Network


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

Hotspot


Dive into the research topics where Sean Kim is active.

Publication


Featured researches published by Sean Kim.


Current Drug Metabolism | 2006

Evaluation of 170 Xenobiotics as Transactivators of Human Pregnane X Receptor (hPXR) and Correlation to Known CYP3A4 Drug Interactions

Michael Sinz; Sean Kim; Zhengrong Zhu; Taosheng Chen; Monique N. Anthony; Kenneth E.J. Dickinson; A. D. Rodrigues

The human transcription factor pregnane X receptor (hPXR) is a key regulator of enzyme expression, especially cytochrome P450 3A4 (CYP3A4). Due to the prominence of CYP3A4 in the elimination of many drugs, the development of high throughput in vitro models to predict the effect of drugs on CYP3A4 expression have increased. To better interpret and predict potential drug-drug interactions due to CYP3A4 enzyme induction, we evaluated 170 xenobiotics in a hPXR transactivation assay and compared these results to known clinical drug-drug interactions. Of the 170 xenobiotics tested, 54% of them demonstrated some level of hPXR transactivation. By taking into consideration cell culture conditions (solubility, cytotoxicity, appropriate drug concentration in media), as well as in vivo pharmacokinetics (therapeutic plasma C(max), distribution, route of administration, dosing regimen, liver exposure, potential to inhibit CYP3A4), the risk potential of CYP3A4 enzyme induction for most compounds reduced dramatically. By employing this overall interpretation strategy, the final percentage of compounds predicted to significantly induce CYP3A4 reduced to 5%, all of which are known to cause drug-drug interactions. Also, this is the first report that identifies several potent compounds that have the ability to transactivate hPXR that previously have not been identified, such as terbinafine, diclofenac, sildenafil, glimepiride, montelukast, and ticlopidine.


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.


Clinical Cancer Research | 2008

Expanding the Roles for Pregnane X Receptor in Cancer: Proliferation and Drug Resistance in Ovarian Cancer

Divya Gupta; Madhukumar Venkatesh; Hongwei Wang; Sean Kim; Michael Sinz; Gary L. Goldberg; Kathleen D. Whitney; Clifford Longley; Sridhar Mani

Purpose: We examined the presence of the pregnane X receptor (PXR) and its effects on ovarian cancer cells after activation by its cognate ligand. Experimental Design: SKOV-3 and OVCAR-8 ovarian carcinoma cells were analyzed for expression of PXR by quantitative reverse transcription-PCR and Western blot. Human ovarian cancer tissue was also analyzed for PXR expression by immunochemistry. Ligand (agonist)–induced PXR target genes were analyzed in SKOV-3 cells by quantitative reverse transcription-PCR. SKOV-3 cell proliferation was assessed by MTT assay. In vivo confirmation of in vitro effects of PXR ligands were done in NOD.SCID mice carrying SKOV-3 xenografts. Results: PXR is expressed in ovarian cancer cells. In SKOV-3 cells, PXR is functional and its activation by cognate ligands induces PXR target genes (CYP2B6, CYP3A4, and UGT1A1) but not MDR1 and MRP2. PXR activation in SKOV-3 cells induces cell proliferation and drug resistance. In mice harboring SKOV-3 xenografts, rifampicin (PXR agonist) induces cell proliferation and tumor growth. Conclusion: PXR activation, regardless of the type of ligand agonist present, promotes the “malignant” phenotype of cancer cells. These data serve as the basis for finding novel nontoxic inhibitors of PXR activation as a method to control cell growth and prevent induction of drug resistance.


Journal of Biomolecular Screening | 2004

Correlation of High-Throughput Pregnane X Receptor (PXR) Transactivation and Binding Assays

Zhengrong Zhu; Sean Kim; Taosheng Chen; Jun-Hsiang Lin; Aneka Bell; James W. Bryson; Yves Dubaquie; Ning Yan; Joseph Yanchunas; Dianlin Xie; Robert H. Stoffel; Michael Sinz; Kenneth E.J. Dickinson

Pregnane X receptor (PXR) transactivation and binding assays have been developed into high-throughput assays, which are robust and reproducible (Z′ > 0.5). For most compounds, there was a good correlation between the results of the transactivation and binding assays. EC50 values of compounds in the transactivation assay correlated reasonably well with their IC50 values in the binding assay. However, there were discrepancies with some compounds showing high binding affinity in the binding assay translated into low transactivation. The most likely cause for these discrepancies was an agonist-dependent relationship between binding affinity and transactivation response. In general, compounds that bound to human PXR and transactivated PXR tended to be large hydrophobic molecules.


