Mechanism of action of T-705 ribosyl triphosphate against influenza virus RNA polymerase

H Sangawa, T Komeno, H Nishikawa… - Antimicrobial agents …, 2013 - Am Soc Microbiol
H Sangawa, T Komeno, H Nishikawa, A Yoshida, K Takahashi, N Nomura, Y Furuta
Antimicrobial agents and chemotherapy, 2013Am Soc Microbiol
ABSTRACT T-705 (favipiravir; 6-fluoro-3-hydroxy-2-pyrazinecarboxamide) selectively and
strongly inhibits replication of the influenza virus in vitro and in vivo. T-705 has been shown
to be converted to T-705-4-ribofuranosyl-5-triphosphate (T-705RTP) by intracellular
enzymes and then functions as a nucleotide analog to selectively inhibit RNA-dependent
RNA polymerase (RdRp) of the influenza virus. To elucidate these inhibitory mechanisms,
we analyzed the enzyme kinetics of inhibition using Lineweaver-Burk plots of four natural …
Abstract
T-705 (favipiravir; 6-fluoro-3-hydroxy-2-pyrazinecarboxamide) selectively and strongly inhibits replication of the influenza virus in vitro and in vivo. T-705 has been shown to be converted to T-705-4-ribofuranosyl-5-triphosphate (T-705RTP) by intracellular enzymes and then functions as a nucleotide analog to selectively inhibit RNA-dependent RNA polymerase (RdRp) of the influenza virus. To elucidate these inhibitory mechanisms, we analyzed the enzyme kinetics of inhibition using Lineweaver-Burk plots of four natural nucleoside triphosphates and conducted polyacrylamide gel electrophoresis of the primer extension products initiated from 32P-radiolabeled 5′Cap1 RNA. Enzyme kinetic analysis demonstrated that T-705RTP inhibited the incorporation of ATP and GTP in a competitive manner, which suggests that T-705RTP is recognized as a purine nucleotide by influenza virus RdRp and inhibited the incorporation of UTP and CTP in noncompetitive and mixed-type manners, respectively. Primer extension analysis demonstrated that a single molecule of T-705RTP was incorporated into the nascent RNA strand of the influenza virus and inhibited the subsequent incorporation of nucleotides. These results suggest that a single molecule of T-705RTP is incorporated into the nascent RNA strand as a purine nucleotide analog and inhibits strand extension, even though the natural ribose of T-705RTP has a 3′-OH group, which is essential for forming a covalent bond with the phosphate group.
American Society for Microbiology