Abstract

GS-9219 is a cell-permeable prodrug of the acyclic nucleotide analogue 9-(2-phosphonylmethoxyethyl)guanine (PMEG); the incorporation of the active metabolite PMEG diphosphate (PMEGpp) into DNA results in DNA chain termination due to the lack of a 3'-hydroxyl moiety. We hypothesized that the incorporation of PMEGpp into DNA during repair resynthesis would result in the inhibition of DNA repair and the accumulation of DNA breaks in chronic lymphocytic leukemia (CLL) cells that would activate signaling pathways to cell death. To test this hypothesis, CLL cells were irradiated with UV light to stimulate nucleotide excision repair pathways, enabling the incorporation of PMEGpp into DNA. The combination effects of GS-9219 and DNA-damaging agents and the signaling mechanisms activated in response to DNA repair inhibition by GS-9219, as well as changes in CLL cell viability, were investigated. PMEGpp was incorporated into DNA in CLL cells when nucleotide excision repair was activated by UV. Following PMEGpp incorporation, DNA repair was inhibited, which led to the accumulation of DNA strand breaks. The presence of DNA strand breaks activated the phosphatidylinositol 3-kinase-like protein kinase family members ataxia-telangiectasia mutated and DNA-dependent protein kinase. P53 was phosphorylated and stabilized in response to the inhibition of DNA repair. P53 targeted proteins, Puma and Bax, were up-regulated and activated. The combination of GS-9219 and DNA-damaging agents resulted in more cell death than the sum of the single agents alone. GS-9219 inhibits DNA repair in CLL cells, an action that stimulates signaling pathways for apoptosis induction.

Highlights

  • GS-9219 is a cell-permeable prodrug of the acyclic nucleotide analogue 9(2-phosphonylmethoxyethyl)guanine (PMEG); the incorporation of the active metabolite PMEG diphosphate (PMEGpp) into DNA results in DNA chain termination due to the lack of a 3′-hydroxyl moiety

  • The intracellular metabolism of GS-9219 and the concentration of each metabolite were first studied in chronic lymphocytic leukemia (CLL) cells to determine the optimal concentration of GS-9219 for the investigations of DNA repair inhibition

  • Twenty-one CLL cell samples were incubated with 10 μmol/L [14C]GS-9219 for 24 hours and subjected to high-performance liquid chromatography (HPLC) analysis to determine the concentration of each metabolite

Read more

Summary

Introduction

GS-9219 is a cell-permeable prodrug of the acyclic nucleotide analogue 9(2-phosphonylmethoxyethyl)guanine (PMEG); the incorporation of the active metabolite PMEG diphosphate (PMEGpp) into DNA results in DNA chain termination due to the lack of a 3′-hydroxyl moiety. We hypothesized that the incorporation of PMEGpp into DNA during repair resynthesis would result in the inhibition of DNA repair and the accumulation of DNA breaks in chronic lymphocytic leukemia (CLL) cells that would activate signaling pathways to cell death. The primary mechanism of action of GS-9219 in replicating cells is incorporation of its active metabolite, 9-(2-phosphonylmethoxyethyl)guanine diphosphate (PMEGpp), into DNA and subsequent chain termination due to the lack of a 3′-hydroxy moiety. The accumulation of DNA breaks and the inhibition of DNA repair would subsequently activate signaling pathways to cell death. The double-prodrug design of GS-9219 increased the accumulation of PMEGpp in lymphoid cells and tissues and decreased the systematic toxicity generated from PMEG [13]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call