Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Nikos Pagratis is active.

Publication


Featured researches published by Nikos Pagratis.


Antimicrobial Agents and Chemotherapy | 2016

In Vitro Antiviral Activity and Resistance Profile Characterization of the Hepatitis C Virus NS5A Inhibitor Ledipasvir

Guofeng Cheng; Yang Tian; Brian Doehle; Betty Peng; Amoreena C. Corsa; Yu-Jen Lee; Ruoyu Gong; Mei Yu; Bin Han; Simin Xu; Hadas Dvory-Sobol; Michel Perron; Yili Xu; Hongmei Mo; Nikos Pagratis; John O. Link; William E. Delaney

ABSTRACT Ledipasvir (LDV; GS-5885), a component of Harvoni (a fixed-dose combination of LDV with sofosbuvir [SOF]), is approved to treat chronic hepatitis C virus (HCV) infection. Here, we report key preclinical antiviral properties of LDV, including in vitro potency, in vitro resistance profile, and activity in combination with other anti-HCV agents. LDV has picomolar antiviral activity against genotype 1a and genotype 1b replicons with 50% effective concentration (EC50) values of 0.031 nM and 0.004 nM, respectively. LDV is also active against HCV genotypes 4a, 4d, 5a, and 6a with EC50 values of 0.11 to 1.1 nM. LDV has relatively less in vitro antiviral activity against genotypes 2a, 2b, 3a, and 6e, with EC50 values of 16 to 530 nM. In vitro resistance selection with LDV identified the single Y93H and Q30E resistance-associated variants (RAVs) in the NS5A gene; these RAVs were also observed in patients after a 3-day monotherapy treatment. In vitro antiviral combination studies indicate that LDV has additive to moderately synergistic antiviral activity when combined with other classes of HCV direct-acting antiviral (DAA) agents, including NS3/4A protease inhibitors and the nucleotide NS5B polymerase inhibitor SOF. Furthermore, LDV is active against known NS3 protease and NS5B polymerase inhibitor RAVs with EC50 values equivalent to those for the wild type.


Journal of Medicinal Chemistry | 2010

Identification of Orally Available Naphthyridine Protein Kinase D Inhibitors

Erik Meredith; Ophelia Ardayfio; Kimberly Beattie; Markus Dobler; Istvan J. Enyedy; Christoph Gaul; Vinayak Hosagrahara; Charles Jewell; Keith A. Koch; Wendy Lee; Hansjoerg Lehmann; Timothy A. McKinsey; Karl Miranda; Nikos Pagratis; Margaret R. Pancost; Anup Patnaik; Dillon Phan; Craig F. Plato; Ming Qian; Vasumathy Rajaraman; Chang Rao; Olga Rozhitskaya; Thomas Ruppen; Jie Shi; Sarah Siska; Clayton Springer; Maurice J. van Eis; Richard B. Vega; Anette Von Matt; Lihua Yang

A novel 2,6-naphthyridine was identified by high throughput screen (HTS) as a dual protein kinase C/D (PKC/PKD) inhibitor. PKD inhibition in the heart was proposed as a potential antihypertrophic mechanism with application as a heart failure therapy. As PKC was previously identified as the immediate upstream activator of PKD, PKD vs PKC selectivity was essential to understand the effect of PKD inhibition in models of cardiac hypertrophy and heart failure. The present study describes the modification of the HTS hit to a series of prototype pan-PKD inhibitors with routine 1000-fold PKD vs PKC selectivity. Example compounds inhibited PKD activity in vitro, in cells, and in vivo following oral administration. Their effects on heart morphology and function are discussed herein.


Journal of Medicinal Chemistry | 2010

Identification of Potent and Selective Amidobipyridyl Inhibitors of Protein Kinase D

Erik Meredith; Kimberly Beattie; Robin Burgis; Michael Paul Capparelli; Joseph A. Chapo; Lucian DiPietro; Gabriel G. Gamber; Istvan J. Enyedy; David B. Hood; Vinayak Hosagrahara; Charles Jewell; Keith A. Koch; Wendy Lee; Douglas D. Lemon; Timothy A. McKinsey; Karl Miranda; Nikos Pagratis; Dillon Phan; Craig F. Plato; Chang Rao; Olga Rozhitskaya; Nicolas Soldermann; Clayton Springer; Maurice J. van Eis; Richard B. Vega; Wanlin Yan; Qingming Zhu; Lauren G. Monovich

The synthesis and biological evaluation of potent and selective PKD inhibitors are described herein. The compounds described in the present study selectively inhibit PKD among other putative HDAC kinases. The PKD inhibitors of the present study blunt phosphorylation and subsequent nuclear export of HDAC4/5 in response to diverse agonists. These compounds further establish the central role of PKD as an HDAC4/5 kinase and enhance the current understanding of cardiac myocyte signal transduction. The in vivo efficacy of a representative example compound on heart morphology is reported herein.


Biochimica et Biophysica Acta | 2009

Suppression of HDAC nuclear export and cardiomyocyte hypertrophy by novel irreversible inhibitors of CRM1.

Lauren G. Monovich; Keith A. Koch; Robin Burgis; Ekundayo Osimboni; Thierry Mann; Daniel Wall; Jinhai Gao; Yan Feng; Richard B. Vega; Benjamin A. Turner; David B. Hood; Andy Law; Philip J. Papst; David Koditek; Joseph A. Chapo; Brian G. Reid; Lawrence Melvin; Nikos Pagratis; Timothy A. McKinsey

Histone deacetylase 5 (HDAC5) represses expression of nuclear genes that promote cardiac hypertrophy. Agonism of a variety of G protein coupled receptors (GPCRs) triggers phosphorylation-dependent nuclear export of HDAC5 via the CRM1 nuclear export receptor, resulting in derepression of pro-hypertrophic genes. A cell-based high-throughput screen of a commercial compound collection was employed to identify compounds with the ability to preserve the nuclear fraction of GFP-HDAC5 in primary cardiomyocytes exposed to GPCR agonists. A hit compound potently inhibited agonist-induced GFP-HDAC5 nuclear export in cultured neonatal rat ventricular myocytes (NRVMs). A small set of related compounds was designed and synthesized to evaluate structure-activity relationship (SAR). The results demonstrated that inhibition of HDAC5 nuclear export was a result of compounds irreversibly reacting with a key cysteine residue in CRM1 that is required for its function. CRM1 inhibition by the compounds also resulted in potent suppression of cardiomyocyte hypertrophy. These studies define a novel class of anti-hypertrophic compounds that function through irreversible inhibition of CRM1-dependent nuclear export.


Bioorganic & Medicinal Chemistry Letters | 2011

3,5-Diarylazoles as novel and selective inhibitors of protein kinase D

Gabriel G. Gamber; Erik Meredith; Qingming Zhu; Wanlin Yan; Chang Rao; Michael Paul Capparelli; Robin Burgis; Istvan J. Enyedy; Ji-Hu Zhang; Nicolas Soldermann; Kimberley Beattie; Olga Rozhitskaya; Keith A. Koch; Nikos Pagratis; Vinayak P. Hosagrahara; Richard B. Vega; Timothy A. McKinsey; Lauren G. Monovich

The synthesis and preliminary studies of the SAR of novel 3,5-diarylazole inhibitors of Protein Kinase D (PKD) are reported. Notably, optimized compounds in this class have been found to be active in cellular assays of phosphorylation-dependant HDAC5 nuclear export, orally bioavailable, and highly selective versus a panel of additional putative histone deacetylase (HDAC) kinases. Therefore these compounds could provide attractive tools for the further study of PKD/HDAC5 signaling.


Antimicrobial Agents and Chemotherapy | 2014

A Small-Molecule Inhibitor of Hepatitis C Virus Infectivity

Caroline O. Bush; Maria V. Pokrovskii; Roland Saito; Philip Anthony Morganelli; Eda Canales; Michael O. Clarke; Scott E. Lazerwith; Justin Golde; Brian Reid; Kerim Babaoglu; Nikos Pagratis; Weidong Zhong; William E. Delaney; Matthew Paulson; Rudolf K. F. Beran

ABSTRACT One of the most challenging goals of hepatitis C virus (HCV) research is to develop well-tolerated regimens with high cure rates across a variety of patient populations. Such a regimen will likely require a combination of at least two distinct direct-acting antivirals (DAAs). Combining two or more DAAs with different resistance profiles increases the number of mutations required for viral breakthrough. Currently, most DAAs inhibit HCV replication. We recently reported that the combination of two distinct classes of HCV inhibitors, entry inhibitors and replication inhibitors, prolonged reductions in extracellular HCV in persistently infected cells. We therefore sought to identify new inhibitors targeting aspects of the HCV replication cycle other than RNA replication. We report here the discovery of the first small-molecule HCV infectivity inhibitor, GS-563253, also called HCV infectivity inhibitor 1 (HCV II-1). HCV II-1 is a substituted tetrahydroquinoline that selectively inhibits genotype 1 and 2 HCVs with low-nanomolar 50% effective concentrations. It was identified through a high-throughput screen and subsequent chemical optimization. HCV II-1 only permits the production and release of noninfectious HCV particles from cells. Moreover, infectious HCV is rapidly inactivated in its presence. HCV II-1 resistance mutations map to HCV E2. In addition, HCV-II prevents HCV endosomal fusion, suggesting that it either locks the viral envelope in its prefusion state or promotes a viral envelope conformation change incapable of fusion. Importantly, the discovery of HCV II-1 opens up a new class of HCV inhibitors that prolong viral suppression by HCV replication inhibitors in persistently infected cell cultures.


Journal of Biomolecular Screening | 2015

High-Throughput Kinetic Screening of Hybridomas to Identify High-Affinity Antibodies Using Bio-Layer Interferometry

Latesh Lad; Sheila Clancy; Maria Kovalenko; Chian Liu; Terence Hui; Victoria C. Smith; Nikos Pagratis

Kinetic analysis of antibodies is crucial in both clone selection and characterization. Historically, antibodies in supernatants from hybridomas are selected based on a solid-phase enzyme-linked immunosorbent assay (ELISA) in which the antigen is immobilized on the assay plate. ELISA selects clones based on a combination of antibody concentration in the supernatant and affinity. The antibody concentration in the supernatant can vary significantly and is typically unknown. Using the ELISA method, clones that express high levels of a low-affinity antibody can give an equivalent signal as clones that express low levels of a high-affinity antibody. As a consequence, using the ELISA method, superior clones can be overshadowed by inferior clones. In this study, we have applied Bio-Layer Interferometry to screen hybridoma clones based on disassociation rates using the OctetRED 384 platform. Using the OctetRED platform, we were able to screen 2000 clones within 24 hours and select clones containing high-affinity antibodies for further expansion and subsequent characterization. Using this method, we were able to identify several clones producing high-affinity antibodies that were missed by ELISA.


Journal of Biomolecular Screening | 2011

Screening and Identification of a Novel Class of TGF-β Type 1 Receptor Kinase Inhibitor

Q. Khai Huynh; Sarah Wise; Keith A. Koch; Laurie Castonguay; Brian Reid; Erinn E. Pagratis; David Koditek; Christopher B. Glascock; Kelly R. PittsK.R. Pitts; Benjamin A. Turner; Xiaohong Liu; Magdeleine Hung; Bin Han; Nikos Pagratis

Transforming growth factor β (TGF-β) type I receptor (activin receptor–like kinase 5, ALK5) has been identified as a promising target for fibrotic diseases. To find a novel inhibitor of ALK5, the authors performed a high-throughput screen of a library of 420 000 compounds using dephosphorylated ALK5. From primary hits of 1521 compounds, 555 compounds were confirmed. In total, 124 compounds were then selected for follow-up based on their unique structures and other properties. Repeated concentration–response testing and final interference assays of the above compounds resulted in the discovery of a structurally novel ALK5 inhibitor (compound 8) (N-(thiophen 2-ylmethyl)-3-(3,4,5 trimethoxyphenyl)imidazo[1,2β]pyridazin 6-amine) with a low IC50 value of 0.7 µM. Compound 8 also inhibited the TGF-β-induced nuclear translocation of SMAD with an EC50 value of 0.8 µM. Kinetic analysis revealed that compound 8 inhibited ALK5 via mixed-type inhibition, suggesting that it may bind to ALK5 differently than other published adenosine triphosphate site inhibitors.


Bioorganic & Medicinal Chemistry Letters | 2017

Discovery of potent and selective inhibitors of calmodulin-dependent kinase II (CaMKII)

Dmitry Koltun; Eric Parkhill; Rao Kalla; Thao Perry; Elfatih Elzein; Xiaofen Li; Scott P. Simonovich; Christopher Ziebenhaus; Timothy R. Hansen; Bruno Marchand; WaiLok K. Hung; Leanna Lagpacan; Magdeleine Hung; Ron G. Aoyama; Bernard P. Murray; Jason K. Perry; John R. Somoza; Armando G. Villaseñor; Nikos Pagratis; Jeff Zablocki

We hereby disclose the discovery of inhibitors of CaMKII (7h and 7i) that are highly potent in rat ventricular myocytes, selective against hERG and other off-target kinases, while possessing good CaMKII tissue isoform selectivity (cardiac γ/δ vs. neuronal α/β). In vitro and in vivo ADME/PK studies demonstrated the suitability of these CaMKII inhibitors for PO (7h rat F = 73%) and IV pharmacological studies.


Biochemistry | 2015

Functional Label-Free Assays for Characterizing the in Vitro Mechanism of Action of Small Molecule Modulators of Capsid Assembly

Latesh Lad; Sheila Clancy; David Koditek; Melanie H. Wong; Debi Jin; Anita Niedziela-Majka; Giuseppe A. Papalia; Magdeleine Hung; Stephen R. Yant; John R. Somoza; Eric Hu; Chien-Hung Chou; Winston Tse; Randall L. Halcomb; Roman Sakowicz; Nikos Pagratis

HIV capsid protein is an important target for antiviral drug design. High-throughput screening campaigns have identified two classes of compounds (PF74 and BI64) that directly target HIV capsid, resulting in antiviral activity against HIV-1 and HIV-2 laboratory strains. Using recombinant proteins, we developed a suite of label-free assays to mechanistically understand how these compounds modulate capsid activity. PF74 preferentially binds to the preassembled hexameric capsid form and prevents disruption of higher-order capsid structures by stabilizing capsid intersubunit interactions. BI64 binds only the monomeric capsid and locks the protein in the assembly incompetent monomeric form by disrupting capsid intersubunit interactions. We also used these assays to characterize the interaction between capsid and the host protein cleavage and polyadenylation specific factor 6 (CPSF6). Consistent with recently published results, our assays revealed CPSF6 activates capsid polymerization and preferentially binds to the preassembled hexameric capsid form similar to the small molecule compound, PF74. Furthermore, these label-free assays provide a robust method for facilitating the identification of a different class of small molecule modulators of capsid function.

Collaboration


Dive into the Nikos Pagratis's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Timothy A. McKinsey

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Latesh Lad

University of California

View shared research outputs
Top Co-Authors

Avatar

Khai Huynh

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Roman Sakowicz

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge