The herein proposed Center of Excellence for Translational Research (CETR), which will be named the Antiviral Drug Discovery and Development Center (AD3C) has, at its center, the theme to develop new small molecule therapeutics for emerging and re-emerging viral infections. Translational research will focus on the inhibition of viral replication, especially viral polymerases. Progression pathways will includ target validation, high throughput screening to identify novel chemical scaffolds, and basic virologic research to prove and further probe the exact mechanism of action of identified lead molecules in viral replication. Medicinal chemistry and lead development activities will advance identified compounds down the drug discovery and development pathway, ultimately leading to preclinical evaluation of promising drug candidates for clinical evaluation. The AD3C has four interrelated research projects with a focus on flaviviruses, coronaviruses, alphaviruses and influenza. They will be supported in their efforts by three cores: an Administrative Core (Core A), a Screening Core (SC;Core B) and a Medicinal Chemistry and Lead Development Core (MCLDC;Core C). Organization and interaction between all projects and cores will be mediated out of the Administrative Core, with day to day AD3C operations led by the Core's personnel. An Executive Committee (EC) consisting of all project and core leaders will regularly review the projects'and cores'activities and productivity. Finally, an External Scientific Advisory Board (EAB) of distinguished scientists has been established to annually provide a high level evaluation of AD3C's progress and successes, and aid in refining the Center's activities and direction.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Program--Cooperative Agreements (U19)
Project #
1U19AI109680-01
Application #
8641766
Study Section
Special Emphasis Panel (ZAI1-LR-M (J1))
Program Officer
Beanan, Maureen J
Project Start
2014-03-01
Project End
2019-02-28
Budget Start
2014-03-01
Budget End
2015-02-28
Support Year
1
Fiscal Year
2014
Total Cost
$6,270,702
Indirect Cost
$238,071
Name
University of Alabama Birmingham
Department
Pediatrics
Type
Schools of Medicine
DUNS #
063690705
City
Birmingham
State
AL
Country
United States
Zip Code
35294
Carpentier, Kathryn S; Morrison, Thomas E (2018) Innate immune control of alphavirus infection. Curr Opin Virol 28:53-60
McCarthy, Mary K; Davenport, Bennett J; Reynoso, Glennys V et al. (2018) Chikungunya virus impairs draining lymph node function by inhibiting HEV-mediated lymphocyte recruitment. JCI Insight 3:
Shin, Jin Soo; Jung, Eunhye; Kim, Meehyein et al. (2018) Saracatinib Inhibits Middle East Respiratory Syndrome-Coronavirus Replication In Vitro. Viruses 10:
Johnson, Britney; VanBlargan, Laura A; Xu, Wei et al. (2018) Human IFIT3 Modulates IFIT1 RNA Binding Specificity and Protein Stability. Immunity 48:487-499.e5
Agostini, Maria L; Andres, Erica L; Sims, Amy C et al. (2018) Coronavirus Susceptibility to the Antiviral Remdesivir (GS-5734) Is Mediated by the Viral Polymerase and the Proofreading Exoribonuclease. MBio 9:
Pryke, Kara M; Abraham, Jinu; Sali, Tina M et al. (2017) A Novel Agonist of the TRIF Pathway Induces a Cellular State Refractory to Replication of Zika, Chikungunya, and Dengue Viruses. MBio 8:
McCarthy, Mary K; Morrison, Thomas E (2017) Persistent RNA virus infections: do PAMPS drive chronic disease? Curr Opin Virol 23:8-15
Sheahan, Timothy P; Sims, Amy C; Graham, Rachel L et al. (2017) Broad-spectrum antiviral GS-5734 inhibits both epidemic and zoonotic coronaviruses. Sci Transl Med 9:
Jones, Jennifer E; Long, Kristin M; Whitmore, Alan C et al. (2017) Disruption of the Opal Stop Codon Attenuates Chikungunya Virus-Induced Arthritis and Pathology. MBio 8:
Haese, Nicole N; Broeckel, Rebecca M; Hawman, David W et al. (2016) Animal Models of Chikungunya Virus Infection and Disease. J Infect Dis 214:S482-S487

Showing the most recent 10 out of 24 publications