Network


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

Hotspot


Dive into the research topics where Lewis Whitehead is active.

Publication


Featured researches published by Lewis Whitehead.


Bioorganic & Medicinal Chemistry | 2011

Human HDAC isoform selectivity achieved via exploitation of the acetate release channel with structurally unique small molecule inhibitors.

Lewis Whitehead; Markus Dobler; Branko Radetich; Peter Atadja; Tavina Claiborne; Jonathan E. Grob; Andrew McRiner; Margaret R. Pancost; Anup Patnaik; Wenlin Shao; Michael Shultz; Ritesh Tichkule; Ruben Tommasi; Brian Edward Vash; Ping Wang; Travis Stams

Herein we report the discovery of a family of novel yet simple, amino-acid derived class I HDAC inhibitors that demonstrate isoform selectivity via access to the internal acetate release channel. Isoform selectivity criteria is discussed on the basis of X-ray crystallography and molecular modeling of these novel inhibitors bound to HDAC8, potentially revealing insights into the mechanism of enzymatic function through novel structural features revealed at the atomic level.


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.


Journal of Medicinal Chemistry | 2011

Optimization of the in Vitro Cardiac Safety of Hydroxamate-Based Histone Deacetylase Inhibitors

Michael Shultz; Xueying Cao; Christine Hiu-Tung Chen; Young Shin Cho; Nicole R. Davis; Joe Eckman; Jianmei Fan; Alex Fekete; Brant Firestone; Julie Flynn; Jack Green; Joseph D. Growney; Mats Holmqvist; Meier Hsu; Daniel Jansson; Lei Jiang; Paul Kwon; Gang Liu; Franco Lombardo; Qiang Lu; Dyuti Majumdar; Christopher Meta; Lawrence Blas Perez; Minying Pu; Tim Ramsey; Stacy W. Remiszewski; Suzanne Skolnik; Martin Traebert; Laszlo Urban; Vinita Uttamsingh

Histone deacetylase (HDAC) inhibitors have shown promise in treating various forms of cancer. However, many HDAC inhibitors from diverse structural classes have been associated with QT prolongation in humans. Inhibition of the human ether a-go-go related gene (hERG) channel has been associated with QT prolongation and fatal arrhythmias. To determine if the observed cardiac effects of HDAC inhibitors in humans is due to hERG blockade, a highly potent HDAC inhibitor devoid of hERG activity was required. Starting with dacinostat (LAQ824), a highly potent HDAC inhibitor, we explored the SAR to determine the pharmacophores required for HDAC and hERG inhibition. We disclose here the results of these efforts where a high degree of pharmacophore homology between these two targets was discovered. This similarity prevented traditional strategies for mitigating hERG binding/modulation from being successful and novel approaches for reducing hERG inhibition were required. Using a hERG homology model, two compounds, 11r and 25i, were discovered to be highly efficacious with weak affinity for the hERG and other ion channels.


Journal of Chemical Information and Modeling | 2013

Automated ligand- and structure-based protocol for in silico prediction of human serum albumin binding.

Michelle Lynn Hall; William L. Jorgensen; Lewis Whitehead

Plasma protein binding has a profound impact on the pharmacokinetic and pharmacodynamic properties of many drug candidates and is thus an integral component of drug discovery. Nevertheless, extant methods to examine small-molecule interactions with plasma protein have various limitations, thus creating a need for alternative methods. Herein we present a comprehensive and cross-validated in silico workflow for the prediction of small-molecule binding to Human Serum Albumin (HSA), the most ubiquitous plasma protein. This protocol reliably predicts small-molecule interactions with HSA, including a binding affinity calculation using multiple linear regression methods, binding site prediction using a naive-Bayes classifier, and a three-dimensional binding pose using induced fit docking. Furthermore, this workflow is implemented in a portable and automated format that can be downloaded and used by other end users, either as is or with customization.


Bioorganic & Medicinal Chemistry Letters | 2012

Development of isoform selective PI3-kinase inhibitors as pharmacological tools for elucidating the PI3K pathway

Ian Bruce; Mohammed Akhlaq; Graham Charles Bloomfield; Emma Budd; Brian Cox; Bernard Cuenoud; Peter Finan; Peter Gedeck; Julia Hatto; Judy Hayler; Denise Head; Thomas H. Keller; Louise Kirman; Catherine Leblanc; Darren Le Grand; Clive Mccarthy; Desmond O’Connor; Charles Owen; Mrinalini Sachin Oza; Gaynor Elizabeth Pilgrim; Nicola Press; Lilya Sviridenko; Lewis Whitehead

Using a parallel synthesis approach to target a non-conserved region of the PI3K catalytic domain a pan-PI3K inhibitor 1 was elaborated to provide alpha, delta and gamma isoform selective Class I PI3K inhibitors 21, 24, 26 and 27. The compounds had good cellular activity and were selective against protein kinases and other members of the PI3K superfamily including mTOR and DNA-PK.


Journal of Medicinal Chemistry | 2010

Conformational Refinement of Hydroxamate-Based Histone Deacetylase Inhibitors and Exploration of 3-Piperidin-3-ylindole Analogues of Dacinostat (LAQ824)

Young Shin Cho; Lewis Whitehead; Jianke Li; Christine Hiu-Tung Chen; Lei Jiang; Markus Vögtle; Eric Francotte; Paul Richert; Trixie Wagner; Martin Traebert; Qiang Lu; Xueying Cao; Berengere Dumotier; Jasna Fejzo; Srinivasan Rajan; Ping Wang; Yan Yan-Neale; Wenlin Shao; Peter Atadja; Michael Shultz

Inspired by natural product HDAC inhibitors, we prepared a series of conformationally restrained HDAC inhibitors based on the hydroxamic acid dacinostat (LAQ824, 7). Several scaffolds with improved biochemical and cellular potency, as well as attenuated hERG inhibition, were identified, suggesting that the introduction of molecular rigidity is a viable strategy to enhance HDAC binding and mitigate hERG liability. Further SAR studies around a 3-piperidin-3-ylindole moiety resulted in the discovery of compound 30, for which a unique conformation was speculated to contribute to overcoming increased lipophilicity and attenuating hERG binding. Separation of racemate 30 afforded 32, the R enantiomer, which demonstrated improved potency in both enzyme and cellular assays compared to dacinostat.


Journal of Cheminformatics | 2011

An investigation into pharmaceutically relevant mutagenicity data and the influence on Ames predictive potential

Patrick McCarren; Clayton Springer; Lewis Whitehead

BackgroundIn drug discovery, a positive Ames test for bacterial mutation presents a significant hurdle to advancing a drug to clinical trials. In a previous paper, we discussed success in predicting the genotoxicity of reagent-sized aryl-amines (ArNH2), a structure frequently found in marketed drugs and in drug discovery, using quantum mechanics calculations of the energy required to generate the DNA-reactive nitrenium intermediate (ArNH:+). In this paper we approach the question of what molecular descriptors could improve these predictions and whether external data sets are appropriate for further training.ResultsIn trying to extend and improve this model beyond this quantum mechanical reaction energy, we faced considerable difficulty, which was surprising considering the long history and success of QSAR model development for this test. Other quantum mechanics descriptors were compared to this reaction energy including AM1 semi-empirical orbital energies, nitrenium formation with alternative leaving groups, nitrenium charge, and aryl-amine anion formation energy. Nitrenium formation energy, regardless of the starting species, was found to be the most useful single descriptor. External sets used in other QSAR investigations did not present the same difficulty using the same methods and descriptors. When considering all substructures rather than just aryl-amines, we also noted a significantly lower performance for the Novartis set. The performance gap between Novartis and external sets persists across different descriptors and learning methods. The profiles of the Novartis and external data are significantly different both in aryl-amines and considering all substructures. The Novartis and external data sets are easily separated in an unsupervised clustering using chemical fingerprints. The chemical differences are discussed and visualized using Kohonen Self-Organizing Maps trained on chemical fingerprints, mutagenic substructure prevalence, and molecular weight.ConclusionsDespite extensive work in the area of predicting this particular toxicity, work in designing and publishing more relevant test sets for compounds relevant to drug discovery is still necessary. This work also shows that great care must be taken in using QSAR models to replace experimental evidence. When considering all substructures, a random forest model, which can inherently cover distinct neighborhoods, built on Novartis data and previously reported external data provided a suitable model.


Bioorganic & Medicinal Chemistry | 2011

Avoidance of the Ames test liability for aryl-amines via computation.

Patrick McCarren; Gregory Raymond Bebernitz; Peter Gedeck; Susanne Glowienke; Melissa S. Grondine; Louise Kirman; Jacob Klickstein; Herbert F. Schuster; Lewis Whitehead

Aryl-amines are commonly used synthons in modern drug discovery, however a minority of these chemical templates have the potential to cause toxicity through mutagenicity. The toxicity mostly arises through a series of metabolic steps leading to a reactive electrophilic nitrenium cation intermediate that reacts with DNA nucleotides causing mutation. Highly detailed in silico calculations of the energetics of chemical reactions involved in the metabolic formation of nitrenium cations have been performed. This allowed a critical assessment of the accuracy and reliability of using a theoretical formation energy of the DNA-reactive nitrenium intermediate to correlate with the Ames test response. This study contains the largest data set reported to date, and presents the in silico calculations versus the in vitro Ames response data in the form of beanplots commonly used in statistical analysis. A comparison of this quantum mechanical approach to QSAR and knowledge-based methods is also reported, as well as the calculated formation energies of nitrenium ions for thousands of commercially available aryl-amines generated as a watch-list for medicinal chemists in their synthetic optimization strategies.


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.


PLOS ONE | 2012

Identification of Elongation Factor G as the Conserved Cellular Target of Argyrin B

Beat Nyfeler; Dominic Hoepfner; Deborah Palestrant; Christina A. Kirby; Lewis Whitehead; Robert Yu; Gejing Deng; Ruth E. Caughlan; Angela L. Woods; Adriana K. Jones; S. Whitney Barnes; John R. Walker; Swann Gaulis; Ervan Hauy; Saskia M. Brachmann; Philipp Krastel; Christian Studer; Ralph Riedl; David Estoppey; Thomas Aust; N. Rao Movva; Zuncai Wang; Michael Salcius; Gregory A. Michaud; Gregory McAllister; Leon O. Murphy; John A. Tallarico; Christopher J. Wilson; Charles R. Dean

Argyrins, produced by myxobacteria and actinomycetes, are cyclic octapeptides with antibacterial and antitumor activity. Here, we identify elongation factor G (EF-G) as the cellular target of argyrin B in bacteria, via resistant mutant selection and whole genome sequencing, biophysical binding studies and crystallography. Argyrin B binds a novel allosteric pocket in EF-G, distinct from the known EF-G inhibitor antibiotic fusidic acid, revealing a new mode of protein synthesis inhibition. In eukaryotic cells, argyrin B was found to target mitochondrial elongation factor G1 (EF-G1), the closest homologue of bacterial EF-G. By blocking mitochondrial translation, argyrin B depletes electron transport components and inhibits the growth of yeast and tumor cells. Further supporting direct inhibition of EF-G1, expression of an argyrin B-binding deficient EF-G1 L693Q variant partially rescued argyrin B-sensitivity in tumor cells. In summary, we show that argyrin B is an antibacterial and cytotoxic agent that inhibits the evolutionarily conserved target EF-G, blocking protein synthesis in bacteria and mitochondrial translation in yeast and mammalian cells.

Collaboration


Dive into the Lewis Whitehead's collaboration.

Researchain Logo
Decentralizing Knowledge