Network


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

Hotspot


Dive into the research topics where Thomas Hunsaker is active.

Publication


Featured researches published by Thomas Hunsaker.


Journal of Medicinal Chemistry | 2009

A class of 2,4-bisanilinopyrimidine Aurora A inhibitors with unusually high selectivity against Aurora B.

Ignacio Aliagas-Martin; Dan Burdick; Laura Corson; Jennafer Dotson; Jason Drummond; Carter Fields; Oscar W. Huang; Thomas Hunsaker; Tracy Kleinheinz; Elaine Krueger; Jun Liang; John Moffat; Gail Lewis Phillips; Rebecca Pulk; Thomas E. Rawson; Mark Ultsch; Leslie Walker; Christian Wiesmann; Birong Zhang; Bing-Yan Zhu; Andrea G. Cochran

The two major Aurora kinases carry out critical functions at distinct mitotic stages. Selective inhibitors of these kinases, as well as pan-Aurora inhibitors, show antitumor efficacy and are now under clinical investigation. However, the ATP-binding sites of Aurora A and Aurora B are virtually identical, and the structural basis for selective inhibition has therefore not been clear. We report here a class of bisanilinopyrimidine Aurora A inhibitors with excellent selectivity for Aurora A over Aurora B, both in enzymatic assays and in cellular phenotypic assays. Crystal structures of two of the inhibitors in complex with Aurora A implicate a single amino acid difference in Aurora B as responsible for poor inhibitory activity against this enzyme. Mutation of this residue in Aurora B (E161T) or Aurora A (T217E) is sufficient to swap the inhibition profile, suggesting that this difference might be exploited more generally to achieve high selectivity for Aurora A.


Cancer Research | 2012

An Integrated Genomic Screen Identifies LDHB as an Essential Gene for Triple-Negative Breast Cancer

Mark L. McCleland; Adam S. Adler; Yonglei Shang; Thomas Hunsaker; Tom Truong; David Peterson; Eric Torres; Li Li; Benjamin Haley; Jean-Philippe Stephan; Marcia Belvin; Georgia Hatzivassiliou; Elizabeth Blackwood; Laura Corson; Marie Evangelista; Jiping Zha; Ron Firestein

Breast cancer has been redefined into three clinically relevant subclasses: (i) estrogen/progesterone receptor positive (ER+/PR+), (ii) HER2/ERRB2 positive, and (iii) those lacking expression of all three markers (triple negative or basal-like). While targeted therapies for ER+/PR+ and HER2+ tumors have revolutionized patient treatment and increased lifespan, an urgent need exists for identifying novel targets for triple-negative breast cancers. Here, we used integrative genomic analysis to identify candidate oncogenes in triple-negative breast tumors and assess their function through loss of function screening. Using this approach, we identify lactate dehydrogenase B (LDHB), a component of glycolytic metabolism, as an essential gene in triple-negative breast cancer. Loss of LDHB abrogated cell proliferation in vitro and arrested tumor growth in fully formed tumors in vivo. We find that LDHB and other related glycolysis genes are specifically upregulated in basal-like/triple-negative breast cancers as compared with other subtypes, suggesting that these tumors are distinctly glycolytic. Consistent with this, triple-negative breast cancer cell lines were more dependent on glycolysis for growth than luminal cell lines. Finally, we find that patients with breast cancer and high LDHB expression in their tumors had a poor clinical outcome. While previous studies have focused on the ubiquitous role of LDHA in tumor metabolism and growth, our data reveal that LDHB is upregulated and required only in certain cancer genotypes. These findings suggest that targeting LDHB or other components of lactate metabolism would be of clinical benefit in triple-negative breast cancer.


Bioorganic & Medicinal Chemistry Letters | 2013

Identification of substituted 2-thio-6-oxo-1,6-dihydropyrimidines as inhibitors of human lactate dehydrogenase.

Peter S. Dragovich; Benjamin P. Fauber; Laura Corson; Charles Z. Ding; Charles Eigenbrot; HongXiu Ge; Anthony M. Giannetti; Thomas Hunsaker; Sharada Labadie; Yichin Liu; Shiva Malek; Borlan Pan; David Peterson; Keith Pitts; Hans E. Purkey; Steve Sideris; Mark Ultsch; Erica VanderPorten; Binqing Wei; Qing Xu; Ivana Yen; Qin Yue; Huihui Zhang; Xuying Zhang

A novel 2-thio-6-oxo-1,6-dihydropyrimidine-containing inhibitor of human lactate dehydrogenase (LDH) was identified by high-throughput screening (IC50=8.1 μM). Biochemical, surface plasmon resonance, and saturation transfer difference NMR experiments indicated that the compound specifically associated with human LDHA in a manner that required simultaneous binding of the NADH co-factor. Structural variation of the screening hit resulted in significant improvements in LDHA biochemical inhibition activity (best IC50=0.48 μM). A crystal structure of an optimized compound bound to human LDHA was obtained and explained many of the observed structure-activity relationships.


Journal of Medicinal Chemistry | 2008

A pentacyclic aurora kinase inhibitor (AKI-001) with high in vivo potency and oral bioavailability.

Thomas E. Rawson; Matthias Rüth; Elizabeth Blackwood; Dan Burdick; Laura Corson; Jenna Dotson; Jason Drummond; Carter Fields; Guy Georges; Bernhard Goller; Jason S. Halladay; Thomas Hunsaker; Tracy Kleinheinz; Hans-Willi Krell; Jun Li; Jun Liang; Anja Limberg; Angela McNutt; John Moffat; Gail Lewis Phillips; Yingqing Ran; Brian Safina; Mark Ultsch; Leslie Walker; Christian Wiesmann; Birong Zhang; Aihe Zhou; Bing-Yan Zhu; Petra Rüger; Andrea G. Cochran

Aurora kinase inhibitors have attracted a great deal of interest as a new class of antimitotic agents. We report a novel class of Aurora inhibitors based on a pentacyclic scaffold. A prototype pentacyclic inhibitor 32 (AKI-001) derived from two early lead structures improves upon the best properties of each parent and compares favorably to a previously reported Aurora inhibitor, 39 (VX-680). The inhibitor exhibits low nanomolar potency against both Aurora A and Aurora B enzymes, excellent cellular potency (IC50 < 100 nM), and good oral bioavailability. Phenotypic cellular assays show that both Aurora A and Aurora B are inhibited at inhibitor concentrations sufficient to block proliferation. Importantly, the cellular activity translates to potent inhibition of tumor growth in vivo. An oral dose of 5 mg/kg QD is well tolerated and results in near stasis (92% TGI) in an HCT116 mouse xenograft model.


Bioorganic & Medicinal Chemistry Letters | 2013

Identification of 2-amino-5-aryl-pyrazines as inhibitors of human lactate dehydrogenase.

Benjamin P. Fauber; Peter S. Dragovich; Jinhua Chen; Laura Corson; Charles Z. Ding; Charles Eigenbrot; Anthony M. Giannetti; Thomas Hunsaker; Sharada Labadie; Yichin Liu; Shiva Malek; David Peterson; Keith Pitts; Steven Sideris; Mark Ultsch; Erica VanderPorten; J Wang; Binqing Wei; Ivana Yen; Qin Yue

A 2-amino-5-aryl-pyrazine was identified as an inhibitor of human lactate dehydrogenase A (LDHA) via a biochemical screening campaign. Biochemical and biophysical experiments demonstrated that the compound specifically interacted with human LDHA. Structural variation of the screening hit resulted in improvements in LDHA biochemical inhibition and pharmacokinetic properties. A crystal structure of an improved compound bound to human LDHA was also obtained and it explained many of the observed structure-activity relationships.


Journal of Medicinal Chemistry | 2016

Discovery of (S)-1-(1-(4-Chloro-3-fluorophenyl)-2-hydroxyethyl)-4-(2-((1-methyl-1H-pyrazol-5-yl)amino)pyrimidin-4-yl)pyridin-2(1H)-one (GDC-0994), an Extracellular Signal-Regulated Kinase 1/2 (ERK1/2) Inhibitor in Early Clinical Development.

James F. Blake; Michael Burkard; Jocelyn Chan; Huifen Chen; Kang-Jye Chou; Dolores Díaz; Danette Dudley; John J. Gaudino; Stephen E. Gould; Jonas Grina; Thomas Hunsaker; Lichuan Liu; Matthew Martinson; David Moreno; Lars Mueller; Christine Orr; Patricia Pacheco; Ann Qin; Kevin Rasor; Li Ren; Kirk Robarge; Sheerin Shahidi-Latham; Jeffrey Stults; Francis J. Sullivan; Weiru Wang; JianPing Yin; Aihe Zhou; Marcia Belvin; Mark Merchant; John Moffat

The extracellular signal-regulated kinases ERK1/2 represent an essential node within the RAS/RAF/MEK/ERK signaling cascade that is commonly activated by oncogenic mutations in BRAF or RAS or by upstream oncogenic signaling. While targeting upstream nodes with RAF and MEK inhibitors has proven effective clinically, resistance frequently develops through reactivation of the pathway. Simultaneous targeting of multiple nodes in the pathway, such as MEK and ERK, offers the prospect of enhanced efficacy as well as reduced potential for acquired resistance. Described herein is the discovery and characterization of GDC-0994 (22), an orally bioavailable small molecule inhibitor selective for ERK kinase activity.


Bioorganic & Medicinal Chemistry Letters | 2014

Identification of substituted 3-hydroxy-2-mercaptocyclohex-2-enones as potent inhibitors of human lactate dehydrogenase.

Peter S. Dragovich; Benjamin P. Fauber; Jason Boggs; Jinhua Chen; Laura Corson; Charles Z. Ding; Charles Eigenbrot; HongXiu Ge; Anthony M. Giannetti; Thomas Hunsaker; Sharada Labadie; C Li; Yichin Liu; Shuguang Ma; Shiva Malek; David Peterson; Keith Pitts; Hans E. Purkey; Kirk Robarge; Laurent Salphati; Steven Sideris; Mark Ultsch; Erica VanderPorten; J Wang; Binqing Wei; Qing Xu; Ivana Yen; Qin Yue; Huihui Zhang; Xuying Zhang

A novel class of 3-hydroxy-2-mercaptocyclohex-2-enone-containing inhibitors of human lactate dehydrogenase (LDH) was identified through a high-throughput screening approach. Biochemical and surface plasmon resonance experiments performed with a screening hit (LDHA IC50=1.7 μM) indicated that the compound specifically associated with human LDHA in a manner that required simultaneous binding of the NADH co-factor. Structural variation of this screening hit resulted in significant improvements in LDHA biochemical inhibition activity (best IC50=0.18 μM). Two crystal structures of optimized compounds bound to human LDHA were obtained and explained many of the observed structure-activity relationships. In addition, an optimized inhibitor exhibited good pharmacokinetic properties after oral administration to rats (F=45%).


Cell Cycle | 2013

A high-content cellular senescence screen identifies candidate tumor suppressors, including EPHA3.

Jenni Lahtela; Laura Corson; Annabrita Hemmes; Matthew J. Brauer; Sonja Koopal; James Lee; Thomas Hunsaker; Peter K. Jackson; Emmy W. Verschuren

Activation of a cellular senescence program is a common response to prolonged oncogene activation or tumor suppressor loss, providing a physiological mechanism for tumor suppression in premalignant cells. The link between senescence and tumor suppression supports the hypothesis that a loss-of-function screen measuring bona fide senescence marker activation should identify candidate tumor suppressors. Using a high-content siRNA screening assay for cell morphology and proliferation measures, we identify 12 senescence-regulating kinases and determine their senescence marker signatures, including elevation of senescence-associated β-galactosidase, DNA damage and p53 or p16INK4a expression. Consistent with our hypothesis, SNP array CGH data supports loss of gene copy number of five senescence-suppressing genes across multiple tumor samples. One such candidate is the EPHA3 receptor tyrosine kinase, a gene commonly mutated in human cancer. We demonstrate that selected intracellular EPHA3 tumor-associated point mutations decrease receptor expression level and/or receptor tyrosine kinase (RTK) activity. Our study therefore describes a new strategy to mine for novel candidate tumor suppressors and provides compelling evidence that EPHA3 mutations may promote tumorigenesis only when key senescence-inducing pathways have been inactivated.


PLOS ONE | 2017

Combined MEK and ERK inhibition overcomes therapy-mediated pathway reactivation in RAS mutant tumors

Mark Merchant; John Moffat; Gabriele Schaefer; Jocelyn Chan; Xi Wang; Christine Orr; Jason H. Cheng; Thomas Hunsaker; Lily Shao; Stephanie J. Wang; Marie-Claire Wagle; Eva Lin; Peter M. Haverty; Sheerin Shahidi-Latham; Hai Ngu; Margaret Solon; Jeffrey Eastham-Anderson; Hartmut Koeppen; Shih-Min A. Huang; Jacob B. Schwarz; Marcia Belvin; Daniel C. Kirouac; Melissa R. Junttila

Mitogen-activated protein kinase (MAPK) pathway dysregulation is implicated in >30% of all cancers, rationalizing the development of RAF, MEK and ERK inhibitors. While BRAF and MEK inhibitors improve BRAF mutant melanoma patient outcomes, these inhibitors had limited success in other MAPK dysregulated tumors, with insufficient pathway suppression and likely pathway reactivation. In this study we show that inhibition of either MEK or ERK alone only transiently inhibits the MAPK pathway due to feedback reactivation. Simultaneous targeting of both MEK and ERK nodes results in deeper and more durable suppression of MAPK signaling that is not achievable with any dose of single agent, in tumors where feedback reactivation occurs. Strikingly, combined MEK and ERK inhibition is synergistic in RAS mutant models but only additive in BRAF mutant models where the RAF complex is dissociated from RAS and thus feedback productivity is disabled. We discovered that pathway reactivation in RAS mutant models occurs at the level of CRAF with combination treatment resulting in a markedly more active pool of CRAF. However, distinct from single node targeting, combining MEK and ERK inhibitor treatment effectively blocks the downstream signaling as assessed by transcriptional signatures and phospho-p90RSK. Importantly, these data reveal that MAPK pathway inhibitors whose activity is attenuated due to feedback reactivation can be rescued with sufficient inhibition by using a combination of MEK and ERK inhibitors. The MEK and ERK combination significantly suppresses MAPK pathway output and tumor growth in vivo to a greater extent than the maximum tolerated doses of single agents, and results in improved anti-tumor activity in multiple xenografts as well as in two Kras mutant genetically engineered mouse (GEM) models. Collectively, these data demonstrate that combined MEK and ERK inhibition is functionally unique, yielding greater than additive anti-tumor effects and elucidates a highly effective combination strategy in MAPK-dependent cancer, such as KRAS mutant tumors.


Cancer Cell | 2018

Pharmacological Induction of RAS-GTP Confers RAF Inhibitor Sensitivity in KRAS Mutant Tumors

Ivana Yen; Frances Shanahan; Mark Merchant; Christine Orr; Thomas Hunsaker; Matthew Durk; Hank La; Xiaolin Zhang; Scott E. Martin; Eva Lin; John K. Chan; Yihong Yu; Dhara Amin; Richard M. Neve; Amy Gustafson; Avinashnarayan Venkatanarayan; Scott A. Foster; Joachim Rudolph; Christiaan Klijn; Shiva Malek

Targeting KRAS mutant tumors through inhibition of individual downstream pathways has had limited clinical success. Here we report that RAF inhibitors exhibit little efficacy in KRAS mutant tumors. In combination drug screens, MEK and PI3K inhibitors synergized with pan-RAF inhibitors through an RAS-GTP-dependent mechanism. Broad cell line profiling with RAF/MEK inhibitor combinations revealed synergistic efficacy in KRAS mutant and wild-type tumors, with KRASG13D mutants exhibiting greater synergy versus KRASG12 mutant tumors. Mechanistic studies demonstrate that MEK inhibition induced RAS-GTP levels, RAF dimerization and RAF kinase activity resulting in MEK phosphorylation in synergistic tumor lines regardless of KRAS status. Taken together, our studies uncover a strategy to rewire KRAS mutant tumors to confer sensitivity to RAF kinase inhibition.

Collaboration


Dive into the Thomas Hunsaker's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge