The global health burden of annual influenza epidemics coupled with emerging influenza pandemics highlight the urgent need for new, effective treatments. The majority of current circulating seasonal influenza viruses, as well as highly pathogenic avian influenza viruses such as H5N1 and H7N9, carry the S31N mutant in their M2 genes, which confers resistance to adamantanes (amantadine and rimantadine). Influenza viruses that are resistant to oseltamivir, the only orally available drug, are continuousy on the rise. Thus there is a persistent need for novel anti-influenza drugs. The M2-S31N mutation is persistent in more than 95% of currently circulating influenza A viruses, and it is one of the most conserved viral proteins. Therefore inhibitors targeting M2-S31N will be excellent candidates for the next generation of anti-influenza drugs. This proposal focuses on developing M2-S31N channel blockers with favorable drug-like properties and in vivo activities. We implement a multi-faceted approach that includes structure-based drug design, medicinal chemistry, electrophysiology, and virology to design M2-S31N channel blockers. The goal is to develop broad spectrum antiviral drugs that are active against clinical isolates of influenza A viruses that are resistant to one or both classes of currently approved anti-influenza drugs. In the R21 phase we will focus on optimizing our previously discovered M2-S31N inhibitors as well as exploring novel chemotypes. The inhibitor design will be guided by our previously solved solution NMR structure of M2-S31N. Synthesized molecules will be tested in antiviral assays against clinically relevant influenza A viruses that show resistance to either amantadine or oseltamivir or both. The mechanism of action will be confirmed using electrophysiological assays and solution NMR NOESY spectroscopy. To prioritize compounds for the in vivo studies, we will profile the ADME properties of top lead compounds as well as evaluate their tendencies to elicit drug resistance. In the R33 phase, we will evaluate the combination therapy potential of M2-S31N inhibitors with oseltamivir or ribavirin. The structure-activity relationship studies will e driven by the in vitro ADME properties, cytotoxicity, and synergistic effects with oseltamivir or ribavirin. We will assay the in vivo pharmacokinetic ADME properties of up to five compounds in mice. For efficacy studies, mice will be challenged with a lethal dose of mouse-adapted influenza A viruses, and M2-S31N inhibitors will be administrated by oral gavage or tail vein injection. Survival rate, body weight loss, and other clinical signs will be used to evaluate the i vivo efficacy of M2-S31N inhibitors. Upon successful completion of the proposed studies, we will have at least one compound that is ready for the next stage of studies needed for Investigational New Drug (IND) filling.

Public Health Relevance

The limited efficacy of currently approved antiviral drugs in combating emerging drug-resistant influenza A viruses highlights the immediate need of novel anti-influenza drugs. This application focuses on one of the most conserved viral proteins found in drug-resistant influenza A virus strains, M2-S31N, and seeks developing M2-S31N channel blockers to target the clinical isolates of human influenza A viruses that show resistance to one or both classes of currently approved antiviral drugs. These inhibitors represent the first-in-clas channel blockers that are active against drug-resistant M2 mutants.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Exploratory/Developmental Grants Phase II (R33)
Project #
4R33AI119187-03
Application #
9488852
Study Section
Special Emphasis Panel (ZAI1)
Program Officer
Krafft, Amy
Project Start
2015-07-06
Project End
2020-06-30
Budget Start
2017-07-06
Budget End
2018-06-30
Support Year
3
Fiscal Year
2017
Total Cost
Indirect Cost
Name
University of Arizona
Department
Type
Schools of Pharmacy
DUNS #
806345617
City
Tucson
State
AZ
Country
United States
Zip Code
85721
Drakopoulos, Antonios; Tzitzoglaki, Christina; McGuire, Kelly et al. (2018) Unraveling the Binding, Proton Blockage, and Inhibition of Influenza M2 WT and S31N by Rimantadine Variants. ACS Med Chem Lett 9:198-203
Zhang, Jiantao; Hu, Yanmei; Foley, Christopher et al. (2018) Exploring Ugi-Azide Four-Component Reaction Products for Broad-Spectrum Influenza Antivirals with a High Genetic Barrier to Drug Resistance. Sci Rep 8:4653
Ma, Chunlong; Wang, Jun (2018) Functional studies reveal the similarities and differences between AM2 and BM2 proton channels from influenza viruses. Biochim Biophys Acta Biomembr 1860:272-280
Musharrafieh, Rami; Ma, Chunlong; Wang, Jun (2018) Profiling the in vitro drug-resistance mechanism of influenza A viruses towards the AM2-S31N proton channel blockers. Antiviral Res 153:10-22
Williams, Jonathan K; Shcherbakov, Alexander A; Wang, Jun et al. (2017) Protonation equilibria and pore-opening structure of the dual-histidine influenza B virus M2 transmembrane proton channel from solid-state NMR. J Biol Chem 292:17876-17884
Hu, Yanmei; Zhang, Jiantao; Musharrafieh, Rami et al. (2017) Chemical Genomics Approach Leads to the Identification of Hesperadin, an Aurora B Kinase Inhibitor, as a Broad-Spectrum Influenza Antiviral. Int J Mol Sci 18:
Hu, Yanmei; Sneyd, Hannah; Dekant, Raphael et al. (2017) Influenza A Virus Nucleoprotein: A Highly Conserved Multi-Functional Viral Protein as a Hot Antiviral Drug Target. Curr Top Med Chem 17:2271-2285
Drakopoulos, Antonios; Tzitzoglaki, Christina; Ma, Chulong et al. (2017) Affinity of Rimantadine Enantiomers against Influenza A/M2 Protein Revisited. ACS Med Chem Lett 8:145-150
Barniol-Xicota, Marta; Gazzarrini, Sabrina; Torres, Eva et al. (2017) Slow but Steady Wins the Race: Dissimilarities among New Dual Inhibitors of the Wild-Type and the V27A Mutant M2 Channels of Influenza A Virus. J Med Chem 60:3727-3738
Hu, Yanmei; Musharrafieh, Rami; Ma, Chunlong et al. (2017) An M2-V27A channel blocker demonstrates potent in vitro and in vivo antiviral activities against amantadine-sensitive and -resistant influenza A viruses. Antiviral Res 140:45-54

Showing the most recent 10 out of 16 publications