Network


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

Hotspot


Dive into the research topics where Aaron C. Siegmund is active.

Publication


Featured researches published by Aaron C. Siegmund.


Journal of Medicinal Chemistry | 2008

Discovery and Optimization of Triazolopyridazines as Potent and Selective Inhibitors of the c-Met Kinase.

Brian K. Albrecht; Jean-Christophe Harmange; David Bauer; Loren Berry; Christiane Bode; Alessandro Boezio; April Chen; Deborah Choquette; Isabelle Dussault; Cary Fridrich; Satoko Hirai; Doug Hoffman; Jay Larrow; Paula Kaplan-Lefko; Jasmine Lin; Julia Lohman; Alexander M. Long; Jodi Moriguchi; Anne O'connor; Michele Potashman; Monica Reese; Karen Rex; Aaron C. Siegmund; Kavita Shah; Roman Shimanovich; Stephanie K. Springer; Yohannes Teffera; Yajing Yang; Yihong Zhang; Steven Bellon

Tumorigenesis is a multistep process in which oncogenes play a key role in tumor formation, growth, and maintenance. MET was discovered as an oncogene that is activated by its ligand, hepatocyte growth factor. Deregulated signaling in the c-Met pathway has been observed in multiple tumor types. Herein we report the discovery of potent and selective triazolopyridazine small molecules that inhibit c-Met activity.


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.


Journal of Medicinal Chemistry | 2008

Discovery of a Potent, Selective, and Orally Bioavailable c-Met Inhibitor: 1-(2-Hydroxy-2-methylpropyl)-N-(5-(7-methoxyquinolin-4-yloxy)pyridin-2-yl)-5-methyl-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carboxamide (AMG 458)

Longbin Liu; Aaron C. Siegmund; Ning Xi; Paula Kaplan-Lefko; Karen Rex; April Chen; Jasmine Lin; Jodi Moriguchi; Loren Berry; Liyue Huang; Yohannes Teffera; Yajing Yang; Yihong Zhang; Steven Bellon; Matthew R. Lee; Roman Shimanovich; Annette Bak; Celia Dominguez; Mark H. Norman; Jean-Christophe Harmange; Isabelle Dussault; Tae-Seong Kim

Deregulation of the receptor tyrosine kinase c-Met has been implicated in human cancers. Pyrazolones with N-1 bearing a pendent hydroxyalkyl side chain showed selective inhibition of c-Met over VEGFR2. However, studies revealed the generation of active, nonselective metabolites. Blocking this metabolic hot spot led to the discovery of 17 (AMG 458). When dosed orally, 17 significantly inhibited tumor growth in the NIH3T3/TPR-Met and U-87 MG xenograft models with no adverse effect on body weight.


Journal of Medicinal Chemistry | 2012

Structure-Based Design of Novel Class II c-Met Inhibitors: 2. SAR and Kinase Selectivity Profiles of the Pyrazolone Series

Longbin Liu; Mark H. Norman; Matthew R. Lee; Ning Xi; Aaron C. Siegmund; Alessandro Boezio; Shon Booker; Debbie Choquette; Noel D. D’Angelo; Julie Germain; Kevin Yang; Yajing Yang; Yihong Zhang; Steven Bellon; Douglas A. Whittington; Jean-Christophe Harmange; Celia Dominguez; Tae-Seong Kim; Isabelle Dussault

As part of our effort toward developing an effective therapeutic agent for c-Met-dependent tumors, a pyrazolone-based class II c-Met inhibitor, N-(4-((6,7-dimethoxyquinolin-4-yl)oxy)-3-fluorophenyl)-1,5-dimethyl-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carboxamide (1), was identified. Knowledge of the binding mode of this molecule in both c-Met and VEGFR-2 proteins led to a novel strategy for designing more selective analogues of 1. Along with detailed SAR information, we demonstrate that the low kinase selectivity associated with class II c-Met inhibitors can be improved significantly. This work resulted in the discovery of potent c-Met inhibitors with improved selectivity profiles over VEGFR-2 and IGF-1R that could serve as useful tools to probe the relationship between kinase selectivity and in vivo efficacy in tumor xenograft models. Compound 59e (AMG 458) was ultimately advanced into preclinical safety studies.


Journal of Medicinal Chemistry | 2013

Design, Optimization, and Biological Evaluation of Novel Keto-Benzimidazoles as Potent and Selective Inhibitors of Phosphodiesterase 10A (PDE10A).

Essa Hu; Roxanne Kunz; Ning Chen; Shannon Rumfelt; Aaron C. Siegmund; Kristin L. Andrews; Samer Chmait; Sharon Zhao; Carl D. Davis; Hang Chen; Dianna Lester-Zeiner; Ji Ma; Christopher Biorn; Jianxia Shi; Amy Porter; James J. S. Treanor; Jennifer R. Allen

Our development of PDE10A inhibitors began with an HTS screening hit (1) that exhibited both high p-glycoprotein (P-gp) efflux ratios in rat and human and poor metabolic stability. On the basis of cocrystal structure of 1 in human PDE10A enzyme, we designed a novel keto-benzimidazole 26 with comparable PDE10A potency devoid of efflux liabilities. On target in vivo coverage of PDE10A in rat brain was assessed using our previously reported LC-MS/MS receptor occupancy (RO) technology. Compound 26 achieved 55% RO of PDE10A at 30 mg/kg po and covered PDE10A receptors in rat brain in a dose-dependent manner. Cocrystal structure of 26 in PDE10A confirmed the binding mode of the novel scaffold. Further optimization resulted in the identification of keto-benzimidazole 34, which showed an increased in vivo efficacy of 57% RO in rats at 10 mg/kg po and an improved in vivo rat clearance and oral bioavailability.


Journal of Medicinal Chemistry | 2014

Small Molecule Disruptors of the Glucokinase–Glucokinase Regulatory Protein Interaction: 3. Structure–Activity Relationships within the Aryl Carbinol Region of the N-Arylsulfonamido-N′-arylpiperazine Series

Nobuko Nishimura; Mark H. Norman; Longbin Liu; Kevin C. Yang; Kate S. Ashton; Michael D. Bartberger; Samer Chmait; Jie Chen; Rod Cupples; Christopher Fotsch; Joan Helmering; Steven R. Jordan; Roxanne Kunz; Lewis D. Pennington; Steve F. Poon; Aaron C. Siegmund; Glenn Sivits; David J. Lloyd; Clarence Hale; David J. St. Jean

We have recently reported a novel approach to increase cytosolic glucokinase (GK) levels through the binding of a small molecule to its endogenous inhibitor, glucokinase regulatory protein (GKRP). These initial investigations culminated in the identification of 2-(4-((2S)-4-((6-amino-3-pyridinyl)sulfonyl)-2-(1-propyn-1-yl)-1-piperazinyl)phenyl)-1,1,1,3,3,3-hexafluoro-2-propanol (1, AMG-3969), a compound that effectively enhanced GK translocation and reduced blood glucose levels in diabetic animals. Herein we report the results of our expanded SAR investigations that focused on modifications to the aryl carbinol group of this series. Guided by the X-ray cocrystal structure of compound 1 bound to hGKRP, we identified several potent GK-GKRP disruptors bearing a diverse set of functionalities in the aryl carbinol region. Among them, sulfoximine and pyridinyl derivatives 24 and 29 possessed excellent potency as well as favorable PK properties. When dosed orally in db/db mice, both compounds significantly lowered fed blood glucose levels (up to 58%).


Journal of Medicinal Chemistry | 2015

Discovery and Structure-Guided Optimization of Diarylmethanesulfonamide Disrupters of Glucokinase–Glucokinase Regulatory Protein (GK–GKRP) Binding: Strategic Use of a N → S (nN → σ*S–X) Interaction for Conformational Constraint

Lewis D. Pennington; Michael D. Bartberger; Michael Croghan; Kristin L. Andrews; Kate S. Ashton; Matthew P. Bourbeau; Jie Chen; Samer Chmait; Rod Cupples; Christopher Fotsch; Joan Helmering; Fang-Tsao Hong; Randall W. Hungate; Steven R. Jordan; Ke Kong; Longbin Liu; Klaus Michelsen; Carolyn Moyer; Nobuko Nishimura; Mark H. Norman; Andreas Reichelt; Aaron C. Siegmund; Glenn Sivits; Seifu Tadesse; Christopher M. Tegley; Gwyneth Van; Kevin C. Yang; Guomin Yao; Jiandong Zhang; David J. Lloyd

The HTS-based discovery and structure-guided optimization of a novel series of GKRP-selective GK-GKRP disrupters are revealed. Diarylmethanesulfonamide hit 6 (hGK-hGKRP IC50 = 1.2 μM) was optimized to lead compound 32 (AMG-0696; hGK-hGKRP IC50 = 0.0038 μM). A stabilizing interaction between a nitrogen atom lone pair and an aromatic sulfur system (nN → σ*S-X) in 32 was exploited to conformationally constrain a biaryl linkage and allow contact with key residues in GKRP. Lead compound 32 was shown to induce GK translocation from the nucleus to the cytoplasm in rats (IHC score = 0; 10 mg/kg po, 6 h) and blood glucose reduction in mice (POC = -45%; 100 mg/kg po, 3 h). X-ray analyses of 32 and several precursors bound to GKRP were also obtained. This novel disrupter of GK-GKRP binding enables further exploration of GKRP as a potential therapeutic target for type II diabetes and highlights the value of exploiting unconventional nonbonded interactions in drug design.


Journal of Medicinal Chemistry | 2013

Piperazine Oxadiazole Inhibitors of Acetyl-CoA Carboxylase

Matthew P. Bourbeau; Aaron C. Siegmund; John G. Allen; Hong Shu; Christopher Fotsch; Michael D. Bartberger; Ki-Won Kim; Renee Komorowski; Melissa Graham; James Busby; Minghan Wang; James Meyer; Yang Xu; Kevin Salyers; Mark R. Fielden; Murielle M. Véniant; Wei Gu

Acetyl-CoA carboxylase (ACC) is a target of interest for the treatment of metabolic syndrome. Starting from a biphenyloxadiazole screening hit, a series of piperazine oxadiazole ACC inhibitors was developed. Initial pharmacokinetic liabilities of the piperazine oxadiazoles were overcome by blocking predicted sites of metabolism, resulting in compounds with suitable properties for further in vivo studies. Compound 26 was shown to inhibit malonyl-CoA production in an in vivo pharmacodynamic assay and was advanced to a long-term efficacy study. Prolonged dosing with compound 26 resulted in impaired glucose tolerance in diet-induced obese (DIO) C57BL6 mice, an unexpected finding.


Cancer Research | 2013

Abstract 711: Small molecule compounds that target cell division cycle 7 (Cdc7) kinase inhibit cell proliferation and tumor growth.

Julie M. Bailis; Li Fang; Jessica Orf; Scott Heller; Tammy L. Bush; Matthew P. Bourbeau; Sonia Escobar; Michael J. Frohn; Paul E. Harrington; Faye Hsieh; Alexander J. Pickrell; Kelvin Sham; Aaron C. Siegmund; Helming Tan; Leeanne Zalameda; John G. Allen; Dineli Wickramasinghe

Cdc7 is an essential, serine/threonine protein kinase that activates the initiation of DNA synthesis at replication origins. Cdc7 also promotes cell cycle checkpoint activation in response to replication stress. As a key regulator of S phase entry and progression, Cdc7 kinase is a potential target for cancer therapy, with a distinct mechanism of action from known drugs that inhibit DNA replication. Following a high throughput screen for inhibitors of Cdc7 kinase activity, we investigated structure-activity relationships of azole-based compounds and optimized the compounds for potency and pharmacokinetic properties. Here we present the characterization of one of these compounds as a potent, selective, bioavailable Cdc7 kinase inhibitor. In cells, Cdc7 inhibition decreases MCM2 phosphorylation and DNA synthesis, causes DNA damage, and slows S phase progression. Cdc7 inhibition also induces chromosome missegregation leading to cell lethality in vitro and tumor growth inhibition in vivo. Cdc7 inhibition provides a new approach to target cancers, either as a single agent or in combination with chemotherapy. Citation Format: Julie Bailis, Li Fang, Jessica Orf, Scott Heller, Tammy Bush, Matthew Bourbeau, Sonia Escobar, Michael Frohn, Paul Harrington, Faye Hsieh, Alexander Pickrell, Kelvin Sham, Aaron Siegmund, Helming Tan, Leeanne Zalameda, John Allen, Dineli Wickramasinghe. Small molecule compounds that target cell division cycle 7 (Cdc7) kinase inhibit cell proliferation and tumor growth. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 711. doi:10.1158/1538-7445.AM2013-711


Synlett | 2008

Selective β-Hydroxyethylation at the N-1 Position of a Pyrazolone: Synthesis of Benzyl 1-(β-Hydroxyethyl)-5-methyl-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carboxylate

Aaron C. Siegmund; Daniel Retz; Ning Xi; Celia Dominguez; Roland Bürli; Longbin Liu

Selective 2-hydroxyethylation at the N-1 position of benzyl 5-methyl-3-oxo-2-phenyl-2,3-dihydro-1 H-pyrazole-4-carboxylate with epoxides was achieved using either AlMe 3 or Mg(ClO 4 ) 2 under mild conditions. The epoxide ring opening was both regioselective and stereospecific. Moderate to excellent yields were obtained from mono- and disubstituted epoxides with the exception of CIS-dimethyl-2-butene oxide that gave only a trace amount of the product.

Collaboration


Dive into the Aaron C. Siegmund's collaboration.

Researchain Logo
Decentralizing Knowledge