Network


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

Hotspot


Dive into the research topics where Sichen Chang is active.

Publication


Featured researches published by Sichen Chang.


Journal of Medicinal Chemistry | 2015

Discovery of a Selective and CNS Penetrant Negative Allosteric Modulator of Metabotropic Glutamate Receptor Subtype 3 with Antidepressant and Anxiolytic Activity in Rodents.

Julie L. Engers; Alice L. Rodriguez; Leah C. Konkol; Ryan D. Morrison; Analisa D. Thompson; Frank W. Byers; Anna L. Blobaum; Sichen Chang; Daryl F. Venable; Matthew T. Loch; Colleen M. Niswender; J. Scott Daniels; Carrie K. Jones; P. Jeffrey Conn; Craig W. Lindsley; Kyle A. Emmitte

Previous preclinical work has demonstrated the therapeutic potential of antagonists of the group II metabotropic glutamate receptors (mGlus). Still, compounds that are selective for the individual group II mGlus (mGlu2 and mGlu3) have been scarce. There remains a need for such compounds with the balance of properties suitable for convenient use in a wide array of rodent behavioral studies. We describe here the discovery of a selective mGlu3 NAM 106 (VU0650786) suitable for in vivo work. Compound 106 is a member of a series of 5-aryl-6,7-dihydropyrazolo[1,5-a]pyrazine-4(5H)-one compounds originally identified as a mGlu5 positive allosteric modulator (PAM) chemotype. Its suitability for use in rodent behavioral models has been established by extensive in vivo PK studies, and the behavioral experiments presented here with compound 106 represent the first examples in which an mGlu3 NAM has demonstrated efficacy in models where prior efficacy had previously been noted with nonselective group II antagonists.


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.


Bioorganic & Medicinal Chemistry Letters | 2016

Discovery and optimization of a novel series of highly CNS penetrant M4 PAMs based on a 5,6-dimethyl-4-(piperidin-1-yl)thieno[2,3-d]pyrimidine core

Michael R. Wood; Meredith J. Noetzel; Julie L. Engers; Katrina A. Bollinger; Bruce J. Melancon; James C. Tarr; Changho Han; Mary West; Alison R. Gregro; Atin Lamsal; Sichen Chang; Sonia Ajmera; Emery Smith; Peter Chase; Peter Hodder; Michael Bubser; Carrie K. Jones; Corey R. Hopkins; Kyle A. Emmitte; Colleen M. Niswender; Michael W. Wood; 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 5,6-dimethyl-4-(piperidin-1-yl)thieno[2,3-d]pyrimidine core, identified from an MLPCN functional high-throughput screen. The HTS hit was potent and selective, but not CNS penetrant. Potency was maintained, while CNS penetration was improved (rat brain:plasma Kp=0.74), within the original core after several rounds of optimization; however, the thieno[2,3-d]pyrimidine core was subject to extensive oxidative metabolism. Ultimately, we identified a 6-fluoroquinazoline core replacement that afforded good M4 PAM potency, muscarinic receptor subtype selectivity and CNS penetration (rat brain:plasma Kp>10). Moreover, this campaign provided fundamentally distinct M4 PAM chemotypes, greatly expanding the available structural diversity for this exciting CNS target.


Journal of Medicinal Chemistry | 2015

Design of 4-Oxo-1-aryl-1,4-dihydroquinoline-3-carboxamides as Selective Negative Allosteric Modulators of Metabotropic Glutamate Receptor Subtype 2

Andrew S. Felts; Alice L. Rodriguez; Katrina A. Smith; Julie L. Engers; Ryan D. Morrison; Frank W. Byers; Blobaum Al; Locuson Cw; Sichen Chang; Daryl F. Venable; Colleen M. Niswender; Daniels Js; P.J. Conn; Craig W. Lindsley; Kyle A. Emmitte

Both orthosteric and allosteric antagonists of the group II metabotropic glutamate receptors (mGlus) have been used to establish a link between mGlu2/3 inhibition and a variety of CNS diseases and disorders. Though these tools typically have good selectivity for mGlu2/3 versus the remaining six members of the mGlu family, compounds that are selective for only one of the individual group II mGlus have proved elusive. Herein we report on the discovery of a potent and highly selective mGlu2 negative allosteric modulator 58 (VU6001192) from a series of 4-oxo-1-aryl-1,4-dihydroquinoline-3-carboxamides. The concept for the design of this series centered on morphing a quinoline series recently disclosed in the patent literature into a chemotype previously used for the preparation of muscarinic acetylcholine receptor subtype 1 positive allosteric modulators. Compound 58 exhibits a favorable profile and will be a useful tool for understanding the biological implications of selective inhibition of mGlu2 in the CNS.


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).


Bioorganic & Medicinal Chemistry Letters | 2016

Discovery and SAR of a novel series of potent, CNS penetrant M4 PAMs based on a non-enolizable ketone core: Challenges in disposition.

Michael R. Wood; Meredith J. Noetzel; James C. Tarr; Alice L. Rodriguez; Atin Lamsal; Sichen Chang; Jarrett J. Foster; Emery Smith; Peter Chase; Peter Hodder; Darren W. Engers; Colleen M. Niswender; Nicholas J. Brandon; Michael W. Wood; Mark E. Duggan; P. Jeffrey Conn; Thomas M. Bridges; Craig W. Lindsley

This Letter describes the chemical optimization of a novel series of M4 PAMs based on a non-enolizable ketone core, identified from an MLPCN functional high-throughput screen. The HTS hit was potent, selective and CNS penetrant; however, the compound was highly cleared in vitro and in vivo. SAR provided analogs for which M4 PAM potency and CNS exposure were maintained; yet, clearance remained high. Metabolite identification studies demonstrated that this series was subject to rapid, and near quantitative, reductive metabolism to the corresponding secondary alcohol metabolite that was devoid of M4 PAM activity.


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.


Bioorganic & Medicinal Chemistry Letters | 2017

Optimization of M 4 positive allosteric modulators (PAMs): The discovery of VU0476406, a non-human primate in vivo tool compound for translational pharmacology

Bruce J. Melancon; Michael R. Wood; Meredith J. Noetzel; Kellie D. Nance; Eileen M. Engelberg; Changho Han; Atin Lamsal; Sichen Chang; Hyekyung P. Cho; Frank W. Byers; Michael Bubser; Carrie K. Jones; Colleen M. Niswender; Michael W. Wood; Darren W. Engers; Dedong Wu; Nicholas J. Brandon; Mark E. Duggan; P. Jeffrey Conn; Thomas M. Bridges; Craig W. Lindsley

This letter describes the further chemical optimization of the 5-amino-thieno[2,3-c]pyridazine series (VU0467154/VU0467485) of M4 positive allosteric modulators (PAMs), developed via iterative parallel synthesis, culminating in the discovery of the non-human primate (NHP) in vivo tool compound, VU0476406 (8p). VU0476406 is an important in vivo tool compound to enable translation of pharmacodynamics from rodent to NHP, and while data related to a Parkinsons disease model has been reported with 8p, this is the first disclosure of the optimization and discovery of VU0476406, as well as detailed pharmacology and DMPK properties.


ACS Medicinal Chemistry Letters | 2017

Design and Synthesis of mGlu2 NAMs with Improved Potency and CNS Penetration Based on a Truncated Picolinamide Core

Katrina A. Bollinger; Andrew S. Felts; Christopher J. Brassard; Julie L. Engers; Alice L. Rodriguez; Rebecca L. Weiner; Hyekyung P. Cho; Sichen Chang; Michael Bubser; Carrie K. Jones; Anna L. Blobaum; Colleen M. Niswender; P. Jeffrey Conn; Kyle A. Emmitte; Craig W. Lindsley

Herein, we detail the optimization of the mGlu2 negative allosteric modulator (NAM), VU6001192, by a reductionist approach to afford a novel, simplified mGlu2 NAM scaffold. This new chemotype not only affords potent and selective mGlu2 inhibition, as exemplified by VU6001966 (mGlu2 IC50 = 78 nM, mGlu3 IC50 > 30 μM), but also excellent central nervous system (CNS) penetration (Kp = 1.9, Kp,uu = 0.78), a feature devoid in all previously disclosed mGlu2 NAMs (Kps ≈ 0.3, Kp,uus ≈ 0.1). Moreover, this series, based on overall properties, represents an exciting lead series for potential mGlu2 PET tracer development.

Collaboration


Dive into the Sichen Chang'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

Atin Lamsal

Vanderbilt University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge