Archives

  • 2018-07
  • 2018-10
  • 2018-11
  • 2019-04
  • 2019-05
  • 2019-06
  • 2019-07
  • 2019-08
  • 2019-09
  • 2019-10
  • 2019-11
  • 2019-12
  • 2020-01
  • 2020-02
  • 2020-03
  • 2020-04
  • 2020-05
  • 2020-06
  • 2020-07
  • 2020-08
  • 2020-09
  • 2020-10
  • 2020-11
  • 2020-12
  • 2021-01
  • 2021-02
  • 2021-03
  • 2021-04
  • 2021-05
  • 2021-06
  • 2021-07
  • 2021-08
  • 2021-09
  • 2021-10
  • 2021-11
  • 2021-12
  • 2022-01
  • 2022-02
  • 2022-03
  • 2022-04
  • 2022-05
  • 2022-06
  • 2022-07
  • 2022-08
  • 2022-09
  • 2022-10
  • 2022-11
  • 2022-12
  • 2023-01
  • 2023-02
  • 2023-03
  • 2023-04
  • 2023-05
  • 2023-06
  • 2023-08
  • 2023-09
  • 2023-10
  • 2023-11
  • 2023-12
  • 2024-01
  • 2024-02
  • 2024-03
  • We also demonstrated that inhibition of NAAG hydrolysis

    2022-08-12

    We also demonstrated that inhibition of NAAG hydrolysis to suppress glutamate production through a GCPII inhibitor is a viable target for cancer therapy. GCPII is also known as N-acetyl-L-aspartyl-L-glutamate peptidase I (NAALADase I) or NAAG peptidase (Pinto et al., 1996), and its increased expression was found in poorly differentiated, metastatic, and hormone-refractory prostate cancers (Bouchelouche et al., 2010, Ross et al., 2003) or associated with higher tumor grading (G3/G4) of non-small-cell lung cancer (Schmidt et al., 2017). 2-PMPA, a potent and selective inhibitor of GCPII (Jackson et al., 1996), has been found to effectively diminish extracellular glutamate directly by reducing the amount of glutamate released from the NAAG hydrolysis reaction in in vitro and in vivo models of stroke (Slusher et al., 1999, Tsukamoto et al., 2005). Although there now exist numerous inhibitors with various forms and efficacies, 2-PMPA is the first selective GCPII inhibitor, as well as the one with the greatest binding affinity (Zhou et al., 2005). Hence, we selected 2-PMPA for this study, not only because it has the greatest binding affinity but also because it is the most extensively used inhibitor and has been shown to be effective in several in vivo models (Baslow et al., 2005, Harada et al., 2000, Zhou et al., 2005). Taking advantage of the availability of 2-PMPA, our study demonstrated the efficacy of this inhibitor in diminishing tumor growth of patient-derived, recurrent OVCA and pancreatic cancer orthotopic tumors. Importantly, in the pancreatic cancer model expressing both GCPII and glutaminase, the combination of 2-PMPA and CB-839, a glutaminase inhibitor currently undergoing clinical trials (Harding et al., 2015), results in a significant decrease in tumor weights as compared to either treatment alone. Thus, these findings suggest that the GCPII inhibitor could be valuable in developing anticancer agents. However, not all cancer RK-33 receptor express GCPII (even within the same cancer type). Our findings of the role of NAAG as a glutamate provider are only applicable to GCPII-positive cancers. For the GCPII-negative cancers, we suspect that NAAG could be taking on another role to promote cell growth through an alternative pathway in these GCPII-negative cells, thereby precluding the role of NAAG as a glutamate provider. NAAG is known to activate the metabotropic glutamate receptor (mGlu3) (Fricker et al., 2009, Wroblewska et al., 1997, Wroblewska et al., 1998), and previous studies have shown that mGlu3 expression is associated with glioma growth and poor GBM survival rates (Arcella et al., 2005, Aronica et al., 2003, Brocke et al., 2010, Ciceroni et al., 2008, Ciceroni et al., 2013, D’Onofrio et al., 2003, Stepulak et al., 2009, Zhou et al., 2014). The alternative pathway could be achieved through NAAG acting as an agonist to mGlu3 receptors. We have not deeply explored exactly how NAAG promotes cancer cell growth through this pathway, which will be the focus of our future work. In conclusion, the neurotransmitter NAAG was found to be elevated in higher grade cancers and strongly mirrored tumor sizes in vivo, making NAAG a potential metabolite monitor for cancer progression, whereby NAAG concentration spikes can be detected in advance of any surges in tumor growth. Importantly, we uncovered that NAAG serves as an important reservoir to provide glutamate to cancer cells through GCPII when glutamate production from other sources is limited. GCPII was thus identified as a viable target for cancer therapy, either alone or in combination with glutaminase inhibition. Together, these results exhibit the multi-faceted roles of NAAG in cancer, which can be exploited to improve patients’ clinical outcomes.
    STAR★Methods
    Acknowledgments This work was supported by the NIH grants R01-CA193895, R01-CA112314, 1S10OD025226-01, and UL1 TR 001079; Hopkins-Allegheny Health Network Cancer Research Fund; and the Doris M. Weinstein Pancreatic Cancer Research Fund (to A.L.). Special thanks to Michel Soudée for his helpful editing.