29 results on '"Felix Vajdos"'
Search Results
2. Demonstration of In Vitro to In Vivo Translation of a TYK2 Inhibitor That Shows Cross Species Potency Differences
- Author
-
Martin E. Dowty, James F. Smith, Tsung H. Lin, Brian S. Gerstenberger, Matthew C. Griffor, Brett D. Hollingshead, Roger S. Gifford, Martin Hegen, Felix Vajdos, John D. Knafels, Andrew Fensome, Mary Ellen Banker, and James D. Clark
- Subjects
Mutant ,Immunology ,lcsh:Medicine ,Article ,Mice ,Adenosine Triphosphate ,Dogs ,TYK2 Kinase ,Protein Domains ,Species Specificity ,In vivo ,Potency ,Animals ,Humans ,Amino Acid Sequence ,Binding site ,lcsh:Science ,Protein Kinase Inhibitors ,Multidisciplinary ,Binding Sites ,Sequence Homology, Amino Acid ,Drug discovery ,Kinase ,Chemistry ,lcsh:R ,Janus Kinase 1 ,In vitro ,Biochemistry ,Mutation ,Macaca ,lcsh:Q - Abstract
Translation of modulation of drug target activity to therapeutic effect is a critical aspect for all drug discovery programs. In this work we describe the profiling of a non-receptor tyrosine-protein kinase (TYK2) inhibitor which shows a functionally relevant potency shift between human and preclinical species (e.g. murine, dog, macaque) in both biochemical and cellular assays. Comparison of the structure and sequence homology of TYK2 between human and preclinical species within the ATP binding site highlights a single amino acid (I960 → V) responsible for the potency shift. Through TYK2 kinase domain mutants and a TYK2 980I knock-in mouse model, we demonstrate that this single amino acid change drives a functionally relevant potency difference that exists between human and all evaluated preclinical species, for a series of TYK2 inhibitors which target the ATP binding site.
- Published
- 2020
- Full Text
- View/download PDF
3. Dual Inhibition of TYK2 and JAK1 for the Treatment of Autoimmune Diseases: Discovery of ((S)-2,2-Difluorocyclopropyl)((1R,5S)-3-(2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)-3,8-diazabicyclo[3.2.1]octan-8-yl)methanone (PF-06700841)
- Author
-
Brett D. Hollingshead, Fabien Vincent, Tsung H. Lin, John I. Trujillo, Betsy Pierce, Andrew Fensome, Brian S. Gerstenberger, Martin E. Dowty, Matthew Frank Brown, Xin Yang, David C. Limburg, Martin Hegen, John D. Knafels, Felix Vajdos, Andrew C. Flick, Mary Ellen Banker, Raman Sharma, Li Xing, Jason Jussif, Catherine M. Ambler, Ivan Viktorovich Efremov, James D. Clark, Liying Zhang, Peter T. Symanowicz, Eddine Saiah, Jill Chrencik, Jean-Baptiste Telliez, Xiaojing Yang, Eric P. Arnold, Ariamala Gopalsamy, Zhao-Kui Wan, Matthew Merrill Hayward, and David W. Lin
- Subjects
0301 basic medicine ,Janus kinase 1 ,Chemistry ,medicine.medical_treatment ,Interleukin ,Arthritis ,Pharmacology ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Cytokine ,Interferon ,Tyrosine kinase 2 ,030220 oncology & carcinogenesis ,Drug Discovery ,medicine ,STAT protein ,Molecular Medicine ,Janus kinase ,medicine.drug - Abstract
Cytokine signaling is an important characteristic of autoimmune diseases. Many pro-inflammatory cytokines signal through the Janus kinase (JAK)/Signal transducer and activator of transcription (STAT) pathway. JAK1 is important for the γ-common chain cytokines, interleukin (IL)-6, and type-I interferon (IFN) family, while TYK2 in addition to type-I IFN signaling also plays a role in IL-23 and IL-12 signaling. Intervention with monoclonal antibodies (mAbs) or JAK1 inhibitors has demonstrated efficacy in Phase III psoriasis, psoriatic arthritis, inflammatory bowel disease, and rheumatoid arthritis studies, leading to multiple drug approvals. We hypothesized that a dual JAK1/TYK2 inhibitor will provide additional efficacy, while managing risk by optimizing selectivity against JAK2 driven hematopoietic changes. Our program began with a conformationally constrained piperazinyl-pyrimidine Type 1 ATP site inhibitor, subsequent work led to the discovery of PF-06700841 (compound 23), which is in Phase II clinical d...
- Published
- 2018
- Full Text
- View/download PDF
4. Aminomethyl-Derived Beta Secretase (BACE1) Inhibitors: Engaging Gly230 without an Anilide Functionality
- Author
-
Felix Vajdos, Eric Benvenuti, Leanne M. Buzon, Brian T. O’Neill, Gabriela Barreiro, David Riddell, Kevin Atchison, Charles E. Nolan, Ashley Robshaw, Eric F. Johnson, Kevin D. Parris, Shawn D. Doran, Michael Aaron Brodney, Xinjun Hou, Luis Martinez-Alsina, Kimberly Lapham, Mei-Hui Hsu, Elizabeth Mary Beck, Christopher Ryan Butler, Kevin Ogilvie, Cathleen Gonzales, Lorraine F. Lanyon, and Christopher John Helal
- Subjects
Male ,0301 basic medicine ,Patch-Clamp Techniques ,Stereochemistry ,Glycine ,Article ,Mice ,Structure-Activity Relationship ,03 medical and health sciences ,0302 clinical medicine ,Tandem Mass Spectrometry ,Drug Discovery ,Animals ,Moiety ,Potency ,Structure–activity relationship ,Anilides ,Amino Acid Sequence ,Enzyme Inhibitors ,Binding site ,Structural motif ,Chromatography, High Pressure Liquid ,ADME ,biology ,Chemistry ,Brain ,030104 developmental biology ,biology.protein ,Molecular Medicine ,Amyloid Precursor Protein Secretases ,Crystallization ,Linker ,Amyloid precursor protein secretase ,030217 neurology & neurosurgery - Abstract
A growing subset of β-secretase (BACE1) inhibitors for the treatment of Alzheimer's disease (AD) utilizes an anilide chemotype that engages a key residue (Gly230) in the BACE1 binding site. Although the anilide moiety affords excellent potency, it simultaneously introduces a third hydrogen bond donor that limits brain availability and provides a potential metabolic site leading to the formation of an aniline, a structural motif of prospective safety concern. We report herein an alternative aminomethyl linker that delivers similar potency and improved brain penetration relative to the amide moiety. Optimization of this series identified analogues with an excellent balance of ADME properties and potency; however, potential drug-drug interactions (DDI) were predicted based on CYP 2D6 affinities. Generation and analysis of key BACE1 and CYP 2D6 crystal structures identified strategies to obviate the DDI liability, leading to compound 16, which exhibits robust in vivo efficacy as a BACE1 inhibitor.
- Published
- 2016
- Full Text
- View/download PDF
5. Design and optimization of a series of 4-(3-azabicyclo[3.1.0]hexan-3-yl)pyrimidin-2-amines: Dual inhibitors of TYK2 and JAK1
- Author
-
David C. Limburg, Andrew Fensome, Fabien Vincent, Betsy Pierce, Martin E. Dowty, Mary Ellen Banker, Raman Sharma, Xin Yang, Ariamala Gopalsamy, Roger S. Gifford, Brian S. Gerstenberger, Andrew C. Flick, Eric P. Arnold, Jason Jussif, Ivan Viktorovich Efremov, Xiaojing Yang, Martin Hegen, Zhao-Kui Wan, Felix Vajdos, Catherine M. Ambler, James D. Clark, Tsung H. Lin, John I. Trujillo, and Li Xing
- Subjects
Pyrimidine ,Clinical Biochemistry ,Rat model ,Pharmaceutical Science ,01 natural sciences ,Biochemistry ,Structure-Activity Relationship ,chemistry.chemical_compound ,Drug Discovery ,Animals ,Humans ,Protein Kinase Inhibitors ,Molecular Biology ,ADME ,TYK2 Kinase ,Dose-Response Relationship, Drug ,Molecular Structure ,010405 organic chemistry ,Organic Chemistry ,Janus Kinase 1 ,Arthritis, Experimental ,Combinatorial chemistry ,Rats ,0104 chemical sciences ,Disease Models, Animal ,010404 medicinal & biomolecular chemistry ,chemistry ,Rats, Inbred Lew ,Drug Design ,Molecular Medicine ,Female ,Selectivity ,Transferase inhibitor - Abstract
Herein, we disclose a new series of TYK2/ JAK1 inhibitors based upon a 3.1.0 azabicyclic substituted pyrimidine scaffold. We illustrate the use of structure-based drug design for the initial design and subsequent optimization of this series of compounds. One advanced example 19 met program objectives for potency, selectivity and ADME, and demonstrated oral activity in the adjuvant-induced arthritis rat model.
- Published
- 2020
- Full Text
- View/download PDF
6. Discovery of 3-Cyano- N-(3-(1-isobutyrylpiperidin-4-yl)-1-methyl-4-(trifluoromethyl)-1 H-pyrrolo[2,3- b]pyridin-5-yl)benzamide: A Potent, Selective, and Orally Bioavailable Retinoic Acid Receptor-Related Orphan Receptor C2 Inverse Agonist
- Author
-
Martin Hegen, David C. Wood, Patrik Rhönnstad, Felix Vajdos, Atli Thorarensen, Mark E. Schnute, Timothy Braden, Kimberly Crouse, Maria Sjöberg, John I. Trujillo, Ravi G. Kurumbail, Bolette Husman, Konrad F. Koehler, Mattias Wennerstål, Tomas Bonn, Joakim Löfstedt, Eva Backström-Rydin, Bo Carlsson, Aron Sundell, Ming Z. Chen, Steven E. Heasley, John David Trzupek, Annika Goos-Nilsson, Carol A. Menard, Peter Harris, James R. Kiefer, Martin Bengtsson, Leon P. Collis, Michael J. Prinsen, Philippe Nuhant, Jennifer Alley, Scott A. Long, Alexander E. Hromockyj, Andrew C. Flick, Johnny Sandberg, Christoph W. Zapf, Edouard Zamaratski, Xiao Hu, Lee Napierata, Björn Kauppi, Nicole Caspers, Kimberly F. Fennell, Robert E. Kyne, Gabriel Berstein, Neelu Kaila, Lars Kruger, Wei Li, Li Xing, Ray Unwalla, Elisabet Kallin, Matthew J. Pelc, Susan Fish, James Robert Blinn, Hjalmar Gullberg, Marvin J. Meyers, Scot Richard Mente, Chulho Choi, Falgun Shah, Mathias Färnegårdh, Dean Messing, Peter G. Jones, Yajuan Zhao, Alexandria P. Taylor, Maria Sandström, Charles W. Bolten, Daniel Nöteberg, Robin A. Weinberg, Tomasz Janosik, John D. Knafels, and Anna Wilhelmsson
- Subjects
0301 basic medicine ,Drug Inverse Agonism ,Stereochemistry ,Pyridines ,Retinoic acid ,Drug Evaluation, Preclinical ,Administration, Oral ,Biological Availability ,01 natural sciences ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,RAR-related orphan receptor gamma ,Drug Discovery ,Inverse agonist ,Animals ,Humans ,Benzamide ,Receptor ,Orphan receptor ,010405 organic chemistry ,Nuclear Receptor Subfamily 1, Group F, Member 3 ,0104 chemical sciences ,Retinoic acid receptor ,030104 developmental biology ,chemistry ,Nuclear receptor ,Drug Design ,Molecular Medicine ,Th17 Cells - Abstract
The nuclear hormone receptor retinoic acid receptor-related orphan C2 (RORC2, also known as RORγt) is a promising target for the treatment of autoimmune diseases. A small molecule, inverse agonist of the receptor is anticipated to reduce production of IL-17, a key proinflammatory cytokine. Through a high-throughput screening approach, we identified a molecule displaying promising binding affinity for RORC2, inhibition of IL-17 production in Th17 cells, and selectivity against the related RORA and RORB receptor isoforms. Lead optimization to improve the potency and metabolic stability of this hit focused on two key design strategies, namely, iterative optimization driven by increasing lipophilic efficiency and structure-guided conformational restriction to achieve optimal ground state energetics and maximize receptor residence time. This approach successfully identified 3-cyano- N-(3-(1-isobutyrylpiperidin-4-yl)-1-methyl-4-(trifluoromethyl)-1 H-pyrrolo[2,3- b]pyridin-5-yl)benzamide as a potent and selective RORC2 inverse agonist, demonstrating good metabolic stability, oral bioavailability, and the ability to reduce IL-17 levels and skin inflammation in a preclinical in vivo animal model upon oral administration.
- Published
- 2018
7. Discovery of Potent and Selective Periphery-Restricted Quinazoline Inhibitors of the Cyclic Nucleotide Phosphodiesterase PDE1
- Author
-
Eddie Yang, Jennifer L. Liras, Christine C. Orozco, Christopher W. am Ende, Matthew A. Movsesian, Jayvardhan Pandit, John M. Humphrey, Stephen Jenkinson, Frank S. Menniti, Thomas Allen Chappie, Spiros Liras, Stacey L. Becker, Felix Vajdos, and Fabrice Vandeput
- Subjects
0301 basic medicine ,Models, Molecular ,Phosphodiesterase Inhibitors ,Protein Conformation ,030204 cardiovascular system & hematology ,PDE1 ,Human myocardium ,Cocrystal ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Drug Discovery ,Quinazoline ,Ic50 values ,Cyclic AMP ,Humans ,Cyclic nucleotide phosphodiesterase ,Molecular Structure ,Myocardium ,Cyclic Nucleotide Phosphodiesterases, Type 1 ,030104 developmental biology ,chemistry ,Biochemistry ,Plasma concentration ,Quinazolines ,Molecular Medicine - Abstract
We disclose the discovery and X-ray cocrystal data of potent, selective quinazoline inhibitors of PDE1. Inhibitor (S)-3 readily attains free plasma concentrations above PDE1 IC50 values and has restricted brain access. The racemic compound 3 inhibits >75% of PDE hydrolytic activity in soluble samples of human myocardium, consistent with heightened PDE1 activity in this tissue. These compounds represent promising new tools to probe the value of PDE1 inhibition in the treatment of cardiovascular disease.
- Published
- 2018
8. Discovery of a Series of Efficient, Centrally Efficacious BACE1 Inhibitors through Structure-Based Drug Design
- Author
-
Christopher John Helal, Kevin Atchison, Christopher Ryan Butler, Cathleen Gonzales, Ricardo Lira, Shawn D. Doran, Feng Pan, Felix Vajdos, Alison H. Varghese, Gabriela Barreiro, Michael Aaron Brodney, Jinlong Wang, Elizabeth Mary Beck, Luis Martinez-Alsina, Yong Zhang, Kevin D. Parris, David Riddell, Joseph Michael Young, Leanne M. Buzon, Ashley Robshaw, Kevin Ogilvie, Jason K. Dutra, Brian T. O’Neill, John C. Murray, and Charles E. Nolan
- Subjects
Male ,Models, Molecular ,Drug ,media_common.quotation_subject ,Amidines ,Cathepsin D ,Plaque, Amyloid ,Pharmacology ,Mice ,Dogs ,Alzheimer Disease ,Oral administration ,mental disorders ,Drug Discovery ,medicine ,Animals ,Aspartic Acid Endopeptidases ,Humans ,Potency ,Sulfhydryl Compounds ,Enzyme Inhibitors ,Rats, Wistar ,media_common ,ADME ,Amyloid beta-Peptides ,Retinal pigment epithelium ,biology ,Chemistry ,Brain ,medicine.disease ,Rats ,medicine.anatomical_structure ,Drug Design ,biology.protein ,Molecular Medicine ,Amyloid Precursor Protein Secretases ,Alzheimer's disease ,Amyloid precursor protein secretase - Abstract
The identification of centrally efficacious β-secretase (BACE1) inhibitors for the treatment of Alzheimer's 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.
- Published
- 2015
- Full Text
- View/download PDF
9. 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
- Author
-
Louis Leung, Jonathan Langille, John I. Trujillo, Paul B. Balbo, Jason Jussif, Jill Chrencik, Martin Hegen, Felix Vajdos, Atli Thorarensen, Sarah Soucy, Adam M. Gilbert, Jean-Baptiste Telliez, Tsung Lin, Ray Unwalla, Matthew Merrill Hayward, Justin I. Montgomery, Sidney Liang, Fabien Vincent, Robert M. Czerwinski, Ye Che, Martin E. Dowty, Mary Ellen Banker, Xin Yang, Matthew Frank Brown, Agustin Casimiro-Garcia, Jotham Wadsworth Coe, and Brian Juba
- Subjects
0301 basic medicine ,Gene isoform ,Administration, Oral ,Pharmacology ,01 natural sciences ,03 medical and health sciences ,In vivo ,Drug Discovery ,Humans ,Pyrroles ,Protein Kinase Inhibitors ,chemistry.chemical_classification ,010405 organic chemistry ,Janus kinase 3 ,Janus Kinase 3 ,0104 chemical sciences ,Safety profile ,030104 developmental biology ,Orally active ,Enzyme ,Pyrimidines ,Biochemistry ,chemistry ,Pharmacodynamics ,Drug Design ,Molecular Medicine ,Janus kinase ,Signal Transduction - Abstract
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.
- Published
- 2017
10. Decreasing the Rate of Metabolic Ketone Reduction in the Discovery of a Clinical Acetyl-CoA Carboxylase Inhibitor for the Treatment of Diabetes
- Author
-
Laurel Sweet, Carine Beysen, William P. Esler, David Price, Alan M. Mathiowetz, David A. Griffith, Daniel W. Kung, Scott W. Bagley, Scott M. Turner, Gabriele E. Sonnenberg, Eric Ravussin, Santos Carvajal-Gonzalez, Felix Vajdos, Chris Limberakis, James A. Southers, Paul A. Amor, Shawn D. Doran, and Kirk McPherson
- Subjects
Adult ,Male ,Models, Molecular ,Ketone ,Article ,Rats, Sprague-Dawley ,Structure-Activity Relationship ,Young Adult ,Dogs ,Double-Blind Method ,In vivo ,Microsomes ,Drug Discovery ,medicine ,Animals ,Humans ,Rats, Wistar ,Beta oxidation ,Cells, Cultured ,chemistry.chemical_classification ,Cross-Over Studies ,Molecular Structure ,Lipogenesis ,Acetyl-CoA carboxylase ,Type 2 Diabetes Mellitus ,Ketones ,Middle Aged ,medicine.disease ,3. Good health ,Pyruvate carboxylase ,Rats ,Malonyl Coenzyme A ,chemistry ,Biochemistry ,Diabetes Mellitus, Type 2 ,Area Under Curve ,Hepatocytes ,Molecular Medicine ,Steatosis ,Acetyl-CoA Carboxylase - Abstract
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.
