Network


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

Hotspot


Dive into the research topics where Bradley K. Wong is active.

Publication


Featured researches published by Bradley K. Wong.


Nature Medicine | 2000

A peptide–doxorubicin 'prodrug' activated by prostate-specific antigen selectively kills prostate tumor cells positive for prostate-specific antigen in vivo

Deborah Defeo-Jones; Victor M. Garsky; Bradley K. Wong; Dong-Mei Feng; Trina Bolyar; Kathleen M. Haskell; David M. Kiefer; Karen R. Leander; Elizabeth McAvoy; Patricia K. Lumma; Jenny Miu-Chun Wai; Edith T. Senderak; Sherri L. Motzel; Kevin P. Keenan; Matthew J. van Zwieten; Jiunn H. Lin; Roger M. Freidinger; Joel R. Huff; Allen Oliff; Raymond E. Jones

We covalently linked doxorubicin with a peptide that is hydrolyzable by prostate-specific antigen. In the presence of prostate tumor cells secreting prostate-specific antigen, the peptide moiety of this conjugate, L-377,202, was hydrolyzed, resulting in the release of leucine-doxorubicin and doxorubicin, which are both very cytotoxic to cancer cells. However, L-377,202 was much less cytotoxic than conventional doxorubicin to cells in culture that do not secrete prostate-specific antigen. L-377,202 was approximately 15 times more effective than was conventional doxorubicin at inhibiting the growth of human prostate cancer tumors in nude mice when both drugs were used at their maximally tolerated doses. Nude mice inoculated with human prostate tumor cells secreting prostate-specific antigen showed considerable reductions in tumor burden with minimal total body weight loss when treated with L-377,202. This improvement in therapeutic index correlated with the selective localization of leucine–doxorubicin and free doxorubicin in tissues secreting prostate-specific antigen after exposure to L-377,202.


Current Drug Metabolism | 2012

Enzyme kinetics of cytochrome P450-mediated reactions.

Magang Shou; Yuh Lin; Ping Lu; Cuyue Tang; Qin Mei; Dan Cui; Wei Tang; Jason S. Ngui; C. Charles Lin; Rominder Singh; Bradley K. Wong; James A. Yergey; Jiunn H. Lin; Paul G. Pearson; Thomas A. Baillie; A. David Rodrigues; Thomas H. Rushmore

The most common drug-drug interactions may be understood in terms of alterations of metabolism, associated primarily with changes in the activity of cytochrome P450 (CYP) enzymes. Kinetic parameters such as Km, Vmax, Ki and Ka, which describe metabolism-based drug interactions, are usually determined by appropriate kinetic models and may be used to predict the pharmacokinetic consequences of exposure to one or multiple drugs. According to classic Michaelis-Menten (M-M) kinetics, one binding site models can be employed to simply interpret inhibition (pure competitive, non-competitive and uncompetitive) or activation of the enzyme. However, some cytochromes P450, in particular CYP3A4, exhibit unusual kinetic characteristics. In this instance, the changes in apparent kinetic constants in the presence of inhibitor or activator or second substrate do not obey the rules of M-M kinetics, and the resulting kinetics are not straightforward and hamper mechanistic interpretation of the interaction in question. These unusual kinetics include substrate activation (autoactivation), substrate inhibition, partial inhibition, activation, differential kinetics and others. To address this problem, several kinetic models can be proposed, based upon the assumption that multiple substrate binding sites exist at the active site of a particular P450, and the resulting kinetic constants are, therefore, solved to adequately describe the observed interaction between multiple drugs. The following is an overview of some cytochrome P450-mediated classic and atypical enzyme kinetics, and the associated kinetic models. Applications of these kinetic models can provide some new insights into the mechanism of P450-mediated drug-drug interactions.


Drug Metabolism and Disposition | 2009

An Inhibitory Metabolite Leads to Dose- and Time-Dependent Pharmacokinetics of (R)-N-{1-(3-(4-Ethoxy-phenyl)-4-oxo-3,4- dihydro-pyrido(2,3-d)pyrimidin-2-yl)-ethyl}-N-pyridin-3-yl-methyl-2- (4-trifluoromethoxy-phenyl)-acetamide (AMG 487) in Human Subjects After Multiple Dosing

George Tonn; Simon G. Wong; Sylvia C. Wong; Michael G. Johnson; Ji Ma; Robert Cho; Leslie C. Floren; Kathryn Kersey; Karen Berry; Andrew P. Marcus; Xuemei Wang; Bettina Van Lengerich; Julio C. Medina; Paul G. Pearson; Bradley K. Wong

(R)-N-{1-[3-(4-Ethoxy-phenyl)-4-oxo-3,4-dihydro-pyrido[2,3-d]-pyrimidin-2-yl]-ethyl}-N-pyridin-3-yl-methyl-2-(4-trifluoromethoxyphenyl)-acetamide (AMG 487) is a potent and selective orally bioavailable chemokine (C-X-C motif) receptor 3 (CXCR3) antagonist that displays dose- and time-dependent pharmacokinetics in human subjects after multiple oral dosing. Although AMG 487 exhibited linear pharmacokinetics on both days 1 and 7 at the 25-mg dose, dose- and time-dependent kinetics were evident at the two higher doses. Nonlinear kinetics were more pronounced after multiple dosing. Area under the plasma concentration-time curve from 0 to 24 h [AUC(0–24 h)] increased 96-fold with a 10-fold increase in dose on day 7 compared with a 28-fold increase in AUC(0–24 h) on day 1. These changes were correlated with time- and dose-dependent decreases in the metabolite to parent plasma concentrations, suggesting that these changes result from a decrease in the oral clearance (CL) of AMG 487 (e.g., intestinal/hepatic first-pass metabolism and systemic CL). The biotransformation of AMG 487 is dependent on CYP3A and results in the formation of two primary metabolites, a pyridyl N-oxide AMG 487 (M1) and an O-deethylated AMG 487 (M2). One of these metabolites, M2, undergoes further metabolism by CYP3A. M2 has also been demonstrated to inhibit CYP3A in a competitive (Ki = 0.75 μM) manner as well as via mechanism-based inhibition (unbound KI = 1.4 μM, kinact = 0.041 min–1). Data from this study implicate M2-mediated CYP3A mechanism-based inhibition as the proximal cause for the time-dependent pharmacokinetics of AMG 487. However, the sequential metabolism of M2, nonlinear AMG 487 pharmacokinetics, and the inability to accurately determine the role of intestinal AMG 487 metabolism complicates the correlation between M2 plasma concentrations and the time-dependent AMG 487 pharmacokinetic changes.


Drug Metabolism and Disposition | 2012

Sequential Metabolism of AMG 487, a Novel CXCR3 Antagonist, Results in Formation of Quinone Reactive Metabolites That Covalently Modify CYP3A4 Cys239 and Cause Time-Dependent Inhibition of the Enzyme

Kirk R. Henne; Thuy Tran; Brooke M. VandenBrink; Dan A. Rock; Divesh Aidasani; Raju Subramanian; Andrew K. Mason; David M. Stresser; Yohannes Teffera; Simon Wong; Michael G. Johnson; Xiaoqi Chen; George Tonn; Bradley K. Wong

CYP3A4-mediated biotransformation of (R)-N-(1-(3-(4-ethoxyphenyl)-4-oxo-3,4-dihydropyrido[2,3-d]pyrimidin-2-yl)ethyl)-N-(pyridin-3-ylmethyl)-2-(4-(trifluoromethoxy)phenyl)acetamide (AMG 487) was previously shown to generate an inhibitory metabolite linked to dose- and time-dependent pharmacokinetics in humans. Although in vitro activity loss assays failed to demonstrate CYP3A4 time-dependent inhibition (TDI) with AMG 487, its M2 phenol metabolite readily produced TDI when remaining activity was assessed using either midazolam or testosterone (KI = 0.73–0.74 μM, kinact = 0.088–0.099 min−1). TDI investigations using an IC50 shift method successfully produced inhibition attributable to AMG 487, but only when preincubations were extended from 30 to 90 min. The shift magnitude was ∼3× for midazolam activity, but no shift was observed for testosterone activity. Subsequent partition ratio determinations conducted for M2 using recombinant CYP3A4 showed that inactivation was a relatively inefficient process (r = 36). CYP3A4-mediated biotransformation of [3H]M2 in the presence of GSH led to identification of two new metabolites, M4 and M5, which shifted focus away from M2 being directly responsible for TDI. M4 (hydroxylated M2) was further metabolized to form reactive intermediates that, upon reaction with GSH, produced isomeric adducts, collectively designated M5. Incubations conducted in the presence of [18O]H2O confirmed incorporation of oxygen from O2 for the majority of M4 and M5 formed (>75%). Further evidence of a primary role for M4 in CYP3A4 TDI was generated by protein labeling and proteolysis experiments, in which M4 was found to be covalently bound to Cys239 of CYP3A4. These investigations confirmed a primarily role for M4 in CYP3A4 inactivation, suggesting that a more complex metabolic pathway was responsible for generation of inhibitory metabolites affecting AMG 487 human pharmacokinetics.


Drug Metabolism and Disposition | 2008

An Inhibitory Metabolite Leads To Dose- And Time-Dependent Pharmacokinetics Of AMG 487 In Human Subjects Following Multiple Dosing

George Tonn; Simon Wong; Sylvia C. Wong; Michael G. Johnson; Ji Ma; Robert Cho; Leslie C. Floren; Kathryn Kersey; Karen Berry; Andrew P. Marcus; Xuemei Wang; Bettina Van Lengerich; Julio C. Medina; Paul G. Pearson; Bradley K. Wong

(R)-N-{1-[3-(4-Ethoxy-phenyl)-4-oxo-3,4-dihydro-pyrido[2,3-d]-pyrimidin-2-yl]-ethyl}-N-pyridin-3-yl-methyl-2-(4-trifluoromethoxyphenyl)-acetamide (AMG 487) is a potent and selective orally bioavailable chemokine (C-X-C motif) receptor 3 (CXCR3) antagonist that displays dose- and time-dependent pharmacokinetics in human subjects after multiple oral dosing. Although AMG 487 exhibited linear pharmacokinetics on both days 1 and 7 at the 25-mg dose, dose- and time-dependent kinetics were evident at the two higher doses. Nonlinear kinetics were more pronounced after multiple dosing. Area under the plasma concentration-time curve from 0 to 24 h [AUC(0–24 h)] increased 96-fold with a 10-fold increase in dose on day 7 compared with a 28-fold increase in AUC(0–24 h) on day 1. These changes were correlated with time- and dose-dependent decreases in the metabolite to parent plasma concentrations, suggesting that these changes result from a decrease in the oral clearance (CL) of AMG 487 (e.g., intestinal/hepatic first-pass metabolism and systemic CL). The biotransformation of AMG 487 is dependent on CYP3A and results in the formation of two primary metabolites, a pyridyl N-oxide AMG 487 (M1) and an O-deethylated AMG 487 (M2). One of these metabolites, M2, undergoes further metabolism by CYP3A. M2 has also been demonstrated to inhibit CYP3A in a competitive (Ki = 0.75 μM) manner as well as via mechanism-based inhibition (unbound KI = 1.4 μM, kinact = 0.041 min–1). Data from this study implicate M2-mediated CYP3A mechanism-based inhibition as the proximal cause for the time-dependent pharmacokinetics of AMG 487. However, the sequential metabolism of M2, nonlinear AMG 487 pharmacokinetics, and the inability to accurately determine the role of intestinal AMG 487 metabolism complicates the correlation between M2 plasma concentrations and the time-dependent AMG 487 pharmacokinetic changes.


Archive | 2001

Design and Synthesis of a Selective PSA Cleavable Peptide-Doxorubicin Prodrug Which Targets PSA Positive Tumor Cells

Victor M. Garsky; Patricia K. Lumma; Dong-Mei Feng; Jenny Miu-Chun Wai; Mohinder K. Sardana; Harri G. Ramjit; Bradley K. Wong; Allen Oliff; Raymond E. Jones; Deborah Defeo-Jones; Roger M. Freidinger

Prostate cancer is the second leading cause of cancer mortality in males. It is estimated that 37,000 men died of prostate carcinoma in the United States in 1999 [1]. While cancer that is confined to the prostate can be treated with surgery or radiation, the prognosis for metastatic disease is poor.


Proceedings of the National Academy of Sciences of the United States of America | 2004

A naphthyridine carboxamide provides evidence for discordant resistance between mechanistically identical inhibitors of HIV-1 integrase

Daria J. Hazuda; Neville J. Anthony; Robert P. Gomez; Samson M. Jolly; John S. Wai; Linghang Zhuang; Thorsten E. Fisher; Mark W. Embrey; James P. Guare; Melissa S. Egbertson; Joseph P. Vacca; Joel R. Huff; Peter J. Felock; Marc Witmer; Kara A. Stillmock; Robert Danovich; Jay A. Grobler; Michael D. Miller; Amy S. Espeseth; Lixia Jin; I-Wu Chen; Jiunn H. Lin; Kelem Kassahun; Joan D. Ellis; Bradley K. Wong; Wei Xu; Paul G. Pearson; William A. Schleif; Riccardo Cortese; Emilio A. Emini


Journal of Medicinal Chemistry | 2007

Potent, orally bioavailable calcitonin gene-related peptide receptor antagonists for the treatment of migraine: discovery of N-[(3R,6S)-6-(2,3-difluorophenyl)-2-oxo-1- (2,2,2-trifluoroethyl)azepan-3-yl]-4- (2-oxo-2,3-dihydro-1H-imidazo[4,5-b]pyridin- 1-yl)piperidine-1-carboxamide (MK-0974).

Daniel V. Paone; Anthony W. Shaw; Diem N. Nguyen; Christopher S. Burgey; James Z. Deng; Stefanie A. Kane; Kenneth S. Koblan; Christopher A. Salvatore; Scott D. Mosser; Victor K. Johnston; Bradley K. Wong; Cynthia Miller-Stein; James C. Hershey; Samuel Graham; and Joseph P. Vacca; Theresa M. Williams


Journal of Medicinal Chemistry | 2004

Discovery and Evaluation of Potent P1 Aryl Heterocycle-Based Thrombin Inhibitors

Mary Beth Young; James C. Barrow; Kristen L. Glass; George F. Lundell; Christina L. Newton; Janetta M. Pellicore; Kenneth E. Rittle; Harold G. Selnick; Kenneth J. Stauffer; Joseph P. Vacca; Peter D. Williams; Dennis Bohn; Franklin C. Clayton; Jacquelynn J. Cook; Julie A. Krueger; Lawrence C. Kuo; S. Dale Lewis; Bobby J. Lucas; Daniel R. McMasters; Cynthia Miller-Stein; Beth Pietrak; Audrey A. Wallace; Rebecca B. White; Bradley K. Wong; Youwei Yan; Philippe G. Nantermet


Journal of Medicinal Chemistry | 2003

Metabolism-directed optimization of 3-aminopyrazinone acetamide thrombin inhibitors. Development of an orally bioavailable series containing P1 and P3 pyridines.

Christopher S. Burgey; Kyle A. Robinson; Terry A. Lyle; Philip E. Sanderson; S. Dale Lewis; Bobby J. Lucas; Julie A. Krueger; Rominder Singh; Cynthia Miller-Stein; Rebecca B. White; Bradley K. Wong; Elizabeth A. Lyle; Peter D. Williams; Craig A. Coburn; Bruce D. Dorsey; James C. Barrow; Maria T. Stranieri; Marie Holahan; Gary R. Sitko; Jacquelynn J. Cook; Daniel R. McMasters; Colleen McDonough; William M. Sanders; Audrey A. Wallace; Franklin C. Clayton; Dennis Bohn; Yvonne M. Leonard; Theodore J. Detwiler; Joseph J. Lynch; Youwei Yan

Researchain Logo
Decentralizing Knowledge