Ellen Chung
Bristol-Myers Squibb
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Ellen Chung.
Gastroenterology | 2014
Gregory T. Everson; Karen Sims; Maribel Rodriguez-Torres; Christophe Hézode; Eric Lawitz; Marc Bourlière; V. Loustaud-Ratti; Vinod K. Rustgi; Howard Schwartz; Harvey A Tatum; Patrick Marcellin; Stanislas Pol; Paul J. Thuluvath; Timothy Eley; Xiaodong Wang; Shu-Pang Huang; Fiona McPhee; Megan Wind-Rotolo; Ellen Chung; Claudio Pasquinelli; Dennis M. Grasela; David F. Gardiner
BACKGROUND & AIMS The combination of peginterferon and ribavirin with telaprevir or boceprevir is the standard treatment of hepatitis C virus (HCV) genotype 1 infection. However, these drugs are not well tolerated because of their side effects and suboptimal virologic responses. In a phase 2a, open-label study, we examined the safety and efficacy of an interferon-free, ribavirin-free regimen of direct-acting antivirals, comprising daclatasvir (an NS5A replication complex inhibitor), asunaprevir (an NS3 protease inhibitor), and BMS-791325 (a non-nucleoside NS5B inhibitor), in patients with chronic HCV infection. METHODS We analyzed data from 66 treatment-naive patients with HCV genotype 1 infection without cirrhosis who were assigned randomly to groups given daclatasvir (60 mg, once daily), asunaprevir (200 mg, twice daily), and BMS-791325 (75 or 150 mg, twice daily) for 12 or 24 weeks. The primary end point was an HCV-RNA level less than 25 IU/mL at 12 weeks after treatment (sustained virologic response at 12 weeks [SVR12]). RESULTS In 64 patients, HCV-RNA levels were less than 25 IU/mL by week 4 of treatment (including 48 of 49 patients with HCV genotype 1a infection and 45 of 46 patients with the non-CC interleukin 28B genotype). Sixty-one patients (92%) achieved SVR12, based on a modified intention-to-treat analysis. Virologic responses were similar between 12 and 24 weeks of treatment. During the study, 2 patients experienced viral breakthrough and 1 patient relapsed. There were no grade 3-4 increases in levels of alanine or aspartate aminotransferases or bilirubin; there were no deaths or discontinuations resulting from serious adverse events or adverse events related to the treatment regimen. The most common adverse events were headache, asthenia, and gastrointestinal symptoms. CONCLUSIONS In a phase 2a study, the all-oral, interferon-free, and ribavirin-free regimen of daclatasvir, asunaprevir, and BMS-791325 was well tolerated and achieved high rates of SVR12 in patients with HCV genotype 1 infection. Further studies of this regimen are warranted. ClinicalTrials.gov, number NCT01455090.
Journal of Hepatology | 2015
Tarek Hassanein; Karen Sims; Michael Bennett; Norman Gitlin; Eric Lawitz; Tuan Nguyen; Lynn R. Webster; Zobair M. Younossi; Howard Schwartz; Paul J. Thuluvath; Helen Zhou; Bhaskar Rege; Fiona McPhee; Nannan Zhou; Megan Wind-Rotolo; Ellen Chung; Amber Griffies; Dennis M. Grasela; David F. Gardiner
Please cite this article as: Hassanein, T., Sims, K.D., Bennett, M., Gitlin, N., Lawitz, E., Nguyen, T., Webster, L., Younossi, Z., Schwartz, H., Thuluvath, P.J., Zhou, H., Rege, B., McPhee, F., Zhou, N., Wind-Rotolo, M., Chung, E., Griffies, A., Grasela, D.M., Gardiner, D.F., A Randomized Trial of Daclatasvir in Combination With Asunaprevir and Beclabuvir in Patients With Chronic Hepatitis C Virus Genotype 4 Infection, Journal of Hepatology (2015), doi: http://dx.doi.org/10.1016/j.jhep.2014.12.025
Journal of Hepatology | 2013
Gregory T. Everson; Karen Sims; Maribel Rodriguez-Torres; Christophe Hézode; Eric Lawitz; Marc Bourlière; V. Loustaud-Ratti; Vinod K. Rustgi; Howard Schwartz; Harvey A Tatum; Patrick Marcellin; Stanislas Pol; Paul J. Thuluvath; Timothy Eley; X. Wang; S.-P. Huang; Fiona McPhee; Megan Wind-Rotolo; Ellen Chung; Claudio Pasquinelli; Dennis M. Grasela; David F. Gardiner
13.6±1.8 g/dL [8.8–17.5], respectively. After 12W, a complete early virological response was obtained in 34 (83%) boceprevir patients and in 35 (61%) telaprevir patients (p = 0.026). Among 17 boceprevir and 16 telaprevir patients, 14 (82%) and 7 (43%) achieved an end of treatment response (EOT) with an undetetectable viral load, respectively (p = 0.032). Among 9 boceprevir and 5 telaprevir patients, 6 and 1 achieved SVR12, respectively. Among 6 patients in the boceprevir group, 3 achieved SVR24. In the telaprevir group, 29 patients discontinued therapy (serious adverse events, n = 13; virological breakthrough, n = 6; non-response, n = 9). In the boceprevir group, 14 patients discontinued therapy (serious adverse events, n = 5; virological breakthrough, n = 2; non-response, n = 4; retransplantation, n = 1). Four patients died in a context of infectious disorders: boceprevir, n = 2 (W20/W24); telaprevir, n = 2 (W2/W9). The most common side effect was anemia in 85% of patients: 95% and 96% in boceprevir and telaprevir groups received erythropoietin alone or combined with ribavirin dose reduction. Conclusion: In liver transplanted patients, EOT rate was 82% and 38% with boceprevir and telaprevir, respectively. Among the overall population, 44% of patients discontinued therapy because of treatment failure or occurrence of serious adverse events.
Antiviral Therapy | 2011
Jenny Zhang; Ellen Chung; Cynthia Yones; Anna Persson; Lisa Mahnke; Timothy Eley; Xiaohui Xu; Richard Bertz
BACKGROUND Coadministration of oral contraceptives with protease inhibitors is complicated by drug interactions. An open-label three-period single-sequence study assessed the effect of coadministration of atazanavir (ATV)/ritonavir (RTV) with Ortho Tri-Cyclen(®) and with Ortho Tri-Cyclen(®) LO (Ortho-McNeil-Janssen Pharmaceuticals, Inc., Raritan, NJ, USA) on the pharmacokinetics (PK) of ethinyl estradiol (EE), norgestimate (NGM), ATV and RTV. METHODS A total of 20 healthy women aged 18-45 years received study treatments. In the lead-in period and in period 1, participants received a full cycle of Ortho Tri-Cyclen (EE 35 μg with NGM 0.18/0.215/0.25 mg) from days 1-28. In period 2, participants received a full cycle of Ortho Tri-Cyclen LO (EE 25 μg with NGM 0.18/0.215/0.25 mg) plus ATV/RTV (300/100 mg once daily) on days 29-42. PK assessments were performed on days 14 and 42 in periods 1 and 2, respectively. RESULTS ATV/RTV with dose-normalized EE/NGM resulted in geometric mean reductions of 16% in EE peak plasma concentration (C(max)), 19% in EE area under the concentration-time curve for a dosing interval (AUC([τ])) and 37% in EE lowest plasma concentration (C(min)), compared with EE 35 μg with NGM in the absence of ATV/RTV. NGM with EE and ATV/RTV 300/100 mg once daily resulted in increases of approximately 68%, 85% and 102% in 17-deacetyl NGM C(max), AUC((τ)) and C(min), respectively. Two participants discontinued the study because of adverse events. CONCLUSIONS ATV/RTV with Ortho Tri-Cyclen was well-tolerated and reductions in EE were not predicted to decrease contraceptive efficacy if the formulation contained ≥30 μg of EE.
Aids Patient Care and Stds | 2011
Xiaodong Wang; Marta Boffito; Jenny Zhang; Ellen Chung; Li Zhu; Yaoshi Wu; Kristine B. Patterson; Angela D. M. Kashuba; Pablo Tebas; Michael Child; Lisa Mahnke; Richard Bertz
Significant pharmacokinetic interactions can result between acid-suppressing agents and some protease inhibitors (PIs) in the management of HIV infection. In healthy subjects, famotidine, an H(2)-receptor antagonist, reduces exposures of atazanavir by 4-28% at doses of 20-40 mg twice daily. This study evaluated the effect of famotidine 20-40 mg twice daily on the pharmacokinetics of atazanavir/ritonavir 300/100 mg once daily with and without tenofovir disoproxil fumarate (TDF) 300 mg in HIV-infected patients (n=40; 87.5% male; mean age 42, range 26-63 years; 55% white). Coadministration of famotidine 40 mg and atazanavir/ritonavir to HIV-infected patients reduced exposures of atazanavir by approximately 20%. This is comparable to reductions seen in HIV-uninfected subjects. Coadministration of famotidine 20 mg had less impact on atazanavir exposures, with no reduction of atazanavir geometric mean plasma concentration at 24 h postdose (C(min)). In the presence of TDF, administration of famotidine 20-40 mg twice daily 2 h after and 10 h before atazanavir/ritonavir reduced exposures of atazanavir by 19-25%. However, all individual atazanavir C(min) values remained at least five-fold above the population mean protein-binding adjusted 90% maximum effect (EC(90)) against wild-type HIV (14 ng/mL). No viral load rebound was observed at end of study. The results confirmed that coadministration of an H(2)-receptor antagonist with atazanavir/ritonavir in HIV-infected patients resulted in similar magnitude of reductions in atazanavir exposures as in healthy subjects. This supports the current dose recommendations for coadministration of an H(2)-receptor antagonist with atazanavir/ritonavir.
Pharmacotherapy | 2013
Richard Bertz; Anna Persson; Ellen Chung; Li Zhu; Jenny Zhang; Donnie McGrath; Dennis M. Grasela
To investigate the pharmacokinetic and pharmacodynamic relationships of the human immunodeficiency virus (HIV)–protease inhibitor atazanavir (ATV) in the presence and absence of the pharmacokinetic booster ritonavir, utilizing ATV plasma trough concentrations (Ctrough) and clinical biomarkers of antiviral efficacy and safety over 48 weeks.
The Journal of Clinical Pharmacology | 2017
Li Zhu; Roger J. M. Brüggemann; Jonathan Uy; Angela Colbers; Matthew Hruska; Ellen Chung; Karen Sims; Blisse Vakkalagadda; Xiaohui Xu; Ron H.N. van Schaik; David M. Burger; Richard Bertz
Voriconazole, a broad‐spectrum triazole antifungal agent, is metabolized by cytochrome P450 (CYP) 2C19 and, to a lesser extent, by CYP3A. Genetic polymorphism of CYP2C19 not only plays a prominent role in its disposition but may also influence potential drug interactions with CYP450 modulators such as ritonavir. This study assessed 2‐way drug interactions of voriconazole added on to ritonavir‐boosted atazanavir in both CYP2C19 extensive‐metabolizer (EM) and poor‐metabolizer (PM) healthy subjects. Each subject received voriconazole alone on days 1–3, followed by a 7‐day washout. Atazanavir/ritonavir 300/100 mg once daily was given on days 11–30 and voriconazole on days 21–30. Voriconazole doses were 200 mg (400 mg on days 1 and 21) twice daily and 50 mg (100 mg on days 1 and 21) twice daily for CYP2C19 EM and PM subjects, respectively. On coadministration, voriconazole AUC and Cmin decreased by 33% (90%CI, 22%–42%) and 39% (90%CI, 28%–49%), respectively, in CYP2C19 EMs, whereas voriconazole Cmax and AUC increased 4.4‐fold (90%CI, 3.6‐fold to 5.4‐fold) and 5.6‐fold (90%CI, 4.5‐fold to 7.0‐fold), respectively, in PMs. Adding voriconazole resulted in a 20%–30% decrease in atazanavir Cmin in both EMs and PMs. Ritonavir exposure was generally unchanged in either population. The safety and tolerability profiles of the combination were comparable with atazanavir/ritonavir and voriconazole administered alone. The most frequent adverse events with voriconazole were visual disturbance and headache. Coadministration of voriconazole and atazanavir/ritonavir is not recommended unless the benefit/risk to the patient justifies the use of the combination.
Journal of Hepatology | 2013
X. Wang; W. Li; S.-P. Huang; Bing He; Ellen Chung; A. Griffies; E. Cooney; Eric Hughes; H. Kandoussi; Karen Sims; David F. Gardiner; Richard Bertz; Timothy Eley
Journal of Hepatology | 2015
Bing He; Karen Sims; Ellen Chung; W.-L. Luo; J. Pursley; Megan Wind-Rotolo; Frank LaCreta; Richard Bertz; Malaz AbuTarif
Gastroenterology | 2014
Tarek Hassanein; Gregory T. Everson; Karen Sims; Michael T. Bennett; Norman Gitlin; Eric Lawitz; Tuan T. Nguyen; Lynn R. Webster; Zobair M. Younossi; Howard Schwartz; Paul J. Thuluvath; Helen Zhou; Bhaskar Rege; Fiona McPhee; Megan Wind-Rotolo; Ellen Chung; Amber Griffies; Dennis M. Grasela; David F. Gardiner