- Published
- 2014
11. Small-molecule phosphodiesterase probes: discovery of potent and selective CNS-penetrable quinazoline inhibitors of PDE1
- Author
-
Artem G. Evdokimov, John M. Humphrey, Christopher W. am Ende, Brian Samas, Lorraine A. Lebel, Frank S. Menniti, Mahmoud N. Mansour, Eddie Yang, Eric P. Arnold, Stephen Jenkinson, Spiros Liras, Felix Vajdos, Jenna L. Head, Samuel P. Simons, and Jayvardhan Pandit
- Subjects
Pharmacology ,chemistry.chemical_classification ,Organic Chemistry ,Pharmaceutical Science ,Phosphodiesterase ,PDE1 ,Biology ,Blood–brain barrier ,Biochemistry ,Small molecule ,Calcium in biology ,Cyclic nucleotide ,chemistry.chemical_compound ,medicine.anatomical_structure ,Enzyme ,chemistry ,Drug Discovery ,medicine ,Quinazoline ,Molecular Medicine - Abstract
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.
- Published
- 2014
- Full Text
- View/download PDF
12. Discovery and synthesis of novel 4-aminopyrrolopyrimidine Tie-2 kinase inhibitors for the treatment of solid tumors
- Author
-
Jean S. Beebe, William M. Hungerford, Nandini Chaturbhai Patel, Heather N. Frost, Susan Deborah Lagreca, Cathy Soderstrom, Shefali Kakar, Doug Savage, Matthew David Wessel, Matthew A. Marx, Merin Boehm, Thompson Carl Brian, Jinshan M. Chen, Connell Richard D, Nandell F. Keene, Elizabeth Knauth, Gary Borzillo, Aaron Kanter, Yong Lu, Joel Morris, Martin A. Berliner, Patrick Vincent, Bruce D. Cohen, George T. Tkalcevic, Joel T. Arcari, Deborah A. Gordon, James J. Valentine, Tracey Clark, Louis Martinez-Alsina, Vincent Bernardo, Felix Vajdos, and Norma Jacqueline Tom
- Subjects
Male ,Benzimidazole ,Clinical Biochemistry ,Pharmaceutical Science ,Antineoplastic Agents ,Biochemistry ,Rats, Sprague-Dawley ,Structure-Activity Relationship ,chemistry.chemical_compound ,Cell Line, Tumor ,Neoplasms ,Drug Discovery ,Animals ,Humans ,Structure–activity relationship ,Pyrroles ,Protein Kinase Inhibitors ,Molecular Biology ,Cell Proliferation ,4-aminopyrrolopyrimidine ,Dose-Response Relationship, Drug ,Molecular Structure ,Drug discovery ,Kinase ,Organic Chemistry ,Receptor, TIE-2 ,Xenograft Model Antitumor Assays ,In vitro ,Rats ,Pyrimidines ,chemistry ,Cell culture ,Molecular Medicine ,Growth inhibition - Abstract
The synthesis and biological evaluation of novel Tie-2 kinase inhibitors are presented. Based on the pyrrolopyrimidine chemotype, several new series are described, including the benzimidazole series by linking a benzimidazole to the C5-position of the 4-amino-pyrrolopyrimidine core and the ketophenyl series synthesized by incorporating a ketophenyl group to the C5-position. Medicinal chemistry efforts led to potent Tie-2 inhibitors. Compound 15, a ketophenyl pyrrolopyrimidine urea analog with improved physicochemical properties, demonstrated favorable in vitro attributes as well as dose responsive and robust oral tumor growth inhibition in animal models.
- Published
- 2013
- Full Text
- View/download PDF
13. Deconstruction of Activity-Dependent Covalent Modification of Heme in Human Neutrophil Myeloperoxidase by Multistage Mass Spectrometry (MS4)
- Author
-
Kay Ahn, Steven J. Conrad, Alison H. Varghese, Roger B. Ruggeri, Felix Vajdos, Andrew J. Bessire, Philip A. Carpino, Xidong Feng, James J. Conboy, Samantha N. Spath, Sergey V. Filippov, Cristiano Ruch Werneck Guimarães, and Kieran F. Geoghegan
- Subjects
Models, Molecular ,Protein mass spectrometry ,Neutrophils ,Molecular Sequence Data ,Heme ,Crystallography, X-Ray ,Mass spectrometry ,Tandem mass spectrometry ,Orbitrap ,Peptide Mapping ,Biochemistry ,Sample preparation in mass spectrometry ,law.invention ,chemistry.chemical_compound ,Tandem Mass Spectrometry ,law ,Catalytic Domain ,Humans ,Amino Acid Sequence ,Peroxidase ,Chromatography ,biology ,Peptide Fragments ,Molecular Weight ,Models, Chemical ,chemistry ,Myeloperoxidase ,biology.protein ,Ion trap ,Oxidation-Reduction ,Chromatography, Liquid - Abstract
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.
- Published
- 2012
- Full Text
- View/download PDF
14. Discovery of spirocyclic-diamine inhibitors of mammalian acetyl CoA-carboxylase
- Author
-
Kathrine Loomis, Kristin L. Rockwell, Meihua Tu, Paul A. Amor, Kirk McPherson, William P. Esler, Tereece Banks, Aaron C. Smith, Shawn Cabral, James J. Valentine, Susan Tapley, Kristen Ford, Colin R. Rose, Mark Niosi, Felix Vajdos, Carmen N. Garcia-Irizarry, Shawn D. Doran, James A. Southers, Scott W. Bagley, Margaret S. Landis, Alan M. Mathiowetz, Daniel W. Kung, and David A. Griffith
- Subjects
Gene isoform ,Stereochemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Biochemistry ,Models, Biological ,Small Molecule Libraries ,chemistry.chemical_compound ,Enzyme activator ,Inhibitory Concentration 50 ,Acetyl Coenzyme A ,Diamine ,Drug Discovery ,Animals ,Humans ,Spiro Compounds ,Enzyme Inhibitors ,Molecular Biology ,Molecular Structure ,Drug discovery ,Organic Chemistry ,Acetyl-CoA carboxylase ,Pyruvate carboxylase ,Rats ,Enzyme Activation ,chemistry ,Lipogenesis ,Hepatocytes ,Molecular Medicine ,Lead compound ,Acetyl-CoA Carboxylase - Abstract
A novel series of spirocyclic-diamine based, isoform non-selective inhibitors of acetyl-CoA carboxylase (ACC) is described. These spirodiamine derivatives were discovered by design of a library to mimic the structural rigidity and hydrogen-bonding pattern observed in the co-crystal structure of spirochromanone inhibitor I. The lead compound 3.5.1 inhibited de novo lipogenesis in rat hepatocytes, with an IC50 of 0.30 μM.
- Published
- 2015
15. Suppressor of Fused Regulates Gli Activity through a Dual Binding Mechanism
- Author
-
Frederic J. de Sauvage, Felix Vajdos, Mark Merchant, Jennifer Cannon, Mark Ultsch, Henry R. Maun, Ulrich Wendt, William Desmarais, Abraham M. de Vos, and Robert A. Lazarus
- Subjects
DNA Mutational Analysis ,Repressor ,Biology ,Zinc Finger Protein GLI1 ,Protein Structure, Secondary ,Genes, Reporter ,GLI1 ,Chlorocebus aethiops ,Animals ,Humans ,Binding site ,Molecular Biology ,Transcription factor ,Psychological repression ,Transcriptional Regulation ,Zinc finger ,Binding Sites ,Cell Biology ,Molecular biology ,Hedgehog signaling pathway ,Protein Structure, Tertiary ,Repressor Proteins ,Gene Expression Regulation ,COS Cells ,biology.protein ,Transcription Factors - Abstract
The Hedgehog pathway drives proliferation and differentiation by activating the Gli/Ci family of zinc finger transcription factors. Gli/Ci proteins form Hedgehog signaling complexes with other signaling components, including the kinesin-like protein Costal-2, the serine-threonine kinase Fused, and Suppressor of Fused [Su(fu)]. In these complexes Gli/Ci proteins are regulated by cytoplasmic sequestration, phosphorylation, and proteolysis. Here we characterize structural and functional determinants of Su(fu) required for Gli regulation and show that Su(fu) contains at least two distinct domains: a highly conserved carboxy-terminal region required for binding to the amino-terminal ends of the Gli proteins and a unique amino-terminal domain that binds the carboxy-terminal tail of Gli1. While each domain is capable of binding to different Gli1 regions independently, interactions between Su(fu) and Gli1 at both sites are required for cytoplasmic tethering and repression of Gli1. Furthermore, we have solved the crystal structure of the amino-terminal domain of human Su(fu)(27-268) at 2.65 A resolution. This domain forms a concave pocket with a prominent acidic patch. Mutation at Asp(159) in the acidic patch disrupts Gli1 tethering and repression while not strongly disrupting binding, indicating that the amino-terminal domain of Su(fu) likely impacts Gli binding through a mechanism distinct from that for tethering and repression. These studies provide a structural basis for understanding the function of Su(fu).
- Published
- 2004
- Full Text
- View/download PDF
16. Structural insights into the catalytic mechanism of cyclophilin A
- Author
-
Felix Vajdos, Su Li, Bruce R. Howard, Christopher P. Hill, and Wesley I. Sundquist
- Subjects
Models, Molecular ,Proline ,Protein family ,Macromolecular Substances ,Protein Conformation ,Stereochemistry ,Molecular Sequence Data ,Cypa ,Isomerase ,Arginine ,Crystallography, X-Ray ,Catalysis ,Viral Proteins ,Cyclophilin A ,Capsid ,Protein structure ,Structural Biology ,Amino Acid Sequence ,Molecular Biology ,Binding Sites ,biology ,Chemistry ,Active site ,biology.organism_classification ,Mutation ,HIV-1 ,biology.protein ,Protein folding - Abstract
Cyclophilins constitute a ubiquitous protein family whose functions include protein folding, transport and signaling. They possess both sequence-specific binding and proline cis-trans isomerase activities, as exemplified by the interaction between cyclophilin A (CypA) and the HIV-1 CA protein. Here, we report crystal structures of CypA in complex with HIV-1 CA protein variants that bind preferentially with the substrate proline residue in either the cis or the trans conformation. Cis- and trans-Pro substrates are accommodated within the enzyme active site by rearrangement of their N-terminal residues and with minimal distortions in the path of the main chain. CypA Arg55 guanidinium group probably facilitates catalysis by anchoring the substrate proline oxygen and stabilizing sp3 hybridization of the proline nitrogen in the transition state.
- Published
- 2003
- Full Text
- View/download PDF
17. Comprehensive Functional Maps of the Antigen-binding Site of an Anti-ErbB2 Antibody Obtained with Shotgun Scanning Mutagenesis
- Author
-
Camellia W. Adams, Sachdev S. Sidhu, Abraham M. de Vos, Timothy N Breece, Felix Vajdos, and Leonard G. Presta
- Subjects
Models, Molecular ,Receptor, ErbB-2 ,Molecular Sequence Data ,Mutagenesis (molecular biology technique) ,Enzyme-Linked Immunosorbent Assay ,Complementarity determining region ,Biology ,Crystallography, X-Ray ,Antibodies ,Epitope ,Antigen-Antibody Reactions ,Epitopes ,Peptide Library ,Structural Biology ,Point Mutation ,Genomic library ,Amino Acid Sequence ,Binding site ,Peptide library ,Molecular Biology ,Peptide sequence ,Gene Library ,Alanine ,Binding Sites ,Molecular biology ,Biochemistry ,Mutagenesis ,Protein Binding - Abstract
Shotgun scanning combinatorial mutagenesis was used to study the antigen-binding site of Fab2C4, a humanized monoclonal antibody fragment that binds to the extracellular domain of the human oncogene product ErbB2. Essentially all the residues in the Fab2C4 complementarity determining regions (CDRs) were alanine-scanned using phage-displayed libraries that preferentially allowed side-chains to vary as the wild-type or alanine. A separate homolog-scan was performed using libraries that allowed side-chains to vary only as the wild-type or a similar amino acid residue. Following binding selections to isolate functional clones, DNA sequencing was used to determine the wild-type/mutant ratios at each varied position, and these ratios were used to assess the contributions of each side-chain to antigen binding. The alanine-scan revealed that most of the side-chains that contribute to antigen binding are located in the heavy chain, and the Fab2C4 three-dimensional structure revealed that these residues fall into two groups. The first group consists of solvent-exposed residues which likely make energetically favorable contacts with the antigen and thus comprise the functional-binding epitope. The second group consists of buried residues with side-chains that pack against other CDR residues and apparently act as scaffolding to maintain the functional epitope in a binding-competent conformation. The homolog-scan involved subtle mutations, and as a result, only a subset of the side-chains that were intolerant to alanine substitutions were also intolerant to homologous substitutions. In particular, the 610 A2 functional epitope surface revealed by alanine-scanning shrunk to only 369 A2 when mapped with homologous substitutions, suggesting that this smaller subset of side-chains may be involved in more precise contacts with the antigen. The results validate shotgun scanning as a rapid and accurate method for determining the functional contributions of individual side-chains involved in protein-protein interactions.
- Published
- 2002
- Full Text
- View/download PDF
18. Crystal Structure of Human Insulin-like Growth Factor-1: Detergent Binding Inhibits Binding Protein Interactions
- Author
-
Kurt Deshayes, Mark Ultsch, Felix Vajdos, Nicholas J. Skelton, Jun Liu, A.M. de Vos, and M.L. Schaffer
- Subjects
Models, Molecular ,Crystallography ,Anomalous scattering ,Chemistry ,Binding protein ,Dimer ,Detergents ,Cholic Acids ,Plasma protein binding ,Crystal structure ,Biochemistry ,Recombinant Proteins ,Insulin-Like Growth Factor Binding Protein 1 ,chemistry.chemical_compound ,Insulin-Like Growth Factor Binding Protein 3 ,Chaps ,Biophysics ,Humans ,Molecule ,Insulin-Like Growth Factor I ,Linker ,Protein Binding - Abstract
Despite efforts spanning considerably more than a decade, a high-resolution view of the family of proteins known as insulin-like growth factors (IGFs) has remained elusive. IGF-1 consists of three helical segments which are connected by a 12-residue linker known as the C-region. NMR studies of members of this family reveal a dynamic structure with a topology resembling insulin but little structural definition in the C-region. We have crystallized IGF-1 in the presence of the detergent deoxy big CHAPS, and determined its structure at 1.8 A resolution by multiwavelength anomalous diffraction, exploiting the anomalous scattering of a single bromide ion and six of the seven sulfur atoms of IGF-1. The structure reveals a well-defined conformation for much of the C-region, which extends away from the core of IGF-1 and has residues known to be involved in receptor binding prominently displayed in a type II beta-turn. In the crystal, these residues form a dimer interface, but analytical ultracentrifugation experiments demonstrate that at physiological concentrations IGF-1 is monomeric. A single detergent molecule contacts residues known to be important for IGF-1 binding protein (IGFBP) interactions. Biophysical and biochemical data show that the detergent binds to IGF-1 specifically and blocks binding of IGFBP-1 and IGFBP-3.
- Published
- 2001
- Full Text
- View/download PDF
19. Crystal Structure of Human Cyclophilin A Bound to the Amino-Terminal Domain of HIV-1 Capsid
- Author
-
Sanghee Yoo, Megan Houseweart, T.R. Gamble, Wesley I. Sundquist, Christopher P. Hill, Felix Vajdos, and David K. Worthylake
- Subjects
Models, Molecular ,viruses ,Gene Products, gag ,Crystallography, X-Ray ,General Biochemistry, Genetics and Molecular Biology ,Cyclophilin A ,Capsid ,Prolyl isomerase ,Humans ,Cyclophilin ,Amino Acid Isomerases ,biology ,Biochemistry, Genetics and Molecular Biology(all) ,Binding protein ,Capsomere ,Virion ,Active site ,Peptidylprolyl Isomerase ,biochemical phenomena, metabolism, and nutrition ,Molecular biology ,Protein Structure, Tertiary ,HIV-1 ,biology.protein ,Biophysics ,TRIM5alpha ,Carrier Proteins ,Protein Binding - Abstract
The HIV-1 capsid protein forms the conical core structure at the center of the mature virion. Capsid also binds the human peptidyl prolyl isomerase, cyclophilin A, thereby packaging the enzyme into the virion. Cyclophilin A subsequently performs an essential function in HIV-1 replication, possibly helping to disassemble the capsid core upon infection. We report the 2.36 A crystal structure of the N-terminal domain of HIV-1 capsid (residues 1–151) in complex with human cyclophilin A. A single exposed capsid loop (residues 85–93) binds in the enzyme's active site, and Pro-90 adopts an unprecedented trans conformation. The structure suggests how cyclophilin A can act as a sequence-specific binding protein and a nonspecific prolyl isomerase. In the crystal lattice, capsid molecules assemble into continuous planar strips. Side by side association of these strips may allow capsid to form the surface of the viral core. Cyclophilin A could then function by weakening the association between capsid strips, thereby promoting disassembly of the viral core.
- Published
- 1996
- Full Text
- View/download PDF
20. How to measure and predict the molar absorption coefficient of a protein
- Author
-
Gerald R. Grimsley, T Gray, C. N. Pace, Felix Vajdos, and L. R. Fee
- Subjects
Chemical Phenomena ,Cystine ,Analytical chemistry ,1-Propanol ,Guanidines ,Biochemistry ,Propanol ,Absorbance ,chemistry.chemical_compound ,Urea ,Guanidine ,Molecular Biology ,Chromatography ,Chemistry, Physical ,Tryptophan ,Proteins ,Water ,Molar absorptivity ,chemistry ,Solvents ,Tyrosine ,Spectrophotometry, Ultraviolet ,Research Article - Abstract
The molar absorption coefficient, epsilon, of a protein is usually based on concentrations measured by dry weight, nitrogen, or amino acid analysis. The studies reported here suggest that the Edelhoch method is the best method for measuring epsilon for a protein. (This method is described by Gill and von Hippel [1989, Anal Biochem 182:319-326] and is based on data from Edelhoch [1967, Biochemistry 6:1948-1954]). The absorbance of a protein at 280 nm depends on the content of Trp, Tyr, and cystine (disulfide bonds). The average epsilon values for these chromophores in a sample of 18 well-characterized proteins have been estimated, and the epsilon values in water, propanol, 6 M guanidine hydrochloride (GdnHCl), and 8 M urea have been measured. For Trp, the average epsilon values for the proteins are less than the epsilon values measured in any of the solvents. For Tyr, the average epsilon values for the proteins are intermediate between those measured in 6 M GdnHCl and those measured in propanol. Based on a sample of 116 measured epsilon values for 80 proteins, the epsilon at 280 nm of a folded protein in water, epsilon (280), can best be predicted with this equation: epsilon (280) (M-1 cm-1) = (#Trp)(5,500) + (#Tyr)(1,490) + (#cystine)(125) These epsilon (280) values are quite reliable for proteins containing Trp residues, and less reliable for proteins that do not. However, the Edelhoch method is convenient and accurate, and the best approach is to measure rather than predict epsilon.
- Published
- 1995
- Full Text
- View/download PDF
21. 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
- Author
-
Jane M. Withka, Ivan Viktorovich Efremov, Jianhua Liu, Alison H. Varghese, Felix Vajdos, Kevin Ogilvie, Kris A. Borzilleri, Michael Aaron Brodney, Claude Ambroise, Gabriela Barreiro, Eva Hajos-Korcsok, Lorraine F. Lanyon, Curt Christoffersen, Christine E. Oborski, Katherine Fisher, Charles E. Nolan, John C. Murray, and Brian T. O’Neill
- Subjects
Drug ,Male ,Models, Molecular ,media_common.quotation_subject ,In silico ,ATP-binding cassette transporter ,Pharmacology ,Crystallography, X-Ray ,Transfection ,Permeability ,Madin Darby Canine Kidney Cells ,chemistry.chemical_compound ,Mice ,Structure-Activity Relationship ,Dogs ,Alzheimer Disease ,Drug Discovery ,Structure–activity relationship ,Animals ,Aspartic Acid Endopeptidases ,Humans ,Spiro Compounds ,ATP Binding Cassette Transporter, Subfamily B, Member 1 ,IC50 ,Sulfamide ,media_common ,Mice, Knockout ,Aza Compounds ,Sulfonamides ,Amyloid beta-Peptides ,Molecular Structure ,Chemistry ,Brain ,Stereoisomerism ,Drug Design ,Knockout mouse ,Microsomes, Liver ,Molecular Medicine ,Female ,Efflux ,Amyloid Precursor Protein Secretases - Abstract
β-Secretase 1 (BACE-1) is an attractive therapeutic target for the treatment and prevention of Alzheimer's 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.
- Published
- 2012
22. Discovery and optimization of a novel spiropyrrolidine inhibitor of β-secretase (BACE1) through fragment-based drug design
- Author
-
Jason K. Dutra, Jayvardhan Pandit, Hong Wang, Mahmoud N. Mansour, Christine E. Oborski, Binqing Wei, Alexander S. McColl, Theresa J. O’Sullivan, Steven W. Goldstein, Steven Capetta, Stephen Noell, Jane M. Withka, Dorff Peter H, Hou Chen, Thomas N. O'Connell, Felix Vajdos, Ivan Viktorovich Efremov, and Kris A. Borzilleri
- Subjects
Drug ,Models, Molecular ,Indoles ,Pyrrolidines ,Stereochemistry ,media_common.quotation_subject ,Stereoisomerism ,Crystallography, X-Ray ,chemistry.chemical_compound ,Structure-Activity Relationship ,Fragment (logic) ,Drug Discovery ,Structure–activity relationship ,Aspartic Acid Endopeptidases ,Humans ,Spiro Compounds ,media_common ,chemistry.chemical_classification ,Ligand efficiency ,Molecular Structure ,Chemistry ,Enzyme ,Biochemistry ,Drug Design ,β secretase ,Molecular Medicine ,Amyloid Precursor Protein Secretases ,Lead compound - Abstract
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 Alzheimer’s 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 Pfizer’s 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.
- Published
- 2012
23. Structure-guided Inhibitor Design for Human Acetyl-coenzyme A Carboxylase by Interspecies Active Site Conversion
- Author
-
James H. Harwood, Allan R. Reyes, Jeffrey W. Corbett, Kenneth J. DiRico, James A. Landro, Erick Marr, Marie Anderson, Meihua Tu, Francis Rajamohan, and Felix Vajdos
- Subjects
Saccharomyces cerevisiae Proteins ,Saccharomyces cerevisiae ,Spodoptera ,Crystallography, X-Ray ,Biochemistry ,Cell Line ,Structure-Activity Relationship ,Species Specificity ,Catalytic Domain ,Structure–activity relationship ,Animals ,Humans ,Enzyme Inhibitors ,Site-directed mutagenesis ,Molecular Biology ,chemistry.chemical_classification ,biology ,Acetyl-CoA carboxylase ,Active site ,Molecular Bases of Disease ,Cell Biology ,biology.organism_classification ,Yeast ,Enzyme ,Structural biology ,chemistry ,Structural Homology, Protein ,Drug Design ,biology.protein ,Acetyl-CoA Carboxylase - Abstract
Inhibition of acetyl-CoA carboxylases (ACCs), a crucial enzyme for fatty acid metabolism, has been shown to promote fatty acid oxidation and reduce body fat in animal models. Therefore, ACCs are attractive targets for structure-based inhibitor design, particularly the carboxyltransferase (CT) domain, which is the primary site for inhibitor interaction. We have cloned, expressed, and purified the CT domain of human ACC2 using baculovirus-mediated insect cell expression system. However, attempts to crystallize the human ACC2 CT domain have not been successful in our hands. Hence, we have been using the available crystal structure of yeast CT domain to design human ACC inhibitors. Unfortunately, as the selectivity of the lead series has increased against the full-length human enzyme, the potency against the yeast enzyme has decreased significantly. This loss of potency against the yeast enzyme correlated with a complete lack of binding of the human-specific compounds to crystals of the yeast CT domain. Here, we address this problem by converting nine key active site residues of the yeast CT domain to the corresponding human residues. The resulting humanized yeast ACC-CT (yCT-H9) protein exhibits biochemical and biophysical properties closer to the human CT domain and binding to human specific compounds. We report high resolution crystal structures of yCT-H9 complexed with inhibitors that show a preference for the human CT domain. These structures offer insights that explain the species selectivity of ACC inhibitors and may guide future drug design programs.
- Published
- 2011
24. P3‐282: β‐secretase inhibitors for treatment of Alzheimer's disease
- Author
-
Eva Hajos-Korcsok, Karen J. Coffman, Claude Ambroise, Michael Aaron Brodney, Christopher John Helal, Kris A. Borzilleri, Kevin Ogilvie, Gregory W. Kauffman, John D. Murray, Stephen Noell, Dane R. Liston, Luis Martinez-Alsina, Christine E. Oborski, Jane M. Withka, Lorraine F. Lanyon, JianHua Liu, Felix Vajdos, Ivan Viktorovich Efremov, Jamison B. Tuttle, Brian T. O’Neill, Yasong Lu, Frederick R. Nelson, and Charles E. Nolan
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,business.industry ,Health Policy ,β secretase ,Medicine ,Neurology (clinical) ,Disease ,Geriatrics and Gerontology ,Pharmacology ,business - Published
- 2010
- Full Text
- View/download PDF
25. Structure-Based Design of BACE Inhibitors: Technical and Practical Aspects of Preparation, 3-Dimensional Structure, and Computational Analysis
- Author
-
Alfredo G. Tomasselli, Veerabahu Shanmugasundaram, and Felix Vajdos
- Subjects
Theoretical computer science ,Computer science ,Structure (category theory) ,Structure based ,Computational analysis ,Combinatorial chemistry - Published
- 2010
- Full Text
- View/download PDF
26. Discovery of small molecule isozyme non-specific inhibitors of mammalian acetyl-CoA carboxylase 1 and 2
- Author
-
H. James Harwood, R. Kirk McPherson, Susan Tapley, Leanne M. Buzon, Kevin Daniel Freeman-Cook, Steven B. Coffey, Richard L. Elliott, Darcy Kohls, Liang Tong, Felix Vajdos, Meihua Tu, Eliot Sugarman, Shawn D. Doran, Francis Rajamohan, Eric S. Marr, Hailong Zhang, Wei Song, Kenneth J. DiRico, Jeffrey W. Corbett, Janet A. Houser, Sharad B. Murdande, William P. Esler, Martha L. Minich, and Christopher J. Jones
- Subjects
Models, Molecular ,Stereochemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Biochemistry ,Isozyme ,Small Molecule Libraries ,Structure-Activity Relationship ,Drug Discovery ,Transferase ,Animals ,Humans ,Enzyme Inhibitors ,Molecular Biology ,biology ,Drug discovery ,Chemistry ,Organic Chemistry ,Acetyl-CoA carboxylase ,Acetyl-CoA Carboxylase 1 ,Small molecule ,Pyruvate carboxylase ,Rats ,Isoenzymes ,Enzyme inhibitor ,biology.protein ,Molecular Medicine ,Acetyl-CoA Carboxylase - Abstract
Screening Pfizer's 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.
- Published
- 2009
27. Antitumor activity and pharmacology of a selective focal adhesion kinase inhibitor, PF-562,271
- Author
-
Susan Deborah Lagreca, Daniel Tyler Richter, Kevin Coleman, Matt Wessel, John Charles Kath, Catherine Angela Hulford, Martin A. Berliner, Erika Schmitt, Walter Gregory Roberts, Jing Lin, Matt Griffor, Michael Joseph Luzzio, Eric S. Marr, Pamela Whalen, Christofer Autry, Beth Cooper, Jitesh P. Jani, Earling Emerson, Nandini Chaturbhai Patel, Luis Martinez-Alsina, Lili Yao, Marianne J. Lorenzen, Felix Vajdos, and Ethan Ung
- Subjects
Cancer Research ,Indoles ,PTK2 ,Mice, Nude ,Antineoplastic Agents ,Apoptosis ,Biology ,Focal adhesion ,Mice ,In vivo ,Cell Line, Tumor ,Animals ,Humans ,Phosphorylation ,Protein Kinase Inhibitors ,Sulfonamides ,Cell adhesion molecule ,Kinase ,Biological activity ,Xenograft Model Antitumor Assays ,Oncology ,Biochemistry ,Models, Chemical ,Focal Adhesion Protein-Tyrosine Kinases ,Cancer research ,Female ,Glioblastoma ,Tyrosine kinase - Abstract
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 IC50 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 IC50 of 5 nmol/L. PF-562,271 was evaluated against multiple kinases and displays >100× selectivity against a long list of nontarget kinases. PF-562,271 inhibits FAK phosphorylation in vivo in a dose-dependent fashion (calculated EC50 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. [Cancer Res 2008;68(6):1935–44]
- Published
- 2008
28. The 2.0 A crystal structure of the ERalpha ligand-binding domain complexed with lasofoxifene
- Author
-
Felix Vajdos, Dennis E. Danley, Kieran F. Geoghegan, Jayvardhan Pandit, Mark Ammirati, Peter K. LeMotte, Lise R. Hoth, and Samuel P. Simons
- Subjects
Selective Estrogen Receptor Modulators ,Pyrrolidines ,Tetrahydronaphthalenes ,Stereochemistry ,Estrogen receptor ,Plasma protein binding ,Crystallography, X-Ray ,Biochemistry ,Protein Structure, Secondary ,Article ,Coactivator ,medicine ,Molecular Biology ,Molecular Structure ,Chemistry ,Estrogen Receptor alpha ,Lasofoxifene ,Hydrogen Bonding ,Protein Structure, Tertiary ,Nuclear receptor ,Selective estrogen receptor modulator ,Salt bridge ,Estrogen receptor alpha ,hormones, hormone substitutes, and hormone antagonists ,medicine.drug ,Protein Binding - Abstract
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 alpha at a resolution of 2.0 A. 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 ERalpha/SERM recognition, and add a new dimension to efforts to rationalize differences between the biological activity profiles exhibited by these important pharmacological agents.
- Published
- 2007
29. Abstract A86: Design, synthesis, and SAR of focal adhesion kinase (FAK) inhibitors
- Author
-
Roberts, Walter Gregory, primary, Berliner, Martin, additional, Coleman, Kevin, additional, Emerson, Erling, additional, Griffor, Matt, additional, Hulford, Catherine, additional, Jani, Jitesh, additional, Kath, John, additional, LaGreca, Susan, additional, Lin, Jing, additional, Lorenzen, Marianne J., additional, Marr, Eric, additional, Martinez‐Alsina, Luis, additional, Patel, Nandini, additional, Richter, Daniel, additional, Roberts, Erika, additional, Autry, Christopher, additional, Ung, Ethan, additional, Felix, Vajdos, additional, Vetelino, Beth, additional, Wessel, Matthew, additional, Whalen, Pamela, additional, Xu, Huiping, additional, and Yao, Lili, additional
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.