Network


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

Hotspot


Dive into the research topics where Felix Vajdos is active.

Publication


Featured researches published by Felix Vajdos.


Cancer Research | 2008

Antitumor Activity and Pharmacology of a Selective Focal Adhesion Kinase Inhibitor, PF-562,271

Walter Gregory Roberts; Ethan Ung; Pamela Whalen; Beth Cooper; Catherine A. Hulford; Christofer Autry; Daniel T. Richter; Earling Emerson; Jing Lin; John Charles Kath; Kevin Coleman; Lili Yao; Luis Martinez-Alsina; Marianne Lorenzen; Martin A. Berliner; Michael Joseph Luzzio; Nandini Chaturbhai Patel; Erika Schmitt; Susan Deborah Lagreca; Jitesh P. Jani; Matt Wessel; Eric S. Marr; Matt Griffor; Felix Vajdos

Cancer cells are characterized by the ability to grow in an anchorage-independent manner. The activity of the nonreceptor tyrosine kinase, focal adhesion kinase (FAK), is thought to contribute to this phenotype. FAK localizes in focal adhesion plaques and has a role as a scaffolding and signaling protein for other adhesion molecules. Recent studies show a strong correlation between increased FAK expression and phosphorylation status and the invasive phenotype of aggressive human tumors. PF-562,271 is a potent, ATP-competitive, reversible inhibitor of FAK and Pyk2 catalytic activity with a IC(50) of 1.5 and 14 nmol/L, respectively. Additionally, PF-562,271 displayed robust inhibition in an inducible cell-based assay measuring phospho-FAK with an IC(50) of 5 nmol/L. PF-562,271 was evaluated against multiple kinases and displays >100x selectivity against a long list of nontarget kinases. PF-562,271 inhibits FAK phosphorylation in vivo in a dose-dependent fashion (calculated EC(50) of 93 ng/mL, total) after p.o. administration to tumor-bearing mice. In vivo inhibition of FAK phosphorylation (>50%) was sustained for >4 hours with a single p.o. dose of 33 mg/kg. Antitumor efficacy and regressions were observed in multiple human s.c. xenograft models. No weight loss, morbidity, or mortality were observed in any in vivo experiment. Tumor growth inhibition was dose and drug exposure dependent. Taken together, these data show that kinase inhibition with an ATP-competitive small molecule inhibitor of FAK decreases the phospho-status in vivo, resulting in robust antitumor activity.


Journal of Medicinal Chemistry | 2012

Spirocyclic Sulfamides as β-Secretase 1 (BACE-1) Inhibitors for the Treatment of Alzheimer’s Disease: Utilization of Structure Based Drug Design, WaterMap, and CNS Penetration Studies To Identify Centrally Efficacious Inhibitors

Michael Aaron Brodney; Gabriela Barreiro; Kevin Ogilvie; Eva Hajos-Korcsok; John C. Murray; Felix Vajdos; Claude Ambroise; Curt Christoffersen; Katherine Fisher; Lorraine Lanyon; JianHua Liu; Charles E. Nolan; Jane M. Withka; Kris A. Borzilleri; Ivan Viktorovich Efremov; Christine E. Oborski; Alison H. Varghese; Brian T. O’Neill

β-Secretase 1 (BACE-1) is an attractive therapeutic target for the treatment and prevention of Alzheimers disease (AD). Herein, we describe the discovery of a novel class of BACE-1 inhibitors represented by sulfamide 14g, using a medicinal chemistry strategy to optimize central nervous system (CNS) penetration by minimizing hydrogen bond donors (HBDs) and reducing P-glycoprotein (P-gp) mediated efflux. We have also taken advantage of the combination of structure based drug design (SBDD) to guide the optimization of the sulfamide analogues and the in silico tool WaterMap to explain the observed SAR. Compound 14g is a potent inhibitor of BACE-1 with excellent permeability and a moderate P-gp liability. Administration of 14g to mice produced a significant, dose-dependent reduction in central Aβ(X-40) levels at a free drug exposure equivalent to the whole cell IC(50) (100 nM). Furthermore, studies of the P-gp knockout mouse provided evidence that efflux transporters affected the amount of Aβ lowering versus that observed in wild-type (WT) mouse at an equivalent dose.


Journal of Medicinal Chemistry | 2012

Discovery and Optimization of a Novel Spiropyrrolidine Inhibitor of β-Secretase (BACE1) through Fragment-Based Drug Design

Ivan Viktorovich Efremov; Felix Vajdos; Kris A. Borzilleri; Steven Capetta; Hou Chen; Peter Hans Dorff; Jason K. Dutra; Steven Wayne Goldstein; Mahmoud N. Mansour; Alexander S. McColl; Stephen Noell; Christine E. Oborski; Thomas N. O’Connell; Theresa J. O’Sullivan; Jayvardhan Pandit; Hong Wang; BinQing Wei; Jane M. Withka

The aspartyl protease β-secretase, or BACE, has been demonstrated to be a key factor in the proteolytic formation of Aβ-peptide, a major component of plaques in the brains of Alzheimers disease (AD) patients, and inhibition of this enzyme has emerged as a major strategy for pharmacologic intervention in AD. An X-ray-based fragment screen of Pfizers proprietary fragment collection has resulted in the identification of a novel BACE binder featuring spiropyrrolidine framework. Although exhibiting only weak inhibitory activity against the BACE enzyme, the small compound was verified by biophysical and NMR-based methods as a bona fide BACE inhibitor. Subsequent optimization of the lead compound, relying heavily on structure-based drug design and computational prediction of physiochemical properties, resulted in a nearly 1000-fold improvement in potency while maintaining ligand efficiency and properties predictive of good permeability and low P-gp liability.


Bioorganic & Medicinal Chemistry Letters | 2010

Discovery of small molecule isozyme non-specific inhibitors of mammalian acetyl-CoA carboxylase 1 and 2.

Jeffrey W. Corbett; Kevin Daniel Freeman-Cook; Richard L. Elliott; Felix Vajdos; Francis Rajamohan; D Kohls; Eric S. Marr; Hailong Zhang; Liang Tong; Meihua Tu; S Murdande; Shawn D. Doran; Janet A. Houser; Wei Song; C.J Jones; Steven B. Coffey; Leanne M. Buzon; Martha L. Minich; Kenneth J. DiRico; Susan Tapley; R.K. McPherson; E Sugarman; H.J Harwood; William Paul Esler

Screening Pfizers compound library resulted in the identification of weak acetyl-CoA carboxylase inhibitors, from which were obtained rACC1 CT-domain co-crystal structures. Utilizing HTS hits and structure-based drug discovery, a more rigid inhibitor was designed and led to the discovery of sub-micromolar, spirochromanone non-specific ACC inhibitors. Low nanomolar, non-specific ACC-isozyme inhibitors that exhibited good rat pharmacokinetics were obtained from this chemotype.


Protein Science | 2007

The 2.0 Å crystal structure of the ERα ligand‐binding domain complexed with lasofoxifene

Felix Vajdos; Lise R. Hoth; Kieran F. Geoghegan; Samuel P. Simons; Peter K. LeMotte; Dennis E. Danley; Mark Ammirati; Jayvardhan Pandit

Lasofoxifene is a new and potent selective estrogen receptor modulator (SERM). The structural basis of its interaction with the estrogen receptor has been investigated by crystallographic analysis of its complex with the ligand‐binding domain of estrogen receptor α at a resolution of 2.0 Å. As with other SERMs, lasofoxifene diverts the receptor from its agonist‐bound conformation by displacing the C‐terminal AF‐2 helix into the site at which the LXXLL motif of coactivator proteins would otherwise be able to bind. Lasofoxifene achieves this effect by occupying the space normally filled by residue Leu 540, as well as by modulating the conformation of residues of helix 11 (His 524, Leu 525). A well‐defined salt bridge between lasofoxifene and Asp 351 suggests that charge neutralization in this region of the receptor may explain the some of the antiestrogenic effects of lasofoxifene. The results suggest general features of ERα/SERM recognition, and add a new dimension to efforts to rationalize differences between the biological activity profiles exhibited by these important pharmacological agents.


Journal of Medicinal Chemistry | 2015

Discovery of a Series of Efficient, Centrally Efficacious BACE1 Inhibitors through Structure-Based Drug Design.

Christopher Ryan Butler; Michael Aaron Brodney; Elizabeth Mary Beck; Gabriela Barreiro; Charles E. Nolan; Feng Pan; Felix Vajdos; Kevin Parris; Alison H. Varghese; Christopher John Helal; Ricardo Lira; Shawn D. Doran; David Riddell; Leanne M. Buzon; Jason K. Dutra; Luis Martinez-Alsina; Kevin Ogilvie; John C. Murray; Joseph M. Young; Kevin Atchison; Ashley Robshaw; Cathleen Gonzales; Jinlong Wang; Yong Zhang; Brian T. O’Neill

The identification of centrally efficacious β-secretase (BACE1) inhibitors for the treatment of Alzheimers disease (AD) has historically been thwarted by an inability to maintain alignment of potency, brain availability, and desired absorption, distribution, metabolism, and excretion (ADME) properties. In this paper, we describe a series of truncated, fused thioamidines that are efficiently selective in garnering BACE1 activity without simultaneously inhibiting the closely related cathepsin D or negatively impacting brain penetration and ADME alignment, as exemplified by 36. Upon oral administration, these inhibitors exhibit robust brain availability and are efficacious in lowering central Amyloid β (Aβ) levels in mouse and dog. In addition, chronic treatment in aged PS1/APP mice effects a decrease in the number and size of Aβ-derived plaques. Most importantly, evaluation of 36 in a 2-week exploratory toxicology study revealed no accumulation of autofluorescent material in retinal pigment epithelium or histology findings in the eye, issues observed with earlier BACE1 inhibitors.


Journal of Medicinal Chemistry | 2014

Decreasing the Rate of Metabolic Ketone Reduction in the Discovery of a Clinical Acetyl-CoA Carboxylase Inhibitor for the Treatment of Diabetes

David A. Griffith; Daniel W. Kung; William Esler; Paul Amor; Scott W. Bagley; Carine Beysen; Santos Carvajal-Gonzalez; Shawn D. Doran; Chris Limberakis; Alan M. Mathiowetz; Kirk McPherson; David A. Price; Eric Ravussin; Gabriele Sonnenberg; James A. Southers; Laurel Sweet; Scott M. Turner; Felix Vajdos

Acetyl-CoA carboxylase (ACC) inhibitors offer significant potential for the treatment of type 2 diabetes mellitus (T2DM), hepatic steatosis, and cancer. However, the identification of tool compounds suitable to test the hypothesis in human trials has been challenging. An advanced series of spirocyclic ketone-containing ACC inhibitors recently reported by Pfizer were metabolized in vivo by ketone reduction, which complicated human pharmacology projections. We disclose that this metabolic reduction can be greatly attenuated through introduction of steric hindrance adjacent to the ketone carbonyl. Incorporation of weakly basic functionality improved solubility and led to the identification of 9 as a clinical candidate for the treatment of T2DM. Phase I clinical studies demonstrated dose-proportional increases in exposure, single-dose inhibition of de novo lipogenesis (DNL), and changes in indirect calorimetry consistent with increased whole-body fatty acid oxidation. This demonstration of target engagement validates the use of compound 9 to evaluate the role of DNL in human disease.


Journal of Medicinal Chemistry | 2017

Design of a Janus Kinase 3 (JAK3) Specific Inhibitor 1-((2S,5R)-5-((7H-Pyrrolo[2,3-d]pyrimidin-4-yl)amino)-2-methylpiperidin-1-yl)prop-2-en-1-one (PF-06651600) Allowing for the Interrogation of JAK3 Signaling in Humans

Atli Thorarensen; Martin E. Dowty; Mary Ellen Banker; Brian Juba; Jason Jussif; Tsung Lin; Fabien Vincent; Robert M. Czerwinski; Agustin Casimiro-Garcia; Ray Unwalla; John I. Trujillo; Sidney Xi Liang; Paul Balbo; Ye Che; Adam M. Gilbert; Matthew Frank Brown; Matthew Merrill Hayward; Justin Ian Montgomery; Louis Leung; Xin Yang; Sarah Soucy; Martin Hegen; Jotham Wadsworth Coe; Jonathan Langille; Felix Vajdos; Jill Chrencik; Jean-Baptiste Telliez

Significant work has been dedicated to the discovery of JAK kinase inhibitors resulting in several compounds entering clinical development and two FDA approved NMEs. However, despite significant effort during the past 2 decades, identification of highly selective JAK3 inhibitors has eluded the scientific community. A significant effort within our research organization has resulted in the identification of the first orally active JAK3 specific inhibitor, which achieves JAK isoform specificity through covalent interaction with a unique JAK3 residue Cys-909. The relatively rapid resynthesis rate of the JAK3 enzyme presented a unique challenge in the design of covalent inhibitors with appropriate pharmacodynamics properties coupled with limited unwanted off-target reactivity. This effort resulted in the identification of 11 (PF-06651600), a potent and low clearance compound with demonstrated in vivo efficacy. The favorable efficacy and safety profile of this JAK3-specific inhibitor 11 led to its evaluation in several human clinical studies.


Biochemistry | 2012

Deconstruction of activity-dependent covalent modification of heme in human neutrophil myeloperoxidase by multistage mass spectrometry (MS(4)).

Kieran F. Geoghegan; Alison H. Varghese; Xidong Feng; Andrew J. Bessire; James J. Conboy; Roger Benjamin Ruggeri; Kay Ahn; Samantha N. Spath; Sergey V. Filippov; Steven J. Conrad; Philip A. Carpino; Cristiano R. W. Guimarães; Felix Vajdos

Myeloperoxidase (MPO) is known to be inactivated and covalently modified by treatment with hydrogen peroxide and agents similar to 3-(2-ethoxypropyl)-2-thioxo-2,3-dihydro-1H-purin-6(9H)-one (1), a 254.08 Da derivative of 2-thioxanthine. Peptide mapping by liquid chromatography and mass spectrometry detected modification by 1 in a labile peptide-heme-peptide fragment of the enzyme, accompanied by a mass increase of 252.08 Da. The loss of two hydrogen atoms was consistent with mechanism-based oxidative coupling. Multistage mass spectrometry (MS(4)) of the modified fragment in an ion trap/Orbitrap spectrometer demonstrated that 1 was coupled directly to heme. Use of a 10 amu window delivered the full isotopic envelope of each precursor ion to collision-induced dissociation, preserving definitive isotopic profiles for iron-containing fragments through successive steps of multistage mass spectrometry. Iron isotope signatures and accurate mass measurements supported the structural assignments. Crystallographic analysis confirmed linkage between the methyl substituent of the heme pyrrole D ring and the sulfur atom of 1. The final orientation of 1 perpendicular to the plane of the heme ring suggested a mechanism consisting of two consecutive one-electron oxidations of 1 by MPO. Multistage mass spectrometry using stage-specific collision energies permits stepwise deconstruction of modifications of heme enzymes containing covalent links between the heme group and the polypeptide chain.


MedChemComm | 2014

Small-molecule phosphodiesterase probes: discovery of potent and selective CNS-penetrable quinazoline inhibitors of PDE1

John Michael Humphrey; Eddie Yang; Christopher W. am Ende; Eric P. Arnold; Jenna L. Head; Stephen Jenkinson; Lorraine A. Lebel; Spiros Liras; Jayvardhan Pandit; Brian Samas; Felix Vajdos; Samuel P. Simons; Artem G. Evdokimov; Mahmoud N. Mansour; Frank S. Menniti

PDE1 is a family of calcium-activated, dual substrate phosphodiesterases expressed in both the CNS and periphery that play a role in the integration of intracellular calcium and cyclic nucleotide signaling cascades. Exploration of the potential in targeting this family of enzymes to treat neuropsychiatric disorders has been hampered by a lack of potent, selective, and brain penetrable PDE1 inhibitors. To identify such compounds we used high-throughput screening, structure-based design, and targeted synthetic chemistry to discover the 4-aminoquinazoline 7a (PF-04471141) and the 4-indanylquinazoline 27 (PF-04822163) each of which are PDE1 inhibitors that readily cross the blood brain barrier. These quinazoline-based PDE1-selective inhibitors represent valuable new tools to study the biological processes regulated by PDE1 and to begin to determine the potential therapeutic utility of such compounds to treat neuropsychiatric disorders.

Researchain Logo
Decentralizing Knowledge