Network


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

Hotspot


Dive into the research topics where Lawrence B. Snyder is active.

Publication


Featured researches published by Lawrence B. Snyder.


Nature | 2010

Chemical genetics strategy identifies an HCV NS5A inhibitor with a potent clinical effect

Min Gao; Richard E. Nettles; Makonen Belema; Lawrence B. Snyder; Van N. Nguyen; Robert A. Fridell; Michael H. Serrano-Wu; David R. Langley; Jin-Hua Sun; Donald R. O'Boyle; Julie A. Lemm; Chunfu Wang; Jay O. Knipe; Caly Chien; Richard J. Colonno; Dennis M. Grasela; Nicholas A. Meanwell; Lawrence G. Hamann

The worldwide prevalence of chronic hepatitis C virus (HCV) infection is estimated to be approaching 200 million people. Current therapy relies upon a combination of pegylated interferon-α and ribavirin, a poorly tolerated regimen typically associated with less than 50% sustained virological response rate in those infected with genotype 1 virus. The development of direct-acting antiviral agents to treat HCV has focused predominantly on inhibitors of the viral enzymes NS3 protease and the RNA-dependent RNA polymerase NS5B. Here we describe the profile of BMS-790052, a small molecule inhibitor of the HCV NS5A protein that exhibits picomolar half-maximum effective concentrations (EC50) towards replicons expressing a broad range of HCV genotypes and the JFH-1 genotype 2a infectious virus in cell culture. In a phase I clinical trial in patients chronically infected with HCV, administration of a single 100-mg dose of BMS-790052 was associated with a 3.3 log10 reduction in mean viral load measured 24 h post-dose that was sustained for an additional 120 h in two patients infected with genotype 1b virus. Genotypic analysis of samples taken at baseline, 24 and 144 h post-dose revealed that the major HCV variants observed had substitutions at amino-acid positions identified using the in vitro replicon system. These results provide the first clinical validation of an inhibitor of HCV NS5A, a protein with no known enzymatic function, as an approach to the suppression of virus replication that offers potential as part of a therapeutic regimen based on combinations of HCV inhibitors.


Journal of Virology | 2010

Identification of Hepatitis C Virus NS5A Inhibitors

Julie A. Lemm; Donald R. O'Boyle; Mengping Liu; Peter T. Nower; Richard J. Colonno; Milind Deshpande; Lawrence B. Snyder; Scott Martin; Denis R. St. Laurent; Michael H. Serrano-Wu; Jeffrey L. Romine; Nicholas A. Meanwell; Min Gao

ABSTRACT Using a cell-based replicon screen, we identified a class of compounds with a thiazolidinone core structure as inhibitors of hepatitis C virus (HCV) replication. The concentration of one such compound, BMS-824, that resulted in a 50% inhibition of HCV replicon replication was ∼5 nM, with a therapeutic index of >10,000. The compound showed good specificity for HCV, as it was not active against several other RNA and DNA viruses. Replicon cells resistant to BMS-824 were isolated, and mutations were identified. A combination of amino acid substitutions of leucine to valine at residue 31 (L31V) and glutamine to leucine at residue 54 (Q54L) in NS5A conferred resistance to this chemotype, as did a single substitution of tyrosine to histidine at amino acid 93 (Y93H) in NS5A. To further explore the region(s) of NS5A involved in inhibitor sensitivity, genotype-specific NS5A inhibitors were used to evaluate a series of genotype 1a/1b hybrid replicons. Our results showed that, consistent with resistance mapping, the inhibitor sensitivity domain also mapped to the N terminus of NS5A, but it could be distinguished from the key resistance sites. In addition, we demonstrated that NS5A inhibitors, as well as an active-site inhibitor that specifically binds NS3 protease, could block the hyperphosphorylation of NS5A, which is believed to play an essential role in the viral life cycle. Clinical proof of concept has recently been achieved with derivatives of these NS5A inhibitors, indicating that small molecules targeting a nontraditional viral protein like NS5A, without any known enzymatic activity, can also have profound antiviral effects on HCV-infected subjects.


ACS Medicinal Chemistry Letters | 2011

Inhibitors of HCV NS5A: From Iminothiazolidinones to Symmetrical Stilbenes

Jeffrey L. Romine; Denis R. St. Laurent; John E. Leet; Scott Martin; Michael H. Serrano-Wu; Fukang Yang; Min Gao; Donald R. O’Boyle; Julie A. Lemm; Jin-Hua Sun; Peter T. Nower; Milind Deshpande; Nicholas A. Meanwell; Lawrence B. Snyder

The iminothiazolidinone BMS-858 (2) was identified as a specific inhibitor of HCV replication in a genotype 1b replicon assay via a high-throughput screening campaign. A more potent analogue, BMS-824 (18), was used in resistance mapping studies, which revealed that inhibitory activity was related to disrupting the function of the HCV nonstructural protein 5A. Despite the development of coherent and interpretable SAR, it was subsequently discovered that in DMSO 18 underwent an oxidation and structural rearrangement to afford the thiohydantoin 47, a compound with reduced HCV inhibitory activity. However, HPLC bioassay fractionation studies performed after incubation of 18 in assay media led to the identification of fractions containing a dimeric species 48 that exhibited potent antiviral activity. Excision of the key elements hypothesized to be responsible for antiviral activity based on SAR observations reduced 48 to a simplified, symmetrical, pharmacophore realized most effectively with the stilbene 55, a compound that demonstrated potent inhibition of HCV in a genotype 1b replicon with an EC50 = 86 pM.


Bioorganic & Medicinal Chemistry Letters | 2003

Discovery of the first antibacterial small molecule inhibitors of MurB

Joanne J. Bronson; Kenneth DenBleyker; Paul Falk; Robert A. Mate; Hsu-Tso Ho; Michael J. Pucci; Lawrence B. Snyder

A series of imidazolinone analogues was synthesized and shown to possess potent MurB inhibitory as well as good antibacterial activity.


Journal of General Virology | 2011

The effects of NS5A inhibitors on NS5A phosphorylation, polyprotein processing and localization

Dike Qiu; Julie A. Lemm; Donald R. O’Boyle; Jin-Hua Sun; Peter T. Nower; Van N. Nguyen; Lawrence G. Hamann; Lawrence B. Snyder; Daniel H. Deon; Edward H. Ruediger; Nicholas A. Meanwell; Makonen Belema; Min Gao; Robert A. Fridell

Hepatitis C virus (HCV) non-structural protein 5A (NS5A) is a multi-functional protein that is expressed in basally phosphorylated (p56) and in hyperphosphorylated (p58) forms. NS5A phosphorylation has been implicated in regulating multiple aspects of HCV replication. We recently reported the identification of a class of compounds that potently inhibit HCV RNA replication by targeting NS5A. Although the precise mechanism of inhibition of these compounds is not well understood, one activity that has been described is their ability to block expression of the hyperphosphorylated form of NS5A. Here, we report that an NS5A inhibitor impaired hyperphosphorylation without affecting basal phosphorylation at the C-terminal region of NS5A. This inhibitor activity did not require NS5A domains II and III and was distinct from that of a cellular kinase inhibitor that also blocked NS5A hyperphosphorylation, results that are consistent with an inhibitor-binding site within the N-terminal region of NS5A. In addition, we observed that an NS5A inhibitor promoted the accumulation of an HCV polyprotein intermediate, suggesting that inhibitor binding to NS5A may occur prior to the completion of polyprotein processing. Finally, we observed that NS5A p56 and p58 separated into different membrane fractions during discontinuous sucrose gradient centrifugation, consistent with these NS5A phosphoforms performing distinct replication functions. The p58 localization pattern was disrupted by an NS5A inhibitor. Collectively, our results suggest that NS5A inhibitors probably impact several aspects of HCV expression and regulation. These findings may help to explain the exceptional potency of this class of HCV replication complex inhibitors.


Antimicrobial Agents and Chemotherapy | 2011

Discovery of Potent Hepatitis C Virus NS5A Inhibitors with Dimeric Structures

Julie A. Lemm; John E. Leet; Donald R. O'Boyle; Jeffrey L. Romine; Xiaohua Stella Huang; Daniel R. Schroeder; Jeffrey Alberts; Joseph L. Cantone; Jin-Hua Sun; Peter T. Nower; Scott W. Martin; Michael H. Serrano-Wu; Nicholas A. Meanwell; Lawrence B. Snyder; Min Gao

ABSTRACT The exceptional in vitro potency of the hepatitis C virus (HCV) NS5A inhibitor BMS-790052 has translated into an in vivo effect in proof-of-concept clinical trials. Although the 50% effective concentration (EC50) of the initial lead, the thiazolidinone BMS-824, was ∼10 nM in the replicon assay, it underwent transformation to other inhibitory species after incubation in cell culture medium. The biological profile of BMS-824, including the EC50, the drug concentration required to reduce cell growth by 50% (CC50), and the resistance profile, however, remained unchanged, triggering an investigation to identify the biologically active species. High-performance liquid chromatography (HPLC) biogram fractionation of a sample of BMS-824 incubated in medium revealed that the most active fractions could readily be separated from the parental compound and retained the biological profile of BMS-824. From mass spectral and nuclear magnetic resonance data, the active species was determined to be a dimer of BMS-824 derived from an intermolecular radical-mediated reaction of the parent compound. Based upon an analysis of the structural elements of the dimer deemed necessary for anti-HCV activity, the stilbene derivative BMS-346 was synthesized. This compound exhibited excellent anti-HCV activity and showed a resistance profile similar to that of BMS-824, with changes in compound sensitivity mapped to the N terminus of NS5A. The N terminus of NS5A has been crystallized as a dimer, complementing the symmetry of BMS-346 and allowing a potential mode of inhibition of NS5A to be discussed. Identification of the stable, active pharmacophore associated with these NS5A inhibitors provided the foundation for the design of more potent inhibitors with broad genotype inhibition. This culminated in the identification of BMS-790052, a compound that preserves the symmetry discovered with BMS-346.


Journal of Medicinal Chemistry | 2014

Hepatitis C virus NS5A replication complex inhibitors: the discovery of daclatasvir.

Makonen Belema; Van N. Nguyen; Carol Bachand; Dan H. Deon; Jason Goodrich; Clint A. James; Rico Lavoie; Omar D. Lopez; Alain Martel; Jeffrey L. Romine; Edward H. Ruediger; Lawrence B. Snyder; Denis R. St. Laurent; Fukang Yang; Juliang Zhu; Henry S. Wong; David R. Langley; Stephen P. Adams; Glenn H. Cantor; Anjaneya Chimalakonda; Aberra Fura; Benjamin M. Johnson; Jay O. Knipe; Dawn D. Parker; Kenneth S. Santone; Robert A. Fridell; Julie A. Lemm; Donald R. O’Boyle; Richard J. Colonno; Min Gao

The biphenyl derivatives 2 and 3 are prototypes of a novel class of NS5A replication complex inhibitors that demonstrate high inhibitory potency toward a panel of clinically relevant HCV strains encompassing genotypes 1-6. However, these compounds exhibit poor systemic exposure in rat pharmacokinetic studies after oral dosing. The structure-activity relationship investigations that improved the exposure properties of the parent bis-phenylimidazole chemotype, culminating in the identification of the highly potent NS5A replication complex inhibitor daclatasvir (33) are described. An element critical to success was the realization that the arylglycine cap of 2 could be replaced with an alkylglycine derivative and still maintain the high inhibitory potency of the series if accompanied with a stereoinversion, a finding that enabled a rapid optimization of exposure properties. Compound 33 had EC50 values of 50 and 9 pM toward genotype-1a and -1b replicons, respectively, and oral bioavailabilities of 38-108% in preclinical species. Compound 33 provided clinical proof-of-concept for the NS5A replication complex inhibitor class, and regulatory approval to market it with the NS3/4A protease inhibitor asunaprevir for the treatment of HCV genotype-1b infection has recently been sought in Japan.


Virology | 2013

Characterizations of HCV NS5A replication complex inhibitors

Donald R. O'Boyle; Jin-Hua Sun; Peter T. Nower; Julie A. Lemm; Robert A. Fridell; Chunfu Wang; Jeffrey L. Romine; Makonen Belema; Van N. Nguyen; Denis R. St. Laurent; Michael H. Serrano-Wu; Lawrence B. Snyder; Nicholas A. Meanwell; David R. Langley; Min Gao

The hepatitis C virus NS5A protein is an established and clinically validated target for antiviral intervention by small molecules. Characterizations are presented of compounds identified as potent inhibitors of HCV replication to provide insight into structural elements that interact with the NS5A protein. UV-activated cross linking and affinity isolation was performed with one series to probe the physical interaction between the inhibitors and the NS5A protein expressed in HCV replicon cells. Resistance mapping with the second series was used to determine the functional impact of specific inhibitor subdomains on the interaction with NS5A. The data provide evidence for a direct high-affinity interaction between these inhibitors and the NS5A protein, with the interaction dependent on inhibitor stereochemistry. The functional data supports a model of inhibition that implicates inhibitor binding by covalently combining distinct pharmacophores across an NS5A dimer interface to achieve maximal inhibition of HCV replication.


Journal of Medicinal Chemistry | 2014

HCV NS5A Replication Complex Inhibitors. Part 4.1 Optimization for Genotype 1a Replicon Inhibitory Activity

Denis R. St. Laurent; Michael H. Serrano-Wu; Makonen Belema; Min Ding; Hua Fang; Min Gao; Jason Goodrich; Rudolph G. Krause; Julie A. Lemm; Mengping Liu; Omar D. Lopez; Van N. Nguyen; Peter T. Nower; Donald R. O’Boyle; Bradley C. Pearce; Jeffrey L. Romine; Lourdes Valera; Jin-Hua Sun; Ying-Kai Wang; Fukang Yang; Xuejie Yang; Nicholas A. Meanwell; Lawrence B. Snyder

A series of symmetrical E-stilbene prolinamides that originated from the library-synthesized lead 3 was studied with respect to HCV genotype 1a (G-1a) and genotype 1b (G-1b) replicon inhibition and selectivity against BVDV and cytotoxicity. SAR emerging from an examination of the prolinamide cap region revealed 11 to be a selective HCV NS5A inhibitor exhibiting submicromolar potency against both G-1a and G-1b replicons. Additional structural refinements resulted in the identification of 30 as a potent, dual G-1a/1b HCV NS5A inhibitor.


Bioorganic & Medicinal Chemistry Letters | 2012

HCV NS5A replication complex inhibitors. Part 2: investigation of stilbene prolinamides.

Denis R. St. Laurent; Makonen Belema; Min Gao; Jason Goodrich; Ramesh Kakarla; Jay O. Knipe; Julie A. Lemm; Mengping Liu; Omar D. Lopez; Van N. Nguyen; Peter T. Nower; Donald R. O’Boyle; Yuping Qiu; Jeffrey L. Romine; Michael H. Serrano-Wu; Jin-Hua Sun; Lourdes Valera; Fukang Yang; Xuejie Yang; Nicholas A. Meanwell; Lawrence B. Snyder

In a previous disclosure,(1) we reported the dimerization of an iminothiazolidinone to form 1, a contributor to the observed inhibition of HCV genotype 1b replicon activity. The dimer was isolated via bioassay-guided fractionation experiments and shown to be a potent inhibitor of genotype 1b HCV replication for which resistance mapped to the NS5A protein. The elements responsible for governing HCV inhibitory activity were successfully captured in the structurally simplified stilbene prolinamide 2. We describe herein the early SAR and profiling associated with stilbene prolinamides that culminated in the identification of analogs with PK properties sufficient to warrant continued commitment to this chemotype. These studies represent the key initial steps toward the discovery of daclatasvir (BMS-790052), a compound that has demonstrated clinical proof-of-concept for inhibiting the NS5A replication complex in the treatment of HCV infection.

Collaboration


Dive into the Lawrence B. Snyder's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Min Gao

Bristol-Myers Squibb

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Omar D. Lopez

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge