Network


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

Hotspot


Dive into the research topics where Lixia Jin is active.

Publication


Featured researches published by Lixia Jin.


Journal of Pharmacology and Experimental Therapeutics | 2009

First Demonstration of Cerebrospinal Fluid and Plasma Aβ Lowering with Oral Administration of a β-Site Amyloid Precursor Protein-Cleaving Enzyme 1 Inhibitor in Nonhuman Primates

Sethu Sankaranarayanan; Marie A. Holahan; Dennis Colussi; Ming-Chih Crouthamel; Viswanath Devanarayan; Joan D. Ellis; Amy S. Espeseth; Adam T. Gates; Samuel Graham; Allison R. Gregro; Daria J. Hazuda; Jerome H. Hochman; Katharine M Holloway; Lixia Jin; Jason A. Kahana; Ming-Tain Lai; Janet Lineberger; Georgia B. McGaughey; Keith P. Moore; Philippe G. Nantermet; Beth Pietrak; Eric A. Price; Hemaka A. Rajapakse; Shaun R. Stauffer; Melissa A. Steinbeiser; Guy R. Seabrook; Harold G. Selnick; Xiao-Ping Shi; Matthew G. Stanton; John Swestock

β-Site amyloid precursor protein (APP)-cleaving enzyme (BACE) 1 cleavage of amyloid precursor protein is an essential step in the generation of the potentially neurotoxic and amyloidogenic Aβ42 peptides in Alzheimers disease. Although previous mouse studies have shown brain Aβ lowering after BACE1 inhibition, extension of such studies to nonhuman primates or man was precluded by poor potency, brain penetration, and pharmacokinetics of available inhibitors. In this study, a novel tertiary carbinamine BACE1 inhibitor, tertiary carbinamine (TC)-1, was assessed in a unique cisterna magna ported rhesus monkey model, where the temporal dynamics of Aβ in cerebrospinal fluid (CSF) and plasma could be evaluated. TC-1, a potent inhibitor (IC50 ∼ 0.4 nM), has excellent passive membrane permeability, low susceptibility to P-glycoprotein transport, and lowered brain Aβ levels in a mouse model. Intravenous infusion of TC-1 led to a significant but transient lowering of CSF and plasma Aβ levels in conscious rhesus monkeys because it underwent CYP3A4-mediated metabolism. Oral codosing of TC-1 with ritonavir, a potent CYP3A4 inhibitor, twice daily over 3.5 days in rhesus monkeys led to sustained plasma TC-1 exposure and a significant and sustained reduction in CSF sAPPβ, Aβ40, Aβ42, and plasma Aβ40 levels. CSF Aβ42 lowering showed an EC50 of ∼20 nM with respect to the CSF [TC-1] levels, demonstrating excellent concordance with its potency in a cell-based assay. These results demonstrate the first in vivo proof of concept of CSF Aβ lowering after oral administration of a BACE1 inhibitor in a nonhuman primate.


Journal of Pharmacology and Experimental Therapeutics | 2007

In Vivo β-Secretase 1 Inhibition Leads to Brain Aβ Lowering and Increased α-Secretase Processing of Amyloid Precursor Protein without Effect on Neuregulin-1

Sethu Sankaranarayanan; Eric A. Price; Guoxin Wu; Ming-Chih Crouthamel; Xiao-Ping Shi; Katherine Tugusheva; Keala X. Tyler; Jason A. Kahana; Joan D. Ellis; Lixia Jin; Thomas H. Steele; Shawn J. Stachel; Craig A. Coburn; Adam J. Simon

β-Secretase (BACE) cleavage of amyloid precursor protein (APP) is one of the first steps in the production of amyloid β peptide Aβ42, the putative neurotoxic species in Alzheimers disease. Recent studies have shown that BACE1 knockdown leads to hypomyelination, putatively caused by a decline in neuregulin (NRG)-1 processing. In this study, we have tested a potent cell-permeable BACE1 inhibitor (IC50 ∼ 30 nM) by administering it directly into the lateral ventricles of mice, expressing human wild-type (WT)-APP, to determine the consequences of BACE1 inhibition on brain APP and NRG-1 processing. BACE1 inhibition, in vivo, led to a significant dose- and time-dependent lowering of brain Aβ40 and Aβ42. BACE1 inhibition also led to a robust brain secreted (s)APPβ lowering that was accompanied by an increase in brain sAPPα levels. Although an increase in full-length NRG-1 levels was evident in 15-day-old BACE1 homozygous knockout (KO) (–/–) mice, in agreement with previous studies, this effect was also observed in 15-day-old heterozygous (+/–) mice, but it was not evident in 30-day-old and 2-year-old BACE1 KO (–/–) mice. Thus, BACE1 knockdown led to a transient decrease in NRG-1 processing in mice. Pharmacological inhibition of BACE1 in adult mice, which led to significant Aβ lowering, was without any significant effect on brain NRG-1 processing. Taken together, these results suggest that BACE1 is the major β-site cleavage enzyme for APP and that its inhibition can lower brain Aβ and redirect APP processing via the potentially nonamyloidogenic α-secretase pathway, without significantly altering NRG-1 processing.


