American Association for Cancer Research
Browse
15357163mct170407-sup-182946_2_supp_4322943_3xmg3b.xlsx (55.9 kB)

Table S3 from Metabolite Profiling Reveals the Glutathione Biosynthetic Pathway as a Therapeutic Target in Triple-Negative Breast Cancer

Download (55.9 kB)
dataset
posted on 2023-04-03, 15:01 authored by Alexander Beatty, Lauren S. Fink, Tanu Singh, Alexander Strigun, Erik Peter, Christina M. Ferrer, Emmanuelle Nicolas, Kathy Q. Cai, Timothy P. Moran, Mauricio J. Reginato, Ulrike Rennefahrt, Jeffrey R. Peterson

Table S3. Comparison of the levels of 155 intracellular metabolites across the three metabolic subtypes, related to Figure 1, included as a separate Excel file. The table is ordered according to ontology classes 1 and 2 of the measured metabolite. Explanations for the values presented in each of the columns can be found under the "Legend" tab in the spreadsheet.

History

ARTICLE ABSTRACT

Cancer cells can exhibit altered dependency on specific metabolic pathways and targeting these dependencies is a promising therapeutic strategy. Triple-negative breast cancer (TNBC) is an aggressive and genomically heterogeneous subset of breast cancer that is resistant to existing targeted therapies. To identify metabolic pathway dependencies in TNBC, we first conducted mass spectrometry–based metabolomics of TNBC and control cells. Relative levels of intracellular metabolites distinguished TNBC from nontransformed breast epithelia and revealed two metabolic subtypes within TNBC that correlate with markers of basal-like versus non-basal–like status. Among the distinguishing metabolites, levels of the cellular redox buffer glutathione were lower in TNBC cell lines compared to controls and markedly lower in non-basal–like TNBC. Significantly, these cell lines showed enhanced sensitivity to pharmacologic inhibition of glutathione biosynthesis that was rescued by N-acetylcysteine, demonstrating a dependence on glutathione production to suppress ROS and support tumor cell survival. Consistent with this, patients whose tumors express elevated levels of γ-glutamylcysteine ligase, the rate-limiting enzyme in glutathione biosynthesis, had significantly poorer survival. We find, further, that agents that limit the availability of glutathione precursors enhance both glutathione depletion and TNBC cell killing by γ-glutamylcysteine ligase inhibitors in vitro. Importantly, we demonstrate the ability to this approach to suppress glutathione levels and TNBC xenograft growth in vivo. Overall, these findings support the potential of targeting the glutathione biosynthetic pathway as a therapeutic strategy in TNBC and identify the non-basal-like subset as most likely to respond. Mol Cancer Ther; 17(1); 264–75. ©2017 AACR.

Usage metrics

    Molecular Cancer Therapeutics

    Licence

    Exports

    RefWorks
    BibTeX
    Ref. manager
    Endnote
    DataCite
    NLM
    DC