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:

A combination of novel NSC small molecule inhibitor along with doxorubicin inhibits proliferation of triple-negative breast cancer through metabolic reprogramming

A Correction to this article was published on 09 February 2023

This article has been updated

Abstract

Treatment of patients with triple-negative breast cancer (TNBC) has been challenging due to the absence of well-defined molecular targets and the highly invasive and proliferative nature of TNBC cells. Current treatments against TNBC have shown little promise due to high recurrence rate in patients. Consequently, there is a pressing need for novel and efficacious therapies against TNBC. Here, we report the discovery of a novel small molecule inhibitor (NSC33353) with potent anti-tumor activity against TNBC cells. The anti-proliferative effects of this small molecule inhibitor were determined using 2D and 3D cell proliferation assays. We found that NSC33353 significantly reduces the proliferation of TNBC cells in these assays. Using proteomics, next generation sequencing (NGS), and gene enrichment analysis, we investigated global regulatory pathways affected by this compound in TNBC cells. Proteomics data indicate a significant metabolic reprograming affecting both glycolytic enzymes and energy generation through oxidative phosphorylation. Subsequently, using metabolic (Seahorse) and enzymatic assays, we validated our proteomics and NGS analysis findings. Finally, we showed the inhibitory and anti-tumor effects of this small molecule in vitro and confirmed its inhibitory activity in vivo. Doxorubicin is one of the most effective agents in the treatment of TNBC and resistance to this drug has been a major problem. We show that the combination of NSC33353 and doxorubicin suppresses the growth of TNBC cells synergistically, suggesting that NSC33353 enhances TNBC sensitivity to doxorubicin. In summary, our data indicate that the small molecule inhibitor, NSC33353, exhibits anti-tumor activity in TNBC cells, and works in a synergistic fashion with a well-known chemotherapeutic agent.

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: NSC33353 inhibits breast cancer cell proliferation.
Fig. 2: NSC33353 induces apoptosis in TNBC cells.
Fig. 3: NSC33353 inhibitor suppressed migration and invasion of TNBC cells by inhibiting the EMT pathway.
Fig. 4: GSEA analyses showed that NSC33353-treated TNBC cells demonstrated significantly negative glycolysis and oxidative phosphorylation enrichment compared with controls.
Fig. 5: NSC33353 treatment suppressed glycolysis and OXPHOS in TNBC cells.
Fig. 6: NSC33353 treatment significantly suppressed the TNBC tumor growth and suppressed the lung metastasis in vivo.
Fig. 7: NSC33353 treatment reduced the expression of glycolytic, OXPHOS, EMT, angiogenesis, and proliferation markers in vivo.
Fig. 8: Combined treatment with NSC33353 and doxorubicin induced apoptosis in TNBC cells.

Similar content being viewed by others

Change history

References

  1. Yin L, Duan J-J, Bian X-W, Yu S-C. Triple-negative breast cancer molecular subtyping and treatment progress. Breast Cancer Res. 2020;22:61.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Foulkes WD, Smith IE, Reis-Filho JS. Triple-negative breast cancer. N. Engl J Med. 2010;363:1938–48.

    Article  CAS  PubMed  Google Scholar 

  3. Marra A, Trapani D, Viale G, Criscitiello C, Curigliano G. Practical classification of triple-negative breast cancer: intratumoral heterogeneity, mechanisms of drug resistance, and novel therapies. NPJ Breast Cancer. 2020;6:1–16.

    Article  Google Scholar 

  4. 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;13:674–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Arumugam A, Subramani R, Nandy SB, Terreros D, Dwivedi AK, Saltzstein E, et al. Silencing growth hormone receptor inhibits estrogen receptor negative breast cancer through ATP-binding cassette sub-family G member 2. Exp Mol Med. 2019;51:1–13.

    Article  CAS  PubMed  Google Scholar 

  6. Guestini F, Ono K, Miyashita M, Ishida T, Ohuchi N, Nakagawa S, et al. Impact of Topoisomerase IIα, PTEN, ABCC1/MRP1, and KI67 on triple-negative breast cancer patients treated with neoadjuvant chemotherapy. Breast Cancer Res Treat. 2019;173:275–88.

    Article  CAS  PubMed  Google Scholar 

  7. Yamada A, Ishikawa T, Ota I, Kimura M, Shimizu D, Tanabe M, et al. High expression of ATP-binding cassette transporter ABCC11 in breast tumors is associated with aggressive subtypes and low disease-free survival. Breast Cancer Res Treat. 2013;137:773–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Clara JA, Monge C, Yang Y, Takebe N. Targeting signalling pathways and the immune microenvironment of cancer stem cells—A clinical update. Nat Rev Clin Oncol. 2020;17:204–32.

    Article  PubMed  Google Scholar 

  9. Ma F, Li H, Wang H, Shi X, Fan Y, Ding X, et al. Enriched CD44+/CD24− population drives the aggressive phenotypes presented in triple-negative breast cancer (TNBC). Cancer Lett. 2014;353:153–9.

    Article  CAS  PubMed  Google Scholar 

  10. Tan E, Yan M, Campo L, Han C, Takano E, Turley H, et al. The key hypoxia regulated gene CAIX is upregulated in basal-like breast tumours and is associated with resistance to chemotherapy. Br J Cancer. 2009;100:405–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Coradini D, Biganzoli E, Ardoino I, Ambrogi F, Boracchi P, Demicheli R, et al. p53 status identifies triple-negative breast cancer patients who do not respond to adjuvant chemotherapy. Breast. 2015;24:294–7.

    Article  PubMed  Google Scholar 

  12. Garrido-Castro AC, Lin NU, Polyak K. Insights into molecular classifications of triple-negative breast cancer: improving patient selection for treatment. Cancer Discov. 2019;9:176–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. He J, McLaughlin RP, van der Noord V, Foekens JA, Martens JWM, van Westen G, et al. Multi-targeted kinase inhibition alleviates mTOR inhibitor resistance in triple-negative breast cancer. Breast Cancer Res Treat. 2019;178:263–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Kim J-Y, Jung HH, Ahn S, Bae S, Lee SK, Kim SW, et al. The relationship between nuclear factor (NF)-κB family gene expression and prognosis in triple-negative breast cancer (TNBC) patients receiving adjuvant doxorubicin treatment. Sci Rep. 2016;6:31804.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Stover DG, Del Alcazar CRG, Brock J, Guo H, Overmoyer B, Balko J, et al. Phase II study of ruxolitinib, a selective JAK1/2 inhibitor, in patients with metastatic triple-negative breast cancer. NPJ Breast Cancer. 2018;4:1–9.

    Article  CAS  Google Scholar 

  16. Adams S, Diamond JR, Hamilton E, Pohlmann PR, Tolaney SM, Chang C-W, et al. Atezolizumab plus nab-paclitaxel in the treatment of metastatic triple-negative breast cancer with 2-year survival follow-up: a phase 1b clinical trial. JAMA Oncol. 2019;5:334–42.

    Article  PubMed  Google Scholar 

  17. Tolaney SM, Kalinsky K, Kaklamani VG, D'Adamo DR, Aktan G, Tsai ML, et al. A phase Ib/II study of eribulin (ERI) plus pembrolizumab (PEMBRO) in metastatic triple-negative breast cancer (mTNBC)(ENHANCE 1). Cancer Res. 2021;7:3061–8.

    Google Scholar 

  18. Rugo HS, Loi S, Adams S, Schmid P, Schneeweiss A, Barrios CH, et al. Performance of PD-L1 immunohistochemistry (IHC) assays in unresectable locally advanced or metastatic triple-negative breast cancer (mTNBC): post-hoc analysis of IMpassion130. Ann Oncol. 2019;30:v858–59.

    Article  Google Scholar 

  19. Syn NL, Teng MW, Mok TS, Soo RA. De-novo and acquired resistance to immune checkpoint targeting. Lancet Oncol. 2017;18:e731–41.

    Article  PubMed  Google Scholar 

  20. Martinez-Outschoorn UE, Peiris-Pagés M, Pestell RG, Sotgia F, Lisanti MP. Cancer metabolism: a therapeutic perspective. Nat Rev Clin Oncol. 2017;14:11–31.

    Article  CAS  PubMed  Google Scholar 

  21. Pavlova NN, Thompson CB. The emerging hallmarks of cancer metabolism. Cell Metab. 2016;23:27–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Hakimi AA, Reznik E, Lee C-H, Creighton CJ, Brannon AR, Luna A, et al. An integrated metabolic atlas of clear cell renal cell carcinoma. Cancer Cell. 2016;29:104–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Kim J, DeBerardinis RJ. Mechanisms and implications of metabolic heterogeneity in cancer. Cell Metab. 2019;30:434–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Reznik E, Luna A, Aksoy BA, Liu EM, La K, Ostrovnaya I, et al. A landscape of metabolic variation across tumor types. Cell Syst. 2018;6:301–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Warburg O. On the origin of cancer cells. Science. 1956;123:309–14.

    Article  CAS  PubMed  Google Scholar 

  26. Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324:1029–33.

    Article  Google Scholar 

  27. 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 

  28. Lee C-K, Jeong S-H, Jang C, Bae H, Kim YH, Park I, et al. Tumor metastasis to lymph nodes requires YAP-dependent metabolic adaptation. Science 2019;363:644–9.

    Article  CAS  PubMed  Google Scholar 

  29. LeBleu VS, O’Connell JT, Herrera KNG, Wikman H, Pantel K, Haigis MC, et al. PGC-1α mediates mitochondrial biogenesis and oxidative phosphorylation in cancer cells to promote metastasis. Nat Cell Biol. 2014;16:992–1003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lehuédé C, Dupuy F, Rabinovitch R, Jones RG, Siegel PM. Metabolic plasticity as a determinant of tumor growth and metastasis. Cancer Res. 2016;76:5201–8.

    Article  PubMed  Google Scholar 

  31. Simões RV, Serganova IS, Kruchevsky N, Leftin A, Shestov AA, Thaler HT, et al. Metabolic plasticity of metastatic breast cancer cells: adaptation to changes in the microenvironment. Neoplasia. 2015;17:671–84.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Wright HJ, Hou J, Xu B, Cortez M, Potma EO, Tromberg BJ, et al. CDCP1 drives triple-negative breast cancer metastasis through reduction of lipid-droplet abundance and stimulation of fatty acid oxidation. Proc Natl Acad Sci. 2017;114:E6556–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Han T, Kang D, Ji D, Wang X, Zhan W, Fu M, et al. How does cancer cell metabolism affect tumor migration and invasion? Cell Adhes Migr. 2013;7:395–403.

    Article  Google Scholar 

  34. Christiansen JJ, Rajasekaran AK. Reassessing epithelial to mesenchymal transition as a prerequisite for carcinoma invasion and metastasis. Cancer Res. 2006;66:8319–26.

    Article  CAS  PubMed  Google Scholar 

  35. Gumbiner BM. Regulation of cadherin-mediated adhesion in morphogenesis. Nat Rev Mol cell Biol. 2005;6:622–34.

    Article  CAS  PubMed  Google Scholar 

  36. Kota S, Hou S, Guerrant W, Madoux F, Troutman S, Fernandez-Vega V, et al. A novel three-dimensional high-throughput screening approach identifies inducers of a mutant KRAS selective lethal phenotype. Oncogene. 2018;37:4372–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Keung MY, Wu Y, Badar F, Vadgama JV. Response of breast cancer cells to PARP inhibitors is independent of BRCA status. J Clin Med. 2020;9:940.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zhao H, Yang Q, Hu Y, Zhang J. Antitumor effects and mechanisms of olaparib in combination with carboplatin and BKM120 on human triple‑negative breast cancer cells. Oncol Rep. 2018;40:3223–34.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Gao H, Korn JM, Ferretti S, Monahan JE, Wang Y, Singh M, et al. High-throughput screening using patient-derived tumor xenografts to predict clinical trial drug response. Nat Med. 2015;21:1318–25.

    Article  CAS  PubMed  Google Scholar 

  40. Momeny M, Yousefi H, Eyvani H, Moghaddaskho F, Salehi A, Esmaeili F, et al. Blockade of nuclear factor-κB (NF-κB) pathway inhibits growth and induces apoptosis in chemoresistant ovarian carcinoma cells. Int J Biochem Cell Biol. 2018;99:1–9.

    Article  CAS  PubMed  Google Scholar 

  41. Chiarugi P, Giannoni E. Anoikis: a necessary death program for anchorage-dependent cells. Biochemical Pharmacol. 2008;76:1352–64.

    Article  CAS  Google Scholar 

  42. Tajbakhsh A, Rivandi M, Abedini S, Pasdar A, Sahebkar A. Regulators and mechanisms of anoikis in triple-negative breast cancer (TNBC): A review. Crit Rev Oncol/Hematol. 2019;140:17–27.

    Article  PubMed  Google Scholar 

  43. Clark AS, Karasic TB, DeMichele A, Vaughn DJ, O’Hara M, Perini R, et al. Palbociclib (PD0332991)—a selective and potent cyclin-dependent kinase inhibitor: a review of pharmacodynamics and clinical development. JAMA Oncol. 2016;2:253–60.

    Article  PubMed  Google Scholar 

  44. Álvarez-Fernández M, Malumbres M. Mechanisms of sensitivity and resistance to CDK4/6 inhibition. Cancer Cell. 2020;37:514–29.

    Article  PubMed  Google Scholar 

  45. Tadesse S, Caldon EC, Tilley W, Wang S. Cyclin-dependent kinase 2 inhibitors in cancer therapy: an update. J Medicinal Chem. 2018;62:4233–51.

    Article  Google Scholar 

  46. Buckley MF, Sweeney K, Hamilton J, Sini R, Manning D, Nicholson R, et al. Expression and amplification of cyclin genes in human breast cancer. Oncogene. 1993;8:2127–33.

    CAS  PubMed  Google Scholar 

  47. Gillett C, Fantl V, Smith R, Fisher C, Bartek J, Dickson C, et al. Amplification and overexpression of cyclin D1 in breast cancer detected by immunohistochemical staining. Cancer Res. 1994;54:1812–7.

    CAS  PubMed  Google Scholar 

  48. Androic I, Krämer A, Yan R, Rödel F, Gätje R, Kaufmann M, et al. Targeting cyclin B1 inhibits proliferation and sensitizes breast cancer cells to taxol. BMC Cancer. 2008;8:1–11.

    Article  Google Scholar 

  49. Chaitanya GV, Steven AJ, Babu PP. PARP-1 cleavage fragments: signatures of cell-death proteases in neurodegeneration. Cell Commun Signal: CCS. 2010;8:31–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Goodwin CM, Rossanese OW, Olejniczak ET, Fesik SW. Myeloid cell leukemia-1 is an important apoptotic survival factor in triple-negative breast cancer. Cell Death Differ. 2015;22:2098–106.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Olopade OI, Adeyanju MO, Safa AR, Hagos F, Mick R, Thompson CB, et al. Overexpression of BCL-x protein in primary breast cancer is associated with high tumor grade and nodal metastases. Cancer J Sci Am. 1997;3:230–7.

    CAS  PubMed  Google Scholar 

  52. Youle RJ, Strasser A. The BCL-2 protein family: opposing activities that mediate cell death. Nat Rev Mol Cell Biol. 2008;9:47–59.

    Article  CAS  PubMed  Google Scholar 

  53. Lopez J, Tait S. Mitochondrial apoptosis: killing cancer using the enemy within. Br J Cancer. 2015;112:957–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Yu J, Zhang L. PUMA, a potent killer with or without p53. Oncogene. 2008;27:S71–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Xu J, Chen Y, Olopade OI. MYC and breast cancer. Genes Cancer. 2010;1:629–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Deveraux QL, Reed JC. IAP family proteins—suppressors of apoptosis. Genes Dev. 1999;13:239–52.

    Article  CAS  PubMed  Google Scholar 

  57. Tamm I, Kornblau SM, Segall H, Krajewski S, Welsh K, Kitada S, et al. Expression and prognostic significance of IAP-family genes in human cancers and myeloid leukemias. Clin Cancer Res. 2000;6:1796–803.

    CAS  PubMed  Google Scholar 

  58. Flygare JA, Beresini M, Budha N, Chan H, Chan IT, Cheeti S, et al. Discovery of a potent small-molecule antagonist of inhibitor of apoptosis (IAP) proteins and clinical candidate for the treatment of cancer (GDC-0152). J Medicinal Chem. 2012;55:4101–13.

    Article  CAS  Google Scholar 

  59. Moriai R, Tsuji N, Moriai M, Kobayashi D, Watanabe N. Survivin plays as a resistant factor against tamoxifen-induced apoptosis in human breast cancer cells. Breast Cancer Res Treat. 2009;117:261–71.

    Article  CAS  PubMed  Google Scholar 

  60. Nestal de Moraes G, Delbue D, Silva KL, Robaina MC, Khongkow P, Gomes AR, et al. FOXM1 targets XIAP and Survivin to modulate breast cancer survival and chemoresistance. Cell Signal. 2015;27:2496–505.

    Article  CAS  PubMed  Google Scholar 

  61. Span PN, Sweep FC, Wiegerinck ET, Tjan-Heijnen VC, Manders P, Beex LV, et al. Survivin is an independent prognostic marker for risk stratification of breast cancer patients. Clin Chem. 2004;50:1986–93.

    Article  CAS  PubMed  Google Scholar 

  62. Zhang Y, Zhu J, Tang Y, Li F, Zhou H, Peng B, et al. X-linked inhibitor of apoptosis positive nuclear labeling: a new independent prognostic biomarker of breast invasive ductal carcinoma. Diagnostic Pathol. 2011;6:1–9.

    Article  Google Scholar 

  63. Brabletz T, Kalluri R, Nieto MA, Weinberg RA. EMT in cancer. Nat Rev Cancer. 2018;18:128–34.

    Article  CAS  PubMed  Google Scholar 

  64. Ribatti D, Tamma R, Annese T. Epithelial-mesenchymal transition in cancer: a historical overview. Transl Oncol. 2020;13:100773.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Bonnomet A, Syne L, Brysse A, Feyereisen E, Thompson E, Noël A, et al. A dynamic in vivo model of epithelial-to-mesenchymal transitions in circulating tumor cells and metastases of breast cancer. Oncogene. 2012;31:3741–53.

    Article  CAS  PubMed  Google Scholar 

  66. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci. 2005;102:15545–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Perou CM, Sørlie T, Eisen MB, Van De Rijn M, Jeffrey SS, Rees CA, et al. Molecular portraits of human breast tumours. Nature. 2000;406:747–52.

    Article  CAS  PubMed  Google Scholar 

  68. Hussein YR, Bandyopadhyay S, Semaan A, Ahmed Q, Albashiti B, Jazaerly T, et al. Glut-1 expression correlates with basal-like breast cancer. Transl Oncol. 2011;4:321.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Oh S, Kim H, Nam K, Shin I. Glut1 promotes cell proliferation, migration and invasion by regulating epidermal growth factor receptor and integrin signaling in triple-negative breast cancer cells. BMB Rep. 2017;50:132.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Avanzato D, Pupo E, Ducano N, Isella C, Bertalot G, Luise C, et al. High USP6NL levels in breast cancer sustain chronic AKT phosphorylation and GLUT1 stability fueling aerobic glycolysis. Cancer Res. 2018;78:3432–44.

    Article  CAS  PubMed  Google Scholar 

  71. Dang CV, Le A, Gao P. MYC-induced cancer cell energy metabolism and therapeutic opportunities. Clin Cancer Res. 2009;15:6479–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Network CGA. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490:61.

    Article  Google Scholar 

  73. Huang X, Li X, Xie X, Ye F, Chen B, Song C, et al. High expressions of LDHA and AMPK as prognostic biomarkers for breast cancer. Breast. 2016;30:39–46.

    Article  PubMed  Google Scholar 

  74. Jin XJ, Chen XJ, Zhang ZF, Hu WS, Ou RY, Li S, et al. Long noncoding RNA SNHG12 promotes the progression of cervical cancer via modulating miR‐125b/STAT3 axis. J Cell Physiol. 2019;234:6624–32.

    Article  CAS  PubMed  Google Scholar 

  75. Masson N, Ratcliffe PJ. Hypoxia signaling pathways in cancer metabolism: the importance of co-selecting interconnected physiological pathways. Cancer Metab. 2014;2:1–17.

    Article  Google Scholar 

  76. Pinheiro C, Albergaria A, Paredes J, Sousa B, Dufloth R, Vieira D, et al. Monocarboxylate transporter 1 is up‐regulated in basal‐like breast carcinoma. Histopathology. 2010;56:860–7.

    Article  PubMed  Google Scholar 

  77. Andrzejewski S, Klimcakova E, Johnson RM, Tabariès S, Annis MG, McGuirk S, et al. PGC-1α promotes breast cancer metastasis and confers bioenergetic flexibility against metabolic drugs. Cell Metab. 2017;26:778–87.e775.

    Article  CAS  PubMed  Google Scholar 

  78. Shen L, O’Shea JM, Kaadige MR, Cunha S, Wilde BR, Cohen AL, et al. Metabolic reprogramming in triple-negative breast cancer through Myc suppression of TXNIP. Proc Natl Acad Sci. 2015;112:5425–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Curtis C, Shah SP, Chin S-F, Turashvili G, Rueda OM, Dunning MJ, et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 2012;486:346–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Garcia D, Shaw RJ. AMPK: mechanisms of cellular energy sensing and restoration of metabolic balance. Mol Cell. 2017;66:789–800.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Hardie DG. AMP-activated protein kinase: an energy sensor that regulates all aspects of cell function. Genes Dev. 2011;25:1895–908.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Longley D, Johnston P. Molecular mechanisms of drug resistance. J Pathol: A J Pathological Soc Gt Br Irel. 2005;205:275–92.

    Article  CAS  Google Scholar 

  83. Zhou M, Zhao Y, Ding Y, Liu H, Liu Z, Fodstad O, et al. Warburg effect in chemosensitivity: targeting lactate dehydrogenase-A re-sensitizes taxol-resistant cancer cells to taxol. Mol Cancer. 2010;9:1–12.

    Article  Google Scholar 

  84. Barrett-Lee P, Dixon J, Farrell C, Jones A, Leonard R, Murray N, et al. Expert opinion on the use of anthracyclines in patients with advanced breast cancer at cardiac risk. Ann Oncol. 2009;20:816–27.

    Article  CAS  PubMed  Google Scholar 

  85. Andre F, Zielinski C. Optimal strategies for the treatment of metastatic triple-negative breast cancer with currently approved agents. Ann Oncol. 2012;23:vi46–51.

    Article  PubMed  Google Scholar 

  86. Christowitz C, Davis T, Isaacs A, van Niekerk G, Hattingh S, Engelbrecht A-M. Mechanisms of doxorubicin-induced drug resistance and drug resistant tumour growth in a murine breast tumour model. BMC Cancer. 2019;19:757.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Bianchini G, De Angelis C, Licata L, Gianni L. Treatment landscape of triple-negative breast cancer—expanded options, evolving needs. Nat Rev Clin Oncol. 2021;19:1–23.

    Google Scholar 

  88. Lev S. Targeted therapy and drug resistance in triple-negative breast cancer: the EGFR axis. Biochemical Soc Trans. 2020;48:657–65.

    Article  CAS  Google Scholar 

  89. Won KA, Spruck C. Triple‑negative breast cancer therapy: Current and future perspectives. Int J Oncol. 2020;57:1245–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. 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;13:4429–34.

    Article  PubMed  Google Scholar 

  91. Lin NU, Claus E, Sohl J, Razzak AR, Arnaout A, Winer EP. Sites of distant recurrence and clinical outcomes in patients with metastatic triple‐negative breast cancer: high incidence of central nervous system metastases. Cancer. 2008;113:2638–45.

    Article  PubMed  Google Scholar 

  92. Yin L, Duan J-J, Bian X-W, Yu S-C. Triple-negative breast cancer molecular subtyping and treatment progress. Breast Cancer Res. 2020;22:1–13.

    Article  Google Scholar 

  93. Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial–mesenchymal transition. Nat Rev Mol Cell Biol. 2014;15:178–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Prasad CP, Rath G, Mathur S, Bhatnagar D, Parshad R, Ralhan R. Expression analysis of E-cadherin, Slug and GSK3β in invasive ductal carcinoma of breast. BMC Cancer. 2009;9:1–10.

    Article  Google Scholar 

  95. Hennig G, Behrens J, Truss M, Frisch S, Reichmann E, Birchmeier W. Progression of carcinoma cells is associated with alterations in chromatin structure and factor binding at the E-cadherin promoter in vivo. Oncogene. 1995;11:475–84.

    CAS  PubMed  Google Scholar 

  96. Cano A, Pérez-Moreno MA, Rodrigo I, Locascio A, Blanco MJ, del Barrio MG, et al. The transcription factor snail controls epithelial–mesenchymal transitions by repressing E-cadherin expression. Nat Cell Biol. 2000;2:76–83.

