Network


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

Hotspot


Dive into the research topics where Tao Osgood is active.

Publication


Featured researches published by Tao Osgood.


Journal of Medicinal Chemistry | 2012

Structure-based design of novel inhibitors of the MDM2-p53 interaction.

Yosup Rew; Daqing Sun; Felix Gonzalez-Lopez de Turiso; Michael D. Bartberger; Hilary P. Beck; Jude Canon; Ada Chen; David Chow; Jeffrey Deignan; Brian M. Fox; Darin Gustin; Xin Huang; Min Jiang; Xianyun Jiao; Lixia Jin; Frank Kayser; David J. Kopecky; Yihong Li; Mei-Chu Lo; Alexander M. Long; Klaus Michelsen; Jonathan D. Oliner; Tao Osgood; Mark L. Ragains; Anne Y. Saiki; Steve Schneider; Maria M. Toteva; Peter Yakowec; Xuelei Yan; Qiuping Ye

Structure-based rational design led to the discovery of novel inhibitors of the MDM2-p53 protein-protein interaction. The affinity of these compounds for MDM2 was improved through conformational control of both the piperidinone ring and the appended N-alkyl substituent. Optimization afforded 29 (AM-8553), a potent and selective MDM2 inhibitor with excellent pharmacokinetic properties and in vivo efficacy.


Journal of Medicinal Chemistry | 2014

Discovery of AMG 232, a Potent, Selective, and Orally Bioavailable MDM2–p53 Inhibitor in Clinical Development

Daqing Sun; Zhihong Li; Yosup Rew; Michael W. Gribble; Michael D. Bartberger; Hilary P. Beck; Jude Canon; Ada Chen; Xiaoqi Chen; David Chow; Jeffrey Deignan; Jason Duquette; John Eksterowicz; Benjamin Fisher; Brian M. Fox; Jiasheng Fu; Ana Z. Gonzalez; Felix Gonzalez-Lopez de Turiso; Jonathan B. Houze; Xin Huang; Min Jiang; Lixia Jin; Frank Kayser; Jiwen Liu; Mei-Chu Lo; Alexander M. Long; Brian Lucas; Lawrence R. McGee; Joel McIntosh; Jeff Mihalic

We recently reported the discovery of AM-8553 (1), a potent and selective piperidinone inhibitor of the MDM2-p53 interaction. Continued research investigation of the N-alkyl substituent of this series, focused in particular on a previously underutilized interaction in a shallow cleft on the MDM2 surface, led to the discovery of a one-carbon tethered sulfone which gave rise to substantial improvements in biochemical and cellular potency. Further investigation produced AMG 232 (2), which is currently being evaluated in human clinical trials for the treatment of cancer. Compound 2 is an extremely potent MDM2 inhibitor (SPR KD = 0.045 nM, SJSA-1 EdU IC50 = 9.1 nM), with remarkable pharmacokinetic properties and in vivo antitumor activity in the SJSA-1 osteosarcoma xenograft model (ED50 = 9.1 mg/kg).


Journal of Medicinal Chemistry | 2013

Rational Design and Binding Mode Duality of MDM2–p53 Inhibitors

Felix Gonzalez-Lopez de Turiso; Daqing Sun; Yosup Rew; Michael D. Bartberger; Hilary P. Beck; Jude Canon; Ada Chen; David Chow; Tiffany L. Correll; Xin Huang; Lisa Julian; Frank Kayser; Mei-Chu Lo; Alexander M. Long; Dustin L. McMinn; Jonathan D. Oliner; Tao Osgood; Jay P. Powers; Anne Y. Saiki; Steve Schneider; Paul Shaffer; Shou-Hua Xiao; Peter Yakowec; Xuelei Yan; Qiuping Ye; Dongyin Yu; Xiaoning Zhao; Jing Zhou; Julio C. Medina; Steven H. Olson

Structural analysis of both the MDM2-p53 protein-protein interaction and several small molecules bound to MDM2 led to the design and synthesis of tetrasubstituted morpholinone 10, an MDM2 inhibitor with a biochemical IC50 of 1.0 μM. The cocrystal structure of 10 with MDM2 inspired two independent optimization strategies and resulted in the discovery of morpholinones 16 and 27 possessing distinct binding modes. Both analogues were potent MDM2 inhibitors in biochemical and cellular assays, and morpholinone 27 (IC50 = 0.10 μM) also displayed suitable PK profile for in vivo animal experiments. A pharmacodynamic (PD) experiment in mice implanted with human SJSA-1 tumors showed p21(WAF1) mRNA induction (2.7-fold over vehicle) upon oral dosing of 27 at 300 mg/kg.


Molecular Cancer Therapeutics | 2009

Therapeutic potential of hepatocyte growth factor/scatter factor neutralizing antibodies: Inhibition of tumor growth in both autocrine and paracrine hepatocyte growth factor/scatter factor:c-Met-driven models of leiomyosarcoma

Chong-Feng Gao; Qian Xie; Yu-Wen Zhang; Yanli Su; Ping Zhao; Brian Cao; Kyle A. Furge; Jan Sun; Karen Rex; Tao Osgood; Angela Coxon; Teresa L. Burgess; George F. Vande Woude

Hepatocyte growth factor/scatter factor (HGF/SF) and its receptor, c-Met, have been implicated in the growth and progression of a variety of solid human tumors. Thus, inhibiting HGF/SF:c-Met signaling may provide a novel therapeutic approach for treating human tumors. We have generated and characterized fully human monoclonal antibodies that bind to and neutralize human HGF/SF. In this study, we tested the effects of the investigational, human anti-human HGF/SF monoclonal antibody, AMG 102, and a mixture of mouse anti-human HGF/SF monoclonal antibodies (Amix) on HGF/SF-mediated cell migration, proliferation, and invasion in vitro. Both agents had high HGF/SF-neutralizing activity in these cell-based assays. The HGF/SF:c-Met pathway has been implicated in the growth of sarcomas; thus, we also investigated the effect of AMG 102 on the growth of human leiomyosarcoma (SK-LMS-1) in HGF/SF transgenic C3H severe combined immunodeficient mice engineered to express high levels of human HGF/SF, as well as tumor growth of an autocrine variant of the SK-LMS-1 cell line (SK-LMS-1TO) in nude mice. The results indicate that interrupting autocrine and/or paracrine HGF/SF:c-Met signaling with AMG 102 has profound antitumor effects. These findings suggest that blocking HGF/SF:c-Met signaling may provide a potent intervention strategy to treat patients with HGF/SF:c-Met–dependent tumors. [Mol Cancer Ther 2009;8(10):2803–10]


Journal of Medicinal Chemistry | 2014

Selective and potent morpholinone inhibitors of the MDM2-p53 protein-protein interaction.

Ana Z. Gonzalez; John Eksterowicz; Michael D. Bartberger; Hilary P. Beck; Jude Canon; Ada Chen; David Chow; Jason Duquette; Brian M. Fox; Jiasheng Fu; Xin Huang; Jonathan B. Houze; Lixia Jin; Yihong Li; Zhihong Li; Yun Ling; Mei-Chu Lo; Alexander M. Long; Lawrence R. McGee; Joel McIntosh; Dustin L. McMinn; Jonathan D. Oliner; Tao Osgood; Yosup Rew; Anne Y. Saiki; Paul Shaffer; Sarah Wortman; Peter Yakowec; Xuelei Yan; Qiuping Ye

We previously reported the discovery of AMG 232, a highly potent and selective piperidinone inhibitor of the MDM2-p53 interaction. Our continued search for potent and diverse analogues led to the discovery of novel morpholinone MDM2 inhibitors. This change to a morpholinone core has a significant impact on both potency and metabolic stability compared to the piperidinone series. Within this morpholinone series, AM-8735 emerged as an inhibitor with remarkable biochemical potency (HTRF IC50 = 0.4 nM) and cellular potency (SJSA-1 EdU IC50 = 25 nM), as well as pharmacokinetic properties. Compound 4 also shows excellent antitumor activity in the SJSA-1 osteosarcoma xenograft model with an ED50 of 41 mg/kg. Lead optimization toward the discovery of this inhibitor as well as key differences between the morpholinone and the piperidinone series will be described herein.


Journal of Medicinal Chemistry | 2014

Novel Inhibitors of the MDM2-p53 Interaction Featuring Hydrogen Bond Acceptors as Carboxylic Acid Isosteres.

Ana Z. Gonzalez; Zhihong Li; Hilary P. Beck; Jude Canon; Ada Chen; David Chow; Jason Duquette; John Eksterowicz; Brian M. Fox; Jiasheng Fu; Xin Huang; Jonathan B. Houze; Lixia Jin; Yihong Li; Yun Ling; Mei-Chu Lo; Alexander M. Long; Lawrence R. McGee; Joel McIntosh; Jonathan D. Oliner; Tao Osgood; Yosup Rew; Anne Y. Saiki; Paul Shaffer; Sarah Wortman; Peter Yakowec; Xuelei Yan; Qiuping Ye; Dongyin Yu; Xiaoning Zhao

We previously reported the discovery of potent and selective morpholinone and piperidinone inhibitors of the MDM2-p53 interaction. These inhibitors have in common a carboxylic acid moiety that engages in an electrostatic interaction with MDM2-His96. Our continued search for potent and diverse inhibitors led to the discovery of novel replacements for these acids uncovering new interactions with the MDM2 protein. In particular, using pyridine or thiazole as isosteres of the carboxylic acid moiety resulted in very potent analogues. From these, AM-6761 (4) emerged as a potent inhibitor with remarkable biochemical (HTRF IC50 = 0.1 nM) and cellular potency (SJSA-1 EdU IC50 = 16 nM), as well as favorable pharmacokinetic properties. Compound 4 also shows excellent antitumor activity in the SJSA-1 osteosarcoma xenograft model with an ED50 of 11 mg/kg. Optimization efforts toward the discovery of these inhibitors as well as the new interactions observed with the MDM2 protein are described herein.


Bone | 2010

Inhibition of RANKL increases the anti-tumor effect of the EGFR inhibitor panitumumab in a murine model of bone metastasis

Jude Canon; Rebecca Bryant; Martine Roudier; Tao Osgood; Jon Jones; Robert E. Miller; Angela Coxon; Robert Radinsky; William C. Dougall

Bone metastases cause severe skeletal complications and are associated with osteoclast-mediated bone destruction. RANKL is essential for osteoclast formation, function, and survival, and is the primary effector of tumor-induced osteoclastogenesis and osteolysis. RANKL inhibition by its soluble decoy receptor osteoprotegerin (OPG) prevents tumor-induced osteolysis and decreases skeletal tumor burden. Because osteoclast-mediated bone resorption releases growth factors from the bone matrix, the host bone microenvironment induces a vicious cycle of bone destruction and tumor proliferation and survival. A prediction of this vicious cycle hypothesis is that targeting the host bone microenvironment by osteoclast inhibition would reduce tumor growth and survival and may enhance the anti-tumor effects of targeted therapies. The epidermal growth factor receptor (EGFR) pathway regulates critical processes such as cell growth and survival, and anti-EGFR therapies can cause tumor cell arrest and apoptosis. We evaluated whether reduction of osteolysis by RANKL inhibition could enhance the anti-tumor effects of an anti-EGFR antibody (panitumumab) in a novel murine model of human A431 epidermoid carcinoma bone metastasis. Skeletal tumor progression was assessed longitudinally by bioluminescence imaging. RANKL inhibition by OPG-Fc treatment resulted in a reduction in tumor progression in bony sites. OPG-Fc treatment also caused a dose-dependent reduction in tumor-induced osteolysis, supporting the essential role of RANKL in this process. In combination, RANKL inhibition increased the anti-tumor efficacy of an anti-EGFR antibody, and completely blocked tumor-induced bone breakdown, demonstrating that addition of the indirect anti-tumor effect of RANKL inhibition increases the anti-tumor efficacy of panitumumab, a targeted anti-EGFR antibody.


Molecular Cancer Therapeutics | 2015

The MDM2 Inhibitor AMG 232 Demonstrates Robust Antitumor Efficacy and Potentiates the Activity of p53-Inducing Cytotoxic Agents

Jude Canon; Tao Osgood; Steven H. Olson; Anne Y. Saiki; Rebecca Robertson; Dongyin Yu; John Eksterowicz; Qiuping Ye; Lixia Jin; Ada Chen; Jing Zhou; David Cordover; Stephen Kaufman; Richard Kendall; Jonathan D. Oliner; Angela Coxon; Robert Radinsky

p53 is a critical tumor suppressor and is the most frequently inactivated gene in human cancer. Inhibition of the interaction of p53 with its negative regulator MDM2 represents a promising clinical strategy to treat p53 wild-type tumors. AMG 232 is a potential best-in-class inhibitor of the MDM2–p53 interaction and is currently in clinical trials. We characterized the activity of AMG 232 and its effect on p53 signaling in several preclinical tumor models. AMG 232 binds the MDM2 protein with picomolar affinity and robustly induces p53 activity, leading to cell-cycle arrest and inhibition of tumor cell proliferation. AMG 232 treatment inhibited the in vivo growth of several tumor xenografts and led to complete and durable regression of MDM2-amplified SJSA-1 tumors via growth arrest and induction of apoptosis. Therapeutic combination studies of AMG 232 with chemotherapies that induce DNA damage and p53 activity resulted in significantly superior antitumor efficacy and regression, and markedly increased activation of p53 signaling in tumors. These preclinical data support the further evaluation of AMG 232 in clinical trials as both a monotherapy and in combination with standard-of-care cytotoxics. Mol Cancer Ther; 14(3); 649–58. ©2015 AACR.


Journal of Endocrinological Investigation | 2011

Anti-tumor activity of motesanib in a medullary thyroid cancer model

Angela Coxon; James Bready; Stephen Kaufman; Juan Estrada; Tao Osgood; Jude Canon; Ling Wang; Robert Radinsky; Rick Kendall; Paul E. Hughes; Anthony Polverino

Background: Medullary thyroid cancer (MTC) is frequently associated with mutations in the tyrosine kinase Ret and with increased expression of vascular endothelial growth factor (VEGF) and VEGF receptor 2 (VEGFR2). Motesanib is an investigational, orally administered small molecule antagonist of VEGFR1, 2, and 3; platelet-derived growth factor receptor (PDGFR); Kit; and possibly Ret. Aim: The aim of this study was to investigate the effects of motesanib on wild-type and mutant Ret activity in vitro and on tumor xenograft growth in a mouse model of MTC. Methods/Results: In cellular phosphorylation assays, motesanib inhibited the activity of wild-type Ret (IC50=66 nM), while it had limited activity against mutant Ret C634W (IC50=1100 nM) or Ret M918T (IC50>2500 nM). In vivo, motesanib significantly inhibited the growth of TT tumor cell xenografts (expressing Ret C634W) and significantly reduced tumor blood vessel area and tumor cell proliferation, compared with control. Treatment with motesanib resulted in substantial inhibition of Ret tyrosine phosphorylation in TT xenografts and, at comparable doses, in equivalent inhibition of VEGFR2 phosphorylation in both TT xenografts and in mouse lung tissue. Conclusions: The results of this study demonstrate that motesanib inhibited thyroid tumor xenograft growth predominantly through inhibition of angiogenesis and possibly via a direct inhibition of VEGFR2 and Ret expressed on tumor cells. These data suggest that targeting angiogenesis pathways and specifically the VEGF pathway may represent a novel therapeutic approach in the treatment of MTC.


Journal of Medicinal Chemistry | 2014

Discovery of AM-7209, a Potent and Selective 4-Amidobenzoic Acid Inhibitor of the MDM2-p53 Interaction.

Yosup Rew; Daqing Sun; Xuelei Yan; Hilary P. Beck; Jude Canon; Ada Chen; Jason Duquette; John Eksterowicz; Brian M. Fox; Jiasheng Fu; Ana Z. Gonzalez; Jonathan B. Houze; Xin Huang; Min Jiang; Lixia Jin; Yihong Li; Zhihong Li; Yun Ling; Mei-Chu Lo; Alexander M. Long; Lawrence R. McGee; Joel McIntosh; Jonathan D. Oliner; Tao Osgood; Anne Y. Saiki; Paul Shaffer; Yu Chung Wang; Sarah Wortman; Peter Yakowec; Qiuping Ye

Structure-based rational design and extensive structure-activity relationship studies led to the discovery of AMG 232 (1), a potent piperidinone inhibitor of the MDM2-p53 association, which is currently being evaluated in human clinical trials for the treatment of cancer. Further modifications of 1, including replacing the carboxylic acid with a 4-amidobenzoic acid, afforded AM-7209 (25), featuring improved potency (KD from ITC competition was 38 pM, SJSA-1 EdU IC50 = 1.6 nM), remarkable pharmacokinetic properties, and in vivo antitumor activity in both the SJSA-1 osteosarcoma xenograft model (ED50 = 2.6 mg/kg QD) and the HCT-116 colorectal carcinoma xenograft model (ED50 = 10 mg/kg QD). In addition, 25 possesses distinct mechanisms of elimination compared to 1.

Collaboration


Dive into the Tao Osgood's collaboration.

Researchain Logo
Decentralizing Knowledge