Network


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

Hotspot


Dive into the research topics where Matthew P. Bourbeau is active.

Publication


Featured researches published by Matthew P. Bourbeau.


Journal of Medicinal Chemistry | 2014

Discovery of clinical candidate 1-(4-(3-(4-(1H-benzo[d]imidazole-2-carbonyl)phenoxy)pyrazin-2-yl)piperidin-1-yl)ethanone (AMG 579), a potent, selective, and efficacious inhibitor of phosphodiesterase 10A (PDE10A).

Essa Hu; Ning Chen; Matthew P. Bourbeau; Paul E. Harrington; Kaustav Biswas; Roxanne Kunz; Kristin L. Andrews; Samer Chmait; Xiaoning Zhao; Carl D. Davis; Ji Ma; Jianxia Shi; Dianna Lester-Zeiner; Jean Danao; Jessica Able; Madelyn Cueva; Santosh Talreja; Thomas Kornecook; Hang Chen; Amy Porter; Randall W. Hungate; James J. S. Treanor; Jennifer R. Allen

We report the identification of a PDE10A clinical candidate by optimizing potency and in vivo efficacy of promising keto-benzimidazole leads 1 and 2. Significant increase in biochemical potency was observed when the saturated rings on morpholine 1 and N-acetyl piperazine 2 were changed by a single atom to tetrahydropyran 3 and N-acetyl piperidine 5. A second single atom modification from pyrazines 3 and 5 to pyridines 4 and 6 improved the inhibitory activity of 4 but not 6. In the in vivo LC-MS/MS target occupancy (TO) study at 10 mg/kg, 3, 5, and 6 achieved 86-91% occupancy of PDE10A in the brain. Furthermore, both CNS TO and efficacy in PCP-LMA behavioral model were observed in a dose dependent manner. With superior in vivo TO, in vivo efficacy and in vivo PK profiles in multiple preclinical species, compound 5 (AMG 579) was advanced as our PDE10A clinical candidate.


Journal of Medicinal Chemistry | 2015

Recent advances in the development of acetyl-CoA carboxylase (ACC) inhibitors for the treatment of metabolic disease.

Matthew P. Bourbeau; Michael D. Bartberger

The development of acetyl-CoA carboxylase (ACC) inhibitors for the treatment of metabolic disease has been pursued by the pharmaceutical industry for some time. A number of recent disclosures describing potent ACC inhibitors have been reported by multiple research groups. Unlike many prior publications in this area, more recent publications contain a significant amount of in vivo efficacy data generated by long-term experiments in rodent models of metabolic disease. Additionally, one compound has been advanced to human clinical studies. The results from these studies should allow researchers to better gauge the potential utility of ACC inhibition for the treatment of human disease.


Chemical Research in Toxicology | 2010

Cytochrome P450-Mediated Epoxidation of 2-Aminothiazole-Based AKT Inhibitors: Identification of Novel GSH Adducts and Reduction of Metabolic Activation through Structural Changes Guided by in Silico and in Vitro Screening

Raju Subramanian; Matthew R. Lee; John G. Allen; Matthew P. Bourbeau; Christopher Fotsch; Fang-Tsao Hong; Seifu Tadesse; Guomin Yao; Chester Chenguang Yuan; Sekhar Surapaneni; Gary L. Skiles; Xianghong Wang; G. Erich Wohlhieter; Qingping Zeng; Yihong Zhou; Xiaochun Zhu; Chun Li

A 2-aminothiazole derivative 1 was developed as a potential inhibitor of the oncology target AKT, a serine/threonine kinase. When incubated in rat and human liver microsomes in the presence of NADPH, 1 underwent significant metabolic activation on its 2-aminothiazole ring, leading to substantial covalent protein binding. Upon addition of glutathione, covalent binding was reduced significantly, and multiple glutathione adducts were detected. Novel metabolites from the in vitro incubates were characterized by LC-MS and NMR to discern the mechanism of bioactivation. An in silico model was developed based on the proposed mechanism and was employed to predict bioactivation in 23 structural analogues. The predictions were confirmed empirically for the bioactivation liability, in vitro, by LC-MS methods screening for glutathione incorporation. New compounds were identified with a low propensity for bioactivation.


Bioorganic & Medicinal Chemistry Letters | 2010

2-Aminothiadiazole inhibitors of AKT1 as potential cancer therapeutics.

Qingping Zeng; Matthew P. Bourbeau; G. Erich Wohlhieter; Guomin Yao; Holger Monenschein; James T. Rider; Matthew R. Lee; Shiwen Zhang; Julie A. Lofgren; Daniel J. Freeman; Chun Li; Elizabeth Tominey; Xin Huang; Douglas Hoffman; Harvey Yamane; Andrew Tasker; Celia Dominguez; Vellarkad N. Viswanadhan; Randall W. Hungate; Xiaoling Zhang

A series of 2-aminothiadiazole of inhibitors of AKT1 is described. SAR relationships are discussed, along with selectivity for protein kinase A (PKA) and cyclin-dependent kinase 2 (CDK2). Moderate selectivity observed in several compounds for AKT1 versus PKA is rationalized by X-ray crystallographic analysis. Key compounds showed activity in cellular assays measuring phosphorylation of two AKT substrates, PRAS40 and FKHRL1. Compound 30 was advanced to a mouse liver PD assay, where it showed dose-dependent inhibition of AKT activity, as measured by the inhibition of phospho-PRAS40.


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.


Annual Reports in Medicinal Chemistry | 2010

Recent Advances in Acetyl-CoA Carboxylase Inhibitors

Matthew P. Bourbeau; John G. Allen; Wei Gu

Publisher Summary This chapter discusses the recent advances made in acetyl– CoA carboxylase inhibitors. Significant progress has been made in the development of potent inhibitors of the two acetyl-CoA carboxylases (ACC) isoforms. A number of different chemotypes have been designed, some of which possess selectivity for ACC1 or ACC2. Published mouse knockout (KO) studies from two research groups, using different constructs and different backgrounds, have provided conflicting results. However, limited in vivo efficacy studies suggest that a beneficial effect may be seen in DIO mouse models of diabetes when treated with an ACC1/2 dual inhibitor, and thus ACC inhibition still remains a promising therapeutic approach. It remains to be seen whether ACC1 or ACC2 selective inhibitors will show beneficial in vivo effects and whether inhibition of ACC will be a useful treatment for metabolic disease in humans. This chapter also discusses the available data showing the efficacy of ACC inhibition in long-term studies.


Bioorganic & Medicinal Chemistry Letters | 2010

Azole-based inhibitors of AKT/PKB for the treatment of cancer.

Qingping Zeng; John G. Allen; Matthew P. Bourbeau; Xianghong Wang; Guomin Yao; Seifu Tadesse; James T. Rider; Chester Chenguang Yuan; Fang-Tsao Hong; Matthew R. Lee; Shiwen Zhang; Julie A. Lofgren; Daniel J. Freeman; Suijin Yang; Chun Li; Elizabeth Tominey; Xin Huang; Douglas Hoffman; Harvey Yamane; Christopher Fotsch; Celia Dominguez; Randall W. Hungate; Xiaoling Zhang

Through a combination of screening and structure-based rational design, we have discovered a series of N(1)-(5-(heterocyclyl)-thiazol-2-yl)-3-(4-trifluoromethylphenyl)-1,2-propanediamines that were developed into potent ATP competitive inhibitors of AKT. Studies of linker strand-binding adenine isosteres identified SAR trends in potency and selectivity that were consistent with binding interactions observed in structures of the inhibitors bound to AKT1 and to the counter-screening target PKA. One compound was shown to have acceptable pharmacokinetic properties and to be a potent inhibitor of AKT signaling and of in vivo xenograft tumor growth in a preclinical model of glioblastoma.


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.


Journal of Organic Chemistry | 2014

Nonracemic Synthesis of GK–GKRP Disruptor AMG-3969

Matthew P. Bourbeau; Kate S. Ashton; Jie Yan; David J. St. Jean

A nonracemic synthesis of the glucokinase-glucokinase regulatory protein disruptor AMG-3969 (5) is reported. Key features of the synthetic approach are an asymmetric synthesis of the 2-alkynyl piperazine core via a base-promoted isomerization and a revised approach to the synthesis of the aminopyridinesulfonamide with an improved safety profile.


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

Collaboration


Dive into the Matthew P. Bourbeau's collaboration.

Researchain Logo
Decentralizing Knowledge