    Article  CAS  PubMed  Google Scholar 

  97. Hajra KM, Chen DY, Fearon ER. The SLUG zinc-finger protein represses E-cadherin in breast cancer. Cancer Res. 2002;62:1613–8.

    CAS  PubMed  Google Scholar 

  98. Eger A, Aigner K, Sonderegger S, Dampier B, Oehler S, Schreiber M, et al. DeltaEF1 is a transcriptional repressor of E-cadherin and regulates epithelial plasticity in breast cancer cells. Oncogene. 2005;24:2375–85.

    Article  CAS  PubMed  Google Scholar 

  99. Grooteclaes ML, Frisch SM. Evidence for a function of CtBP in epithelial gene regulation and anoikis. Oncogene. 2000;19:3823–8.

    Article  CAS  PubMed  Google Scholar 

  100. Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139:871–90.

    Article  CAS  PubMed  Google Scholar 

  101. Karihtala P, Auvinen P, Kauppila S, Haapasaari K-M, Jukkola-Vuorinen A, Soini Y. Vimentin, zeb1 and Sip1 are up-regulated in triple-negative and basal-like breast cancers: association with an aggressive tumour phenotype. Breast Cancer Res Treat. 2013;138:81–90.

    Article  CAS  PubMed  Google Scholar 

  102. Ferrari-Amorotti G, Chiodoni C, Shen F, Cattelani S, Soliera AR, Manzotti G, et al. Suppression of invasion and metastasis of triple-negative breast cancer lines by pharmacological or genetic inhibition of slug activity. Neoplasia. 2014;16:1047–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Shih J-Y, Tsai M-F, Chang T-H, Chang Y-L, Yuan A, Yu C-J, et al. Transcription repressor slug promotes carcinoma invasion and predicts outcome of patients with lung adenocarcinoma. Clin Cancer Res. 2005;11:8070–8.

    Article  CAS  PubMed  Google Scholar 

  104. Shioiri M, Shida T, Koda K, Oda K, Seike K, Nishimura M, et al. Slug expression is an independent prognostic parameter for poor survival in colorectal carcinoma patients. Br J Cancer. 2006;94:1816–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Coller HA. Is cancer a metabolic disease? Am J Pathol. 2014;184:4–17.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Wallace DC. Mitochondria and cancer. Nat Rev Cancer. 2012;12:685–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Lunetti P, Di Giacomo M, Vergara D, De Domenico S, Maffia M, Zara V, et al. Metabolic reprogramming in breast cancer results in distinct mitochondrial bioenergetics between luminal and basal subtypes. FEBS J. 2019;286:688–709.

    Article  CAS  PubMed  Google Scholar 

  108. Dupuy F, Tabariès S, Andrzejewski S, Dong Z, Blagih J, Annis MG, et al. PDK1-dependent metabolic reprogramming dictates metastatic potential in breast cancer. Cell Metab. 2015;22:577–89.

    Article  CAS  PubMed  Google Scholar 

  109. Kim S, Kim DH, Jung WH, Koo JS. Metabolic phenotypes in triple-negative breast cancer. Tumour Biol. 2013;34:1699–712.

    Article  CAS  PubMed  Google Scholar 

  110. Urra FA, Muñoz F, Córdova-Delgado M, Ramírez MP, Peña-Ahumada B, Rios M, et al. FR58P1a; a new uncoupler of OXPHOS that inhibits migration in triple-negative breast cancer cells via Sirt1/AMPK/β1-integrin pathway. Sci Rep. 2018;8:13190.

    Article  PubMed  PubMed Central  Google Scholar 

  111. DeBerardinis RJ, Chandel NS. Fundamentals of cancer metabolism. Sci Adv. 2016;2:e1600200.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Vander Heiden MG, DeBerardinis RJ. Understanding the intersections between metabolism and cancer biology. Cell. 2017;168:657–69.

    Article  PubMed Central  Google Scholar 

  113. Avagliano A, Ruocco MR, Aliotta F, Belviso I, Accurso A, Masone S, et al. Mitochondrial flexibility of breast cancers: a growth advantage and a therapeutic opportunity. Cells. 2019;8:401.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Birsoy K, Wang T, Chen WW, Freinkman E, Abu-Remaileh M, Sabatini DM. An essential role of the mitochondrial electron transport chain in cell proliferation is to enable aspartate synthesis. Cell. 2015;162:540–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Lane AN, Fan TW. Regulation of mammalian nucleotide metabolism and biosynthesis. Nucleic Acids Res. 2015;43:2466–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Sullivan LB, Gui DY, Hosios AM, Bush LN, Freinkman E, Vander, et al. Supporting aspartate biosynthesis is an essential function of respiration in proliferating cells. Cell. 2015;162:552–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. O’Neill S, Porter RK, McNamee N, Martinez VG, O’Driscoll L. 2-Deoxy-D-Glucose inhibits aggressive triple-negative breast cancer cells by targeting glycolysis and the cancer stem cell phenotype. Sci Rep. 2019;9:1–11.

    Article  Google Scholar 

  118. Wang L, Chan JY, Zhou X, Cui G, Yan Z, Wang L, et al. A novel agent enhances the chemotherapeutic efficacy of doxorubicin in MCF-7 breast cancer cells. Front Pharm. 2016;7:249.

    Article  Google Scholar 

  119. Marinello PC, Panis C, Silva TNX, Binato R, Abdelhay E, Rodrigues JA, et al. Metformin prevention of doxorubicin resistance in MCF-7 and MDA-MB-231 involves oxidative stress generation and modulation of cell adaptation genes. Sci Rep. 2019;9:5864.

    Article  PubMed  PubMed Central  Google Scholar 

  120. Andrzejewski S, Siegel PM, St-Pierre J. Metabolic profiles associated with metformin efficacy in cancer. Front Endocrinol. 2018;9:372–372.

    Article  Google Scholar 

  121. Lee K-M, Giltnane JM, Balko JM, Schwarz LJ, Guerrero-Zotano AL, Hutchinson KE, et al. MYC and MCL1 cooperatively promote chemotherapy-resistant breast cancer stem cells via regulation of mitochondrial oxidative phosphorylation. Cell Metab. 2017;26:633–47.e637.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Franken NA, Rodermond HM, Stap J, Haveman J, Van Bree C. Clonogenic assay of cells in vitro. Nat Protoc. 2006;1:2315–9.

    Article  CAS  PubMed  Google Scholar 

  123. Sher I, Adham SA, Petrik J, Coomber BL. Autocrine VEGF‐A/KDR loop protects epithelial ovarian carcinoma cells from anoikis. Int J Cancer. 2009;124:553–61.

    Article  CAS  PubMed  Google Scholar 

  124. Spinazzi M, Casarin A, Pertegato V, Salviati L, Angelini C. Assessment of mitochondrial respiratory chain enzymatic activities on tissues and cultured cells. Nat Protoc. 2012;7:1235–46.

    Article  CAS  PubMed  Google Scholar 

  125. Paredes J, Zabaleta J, Garai J, Ji P, Imtiaz S, Spagnardi M, et al. Immune-related gene expression and cytokine secretion is reduced among African American colon cancer patients. Front Oncol. 2020;10:1498.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Pascale CL, Martinez AN, Carr C, Sawyer DM, Ribeiro-Alves M, Chen M, et al. Treatment with dimethyl fumarate reduces the formation and rupture of intracranial aneurysms: Role of Nrf2 activation. J Cereb Blood Flow Metab. 2019;40:1077–89.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Funding

This work is supported by funds from LSUHSC School of Medicine, Fred G. Brazda foundation, and from NIH grants P20CA202922, P20GM121288-01, P30GM114732 to JZ.

Author information

Authors and Affiliations

Authors

Contributions

Methodology: HY, MK, LL, SO, JGa, MZ, and DWy; Formal analysis: JZ, JGu, DWo and SA; Investigation: JJ, JK, QY, and SA; Original draft preparation: HY, SA; review and editing: SA, DWo, Supervision: SA.

Corresponding author

Correspondence to Suresh K. Alahari.

Ethics declarations

Competing interests

The authors declare no competing interests.

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

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

Yousefi, H., Khosla, M., Lauterboeck, L. et al. A combination of novel NSC small molecule inhibitor along with doxorubicin inhibits proliferation of triple-negative breast cancer through metabolic reprogramming. Oncogene 41, 5076–5091 (2022). https://doi.org/10.1038/s41388-022-02497-2

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-022-02497-2

This article is cited by

Search

Quick links