Network


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

Hotspot


Dive into the research topics where Mary Zoeckler is active.

Publication


Featured researches published by Mary Zoeckler.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Inhibition of influenza virus replication via small molecules that induce the formation of higher-order nucleoprotein oligomers

Samuel W. Gerritz; Christopher Cianci; Sean Kim; Bradley C. Pearce; Carol Deminie; Linda F. Discotto; Brian McAuliffe; B Minassian; Shuhao Shi; Shirong Zhu; Weixu Zhai; Annapurna Pendri; Guo Li; Michael A. Poss; Suzanne Edavettal; Patricia A. McDonnell; Hal A. Lewis; Klaus Maskos; Mario Mörtl; Reiner Kiefersauer; Stefan Steinbacher; Eric T. Baldwin; William Metzler; James Bryson; Matthew D. Healy; Thomas Philip; Mary Zoeckler; Richard Schartman; Michael Sinz; Victor H. Leyva-Grado

Influenza nucleoprotein (NP) plays multiple roles in the virus life cycle, including an essential function in viral replication as an integral component of the ribonucleoprotein complex, associating with viral RNA and polymerase within the viral core. The multifunctional nature of NP makes it an attractive target for antiviral intervention, and inhibitors targeting this protein have recently been reported. In a parallel effort, we discovered a structurally similar series of influenza replication inhibitors and show that they interfere with NP-dependent processes via formation of higher-order NP oligomers. Support for this unique mechanism is provided by site-directed mutagenesis studies, biophysical characterization of the oligomeric ligand:NP complex, and an X-ray cocrystal structure of an NP dimer of trimers (or hexamer) comprising three NP_A:NP_B dimeric subunits. Each NP_A:NP_B dimeric subunit contains two ligands that bridge two composite, protein-spanning binding sites in an antiparallel orientation to form a stable quaternary complex. Optimization of the initial screening hit produced an analog that protects mice from influenza-induced weight loss and mortality by reducing viral titers to undetectable levels throughout the course of treatment.


Drug Metabolism and Disposition | 2010

Evaluation of cynomolgus monkey pregnane X receptor, primary hepatocyte, and in vivo pharmacokinetic changes in predicting human CYP3A4 induction.

Sean Kim; Joseph E. Dinchuk; Monique N. Anthony; Tami Orcutt; Mary Zoeckler; Mary B. Sauer; Kathleen W. Mosure; Ragini Vuppugalla; James E. Grace; Jean Simmermacher; Heidi Dulac; Jennifer Pizzano; Michael Sinz

Monkeys have been proposed as an animal model to predict the magnitude of human clinical drug-drug interactions caused by CYP3A4 enzyme induction. To evaluate whether the cynomolgus monkey can be an effective in vivo model, human CYP3A4 inducers were evaluated both in vitro and in vivo. First, a full-length pregnane X receptor (PXR) was cloned from the cynomolgus monkey, and the sequence was compared with those of rhesus monkey and human PXR. Cynomolgus and rhesus monkey PXR differed by only one amino acid (A68V), and both were highly homologous to human PXR (∼96%). When the transactivation profiles of 30 compounds, including known inducers of CYP3A4, were compared between cynomolgus and human PXR, a high degree of correlation with EC50 values was observed. These results suggest that cynomolgus and human PXR respond in a similar fashion to these ligands. Second, two known human CYP3A4 inducers, rifampicin and hyperforin, were tested in monkey and human primary hepatocytes for induction of CYP3A enzymes. Both monkey and human hepatocytes responded similarly to the inducers and resulted in increased RNA and enzyme activity changes of CYP3A8 and CYP3A4, respectively. Lastly, in vivo induction of CYP3A8 by rifampicin and hyperforin was shown by significant reductions of midazolam exposure that were comparable with those in humans. These results show that the cynomolgus monkey can be a predictive in vivo animal model of PXR-mediated induction of human CYP3A4 and can provide a useful assessment of the resulting pharmacokinetic changes of affected drugs.


Bioorganic & Medicinal Chemistry Letters | 2001

Synthesis of metabolically blocked paclitaxel analogues

Mark D. Wittman; Thomas J. Altstadt; Craig R. Fairchild; Stephen Hansel; Kathy A. Johnston; John F. Kadow; Byron H. Long; William C. Rose; Dolatrai M. Vyas; Mu-Jen Wu; Mary Zoeckler

The stereospecific syntheses of the metabolically blocked 6-alpha-F, Cl, Br paclitaxel, and 6-alpha-F-10-acetyldocetaxel are described and in vitro and in vivo activity is presented.


Journal of Pharmacology and Experimental Therapeutics | 2011

Pharmacokinetic-Pharmacodynamic Modeling of Rifampicin-Mediated Cyp3a11 Induction in Steroid and Xenobiotic X Receptor Humanized Mice

Joseph J. Raybon; Devin Pray; Daniel G Morgan; Mary Zoeckler; Ming Zheng; Michael Sinz; Sean Kim

The purpose of this study was to develop a mechanistic pharmacokinetic-pharmacodynamic (PK-PD) model to describe the effects of rifampicin on hepatic Cyp3a11 RNA, enzymatic activity, and triazolam pharmacokinetics. Rifampicin was administered to steroid and xenobiotic X receptor (SXR) humanized mice at 10 mg/kg p.o. (every day for 3 days) followed by triazolam (4 mg/kg p.o.) 24 h after the last dose of rifampicin. Rifampicin and triazolam concentrations and Cyp3a11 RNA expression and activity in the liver were measured over the 4-day period. Elevations in Cyp3a11 RNA expression were observed 24 h after the first dose of rifampicin, reaching a maximum (∼10 times baseline) after the third dose and were sustained until day 4 and began declining 48 h after the last rifampicin dose. Similar changes in enzymatic activity were also observed. The triazolam serum area under the curve (AUC) was 5-fold lower in mice pretreated with rifampicin, consistent with enzyme induction. The final PK-PD model incorporated rifampicin liver concentration as the driving force for the time-delayed Cyp3a11 induction governed by in vitro potency estimates, which in turn regulated the turnover of enzyme activity. The PK-PD model was able to recapitulate the delayed induction of Cyp3a11 mRNA and enzymatic activity by rifampicin. Furthermore, the model was able to accurately anticipate the reduction in the triazolam plasma AUC by integrating a ratio of the predicted induced enzyme activity and basal activity into the equations describing triazolam pharmacokinetics. In conjunction with the SXR humanized mouse model, this mathematical approach may serve as a tool for predicting clinically relevant drug-drug interactions via pregnane X receptor-mediated enzyme induction and possibly extended to other induction pathways (e.g., constitutive androstane receptor).


Journal of Medicinal Chemistry | 2005

Discovery of a (1H-benzoimidazol-2-yl)-1H-pyridin-2-one (BMS-536924) inhibitor of insulin-like growth factor I receptor kinase with in vivo antitumor activity.

Mark D. Wittman; Joan M. Carboni; Ricardo M. Attar; Balu Balasubramanian; Praveen Balimane; Patrick Brassil; Francis Beaulieu; Chiehying Chang; Wendy Clarke; Janet Dell; Jeffrey Eummer; David B. Frennesson; Marco M. Gottardis; Ann Greer; Steven Hansel; Warren Hurlburt; Bruce L. Jacobson; Subramaniam Krishnananthan; Francis Y. Lee; Aixin Li; Tai-An Lin; Peiying Liu; Carl Ouellet; Xiaopeng Sang; Mark G. Saulnier; Karen Stoffan; Yax Sun; Upender Velaparthi; Henry Wong; Zheng Yang


Cancer Research | 1997

Effect of Linker Variation on the Stability, Potency, and Efficacy of Carcinoma-reactive BR64-Doxorubicin Immunoconjugates

Pamela A. Trail; David Willner; Jay O. Knipe; Arris J. Henderson; Shirley J. Lasch; Mary Zoeckler; Mark D. TrailSmith; Terrence W. Doyle; H. Dalton King; Anna Maria Casazza; Gary R. Braslawsky; Joseph P. Brown; Sandra J. Hofstead; Robert S. Greenfield; Raymond A. Firestone; Kathleen W. Mosure; Kathleen F. Kadow; Michael B. Yang; Karl Erik Hellström; Ingegerd Hellström


Cancer Research | 1992

Antigen-specific Activity of Carcinoma-reactive BR64-Doxorubicin Conjugates Evaluated in Vitro and in Human Tumor Xenograft Models

Pamela A. Trail; David Willner; Shirley J. Lasch; Aris J. Henderson; Robert S. Greenfield; Dalton King; Mary Zoeckler; Gary R. Braslawsky


Bioorganic & Medicinal Chemistry | 2003

The discovery of BMS-275183: an orally efficacious novel taxane

Harold Mastalerz; Donald Cook; Craig R. Fairchild; Steven Hansel; Walter Lewis Johnson; John F. Kadow; Byron H. Long; William C. Rose; James G. Tarrant; Mu-Jen Wu; May Quifen Xue; Guifen Zhang; Mary Zoeckler; Dolatrai M. Vyas


Bioorganic & Medicinal Chemistry Letters | 2007

Imidazole moiety replacements in the 3 -(1H -benzo[d]imidazol-2 -yl )pyridin -2 (1H) -one inhibitors of insulin-like growth factor receptor-1 (IGF-1R) to improve cytochrome P450 profile

Upender Velaparthi; Peiying Liu; Balu Balasubramanian; Joan M. Carboni; Ricardo M. Attar; Marco M. Gottardis; Aixin Li; Ann Greer; Mary Zoeckler; Mark D. Wittman; Dolatrai M. Vyas


Bioorganic & Medicinal Chemistry Letters | 2007

Novel 1H-(benzimidazol-2-yl)-1H-pyridin-2-one inhibitors of insulin-like growth factor I (IGF-1R) kinase

Mark D. Wittman; Balu Balasubramanian; Karen Stoffan; Upender Velaparthi; Pieying Liu; Subramaniam Krishnanathan; Joan M. Carboni; Aixin Li; Ann Greer; Ricardo M. Attar; Marco M. Gottardis; Chiehying Chang; Bruce L. Jacobson; Yax Sun; Steven Hansel; Mary Zoeckler; Dolatrai M. Vyas

Collaboration


Dive into the Mary Zoeckler's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aixin Li

Bristol-Myers Squibb

View shared research outputs
Researchain Logo
Decentralizing Knowledge