 2001;20(50):7318–25.

    Article  CAS  Google Scholar 

  27. Cui X, Kim HJ, Kuiatse I, Kim H, Brown PH, Lee AV. Epidermal growth factor induces insulin receptor substrate-2 in breast cancer cells via c-Jun NH(2)-terminal kinase/activator protein-1 signaling to regulate cell migration. Can Res. 2006;66(10):5304–13.

    Article  CAS  Google Scholar 

  28. Pankratz SL, Tan EY, Fine Y, Mercurio AM, Shaw LM. Insulin receptor substrate-2 regulates aerobic glycolysis in mouse mammary tumor cells via glucose transporter 1. J Biol Chem. 2009;284(4):2031–7.

    Article  CAS  Google Scholar 

  29. Tonetti M, Sturla L, Bisso A, Benatti U, De Flora A. Synthesis of GDP-L-fucose by the human FX protein. J Biol Chem. 1996;271(44):27274–9.

    Article  CAS  Google Scholar 

  30. Sun J, Thurin J, Cooper HS, Wang P, Mackiewicz M, Steplewski Z, et al. Elevated expression of H type GDP-l-fucose: β-d-galactoside α-2-l-fucosyltransferase is associated with human colon adenocarcinoma progression. Proc Natl Acad Sci USA. 1995;92(12):5724–8.

    Article  CAS  Google Scholar 

  31. Sun Y, Liu X, Zhang Q, Mao X, Feng L, Su P, et al. Oncogenic potential of TSTA3 in breast cancer and its regulation by the tumor suppressors miR-125a-5p and miR-125b. Tumour Biol. 2016;37(4):4963–72.

    Article  CAS  Google Scholar 

  32. Owczarek TB, Suchanski J, Pula B, Kmiecik AM, Chadalski M, Jethon A, et al. Galactosylceramide affects tumorigenic and metastatic properties of breast cancer cells as an anti-apoptotic molecule. PLoS One. 2013;8(12): e84191.

    Article  Google Scholar 

  33. Gresser I. Biologic effects of interferons. J Invest Dermatol. 1990;95(6 Suppl):66s–71s.

    Article  CAS  Google Scholar 

  34. Castro F, Cardoso AP, Gonçalves RM, Serre K, Oliveira MJ. Interferon-gamma at the crossroads of tumor immune surveillance or evasion. Front Immunol. 2018;9:847.

    Article  Google Scholar 

  35. Mojic M, Takeda K, Hayakawa Y. The dark side of IFN-γ: its role in promoting cancer immunoevasion. Int J Mol Sci. 2017;19(1):89.

    Article  Google Scholar 

  36. Siska PJ, Rathmell JC. Metabolic signaling drives IFN-γ. Cell Metab. 2016;24(5):651–2.

    Article  CAS  Google Scholar 

  37. Jiang B. Aerobic glycolysis and high level of lactate in cancer metabolism and microenvironment. Genes Dis. 2017;4(1):25–7.

    Article  Google Scholar 

  38. Zhang Y, Yu G, Chu H, Wang X, Xiong L, Cai G, et al. Macrophage-associated PGK1 phosphorylation promotes aerobic glycolysis and tumorigenesis. Mol Cell. 2018;71(2):201-15.e7.

    Article  CAS  Google Scholar 

  39. Jiang Z, Liu Z, Li M, Chen C, Wang X. Increased glycolysis correlates with elevated immune activity in tumor immune microenvironment. EBioMedicine. 2019;42:431–42.

    Article  Google Scholar 

  40. Deberardinis RJ, Sayed N, Ditsworth D, Thompson CB. Brick by brick: metabolism and tumor cell growth. Curr Opin Genet Dev. 2008;18(1):54–61.

    Article  CAS  Google Scholar 

  41. Ohashi T, Akazawa T, Aoki M, Kuze B, Mizuta K, Ito Y, et al. Dichloroacetate improves immune dysfunction caused by tumor-secreted lactic acid and increases antitumor immunoreactivity. Int J Cancer. 2013;133(5):1107–18.

    Article  CAS  Google Scholar 

  42. Chang CH, Qiu J, O’Sullivan D, Buck MD, Noguchi T, Curtis JD, et al. Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell. 2015;162(6):1229–41.

    Article  CAS  Google Scholar 

  43. Vasaikar S, Huang C, Wang X, Petyuk VA, Savage SR, Wen B, et al. Proteogenomic analysis of human colon cancer reveals new therapeutic opportunities. Cell. 2019;177(4):1035-49.e19.

    Article  CAS  Google Scholar 

  44. Cascone T, McKenzie JA, Mbofung RM, Punt S, Wang Z, Xu C, et al. Increased tumor glycolysis characterizes immune resistance to adoptive T cell therapy. Cell Metab. 2018;27(5):977-87.e4.

    Article  CAS  Google Scholar 

  45. Rooney MS, Shukla SA, Wu CJ, Getz G, Hacohen N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell. 2015;160(1–2):48–61.

    Article  CAS  Google Scholar 

  46. Moossavi M, Parsamanesh N, Bahrami A, Atkin SL, Sahebkar A. Role of the NLRP3 inflammasome in cancer. Mol Cancer. 2018;17(1):158.

    Article  CAS  Google Scholar 

  47. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12(4):252–64.

    Article  CAS  Google Scholar 

  48. Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515(7528):568–71.

    Article  CAS  Google Scholar 

  49. Karn T, Jiang T, Hatzis C, Sänger N, El-Balat A, Rody A, et al. Association between genomic metrics and immune infiltration in triple-negative breast cancer. JAMA Oncol. 2017;3(12):1707–11.

    Article  Google Scholar 

  50. Kon E, Benhar I. Immune checkpoint inhibitor combinations: current efforts and important aspects for success. Drug Resist Updates. 2019;45:13–29.

    Article  Google Scholar 

  51. Francisco LM, Salinas VH, Brown KE, Vanguri VK, Freeman GJ, Kuchroo VK, et al. PD-L1 regulates the development, maintenance, and function of induced regulatory T cells. J Exp Med. 2009;206(13):3015–29.

    Article  CAS  Google Scholar 

  52. Bhattacharya B, Mohd Omar MF, Soong R. The Warburg effect and drug resistance. Br J Pharmacol. 2016;173(6):970–9.

    Article  CAS  Google Scholar 

  53. He M, Jin Q, Chen C, Liu Y, Ye X, Jiang Y, et al. The miR-186-3p/EREG axis orchestrates tamoxifen resistance and aerobic glycolysis in breast cancer cells. Oncogene. 2019;38(28):5551–65.

    Article  CAS  Google Scholar 

  54. Metzger-Filho O, Tutt A, de Azambuja E, Saini KS, Viale G, Loi S, et al. Dissecting the heterogeneity of triple-negative breast cancer. J Clin Oncol. 2012;30(15):1879–87.

    Article  CAS  Google Scholar 

  55. Asghar US, Barr AR, Cutts R, Beaney M, Babina I, Sampath D, et al. Single-cell dynamics determines response to CDK4/6 inhibition in triple-negative breast cancer. Clin Cancer Res. 2017;23(18):5561–72.

    Article  CAS  Google Scholar 

  56. Teo ZL, Versaci S, Dushyanthen S, Caramia F, Savas P, Mintoff CP, et al. Combined CDK4/6 and PI3Kα inhibition is synergistic and immunogenic in triple-negative breast cancer. Cancer Res. 2017;77(22):6340–52.

    Article  CAS  Google Scholar 

  57. Mao Z, Zhang W. Role of mTOR in glucose and lipid metabolism. Int J Mol Sci. 2018;19(7):2043.

    Article  Google Scholar 

  58. Yamamoto T, Kanaya N, Somlo G, Chen S. Synergistic anti-cancer activity of CDK4/6 inhibitor palbociclib and dual mTOR kinase inhibitor MLN0128 in pRb-expressing ER-negative breast cancer. Breast Cancer Res Treat. 2019;174(3):615–25.

    Article  CAS  Google Scholar 

  59. Lanceta L, Lypova N, O’Neill C, Li X, Rouchka E, Chesney J, et al. Differential gene expression analysis of palbociclib-resistant TNBC via RNA-seq. Breast Cancer Res Treat. 2021;186(3):677–86.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Funding

This project was financially supported by the National Science Foundation of China (grant number: 81871295, 82001740). Ji-min Cao funded the journal’s Rapid Service fees.

Editorial Assistance

We gratefully acknowledge contributions from the GEO, TCGA, and METABRIC databases. The English in this document has been checked by at least two professional editors of Textcheck (http://www.textcheck.com/) both native speakers of English.

Author Contributions

All authors were involved in drafting the article or revising it critically for important content. Jian Zheng and Ji-Min Cao developed the methodology and acquired the related data. Conception and design of the study: Jian Zheng and Ji-Min Cao. Acquisition of data: Yi-Fan Zhang, Guo-Hui Han, Meng-Ying Fan, Ming-Hui Du, Guo-Chen Zhang and Yue Feng. Analysis and interpretation of data: Run-Qi Chen, Jun Qiao, Jian Zheng, Bo Zhang, Yi-Fan Zhang and Sheng-Xiao Zhang. Jian Zheng collected specimens and carried out in vitro assays. Drafting the article: Jian Zheng, Yi-Fan Zhang and Sheng-Xiao Zhang. Revising the article critically: Ji-Min Cao, Sheng-Xiao Zhang, and Guo-Hui Han. All authors contributed to the article and approved the submitted version.

Disclosures

All named authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Compliance with Ethics Guidelines

The study was approved by the Ethics Review Board at Shanxi Cancer Hospital. All patients provided written informed consent for the experimental analysis of their excised tissues.

Data Availability

The datasets analyzed for this study can be found in the [TCGA database] [https://cancergenome.nih.gov], [METABRIC database] [http://molonc.bccrc.ca/aparicio-lab/research/metabric/] and [GTEx database] [https://gtexportal.org/]. All data generated or analyzed during this study are included as supplementary information files.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ji-Min Cao.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (PDF 703 KB)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zheng, J., Zhang, YF., Han, GH. et al. Identification and Validation of a Novel Glycolysis-Related Gene Signature for Predicting the Prognosis and Therapeutic Response in Triple-Negative Breast Cancer. Adv Ther 40, 310–330 (2023). https://doi.org/10.1007/s12325-022-02330-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12325-022-02330-y

Keywords

Navigation