Network


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

Hotspot


Dive into the research topics where Richard E. Nettles is active.

Publication


Featured researches published by Richard E. Nettles.


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 200u2009million 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 3.3u2009log10 reduction in mean viral load measured 24u2009h post-dose that was sustained for an additional 120u2009h in two patients infected with genotype 1b virus. Genotypic analysis of samples taken at baseline, 24 and 144u2009h 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.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Modeling shows that the NS5A inhibitor daclatasvir has two modes of action and yields a shorter estimate of the hepatitis C virus half-life

Jeremie Guedj; Harel Dahari; Libin Rong; Natasha D. Sansone; Richard E. Nettles; Scott J. Cotler; Thomas J. Layden; Susan L. Uprichard; Alan S. Perelson

The nonstructural 5A (NS5A) protein is a target for drug development against hepatitis C virus (HCV). Interestingly, the NS5A inhibitor daclatasvir (BMS-790052) caused a decrease in serum HCV RNA levels by about two orders of magnitude within 6 h of administration. However, NS5A has no known enzymatic functions, making it difficult to understand daclatasvir’s mode of action (MOA) and to estimate its antiviral effectiveness. Modeling viral kinetics during therapy has provided important insights into the MOA and effectiveness of a variety of anti-HCV agents. Here, we show that understanding the effects of daclatasvir in vivo requires a multiscale model that incorporates drug effects on the HCV intracellular lifecycle, and we validated this approach with in vitro HCV infection experiments. The model predicts that daclatasvir efficiently blocks two distinct stages of the viral lifecycle, namely viral RNA synthesis and virion assembly/secretion with mean effectiveness of 99% and 99.8%, respectively, and yields a more precise estimate of the serum HCV half-life, 45 min, i.e., around four times shorter than previous estimates. Intracellular HCV RNA in HCV-infected cells treated with daclatasvir and the HCV polymerase inhibitor NM107 showed a similar pattern of decline. However, daclatasvir treatment led to an immediate and rapid decline of extracellular HCV titers compared to a delayed (6–9 h) and slower decline with NM107, confirming an effect of daclatasvir on both viral replication and assembly/secretion. The multiscale modeling approach, validated with in vitro kinetic experiments, brings a unique conceptual framework for understanding the mechanism of action of a variety of agents in development for the treatment of HCV.


Hepatology | 2011

Genotypic and phenotypic analysis of variants resistant to hepatitis C virus nonstructural protein 5A replication complex inhibitor BMS‐790052 in Humans: In Vitro and In Vivo Correlations

Robert A. Fridell; Chunfu Wang; Jin-Hua Sun; Donald R. O'Boyle; Peter T. Nower; Lourdes Valera; Dike Qiu; Susan B. Roberts; Xin Huang; Bernadette Kienzle; Marc Bifano; Richard E. Nettles; Min Gao

The NS5A replication complex inhibitor, BMS‐790052, inhibits hepatitis C virus (HCV) replication with picomolar potency in preclinical assays. This potency translated in vivo to a substantial antiviral effect in a single‐ascending dose study and a 14‐day multiple‐ascending dose (MAD) monotherapy study. However, HCV RNA remained detectable in genotype 1a–infected patients at the end of the MAD study. In contrast, viral breakthrough was observed less often in patients infected with genotype 1b, and, in several patients, HCV RNA declined and remained below the level of quantitation (<25 IU/mL) through the duration of treatment. Here, we report on the results of the genotypic and phenotypic analyses of resistant variants in 24 genotype 1–infected patients who received BMS‐790052 (1, 10, 30, 60, and 100 mg, once‐daily or 30 mg twice‐daily) in the 14‐day MAD study. Sequence analysis was performed on viral complementary DNA isolated from serum specimens collected at baseline and days 1 (4, 8, and 12 hours), 2, 4, 7, and 14 postdosing. Analyses of the sequence variants (1) established a correlation between resistant variants emerging in vivo with BMS‐790052 treatment and those observed in the in vitro replicon system (major substitutions at residues 28, 30, 31, and 93 for genotype 1a and residues 31 and 93 for genotype 1b); (2) determined the prevalence of variants at baseline and the emergence of resistance at different times during dosing; and (3) revealed the resistance profile and replicative ability (i.e., fitness) of the variants. Conclusion: Although resistance emerged during monotherapy with BMS‐790052, the substantial anti‐HCV effect of this compound makes it an excellent candidate for effective combination therapy. (HEPATOLOGY 2011)


Hepatology | 2011

Multiple ascending dose study of BMS‐790052, a nonstructural protein 5A replication complex inhibitor, in patients infected with hepatitis C virus genotype 1

Richard E. Nettles; Min Gao; Marc Bifano; Ellen Chung; Anna Persson; Thomas Marbury; Ronald Goldwater; Michael Demicco; Maribel Rodriguez-Torres; Apinya Vutikullird; Ernesto Fuentes; Eric Lawitz; Juan Carlos Lopez-Talavera; Dennis M. Grasela

The antiviral activity, resistance profile, pharmacokinetics (PK), safety, and tolerability of BMS‐790052, a nonstructural protein 5A (NS5A) replication complex inhibitor, were evaluated in a double‐blind, placebo‐controlled, sequential panel, multiple ascending dose study. Thirty patients with chronic hepatitis C virus (HCV) genotype 1 infection were randomized to receive a 14‐day course of BMS‐790052 (1, 10, 30, 60, or 100 mg once daily or 30 mg twice daily) or placebo in a ratio of 4:1. The mean maximum decline from baseline in HCV RNA ranged from 2.8 to 4.1 log10 IU/mL; the placebo group showed no evidence of antiviral activity. Most patients experienced viral rebound on or before day 7 of treatment with BMS‐790052 monotherapy; viral rebound was associated with viral variants that had been previously implicated in resistance development in the in vitro replicon system. The PK profile was supportive of once‐daily dosing with median peak plasma concentrations at 1‐2 hours postdose and mean terminal half‐life of 12‐15 hours. Steady state was achieved following 3‐4 days of daily dosing. BMS‐790052 was well tolerated in all dose groups, with adverse events occurring with a similar frequency in BMS‐790052‐ and placebo‐treated groups. There were no clinically relevant changes in vital signs, laboratory, or electrocardiogram parameters. Conclusion: BMS‐7590052 is the first NS5A replication complex inhibitor with multiple dose proof‐of‐concept in clinic. At doses of 1‐100 mg daily, BMS‐790052 was well tolerated, had a PK profile supportive of once‐daily dosing, and produced a rapid and substantial decrease in HCV‐RNA levels in patients chronically infected with HCV genotype 1. (HEPATOLOGY 2011


The Journal of Infectious Diseases | 2012

Pharmacodynamics, safety, and pharmacokinetics of BMS-663068, an oral HIV-1 attachment inhibitor in HIV-1-infected subjects.

Richard E. Nettles; Dirk Schürmann; Li Zhu; Michele Stonier; Shu-Pang Huang; Ih Chang; Caly Chien; Mark Krystal; Megan Wind-Rotolo; Neelanjana Ray; George J. Hanna; Richard Bertz; Dennis M. Grasela

BACKGROUNDnBMS-663068 is a prodrug of the small-molecule inhibitor BMS-626529, which inhibits human immunodeficiency virus type 1 (HIV-1) infection by binding to gp120 and interfering with the attachment of virus to CD4+ T-cells.nnnMETHODSnFifty HIV-1-infected subjects were randomized to 1 of 5 regimen groups (600 mg BMS-663068 plus 100 mg ritonavir every 12 hours [Q12H], 1200 mg BMS-663068 plus 100 mg ritonavir every bedtime, 1200 mg BMS-663068 plus 100 mg ritonavir Q12H, 1200 mg BMS-663068 Q12H plus 100 mg ritonavir every morning, or 1200 mg BMS-663068 Q12H) for 8 days in this open-label, multiple-dose, parallel study. The study assessed the pharmacodynamics, pharmacokinetics, and safety of BMS-663068.nnnRESULTSnThe maximum median decrease in plasma HIV-1 RNA load from baseline ranged from 1.21 to 1.73 log(10) copies/mL. Plasma concentrations of BMS-626529 were not associated with an antiviral response, while low baseline inhibitory concentrations and the minimum and average steady-state BMS-626529 plasma concentrations, when adjusted by the baseline protein binding-adjusted 90% inhibitory concentration (inhibitory quotient), were linked with antiviral response. BMS-663068 was generally well tolerated.nnnCONCLUSIONSnAdministration of BMS-663068 for 8 days with or without ritonavir resulted in substantial declines in plasma HIV-1 RNA levels and was generally well tolerated. Longer-term clinical trials of BMS-663068 as part of combination antiretroviral therapy are warranted. Clinical Trials Registration.NCT01009814.


Antimicrobial Agents and Chemotherapy | 2011

Antiviral Activity, Pharmacokinetics, and Safety of BMS-488043, a Novel Oral Small-Molecule HIV-1 Attachment Inhibitor, in HIV-1-Infected Subjects

George J. Hanna; Jacob Lalezari; James Hellinger; David A. Wohl; Richard E. Nettles; Anna Persson; Mark Krystal; Pin-Fang Lin; Richard J. Colonno; Dennis M. Grasela

ABSTRACT BMS-488043 is a novel and unique oral small-molecule inhibitor of the attachment of human immunodeficiency virus type 1 (HIV-1) to CD4+ lymphocytes. The antiviral activity, pharmacokinetics, viral susceptibility, and safety of BMS-488043 were evaluated in an 8-day monotherapy trial. Thirty HIV-1-infected study subjects were randomly assigned to sequential, safety-guided dose panels of 800 and 1,800 mg BMS-488043 or a matched placebo in a 4:1 ratio, and the drug was administered every 12 h with a high-fat meal for 7 days and on the morning of day 8. Dose-related, albeit less-than-dose-proportional, increases in plasma BMS-488043 concentrations were observed. Mean plasma HIV-1 RNA decreases from the baseline for the BMS-488043 800- and 1,800-mg dose groups on day 8 were 0.72 and 0.96 log10 copies/ml, respectively, compared with 0.02 log10 copies/ml for the placebo group. A lower baseline BMS-488043 50% effective concentration (EC50) in the active-treatment groups was predictive of a greater antiviral response. Although absolute drug exposure was not associated with an antiviral response, the trough concentration (C trough), adjusted by the baseline EC50 (C trough/EC50), was associated with antiviral activity. During dosing, four subjects experienced >10-fold reductions in viral susceptibility to BMS-488043, providing further support of the direct antiviral mechanism of BMS-488043. BMS-488043 was generally safe and well tolerated. These results suggest that further development of this novel class of oral HIV-1 attachment inhibitors is warranted.


Antiviral Therapy | 2011

The effect of efavirenz on the pharmacokinetics of an oral contraceptive containing ethinyl estradiol and norgestimate in healthy HIV-negative women.

Heather Sevinsky; Timothy Eley; Anna Persson; Dennis Garner; Cynthia Yones; Richard E. Nettles; Kathryn Krantz; Richard Bertz; Jenny Zhang

BACKGROUNDnWomen of childbearing age represent a growing proportion of people living with HIV. Preventing pregnancy is important in HIV-infected women receiving efavirenz as part of their antiretroviral therapy.nnnMETHODSnThe effects of coadministration of efavirenz (600 mg once daily) on the pharmacokinetics (PK) of the active components (ethinyl estradiol [EE] and 17-deacetyl norgestimate [NGMN]) of Ortho Cyclen(®) (Ortho-McNeil-Janssen Pharmaceuticals, Inc., Raritan, NJ, USA) were investigated in 28 healthy HIV-negative women. The peak plasma concentration (C(max)), area under the concentration-time curve for a dosing interval (AUC([τ])), and lowest plasma concentration (C(min)) for EE and NGMN during cycles of treatment with Ortho Cyclen with and without coadministration of efavirenz were compared. Additionally, a post hoc exploratory analysis was conducted to assess the effect of efavirenz on the PK of an additional progestin, levonorgestrel (LNG).nnnRESULTSnExposures to EE were similar during coadministration of efavirenz and Ortho Cyclen to those during administration of Ortho Cyclen alone. Exposures to NGMN were substantially decreased following coadministration of efavirenz and Ortho Cyclen (adjusted geometric means for C(max), AUC([τ]) and C(min) decreased by 46%, 64% and 82%, respectively) compared with Ortho Cyclen alone. Consistent with NGMN, LNG exposures were decreased 80-86% by efavirenz.nnnCONCLUSIONSnAlthough efavirenz had no significant effect on the PK of EE, exposures to the progestin components of Ortho Cyclen, NGMN and LNG, were substantially reduced. The results reinforce the need to use reliable methods of barrier contraception, even when taking oral contraceptives and efavirenz.


Journal of Pharmaceutical Sciences | 2013

Compartmental absorption modeling and site of absorption studies to determine feasibility of an extended‐release formulation of an HIV‐1 attachment inhibitor phosphate ester prodrug

Jonathan Brown; Caly Chien; Peter Timmins; Andrew B. Dennis; Walter J. Doll; Erik P. Sandefer; Richard C. Page; Richard E. Nettles; Li Zhu; Dennis M. Grasela

BMS-663068 is a phosphonooxymethyl ester prodrug under development for the treatment of HIV/AIDS. The prodrug is designed to overcome the solubility-limited bioavailability of the active moiety, BMS-626529. BMS-663068 is not absorbed from the gastrointestinal (GI) tract and requires enzymatic conversion by alkaline phosphatase to BMS-626529 immediately before absorption. In the light of the known short in vivo half-life of BMS-626529, compartmental absorption modeling was used to predict the potential feasibility of extended-release (ER) delivery to achieve target Cmax :Cmin ratios. To further refine the model with respect to colonic absorption, the regional absorption of BMS-626529 following delivery of BMS-663068 to upper and lower GI sites was characterized through a site of absorption study in human subjects. A refined model was subsequently applied to guide the development of ER tablet formulations. Comparisons of results from the refined model to the in vivo human pharmacokinetic data for three selected ER formulations demonstrate the utility of the model in predicting feasibility of ER delivery and in directing formulation development.


Journal of Antimicrobial Chemotherapy | 2006

HIV-1 viral load blips are of limited clinical significance

Patricia K. Lee; Tara L. Kieffer; Robert F. Siliciano; Richard E. Nettles


The Journal of Infectious Diseases | 2006

Gilbert syndrome and the development of antiretroviral therapy-associated hyperbilirubinemia: genetic screening is unnecessary.

Richard E. Nettles; Michael Child; Richard Bertz; Steven Schnittman

Collaboration


Dive into the Richard E. Nettles's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Li Zhu

Bristol-Myers Squibb

View shared research outputs
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
Researchain Logo
Decentralizing Knowledge