Peiwen Yu
Exelixis
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Peiwen Yu.
Molecular Cancer Therapeutics | 2011
F. Michael Yakes; Jason Chen; Jenny Tan; Kyoko Yamaguchi; Yongchang Shi; Peiwen Yu; Fawn Qian; Felix Chu; Frauke Bentzien; Belinda Cancilla; Jessica Orf; Andrew You; A. Douglas Laird; Stefan Engst; Lillian Lee; Justin Lesch; Yu-Chien Chou; Alison Joly
The signaling pathway of the receptor tyrosine kinase MET and its ligand hepatocyte growth factor (HGF) is important for cell growth, survival, and motility and is functionally linked to the signaling pathway of VEGF, which is widely recognized as a key effector in angiogenesis and cancer progression. Dysregulation of the MET/VEGF axis is found in a number of human malignancies and has been associated with tumorigenesis. Cabozantinib (XL184) is a small-molecule kinase inhibitor with potent activity toward MET and VEGF receptor 2 (VEGFR2), as well as a number of other receptor tyrosine kinases that have also been implicated in tumor pathobiology, including RET, KIT, AXL, and FLT3. Treatment with cabozantinib inhibited MET and VEGFR2 phosphorylation in vitro and in tumor models in vivo and led to significant reductions in cell invasion in vitro. In mouse models, cabozantinib dramatically altered tumor pathology, resulting in decreased tumor and endothelial cell proliferation coupled with increased apoptosis and dose-dependent inhibition of tumor growth in breast, lung, and glioma tumor models. Importantly, treatment with cabozantinib did not increase lung tumor burden in an experimental model of metastasis, which has been observed with inhibitors of VEGF signaling that do not target MET. Collectively, these data suggest that cabozantinib is a promising agent for inhibiting tumor angiogenesis and metastasis in cancers with dysregulated MET and VEGFR signaling. Mol Cancer Ther; 10(12); 2298–308. ©2011 AACR.
Clinical Cancer Research | 2007
Steven Gendreau; Richard Ventura; Paul Keast; A. Douglas Laird; F. Michael Yakes; Wentao Zhang; Frauke Bentzien; Belinda Cancilla; Jeffery Lutman; Felix Chu; Lisa Jackman; Yongchang Shi; Peiwen Yu; Jing Wang; Dana T. Aftab; Christopher Jaeger; Stephanie Meyer; Anushka de Costa; Kelly Engell; Jason Chen; Jean-Francois Martini; Alison Joly
Purpose: Agents inhibiting the epidermal growth factor receptor (EGFR) have shown clinical benefit in a subset of non–small cell lung cancer patients expressing amplified or mutationally activated EGFR. However, responsive patients can relapse as a result of selection for EGFR gene mutations that confer resistance to ATP competitive EGFR inhibitors, such as erlotinib and gefitinib. We describe here the activity of EXEL-7647 (XL647), a novel spectrum-selective kinase inhibitor with potent activity against the EGF and vascular endothelial growth factor receptor tyrosine kinase families, against both wild-type (WT) and mutant EGFR in vitro and in vivo. Experimental Design: The activity of EGFR inhibitors against WT and mutant EGFRs and their effect on downstream signal transduction was examined in cellular assays and in vivo using A431 and MDA-MB-231 (WT EGFR) and H1975 (L858R and T790M mutant EGFR) xenograft tumors. Results: EXEL-7647 shows potent and long-lived inhibition of the WT EGFR in vivo. In addition, EXEL-7647 inhibits cellular proliferation and EGFR pathway activation in the erlotinib-resistant H1975 cell line that harbors a double mutation (L858R and T790M) in the EGFR gene. In vivo efficacy studies show that EXEL-7647 substantially inhibited the growth of H1975 xenograft tumors and reduced both tumor EGFR signaling and tumor vessel density. Additionally, EXEL-7647, in contrast to erlotinib, substantially inhibited the growth and vascularization of MDA-MB-231 xenografts, a model which is more reliant on signaling through vascular endothelial growth factor receptors. Conclusions: These studies provide a preclinical basis for clinical trials of XL647 in solid tumors and in patients bearing tumors that are resistant to existing EGFR-targeted therapies.
