Network


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

Hotspot


Dive into the research topics where Scott Throner is active.

Publication


Featured researches published by Scott Throner.


Bioorganic & Medicinal Chemistry Letters | 2011

Discovery of novel positive allosteric modulators of the metabotropic glutamate receptor 5 (mGlu5)

Jeffrey G. Varnes; Andrew P. Marcus; Russell C. Mauger; Scott Throner; Valerie Hoesch; Megan M. King; Xia Wang; Linda A. Sygowski; Nathan Spear; Reto Gadient; Dean G. Brown; James B. Campbell

Novel in vitro mGlu(5) positive allosteric modulators with good potency, solubility, and low lipophilicity are described. Compounds were identified which did not rely on the phenylacetylene and carbonyl functionalities previously observed to be required for in vitro activity. Investigation of the allosteric binding requirements of a series of dihydroquinolinone analogs led to phenylacetylene azachromanone 4 (EC(50) 11.5 nM). Because of risks associated with potential metabolic and toxicological liabilities of the phenylacetylene, this moiety was successfully replaced with a phenoxymethyl group (27; EC(50) 156.3 nM). Derivation of a second-generation of mGlu(5) PAMs lacking a ketone carbonyl resulted in azaindoline (33), azabenzimidazole (36), and N-methyl 8-azaoxazine (39) phenylacetylenes. By scoping nitrogen substituents and phenylacetylene replacements in 39, we identified phenoxymethyl 8-azaoxazine 47 (EC(50) 50.1 nM) as a potent and soluble mGlu(5) PAM devoid of both undesirable phenylacetylene and carbonyl functionalities.


Bioorganic & Medicinal Chemistry Letters | 2010

Discovery of 8-azabicyclo[3.2.1]octan-3-yloxy-benzamides as selective antagonists of the kappa opioid receptor. Part 1

Todd Andrew Brugel; Reed W. Smith; Michael Balestra; Christopher Becker; Thalia Daniels; Tiffany N. Hoerter; Gerard M. Koether; Scott Throner; Laura M. Panko; James Folmer; Joseph Cacciola; Angela M. Hunter; Ruifeng Liu; Philip D. Edwards; Dean G. Brown; John C. Gordon; Norman C. Ledonne; Mark R. Pietras; Patricia Schroeder; Linda A. Sygowski; Lee T. Hirata; Anna Zacco; Matthew F. Peters

Initial high throughput screening efforts identified highly potent and selective kappa opioid receptor antagonist 3 (κ IC(50)=77 nM; μ:κ and δ:κ IC(50) ratios>400) which lacked CNS exposure in vivo. Modification of this scaffold resulted in development of a series of 8-azabicyclo[3.2.1]octan-3-yloxy-benzamides showing potent and selectivity κ antagonism as well as good brain exposure. Analog 6c (κ IC(50)=20 nM; μ:κ=36, δ:κ=415) was also shown to reverse κ-agonist induced rat diuresis in vivo.


Bioorganic & Medicinal Chemistry Letters | 2010

SAR development of a series of 8-azabicyclo[3.2.1]octan-3-yloxy-benzamides as kappa opioid receptor antagonists. Part 2

Todd Andrew Brugel; Reed W. Smith; Michael Balestra; Christopher Becker; Thalia Daniels; Gerard M. Koether; Scott Throner; Laura M. Panko; Dean G. Brown; Ruifeng Liu; John C. Gordon; Matthew F. Peters

Further structure activity relationship studies on a previously reported 8-azabicyclo[3.2.1]octan-3-yloxy-benzamide series of potent and selective kappa opioid receptor antagonists is discussed. Modification of the pendant N-substitution to include a cyclohexylurea moiety produced analogs with greater in vitro opioid and hERG selectivity such as 12 (kappa IC50=172 nM, mu:kappa ratio=93, delta:kappa ratio=>174, hERG IC50=>33 microM). Changes to the linker conformation and identity as well as to the benzamide ring moiety were also investigated.


Bioorganic & Medicinal Chemistry Letters | 2015

Discovery of AZ0108, an orally bioavailable phthalazinone PARP inhibitor that blocks centrosome clustering

Jeffrey W. Johannes; Lynsie Almeida; Kevin Daly; Andrew D. Ferguson; Shaun Grosskurth; Huiping Guan; Tina Howard; Stephanos Ioannidis; Steven Kazmirski; Michelle Lamb; Nicholas A. Larsen; Paul Lyne; Keith Mikule; Claude Ogoe; Bo Peng; Philip Petteruti; Jon Read; Nancy Su; Mark Sylvester; Scott Throner; Wenxian Wang; Xin Wang; Jiaquan Wu; Qing Ye; Yan Yu; Xiaolan Zheng; David Scott

The propensity for cancer cells to accumulate additional centrosomes relative to normal cells could be exploited for therapeutic benefit in oncology. Following literature reports that suggested TNKS1 (tankyrase 1) and PARP16 may be involved with spindle structure and function and may play a role in suppressing multi-polar spindle formation in cells with supernumerary centrosomes, we initiated a phenotypic screen to look for small molecule poly (ADP-ribose) polymerase (PARP) enzyme family inhibitors that could produce a multi-polar spindle phenotype via declustering of centrosomes. Screening of AstraZenecas collection of phthalazinone PARP inhibitors in HeLa cells using high-content screening techniques identified several compounds that produced a multi-polar spindle phenotype at low nanomolar concentrations. Characterization of these compounds across a broad panel of PARP family enzyme assays indicated that they had activity against several PARP family enzymes, including PARP1, 2, 3, 5a, 5b, and 6. Further optimization of these initial hits for improved declustering potency, solubility, permeability, and oral bioavailability resulted in AZ0108, a PARP1, 2, 6 inhibitor that potently inhibits centrosome clustering and is suitable for in vivo efficacy and tolerability studies.


Journal of Medicinal Chemistry | 2014

Discovery of spirofused piperazine and diazepane amides as selective histamine-3 antagonists with in vivo efficacy in a mouse model of cognition.

Dean G. Brown; Peter R. Bernstein; Andrew Griffin; Steve Wesolowski; Denis Labrecque; Maxime C. Tremblay; Mark Sylvester; Russell C. Mauger; Phillip D. Edwards; Scott Throner; James Folmer; Joseph Cacciola; Clay W Scott; Lois Ann Lazor; Mehrnaz Pourashraf; V. Santhakumar; William Potts; Simon Sydserff; Pascall Giguère; Carine Lévesque; Mohammed Dasser; Thierry Groblewski

A new series of potent and selective histamine-3 receptor (H3R) antagonists was identified on the basis of an azaspiro[2.5]octane carboxamide scaffold. Many scaffold modifications were largely tolerated, resulting in nanomolar-potent compounds in the H3R functional assay. Exemplar compound 6s demonstrated a selective profile against a panel of 144 secondary pharmacological receptors, with activity at only σ2 (62% at 10 μM). Compound 6s demonstrated free-plasma exposures above the IC50 (∼50×) with a brain-to-plasma ratio of ∼3 following intravenous dosing in mice. At three doses tested in the mouse novel object recognition model (1, 3, and 10 mg/kg s.c.), 6s demonstrated a statistically significant response compared with the control group. This series represents a new scaffold of H3 receptor antagonists that demonstrates in vivo exposure and efficacy in an animal model of cognition.


ACS Medicinal Chemistry Letters | 2013

Azepines and piperidines with dual norepinephrine dopamine uptake inhibition and antidepressant activity.

Dean G. Brown; Peter R. Bernstein; Ye Wu; Rebecca Urbanek; Christopher Becker; Scott Throner; Bruce T. Dembofsky; Gary Steelman; Lois Ann Lazor; Clay W Scott; Michael W. Wood; Steven Wesolowski; David A. Nugiel; Stephanie Koch; Jian Yu; Donald E. Pivonka; Shuang Li; Carol Thompson; Anna Zacco; Charles S. Elmore; Patricia Schroeder; Jianwei Liu; Christopher Hurley; Stuart Ward; Hazel J. Hunt; Karen Williams; Joseph McLaughlin; Valerie Hoesch; Simon Sydserff; Donna L. Maier

Herein, we describe the discovery of inhibitors of norepinephrine (NET) and dopamine (DAT) transporters with reduced activity relative to serotonin transporters (SERT). Two compounds, 8b and 21a, along with nomifensine were tested in a rodent receptor occupancy study and demonstrated dose-dependent displacement of radiolabeled NET and DAT ligands. These compounds were efficacious in a rat forced swim assay (model of depression) and also had activity in rat spontaneous locomotion assay.


ACS Chemical Neuroscience | 2016

Discovery and Preclinical Validation of [11C]AZ13153556, a Novel Probe for the Histamine Type 3 Receptor

Magnus Schou; Katarina Varnäs; Anders Juréus; Charlotte Ahlgren; Jonas Malmquist; Jenny Häggkvist; Lenke Tari; Steven Wesolowski; Scott Throner; Dean G. Brown; Maria Nilsson; Peter Johnström; Sjoerd J. Finnema; Ryuji Nakao; Nahid Amini; Akihiro Takano; Lars Farde

UNLABELLED The histamine type 3 receptor (H3) is a G protein-coupled receptor implicated in several disorders of the central nervous system. Herein, we describe the radiolabeling and preclinical evaluation of a candidate radioligand for the H3 receptor, 4-(1S,2S)-2-(4-cyclobutylpiperazine-1-carbonyl)cyclopropyl]-N-methyl-benzamide (5), and its comparison with one of the frontrunner radioligands for H3 imaging, namely, GSK189254 (1). Compounds 1 and 5 were radiolabeled with tritium and carbon-11 for in vitro and in vivo imaging experiments. The in vitro binding of [(3)H]1 and [(3)H]5 was examined by (i) saturation binding to rat and nonhuman primate brain tissue homogenate and (ii) in vitro autoradiography on tissue sections from rat, guinea pig, and human brain. The in vivo binding of [(11)C]1 and [(11)C]5 was examined by PET imaging in mice and nonhuman primates. Bmax values obtained from Scatchard analysis of [(3)H]1 and [(3)H]5 binding were in good agreement. Autoradiography with [(3)H]5 on rat, guinea pig, and human brain slices showed specific binding in regions known to be enhanced in H3 receptors, a high degree of colocalization with [(3)H]1, and virtually negligible nonspecific binding in tissue. PET measurements in mice and nonhuman primates demonstrated that [(11)C]5 binds specifically and reversibly to H3 receptors in vivo with low nonspecific binding in brain tissue. Whereas [(11)C]1 showed similar binding characteristics in vivo, the binding kinetics appeared faster for [(11)C]5 than for [(11)C]1. CONCLUSIONS [(11)C]5 has suitable properties for quantification of H3 receptors in nonhuman primate brain and has the potential to offer improved binding kinetics in man compared to [(11)C]1.


Journal of Medicinal Chemistry | 2018

Discovery and Optimization of a Novel Series of Highly Selective JAK1 Kinase Inhibitors

Neil Grimster; Erica Anderson; Marat Alimzhanov; Geraldine A. Bebernitz; Kirsten Bell; Claudio Chuaqui; Tracy L. Deegan; Andrew D. Ferguson; Thomas Gero; Andreas Harsch; Dennis Huszar; Aarti Kawatkar; Jason Grant Kettle; Paul Lyne; Jon Read; Caroline Rivard Costa; Linette Ruston; Patricia Schroeder; Jie Shi; Qibin Su; Scott Throner; Dorin Toader; Melissa Vasbinder; Richard Woessner; Haixia Wang; Allan Wu; Minwei Ye; Weijia Zheng; Michael Zinda

Janus kinases (JAKs) have been demonstrated to be critical in cytokine signaling and have thus been implicated in both cancer and inflammatory diseases. The JAK family consists of four highly homologous members: JAK1-3 and TYK2. The development of small-molecule inhibitors that are selective for a specific family member would represent highly desirable tools for deconvoluting the intricacies of JAK family biology. Herein, we report the discovery of a potent JAK1 inhibitor, 24, which displays ∼1000-fold selectivity over the other highly homologous JAK family members (determined by biochemical assays), while also possessing good selectivity over other kinases (determined by panel screening). Moreover, this compound was demonstrated to be orally bioavailable and possesses acceptable pharmacokinetic parameters. In an in vivo study, the compound was observed to dose dependently modulate the phosphorylation of STAT3 (a downstream marker of JAK1 inhibition).


Cancer Research | 2017

Abstract 979: Discovery of the JAK1 selective kinase inhibitor AZD4205

Jason Grant Kettle; Qibin Su; Neil Grimster; Sameer Kawatkar; Scott Throner; Richard Woessner; Huawei Chen; Geraldine A. Bebernitz; Kristen Bell; Erica Anderson; Linette Ruston; Jon Winter-Holt; Paul Lyne; Melissa Vasbinder; Claudio Chuaqui

Janus kinases are a family of four enzymes; JAK1, JAK2, JAK3 and tyrosine kinase 2 (TYK2) that are critical in cytokine signalling, with constitutive activation of JAK/STAT pathways associated with a wide variety of malignancies. Elevated JAK/STAT signalling leading to increased activation of STAT3 is reported in a wide variety of cancers, including breast, liver, prostate, colorectal, head and neck, oesophageal, pancreatic, bladder, and non-small cell lung, and is implicated in the pathogenesis of diffuse large B-cell lymphoma and nasopharyngeal carcinomas. Overall, up to 70% of human tumours are linked to persistent elevated STAT3 activity which can be associated with poorer prognosis in many of these settings. In addition, elevated pSTAT3 is observed in response to chemotherapy treatment, and also in response to treatment with inhibitors of oncogenic signalling pathways such as EGFR, MAPK and AKT, and is associated with resistance or poorer response to agents targeting these pathways. In many of these cases, JAK1 is believed to be a primary driver of STAT3 phosphorylation and signalling, suggesting inhibition of JAKs as a therapeutic approach to treat these potential resistance mechanisms. The mixed JAK1/2 kinase inhibitor ruxolitinib is approved for the treatment of myeloproliferative neoplasms including intermediate or high risk myelofibrosis and polycythemia vera and has been tested in a variety of tumor settings. Since JAK2 is essential for the signal transduction downstream of erythropoietin, thrombopoietin and related receptors that control erythrocyte and megakaryocyte expansion, dosing of inhibitors that target JAK2 can be limited by toxicities such as thrombocytopenia and anaemia. Starting from a non-kinome selective screening hit, structure-based design was used to optimise a series of aminopyrimidines that led to JAK1-selective candidate drug AZD4205. This compound demonstrates ATP competitive binding with IC50’s in a high ATP concentration enzyme assay against JAK1 of 73 nM (Ki = 2.8 nM), with high selectivity against JAK2 and JAK3 with IC50’s of 13,233 nM and >30,000 nM respectively. In addition it showed potent inhibition of p-STAT3 in a cell based assay of JAK1 activity with an IC50 of 128 nM and excellent selectivity across the kinome. In summary, AZD4205 is a highly potent JAK1-selective kinase inhibitor with excellent preclinical pharmacokinetics with potential for further clinical development. The optimization from screening hit to first disclosure of this candidate drug will be presented. Citation Format: Jason G. Kettle, Qibin Su, Neil Grimster, Sameer Kawatkar, Scott Throner, Richard Woessner, Huawei Chen, Geraldine Bebernitz, Kristen Bell, Erica Anderson, Linette Ruston, Jon Winter-Holt, Paul Lyne, Melissa Vasbinder, Claudio Chuaqui. Discovery of the JAK1 selective kinase inhibitor AZD4205 [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 979. doi:10.1158/1538-7445.AM2017-979


Archive | 2005

Substituted Amino-Pyrimidones and Uses Thereof

Jeffrey S. Albert; Don Andisik; James Arnold; Dean G. Brown; Owen Callaghan; James B. Campbell; Robin Arthur Ellis Carr; Gianni Chessari; Miles Stuart Congreve; Phil Edwards; James Empfield; Martyn Frederickson; Gerard M. Koether; Jennifer R. Krumrine; Russ Mauger; Christopher William Murray; Sahil Joe Patel; Mark Sylvester; Scott Throner

Collaboration


Dive into the Scott Throner'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
Researchain Logo
Decentralizing Knowledge