Network


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

Hotspot


Dive into the research topics where Andrew Tasker is active.

Publication


Featured researches published by Andrew Tasker.


Cancer Research | 2006

AMG 706, an Oral, Multikinase Inhibitor that Selectively Targets Vascular Endothelial Growth Factor, Platelet-Derived Growth Factor, and Kit Receptors, Potently Inhibits Angiogenesis and Induces Regression in Tumor Xenografts

Anthony Polverino; Angela Coxon; Charlie Starnes; Zobedia Diaz; Thomas DeMelfi; Ling Wang; James Bready; Juan Estrada; Russell C. Cattley; Stephen Kaufman; Danlin Chen; Yongmei Gan; Gondi Kumar; James Meyer; Sesha Neervannan; Gonzalo Alva; Jane Talvenheimo; Silvia Montestruque; Andrew Tasker; Vinod F. Patel; Robert Radinsky; Richard Kendall

The growth of solid tumors is dependent on the continued stimulation of endothelial cell proliferation and migration resulting in angiogenesis. The angiogenic process is controlled by a variety of factors of which the vascular endothelial growth factor (VEGF) pathway and its receptors play a pivotal role. Small-molecule inhibitors of VEGF receptors (VEGFR) have been shown to inhibit angiogenesis and tumor growth in preclinical models and in clinical trials. A novel nicotinamide, AMG 706, was identified as a potent, orally bioavailable inhibitor of the VEGFR1/Flt1, VEGFR2/kinase domain receptor/Flk-1, VEGFR3/Flt4, platelet-derived growth factor receptor, and Kit receptors in preclinical models. AMG 706 inhibited human endothelial cell proliferation induced by VEGF, but not by basic fibroblast growth factor in vitro, as well as vascular permeability induced by VEGF in mice. Oral administration of AMG 706 potently inhibited VEGF-induced angiogenesis in the rat corneal model and induced regression of established A431 xenografts. AMG 706 was well tolerated and had no significant effects on body weight or on the general health of the animals. Histologic analysis of tumor xenografts from AMG 706-treated animals revealed an increase in endothelial apoptosis and a reduction in blood vessel area that preceded an increase in tumor cell apoptosis. In summary, AMG 706 is an orally bioavailable, well-tolerated multikinase inhibitor that is presently under clinical investigation for the treatment of human malignancies.


Molecular Cell | 2011

TBK1 Directly Engages Akt/PKB Survival Signaling to Support Oncogenic Transformation

Yi Hung Ou; Michael J. Torres; Rosalyn Ram; Etienne Formstecher; Christina L. Roland; Tzuling Cheng; Rolf A. Brekken; Ryan Wurz; Andrew Tasker; Tony Polverino; Seng Lai Tan; Michael A. White

The innate immune-signaling kinase, TBK1, couples pathogen surveillance to induction of host defense mechanisms. Pathological activation of TBK1 in cancer can overcome programmed cell death cues, enabling cells to survive oncogenic stress. The mechanistic basis of TBK1 prosurvival signaling, however, has been enigmatic. Here, we show that TBK1 directly activates AKT by phosphorylation of the canonical activation loop and hydrophobic motif sites independently of PDK1 and mTORC2. Upon mitogen stimulation, triggering of the innate immune response, re-exposure to glucose, or oncogene activation, TBK1 is recruited to the exocyst, where it activates AKT. In cells lacking TBK1, insulin activates AKT normally, but AKT activation by exocyst-dependent mechanisms is impaired. Discovery and characterization of a 6-aminopyrazolopyrimidine derivative, as a selective low-nanomolar TBK1 inhibitor, indicates that this regulatory arm can be pharmacologically perturbed independently of canonical PI3K/PDK1 signaling. Thus, AKT is a direct TBK1 substrate that connects TBK1 to prosurvival signaling.


Journal of Biological Chemistry | 2008

c-Met inhibitors with novel binding mode show activity against several hereditary papillary renal cell carcinoma-related mutations.

Steven Bellon; Paula Kaplan-Lefko; Yajing Yang; Yihong Zhang; Jodi Moriguchi; Karen Rex; Carol W. Johnson; Paul Rose; Alexander M. Long; Anne O'connor; Yan Gu; Angela Coxon; Tae-Seong Kim; Andrew Tasker; Teresa L. Burgess; Isabelle Dussault

c-Met is a receptor tyrosine kinase often deregulated in human cancers, thus making it an attractive drug target. One mechanism by which c-Met deregulation leads to cancer is through gain-of-function mutations. Therefore, small molecules capable of targeting these mutations could offer therapeutic benefits for affected patients. SU11274 was recently described and reported to inhibit the activity of the wild-type and some mutant forms of c-Met, whereas other mutants are resistant to inhibition. We identified a novel series of c-Met small molecule inhibitors that are active against multiple mutants previously identified in hereditary papillary renal cell carcinoma patients. AM7 is active against wild-type c-Met as well as several mutants, inhibits c-Met-mediated signaling in MKN-45 and U-87 MG cells, and inhibits tumor growth in these two models grown as xenografts. The crystal structures of AM7 and SU11274 bound to unphosphorylated c-Met have been determined. The AM7 structure reveals a novel binding mode compared with other published c-Met inhibitors and SU11274. The molecule binds the kinase linker and then extends into a new hydrophobic binding site. This binding site is created by a significant movement of the C-helix and so represents an inactive conformation of the c-Met kinase. Thus, our results demonstrate that it is possible to identify and design inhibitors that will likely be active against mutants found in different cancers.


Journal of Medicinal Chemistry | 2008

Design, synthesis, and biological evaluation of potent c-Met inhibitors.

Noel D'angelo; Steven Bellon; Shon Booker; Yuan Cheng; Angela Coxon; Celia Dominguez; Ingrid M. Fellows; Douglas Hoffman; Randall W. Hungate; Paula Kaplan-Lefko; Matthew R. Lee; Chun Li; Longbin Liu; Elizabeth Rainbeau; Paul J. Reider; Karen Rex; Aaron C. Siegmund; Yaxiong Sun; Andrew Tasker; Ning Xi; Shimin Xu; Yajing Yang; Yihong Zhang; Teresa L. Burgess; Isabelle Dussault; Tae-Seong Kim

c-Met is a receptor tyrosine kinase that plays a key role in several cellular processes but has also been found to be overexpressed and mutated in different human cancers. Consequently, targeting this enzyme has become an area of intense research in drug discovery. Our studies began with the design and synthesis of novel pyrimidone 7, which was found to be a potent c-Met inhibitor. Subsequent SAR studies identified 22 as a more potent analog, whereas an X-ray crystal structure of 7 bound to c-Met revealed an unexpected binding conformation. This latter finding led to the development of a new series that featured compounds that were more potent both in vitro and in vivo than 22 and also exhibited different binding conformations to c-Met. Novel c-Met inhibitors have been designed, developed, and found to be potent in vitro and in vivo.


Cancer Research | 2006

Discovery and Evaluation of Dual CDK1 and CDK2 Inhibitors

Marc Payton; Grace Tin-Yun Chung; Peter Yakowec; Andrew D. Wong; Dave Powers; Ling Xiong; Nancy R. Zhang; Juan Leal; Tammy L. Bush; Vincent Santora; Ben Askew; Andrew Tasker; Robert Radinsky; Richard Kendall; Steve Coats

In eukaryotic cells, cyclin-dependent kinase (CDK) complexes regulate the temporal progression of cells through the cell cycle. Deregulation in the cell cycle is an essential component in the evolution of cancer. Here, we validate CDK1 and CDK2 as potential therapeutic targets using novel selective small-molecule inhibitors of cyclin B1/CDK1 and cyclin E2/CDK2 enzyme complexes (CDKi). Flow cytometry-based methods were developed to assess intracellular retinoblastoma (Rb) phosphorylation to show inhibition of the CDK pathway. Tumor cells treated with CDK inhibitors showed an overall decrease in cell proliferation, accumulation of cells in G1 and G2, and apoptosis in a cell line-specific manner. Although CDK inhibitors activate p53, the inhibitors were equipotent in arresting the cell cycle in isogenic breast and colon tumor cells lacking p53, suggesting the response is independent of p53. In vivo, the CDK inhibitors prevented the growth of colon and prostate tumors, blocked proliferation of tumor cells, and inhibited Rb phosphorylation. The discovery and evaluation of novel potent and selective CDK1 and CDK2 inhibitors will help delineate the role that CDK complexes play in regulating tumorigenesis.


Journal of Medicinal Chemistry | 2008

Naphthamides as novel and potent vascular endothelial growth factor receptor tyrosine kinase inhibitors: design, synthesis, and evaluation.

Jean-Christophe Harmange; Matthew Weiss; Julie Germain; Anthony Polverino; George Borg; James Bready; Danlin Chen; Deborah Choquette; Angela Coxon; Tom DeMelfi; Lucian DiPietro; Nicholas Doerr; Juan Estrada; Julie Flynn; Russell Graceffa; Shawn P. Harriman; Stephen Kaufman; Daniel S. La; Alexander M. Long; Matthew W. Martin; Sesha Neervannan; Vinod F. Patel; Michele Potashman; Kelly Regal; Phillip M. Roveto; Michael Schrag; Charlie Starnes; Andrew Tasker; Yohannes Teffera; Ling Wang

A series of naphthyl-based compounds were synthesized as potential inhibitors of vascular endothelial growth factor (VEGF) receptors. Investigations of structure-activity relationships led to the identification of a series of naphthamides that are potent inhibitors of the VEGF receptor tyrosine kinase family. Numerous analogues demonstrated low nanomolar inhibition of VEGF-dependent human umbilical vein endothelial cell (HUVEC) proliferation, and of these several compounds possessed favorable pharmacokinetic (PK) profiles. In particular, compound 48 demonstrated significant antitumor efficacy against established HT29 human colon adenocarcinoma xenografts implanted in athymic mice. A full account of the preparation, structure-activity relationships, pharmacokinetic properties, and pharmacology of analogues within this series is presented.


Journal of Medicinal Chemistry | 2008

Evaluation of a Series of Naphthamides as Potent, Orally Active Vascular Endothelial Growth Factor Receptor-2 Tyrosine Kinase Inhibitors¶

Matthew Weiss; Jean-Christophe Harmange; Anthony Polverino; David Bauer; Loren Berry; Virginia Berry; George Borg; James Bready; Danlin Chen; Deborah Choquette; Angela Coxon; Tom DeMelfi; Nicholas Doerr; Juan Estrada; Julie Flynn; Russell Graceffa; Shawn P. Harriman; Stephen Kaufman; Daniel S. La; Alexander M. Long; Sesha Neervannan; Vinod F. Patel; Michele Potashman; Kelly Regal; Phillip M. Roveto; Michael Schrag; Charlie Starnes; Andrew Tasker; Yohannes Teffera; Douglas A. Whittington

We have previously shown N-arylnaphthamides can be potent inhibitors of vascular endothelial growth factor receptors (VEGFRs). N-Alkyl and N-unsubstituted naphthamides were prepared and found to yield nanomolar inhibitors of VEGFR-2 (KDR) with an improved selectivity profile against a panel of tyrosine and serine/threonine kinases. The inhibitory activity of this series was retained at the cellular level. Naphthamides 3, 20, and 22 exhibited good pharmacokinetics following oral dosing and showed potent inhibition of VEGF-induced angiogenesis in the rat corneal model. Once-daily oral administration of 22 for 14 days led to 85% inhibition of established HT29 colon cancer and Calu-6 lung cancer xenografts at doses of 10 and 20 mg/kg, respectively.


Journal of Medicinal Chemistry | 2008

Discovery of highly selective and potent p38 inhibitors based on a phthalazine scaffold.

Brad Herberich; Guo-Qiang Cao; Partha P. Chakrabarti; James Richard Falsey; Liping H. Pettus; Robert M. Rzasa; Anthony B. Reed; Andreas Reichelt; Kelvin Sham; Maya C. Thaman; Ryan Wurz; Shimin Xu; Dawei Zhang; Faye Hsieh; Matthew R. Lee; Rashid Syed; Vivian Li; David Grosfeld; Matthew Plant; Bradley Henkle; Lisa Sherman; Scot Middleton; Lu Min Wong; Andrew Tasker

Investigations into the structure-activity relationships (SAR) of a series of phthalazine-based inhibitors of p38 are described. These efforts originated from quinazoline 1 and through rational design led to the development of a series of orally bioavailable, potent, and selective inhibitors. Kinase selectivity was achieved by exploiting a collection of interactions with p38alpha including close contact to Ala157, occupation of the hydrophobic gatekeeper pocket, and a residue flip with Gly110. Substitutions on the phthalazine influenced the pharmacokinetic properties, of which compound 16 displayed the most desirable profile. Oral dosing (0.03 mg/kg) of 16 in rats 1 h prior to LPS challenge gave a >50% decrease in TNFalpha production.


Journal of Medicinal Chemistry | 2008

Discovery of Aryl Aminoquinazoline Pyridones as Potent, Selective, and Orally Efficacious Inhibitors of Receptor Tyrosine Kinase c-Kit

Essa Hu; Andrew Tasker; Ryan White; Roxanne Kunz; Jason Brooks Human; Ning Chen; Roland W. Bürli; Randall W. Hungate; Perry M. Novak; Andrea Itano; Xuxia Zhang; Violeta Yu; Yen Nguyen; Yanyan Tudor; Matthew Plant; Shaun Flynn; Yang Xu; Kristin L. Meagher; Douglas A. Whittington; Gordon Ng

Inhibition of c-Kit has the potential to treat mast cell associated fibrotic diseases. We report the discovery of several aminoquinazoline pyridones that are potent inhibitors of c-Kit with greater than 200-fold selectivity against KDR, p38, Lck, and Src. In vivo efficacy of pyridone 16 by dose-dependent inhibition of histamine release was demonstrated in a rodent pharmacodynamic model of mast cell activation.


Clinical Cancer Research | 2009

Broad Antitumor Activity in Breast Cancer Xenografts by Motesanib, a Highly Selective, Oral Inhibitor of Vascular Endothelial Growth Factor, Platelet-Derived Growth Factor, and Kit Receptors

Angela Coxon; Tammy L. Bush; Douglas Saffran; Stephen Kaufman; Brian Belmontes; Karen Rex; Paul E. Hughes; Sean Caenepeel; James B. Rottman; Andrew Tasker; Vinod F. Patel; Richard Kendall; Robert Radinsky; Anthony Polverino

Purpose: Angiogenesis plays a critical role in breast cancer development and progression. Vascular endothelial growth factor (VEGF) is a potent angiogenic factor that regulates endothelial cell proliferation and survival. We investigated the effects of motesanib, a novel, oral inhibitor of VEGF receptors 1, 2, and 3; platelet-derived growth factor receptor; and Kit receptor, on the growth of xenografts representing various human breast cancer subtypes. Experimental Design: Athymic nude mice were implanted with MCF-7 (luminal) or MDA-MB-231 (mesenchymal) tumor fragments or Cal-51 (mixed/progenitor) tumor cells. Once tumors were established, animals were randomized to receive increasing doses of motesanib alone or motesanib plus cytotoxic chemotherapy (docetaxel, doxorubicin, or tamoxifen). Results: Across all three xenograft models, motesanib treatment resulted in significant dose-dependent reductions in tumor growth, compared with vehicle-treated controls, and in marked reductions in viable tumor fraction and blood vessel density. No significant effect on body weight was observed with compound treatment compared with control-treated animals. Motesanib did not affect the proliferation of tumor cells in vitro. There was a significantly greater reduction in xenograft tumor growth when motesanib was combined with docetaxel (MDA-MB-231 tumors) or with the estrogen receptor modulator tamoxifen (MCF-7 tumors), compared with either treatment alone, but not when combined with doxorubicin (Cal-51 tumors). Conclusions: Treatment with motesanib alone or in combination with chemotherapy inhibits tumor growth in vivo in various models of human breast cancer. These data suggest that motesanib may have broad utility in the treatment of human breast cancer.

Collaboration


Dive into the Andrew Tasker's collaboration.

Researchain Logo
Decentralizing Knowledge