Bioorganic & Medicinal Chemistry Letters | 2002

Indinavir analogues with blocked metabolism sites as HIV protease inhibitors with improved pharmacological profiles and high potency against PI-resistant viral strains.

Yuan Cheng; Fengqi Zhang; Thomas A. Rano; Zhijian Lu; William A. Schleif; Lori Gabryelski; David B. Olsen; Mark Stahlhut; Carrie A. Rutkowski; Jiunn H. Lin; Lixia Jin; Emilio A. Emini; Kevin T. Chapman; James R. Tata

Indinavir analogues with blocked metabolism sites show highly improved pharmacokinetic profiles in animals. The cis-aminochromanol substituted analogues exhibited excellent potency against both the wild-type (NL4-3) virus and protease inhibitor-resistant HIV strains.


Bioorganic & Medicinal Chemistry Letters | 2008

Synthesis of 5-(1-H or 1-alkyl-5-oxopyrrolidin-3-yl)-8-hydroxy-[1,6]-naphthyridine-7-carboxamide inhibitors of HIV-1 integrase

Jeffrey Y. Melamed; Melissa S. Egbertson; Sandor L. Varga; Joseph P. Vacca; Greg Moyer; Lori Gabryelski; Peter J. Felock; Kara A. Stillmock; Marc Witmer; William A. Schleif; Daria J. Hazuda; Yvonne M. Leonard; Lixia Jin; Joan D. Ellis; Steven D. Young

HIV-1 integrase catalyzes the insertion of viral DNA into the genome of the host cell. Integrase inhibitor N-(4-fluorobenzyl)-8-hydroxy-1,6-naphthyridine-7-carboxamide selectively inhibits the strand transfer process of integration. 4-Substituted pyrrolidinones possessing various groups on the pyrrolidinone nitrogen were introduced at the 5-position of the naphthyridine scaffold. These analogs exhibit excellent activity against viral replication in a cell-based assay. The preparation of these compounds was enabled by a three-step, two-pot reaction sequence from a common butenolide intermediate.


Bioorganic & Medicinal Chemistry Letters | 2003

Design and synthesis of highly potent HIV protease inhibitors with activity against resistant virus

Zhijian Lu; Subharekha Raghavan; Joann Bohn; Mark G. Charest; Mark Stahlhut; Carrie A. Rutkowski; Amy L. Simcoe; David B. Olsen; William A. Schleif; Anthony Carella; Lori Gabryelski; Lixia Jin; Jiunn H. Lin; Emilio A. Emini; Kevin T. Chapman; James R. Tata

A series of highly potent HIV protease inhibitors have been designed and synthesized. These compounds are active against various clinical viral isolates as well as wild-type virus. The synthesis and biological activity of these HIV protease inhibitors are discussed.


Bioorganic & Medicinal Chemistry Letters | 2003

The design, synthesis and evaluation of novel HIV-1 protease inhibitors with high potency against PI-resistant viral strains

Fengqi Zhang; Kevin T. Chapman; William A. Schleif; David B. Olsen; Mark Stahlhut; Carrie A. Rutkowski; Lawrence C. Kuo; Lixia Jin; Jiunn H. Lin; Emilio A. Emini; James R. Tata

Replacement of the pyridylmethyl moiety in indinavir with a pyridyl oxazole yielded HIV-1 protease inhibitors (PI) with greatly improved potency against PI-resistant HIV-1 strains. A meta-methoxy group on the pyridyl ring and a gem-dimethyl methyl linkage afforded compound 10 with notable in vitro antiviral activity against HIV-1 viral strains with reduced susceptibility to the clinically available PIs. Compound 10 also demonstrated favorable in vivo pharmacokinetics in animal models.


Bioorganic & Medicinal Chemistry Letters | 2003

HIV protease inhibitors with picomolar potency against PI-Resistant HIV-1 by extension of the P3 substituent.

Joseph L. Duffy; Thomas A. Rano; Nancy J. Kevin; Kevin T. Chapman; William A. Schleif; David B. Olsen; Mark Stahlhut; Carrie A. Rutkowski; Lawrence C. Kuo; Lixia Jin; Jiunn H. Lin; Emilio A. Emini; James R. Tata

A biaryl pyridylfuran P(3) substituent on the hydroxyethylene isostere scaffold affords HIV protease inhibitors (PIs) with picomolar (IC(50)) potency against the protease enzymes from PI-resistant HIV-1 strains. Inclusion of a gem-dimethyl substituent afforded compound 3 with 100% oral bioavailability (dogs) and more than double the t(1/2) of indinavir. Inhibition of multiple P450 isoforms is dependent on the regiochemistry of the pyridyl nitrogen in these compounds.


Bioorganic & Medicinal Chemistry Letters | 2002

Synthesis and Activity of Novel HIV Protease Inhibitors with Improved Potency Against Multiple PI-Resistant Viral Strains

Joseph L. Duffy; Nancy J. Kevin; Brian A. Kirk; Kevin T. Chapman; William A. Schleif; David B. Olsen; Mark Stahlhut; Carrie A. Rutkowski; Lawrence C. Kuo; Lixia Jin; Jiunn H. Lin; Emilio A. Emini; James R. Tata

Substitution of the t-butylcarboxamide substituent in analogues of the HIV protease inhibitor (PI) Indinavir with a trifluoroethylamide moiety confers greater potency against both the wild-type (NL4-3) virus and PI-resistant HIV. The trifluoroethyl substituent also affords a slower clearance rate in vivo (dogs); however, this may be due to more potent inhibition of at least two P450 isoforms.


Xenobiotica | 2003

Interaction with indinavir to enhance systemic exposure of an investigational HIV protease inhibitor in rats, dogs and monkeys

Lixia Jin; I-Wu Chen; Masato Chiba; Jiunn H. Lin

1. The use of a beneficial interaction between indinavir and compound A, a potent investigational HIV protease inhibitor to enhance systemic exposure of compound A, was investigated. 2. When administrated alone, compound A underwent extensive hepatic first-pass metabolism in rats and monkeys, resulting in low oral bioavailability. 3. In vitro studies with liver microsomes revealed that compound A metabolism was mediated exclusively by CYP3A enzymes in rats, dogs and monkeys. Indinavir, which also was metabolized predominantly by CYP3A enzymes, extensively inhibited compound A metabolism in microsomes, whereas compound A showed weak inhibitory potency on indinavir metabolism. 4. Consistent with in vitro observations, co-administration of the two compounds resulted in a 17-fold increase in oral AUC of compound A in rats owing to the inhibition of metabolism of compound A by indinavir, whereas compound A did not affect indinavir metabolism as indicated by the unchanged indinavir AUC. Similarly, the systemic exposure of compound A in dogs and monkeys was increased substantially following oral co-administration with indinavir by 7- and > 50-fold, respectively. 5. Enhancement in compound A systemic exposure by indinavir in humans, as predicted based on the in vivo animal and in vitro human liver microsomal data, was confirmed in subsequent clinical studies.


Xenobiotica | 2015

Pharmacokinetics and metabolism of AMG 232, a novel orally bioavailable inhibitor of the MDM2–p53 interaction, in rats, dogs and monkeys: in vitro–in vivo correlation

Qiuping Ye; Min Jiang; Wotang T. Huang; Yun Ling; Steven H. Olson; Daqing Sun; Guifen Xu; Xuelei Yan; Bradley K. Wong; Lixia Jin

Abstract 1. AMG 232 is a novel inhibitor of the p53–MDM2 protein–protein interaction currently in Phase I clinical trials for multiple tumor indications. The objectives of the investigations reported in this article were to characterize the pharmacokinetic and drug metabolism properties of AMG 232 in pre-clinical species in vivo and in vitro, and in humans in vitro, and to predict its pharmacokinetics in humans through integrating PKDM data. 2. AMG 232 exhibited low clearance (<0.25 × Qh) and moderate to high oral bioavailability in mice, rats and monkeys (>42%), but high clearance (0.74 × Qh) and low oral exposure in dogs (18%). 3. Biotransformation was the major route of elimination of AMG 232 in rats, with only 7% of intravenously administered 14C-labeled AMG 232 recovered as parent molecule in bile. The major metabolite was an acyl glucuronide as measured by in vivo rat studies and in vitro hepatocyte incubations in multiple species. 4. The in vitro–in vivo correlation of AMG 232 clearance was within 2-fold in pre-clinical species using hepatocytes. AMG 232 was predicted to exhibit low clearance, high volume distribution and long half-life in humans. The predictions are consistent with the preliminary human pharmacokinetic parameters of AMG 232 in clinical trials.

Collaboration


Dive into the Lixia Jin's collaboration.

Top Co-Authors

Avatar

William A. Schleif

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Jiunn H. Lin

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Joan D. Ellis

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Emilio A. Emini

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

David B. Olsen

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Carrie A. Rutkowski

United States Military Academy

View shared research outputs
Researchain Logo
Decentralizing Knowledge