1. Academic Validation
  2. Characterization of the interactions of potent allosteric inhibitors with glutaminase C, a key enzyme in cancer cell glutamine metabolism

Characterization of the interactions of potent allosteric inhibitors with glutaminase C, a key enzyme in cancer cell glutamine metabolism

  • J Biol Chem. 2018 Mar 9;293(10):3535-3545. doi: 10.1074/jbc.M117.810101.
Qingqiu Huang 1 Clint Stalnecker 2 Chengliang Zhang 3 Lee A McDermott 4 5 Prema Iyer 4 5 Jason O'Neill 4 Shawn Reimer 4 Richard A Cerione 6 2 3 William P Katt 3
Affiliations

Affiliations

  • 1 From the Cornell High Energy Synchrotron Source (CHESS) and.
  • 2 Departments of Chemistry and Chemical Biology and.
  • 3 Molecular Medicine, Cornell University, Ithaca, New York 14853 and.
  • 4 the Department of Pharmaceutical Sciences and.
  • 5 Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261.
  • 6 From the Cornell High Energy Synchrotron Source (CHESS) and [email protected].
Abstract

Altered glycolytic flux in Cancer cells (the "Warburg effect") causes their proliferation to rely upon elevated glutamine metabolism ("glutamine addiction"). This requirement is met by the overexpression of Glutaminase C (GAC), which catalyzes the first step in glutamine metabolism and therefore represents a potential therapeutic target. The small molecule CB-839 was reported to be more potent than other allosteric GAC inhibitors, including the parent compound bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl)ethyl (BPTES), and is in clinical trials. Recently, we described the synthesis of BPTES analogs having distinct saturated heterocyclic cores as a replacement for the flexible chain moiety, with improved microsomal stability relative to CB-839 and BPTES. Here, we show that one of these new compounds, UPGL00004, like CB-839, more potently inhibits the enzymatic activity of GAC, compared with BPTES. We also compare the abilities of UPGL00004, CB-839, and BPTES to directly bind to recombinant GAC and demonstrate that UPGL00004 has a similar binding affinity as CB-839 for GAC. We also show that UPGL00004 potently inhibits the growth of triple-negative breast Cancer cells, as well as tumor growth when combined with the anti-vascular endothelial growth factor antibody bevacizumab. Finally, we compare the X-ray crystal structures for UPGL00004 and CB-839 bound to GAC, verifying that UPGL00004 occupies the same binding site as CB-839 or BPTES and that all three inhibitors regulate the enzymatic activity of GAC via a similar allosteric mechanism. These results provide insights regarding the potency of these inhibitors that will be useful in designing novel small-molecules that target a key Enzyme in Cancer cell metabolism.

Keywords

BPTES; CB-839; anticancer drug; cancer; crystallography; glutaminase; metabolism.

Figures
Products