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.


Journal of Medicinal Chemistry | 2008

Discovery and evaluation of 4-(2-(4-chloro-1H-pyrazol-1-yl)ethylamino)-3-(6-(1-(3-fluoropropyl)piperidin-4-yl)-4-methyl-1H-benzo[d]imidazol-2-yl)pyridin-2(1H)-one (BMS-695735), an orally efficacious inhibitor of insulin-like growth factor-1 receptor kinase with broad spectrum in vivo antitumor activity.

Upender Velaparthi; Mark D. Wittman; Peiying Liu; Joan M. Carboni; Francis Y. Lee; Ricardo M. Attar; Praveen Balimane; Wendy Clarke; Michael Sinz; Warren Hurlburt; Karishma Patel; Lorell Discenza; Sean Kim; Marco M. Gottardis; Ann Greer; Aixin Li; Mark G. Saulnier; Zheng Yang; Kurt Zimmermann; George L. Trainor; Dolatrai M. Vyas

We previously reported that 1 (BMS-536924), a benzimidazole inhibitor of the insulin-like growth factor-1 receptor, had demonstrated in vivo antitumor activity. This lead compound was found to have potent CYP3A4 inhibition, CYP3A4 induction mediated by PXR transactivation, poor aqueous solubility, and high plasma protein binding. Herein we disclose the evolution of this chemotype to address these issues. This effort led to 10 (BMS-695735), which exhibits improved ADME properties, a low risk for drug-drug interactions, and in vivo efficacy in multiple xenograft models.


Drug Discovery Today: Technologies | 2006

Stem cells, immortalized cells and primary cells in ADMET assays

Michael Sinz; Sean Kim

Cell-based assays are beginning to replace traditional absorption, distribution, metabolism, elimination and toxicology (ADMET) models employing subcellular fractions in high throughput drug discovery screening and drug development where drugs are characterized and predictions are formulated to forecast in vivo biological outcomes. Significant and continuing advances in stem cell research, new immortalized cell lines and our enhanced ability to predict outcomes from primary cells have increased the ability to employ cell-based assays to study ADMET properties of new drugs.:


Bioorganic & Medicinal Chemistry Letters | 2010

SAR of PXR transactivation in benzimidazole-based IGF-1R kinase inhibitors

Kurt Zimmermann; Mark D. Wittman; Mark G. Saulnier; Upender Velaparthi; Xiaopeng Sang; David B. Frennesson; Charles Struzynski; Steven P. Seitz; Liqi He; Joan M. Carboni; Aixin Li; Ann Greer; Marco M. Gottardis; Ricardo M. Attar; Zheng Yang; Praveen Balimane; Lorell Discenza; Francis Y. Lee; Michael Sinz; Sean Kim; Dolatrai M. Vyas

The SAR of PXR transactivation by 3-(benzimidazol-2-yl)-pyridine-2-one based ATP competitive inhibitors of Insulin-like Growth Factor 1 Receptor kinase (IGF-1R) is discussed. Compounds without PXR transactivation, with in vivo antitumor activity, reduced protein binding and improved oral exposure are 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).


Drug Metabolism Letters | 2008

Quantitative relationship between rifampicin exposure and induction of Cyp3a11 in SXR humanized mice: extrapolation to human CYP3A4 induction potential.

Sean Kim; Devin Pray; Ming Zheng; Danial G. Morgan; Jenifer G. Pizzano; Mary Zoeckler; Anjaneya Chimalakonda; Michael Sinz

The SXR humanized mouse model was used to quantitatively assess an in vivo induction response of the human PXR agonist, rifampicin. Three days of rifampicin treatment increased RNA expression and microsomal enzyme activity of CYP3A11, as well as significantly reduced triazolam plasma exposure. These results indicate that the humanized SXR mouse can be used as a model to predict human CYP3A4 induction and the resulting pharmacokinetic changes of CYP3A4 substrates in humans.

Collaboration


Dive into the Sean Kim'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

Aixin Li

Bristol-Myers Squibb

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Darryl Gilbert

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge