Network


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

Hotspot


Dive into the research topics where Simon Bushell is active.

Publication


Featured researches published by Simon Bushell.


Journal of Virology | 2011

Inhibition of Dengue Virus through Suppression of Host Pyrimidine Biosynthesis

Qing Yin Wang; Simon Bushell; Min Qing; Hao Ying Xu; Aurelio Bonavia; Sandra Nunes; Jing Zhou; Mee Kian Poh; Paola Florez de Sessions; Pornwaratt Niyomrattanakit; Hongping Dong; Keith Hoffmaster; Anne Goh; Shahul Nilar; Wouter Schul; Susan A. Jones; Laura D. Kramer; Teresa Compton; Pei Yong Shi

ABSTRACT Viral replication relies on the host to supply nucleosides. Host enzymes involved in nucleoside biosynthesis are potential targets for antiviral development. Ribavirin (a known antiviral drug) is such an inhibitor that suppresses guanine biosynthesis; depletion of the intracellular GTP pool was shown to be the major mechanism to inhibit flavivirus. Along similar lines, inhibitors of the pyrimidine biosynthesis pathway could be targeted for potential antiviral development. Here we report on a novel antiviral compound (NITD-982) that inhibits host dihydroorotate dehydrogenase (DHODH), an enzyme required for pyrimidine biosynthesis. The inhibitor was identified through screening 1.8 million compounds using a dengue virus (DENV) infection assay. The compound contains an isoxazole-pyrazole core structure, and it inhibited DENV with a 50% effective concentration (EC50) of 2.4 nM and a 50% cytotoxic concentration (CC50) of >5 μM. NITD-982 has a broad antiviral spectrum, inhibiting both flaviviruses and nonflaviviruses with nanomolar EC90s. We also show that (i) the compound inhibited the enzymatic activity of recombinant DHODH, (ii) an NITD-982 analogue directly bound to the DHODH protein, (iii) supplementing the culture medium with uridine reversed the compound-mediated antiviral activity, and (iv) DENV type 2 (DENV-2) variants resistant to brequinar (a known DHODH inhibitor) were cross resistant to NITD-982. Collectively, the results demonstrate that the compound inhibits DENV through depleting the intracellular pyrimidine pool. In contrast to the in vitro potency, the compound did not show any efficacy in the DENV-AG129 mouse model. The lack of in vivo efficacy is likely due to the exogenous uptake of pyrimidine from the diet or to a high plasma protein-binding activity of the current compound.


Journal of Medicinal Chemistry | 2012

Discovery of LFF571: An Investigational Agent for Clostridium difficile Infection

Matthew J. LaMarche; Jennifer A. Leeds; Adam Amaral; Jason T. Brewer; Simon Bushell; Gejing Deng; Janetta M. Dewhurst; Jian Ding; JoAnne Dzink-Fox; Gabriel G. Gamber; Akash K. Jain; Kwangho Lee; Lac Lee; Troy Lister; David McKenney; Steve Mullin; Colin P. Osborne; Deborah Palestrant; Michael A. Patane; Elin M. Rann; Meena Sachdeva; Jian Shao; Stacey Tiamfook; Anna Trzasko; Lewis Whitehead; Aregahegn Yifru; Donghui Yu; Wanlin Yan; Qingming Zhu

Clostridium difficile (C. difficile) is a Gram positive, anaerobic bacterium that infects the lumen of the large intestine and produces toxins. This results in a range of syndromes from mild diarrhea to severe toxic megacolon and death. Alarmingly, the prevalence and severity of C. difficile infection are increasing; thus, associated morbidity and mortality rates are rising. 4-Aminothiazolyl analogues of the antibiotic natural product GE2270 A (1) were designed, synthesized, and optimized for the treatment of C. difficile infection. The medicinal chemistry effort focused on enhancing aqueous solubility relative to that of the natural product and previous development candidates (2, 3) and improving antibacterial activity. Structure-activity relationships, cocrystallographic interactions, pharmacokinetics, and efficacy in animal models of infection were characterized. These studies identified a series of dicarboxylic acid derivatives, which enhanced solubility/efficacy profile by several orders of magnitude compared to previously studied compounds and led to the selection of LFF571 (4) as an investigational new drug for treating C. difficile infection.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Identification of broad-spectrum antiviral compounds and assessment of the druggability of their target for efficacy against respiratory syncytial virus (RSV)

Aurelio Bonavia; Michael Franti; Erin P. Keaney; Kelli Kuhen; Mohindra Seepersaud; Branko Radetich; Jian Shao; Ayako Honda; Janetta M. Dewhurst; Kara Balabanis; James Monroe; Karen Wolff; Colin P. Osborne; Leanne Lanieri; Keith Hoffmaster; Jakal Amin; Judit Markovits; Michelle Broome; Elizabeth Skuba; Ivan Cornella-Taracido; Gerard Joberty; Tewis Bouwmeester; Lawrence G. Hamann; John A. Tallarico; Ruben Tommasi; Teresa Compton; Simon Bushell

The search for novel therapeutic interventions for viral disease is a challenging pursuit, hallmarked by the paucity of antiviral agents currently prescribed. Targeting of viral proteins has the inextricable challenge of rise of resistance. Safe and effective vaccines are not possible for many viral pathogens. New approaches are required to address the unmet medical need in this area. We undertook a cell-based high-throughput screen to identify leads for development of drugs to treat respiratory syncytial virus (RSV), a serious pediatric pathogen. We identified compounds that are potent (nanomolar) inhibitors of RSV in vitro in HEp-2 cells and in primary human bronchial epithelial cells and were shown to act postentry. Interestingly, two scaffolds exhibited broad-spectrum activity among multiple RNA viruses. Using the chemical matter as a probe, we identified the targets and identified a common cellular pathway: the de novo pyrimidine biosynthesis pathway. Both targets were validated in vitro and showed no significant cell cytotoxicity except for activity against proliferative B- and T-type lymphoid cells. Corollary to this finding was to understand the consequences of inhibition of the target to the host. An in vivo assessment for antiviral efficacy failed to demonstrate reduced viral load, but revealed microscopic changes and a trend toward reduced pyrimidine pools and findings in histopathology. We present here a discovery program that includes screen, target identification, validation, and druggability that can be broadly applied to identify and interrogate other host factors for antiviral effect starting from chemical matter of unknown target/mechanism of action.


PLOS ONE | 2015

Englerin A Agonizes the TRPC4/C5 Cation Channels to Inhibit Tumor Cell Line Proliferation.

Cheryl Carson; Pichai Raman; Jennifer Tullai; Lei Xu; Martin Henault; Emily Thomas; Sarita Yeola; Jianmin Lao; Mark McPate; J. Martin Verkuyl; George Marsh; Jason Sarber; Adam Amaral; Scott Bailey; Danuta Lubicka; Helen Pham; Nicolette Miranda; Jian Ding; Hai-Ming Tang; Haisong Ju; Pamela Tranter; Nan Ji; Philipp Krastel; Rishi K. Jain; Andrew M. Schumacher; Joseph Loureiro; Elizabeth George; Giuliano Berellini; Nathan T. Ross; Simon Bushell

Englerin A is a structurally unique natural product reported to selectively inhibit growth of renal cell carcinoma cell lines. A large scale phenotypic cell profiling experiment (CLiP) of englerin A on ¬over 500 well characterized cancer cell lines showed that englerin A inhibits growth of a subset of tumor cell lines from many lineages, not just renal cell carcinomas. Expression of the TRPC4 cation channel was the cell line feature that best correlated with sensitivity to englerin A, suggesting the hypothesis that TRPC4 is the efficacy target for englerin A. Genetic experiments demonstrate that TRPC4 expression is both necessary and sufficient for englerin A induced growth inhibition. Englerin A induces calcium influx and membrane depolarization in cells expressing high levels of TRPC4 or its close ortholog TRPC5. Electrophysiology experiments confirmed that englerin A is a TRPC4 agonist. Both the englerin A induced current and the englerin A induced growth inhibition can be blocked by the TRPC4/C5 inhibitor ML204. These experiments confirm that activation of TRPC4/C5 channels inhibits tumor cell line proliferation and confirms the TRPC4 target hypothesis generated by the cell line profiling. In selectivity assays englerin A weakly inhibits TRPA1, TRPV3/V4, and TRPM8 which suggests that englerin A may bind a common feature of TRP ion channels. In vivo experiments show that englerin A is lethal in rodents near doses needed to activate the TRPC4 channel. This toxicity suggests that englerin A itself is probably unsuitable for further drug development. However, since englerin A can be synthesized in the laboratory, it may be a useful chemical starting point to identify novel modulators of other TRP family channels.


Journal of Medicinal Chemistry | 2011

Antibacterial Optimization of 4-Aminothiazolyl Analogues of the Natural Product GE2270 A: Identification of the Cycloalkylcarboxylic Acids

Matthew J. LaMarche; Jennifer A. Leeds; Kerri Amaral; Jason T. Brewer; Simon Bushell; Janetta M. Dewhurst; JoAnne Dzink-Fox; Eric Gangl; Julie Goldovitz; Akash K. Jain; Steve Mullin; Georg Neckermann; Colin P. Osborne; Deborah Palestrant; Michael A. Patane; Elin M. Rann; Meena Sachdeva; Jian Shao; Stacey Tiamfook; Lewis Whitehead; Donghui Yu

4-Aminothiazolyl analogues of the antibiotic natural product GE2270 A (1) were designed, synthesized, and optimized for their activity against Gram positive bacterial infections. Optimization efforts focused on improving the physicochemical properties (e.g., aqueous solubility and chemical stability) of the 4-aminothiazolyl natural product template while improving the in vitro and in vivo antibacterial activity. Structure-activity relationships were defined, and the solubility and efficacy profiles were improved over those of previous analogues and 1. These studies identified novel, potent, soluble, and efficacious elongation factor-Tu inhibitors, which bear cycloalkylcarboxylic acid side chains, and culminated in the selection of development candidates amide 48 and urethane 58.


Archive | 2008

Aminothiazoles and their Uses

Simon Bushell; Matthew J. LaMarche; Jennifer A. Leeds; Lewis Whitehead


Archive | 2006

N - formyl hydroxylamine compounds as pdf inhibitors

Kathryn Rene Bracken; Simon Bushell; Karl Dean; Charles Francavilla; Rakesh K. Jain; Kwangho Lee; Mohindra Seepersaud; Lei Shu; Arathia Sundaram; Zhengyu Yuan


Proceedings of the National Academy of Sciences of the United States of America | 2011

Organic Synthesis Toward Small-Molecule Probes and Drugs Special Feature: Identification of broad-spectrum antiviral compounds and assessment of the druggability of their target for efficacy against respiratory syncytial virus (RSV)

Aurelio Bonavia; Michael Franti; E. Pusateri Keaney; Kelli Kuhen; Mohindra Seepersaud; Branko Radetich; Jian Shao; Akio Honda; John K. Dewhurst; Kara Balabanis; Jocelyn Rebecca Monroe; Karl Erich Wolff; Colin P. Osborne; Leanne Lanieri; Keith Hoffmaster; J. Amin; Judit Markovits; Matthew R. Broome; Elizabeth Skuba; Ivan Cornella-Taracido; G. Joberty; Tewis Bouwmeester; Lawrence G. Hamann; John A. Tallarico; Ruben Tommasi; Todd Compton; Simon Bushell


Archive | 2013

AMINIOTHIAZOLES AND THEIR USES

Simon Bushell; Matthew J. LaMarche; Jennifer A. Leeds; Lewis Whitehead


Archive | 2007

EFTU INHIBITORS OR AMINOTHIAZOLES AND THEIR USES

Simon Bushell; Matthew J. LaMarche; Michael J. Patane; Lewis Whitehead

Collaboration


Dive into the Simon Bushell's collaboration.

Researchain Logo
Decentralizing Knowledge