Network


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

Hotspot


Dive into the research topics where Clarke B. Taylor is active.

Publication


Featured researches published by Clarke B. Taylor.


Bioorganic & Medicinal Chemistry Letters | 2012

Structure based design of an in vivo active hydroxamic acid inhibitor of P. aeruginosa LpxC

Joseph Scott Warmus; Cheryl L. Quinn; Clarke B. Taylor; Sean T. Murphy; Timothy Allen Johnson; Chris Limberakis; Daniel F. Ortwine; Joel C. Bronstein; Paul J. Pagano; John D. Knafels; Sandra Lightle; Igor Mochalkin; Roger J. Brideau; Terry Podoll

Lipid A is an essential component of the Gram negative outer membrane, which protects the bacterium from attack of many antibiotics. The Lipid A biosynthesis pathway is essential for Gram negative bacterial growth and is unique to these bacteria. The first committed step in Lipid A biosynthesis is catalysis by LpxC, a zinc dependent deacetylase. We show the design of an LpxC inhibitor utilizing a robust model which directed efficient design of picomolar inhibitors. Analysis of physiochemical properties drove design to focus on an optimal lipophilicity profile. Further structure based design took advantage of a conserved water network over the active site, and with the optimal lipophilicity profile, led to an improved LpxC inhibitor with in vivo activity against wild type Pseudomonas aeruginosa.


Bioorganic & Medicinal Chemistry Letters | 2009

Discovery and SAR of benzyl phenyl ethers as inhibitors of bacterial phenylalanyl-tRNA synthetase.

Justin Ian Montgomery; Peter L. Toogood; Kim Marie Hutchings; Jia Liu; Lakshmi Narasimhan; Timothy Braden; Michael R. Dermyer; Angela D. Kulynych; Yvonne Smith; Joseph Scott Warmus; Clarke B. Taylor

A series of benzyl phenyl ethers (BPEs) is described that displays potent inhibition of bacterial phenylalanyl-tRNA synthetase. The synthesis, SAR, and select ADMET data are provided.


OncoImmunology | 2016

Synthetic RORγ agonists regulate multiple pathways to enhance antitumor immunity

Xiao Hu; Xikui Liu; Jacques Moisan; Yahong Wang; Charles Lesch; Chauncey Spooner; Rodney W. Morgan; Elizabeth Zawidzka; David Mertz; Dick Bousley; Kinga Majchrzak; Ilona Kryczek; Clarke B. Taylor; Chad A. Van Huis; Don Skalitzky; Alexander R. Hurd; Thomas D. Aicher; Peter L. Toogood; Gary D. Glick; Chrystal M. Paulos; Weiping Zou; Laura Carter

ABSTRACT RORγt is the key transcription factor controlling the development and function of CD4+ Th17 and CD8+ Tc17 cells. Across a range of human tumors, about 15% of the CD4+ T cell fraction in tumor-infiltrating lymphocytes are RORγ+ cells. To evaluate the role of RORγ in antitumor immunity, we have identified synthetic, small molecule agonists that selectively activate RORγ to a greater extent than the endogenous agonist desmosterol. These RORγ agonists enhance effector function of Type 17 cells by increasing the production of cytokines/chemokines such as IL-17A and GM-CSF, augmenting expression of co-stimulatory receptors like CD137, CD226, and improving survival and cytotoxic activity. RORγ agonists also attenuate immunosuppressive mechanisms by curtailing Treg formation, diminishing CD39 and CD73 expression, and decreasing levels of co-inhibitory receptors including PD-1 and TIGIT on tumor-reactive lymphocytes. The effects of RORγ agonists were not observed in RORγ−/− T cells, underscoring the selective on-target activity of the compounds. In vitro treatment of tumor-specific T cells with RORγ agonists, followed by adoptive transfer to tumor-bearing mice is highly effective at controlling tumor growth while improving T cell survival and maintaining enhanced IL-17A and reduced PD-1 in vivo. The in vitro effects of RORγ agonists translate into single agent, immune system-dependent, antitumor efficacy when compounds are administered orally in syngeneic tumor models. RORγ agonists integrate multiple antitumor mechanisms into a single therapeutic that both increases immune activation and decreases immune suppression resulting in robust inhibition of tumor growth. Thus, RORγ agonists represent a novel immunotherapy approach for cancer.


Cancer Research | 2016

Abstract 565: RORγ agonists regulate immune checkpoint receptors to enhance anti-tumor immunity

Xiao Hu; Xikui Liu; Jacques Moisan; Chrystal M. Paulos; Yahong Wang; Chauncey Spooner; Charles Lesch; Rodney W. Morgan; David Mertz; Dick Bousley; Clarke B. Taylor; Chad A. Van Huis; Don Skalitzky; Thomas D. Aicher; Peter L. Toogood; Laura Carter

As the master transcription factor for Type 17 T cells, RORγt activates a program of gene expression associated with enhancing effector function and overcoming immune suppression. Lycera is developing synthetic, small molecule RORγ agonists for immunotherapy of cancer. We have previously reported that RORγγ agonists increase type 17 cytokine and chemokine production, enhance cell survival and have single agent anti-tumor activity in syngeneic tumor models. Interestingly, transcriptional profiling and cytometry studies revealed that treatment of murine and human Th17 or Tc17 cells with RORγ agonists increases the expression of costimulatory receptors such as CD137 and decreased expression of coinhibitory receptors like PD-1. Given the importance of PD-1 in anti-tumor immunity, we further characterized the effects of RORγ agonist on this pathway. In vitro treatment with RORγ agonists significantly decreases mean fluorescent intensity and percent PD-1+ cells after resting and repetitive restimulation of Type 17 T cells. RORγ agonists do not modulate PD-1 expression in RORγ-/- T cells. In co-cultures of wild type and RORγ-/- T cells in the presence of the agonist, RORγ-/- T cells have reduced PD-1 expression suggesting that RORγ agonists induce a transmissible effect on RORγ-/- T cells. However, ChIP-seq data indicates that RORγt does not directly bind to the promoter or enhancer element of PD-1. Transcriptional and epigenetic profiling experiments have identified several pathways modulated by RORγ agonists that may regulate PD-1 expression. Reduced PD-1 expression following RORγ agonist treatment has a functional impact as treated cells also resist PD-L1-mediated inhibition of cytokine production and proliferation. Importantly, the decreased PD-1 expression observed after in vitro treatment with RORγ agonists is maintained following adoptive transfer of tumor specific T cells. These cells are highly effective at controlling tumor growth without further agonist treatment in vivo, suggesting that in vitro RORγ agonist treatment results in durable epigenetic changes. In summary, RORγ agonists have been shown to decrease checkpoint receptor expression while enhancing cytokine production and promoting long term survival and self-renewal of T cells. These results provide rationales for combining an RORγ agonist with checkpoint inhibitor such as anti-CTLA4 or anti-PD-1. By integrating effects on different effector pathways, RORγ agonists represent a promising immunotherapy approach for the treatment of cancer. Citation Format: Xiao Hu, Xikui Liu, Jacques Moisan, Chrystal Paulos, Yahong Wang, Chauncey Spooner, Charles Lesch, Rodney Morgan, David Mertz, Dick Bousley, Clarke Taylor, Chad Van Huis, Don Skalitzky, Thomas Aicher, Peter Toogood, Laura Carter. RORγ agonists regulate immune checkpoint receptors to enhance anti-tumor immunity. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 565.


Journal for ImmunoTherapy of Cancer | 2015

RORgamma agonists enhance survival and memory of type 17 T cells and improve anti-tumor activity

Xiao Hu; Jacques Moisan; Kinga Majchrzak; Chuck A. Lesch; Yahong Wang; Brian Sanchez; Xikui Liu; Rodney Morgan; David Mertz; Dick Bousley; Chad A. Van Huis; Don Skalitzky; Clarke B. Taylor; Thomas D. Aicher; Peter L. Toogood; Weiping Zou; Gary D. Glick; Chrystal M. Paulos; Laura Carter

Meeting abstracts Enhancing tumor-directed immune responses has emerged as an important therapeutic approach to many cancers. Th17/Tc17 cells can mediate robust anti-tumor responses in rodent models and are associated with improved prognosis in some human cancers. RORgt is the key transcription


Bioorganic & Medicinal Chemistry Letters | 2007

The synthesis and biological evaluation of novel series of nitrile-containing fluoroquinolones as antibacterial agents

Sean T. Murphy; Heather L. Case; Edmund L. Ellsworth; Susan Elizabeth Hagen; Michael D. Huband; Themis Joannides; Chris Limberakis; Keith R. Marotti; Amy M. Ottolini; Mark R. Rauckhorst; Jeremy T. Starr; Michael Andrew Stier; Clarke B. Taylor; Tong Zhu; Adrian Blaser; William A. Denny; Guo Liang Lu; Jeff B. Smaill; Freddy Rivault


Archive | 2009

Hydroxamic acid derivatives useful as antibacterial agents

Matthew Frank Brown; Charles Francis Donovan; Edmund L. Ellsworth; Denton Wade Hoyer; Timothy Allen Johnson; Manjinder S. Lall; Chris Limberakis; Sean T. Murphy; Debra Ann Sherry; Clarke B. Taylor; Joseph Scott Warmus


Journal of Organic Chemistry | 2012

Stereoselective synthesis of (S)-3-(methylamino)-3-((R)-pyrrolidin-3-yl)propanenitrile.

Manjinder S. Lall; Garrett Hoge; Tuan P. Tran; William Kissel; Sean Timothy Murphy; Clarke B. Taylor; Kim Marie Hutchings; Brian Samas; Edmund L. Ellsworth; Timothy Curran; H. D. Hollis Showalter


Archive | 2011

Pyridonyl guanidine F1F0-ATPase inhibitors and therapeutic uses thereof

Gary D. Glick; Alexander R. Hurd; Matthew N. Mattson; Clarke B. Taylor; Chad A. Van Huis


Archive | 2013

Heterocyclic guanidine f1f0-atpase inhibitors

Alexander R. Hurd; Clarke B. Taylor; Peter L. Toogood

Collaboration


Dive into the Clarke B. Taylor's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xikui Liu

University of Michigan

View shared research outputs
Top Co-Authors

Avatar

Chrystal M. Paulos

Medical University of South Carolina

View shared research outputs
Researchain Logo
Decentralizing Knowledge