James P. Snyder

Find an error

Name:
Organization: Emory University
Department: Department of Chemistry
Title:
Co-reporter:Qi Shi, Thomas M. Kaiser, Zackery W. Dentmon, Mariangela Ceruso, Daniela Vullo, Claudiu T. Supuran, and James P. Snyder
ACS Medicinal Chemistry Letters 2015 Volume 6(Issue 5) pp:518
Publication Date(Web):April 3, 2015
DOI:10.1021/acsmedchemlett.5b00062
A method capable of identifying novel synthetic targets for small molecule lead optimization has been developed. The FRESH (FRagment-based Exploitation of modular Synthesis by vHTS) approach relies on a multistep synthetic route to a target series of compounds devised by a close collaboration between synthetic and computational chemists. It combines compound library generation, quantitative structure–acitvity relationship construction, fragment processing, virtual high throughput screening and display of results within the Pipeline Pilot framework. Outcomes enumerate tailored selection of novel synthetic targets with improved potency and optimized physical properties for an emerging compound series. To validate the application of FRESH, three retrospective case studies have been performed to pinpoint reported potent analogues. One prospective case study was performed to demonstrate that FRESH is able to capture additional potent analogues.Keywords: ADME; FRESH; lead optimization; Pipeline Pilot; QSAR; virtual HTS
Co-reporter:David G. I. Kingston and James P. Snyder
Accounts of Chemical Research 2014 Volume 47(Issue 8) pp:2682-2691
Publication Date(Web):July 23, 2014
DOI:10.1021/ar500203h
We surmise that past studies have been handicapped by solubility and membrane permeability issues, but primarily by the existence of an expansive taxane binding pocket and the discrepancy in molecular size between PTX and the pruned analogs. A number of these molecules offer molecular volumes 50–60% that of PTX, fewer contacts with the tubulin protein, severe mismatches with the PTX pharmacophore, lessened capacity to dispel binding site waters contributing to ΔGbind, and unanticipated binding poses. The latter is a critical drawback if molecular designs of simpler PTX structures are based on a perceived or known PTX binding conformation. We conclude that design and synthesis of a highly cytotoxic tubulin-assembly agent based on the paclitaxel pharmacophore remains an unsolved challenge, but one that can be overcome by focus on the architecture of the taxane binding site independent of the effective, but not unique, hand-in-glove match represented by the PTX–tubulin complex.
Co-reporter:Mi-Sun Kim, Lauriane A. Buisson, Dean A. Heathcote, Haipeng Hu, D. Christopher Braddock, Anthony G. M. Barrett, Philip G. Ashton-Rickardt and James P. Snyder  
Organic & Biomolecular Chemistry 2014 vol. 12(Issue 44) pp:8952-8965
Publication Date(Web):23 Sep 2014
DOI:10.1039/C4OB01874E
A structure-based design campaign for non-covalent small molecule inhibitors of human granzyme B was carried out by means of a virtual screening strategy employing three constraints and probe site-mapping with FTMAP to identify ligand “hot spots”. In addition, new scaffolds of diverse structures were subsequently explored with ROCS shape-based superposition methods, following by Glide SP docking, induced fit docking and analysis of QikProp molecular properties. Novel classes of moderately active small molecule blockers (≥25 μM IC50 values) from commercially available libraries were identified, and three novel scaffolds have been synthesized by multi-step procedures. Furthermore, we provide an example of a comprehensive structure-based drug discovery approach to non-covalent inhibitors that relies on the X-ray structure of a covalently bound ligand and suggest that the design path may be compromised by alternative and unknown binding poses.
Co-reporter:Craig Grimmer, Terry W. Moore, Aaron Padwa, Andrew Prussia, Gordon Wells, Shaoxiong Wu, Aiming Sun, and James P. Snyder
Journal of Chemical Information and Modeling 2014 Volume 54(Issue 8) pp:2214-2223
Publication Date(Web):July 24, 2014
DOI:10.1021/ci500204j
Biologically active organic molecules characterized by a high single bond torsional barrier generate isolable isomers (atropisomers) and offer a unique stereochemical component to the design of selective therapeutic agents. The present work presents a nanomolar active inhibitor of myxoviruses, which most likely acts by blocking one or more cellular host proteins but also, serendipitously, exhibits axial chirality with an energy barrier of ΔG⧧ ≥30 kcal/mol. The latter has been probed by variable temperature NMR and microwave irradiation and by high level DFT transition state analysis and force field calculations. Full conformational profiles of the corresponding (aR,S) and (aS,S) atropisomers at ambient temperature were derived by conformer deconvolution with NAMFIS (NMR Analysis by Molecular Flexibility In Solution) methodology to generate seven and eight individual conformations, each assigned a % population. An accurate evaluation of a key torsion angle at the center of the molecules associated with a 3JC–S–C–H coupling constant was obtained by mapping the S–C bond rotation with the MPW1PW91/6-31G-d,p DFT method followed by fitting the resulting dihedral angles and J-values to a Karplus expression. Accordingly, we have developed a complete conformational profile of diastereomeric atropisomers consistent with both high and low rotational barriers. We expect this assessment to assist the rationalization of the selectivity of the two (aR,S) and (aS,S) forms against host proteins, while offering insights into their divergent toxicity behavior.
Co-reporter:Haipeng Hu, Shuli Mao, Julia V. Bugrysheva, Sarah Pruett, Dennis C. Liotta, June R. Scott, James P. Snyder
European Journal of Medicinal Chemistry 2014 Volume 82() pp:120-126
Publication Date(Web):23 July 2014
DOI:10.1016/j.ejmech.2014.05.006
•Homology models of RNase J1/J2 were developed as targets for Group A streptococcus.•High-throughput virtual screening (HTVS) was performed against 2 compound libraries.•Two compounds with 10 μM MIC activity were identified and characterized.•The HTVS strategy lays the groundwork for an extensive scaffold-hopping campaign.Group A streptococcus (GAS) is a Gram-positive bacterium, which can cause multiple types of disease from mild infections of skin and throat to invasive and life-threatening infections. Recently RNase J1 and J2 were found to be essential for the growth of GAS. In order to identify inhibitors against RNase J1/J2, homology models of both the ligand-free apo-form and the ligand-bound holo-form complexes were constructed as templates for high-throughput virtual screening (HTVS). A focused small molecule library and the commercially available Maybridge database were employed as sources for potential inhibitors. A cell-based biological assay identified two compounds with 10 μM MIC activity.
Co-reporter:Terry W. Moore, Shijun Zhu, Ryan Randolph, Mamoru Shoji, and James P. Snyder
ACS Medicinal Chemistry Letters 2014 Volume 5(Issue 4) pp:288-292
Publication Date(Web):January 10, 2014
DOI:10.1021/ml4002453
To address the shortcomings of the natural product curcumin, many groups have created analogues that share similar structural features while displaying superior properties, particularly in anticancer drug discovery. Relatively unexplored have been the mechanisms by which such compounds are metabolized. A comprehensive in vitro study of a curcumin analogue (UBS109) in liver S9 fractions from five different species is presented. Further, we examine the cell-based bioactivity of the major metabolites. In spite of the fact that UBS109 reduces tumor growth in mice, it is quickly metabolized in vitro and 94% protein bound in mouse plasma. The primary monounsaturated metabolite is only modestly bioactive against MDA-MB-231 breast cancer cells. These observations suggest that while the α,β-unsaturated ketone common to curcumin analogues is important for bioactivity, protein binding and tissue distribution may serve to protect UBS109 from full metabolism in vivo while allowing it to exert a pharmacological effect by means of slow drug release.Keywords: curcumin; Curcumin analogue; liver S9 fraction; metabolism; UBS109;
Co-reporter:Dr. Bryan D. Cox;Anthony R. Prosser;Brooke M. Katzman;Dr. Ana A. Alcaraz; Dennis C. Liotta;Dr. Lawrence J. Wilson ;Dr. James P. Snyder
ChemBioChem 2014 Volume 15( Issue 11) pp:1614-1620
Publication Date(Web):
DOI:10.1002/cbic.201402056

Abstract

The CXC chemokine receptor 4 (CXCR4) is involved in chemotaxis and serves as a coreceptor for T-tropic HIV-1 viral entry, thus making this receptor an attractive drug target. Recently, crystal structures of CXCR4 were reported as complexes with the small molecule IT1t and the CVX15 peptide. Follow-up efforts to model different antagonists into the small molecule CXCR4:IT1t crystal structure did not generate poses consistent with either the X-ray crystal structure or site-directed mutagenesis (SDM). Here, we compare the binding pockets of the two CXCR4 crystal structures, revealing differences in helices IV, V, VI, and VII, with major differences for the His203 residue buried in the binding pocket. The small molecule antagonist AMD11070 was docked into both CXCR4 crystal structures. An AMD11070 pose identified from the CXCR4:CVX15 model presented interactions with Asp171, Glu288, Trp94, and Asp97, consistent with published SDM data, thus suggesting it is the bioactive pose. A CXCR4 receptor model was optimized around this pose of AMD11070, and the resulting model correlated HIV-1 inhibition with MM-GBSA docking scores for a congeneric AMD11070-like series. Subsequent NAMFIS NMR results successfully linked the proposed binding pose to an independent experimental structure. These results strongly suggest that not all small molecules will bind to CXCR4 in a similar manner as IT1t. Instead, the CXCR4:CVX15 crystal structure may provide a binding locus for small organic molecules that is more suitable than the secondary IT1t site. This work is expected to provide modeling insights useful for future CXCR4 antagonist and X4-tropic HIV-1 based drug design efforts.

Co-reporter:Andrew Brown ; Qi Shi ; Terry W. Moore ; Younghyoun Yoon ; Andrew Prussia ; Clinton Maddox ; Dennis C. Liotta ; Hyunsuk Shim
Journal of Medicinal Chemistry 2013 Volume 56(Issue 9) pp:3456-3466
Publication Date(Web):April 3, 2013
DOI:10.1021/jm4002692
Curcumin is a biologically active component of curry powder. A structurally related class of mimetics possesses similar anti-inflammatory and anticancer properties. Mechanism has been examined by exploring kinase inhibition trends. In a screen of 50 kinases relevant to many forms of cancer, one member of the series (4, EF31) showed ≥85% inhibition for 10 of the enzymes at 5 μM, while 22 of the proteins were blocked at ≥40%. IC50 values for an expanded set of curcumin analogues established a rank order of potencies, and analyses of IKKβ and AKT2 enzyme kinetics for 4 revealed a mixed inhibition model, ATP competition dominating. Our curcumin mimetics are generally selective for Ser/Thr kinases. Both selectivity and potency trends are compatible with protein sequence comparisons, while modeled kinase binding site geometries deliver a reasonable correlation with mixed inhibition. Overall, these analogues are shown to be pleiotropic inhibitors that operate at multiple points along cell signaling pathways.
Co-reporter:Dr. Suazette Reid Mooring;Dr. Jin Liu;Dr. Zhongxing Liang;Jeffrey Ahn;Samuel Hong;Dr. Younghyoun Yoon;Dr. James P. Snyder;Dr. Hyunsuk Shim
ChemMedChem 2013 Volume 8( Issue 4) pp:622-632
Publication Date(Web):
DOI:10.1002/cmdc.201200582

Abstract

The interaction of CXCR4 with CXCL12 (SDF-1) plays a critical role in cancer metastasis by facilitating the homing of tumor cells to metastatic sites. Based on our previously published work on CXCR4 antagonists, we have synthesized a series of aryl sulfonamides that inhibit the CXCR4/CXCL12 interaction. Analogue bioactivities were assessed with binding affinity and Matrigel invasion assays. Computer modeling was employed to evaluate a selection of the new analogues docked into the CXCR4 X-ray structure and to rationalize discrepancies between the affinity and Matrigel in vitro assays. A lead compound displays nanomolar potency in the binding affinity assay (IC50=8.0 nM) and the Matrigel invasion assay (100 % blockade of invasion at 10 nM). These data demonstrate that benzenesulfonamides are a unique class of CXCR4 inhibitors with high potency.

Co-reporter:Haipeng Hu and James P. Snyder
Journal of Chemical Information and Modeling 2012 Volume 52(Issue 12) pp:3190-3199
Publication Date(Web):November 19, 2012
DOI:10.1021/ci300287t
We describe the application of different methods in the development of QSAR models for IKKA and IKKB inhibition. The results show that the best QSAR models provide highly accurate predictions for existing IkB-kinase (IKK) inhibitors. The exceptions, corresponding to 5% of the known collection of inhibitors, are five classes of compounds incorporating the nitrile or sulfonamide moieties, small compounds with molecular weights of less than 300, and two classes of blockers considered to be type II kinase inhibitors. Comparison of our novel IKKB homology model and the recently reported IKKB crystal structure implies that a predictive protein–antagonist complex structure is more likely to exist as an inactive form in the crystalline state as observed in the recent protein X-ray structure.
Co-reporter:Qi Shi, Shaoman Yin, Stefan Kaluz, Nanting Ni, Narra Sarojini Devi, Jiyoung Mun, Danzhu Wang, Krishna Damera, Weixuan Chen, Sarah Burroughs, Suazette Reid Mooring, Mark M. Goodman, Erwin G. Van Meir, Binghe Wang, and James P. Snyder
ACS Medicinal Chemistry Letters 2012 Volume 3(Issue 8) pp:620
Publication Date(Web):June 21, 2012
DOI:10.1021/ml300042k
Hypoxia inducible factors (HIFs) are transcription factors that activate expression of multiple gene products and promote tumor adaptation to a hypoxic environment. To become transcriptionally active, HIFs associate with cofactors p300 or CBP. Previously, we found that arylsulfonamides can antagonize HIF transcription in a bioassay, block the p300/HIF-1α interaction, and exert potent anticancer activity in several animal models. In the present work, KCN1-bead affinity pull down, 14C-labeled KCN1 binding, and KCN1-surface plasmon resonance measurements provide initial support for a mechanism in which KCN1 can bind to the CH1 domain of p300 and likely prevent the p300/HIF-1α assembly. Using a previously reported NMR structure of the p300/HIF-1α complex, we have identified potential binding sites in the p300-CH1 domain. A two-site binding model coupled with IC50 values has allowed establishment of a modest ROC-based enrichment and creation of a guide for future analogue synthesis.Keywords: binding model; HIF arylsulfonamide inhibitors; hypoxia; KCN1; p300; QSAR; solid tumors
Co-reporter:Jullien Rey;Timothy J. C. O'Riordan;Haipeng Hu;Andrew J. P. White;Anthony G. M. Barrett
European Journal of Organic Chemistry 2012 Volume 2012( Issue 20) pp:3781-3794
Publication Date(Web):
DOI:10.1002/ejoc.201200190

