Network


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

Hotspot


Dive into the research topics where Vincent B. Luscombe is active.

Publication


Featured researches published by Vincent B. Luscombe.


ACS Medicinal Chemistry Letters | 2017

Discovery of VU0467485/AZ13713945: An M4 PAM Evaluated as a Preclinical Candidate for the Treatment of Schizophrenia

Michael R. Wood; Meredith J. Noetzel; Bruce J. Melancon; Michael S. Poslusney; Kellie D. Nance; Miguel A. Hurtado; Vincent B. Luscombe; Rebecca L. Weiner; Alice L. Rodriguez; Atin Lamsal; Sichen Chang; Michael Bubser; Anna L. Blobaum; Darren W. Engers; Colleen M. Niswender; Carrie K. Jones; Nicholas J. Brandon; Michael W. Wood; Mark E. Duggan; P. Jeffrey Conn; Thomas M. Bridges; Craig W. Lindsley

Herein, we report the structure-activity relationships within a series of potent, selective, and orally bioavailable muscarinic acetylcholine receptor 4 (M4) positive allosteric modulators (PAMs). Compound 6c (VU0467485) possesses robust in vitro M4 PAM potency across species and in vivo efficacy in preclinical models of schizophrenia. Coupled with an attractive DMPK profile and suitable predicted human PK, 6c (VU0467485) was evaluated as a preclinical development candidate.


ACS Chemical Neuroscience | 2017

Diverse Effects on M1 Signaling and Adverse Effect Liability within a Series of M1 Ago-PAMs

Jerri M. Rook; Masahito Abe; Hyekyung P. Cho; Kellie D. Nance; Vincent B. Luscombe; Jeffrey J. Adams; Jonathan W. Dickerson; Daniel H. Remke; Pedro M. Garcia-Barrantes; Darren W. Engers; Julie L. Engers; Sichen Chang; Jarrett J. Foster; Anna L. Blobaum; Colleen M. Niswender; Carrie K. Jones; P. Jeffrey Conn; Craig W. Lindsley

Both historical clinical and recent preclinical data suggest that the M1 muscarinic acetylcholine receptor is an exciting target for the treatment of Alzheimers disease and the cognitive and negative symptom clusters in schizophrenia; however, early drug discovery efforts targeting the orthosteric binding site have failed to afford selective M1 activation. Efforts then shifted to focus on selective activation of M1 via either allosteric agonists or positive allosteric modulators (PAMs). While M1 PAMs have robust efficacy in rodent models, some chemotypes can induce cholinergic adverse effects (AEs) that could limit their clinical utility. Here, we report studies aimed at understanding the subtle structural and pharmacological nuances that differentiate efficacy from adverse effect liability within an indole-based series of M1 ago-PAMs. Our data demonstrate that closely related M1 PAMs can display striking differences in their in vivo activities, especially their propensities to induce adverse effects. We report the discovery of a novel PAM in this series that is devoid of observable adverse effect liability. Interestingly, the molecular pharmacology profile of this novel PAM is similar to that of a representative M1 PAM that induces severe AEs. For instance, both compounds are potent ago-PAMs that demonstrate significant interaction with the orthosteric site (either bitopic or negative cooperativity). However, there are subtle differences in efficacies of the compounds at potentiating M1 responses, agonist potencies, and abilities to induce receptor internalization. While these differences may contribute to the differential in vivo profiles of these compounds, the in vitro differences are relatively subtle and highlight the complexities of allosteric modulators and the need to focus on in vivo phenotypic screening to identify safe and effective M1 PAMs.


Bioorganic & Medicinal Chemistry Letters | 2017

Challenges in the development of an M4 PAM in vivo tool compound: The discovery of VU0467154 and unexpected DMPK profiles of close analogs

Michael R. Wood; Meredith J. Noetzel; Michael S. Poslusney; Bruce J. Melancon; James C. Tarr; Atin Lamsal; Sichen Chang; Vincent B. Luscombe; Rebecca L. Weiner; Hyekyung P. Cho; Michael Bubser; Carrie K. Jones; Colleen M. Niswender; Michael W. Wood; Darren W. Engers; Nicholas J. Brandon; Mark E. Duggan; P. Jeffrey Conn; Thomas M. Bridges; Craig W. Lindsley

This letter describes the chemical optimization of a novel series of M4 positive allosteric modulators (PAMs) based on a 5-amino-thieno[2,3-c]pyridazine core, developed via iterative parallel synthesis, and culminating in the highly utilized rodent in vivo tool compound, VU0467154 (5). This is the first report of the optimization campaign (SAR and DMPK profiling) that led to the discovery of VU0467154, and details all of the challenges faced in allosteric modulator programs (steep SAR, species differences in PAM pharmacology and subtle structural changes affecting CNS penetration).


Journal of Medicinal Chemistry | 2017

Discovery of N-(5-Fluoropyridin-2-yl)-6-methyl-4-(pyrimidin-5-yloxy)picolinamide (VU0424238): A Novel Negative Allosteric Modulator of Metabotropic Glutamate Receptor Subtype 5 Selected for Clinical Evaluation

Andrew S. Felts; Alice L. Rodriguez; Anna L. Blobaum; Ryan D. Morrison; Brittney S. Bates; Analisa Thompson Gray; Jerri M. Rook; M. N. Tantawy; Frank W. Byers; Sichen Chang; Daryl F. Venable; Vincent B. Luscombe; Gilles Tamagnan; Colleen M. Niswender; J. Scott Daniels; Carrie K. Jones; P. Jeffrey Conn; Craig W. Lindsley; Kyle A. Emmitte

Preclinical evidence in support of the potential utility of mGlu5 NAMs for the treatment of a variety of psychiatric and neurodegenerative disorders is extensive, and multiple such molecules have entered clinical trials. Despite some promising results from clinical studies, no small molecule mGlu5 NAM has yet to reach market. Here we present the discovery and evaluation of N-(5-fluoropyridin-2-yl)-6-methyl-4-(pyrimidin-5-yloxy)picolinamide (27, VU0424238), a compound selected for clinical evaluation. Compound 27 is more than 900-fold selective for mGlu5 versus the other mGlu receptors, and binding studies established a Ki value of 4.4 nM at a known allosteric binding site. Compound 27 had a clearance of 19.3 and 15.5 mL/min/kg in rats and cynomolgus monkeys, respectively. Imaging studies using a known mGlu5 PET ligand demonstrated 50% receptor occupancy at an oral dose of 0.8 mg/kg in rats and an intravenous dose of 0.06 mg/kg in baboons.


ACS Medicinal Chemistry Letters | 2017

Discovery of VU6005649, a CNS Penetrant mGlu7/8 Receptor PAM Derived from a Series of Pyrazolo[1,5-a]pyrimidines

Masahito Abe; Mabel Seto; Rocco G. Gogliotti; Matthew T. Loch; Katrina A. Bollinger; Sichen Chang; Eileen M. Engelberg; Vincent B. Luscombe; Joel M. Harp; Michael Bubser; Darren W. Engers; Carrie K. Jones; Alice L. Rodriguez; Anna L. Blobaum; P. Jeffrey Conn; Colleen M. Niswender; Craig W. Lindsley

Herein, we report the structure-activity relationships within a series of mGlu7 PAMs based on a pyrazolo[1,5-a]pyrimidine core with excellent CNS penetration (Kps > 1 and Kp,uus > 1). Analogues in this series proved to display a range of Group III mGlu receptor selectivity, but VU6005649 emerged as the first dual mGlu7/8 PAM, filling a void in the Group III mGlu receptor PAM toolbox and demonstrating in vivo efficacy in a mouse contextual fear conditioning model.


ACS Medicinal Chemistry Letters | 2017

VU6010608, a Novel mGlu7 NAM from a Series of N-(2-(1H-1,2,4-Triazol-1-yl)-5-(trifluoromethoxy)phenyl)benzamides

Carson W. Reed; Kevin M. McGowan; Paul K. Spearing; Branden J. Stansley; Hanna F. Roenfanz; Darren W. Engers; Alice L. Rodriguez; Eileen M. Engelberg; Vincent B. Luscombe; Matthew T. Loch; Daniel H. Remke; Jerri M. Rook; Anna L. Blobaum; P. Jeffrey Conn; Colleen M. Niswender; Craig W. Lindsley

Herein, we report the structure-activity relationships within a series of mGlu7 NAMs based on an N-(2-(1H-1,2,4-triazol-1-yl)-5-(trifluoromethoxy)phenyl)benzamide core with excellent CNS penetration (Kp 1.9-5.8 and Kp,uu 0.4-1.4). Analogues in this series displayed steep SAR. Of these, VU6010608 (11a) emerged with robust efficacy in blocking high frequency stimulated long-term potentiation in electrophysiology studies.


Bioorganic & Medicinal Chemistry Letters | 2017

Synthesis and evaluation of 4,6-disubstituted pyrimidines as CNS penetrant pan-muscarinic antagonists with a novel chemotype

Aaron M. Bender; Rebecca L. Weiner; Vincent B. Luscombe; Hyekyung P. Cho; Colleen M. Niswender; Darren W. Engers; Thomas M. Bridges; P. Jeffrey Conn; Craig W. Lindsley

This letter describes the synthesis and structure activity relationship (SAR) studies of structurally novel M4 antagonists, based on a 4,6-disubstituted core, identified from a high-throughput screening campaign. A multi-dimensional optimization effort enhanced potency at both human and rat M4 (IC50s<300nM), with no substantial species differences noted. Moreover, CNS penetration proved attractive for this series (brain:plasma Kp,uu=0.87), while other DMPK attributes were addressed in the course of the optimization effort, providing low in vivo clearance in rat (CLp=5.37mL/min/kg). Surprisingly, this series displayed pan-muscarinic antagonist activity across M1-5, despite the absence of the prototypical basic or quaternary amine moiety, thus offering a new chemotype from which to develop a next generation of pan-muscarinic antagonist agents.


Bioorganic & Medicinal Chemistry Letters | 2017

Discovery and optimization of 3-(4-aryl/heteroarylsulfonyl)piperazin-1-yl)-6-(piperidin-1-yl)pyridazines as novel, CNS penetrant pan-muscarinic antagonists

Aaron M. Bender; Rebecca L. Weiner; Vincent B. Luscombe; Sonia Ajmera; Hyekyung P. Cho; Sichen Chang; Xiaoyan Zhan; Alice L. Rodriguez; Colleen M. Niswender; Darren W. Engers; Thomas M. Bridges; P. Jeffrey Conn; Craig W. Lindsley

This letter describes the synthesis and structure activity relationship (SAR) studies of structurally novel M4 antagonists, based on a 3-(4-aryl/heteroarylsulfonyl)piperazin-1-yl)-6-(piperidin-1-yl)pyridazine core, identified from a high-throughput screening campaign. A multi-dimensional optimization effort enhanced potency at human M4 (hM4 IC50s<200nM), with only moderate species differences noted, and with enantioselective inhibition. Moreover, CNS penetration proved attractive for this series (rat brain:plasma Kp=2.1, Kp,uu=1.1). Despite the absence of the prototypical mAChR antagonist basic or quaternary amine moiety, this series displayed pan-muscarinic antagonist activity across M1-5 (with 9- to 16-fold functional selectivity at best). This series further expands the chemical diversity of mAChR antagonists.


Bioorganic & Medicinal Chemistry Letters | 2017

Challenges in the development of an M4 PAM preclinical candidate: The discovery, SAR, and biological characterization of a series of azetidine-derived tertiary amides

James C. Tarr; Michael R. Wood; Meredith J. Noetzel; Bruce J. Melancon; Atin Lamsal; Vincent B. Luscombe; Alice L. Rodriguez; Frank W. Byers; Sichen Chang; Hyekyung P. Cho; Darren W. Engers; Carrie K. Jones; Colleen M. Niswender; Michael W. Wood; Nicholas J. Brandon; Mark E. Duggan; P. Jeffrey Conn; Thomas M. Bridges; Craig W. Lindsley

Herein we describe the continued optimization of M4 positive allosteric modulators (PAMs) within the 5-amino-thieno[2,3-c]pyridazine series of compounds. In this letter, we disclose our studies on tertiary amides derived from substituted azetidines. This series provided excellent CNS penetration, which had been challenging to consistently achieve in other amide series. Efforts to mitigate high clearance, aided by metabolic softspot analysis, were unsuccessful and precluded this series from further consideration as a preclinical candidate. In the course of this study, we found that potassium tetrafluoroborate salts could be engaged in a tosyl hydrazone reductive cross coupling reaction, a previously unreported transformation, which expands the synthetic utility of the methodology.


Journal of Medicinal Chemistry | 2018

Discovery of novel central nervous system penetrant metabotropic glutamate receptor subtype 2 (mGlu2) Negative Allosteric Modulators (NAMs) based on functionalized pyrazolo[1,5-a]pyrimidine-5-carboxamide and thieno[3,2-b]pyridine-5-carboxamide cores

Elizabeth S. Childress; Joshua M. Wieting; Andrew S. Felts; Megan M Breiner; Madeline F Long; Vincent B. Luscombe; Alice L. Rodriguez; Hyekyung P. Cho; Anna L. Blobaum; Colleen M. Niswender; Kyle A. Emmitte; P. Jeffrey Conn; Craig W. Lindsley

A scaffold hopping exercise from a monocyclic mGlu2 NAM with poor rodent PK led to two novel heterobicyclic series of mGlu2 NAMs based on either a functionalized pyrazolo[1,5- a]pyrimidine-5-carboxamide core or a thieno[3,2- b]pyridine-5-carboxamide core. These novel analogues possess enhanced rodent PK, while also maintaining good mGlu2 NAM potency, selectivity (versus mGlu3 and the remaining six mGlu receptors), and high CNS penetration. Interestingly, SAR was divergent between the new 5,6-heterobicyclic systems.

Collaboration


Dive into the Vincent B. Luscombe'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