Network


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

Hotspot


Dive into the research topics where Tian Wu is active.

Publication


Featured researches published by Tian Wu.


Journal of Medicinal Chemistry | 2011

Discovery and Optimization of a Series of Benzothiazole Phosphoinositide 3-Kinase (PI3K)/Mammalian Target of Rapamycin (mTOR) Dual Inhibitors

Noel D. D’Angelo; Tae-Seong Kim; Kristin L. Andrews; Shon Booker; Sean Caenepeel; Kui Chen; Derin C. D’Amico; Daniel J. Freeman; Jian Jiang; Longbin Liu; John D. McCarter; Tisha San Miguel; Erin L. Mullady; Michael L. Schrag; Raju Subramanian; Jin Tang; Robert C. Wahl; Ling Wang; Douglas A. Whittington; Tian Wu; Ning Xi; Yang Xu; Peter Yakowec; Kevin Yang; Leeanne Zalameda; Nancy R. Zhang; Paul E. Hughes; Mark H. Norman

Phosphoinositide 3-kinase α (PI3Kα) is a lipid kinase that plays a key regulatory role in several cellular processes. The mutation or amplification of this kinase in humans has been implicated in the growth of multiple tumor types. Consequently, PI3Kα has become a target of intense research for drug discovery. Our studies began with the identification of benzothiazole compound 1 from a high throughput screen. Extensive SAR studies led to the discovery of sulfonamide 45 as an early lead, based on its in vitro cellular potency. Subsequent modifications of the central pyrimidine ring dramatically improved enzyme and cellular potency and led to the identification of chloropyridine 70. Further arylsulfonamide SAR studies optimized in vitro clearance and led to the identification of 82 as a potent dual inhibitor of PI3K and mTOR. This molecule exhibited potent enzyme and cell activity, low clearance, and high oral bioavailability. In addition, compound 82 demonstrated tumor growth inhibition in U-87 MG, A549, and HCT116 tumor xenograft models.


Journal of Medicinal Chemistry | 2012

Structure-based design of a novel series of potent, selective inhibitors of the class I phosphatidylinositol 3-kinases.

Adrian L. Smith; Noel D'angelo; Yunxin Y. Bo; Shon Booker; Victor J. Cee; Brad Herberich; Fang-Tsao Hong; Claire L.M. Jackson; Brian A. Lanman; Longbin Liu; Nobuko Nishimura; Liping H. Pettus; Anthony B. Reed; Seifu Tadesse; Nuria A. Tamayo; Ryan Wurz; Kevin Yang; Kristin L. Andrews; Douglas A. Whittington; John D. McCarter; Tisha San Miguel; Leeanne Zalameda; Jian Jiang; Raju Subramanian; Erin L. Mullady; Sean Caenepeel; Daniel J. Freeman; Ling Wang; Nancy R. Zhang; Tian Wu

A highly selective series of inhibitors of the class I phosphatidylinositol 3-kinases (PI3Ks) has been designed and synthesized. Starting from the dual PI3K/mTOR inhibitor 5, a structure-based approach was used to improve potency and selectivity, resulting in the identification of 54 as a potent inhibitor of the class I PI3Ks with excellent selectivity over mTOR, related phosphatidylinositol kinases, and a broad panel of protein kinases. Compound 54 demonstrated a robust PD-PK relationship inhibiting the PI3K/Akt pathway in vivo in a mouse model, and it potently inhibited tumor growth in a U-87 MG xenograft model with an activated PI3K/Akt pathway.


Journal of Medicinal Chemistry | 2012

Selective Class I Phosphoinositide 3-Kinase Inhibitors: Optimization of a Series of Pyridyltriazines Leading to the Identification of a Clinical Candidate, AMG 511

Mark H. Norman; Kristin L. Andrews; Yunxin Y. Bo; Shon Booker; Sean Caenepeel; Victor J. Cee; Noel D. D’Angelo; Daniel J. Freeman; Bradley J. Herberich; Fang-Tsao Hong; Claire L.M. Jackson; Jian Jiang; Brian A. Lanman; Longbin Liu; John D. McCarter; Erin L. Mullady; Nobuko Nishimura; Liping H. Pettus; Anthony B. Reed; Tisha San Miguel; Adrian L. Smith; Markian Stec; Seifu Tadesse; Andrew Tasker; Divesh Aidasani; Xiaochun Zhu; Raju Subramanian; Nuria A. Tamayo; Ling Wang; Douglas A. Whittington

The phosphoinositide 3-kinase family catalyzes the phosphorylation of phosphatidylinositol-4,5-diphosphate to phosphatidylinositol-3,4,5-triphosphate, a secondary messenger which plays a critical role in important cellular functions such as metabolism, cell growth, and cell survival. Our efforts to identify potent, efficacious, and orally available phosphatidylinositol 3-kinase (PI3K) inhibitors as potential cancer therapeutics have resulted in the discovery of 4-(2-((6-methoxypyridin-3-yl)amino)-5-((4-(methylsulfonyl)piperazin-1-yl)methyl)pyridin-3-yl)-6-methyl-1,3,5-triazin-2-amine (1). In this paper, we describe the optimization of compound 1, which led to the design and synthesis of pyridyltriazine 31, a potent pan inhibitor of class I PI3Ks with a superior pharmacokinetic profile. Compound 31 was shown to potently block the targeted PI3K pathway in a mouse liver pharmacodynamic model and inhibit tumor growth in a U87 malignant glioma glioblastoma xenograft model. On the basis of its excellent in vivo efficacy and pharmacokinetic profile, compound 31 was selected for further evaluation as a clinical candidate and was designated AMG 511.


ACS Medicinal Chemistry Letters | 2015

Oxopyrido[2,3-d]pyrimidines as Covalent L858R/T790M Mutant Selective Epidermal Growth Factor Receptor (EGFR) Inhibitors

Ryan Wurz; Liping H. Pettus; Kate S. Ashton; James Brown; Jian Jeffrey Chen; Brad Herberich; Fang-Tsao Hong; Essa Hu-Harrington; Tom Nguyen; David J. St. Jean; Seifu Tadesse; David Bauer; Michele Kubryk; Jinghui Zhan; Keegan Cooke; Petia Mitchell; Kristin L. Andrews; Faye Hsieh; Dean Hickman; Nataraj Kalyanaraman; Tian Wu; Darren L. Reid; Edward K. Lobenhofer; Dina A. Andrews; Nancy E. Everds; Roberto E. Guzman; Andrew T. Parsons; Simon J. Hedley; Jason Tedrow; Oliver R. Thiel

In nonsmall cell lung cancer (NSCLC), the threonine(790)-methionine(790) (T790M) point mutation of EGFR kinase is one of the leading causes of acquired resistance to the first generation tyrosine kinase inhibitors (TKIs), such as gefitinib and erlotinib. Herein, we describe the optimization of a series of 7-oxopyrido[2,3-d]pyrimidinyl-derived irreversible inhibitors of EGFR kinase. This led to the discovery of compound 24 which potently inhibits gefitinib-resistant EGFR(L858R,T790M) with 100-fold selectivity over wild-type EGFR. Compound 24 displays strong antiproliferative activity against the H1975 nonsmall cell lung cancer cell line, the first line mutant HCC827 cell line, and promising antitumor activity in an EGFR(L858R,T790M) driven H1975 xenograft model sparing the side effects associated with the inhibition of wild-type EGFR.


Journal of Medicinal Chemistry | 2016

Discovery and Optimization of Quinazolinone-pyrrolopyrrolones as Potent and Orally Bioavailable Pan-Pim Kinase Inhibitors

Liping H. Pettus; Kristin L. Andrews; Shon Booker; Jie Chen; Victor J. Cee; Frank Chavez; Yuping Chen; Heather Eastwood; Nadia Guerrero; Bradley J. Herberich; Dean Hickman; Brian A. Lanman; Jimmy Laszlo; Matthew R. Lee; J. Russell Lipford; Bethany Mattson; Christopher Mohr; Yen Nguyen; Mark H. Norman; David Powers; Anthony B. Reed; Karen Rex; Christine Sastri; Nuria A. Tamayo; Paul Wang; Jeffrey T. Winston; Bin Wu; Tian Wu; Ryan Wurz; Yang Xu

The high expression of proviral insertion site of Moloney murine leukemia virus kinases (Pim-1, -2, and -3) in cancers, particularly the hematopoietic malignancies, is believed to play a role in promoting cell survival and proliferation while suppressing apoptosis. The three isoforms of Pim protein appear largely redundant in their oncogenic functions. Thus, a pan-Pim kinase inhibitor is highly desirable. However, cell active pan-Pim inhibitors have proven difficult to develop because Pim-2 has a low Km for ATP and therefore requires a very potent inhibitor to effectively block the kinase activity at cellular ATP concentrations. Herein, we report a series of quinazolinone-pyrrolopyrrolones as potent and selective pan-Pim inhibitors. In particular, compound 17 is orally efficacious in a mouse xenograft model (KMS-12 BM) of multiple myeloma, with 93% tumor growth inhibition at 50 mg/kg QD upon oral dosing.


ACS Medicinal Chemistry Letters | 2016

Discovery and Optimization of Macrocyclic Quinoxaline-pyrrolo-dihydropiperidinones as Potent Pim-1/2 Kinase Inhibitors.

Victor J. Cee; Frank Chavez; Bradley J. Herberich; Brian A. Lanman; Liping H. Pettus; Anthony B. Reed; Bin Wu; Ryan Wurz; Kristin L. Andrews; Jie Chen; Dean Hickman; Jimmy Laszlo; Matthew R. Lee; Nadia Guerrero; Bethany Mattson; Yen Nguyen; Christopher Mohr; Karen Rex; Christine Sastri; Paul Wang; Qiong Wu; Tian Wu; Yang Xu; Yihong Zhou; Jeffrey T. Winston; J. Russell Lipford; Andrew Tasker; Hui-Ling Wang

The identification of Pim-1/2 kinase overexpression in B-cell malignancies suggests that Pim kinase inhibitors will have utility in the treatment of lymphoma, leukemia, and multiple myeloma. Starting from a moderately potent quinoxaline-dihydropyrrolopiperidinone lead, we recognized the potential for macrocyclization and developed a series of 13-membered macrocycles. The structure-activity relationships of the macrocyclic linker were systematically explored, leading to the identification of 9c as a potent, subnanomolar inhibitor of Pim-1 and -2. This molecule also potently inhibited Pim kinase activity in KMS-12-BM, a multiple myeloma cell line with relatively high endogenous levels of Pim-1/2, both in vitro (pBAD IC50 = 25 nM) and in vivo (pBAD EC50 = 30 nM, unbound), and a 100 mg/kg daily dose was found to completely arrest the growth of KMS-12-BM xenografts in mice.


Bioorganic & Medicinal Chemistry Letters | 2015

The imidazo[1,2-a]pyridine ring system as a scaffold for potent dual phosphoinositide-3-kinase (PI3K)/mammalian target of rapamycin (mTOR) inhibitors.

Markian Stec; Kristin L. Andrews; Yunxin Bo; Sean Caenepeel; Hongyu Liao; John D. McCarter; Erin L. Mullady; Tisha San Miguel; Raju Subramanian; Nuria A. Tamayo; Douglas A. Whittington; Ling Wang; Tian Wu; Leeanne Zalameda; Nancy Zhang; Paul E. Hughes; Mark H. Norman

Based on lead compound 1, which was discovered from a high-throughput screen, a series of PI3Kα/mTOR inhibitors were evaluated that contained an imidazo[1,2-a]pyridine as a core replacement for the benzimidazole contained in 1. By exploring various ring systems that occupy the affinity pocket, two fragments containing a methoxypyridine were identified that gave <100 nM potency toward PI3Kα in enzyme and cellular assays with moderate stability in rat and human liver microsomes. With the two methoxypyridine groups selected to occupy the affinity pocket, analogs were prepared with various fragments intended to occupy the ribose pocket of PI3Kα and mTOR. From these analogs, tertiary alcohol 18 was chosen for in vivo pharmacodynamic evaluation based on its potency in the PI3Kα cellular assay, microsomal stability, and in vivo pharmacokinetic properties. In a mouse liver pharmacodynamic assay, compound 18 showed 56% inhibition of HFG-induced AKT (Ser473) phosphorylation at a 30 mg/kg dose.


Toxicological Sciences | 2014

P450-Mediated O-Demethylated Metabolite Is Responsible for Rat Hepatobiliary Toxicity of Pyridyltriazine-Containing PI3K Inhibitors

Raju Subramanian; Divesh Aidasani; Keith Bailey; Dan Branstetter; Nancy E. Everds; Jian Jiang; Mark H. Norman; Ronya Primack; Gary L. Skiles; Irene Soto; Markian Stec; Mylo Wagner; Tian Wu; Xiaochun Zhu; Hervé Lebrec

The dysregulation of phosphatidylinositol 3-kinase (PI3K)-dependent pathways is implicated in several human cancers making it an attractive target for small molecule PI3K inhibitors. A series of potent pyridyltriazine-containing inhibitors of class Ia PI3Ks were synthesized and a subset of compounds was evaluated in exploratory repeat-dose rat toxicology studies. Daily oral dosing of compound 1: in Sprague Dawley rats for four consecutive days was associated with hepatobiliary toxicity that included biliary epithelial hyperplasia and hypertrophy, periductular edema, biliary stasis, and acute peribiliary inflammatory infiltrates. These histological changes were associated with clinical pathology changes that included increased serum liver enzymes, total bile acids, and bilirubin. The predominant clearance pathway of 1: was shown in vitro and in a bile-duct cannulated rat (14)C-ADME study to be P450-mediated oxidative metabolism. An O-demethylated pyridine metabolite, M3: , was identified as a candidate proximal metabolite that caused the hepatotoxicity. Co-administration of the pan-P450 inhibitor 1-aminobenzotriazole with 1: to rats significantly reduced the formation of M3: and prevented liver toxicity, whereas direct administration of M3: reproduced the toxicity. Structural changes were introduced to 1: to make the methoxypyridine ring less susceptible to P450 oxidation (compound 2: ), and addition of a methyl group to the benzylic carbon (compound 3: ) improved the pharmacokinetic profile. These changes culminated in the successful design of a clinical candidate 3: (AMG 511) that was devoid of liver toxicity in a 14-day rat toxicity study. Herein, we describe how a metabolism-based structure-activity relationship analysis allowed for the successful identification of a PI3K inhibitor devoid of off-target toxicity.


Cancer Research | 2015

Abstract 5398: In vivo development of pan-Pim kinase small molecule inhibitors

Bethany Mattson; Christine Sastri; Nadia Guerrero; Dean Hickman; Jie Chen; Tian Wu; Hui-Ling Wang; Andrew Taskar; Brian A. Lanman; Anthony B. Reed; Jude Canon; J. Russell Lipford; Karen Rex

Pim-1,-2, and -3 are constitutively active serine-threonine kinases which are partially redundant and regulate multiple pathways important for tumor growth and survival. One or more of the human Pims are over-expressed in multiple hematological tumor types (e.g. multiple myeloma (MM), NHL and AML) and in some solid tumors (e.g. prostate and SCLC). Pim over-expression correlates with malignancy and poor prognosis in several indications. Our goal was to generate a pan Pim kinase inhibitor with acceptable physical chemical properties and in vivo anti-tumor efficacy. Here we present data on two ATP-competitive, orally bioavailable pan Pim inhibitors, Compound I and Compound II. These inhibitors have potent enzymatic and cellular activity, acceptable pharmacokinetic properties (PK) and robust in vivo efficacy. In a kinase enzyme assay Compound I inhibits Pim-1 and Pim-2 activity with 0.4 nM and 0.7 nM IC50s, respectively, while Compound II is even more potent with Pim-1 and Pim-2 IC50s of 0.1 nM/0.1 nM. In a cellular assay which measures inhibition of the Pim downstream substrate phospho-BAD (p-BAD), compounds I and II demonstrate IC50s of 56 and 16 nM, respectively. In an in vivo pharmacodynamic assay (PD) to demonstrate on-target Pim activity, compounds I and II significantly inhibited p-BAD in KMS-12-BM multiple myeloma tumors for 16 hours post dose. Treatment of KMS-12-BM tumor xenografts with Compound I demonstrated robust in vivo anti-tumor efficacy resulting in 23% tumor regression at 50 mg/kg BID and tumor stasis at 100 mg/kg QD. Compound II demonstrated improved PK properties leading to greater anti-tumor efficacy of 33% tumor regression at 100 mg/kg QD and tumor stasis at 50 mg/kg QD. Compound II showed efficacy in an orthotopic model of multiple myeloma and in models of AML and DLBCL. Combination treatment of Compound II and the standard of care Dexamethasone in the multiple myeloma RPMI-8226 xenograft model demonstrated enhanced tumor growth inhibition compared to either single agent activity. In summary, Compound I and II are potent and selective inhibitors of Pim kinases with excellent in vivo properties. Pim kinase inhibitors, either as monotherapy or in combination with dexamethasone, may be effective clinical strategies for certain cancer patients. Citation Format: Bethany Mattson, Christine. E. Sastri, Nadia Guerrero, Dean Hickman, Jie Chen, Tian Wu, Hui-Ling Wang, Andrew Taskar, Brian Lanman, Anthony B. Reed, Jude Canon, J. Russell Lipford, Karen Rex. In vivo development of pan-Pim kinase small molecule inhibitors. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 5398. doi:10.1158/1538-7445.AM2015-5398


Bioorganic & Medicinal Chemistry Letters | 2014

Phosphoinositide-3-kinase inhibitors: Evaluation of substituted alcohols as replacements for the piperazine sulfonamide portion of AMG 511

Brian A. Lanman; Anthony B. Reed; Victor J. Cee; Fang-Tsao Hong; Liping H. Pettus; Ryan Wurz; Kristin L. Andrews; Jian Jiang; John D. McCarter; Erin L. Mullady; Tisha San Miguel; Raju Subramanian; Ling Wang; Douglas A. Whittington; Tian Wu; Leeanne Zalameda; Nancy Zhang; Andrew Tasker; Paul E. Hughes; Mark H. Norman

Replacement of the piperazine sulfonamide portion of the PI3Kα inhibitor AMG 511 (1) with a range of aliphatic alcohols led to the identification of a truncated gem-dimethylbenzylic alcohol analog, 2-(5-(4-amino-6-methyl-1,3,5-triazin-2-yl)-6-((5-fluoro-6-methoxypyridin-3-yl)amino)pyridin-3-yl)propan-2-ol (7). This compound possessed good in vitro efficacy and pharmacokinetic parameters and demonstrated an EC50 of 239 ng/mL in a mouse liver pharmacodynamic model measuring the inhibition of hepatocyte growth factor (HGF)-induced Akt Ser473 phosphorylation in CD1 nude mice 6 h post-oral dosing.

Researchain Logo
Decentralizing Knowledge