Clinical Cancer Research | 2012
Harvey Wong; Laurent Vernillet; Amy C. Peterson; Joeseph A Ware; Lillian Lee; Jean-Francois Martini; Peiwen Yu; Congfen Li; Geoffrey Del Rosario; Edna F. Choo; Klaus P. Hoeflich; Yongchang Shi; Blake T. Aftab; Ron G Aoyama; Sanh Tan Lam; Marcia Belvin; John Prescott
Purpose: GDC-0973 is a potent and selective mitogen-activated protein (MAP)/extracellular signal–regulated kinase (ERK) kinase (MEK) inhibitor. Pharmacokinetic–pharmacodynamic (PK–PD) modeling was used to relate GDC-0973 plasma and tumor concentrations, tumor pharmacodynamics and antitumor efficacy to establish pharmacokinetic endpoints and predict active doses in the clinic. Experimental Design: A PK–PD model was used to characterize GDC-0973 tumor disposition and in vivo potency in WM-266-4 xenograft mice. Simulations were conducted using the PK–PD model along with human pharmacokinetics to identify a target plasma concentration and predict active doses. In vivo potency and antitumor efficacy were characterized in A375 melanoma xenograft mice, and a population-based integrated PK–PD-efficacy model was used to relate tumor pharmacodynamics (%pERK decrease) to antitumor activity. Results: GDC-0973 showed a sustained tumor pharmacodynamic response due to longer residence in tumor than in plasma. Following single doses of GDC-0973, estimated in vivo IC50 values of %pERK decrease based on tumor concentrations in xenograft mice were 0.78 (WM-266-4) and 0.52 μmol/L (A375). Following multiple doses of GDC-0973, the estimated in vivo IC50 value in WM-266-4 increased (3.89 μmol/L). Human simulations predicted a minimum target plasma concentration of 83 nmol/L and an active dose range of 28 to 112 mg. The steep relationship between tumor pharmacodynamics (%pERK decrease) and antitumor efficacy suggests a pathway modulation threshold beyond which antitumor efficacy switches on. Conclusions: Clinical observations of %pERK decrease and antitumor activity were consistent with model predictions. This article illustrates how PK–PD modeling can improve the translation of preclinical data to humans by providing a means to integrate preclinical and early clinical data. Clin Cancer Res; 18(11); 3090–9. ©2012 AACR.
ACS Medicinal Chemistry Letters | 2012
Kenneth D. Rice; Naing Aay; Neel Kumar Anand; Charles M. Blazey; Owen Joseph Bowles; Joerg Bussenius; Simona Costanzo; Jeffry Kimo Curtis; Steven Charles Defina; Larisa Dubenko; Stefan Engst; Anagha Abhijit Joshi; Abigail R. Kennedy; Angie I. Kim; Elena S. Koltun; Julie Lougheed; Jean-Claire Limun Manalo; Jean‐Francois Martini; John M. Nuss; Csaba J. Peto; Tsze H. Tsang; Peiwen Yu; Stuart Johnston
The ERK/MAP kinase cascade is a key mechanism subject to dysregulation in cancer and is constitutively activated or highly upregulated in many tumor types. Mutations associated with upstream pathway components RAS and Raf occur frequently and contribute to the oncogenic phenotype through activation of MEK and then ERK. Inhibitors of MEK have been shown to effectively block upregulated ERK/MAPK signaling in a range of cancer cell lines and have further demonstrated early evidence of efficacy in the clinic for the treatment of cancer. Guided by structural insight, a strategy aimed at the identification of an optimal diphenylamine-based MEK inhibitor with an improved metabolism and safety profile versus PD-0325901 led to the discovery of development candidate 1-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-[(2S)-piperidin-2-yl]azetidin-3-ol (XL518, GDC-0973) (1). XL518 exhibits robust in vitro and in vivo potency and efficacy in preclinical models with sustained duration of action and is currently in early stage clinical trials.
Bioorganic & Medicinal Chemistry Letters | 2012
Elena S. Koltun; Amy Lew Tsuhako; David S. Brown; Naing Aay; Arlyn Arcalas; Vicky Chan; Hongwang Du; Stefan Engst; Kim Ferguson; Maurizio Franzini; Adam Antoni Galan; Charles R. Holst; Ping Huang; Brian Kane; Moon Hwan Kim; Jia Li; David Markby; Manisha Mohan; Kevin Noson; Arthur Plonowski; Steven Richards; Scott Robertson; Kenneth James Shaw; Gordon Mark Stott; Thomas J. Stout; Jenny Young; Peiwen Yu; Cristiana A. Zaharia; Wentao Zhang; Peiwen Zhou
CDC7 is a serine/threonine kinase that has been shown to be required for the initiation and maintenance of DNA replication. Up-regulation of CDC7 is detected in multiple tumor cell lines, with inhibition of CDC7 resulting in cell cycle arrest. In this paper, we disclose the discovery of a potent and selective CDC7 inhibitor, XL413 (14), which was advanced into Phase 1 clinical trials. Starting from advanced lead 3, described in a preceding communication, we optimized the CDC7 potency and selectivity to demonstrate in vitro CDC7 dependent cell cycle arrest and in vivo tumor growth inhibition in a Colo-205 xenograft model.
Molecular Cancer Therapeutics | 2014
Peiwen Yu; Laird Ad; Du X; Wu J; Won Ka; Yamaguchi K; Hsu Pp; Fawn Qian; Jaeger Ct; Wentao Zhang; Buhr Ca; Shen P; Abulafia W; Jason Chen; Young J; Plonowski A; Yakes Fm; Felix Chu; Lee M; Bentzien F; Lam St; Dale S; David J. Matthews; Peter Lamb; Paul Foster
Activation of the PI3K (phosphoinositide 3-kinase) pathway is a frequent occurrence in human tumors and is thought to promote growth, survival, and resistance to diverse therapies. Here, we report pharmacologic characterization of the pyridopyrimidinone derivative XL765 (SAR245409), a potent and highly selective pan inhibitor of class I PI3Ks (α, β, γ, and δ) with activity against mTOR. Broad kinase selectivity profiling of >130 protein kinases revealed that XL765 is highly selective for class I PI3Ks and mTOR over other kinases. In cellular assays, XL765 inhibits the formation of PIP3 in the membrane, and inhibits phosphorylation of AKT, p70S6K, and S6 phosphorylation in multiple tumor cell lines with different genetic alterations affecting the PI3K pathway. In a panel of tumor cell lines, XL765 inhibits proliferation with a wide range of potencies, with evidence of an impact of genotype on sensitivity. In mouse xenograft models, oral administration of XL765 results in dose-dependent inhibition of phosphorylation of AKT, p70S6K, and S6 with a duration of action of approximately 24 hours. Repeat dose administration of XL765 results in significant tumor growth inhibition in multiple human xenograft models in nude mice that is associated with antiproliferative, antiangiogenic, and proapoptotic effects. Mol Cancer Ther; 13(5); 1078–91. ©2014 AACR.
Bioorganic & Medicinal Chemistry Letters | 2012
Amy Lew Tsuhako; David S. Brown; Elena S. Koltun; Naing Aay; Arlyn Arcalas; Chan; Hongwang Du; Stefan Engst; Maurizio Franzini; Adam Antoni Galan; Ping Huang; S Johnston; Brian Kane; Moon Hwan Kim; A.D Laird; R Lin; L Mock; I Ngan; Michael Pack; Gordon Mark Stott; Thomas J. Stout; Peiwen Yu; Cristiana A. Zaharia; Wentao Zhang; Peiwen Zhou; John M. Nuss; Patrick Kearney; Wei Xu
A series of substituted benzofuropyrimidinones with pan-PIM activities and excellent selectivity against a panel of diverse kinases is described. Initial exploration identified aryl benzofuropyrimidinones that were potent, but had cell permeability limitation. Using X-ray crystal structures of the bound PIM-1 complexes with 3, 5m, and 6d, we were able to guide the SAR and identify the alkyl benzofuropyrimidinone (6l) with good PIM potencies, permeability, and oral exposure.
Journal of Medicinal Chemistry | 2012
James W. Leahy; Chris A. Buhr; Henry William Beecroft Johnson; Byung Gyu Kim; Tae-Gon Baik; Jonah Cannoy; Timothy Patrick Forsyth; Joon Won Jeong; Matthew Sangyup Lee; Sunghoon Ma; Kevin Noson; Longcheng Wang; Matthew A. Williams; John M. Nuss; Eric Brooks; Paul A. Foster; Leanne Goon; Nathan Heald; Charles R. Holst; Christopher Jaeger; Scott Lam; Julie Lougheed; Lam Nguyen; Arthur Plonowski; Joanne Song; Thomas J. Stout; Xiang Wu; Michael Yakes; Peiwen Yu; Wentao Zhang
The phosphoinositide 3-kinases (PI3Ks) have been linked to an extraordinarily diversified group of cellular functions making these enzymes compelling targets for the treatment of disease. A large body of evidence has linked PI3Kγ to the modulation of autoimmune and inflammatory processes making it an intriguing target for drug discovery. Our high-throughput screening (HTS) campaign revealed two hits that were nominated for further optimization studies. The in vitro activity of the first HTS hit, designated as the sulfonylpiperazine scaffold, was optimized utilizing structure-based design. However, nonoptimal pharmacokinetic properties precluded this series from further studies. An overlay of the X-ray structures of the sulfonylpiperazine scaffold and the second HTS hit within their complexes with PI3Kγ revealed a high degree of overlap. This feature was utilized to design a series of hybrid analogues including advanced leads such as 31 with desirable potency, selectivity, and oral bioavailability.
Molecular Cancer Therapeutics | 2015
Paul Foster; Yamaguchi K; Hsu Pp; Fawn Qian; Du X; Wu J; Won Ka; Peiwen Yu; Jaeger Ct; Wentao Zhang; Marlowe Ck; Keast P; Abulafia W; Jason Chen; Young J; Plonowski A; Yakes Fm; Felix Chu; Engell K; Bentzien F; Lam St; Dale S; Yturralde O; David J. Matthews; Peter Lamb; Laird Ad
Dysregulation of PI3K/PTEN pathway components, resulting in hyperactivated PI3K signaling, is frequently observed in various cancers and correlates with tumor growth and survival. Resistance to a variety of anticancer therapies, including receptor tyrosine kinase (RTK) inhibitors and chemotherapeutic agents, has been attributed to the absence or attenuation of downregulating signals along the PI3K/PTEN pathway. Thus, PI3K inhibitors have therapeutic potential as single agents and in combination with other therapies for a variety of cancer indications. XL147 (SAR245408) is a potent and highly selective inhibitor of class I PI3Ks (α, β, γ, and δ). Moreover, broad kinase selectivity profiling of >130 protein kinases revealed that XL147 is highly selective for class I PI3Ks over other kinases. In cellular assays, XL147 inhibits the formation of PIP3 in the membrane, and inhibits phosphorylation of AKT, p70S6K, and S6 in multiple tumor cell lines with diverse genetic alterations affecting the PI3K pathway. In a panel of tumor cell lines, XL147 inhibits proliferation with a wide range of potencies, with evidence of an impact of genotype on sensitivity. In mouse xenograft models, oral administration of XL147 results in dose-dependent inhibition of phosphorylation of AKT, p70S6K, and S6 with a duration of action of at least 24 hours. Repeat-dose administration of XL147 results in significant tumor growth inhibition in multiple human xenograft models in nude mice. Administration of XL147 in combination with chemotherapeutic agents results in antitumor activity in xenograft models that is enhanced over that observed with the corresponding single agents. Mol Cancer Ther; 14(4); 931–40. ©2015 AACR.
Bioorganic & Medicinal Chemistry Letters | 2012
Moon Hwan Kim; Amy Lew Tsuhako; Erick Wang Co; Dana T. Aftab; Frauke Bentzien; Jason Chen; Wei Cheng; Stefan Engst; Levina Goon; Rhett Ronald Klein; Donna T. Le; Morrison B. Mac; Jason Jevious Parks; Fawn Qian; Monica Rodriquez; Thomas J. Stout; Jeffrey H. Till; Kwang-Ai Won; Xiang Wu; F. Michael Yakes; Peiwen Yu; Wentao Zhang; Yeping Zhao; Peter Lamb; John M. Nuss; Wei Xu
Variously substituted indolin-2-ones were synthesized and evaluated for activity against KDR, Flt-1, FGFR-1 and PDGFR. Extension at the 5-position of the oxindole ring with ethyl piperidine (compound 7i) proved to be the most beneficial for attaining both biochemical and cellular potencies. Further optimization of 7i to balance biochemical and cellular potencies with favorable ADME/ PK properties led to the identification of 8h, a compound with a clean CYP profile, acceptable pharmacokinetic and toxicity profiles, and robust efficacy in multiple xenograft tumor models.