Elliott Nickbarg
Merck & Co.
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Elliott Nickbarg.
Biochemical Pharmacology | 2011
Li Xiao; Elliott Nickbarg; Wenyan Wang; Ann Thomas; Michael Ziebell; Winfred W. Prosise; Charles A. Lesburg; Shane Taremi; Valerie L. Gerlach; Hung V. Le; K.-C. Cheng
The pregnane X-receptor (PXR) is a promiscuous nuclear receptor primarily responsible for the induction of genes from the cytochrome P450 3A family. In this study, we used a previously described PXR/SRC tethered protein to establish two in vitro assays for identifying PXR ligands: automated ligand identification system (ALIS) and temperature-dependent circular dichroism (TdCD). Kd values determined by ALIS and TdCD showed good correlations with the EC50 values determined by a PXR luciferase reporter-gene assay for 37 marketed drugs. The same set of compounds was modeled into the PXR ligand-binding domain that takes into consideration the structural variations of five published X-ray structures of PXR-ligand complexes. Major findings from our in silico analysis are as follows. First, the primary determinants for non-binders of PXR are molecular size and shape of the compounds. Low molecular weight (MW<300) compounds were in general found to be non-binders, and those molecules that do not match the shape of the PXR ligand-binding site may also act as a non-binder. Secondly, the favorable hydrophobic interactions, mostly through aromatic π-π interactions, and the presence of suitable hydrogen bond(s) between the compounds and PXR are attributes of strong binders. Thirdly, the structures of the PXR binding domain possess the flexibility that accommodates structurally diverse compounds, while some of the strong binders may also adapt flexible conformations for fitting into the binding site. The results from this study provide a molecular basis for future efforts in reducing/abolishing the PXR-dependent CYP3A4 induction liability.
Biochemistry | 2010
Payal R. Sheth; Gerald W. Shipps; Wolfgang Seghezzi; Catherine Smith; Cheng-Chi Chuang; David Paul Sanden; Andrea D. Basso; Lev Vilenchik; Kimberly Gray; D. Allen Annis; Elliott Nickbarg; Yao Ma; Brian R. Lahue; Ronald Herbst; Hung V. Le
Affinity selection-mass spectrometry (AS-MS) screening of kinesin spindle protein (KSP) followed by enzyme inhibition studies and temperature-dependent circular dichroism (TdCD) characterization was utilized to identify a series of benzimidazole compounds. This series also binds in the presence of Ispinesib, a known anticancer KSP inhibitor in phase I/II clinical trials for breast cancer. TdCD and AS-MS analyses support simultaneous binding implying existence of a novel non-Ispinesib binding pocket within KSP. Additional TdCD analyses demonstrate direct binding of these compounds to Ispinesib-resistant mutants (D130V, A133D, and A133D + D130V double mutant), further strengthening the hypothesis that the compounds bind to a distinct binding pocket. Also importantly, binding to this pocket causes uncompetitive inhibition of KSP ATPase activity. The uncompetitive inhibition with respect to ATP is also confirmed by the requirement of nucleotide for binding of the compounds. After preliminary affinity optimization, the benzimidazole series exhibited distinctive antimitotic activity as evidenced by blockade of bipolar spindle formation and appearance of monoasters. Cancer cell growth inhibition was also demonstrated either as a single agent or in combination with Ispinesib. The combination was additive as predicted by the binding studies using TdCD and AS-MS analyses. The available data support the existence of a KSP inhibitory site hitherto unknown in the literature. The data also suggest that targeting this novel site could be a productive strategy for eluding Ispinesib-resistant tumors. Finally, AS-MS and TdCD techniques are general in scope and may enable screening other targets in the presence of known drugs, clinical candidates, or tool compounds that bind to the protein of interest in an effort to identify potency-enhancing small molecules that increase efficacy and impede resistance in combination therapy.
Journal of Biomolecular Screening | 2011
Joost C.M. Uitdehaag; Cecile M. Sünnen; Antoon M. van Doornmalen; Nikki de Rouw; Arthur Oubrie; Rita Azevedo; Michael Ziebell; Elliott Nickbarg; Willem-Jan Karstens; Simone Ruygrok
Over the past years, improvements in high-throughput screening (HTS) technology and compound libraries have resulted in a dramatic increase in the amounts of good-quality screening hits, and there is a growing need for follow-on hit profiling assays with medium throughput to further triage hits. Here the authors present such assays for the colony-stimulating factor 1 receptor (CSF1R, Fms), including tests for cellular activity and a homogeneous assay to measure affinity for inactive CSF1R. They also present a high-throughput assay to measure target residence time, which is based on competitive binding kinetics. To better fit koff rates, they present a modified mathematical model for competitive kinetics. In all assays, they profiled eight reference inhibitors (imatinib, sorafenib, sunitinib, tandutinib, dasatinib, GW2580, Ki20227, and J&J’s pyrido[2,3-d]pyrimidin-5-one). Using the known biochemical selectivities of these inhibitors, which can be quantified using metrics such as the selectivity entropy, the authors have determined which assay readout best predicts hit selectivity. Their profiling shows surprisingly that imatinib has a preference for the active form of CSF1R and that Ki20227 has an unusually slow target dissociation rate. This confirms that follow-on hit profiling is essential to ensure that the best hits are selected for lead optimization.
Journal of Biomolecular Screening | 2016
Victoria Kutilek; Christine L. Andrews; Matthew Richards; Zangwei Xu; Tianxiao Sun; Yiping Chen; Andrew Hashke; Nadya Smotrov; Rafael Fernandez; Elliott Nickbarg; Chad Chamberlin; Berengere Sauvagnat; Patrick J. Curran; Ryan Boinay; Peter Saradjian; Samantha J. Allen; Noel Byrne; Nathaniel L. Elsen; Rachael E. Ford; Dawn L. Hall; Maria Kornienko; Keith W. Rickert; Sujata Sharma; Jennifer M. Shipman; Kevin J. Lumb; Kevin Coleman; Peter J. Dandliker; Ilona Kariv; Bruce A. Beutel
The primary objective of early drug discovery is to associate druggable target space with a desired phenotype. The inability to efficiently associate these often leads to failure early in the drug discovery process. In this proof-of-concept study, the most tractable starting points for drug discovery within the NF-κB pathway model system were identified by integrating affinity selection–mass spectrometry (AS-MS) with functional cellular assays. The AS-MS platform Automated Ligand Identification System (ALIS) was used to rapidly screen 15 NF-κB proteins in parallel against large-compound libraries. ALIS identified 382 target-selective compounds binding to 14 of the 15 proteins. Without any chemical optimization, 22 of the 382 target-selective compounds exhibited a cellular phenotype consistent with the respective target associated in ALIS. Further studies on structurally related compounds distinguished two chemical series that exhibited a preliminary structure-activity relationship and confirmed target-driven cellular activity to NF-κB1/p105 and TRAF5, respectively. These two series represent new drug discovery opportunities for chemical optimization. The results described herein demonstrate the power of combining ALIS with cell functional assays in a high-throughput, target-based approach to determine the most tractable drug discovery opportunities within a pathway.
Proceedings of the National Academy of Sciences of the United States of America | 2017
Hua-Poo Su; Keith Rickert; Christine Burlein; Kartik Narayan; Marina Bukhtiyarova; Danielle M. Hurzy; Craig A. Stump; Xu-Fang Zhang; John Reid; Alicja Krasowska-Zoladek; Srivanya Tummala; Jennifer M. Shipman; Maria Kornienko; Peter Lemaire; Daniel Krosky; Amanda Heller; Abdelghani Abe Achab; Chad Chamberlin; Peter Saradjian; Berengere Sauvagnat; Xianshu Yang; Michael Ziebell; Elliott Nickbarg; John M. Sanders; Mark T. Bilodeau; Steven S. Carroll; Kevin J. Lumb; Stephen M. Soisson; Darrell A. Henze; Andrew John Cooke
Significance Signal transduction through Tropomyosin-related kinase A (TrkA), a receptor tyrosine kinase, is a target for inhibition of chronic pain and could lead to a new class of drugs against pain. Selectivity against kinases can be difficult to achieve, especially against members of the same kinase family. Structures of the compounds bound to TrkA show a binding site comprised of the kinase, which is conserved among the Trk family, and the juxtamembrane (JM), which is not well conserved. Depending on their chemical substructure, the region of the juxtamembrane that interacts with the compounds can be different, leading to differences in specificity. This study emphasizes the importance of including residues beyond the catalytic domain for small-molecule screening, importance of screening by affinity, and structural characterization to understand binding interactions. Current therapies for chronic pain can have insufficient efficacy and lead to side effects, necessitating research of novel targets against pain. Although originally identified as an oncogene, Tropomyosin-related kinase A (TrkA) is linked to pain and elevated levels of NGF (the ligand for TrkA) are associated with chronic pain. Antibodies that block TrkA interaction with its ligand, NGF, are in clinical trials for pain relief. Here, we describe the identification of TrkA-specific inhibitors and the structural basis for their selectivity over other Trk family kinases. The X-ray structures reveal a binding site outside the kinase active site that uses residues from the kinase domain and the juxtamembrane region. Three modes of binding with the juxtamembrane region are characterized through a series of ligand-bound complexes. The structures indicate a critical pharmacophore on the compounds that leads to the distinct binding modes. The mode of interaction can allow TrkA selectivity over TrkB and TrkC or promiscuous, pan-Trk inhibition. This finding highlights the difficulty in characterizing the structure-activity relationship of a chemical series in the absence of structural information because of substantial differences in the interacting residues. These structures illustrate the flexibility of binding to sequences outside of—but adjacent to—the kinase domain of TrkA. This knowledge allows development of compounds with specificity for TrkA or the family of Trk proteins.
ACS Chemical Biology | 2018
Noreen F. Rizvi; John A. Howe; Ali Nahvi; Daniel J. Klein; Thierry O. Fischmann; Hai-Young Kim; Mark A. McCoy; Scott S. Walker; Alan Hruza; Matthew Richards; Chad Chamberlin; Peter Saradjian; Margaret T. Butko; Gabriel Mercado; Julja Burchard; Corey Strickland; Peter J. Dandliker; Graham F. Smith; Elliott Nickbarg
Recent advances in understanding the relevance of noncoding RNA (ncRNA) to disease have increased interest in drugging ncRNA with small molecules. The recent discovery of ribocil, a structurally distinct synthetic mimic of the natural ligand of the flavin mononucleotide (FMN) riboswitch, has revealed the potential chemical diversity of small molecules that target ncRNA. Affinity-selection mass spectrometry (AS-MS) is theoretically applicable to high-throughput screening (HTS) of small molecules binding to ncRNA. Here, we report the first application of the Automated Ligand Detection System (ALIS), an indirect AS-MS technique, for the selective detection of small molecule-ncRNA interactions, high-throughput screening against large unbiased small-molecule libraries, and identification and characterization of novel compounds (structurally distinct from both FMN and ribocil) that target the FMN riboswitch. Crystal structures reveal that different compounds induce various conformations of the FMN riboswitch, leading to different activity profiles. Our findings validate the ALIS platform for HTS screening for RNA-binding small molecules and further demonstrate that ncRNA can be broadly targeted by chemically diverse yet selective small molecules as therapeutics.
ACS Chemical Biology | 2017
John P. Santa Maria; Yumi Park; Lihu Yang; Nicholas J. Murgolo; Michael D. Altman; Paul Zuck; Greg Adam; Chad Chamberlin; Peter Saradjian; Peter J. Dandliker; Helena I. Boshoff; Clifton E. Barry; Charles G. Garlisi; David B. Olsen; Katherine Young; Meir Glick; Elliott Nickbarg; Peter S. Kutchukian
Though phenotypic and target-based high-throughput screening approaches have been employed to discover new antibiotics, the identification of promising therapeutic candidates remains challenging. Each approach provides different information, and understanding their results can provide hypotheses for a mechanism of action (MoA) and reveal actionable chemical matter. Here, we describe a framework for identifying efficacy targets of bioactive compounds. High throughput biophysical profiling against a broad range of targets coupled with machine learning was employed to identify chemical features with predicted efficacy targets for a given phenotypic screen. We validate the approach on data from a set of 55 000 compounds in 24 historical internal antibacterial phenotypic screens and 636 bacterial targets screened in high-throughput biophysical binding assays. Models were built to reveal the relationships between phenotype, target, and chemotype, which recapitulated mechanisms for known antibacterials. We also prospectively identified novel inhibitors of dihydrofolate reductase with nanomolar antibacterial efficacy against Mycobacterium tuberculosis. Molecular modeling provided structural insight into target-ligand interactions underlying selective killing activity toward mycobacteria over human cells.
SLAS DISCOVERY: Advancing Life Sciences R&D | 2018
Deborah A. Flusberg; Noreen F. Rizvi; Victoria Kutilek; Christine L. Andrews; Peter Saradjian; Chad Chamberlin; Patrick J. Curran; Brooke Swalm; Sam Kattar; Graham F. Smith; Peter J. Dandliker; Elliott Nickbarg; Jennifer O’Neil
The Myc oncogene is overexpressed in many cancers, yet targeting it for cancer therapy has remained elusive. One strategy for inhibition of Myc expression is through stabilization of the G-quadruplex (G4), a G-rich DNA secondary structure found within the Myc promoter; stabilization of G4s has been shown to halt transcription of downstream gene products. Here we used the Automated Ligand Identification System (ALIS), an affinity selection–mass spectrometry method, to identify compounds that bind to the Myc G4 out of a pool of compounds that had previously been shown to inhibit Myc expression in a reporter screen. Using an ALIS-based screen, we identified hits that bound to the Myc G4, a small subset of which bound preferentially relative to G4s from the promoters of five other genes. To determine functionality and specificity of the Myc G4-binding compounds in cell-based assays, we compared inhibition of Myc expression in cells with and without Myc G4 regulation. Several compounds inhibited Myc expression only in the Myc G4-containing line, and one compound was verified to function through Myc G4 binding. Our study demonstrates that ALIS can be used to identify selective nucleic acid-binding compounds from phenotypic screen hits, increasing the pool of drug targets beyond proteins.
Combinatorial Chemistry & High Throughput Screening | 2012
Charles E. Whitehurst; Zhiping Yao; Mingxuan Zhang; Shane Taremi; Lisa Wojcik; Julie M. Strizki; Jack D. Bracken; Cliff C. Cheng; Xianshu Yang; Gerald W. Shipps; Michael R. Ziebell; Elliott Nickbarg
ACS Chemical Biology | 2017
Scott S. Walker; David Degen; Elliott Nickbarg; Donna M. Carr; Aileen Soriano; Mihir Mandal; Ronald E. Painter; Payal R. Sheth; Li Xiao; Xinwei Sher; Nicholas J. Murgolo; Jing Su; David B. Olsen; Richard H. Ebright; Katherine Young