Network


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

Hotspot


Dive into the research topics where Jay A. Grobler is active.

Publication


Featured researches published by Jay A. Grobler.


Proceedings of the National Academy of Sciences of the United States of America | 2002

Diketo acid inhibitor mechanism and HIV-1 integrase: Implications for metal binding in the active site of phosphotransferase enzymes

Jay A. Grobler; Kara A. Stillmock; Binghua Hu; Marc Witmer; Peter J. Felock; Amy S. Espeseth; Abigail Wolfe; Melissa S. Egbertson; Michele Bourgeois; Jeffrey Y. Melamed; John S. Wai; Steve Young; Joseph P. Vacca; Daria J. Hazuda

The process of integrating the reverse-transcribed HIV-1 DNA into the host chromosomal DNA is catalyzed by the virally encoded enzyme integrase (IN). Integration requires two metal-dependent reactions, 3′ end processing and strand transfer. Compounds that contain a diketo acid moiety have been shown to selectively inhibit the strand transfer reaction of IN in vitro and in infected cells and are effective as inhibitors of HIV-1 replication. To characterize the molecular basis of inhibition, we used functional assays and binding assays to evaluate a series of structurally related analogs. These studies focused on investigating the role of the conserved carboxylate and metal binding. We demonstrate that an acidic moiety such as a carboxylate or isosteric heterocycle is not required for binding to the enzyme complex but is essential for inhibition and confers distinct metal-dependent properties on the inhibitor. Binding requires divalent metal and resistance is metal dependent with active site mutants displaying resistance only when the enzymes are evaluated in the context of Mg2+. The mechanism of action of these inhibitors is therefore likely a consequence of the interaction between the acid moiety and metal ion(s) in the IN active site, resulting in a functional sequestration of the critical metal cofactor(s). These studies thus have implications for modeling active site inhibitors of IN, designing and evaluating analogs with improved efficacy, and identifying inhibitors of other metal-dependent phosphotransferases.


Antimicrobial Agents and Chemotherapy | 2004

A 7-Deaza-Adenosine Analog Is a Potent and Selective Inhibitor of Hepatitis C Virus Replication with Excellent Pharmacokinetic Properties

David B. Olsen; Anne B. Eldrup; Linda Bartholomew; Balkrishen Bhat; Michele Bosserman; Alessandra Ceccacci; Lawrence F. Colwell; John F. Fay; Osvaldo A. Flores; Krista Getty; Jay A. Grobler; Robert L. Lafemina; Eric J. Markel; Giovanni Migliaccio; Marija Prhavc; Mark Stahlhut; Joanne E. Tomassini; Malcolm Maccoss; Daria J. Hazuda; Steven S. Carroll

ABSTRACT Improved treatments for chronic hepatitis C virus (HCV) infection are needed due to the suboptimal response rates and deleterious side effects associated with current treatment options. The triphosphates of 2′-C-methyl-adenosine and 2′-C-methyl-guanosine were previously shown to be potent inhibitors of the HCV RNA-dependent RNA polymerase (RdRp) that is responsible for the replication of viral RNA in cells. Here we demonstrate that the inclusion of a 7-deaza modification in a series of purine nucleoside triphosphates results in an increase in inhibitory potency against the HCV RdRp and improved pharmacokinetic properties. Notably, incorporation of the 7-deaza modification into 2′-C-methyl-adenosine results in an inhibitor with a 20-fold-increased potency as the 5′-triphosphate in HCV RdRp assays while maintaining the inhibitory potency of the nucleoside in the bicistronic HCV replicon and with reduced cellular toxicity. In contrast, while 7-deaza-2′-C-methyl-GTP also displays enhanced inhibitory potency in enzyme assays, due to poor cellular penetration and/or metabolism, the nucleoside does not inhibit replication of a bicistronic HCV replicon in cell culture. 7-Deaza-2′-C-methyl-adenosine displays promising in vivo pharmacokinetics in three animal species, as well as an acute oral lethal dose in excess of 2,000 mg/kg of body weight in mice. Taken together, these data demonstrate that 7-deaza-2′-C-methyl-adenosine is an attractive candidate for further investigation as a potential treatment for HCV infection.


Antimicrobial Agents and Chemotherapy | 2005

Replication Fitness and NS5B Drug Sensitivity of Diverse Hepatitis C Virus Isolates Characterized by Using a Transient Replication Assay

Steven W. Ludmerer; Donald J. Graham; Evelyn Boots; Edward M. Murray; Amy L. Simcoe; Eric J. Markel; Jay A. Grobler; Osvaldo A. Flores; David B. Olsen; Daria J. Hazuda; Robert L. Lafemina

ABSTRACT The innate genetic variability characteristic of chronic hepatitis C virus (HCV) infection makes drug resistance a concern in the clinical development of HCV inhibitors. To address this, a transient replication assay was developed to evaluate the replication fitness and the drug sensitivity of NS5B sequences isolated from the sera of patients with chronic HCV infection. This novel assay directly compares replication between NS5B isolates, thus bypassing the potential sequence and metabolic differences which may arise with independent replicon cell lines. Patient-derived NS5B sequences were similar to those of the established HCV genotypes, but isolates from each patient shared genetic variability specific to that patient, with additional genetic variability observed across the individual isolates. Every sample provided functional NS5B isolates which supported subgenomic replication, frequently to levels comparable to that of laboratory-optimized replicons. All isolates were equivalently sensitive to an active-site nucleoside inhibitor, but the sensitivities to a panel of nonnucleoside inhibitors which targeted three distinct sites on NS5B varied among the isolates. In con1, the original laboratory-optimized replicon, the NS5B S282T substitution confers resistance to the nucleoside inhibitor but impairs replication. This substitution was engineered into both genotype 1a and genotype 1b isolates. Replication was severely debilitated, demonstrating that no compensatory residues were encoded within these genetically diverse sequences to increase the replication fitness of the mutated replicons. This work describes a transient replicon-based assay that can support the clinical development of compounds which target NS5B and demonstrates its utility by examining several patient-derived NS5B isolates for replication fitness and differential sensitivity to NS5B inhibitors.


Journal of Biological Chemistry | 2007

HIV-1 reverse transcriptase plus-strand initiation exhibits preferential sensitivity to non-nucleoside reverse transcriptase inhibitors in vitro.

Jay A. Grobler; Geetha Dornadula; Michele R. Rice; Amy L. Simcoe; Daria J. Hazuda; Michael D. Miller

Non-nucleoside reverse transcriptase inhibitors (NNRTIs) are highly specific and potent allosteric inhibitors of human immunodeficiency virus type 1 (HIV-1) reverse transcriptase. NNRTIs inhibit reverse transcription in a substrate length-dependent manner in biochemical assays and in cell-based HIV-1 replication assays, suggesting a stochastic inhibitory mechanism. Surprisingly, we observed that NNRTIs potently inhibited plus-strand initiation in vitro under conditions in which little or no inhibition of minus-strand DNA synthesis was observed. In assays that recapitulated the initiation of plus-strand DNA synthesis, greater inhibition was observed with an RNA PPT primer than with a DNA primer of corresponding sequence and with wild-type reverse transcriptase but not with NNRTI-resistant enzymes. Structural elements that dictate sensitivity to NNRTIs were revealed using modified plus-strand initiation substrates. The data presented here suggest that specific inhibition of plus-strand initiation may be an important mechanism by which NNRTIs block HIV-1 replication.


Journal of Virology | 2010

Structural basis for the inhibition of RNase H activity of HIV-1 reverse transcriptase by RNase H active site-directed inhibitors.

Hua-Poo Su; Youwei Yan; G. Sridhar Prasad; Robert F. Smith; Christopher L. Daniels; Pravien Abeywickrema; John C. Reid; H. Marie Loughran; Maria Kornienko; Sujata Sharma; Jay A. Grobler; Bei Xu; Vinod V. Sardana; Timothy J. Allison; Peter D. Williams; Paul L. Darke; Daria J. Hazuda; Sanjeev Munshi

ABSTRACT HIV/AIDS continues to be a menace to public health. Several drugs currently on the market have successfully improved the ability to manage the viral burden in infected patients. However, new drugs are needed to combat the rapid emergence of mutated forms of the virus that are resistant to existing therapies. Currently, approved drugs target three of the four major enzyme activities encoded by the virus that are critical to the HIV life cycle. Although a number of inhibitors of HIV RNase H activity have been reported, few inhibit by directly engaging the RNase H active site. Here, we describe structures of naphthyridinone-containing inhibitors bound to the RNase H active site. This class of compounds binds to the active site via two metal ions that are coordinated by catalytic site residues, D443, E478, D498, and D549. The directionality of the naphthyridinone pharmacophore is restricted by the ordering of D549 and H539 in the RNase H domain. In addition, one of the naphthyridinone-based compounds was found to bind at a second site close to the polymerase active site and non-nucleoside/nucleotide inhibitor sites in a metal-independent manner. Further characterization, using fluorescence-based thermal denaturation and a crystal structure of the isolated RNase H domain reveals that this compound can also bind the RNase H site and retains the metal-dependent binding mode of this class of molecules. These structures provide a means for structurally guided design of novel RNase H inhibitors.


Antimicrobial Agents and Chemotherapy | 2010

Distinct Mutation Pathways of Non-Subtype B HIV-1 during In Vitro Resistance Selection with Nonnucleoside Reverse Transcriptase Inhibitors

Ming-Tain Lai; Meiqing Lu; Peter J. Felock; Renee Hrin; Ying-Jie Wang; Youwei Yan; Sanjeev Munshi; Georgia B Mcgaughey; Robert M. Tynebor; Thomas J. Tucker; Theresa M. Williams; Jay A. Grobler; Daria J. Hazuda; Philip M. McKenna; Michael D. Miller

ABSTRACT Studies were conducted to investigate mutation pathways among subtypes A, B, and C of human immunodeficiency virus type 1 (HIV-1) during resistance selection with nonnucleoside reverse transcriptase inhibitors (NNRTIs) in cell culture under low-multiplicity of infection (MOI) conditions. The results showed that distinct pathways were selected by different virus subtypes under increasing selective pressure of NNRTIs. F227C and Y181C were the major mutations selected by MK-4965 in subtype A and C viruses during resistance selection. With efavirenz (EFV), F227C and V106M were the major mutations responsible for viral breakthrough in subtype A viruses, whereas a single pathway (G190A/V106M) accounted for mutation development in subtype C viruses. Y181C was the dominant mutation in the resistance selection with etravirine (ETV) in subtype A, and E138K/H221Y were the mutations detected in the breakthrough viruses from subtype C viruses with ETV. In subtype B viruses, on the other hand, known NNRTI-associated mutations (e.g., Y181C, P236L, L100I, V179D, and K103N) were selected by the NNRTIs. The susceptibility of the subtype A and B mutant viruses to NNRTIs was determined in order to gain insight into the potential mechanisms of mutation development. Collectively, these results suggest that minor differences may exist in conformation of the residues within the NNRTI binding pocket (NNRTIBP) of reverse transcriptase (RT) among the three subtypes of viruses. Thus, the interactions between NNRTIs and the residues in the NNRTIBPs of different subtypes may not be identical, leading to distinct mutation pathways during resistance selection in cell culture.


Journal of Medicinal Chemistry | 2015

Correction to Discovery of 2-Pyridinone Aminals: A Prodrug Strategy to Advance a Second Generation of HIV-1 Integrase Strand Transfer Inhibitors.

Izzat T. Raheem; Abbas Walji; Daniel Klein; John M. Sanders; David Powell; Pravien Abeywickrema; Guillaume Barbe; Amrith Bennet; Karla G. Childers; Melodie Christensen; Sophie Dorothee Clas; David C. Dubost; Mark W. Embrey; Jay A. Grobler; Michael J. Hafey; Timothy J. Hartingh; Daria J. Hazuda; Jeffrey T. Kuethe; Jamie M. McCabe Dunn; Michael D. Miller; Keith P. Moore; Andrew Nolting; Natasa Pajkovic; Sangita B. Patel; Zuihui Peng; Vanessa Rada; Paul Rearden; John D. Schreier; John T. Sisko; Thomas G. Steele

The search for new molecular constructs that resemble the critical two-metal binding pharmacophore required for HIV integrase strand transfer inhibition represents a vibrant area of research within drug discovery. Here we present the discovery of a new class of HIV integrase strand transfer inhibitors based on the 2-pyridinone core of MK-0536. These efforts led to the identification of two lead compounds with excellent antiviral activity and preclinical pharmacokinetic profiles to support a once-daily human dose prediction. Dose escalating PK studies in dog revealed significant issues with limited oral absorption and required an innovative prodrug strategy to enhance the high-dose plasma exposures of the parent molecules.


Nucleic Acids Research | 2012

Bulged DNA substrates for identifying poxvirus resolvase inhibitors

Matthew J. Culyba; Young Hwang; Sana Attar; Peter B. Madrid; James Bupp; Donna M. Huryn; Luis Sanchez; Jay A. Grobler; Michael Miller; Frederic D. Bushman

Resolvase enzymes that cleave DNA four-way (Holliday) junctions are required for poxvirus replication, but clinically useful inhibitors have not been developed. Here, we report an assay for resolvase cleavage activity based on fluorescence polarization (FP) for high-throughput screening and mechanistic studies. Initial analysis showed that cleavage of a fluorescently labeled Holliday junction substrate did not yield an appreciable change in FP, probably because the cleavage product did not have sufficiently increased mobility to yield a strong FP signal. Iterative optimization yielded a substrate with an off-center DNA bulge, which after cleavage released a labeled short stand and yielded a greatly reduced FP signal. Using this assay, 133 000 compounds were screened, identifying 1-hydroxy-1,8-naphthyridin-2(1H)-one compounds as inhibitors. Structure-activity studies revealed functional parallels to Food and Drug Administration (FDA)-approved drugs targeting the related human immunodeficiency virus integrase enzyme. Some 1-hydroxy-1,8-naphthyridin-2(1H)-one compounds showed anti-poxvirus activity.


ACS Chemical Biology | 2017

Discovery of a Distinct Chemical and Mechanistic Class of Allosteric HIV-1 Integrase Inhibitors with Antiretroviral Activity

Christine Burlein; Cheng Wang; Min Xu; Triveni Bhatt; Mark Stahlhut; Yangsi Ou; Gregory C. Adam; Jeffrey Heath; Daniel Klein; John M. Sanders; Kartik Narayan; Pravien Abeywickrema; Mee Ra Heo; Steven S. Carroll; Jay A. Grobler; Sujata Sharma; Tracy L. Diamond; Antonella Converso; Daniel Krosky

Allosteric integrase inhibitors (ALLINIs) bind to the lens epithelial-derived growth factor (LEDGF) pocket on HIV-1 integrase (IN) and possess potent antiviral effects. Rather than blocking proviral integration, ALLINIs trigger IN conformational changes that have catastrophic effects on viral maturation, rendering the virions assembled in the presence of ALLINIs noninfectious. A high-throughput screen for compounds that disrupt the IN·LEDGF interaction was executed, and extensive triage led to the identification of a t-butylsulfonamide series, as exemplified by 1. The chemical, biochemical, and virological characterization of this series revealed that 1 and its analogs produce an ALLINI-like phenotype through engagement of IN sites distinct from the LEDGF pocket. Key to demonstrating target engagement and differentiating this new series from the existing ALLINIs was the development of a fluorescence polarization probe of IN (FLIPPIN) based on the t-butylsulfonamide series. These findings further solidify the late antiviral mechanism of ALLINIs and point toward opportunities to develop structurally and mechanistically novel antiretroviral agents with unique resistance patterns.


Science | 2000

Inhibitors of strand transfer that prevent integration and inhibit HIV-1 replication in cells

Daria J. Hazuda; Peter J. Felock; Marc Witmer; Abigail Wolfe; Kara A. Stillmock; Jay A. Grobler; Amy S. Espeseth; Lori Gabryelski; William A. Schleif; Carol Blau; Michael D. Miller

Collaboration


Dive into the Jay A. Grobler's collaboration.

Top Co-Authors

Avatar

Peter J. Felock

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Marc Witmer

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Abigail Wolfe

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Eric J. Markel

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Kara A. Stillmock

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Mark W. Embrey

United States Military Academy

View shared research outputs
Researchain Logo
Decentralizing Knowledge