Network


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

Hotspot


Dive into the research topics where Peter T. W. Cheng is active.

Publication


Featured researches published by Peter T. W. Cheng.


Angewandte Chemie | 2016

Ligand‐Promoted Borylation of C(sp3)H Bonds with Palladium(II) Catalysts

Jian He; Heng Jiang; Ryosuke Takise; Ru-Yi Zhu; Gang Chen; Hui‐Xiong Dai; T. G. Murali Dhar; Jun Shi; Hao Zhang; Peter T. W. Cheng; Jin-Quan Yu

A quinoline-based ligand effectively promotes the palladium-catalyzed borylation of C(sp(3))-H bonds. Primary β-C(sp(3))-H bonds in carboxylic acid derivatives as well as secondary C(sp(3))-H bonds in a variety of carbocyclic rings, including cyclopropanes, cyclobutanes, cyclopentanes, cyclohexanes, and cycloheptanes, can thus be borylated. This directed borylation method complements existing iridium(I)- and rhodium(I)-catalyzed C-H borylation reactions in terms of scope and operational conditions.


Tetrahedron Letters | 1997

A practical method for the preparation of α′-chloroketones of N-carbamate protected-α-aminoacids

Ping Chen; Peter T. W. Cheng; Steven H. Spergel; Robert Zahler; Xuebao Wang; John K. Thottathil; Joel C. Barrish; Richard Polniaszek

Abstract A practical method for the preparation of α-N-BOC-epoxides from protected amino acid esters based on the Kowalski homologation reaction is described. This procedure can be readily performed on a large scale without the use of hazardous reagents and has allowed preparation of epoxides 3 in multi-kilogram quantities.


Mini-reviews in Medicinal Chemistry | 2005

PPARs as targets for metabolic and cardiovascular diseases.

Peter T. W. Cheng; Ranjan Mukherjee

Peroxisome proliferator-activated receptors (PPARs) alpha, gamma and delta (beta) are ligand-activated transcription factors of the nuclear hormone receptor superfamily which have been shown to play key roles in maintaining glucose and lipid homeostasis. The physiological effects of several marketed drugs for the treatment of dyslipidemia (fenofibrate and gemfibrozil) and diabetes (rosiglitazone and pioglitazone) have now been shown to be mediated through PPARalpha and PPARgamma respectively. Over the past few years our understanding of how PPAR ligands and receptors modulate gene expression has greatly increased; this knowledge is being used to design even more potent and efficacious PPAR ligands for the treatment of diabetes, dyslipidemia, atherosclerosis and obesity. This review is a brief survey of the PPAR field which highlights recent progress, with an emphasis on new ligands with novel PPAR profiles, particularly compounds which are co-agonists of PPAalpha, gamma and beta (delta).


Drug Metabolism and Disposition | 2005

STRUCTURAL ELUCIDATION OF HUMAN OXIDATIVE METABOLITES OF MURAGLITAZAR: USE OF MICROBIAL BIOREACTORS IN THE BIOSYNTHESIS OF METABOLITE STANDARDS

Donglu Zhang; Haiying Zhang; Nelly Aranibar; Ronald L. Hanson; Yande Huang; Peter T. W. Cheng; Shung Wu; Samuel J. Bonacorsi; Mingshe Zhu; Arun Swaminathan; W. Griffith Humphreys

Muraglitazar (Pargluva), a dual α/γ peroxisome proliferator-activated receptor activator, is currently in clinical development for treatment of type 2 diabetes. This study describes the structural elucidation of the human oxidative metabolites of muraglitazar through the use of a combination of microbial bioreactors, NMR and accurate mass analyses, and organic synthesis. Plasma, urine, and feces were collected from six healthy subjects following oral administration of 14C-labeled muraglitazar (10 mg, 100 μCi) and pooled samples were analyzed. Approximately 96% of the recovered radioactive dose was found in the feces and 3.5% in the urine. The parent compound represented >85% of the radioactivity in plasma. The fecal radioactivity was distributed among 16 metabolites (M1–M12, M14–M16, and M8a) and the parent drug, of which hydroxylation and O-demethylation metabolites (M5, M10, M11, M14, and M15) represented the prominent human metabolites. The urinary radioactivity was distributed into several peaks including muraglitazar glucuronide (M13) and the parent drug. Low concentrations of metabolites in human samples prevented direct identification of metabolites beyond liquid chromatographic (LC)-mass spectrometric analysis. Microbial strains Cunninghamella elegans and Saccharopolyspora hirsuta produced muraglitazar metabolites that had the same high performance liquid chromatography retention times and the same tandem mass spectrometric (MS/MS) properties as the corresponding human metabolites. The microbial metabolites M9, M10, M11, M14, M15, and M16 were isolated and analyzed by NMR. Based on these LC-MS/MS and NMR analyses, and organic synthesis, the structures of 16 human oxidative metabolites were identified. The oxidative metabolism of muraglitazar was characterized by hydroxylation, O-demethylation, oxazolering opening, and O-demethylation/hydroxylation, as well as O-dealkylation and carboxylic acid formation. This study demonstrated the utility of microbial bioreactors for the identification of metabolites.


Drug Metabolism and Disposition | 2006

Comparative Metabolism of Radiolabeled Muraglitazar in Animals and Humans by Quantitative and Qualitative Metabolite Profiling

Donglu Zhang; Lifei Wang; Nirmala Raghavan; Haiying Zhang; Wenying Li; Peter T. W. Cheng; Ming Yao; Litao Zhang; Mingshe Zhu; Samuel J. Bonacorsi; Suresh Yeola; James Mitroka; Narayanan Hariharan; Vinayak Hosagrahara; Gamini Chandrasena; Wen Chyi Shyu; W. Griffith Humphreys

Muraglitazar (Pargluva), a dual α/γ peroxisome proliferator-activated receptor (PPAR) activator, has both glucose- and lipid-lowering effects in animal models and in patients with diabetes. This study describes the in vivo and in vitro comparative metabolism of [14C]muraglitazar in rats, dogs, monkeys, and humans by quantitative and qualitative metabolite profiling. Metabolite identification and quantification methods used in these studies included liquid chromatography/mass spectrometry (LC/MS), LC/tandem MS, LC/radiodetection, LC/UV, and a newly described mass defect filtering technique in conjunction with high resolution MS. After oral administration of [14C]muraglitazar, absorption was rapid in all species, reaching a concentration peak for parent and total radioactivity in plasma within 1 h. The most abundant component in plasma at all times in all species was the parent drug, and no metabolite was present in greater than 2.5% of the muraglitazar concentrations at 1 h postdose in rats, dogs, and humans. All metabolites observed in human plasma were also present in rats, dogs, or monkeys. Urinary excretion of radioactivity was low (<5% of the dose) in all intact species, and the primary route of elimination was via biliary excretion in rats, monkeys, and humans. Based on recovered doses in urine and bile, muraglitazar showed a very good absorption in rats, monkeys, and humans. The major drug-related components in bile of rats, monkeys, and humans were glucuronides of muraglitazar and its oxidative metabolites. The parent compound was a minor component in bile, suggesting extensive metabolism of the drug. In contrast, the parent drug and oxidative metabolites were the major components in feces, and no glucuronide conjugates were found, suggesting that glucuronide metabolites were excreted in bile and hydrolyzed in the gastrointestinal tract. The metabolites of muraglitazar resulted from both glucuronidation and oxidation. The metabolites in general had greatly reduced activity as PPARα/γ activators relative to muraglitazar. In conclusion, muraglitazar was rapidly absorbed, extensively metabolized through glucuronidation and oxidation, and mainly eliminated in the feces via biliary excretion of glucuronide metabolites in all species studied. Disposition and metabolic pathways were qualitatively similar in rats, dogs, monkeys, and humans.