Abstract

A novel and efficient synthetic strategy to access unique C-2 substituted steroid analogues 3 and 4 is described. The unusual C-2 aryl ether analogues 3 were shown to act as virtual antagonists of LRH-1 and were prepared as single diastereoisomers, employing a fifteen-step sequence from pregnenolone (9). The key steps include the stereoconvergent nucleophilic displacement of an epimeric mixture of 3-keto 2-bromo steroids, chemoselective carbonylation of an enol triflate and conversion of a thiopyridyl ester into an aryl ketone. The related C-2 benzyl analogues 4 were prepared in a similar manner.

Co-reporter:Dr. Jullien Rey;Dr. Haipeng Hu;Dr. Fiona Kyle;Chun-Fui Lai;Dr. Laki Buluwela; R. Charles Coombes; Eric A. Ortlund; Simak Ali; James P. Snyder; Anthony G. M. Barrett
ChemMedChem 2012 Volume 7( Issue 11) pp:
Publication Date(Web):
DOI:10.1002/cmdc.201290058
Co-reporter:Dr. Jullien Rey;Dr. Haipeng Hu;Dr. Fiona Kyle;Chun-Fui Lai;Dr. Laki Buluwela; R. Charles Coombes; Eric A. Ortlund; Simak Ali; James P. Snyder; Anthony G. M. Barrett
ChemMedChem 2012 Volume 7( Issue 11) pp:1909-1914
Publication Date(Web):
DOI:10.1002/cmdc.201200307
Co-reporter:Ashutosh S. Jogalekar ; Krishnan Damodaran ; Frederik H. Kriel ; Won-Hyuk Jung ; Ana A. Alcaraz ; Shi Zhong ; Dennis P. Curran
Journal of the American Chemical Society 2011 Volume 133(Issue 8) pp:2427-2436
Publication Date(Web):February 7, 2011
DOI:10.1021/ja1023817
Dictyostatin (DCT, 1) is a complex, flexible polyketide macrolide that demonstrates potent microtubule-polymerization activity. Both a solution structure (2a) and a possible binding mode for DCT (Conf-1) have been proposed by earlier NMR experiments. In the present study, the conformational landscape of DCT in DMSO-d6 and methanol-d4 was explored using extensive force-field-based conformational searches combined with geometric parameters derived from solution NMR data. The results portray a diversity of conformations for dictyostatin that illustrates the molecule’s flexibility and excludes the previously suggested dominant solution conformation 2a. One conformation present in DMSO-d6 with a 7% population (Conf-2, 0.6 kcal/mol above the global minimum at 298°) also satisfies the TR-NOESY NMR parameters of Canales et al. that characterize the taxane binding-site interaction between DCT and assembled microtubules in water. Application of several docking methods (Glide, Autodock, and RosettaLigand) has identified a low-energy binding model of the DCT/β-tubulin complex (Pose-2/Conf-2) that is gratifyingly compatible with the emerging DCT structure−activity data.
Co-reporter:Dr. Weiqiang Zhan;Dr. Yi Jiang;Shubhada Sharma;Peggy J. Brodie; Susan Bane; David G. I. Kingston; Dennis C. Liotta;Dr. James P. Snyder
Chemistry - A European Journal 2011 Volume 17( Issue 52) pp:14792-14804
Publication Date(Web):
DOI:10.1002/chem.201102630

Abstract

A series of conformationally restrained epothilone analogues with a short bridge between the methyl groups at C6 and C8 was designed to mimic the binding pose assigned to our recently reported EpoA–microtubule binding model. A versatile synthetic route to these bridged epothilone analogues has been successfully devised and implemented. Biological evaluation of the compounds against A2780 human ovarian cancer and PC3 prostate cancer cell lines suggested that the introduction of a bridge between C6–C8 reduced potency by 25–1000 fold in comparison with natural epothilone D. Tubulin assembly measurements indicate these bridged epothilone analogues to be mildly active, but without significant microtubule stabilization capacity. Molecular mechanics and DFT energy evaluations suggest the mild activity of the bridged epo-analogues may be due to internal conformational strain.

Co-reporter:Qiao-Hong Chen, Thota Ganesh, Yi Jiang, Abhijit Banerjee, Shubhada Sharma, Susan Bane, James P. Snyder and David G. I. Kingston  
Chemical Communications 2010 vol. 46(Issue 12) pp:2019-2021
Publication Date(Web):09 Feb 2010
DOI:10.1039/B926174E
An unusual reaction with Grubbs' catalyst during the synthesis of bridged epothilones yielded five-membered internal lactones instead of the expected metathesis products. Three of the lactones have activities comparable to epothilone D.
Co-reporter:Aizhi Zhu ; Weiqiang Zhan ; Zhongxing Liang ; Younghyoun Yoon ; Hua Yang ; Hans E. Grossniklaus ; Jianguo Xu ; Mauricio Rojas ; Mark Lockwood ; James P. Snyder ; Dennis C. Liotta ;Hyunsuk Shim
Journal of Medicinal Chemistry 2010 Volume 53(Issue 24) pp:8556-8568
Publication Date(Web):November 24, 2010
DOI:10.1021/jm100786g
The C-X-C chemokine receptor type 4 (CXCR4)/stromal cell derived factor-1 (SDF-1 or CXCL12) interaction and the resulting cell signaling cascade play a key role in metastasis and inflammation. On the basis of the previously published CXCR4 antagonist 5 (WZ811), a series of novel nonpeptidic anti-CXCR4 small molecules have been designed and synthesized to improve potency. Following a structure−activity profile around 5, more advanced compounds in the N,N′-(1, 4-phenylenebis(methylene)) dipyrimidin-2-amines series were discovered and shown to possess higher CXCR4 binding potential and specificity than 5. Compound 26 (508MCl) is the lead compound and exhibits subnanomolar potency in three in vitro assays including competitive binding, Matrigel invasion and Gαi cyclic adenosine monophosphate (cAMP) modulation signaling. Furthermore, compound 26 displays promising effects by interfering with CXCR4 function in three mouse models: paw inflammation, Matrigel plug angiogenesis, and uveal melanoma micrometastasis. These data demonstrate that dipyrimidine amines are unique CXCR4 antagonists with high potency and specificity.
Co-reporter:Ashutosh S. Jogalekar ; Frederik H. Kriel ; Qi Shi ; Ben Cornett ; Daniel Cicero
Journal of Medicinal Chemistry 2010 Volume 53(Issue 1) pp:155-165
Publication Date(Web):November 6, 2009
DOI:10.1021/jm9015284
(+)-Discodermolide (DDM), a polyketide macrolide from marine sponge, is a potent microtubule assembly promoter. Reported solid-state, solution, and protein-bound DDM conformations reveal the unusual result that a common hairpin conformational motif exists in all three microenvironments. No other flexible microtubule binding agent exhibits such constancy of conformation. In the present study, we combine force-field conformational searches with NMR deconvolution in different solvents to compare DDM conformers with those observed in other environments. While several conformational families are perceived, the hairpin form dominates. The stability of this motif is dictated primarily by steric factors arising from repeated modular segments in DDM composed of the C(Me)-CHX-C(Me) fragment. Furthermore, docking protocols were utilized to probe the DDM binding mode in β-tubulin. A previously suggested pose is substantiated (Pose-1), while an alternative (Pose-2) has been identified. SAR analysis for DDM analogues differentiates the two poses and suggests that Pose-2 is better able to accommodate the biodata.
Co-reporter:Andrew J. Prussia Dr.;Yutao Yang;Matthew T. Geballe Dr. Dr.
ChemBioChem 2010 Volume 11( Issue 1) pp:101-109
Publication Date(Web):
DOI:10.1002/cbic.200900538

Abstract

Cyclostreptin (CS) is a recently discovered natural product with cytotoxic activity caused by microtubule stabilization. It is the only known microtubule-stabilizing agent (MSA) that covalently binds to tubulin. It also exhibits the fast-binding kinetics seen for other MSAs. Through careful peptide digestion and mass spectrometry analysis, Buey et al. found that two amino acids are labeled by CS: Asn228, near the known taxane-binding site, and Thr220, in the type I microtubule pore. This led Buey et al. to propose Thr220 resides at the site previously predicted to be a way station or low-affinity site. By using molecular dynamics simulations and structural considerations of the microtubule pore and tubulin dimer, we conclude that postulation of a low-affinity site is unnecessary to explain the available experimental data. An alternative explanation views the microtubule pore as a structural entity that presents a substantial kinetic barrier to ligand passage to the known taxane-binding site—an entry point to the microtubule lumen that becomes completely blocked if cyclostreptin is bound at Thr220. Simulations of the free dimer also suggest a common mechanism of microtubule stabilization for taxane site MSAs through their conformational effect on the M-loop. Such an effect explains the low tubulin polymerization caused by cyclostreptin in vitro despite its covalent attachment.

Co-reporter:Yutao Yang, Ana A. Alcaraz and James P. Snyder
Journal of Natural Products 2009 Volume 72(Issue 3) pp:422-429
Publication Date(Web):March 6, 2009
DOI:10.1021/np800662j
Nearly 35 years after its discovery and 11 years after FDA approval of paclitaxel (PTX) as a breakthrough anticancer drug, the 3-D structure of the agent bound to its β-tubulin target was proposed to be T-Taxol. The latter bioactive form has recently been challenged by the Ojima group with a structure, “PTX-NY” (“REDOR Taxol”), in which the C-13 side chain is proposed to adopt a different conformation and an alternative hydrogen-bonding pattern in the tubulin binding site. Previously, the two conformers were compared to show that only T-Taxol fits the PTX-derived electron crystallographic density. That work has been extended by molecular mechanics and quantum chemical methods to reveal that the PTX-NY conformation is relatively less stable, on average, by 10−11 kcal/mol. In agreement with NMR studies, an 11 ns molecular dynamics treatment for PTX in an explicit water pool locates T-Taxol along the trajectory, but not PTX-NY. Docking of various PTX conformers into the electron crystallographic binding site of tubulin demonstrates that PTX-NY cannot be accommodated unless the pocket is reorganized in violation of the experimental constraints. Finally, analysis of the structures of T-Taxol and PTX-NY for their capacity to predict the existence of superpotent PTX analogues discloses that only the former forecasts such analogues, as now established by the T-Taxol-inspired synthesis of bridged taxanes. In sum, all empirical criteria support T-Taxol as the bound conformation of PTX on β-tubulin in microtubules.
Co-reporter:Aiming Sun, Yang J. Lu, Haipeng Hu, Mamoru Shoji, Dennis C. Liotta, James P. Snyder
Bioorganic & Medicinal Chemistry Letters 2009 19(23) pp: 6627-6631
Publication Date(Web):
DOI:10.1016/j.bmcl.2009.10.023
Co-reporter:Yesim A. Tahirovic ; Matthew Geballe ; Ewa Gruszecka-Kowalik ; Scott J. Myers ; Polina Lyuboslavsky ; Phuong Le ; Adam French ; Hasan Irier ; Woo-baeg Choi ; Keith Easterling ; Hongjie Yuan ; Lawrence J. Wilson ; Robert Kotloski ; James O. McNamara ; Raymond Dingledine ; Dennis C. Liotta ; Stephen F. Traynelis
Journal of Medicinal Chemistry 2008 Volume 51(Issue 18) pp:5506-5521
Publication Date(Web):August 23, 2008
DOI:10.1021/jm8002153
Enantiomeric propanolamines have been identified as a new class of NR2B-selective NMDA receptor antagonists. The most effective agents are biaryl structures, synthesized in six steps with overall yields ranging from 11−64%. The compounds are potent and selective inhibitors of NR2B-containing recombinant NMDA receptors with IC50 values between 30−100 nM. Potency is strongly controlled by substitution on both rings and the centrally located amine nitrogen. SAR analysis suggests that well-balanced polarity and chain-length factors provide the greatest inhibitory potency. Structural comparisons based on 3D shape analysis and electrostatic complementarity support this conclusion. The antagonists are neuroprotective in both in vitro and in vivo models of ischemic cell death. In addition, some compounds exhibit anticonvulsant properties. Unlike earlier generation NMDA receptor antagonists and some NR2B-selective antagonists, the present series of propanolamines does not cause increased locomotion in rodents. Thus, the NR2B-selective antagonists exhibit a range of therapeutically interesting properties.
Co-reporter:Aiming Sun ; Jeong-Joong Yoon ; Yan Yin ; Andrew Prussia ; Yutao Yang ; Jaeki Min ; Richard K. Plemper
Journal of Medicinal Chemistry 2008 Volume 51(Issue 13) pp:3731-3741
Publication Date(Web):June 5, 2008
DOI:10.1021/jm701239a
Measles virus (MV) is one of the most infectious pathogens known. In spite of the existence of a vaccine, approximately 350000 deaths/year result from MV or associated complications. Antimeasles compounds could conceivably diminish these statistics and provide a therapy that complements vaccine treatment. We recently described a high-throughput screening hit compound 1 (16677) against MV-infected cells with the capacity to eliminate viral reproduction at 250 nM by inhibiting the action of the virus’s RNA-dependent RNA polymerase complex (RdRp). The compound, 1-methyl-3-(trifluoromethyl)-N-[4-sulfonylphenyl]-1H-pyrazole-5-carboxamide, 1 carries a critical CF3 moiety on the 1,2-pyrazole ring. Elaborating on the preliminary structure−activity (SAR) study, the present work presents the synthesis and SAR of a much broader range of low nanomolar nonpeptidic MV inhibitors and speculates on the role of the CF3 functionality.
Co-reporter:Andrew J. Prussia, Richard K. Plemper and James P. Snyder
Biochemistry 2008 Volume 47(Issue 51) pp:13573-13583
Publication Date(Web):November 26, 2008
DOI:10.1021/bi801513p
Previously, we developed a panel of nonpeptidic compounds specifically preventing fusion of the measles virus (MV) with target cells at IC50 values of 0.6−3 μM. Mutations in the MV fusion protein (MV F) that render resistance to these blockers were described. The structural basis for both inhibition and resistance was unclear in the earlier work because of the availability of a structural model for only the postfusion conformation of MV F. We have now developed structural models for both pre- and postfusion conformers of the latter protein trimer. The models allow investigation of the large-scale conformational changes occurring in the MV fusion machinery and, in conjunction with antisera binding studies, provide a rationale for how inhibitors may arrest a conformational intermediate by interfering with the formation of interactions between the heptad repeat B (HR-B) linker and DIII domains. The models also show that resistance to inhibition can be explained by a predicted destabilizing effect of the mutations on the HR-B domain within the trimeric prefusion structure. This viewpoint is supported by the temperature-dependent differential fusion activities of MV F variants harboring these mutations.
Co-reporter:Yan Liang, Sai Venkatesh Pingali, Ashutosh S. Jogalekar, James P. Snyder, Pappannan Thiyagarajan and David G. Lynn
Biochemistry 2008 Volume 47(Issue 38) pp:
Publication Date(Web):August 30, 2008
DOI:10.1021/bi801081c
Amino acid cross-strand pairing interactions along a β-sheet surface have been implicated in protein β-structural assembly and stability, yet the relative contributions have been difficult to evaluate directly. Here we develop the central core sequence of the Aβ peptide associated with Alzheimer’s disease, Aβ(16−22), as an experimental system for evaluating these interactions. The peptide allows for internal comparisons between electrostatic and steric interactions within the β-sheet and an evaluation of these cross-strand pair contributions to β-sheet registry. A morphological transition from fibers to hollow nanotubes arises from changes in β-sheet surface complementarity and provides a convenient indicator of the β-strand strand registry. The intrinsic β-sequence and pair correlations are critical to regulate secondary assembly. These studies provide evidence for a critical desolvation step that is not present in most models of the nucleation-dependent pathway for amyloid assembly.
Co-reporter:Anlys Olivera, Terry W. Moore, Fang Hu, Andrew P. Brown, Aiming Sun, Dennis C. Liotta, James P. Snyder, Younghyoun Yoon, Hyunsuk Shim, Adam I. Marcus, Andrew H. Miller, Thaddeus W.W. Pace
International Immunopharmacology (February 2012) Volume 12(Issue 2) pp:368-377
Publication Date(Web):1 February 2012
DOI:10.1016/j.intimp.2011.12.009
Nuclear factor kappa B (NF-κB) is a key signaling molecule in the elaboration of the inflammatory response. Data indicate that curcumin, a natural ingredient of the curry spice turmeric, acts as a NF-κB inhibitor and exhibits both anti-inflammatory and anti-cancer properties. Curcumin analogs with enhanced activity on NF-κB and other inflammatory signaling pathways have been developed including the synthetic monoketone compound 3,5-Bis(2-fluorobenzylidene)-4-piperidone (EF24). 3,5-Bis(2-pyridinylmethylidene)-4-piperidone (EF31) is a structurally-related curcumin analog whose potency for NF-κB inhibition has yet to be determined. To examine the activity of EF31 compared to EF24 and curcumin, mouse RAW264.7 macrophages were treated with EF31, EF24, curcumin (1–100 μM) or vehicle (DMSO 1%) for 1 h. NF-κB pathway activity was assessed following treatment with lipopolysaccharide (LPS) (1 μg/mL). EF31 (IC50 ~ 5 μM) exhibited significantly more potent inhibition of LPS-induced NF-κB DNA binding compared to both EF24 (IC50 ~ 35 μM) and curcumin (IC50 > 50 μM). In addition, EF31 exhibited greater inhibition of NF-κB nuclear translocation as well as the induction of downstream inflammatory mediators including pro-inflammatory cytokine mRNA and protein (tumor necrosis factor-α, interleukin-1β, and interleukin-6). Regarding the mechanism of these effects on NF-κB, EF31 (IC50 ~ 1.92 μM) exhibited significantly greater inhibition of IκB kinase β compared to EF24 (IC50 ~ 131 μM). Finally, EF31 demonstrated potent toxicity in NF-κB-dependent cancer cell lines while having minimal and reversible toxicity in RAW264.7 macrophages. These data indicate that EF31 is a more potent inhibitor of NF-κB activity than either EF24 or curcumin while exhibiting both anti-inflammatory and anticancer activities. Thus, EF31 represents a promising curcumin analog for further therapeutic development.Highlights► EF31 is a potent inhibitor of Iκβ kinase, NF-κB translocation, and NF-κB DNA-binding activity in RAW264.7 macrophages. ► EF31 blocks the expression of pro-inflammatory cytokine mRNA and protein. ► EF31 inhibits MAPK transcription factors. ► EF31 exhibits potent anti-cancer activity in various cancer cell lines.
Co-reporter:Yanqi Chang, Thota Ganesh, John R. Horton, Astrid Spannhoff, ... Xiaodong Cheng
Journal of Molecular Biology (2 July 2010) Volume 400(Issue 1) pp:1-7
Publication Date(Web):2 July 2010
DOI:10.1016/j.jmb.2010.04.048
Dynamic histone lysine methylation involves the activities of modifying enzymes (writers), enzymes removing modifications (erasers), and readers of the histone code. One common feature of these activities is the recognition of lysines in methylated and unmethylated states, whether they are substrates, reaction products, or binding partners. We applied the concept of adding a lysine mimic to an established inhibitor (BIX-01294) of histone H3 lysine 9 methyltransferases G9a and G9a-like protein by including a 5-aminopentyloxy moiety, which is inserted into the target lysine-binding channel and becomes methylated by G9a-like protein, albeit slowly. The compound enhances its potency in vitro and reduces cell toxicity in vivo. We suggest that adding a lysine or methyl-lysine mimic should be considered in the design of small-molecule inhibitors for other methyl-lysine writers, erasers, and readers.
Co-reporter:Qiao-Hong Chen, Thota Ganesh, Yi Jiang, Abhijit Banerjee, Shubhada Sharma, Susan Bane, James P. Snyder and David G. I. Kingston
Chemical Communications 2010 - vol. 46(Issue 12) pp:NaN2021-2021
Publication Date(Web):2010/02/09
DOI:10.1039/B926174E
An unusual reaction with Grubbs' catalyst during the synthesis of bridged epothilones yielded five-membered internal lactones instead of the expected metathesis products. Three of the lactones have activities comparable to epothilone D.
Co-reporter:Mi-Sun Kim, Lauriane A. Buisson, Dean A. Heathcote, Haipeng Hu, D. Christopher Braddock, Anthony G. M. Barrett, Philip G. Ashton-Rickardt and James P. Snyder
Organic & Biomolecular Chemistry 2014 - vol. 12(Issue 44) pp:NaN8965-8965
Publication Date(Web):2014/09/23
DOI:10.1039/C4OB01874E
A structure-based design campaign for non-covalent small molecule inhibitors of human granzyme B was carried out by means of a virtual screening strategy employing three constraints and probe site-mapping with FTMAP to identify ligand “hot spots”. In addition, new scaffolds of diverse structures were subsequently explored with ROCS shape-based superposition methods, following by Glide SP docking, induced fit docking and analysis of QikProp molecular properties. Novel classes of moderately active small molecule blockers (≥25 μM IC50 values) from commercially available libraries were identified, and three novel scaffolds have been synthesized by multi-step procedures. Furthermore, we provide an example of a comprehensive structure-based drug discovery approach to non-covalent inhibitors that relies on the X-ray structure of a covalently bound ligand and suggest that the design path may be compromised by alternative and unknown binding poses.
PIPERIDINE, 4-[4-(PHENYLMETHYL)-1H-PYRAZOL-3-YL]-
(S)-N1-((1H-Benzo[d]imidazol-2-yl)methyl)-N1-(5,6,7,8-tetrahydroquinolin-8-yl)butane-1,4-diamine
Cyclooxygenase 2
1,1'-[1,4-Phenylenebis(methylene)]bis[1,4,8,11-tetraazacyclotetradecane]