Journal of Pharmacology and Experimental Therapeutics | 2008

Novel Peroxisome Proliferator-Activated Receptor α Agonists Lower Low-Density Lipoprotein and Triglycerides, Raise High-Density Lipoprotein, and Synergistically Increase Cholesterol Excretion with a Liver X Receptor Agonist

Ranjan Mukherjee; Kenneth T. Locke; Bowman Miao; Daniel Meyers; Hossain Monshizadegan; Rongan Zhang; Debra Search; Denise Grimm; Michael Flynn; Kevin M. O'Malley; Litao Zhang; Jun Li; Yan Shi; Lawrence J. Kennedy; Michael A. Blanar; Peter T. W. Cheng; Joseph A. Tino; Rai Ajit Srivastava

The first generation peroxisome proliferator-activated receptor (PPAR) α agonist gemfibrozil reduces the risk of major cardiovascular events; therefore, more potent PPARα agonists for the treatment of cardiovascular diseases have been actively sought. We describe two novel, potent oxybenzylglycine PPARα-selective agonists, BMS-687453 [N-[[3-[[2-(4-chlorophenyl)-5-methyl-4-oxazolyl]methoxy]phenyl]methyl]-N-(methoxycarbonyl)-glycine] and BMS-711939 N-[[5-[[2-(4-chlorophenyl)-5-methyl-4-oxazolyl]methoxy]-2-fluorophenyl]methyl]-N-(methoxycarbonyl)-glycine], that robustly increase apolipoprotein (Apo) A1 and high-density lipoprotein cholesterol in human ApoA1 transgenic mice and lower low-density lipoprotein-cholesterol and triglycerides in fat-fed hamsters. These compounds have much lower potency against mouse PPARα than human PPARα; therefore, they were tested in PPARα-humanized mice that do not express murine PPARα but express human PPARα selectively in the liver. We developed hepatic gene induction as a novel biomarker for efficacy and demonstrate hepatic gene induction at very low doses of these compounds. BMS-711939 induces fecal cholesterol excretion, which is further increased upon cotreatment with a liver X receptor (LXR) agonist. It is surprising that this synergistic increase upon coadministration is also observed in mice that express PPARα in the liver only. BMS-711939 also prevented the LXR agonist-induced elevation of serum triglycerides. Such PPARα agonists could be attractive candidates to explore for the treatment of cardiovascular diseases, especially in combination with a suitable LXR agonist.


Bioorganic & Medicinal Chemistry Letters | 2009

Design, synthesis and structure-activity relationships of azole acids as novel, potent dual PPAR alpha/gamma agonists.

Hongjian Zhang; Denis E. Ryono; Pratik Devasthale; Wei Wang; K O'Malley; Dennis Farrelly; Liqun Gu; Tom Harrity; Michael Cap; Cuixia Chu; Kenneth T. Locke; Litao Zhang; Jonathan Lippy; Lori Kunselman; Nathan Morgan; Neil Flynn; Lisa Moore; Hosagrahara; Pathanjali Kadiyala; Cen Xu; Arthur M. Doweyko; A Bell; Jodi K. Muckelbauer; Robert Zahler; Narayanan Hariharan; Peter T. W. Cheng

The design, synthesis and structure-activity relationships of a novel series of N-phenyl-substituted pyrrole, 1,2-pyrazole and 1,2,3-triazole acid analogs as PPAR ligands are outlined. The triazole acid analogs 3f and 4f were identified as potent dual PPARalpha/gamma agonists both in binding and functional assays in vitro. The 3-oxybenzyl triazole acetic acid analog 3f showed excellent glucose and triglyceride lowering in diabetic db/db mice.


Drug Metabolism and Disposition | 2005

Glucuronidation as a major metabolic clearance pathway of 14C-labeled muraglitazar in humans : Metabolic profiles in subjects with or without bile collection

Lifei Wang; Donglu Zhang; Arun Swaminathan; Yongjun Xue; Peter T. W. Cheng; Shung Wu; Rogelio Mosqueda-Garcia; Catherine Aurang; Donald W. Everett; W. Griffith Humphreys

The metabolism and disposition of 14C-labeled muraglitazar (Pargluva), a novel dual α/γ peroxisome proliferator-activated receptor activator, was investigated in eight healthy male subjects with and without bile collection (groups 1 and 2) after a single 20-mg oral dose. Bile samples were collected for 3 to 8 h after dosing from group 2 subjects in addition to the urine and feces collection. In plasma, the parent compound was the major component, and circulating metabolites, including several glucuronide conjugates, were minor components at all time points. The exposure to parent drug (Cmax and area under the plasma concentration versus time curve) in subjects with bile collection was generally lower than that in subjects without bile collection. The major portion of the radioactive dose was recovered in feces (91% for group 1 and 51% for group 2). In addition, 40% of the dose was recovered in the bile from group 2 subjects. In this 3- to 8-h bile, the glucuronide of muraglitazar (M13, 15% of dose) and the glucuronides of its oxidative metabolites (M17a,b,c, M18a,b,c, and M20, together, 16% of dose) accounted for approximately 80% of the biliary radioactivity; muraglitazar and its O-demethylated metabolite (M15) each accounted for approximately 4% of the dose. In contrast, fecal samples only contained muraglitazar and its oxidative metabolites, suggesting hydrolysis of biliary glucuronides in the intestine before fecal excretion. Thus, the subjects with and without bile collection showed different metabolic profiles of muraglitazar after oral administration, and glucuronidation was not observed as a major pathway of metabolic clearance from subjects with the conventional urine and fecal collection, but was found as a major elimination pathway from subjects with bile collection.


Journal of Medicinal Chemistry | 2010

Discovery of an oxybenzylglycine based peroxisome proliferator activated receptor alpha selective agonist 2-((3-((2-(4-chlorophenyl)-5-methyloxazol-4-yl)methoxy)benzyl)(methoxycarbonyl)amino)acetic acid (BMS-687453).

Jun Li; Lawrence J. Kennedy; Yan Shi; Shiwei Tao; Xiang-Yang Ye; Stephanie Y. Chen; Ying Wang; Andres S. Hernandez; Wei Wang; Pratik Devasthale; Sean Chen; Zhi Lai; Hao Zhang; Shung Wu; Rebecca A. Smirk; Scott A. Bolton; Denis E. Ryono; Huiping Zhang; Ngiap-Kie Lim; Bang-Chi Chen; Kenneth T. Locke; Kevin O’Malley; Litao Zhang; Rai Ajit Srivastava; Bowman Miao; Daniel Meyers; Hossain Monshizadegan; Debra Search; Denise Grimm; Rongan Zhang

An 1,3-oxybenzylglycine based compound 2 (BMS-687453) was discovered to be a potent and selective peroxisome proliferator activated receptor (PPAR) alpha agonist, with an EC(50) of 10 nM for human PPARalpha and approximately 410-fold selectivity vs human PPARgamma in PPAR-GAL4 transactivation assays. Similar potencies and selectivity were also observed in the full length receptor co-transfection assays. Compound 2 has negligible cross-reactivity against a panel of human nuclear hormone receptors including PPARdelta. Compound 2 demonstrated an excellent pharmacological and safety profile in preclinical studies and thus was chosen as a development candidate for the treatment of atherosclerosis and dyslipidemia. The X-ray cocrystal structures of the early lead compound 12 and compound 2 in complex with PPARalpha ligand binding domain (LBD) were determined. The role of the crystal structure of compound 12 with PPARalpha in the development of the SAR that ultimately resulted in the discovery of compound 2 is discussed.


Bioorganic & Medicinal Chemistry Letters | 2008

Discovery of azetidinone acids as conformationally-constrained dual PPARα/γ agonists

Wei Wang; Pratik Devasthale; Dennis Farrelly; Liqun Gu; Thomas Harrity; Michael Cap; Cuixia Chu; Lori Kunselman; Nathan Morgan; Randy Ponticiello; Rachel Zebo; Litao Zhang; Kenneth T. Locke; Jonathan Lippy; Kevin O’Malley; Vinayak Hosagrahara; Lisa Zhang; Pathanjali Kadiyala; Chiehying Chang; Jodi K. Muckelbauer; Arthur M. Doweyko; Robert Zahler; Denis E. Ryono; Narayanan Hariharan; Peter T. W. Cheng

A novel class of azetidinone acid-derived dual PPARalpha/gamma agonists has been synthesized for the treatment of diabetes and dyslipidemia. The preferred stereochemistry in this series for binding and functional agonist activity against both PPARalpha and PPARgamma receptors was shown to be 3S,4S. Synthesis, in vitro and in vivo activities of compounds in this series are described. A high-yielding method for N-arylation of azetidinone esters is also described.

Collaboration


Dive into the Peter T. W. Cheng's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yan Shi

Bristol-Myers Squibb

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wei Wang

Bristol-Myers Squibb

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge