33 results on '"Theodore O. Johnson"'
Search Results
2. Direct control of CAR T cells through small molecule-regulated antibodies
- Author
-
Pascua Edward Derrick, Barbra Sasu, Javier Chaparro-Riggers, Regina Lin, Robert T. Abraham, Spencer Park, Zea Melton, Xiaodi Deng, Theodore O. Johnson, Jaume Pons, Kevin Lindquist, Thomas Van Blarcom, Bijan Boldajipour, Christopher R. Kimberlin, and Yvonne S. L. Mak
- Subjects
0301 basic medicine ,Drug ,T-Lymphocytes ,media_common.quotation_subject ,Science ,Primary Cell Culture ,General Physics and Astronomy ,T-Cell Antigen Receptor Specificity ,Immunotherapy, Adoptive ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Antigens, Neoplasm ,Cell Line, Tumor ,Neoplasms ,Animals ,Humans ,Leukaemia ,Potency ,Cytotoxicity ,Synthetic biology ,X-ray crystallography ,media_common ,Receptors, Chimeric Antigen ,Cell therapies ,Multidisciplinary ,biology ,Chemistry ,General Chemistry ,Single-Domain Antibodies ,Combined Modality Therapy ,Xenograft Model Antitumor Assays ,Small molecule ,Tumor antigen ,Chimeric antigen receptor ,HEK293 Cells ,Methotrexate ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Female ,Protein design ,Antibody ,Conjugate - Abstract
Antibody-based therapeutics have experienced a rapid growth in recent years and are now utilized in various modalities spanning from conventional antibodies, antibody-drug conjugates, bispecific antibodies to chimeric antigen receptor (CAR) T cells. Many next generation antibody therapeutics achieve enhanced potency but often increase the risk of adverse events. Antibody scaffolds capable of exhibiting inducible affinities could reduce the risk of adverse events by enabling a transient suspension of antibody activity. To demonstrate this, we develop conditionally activated, single-module CARs, in which tumor antigen recognition is directly modulated by an FDA-approved small molecule drug. The resulting CAR T cells demonstrate specific cytotoxicity of tumor cells comparable to that of traditional CARs, but the cytotoxicity is reversibly attenuated by the addition of the small molecule. The exogenous control of conditional CAR T cell activity allows continual modulation of therapeutic activity to improve the safety profile of CAR T cells across all disease indications., Many next-generation antibody therapeutics have enhanced potency but the risk of adverse events. Here the authors develop a conditionally activated, single-module CAR.
- Published
- 2021
3. Evaluation of a Published in Silico Model and Construction of a Novel Bayesian Model for Predicting Phospholipidosis Inducing Potential.
- Author
-
Dennis J. Pelletier, Daniel K. Gehlhaar, Anne Tilloy-Ellul, Theodore O. Johnson, and Nigel Greene
- Published
- 2007
- Full Text
- View/download PDF
4. Discovery of N-((3R,4R)-4-Fluoro-1-(6-((3-methoxy-1-methyl-1H-pyrazol-4-yl)amino)-9-methyl-9H-purin-2-yl)pyrrolidine-3-yl)acrylamide (PF-06747775) through Structure-Based Drug Design: A High Affinity Irreversible Inhibitor Targeting Oncogenic EGFR Mutants with Selectivity over Wild-Type EGFR
- Author
-
Chengyi Zhang, Sujin Cho-Schultz, Tran Khanh Tuan, Manli Shi, Rose Ann Ferre, Sherry Niessen, Sajiv Krishnan Nair, Douglas Carl Behenna, Dac M. Dinh, Elaine E. Tseng, Theodore O. Johnson, Cheng Hengmiao, Ru Zhou, Michael Zientek, T. Eric Ballard, Brion W. Murray, Suvi T. M. Orr, James Solowiej, Jennifer Lafontaine, Jean Joo Matthews, Scott L. Weinrich, Paolo Vicini, Deal Judith G, Longqing Liu, John Charles Kath, Pairish Mason Alan, Simon Paul Planken, Louise Bernier, Deepak Dalvie, Yiqin Luo, Martin Paul Edwards, Asako Nagata, Hong Shen, Neal W. Sach, Yuli Wang, Ketan S. Gajiwala, Shuibo Xin, Simon Bailey, Chau Almaden, Robert Steven Kania, and Michelle Hemkens
- Subjects
0301 basic medicine ,Mutation ,biology ,Chemistry ,Mutant ,Wild type ,medicine.disease_cause ,Molecular biology ,respiratory tract diseases ,03 medical and health sciences ,T790M ,030104 developmental biology ,Drug Discovery ,biology.protein ,medicine ,Molecular Medicine ,Kinome ,Epidermal growth factor receptor ,Erlotinib ,ADME ,medicine.drug - Abstract
Mutant epidermal growth factor receptor (EGFR) is a major driver of non-small-cell lung cancer (NSCLC). Marketed first generation inhibitors, such as erlotinib, effect a transient beneficial response in EGFR mutant NSCLC patients before resistance mechanisms render these inhibitors ineffective. Secondary oncogenic EGFR mutations account for approximately 50% of relapses, the most common being the gatekeeper T790M substitution that renders existing therapies ineffective. The discovery of PF-06459988 (1), an irreversible pyrrolopyrimidine inhibitor of EGFR T790M mutants, was recently disclosed.1 Herein, we describe our continued efforts to achieve potency across EGFR oncogenic mutations and improved kinome selectivity, resulting in the discovery of clinical candidate PF-06747775 (21), which provides potent EGFR activity against the four common mutants (exon 19 deletion (Del), L858R, and double mutants T790M/L858R and T790M/Del), selectivity over wild-type EGFR, and desirable ADME properties. Compound 21 is ...
- Published
- 2017
- Full Text
- View/download PDF
5. Discovery of 1-{(3R,4R)-3-[({5-Chloro-2-[(1-methyl-1H-pyrazol-4-yl)amino]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}oxy)methyl]-4-methoxypyrrolidin-1-yl}prop-2-en-1-one (PF-06459988), a Potent, WT Sparing, Irreversible Inhibitor of T790M-Containing EGFR Mutants
- Author
-
Marlena Walls, Tod Smeal, Suvi T. M. Orr, Zhengyu Liu, Cheng Hengmiao, Shuiwang Wang, Kephart Susan Elizabeth, Jean Joo Matthews, Rose Ann Ferre, Neal W. Sach, Scott L. Weinrich, Doug Behenna, Sherry Niessen, Sangita M. Baxi, Deepak Dalvie, Sujin Cho-Schultz, Dac M. Dinh, Kevin Ryan, Jim Solowiej, Elaine E. Tseng, Simon Paul Planken, Sajiv Krishnan Nair, Brion W. Murray, Jun Li Feng, Jennifer Lafontaine, Pairish Mason Alan, Shijian Ren, Michelle Hemkens, Shuibo Xin, Mehran Jalaie, Tran Khanh Tuan, Robert Steven Kania, Sutton Scott Channing, William F. Vernier, Kevin K.-C. Liu, Amy Jackson-Fisher, Beth Lunney, Min-Jean Yin, Ketan S. Gajiwala, Asako Nagata, Haiwei Xu, Michael Zientek, Ru Zhou, Daniel Tyler Richter, Simon Bailey, Martin Paul Edwards, Martha A. Ornelas, Chau Almaden, John Charles Kath, Hong Shen, and Theodore O. Johnson
- Subjects
0301 basic medicine ,Mutation ,010405 organic chemistry ,Stereochemistry ,Chemistry ,Mutant ,medicine.disease_cause ,01 natural sciences ,respiratory tract diseases ,0104 chemical sciences ,03 medical and health sciences ,T790M ,030104 developmental biology ,Gefitinib ,Protein kinase domain ,Drug Discovery ,Cancer research ,medicine ,Molecular Medicine ,Potency ,Reactivity (chemistry) ,Erlotinib ,medicine.drug - Abstract
First generation EGFR TKIs (gefitinib, erlotinib) provide significant clinical benefit for NSCLC cancer patients with oncogenic EGFR mutations. Ultimately, these patients’ disease progresses, often driven by a second-site mutation in the EGFR kinase domain (T790M). Another liability of the first generation drugs is severe adverse events driven by inhibition of WT EGFR. As such, our goal was to develop a highly potent irreversible inhibitor with the largest selectivity ratio between the drug-resistant double mutants (L858R/T790M, Del/T790M) and WT EGFR. A unique approach to develop covalent inhibitors, optimization of reversible binding affinity, served as a cornerstone of this effort. PF-06459988 was discovered as a novel, third generation irreversible inhibitor, which demonstrates (i) high potency and specificity to the T790M-containing double mutant EGFRs, (ii) minimal intrinsic chemical reactivity of the electrophilic warhead, (iii) greatly reduced proteome reactivity relative to earlier irreversible E...
- Published
- 2016
- Full Text
- View/download PDF
6. Resistance to lorlatinib in ROS1 fusion-positive non-small cell lung cancer
- Author
-
Jochen K. Lennerz, Jessica J. Lin, Theodore O. Johnson, Charlotte E. Lee, Justin F. Gainor, Beow Y. Yeap, Harper Hubbeling, Ibiayi Dagogo-Jack, and Alice T. Shaw
- Subjects
Cancer Research ,medicine.drug_class ,business.industry ,ROS1 Fusion Positive ,medicine.disease ,Lorlatinib ,Tyrosine-kinase inhibitor ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,medicine ,ROS1 ,Cancer research ,Non small cell ,business ,Lung cancer ,030215 immunology - Abstract
9611 Background: Lorlatinib is a potent, brain-penetrant ROS1/ALK tyrosine kinase inhibitor (TKI), which has demonstrated efficacy in advanced ROS1 fusion-positive (ROS1+) non-small cell lung cancer (NSCLC), including in patients (pts) previously treated with crizotinib. Despite initial benefit, however, most pts experience disease progression on lorlatinib. Mechanisms of resistance to lorlatinib in ROS1+ NSCLC are poorly understood. Methods: We analyzed repeat tumor biopsies derived from advanced ROS1+ lung cancer pts progressing on lorlatinib. Next-generation sequencing (NGS, n = 17) or whole exome sequencing (n = 1) was performed to detect mutations, indels, and copy number alterations. Results: Sixteen pts underwent a total of 18 repeat tumor biopsies after progression on lorlatinib. Fourteen had received prior crizotinib; two received prior crizotinib and entrectinib. Median duration of therapy on lorlatinib was 13.5 months (95% CI, 8.3-18.4). Among the 18 cases analyzed by sequencing, 7 (38.9%) harbored a ROS1 resistance mutation, including G2032R (4/18, 22.2%), S1986F/L2000V (1/18, 5.6%), L2086F (1/18, 5.6%), and G2032R/S1986F/L2086F (1/18, 5.6%). Of note, ROS1 L2086F was a novel resistance mutation not previously reported in the literature, but analogous to ALK L1256F (a lorlatinib-resistant ALK mutation). Structural modeling studies showed that ROS1 L2086F causes steric interference with binding of lorlatinib, as well as crizotinib and entrectinib. In addition to ROS1 kinase domain mutations, NGS analyses also identified MET copy number gain in a lorlatinib-resistant case, validated by fluorescence in situ hybridization as high-level focal MET amplification (MET/CEP7 copy number ratio 6.3) without a concomitant ROS1 resistance mutation. Duration of therapy on lorlatinib was significantly shorter in pts with a post-lorlatinib ROS1 resistance mutation compared to those without (8.3 vs 18.1 months; p = 0.005). Conclusions: ROS1 resistance mutations are observed in over one-third of cases progressing on lorlatinib, including the solvent front mutation G2032R and a novel L2086F mutation. These findings underscore the importance of developing next-generation ROS1 TKIs with activity against ROS1 mutations, and the need to elucidate ROS1-independent resistance mechanisms.
- Published
- 2020
- Full Text
- View/download PDF
7. Discovery of N-((3R,4R)-4-Fluoro-1-(6-((3-methoxy-1-methyl-1H-pyrazol-4-yl)amino)-9-methyl-9H-purin-2-yl)pyrrolidine-3-yl)acrylamide (PF-06747775) through Structure-Based Drug Design: A High Affinity Irreversible Inhibitor Targeting Oncogenic EGFR Mutants with Selectivity over Wild-Type EGFR
- Author
-
Simon, Planken, Douglas C, Behenna, Sajiv K, Nair, Theodore O, Johnson, Asako, Nagata, Chau, Almaden, Simon, Bailey, T Eric, Ballard, Louise, Bernier, Hengmiao, Cheng, Sujin, Cho-Schultz, Deepak, Dalvie, Judith G, Deal, Dac M, Dinh, Martin P, Edwards, Rose Ann, Ferre, Ketan S, Gajiwala, Michelle, Hemkens, Robert S, Kania, John C, Kath, Jean, Matthews, Brion W, Murray, Sherry, Niessen, Suvi T M, Orr, Mason, Pairish, Neal W, Sach, Hong, Shen, Manli, Shi, James, Solowiej, Khanh, Tran, Elaine, Tseng, Paolo, Vicini, Yuli, Wang, Scott L, Weinrich, Ru, Zhou, Michael, Zientek, Longqing, Liu, Yiqin, Luo, Shuibo, Xin, Chengyi, Zhang, and Jennifer, Lafontaine
- Subjects
Models, Molecular ,Acrylamides ,Lung Neoplasms ,Pyrrolidines ,Halogenation ,Rats ,ErbB Receptors ,Molecular Docking Simulation ,Mice ,Dogs ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Drug Design ,Mutation ,Animals ,Humans ,Lung ,Protein Kinase Inhibitors - Abstract
Mutant epidermal growth factor receptor (EGFR) is a major driver of non-small-cell lung cancer (NSCLC). Marketed first generation inhibitors, such as erlotinib, effect a transient beneficial response in EGFR mutant NSCLC patients before resistance mechanisms render these inhibitors ineffective. Secondary oncogenic EGFR mutations account for approximately 50% of relapses, the most common being the gatekeeper T790M substitution that renders existing therapies ineffective. The discovery of PF-06459988 (1), an irreversible pyrrolopyrimidine inhibitor of EGFR T790M mutants, was recently disclosed.1 Herein, we describe our continued efforts to achieve potency across EGFR oncogenic mutations and improved kinome selectivity, resulting in the discovery of clinical candidate PF-06747775 (21), which provides potent EGFR activity against the four common mutants (exon 19 deletion (Del), L858R, and double mutants T790M/L858R and T790M/Del), selectivity over wild-type EGFR, and desirable ADME properties. Compound 21 is currently being evaluated in phase-I clinical trials of mutant EGFR driven NSCLC.
- Published
- 2017
8. A roadmap to evaluate the proteome-wide selectivity of covalent kinase inhibitors
- Author
-
John Charles Kath, Theodore O. Johnson, Landon R. Whitby, Benjamin F. Cravatt, Baoxian Wei, Chu Wang, Mark E. Schnute, Melissa M. Dix, Lee R. Roberts, Laurence O. Whiteley, Adam M. Gilbert, Jonathan J. Hulce, Sherry Niessen, Erik C. Hett, Chris Joslyn, Matthew Merrill Hayward, Bryan R. Lanning, and John Douhan
- Subjects
Proteome ,Cell Survival ,Proteomics ,01 natural sciences ,Article ,03 medical and health sciences ,Piperidines ,Cell Line, Tumor ,Agammaglobulinaemia Tyrosine Kinase ,Humans ,Cysteine ,Molecular Biology ,Protein Kinase Inhibitors ,030304 developmental biology ,0303 health sciences ,biology ,010405 organic chemistry ,Kinase ,Drug discovery ,Adenine ,Active site ,Cell Biology ,Genes, erbB-1 ,Protein-Tyrosine Kinases ,Small molecule ,0104 chemical sciences ,Kinetics ,Pyrimidines ,Biochemistry ,Covalent bond ,biology.protein ,Pyrazoles ,Signal transduction ,Protein Kinases ,Signal Transduction - Abstract
Kinases are principal components of signal transduction pathways and the focus of intense basic and drug discovery research. Irreversible inhibitors that covalently modify non-catalytic cysteines in kinase active-sites have emerged as valuable probes and approved drugs. Many protein classes, however, possess functional cysteines and therefore understanding the proteome-wide selectivity of covalent kinase inhibitors is imperative. Here, we accomplish this objective using activity-based protein profiling coupled with quantitative mass spectrometry to globally map the targets, both specific and non-specific, of covalent kinase inhibitors in human cells. Many of the specific off-targets represent non-kinase proteins that, interestingly, possess conserved, active-site cysteines. We define windows of selectivity for covalent kinase inhibitors and show that, when these windows are exceeded, rampant proteome-wide reactivity and kinase target-independent cell death conjointly occur. Our findings, taken together, provide an experimental roadmap to illuminate opportunities and surmount challenges for the development of covalent kinase inhibitors.
- Published
- 2014
9. Pyrimidone-based series of glucokinase activators with alternative donor–acceptor motif
- Author
-
Ketan S. Gajiwala, Dongxiang Zeng, Jeffrey A. Pfefferkorn, Francis Bourbonais, Christopher S. Jones, Michael J. Hickey, Christian Perreault, Paul S. Humphries, Robert John Maguire, Jianwei Bian, Shenping Liu, Kevin J. Filipski, Mary Theresa Didiuk, Meihua Tu, Gary Erik Aspnes, David R. Derksen, Robert L. Dow, Angel Guzman-Perez, Richard F. Hank, Theodore O. Johnson, John Litchfield, John D. Knafels, and Karen Atkinson
- Subjects
Models, Molecular ,Stereochemistry ,Amino Acid Motifs ,Clinical Biochemistry ,Allosteric regulation ,Substituent ,Enzyme Activators ,Pharmaceutical Science ,Pyrimidinones ,Biochemistry ,Enzyme activator ,chemistry.chemical_compound ,Allosteric Regulation ,Amide ,Glucokinase ,Drug Discovery ,Animals ,Moiety ,Pyrimidone ,Binding site ,Molecular Biology ,Binding Sites ,Organic Chemistry ,Rats ,chemistry ,Molecular Medicine - Abstract
Glucokinase activators are a class of experimental agents under investigation as a therapy for Type 2 diabetes mellitus. An X-ray crystal structure of a modestly potent agent revealed the potential to substitute the common heterocyclic amide donor–acceptor motif for a pyridone moiety. We have successfully demonstrated that both pyridone and pyrimidone heterocycles can be used as a potent donor–acceptor substituent. Several sub-micromolar analogs that possess the desired partial activator profile were synthesized and characterized. Unfortunately, the most potent activators suffered from sub-optimal pharmacokinetic properties. Nonetheless, these donor–acceptor motifs may find utility in other glucokinase activator series or beyond.
- Published
- 2013
- Full Text
- View/download PDF
10. N-(Pyridin-2-yl) arylsulfonamide inhibitors of 11β-hydroxysteroid dehydrogenase type 1: Strategies to eliminate reactive metabolites
- Author
-
Deepak Dalvie, Yong Wang, Theodore O. Johnson, Michael Siu, Cripps Stephan James, Klaus Ruprecht Dress, Jacqui Elizabeth Hoffman, Taylor Wendy Dianne, Stanley William Kupchinsky, Ping Kang, Sue Zhou, Jacques Ermolieff, Andrea Fanjul, Sajiv Krishnan Nair, Arturo Castro, Jennifer E. Vogel, Jean Joo Matthews, Amy LaPaglia, Cheng Hengmiao, Paul A. Rejto, Christopher Ronald Smith, Martin Paul Edwards, and Natilie Hosea
- Subjects
Stereochemistry ,Clinical Biochemistry ,Aminopyridines ,Pharmaceutical Science ,Dehydrogenase ,Biochemistry ,Structure-Activity Relationship ,chemistry.chemical_compound ,Pharmacokinetics ,11β-hydroxysteroid dehydrogenase type 1 ,11-beta-Hydroxysteroid Dehydrogenase Type 1 ,Drug Discovery ,Humans ,Structure–activity relationship ,Molecular Biology ,Sulfonamides ,biology ,Chemistry ,Organic Chemistry ,HEK 293 cells ,Glutathione ,HEK293 Cells ,Reactive metabolite ,Risk-benefit analysis ,biology.protein ,Molecular Medicine - Abstract
N-(Pyridin-2-yl) arylsulfonamides 1 and 2 (PF-915275) were identified as potent inhibitors of 11β-hydroxysteroid dehydrogenase type 1. A screen for bioactivation revealed that these compounds formed glutathione conjugates. This communication presents the results of a risk benefit analysis carried out to progress 2 (PF-915275) to a clinical study and the strategies used to eliminate reactive metabolites in this series of inhibitors. Based on the proposed mechanism of bioactivation and structure-activity relationships, design efforts led to N-(pyridin-2-yl) arylsulfonamides such as 18 and 20 that maintained potent 11β-hydroxysteroid dehydrogenase type 1 activity, showed exquisite pharmacokinetic profiles, and were negative in the reactive metabolite assay.
- Published
- 2013
- Full Text
- View/download PDF
11. Discovery of the Highly Potent PI3K/mTOR Dual Inhibitor PF-04979064 through Structure-Based Drug Design
- Author
-
Simon Bailey, Cheng Hengmiao, Marlena Walls, Matthew A. Marx, Kevin K.-C. Liu, Jacqui Elizabeth Hoffman, Zhengyu Liu, Jinjiang Zhu, Lisa Guo, John Li, Lance Goulet, Peter A. Wells, Chunze Li, Ying Jiang, Daniel R. Knighton, Min-Jean Yin, Ted William Johnson, Michael Zientek, Sangita M. Baxi, and Theodore O. Johnson
- Subjects
Drug ,chemistry.chemical_classification ,Kinase ,business.industry ,media_common.quotation_subject ,Organic Chemistry ,Pharmacology ,Biochemistry ,In vitro ,chemistry ,Drug Discovery ,Potency ,Medicine ,business ,Protein kinase B ,Aldehyde oxidase ,PI3K/AKT/mTOR pathway ,media_common ,Tricyclic - Abstract
PI3K, AKT, and mTOR are key kinases from PI3K signaling pathway being extensively pursued to treat a variety of cancers in oncology. To search for a structurally differentiated back-up candidate to PF-04691502, which is currently in phase I/II clinical trials for treating solid tumors, a lead optimization effort was carried out with a tricyclic imidazo[1,5]naphthyridine series. Integration of structure-based drug design and physical properties-based optimization yielded a potent and selective PI3K/mTOR dual kinase inhibitor PF-04979064. This manuscript discusses the lead optimization for the tricyclic series, which both improved the in vitro potency and addressed a number of ADMET issues including high metabolic clearance mediated by both P450 and aldehyde oxidase (AO), poor permeability, and poor solubility. An empirical scaling tool was developed to predict human clearance from in vitro human liver S9 assay data for tricyclic derivatives that were AO substrates.
- Published
- 2012
- Full Text
- View/download PDF
12. Characterization of the CHK1 Allosteric Inhibitor Binding Site
- Author
-
Chen Ping, Ya-Li Deng, Chun Luo, Darin Vanderpool, Terri Quenzer, Theodore O. Johnson, Steve Margosiak, James Register, Edward L. Brown, Soneprasith Phonephaly, Gordon Alton, Simon Bergqvist, Stephan Grant, Jacques Ermolieff, and Eugene Rui
- Subjects
Models, Molecular ,animal structures ,Protein Conformation ,DNA damage ,Allosteric regulation ,Crystallography, X-Ray ,environment and public health ,Biochemistry ,Allosteric Regulation ,Catalytic Domain ,Humans ,Transferase ,CHEK1 ,Cloning, Molecular ,Enzyme Inhibitors ,Binding site ,Binding Sites ,biology ,Kinase ,Chemistry ,Surface Plasmon Resonance ,Small molecule ,Peptide Fragments ,Kinetics ,enzymes and coenzymes (carbohydrates) ,Allosteric enzyme ,Checkpoint Kinase 1 ,biology.protein ,biological phenomena, cell phenomena, and immunity ,Protein Kinases - Abstract
Checkpoint kinase 1 (CHK1) is a key element in the DNA damage response pathway and plays a crucial role in the S-G(2)-phase checkpoint. Inhibiting CHK1 is a therapeutic strategy involving abrogation of the G2/M mitotic checkpoint defense of tumor cells toward lethal damage induced by DNA-directed chemotherapeutic agents. To date, most CHK1 inhibition approaches have involved targeting the ATP site of this kinase. In this study, we provide crystallographic and kinetic characterization of two small molecule inhibitors that bind to an allosteric site in the proximity of the CHK1 substrate site. Analysis of kinetic and biophysical data has led to the conclusion that these small molecule allosteric site inhibitors of CHK1 are reversible and are neither ATP- nor peptide substrate-competitive. K(i) values of 1.89 and 0.15 microM, respectively, have been determined for these compounds using a mixed inhibitor kinetic analysis. Cocrystal structures of the inhibitors bound to CHK1 reveal an allosteric site, unique to CHK1, located in the C-terminal domain and consisting of a shallow groove linked to a small hydrophobic pocket. The pocket displays induced fit characteristics in the presence of the two inhibitors. These findings establish the potential for the development of highly selective CHK1 inhibitors.
- Published
- 2009
- Full Text
- View/download PDF
13. N-(Pyridin-2-yl) arylsulfonamide inhibitors of 11β-hydroxysteroid dehydrogenase type 1: Discovery of PF-915275
- Author
-
Cripps Stephan James, Thomas A. Pauly, Natilie Hosea, Martin Paul Edwards, Sajiv Krishnan Nair, Arturo Castro, Jacques Ermolieff, Theodore O. Johnson, Andrea Fanjul, Jean Joo Matthews, Yong Wang, Michael Siu, Amy LaPaglia, Jennifer E. Vogel, and Taylor Wendy Dianne
- Subjects
Clinical Biochemistry ,Aminopyridines ,Pharmaceutical Science ,Dehydrogenase ,Crystallography, X-Ray ,Biochemistry ,Isozyme ,Cell Line ,Structure-Activity Relationship ,Cricetinae ,11-beta-Hydroxysteroid Dehydrogenase Type 1 ,Drug Discovery ,medicine ,Animals ,Humans ,Computer Simulation ,Enzyme Inhibitors ,Molecular Biology ,chemistry.chemical_classification ,Sulfonamides ,Organic Chemistry ,Biological activity ,Ligand (biochemistry) ,Rats ,Enzyme ,chemistry ,Lipophilic efficiency ,Drug Design ,Molecular Medicine ,Cortisone ,hormones, hormone substitutes, and hormone antagonists ,Glucocorticoid ,medicine.drug - Abstract
N-(Pyridin-2-yl) arylsulfonamides are identified as inhibitors of 11β-hydroxysteroid dehydrogenase type 1 (11βHSD1), an enzyme that catalyzes the reduction of the glucocorticoid cortisone to cortisol. Dysregulation of glucocorticoids has been implicated in the pathogenesis of diabetes and the metabolic syndrome. In this Letter, we present the development of an initial lead to an efficient ligand with improved physiochemical properties using a deletion strategy. This strategy allowed for further optimization of potency leading to the discovery of the clinical candidate PF-915275.
- Published
- 2009
- Full Text
- View/download PDF
14. Evaluation of a Published in Silico Model and Construction of a Novel Bayesian Model for Predicting Phospholipidosis Inducing Potential
- Author
-
Nigel Greene, Daniel K. Gehlhaar, Anne Tilloy-Ellul, Theodore O. Johnson, and Dennis J. Pelletier
- Subjects
Phospholipidosis ,Models, Statistical ,Chemistry ,business.industry ,Drug discovery ,Computer science ,General Chemical Engineering ,In silico ,Drug Evaluation, Preclinical ,Dosing regimen ,Bayes Theorem ,General Medicine ,General Chemistry ,Computational biology ,Library and Information Sciences ,Bioinformatics ,Bayesian inference ,Computer Science Applications ,Models, Chemical ,Preclinical testing ,Computer Simulation ,business ,Phospholipids ,Software ,Pharmaceutical industry - Abstract
The identification of phospholipidosis (PPL) during preclinical testing in animals is a recognized problem in the pharmaceutical industry. Depending on the intended indication and dosing regimen, PPL can delay or stop development of a compound in the drug discovery process. Therefore, for programs and projects where a PPL finding would have adverse impact on the success of the project, it would be desirable to be able to rapidly identify and screen out those compounds with the potential to induce PPL as early as possible. Currently, electron microscopy is the gold standard method for identifying phospholipidosis, but it is low-throughput and resource-demanding. Therefore, a low-cost, high-throughput screening strategy is required to overcome these limitations and be applicable in the drug discovery cycle. A recent publication by Ploemen et al. (Exp. Toxicol. Pathol. 2004, 55, 347-55) describes a method using the computed physicochemical properties pKa and ClogP as part of a simple calculation to determine a compound's potential to induce PPL. We have evaluated this method using a set of 201 compounds, both public and proprietary, with known in vivo PPL-inducing ability and have found the overall concordance to be 75%. We have proposed simple modifications to the model rules, which improve the model's concordance to 80%. Finally, we describe the development of a Bayesian model using the same compound set and found its overall concordance to be 83%.
- Published
- 2007
- Full Text
- View/download PDF
15. Effects of Activating Mutations on EGFR Cellular Protein Turnover and Amino Acid Recycling Determined Using SILAC Mass Spectrometry
- Author
-
Sherry Niessen, Michael J. Greig, Jun Li Feng, Manli Shi, Scott L. Weinrich, and Theodore O. Johnson
- Subjects
chemistry.chemical_classification ,Mutation ,biology ,Article Subject ,lcsh:Cytology ,Protein turnover ,Cell Biology ,Bioinformatics ,medicine.disease_cause ,Molecular biology ,Amino acid ,T790M ,chemistry ,Cell culture ,Stable isotope labeling by amino acids in cell culture ,medicine ,biology.protein ,Epidermal growth factor receptor ,lcsh:QH573-671 ,A431 cells ,Research Article - Abstract
Rapid mutations of proteins that are targeted in cancer therapy often lead to drug resistance. Often, the mutation directly affects a drug’s binding site, effectively blocking binding of the drug, but these mutations can have other effects such as changing the protein turnover half-life. Utilizing SILAC MS, we measured the cellular turnover rates of an important non-small cell lung cancer target, epidermal growth factor receptor (EGFR). Wild-type (WT) EGFR, EGFR with a single activating mutant (Del 746–750 or L858R), and the drug-resistant double mutant (L858R/T790M) EGFR were analyzed. In non-small cell lung cancer cell lines, EGFR turnover rates ranged from 28 hours in A431 cells (WT) to 7.5 hours in the PC-9 cells (Del 746–750 mutant). The measurement of EGFR turnover rate in PC-9 cells dosed with irreversible inhibitors has additional complexity due to inhibitor effects on cell viability and results were reported as a range. Finally, essential amino acid recycling (K and R) was measured in different cell lines. The recycling was different in each cell line, but the overall inclusion of the effect of amino acid recycling on calculating EGFR turnover rates resulted in a 10–20% reduction in rates.
- Published
- 2015
16. Protein tyrosine phosphatase 1B inhibitors for diabetes
- Author
-
Michael Robert Jirousek, Theodore O. Johnson, and Jacques Ermolieff
- Subjects
medicine.medical_specialty ,medicine.medical_treatment ,Phosphatase ,Protein tyrosine phosphatase ,Type 2 diabetes ,Structure-Activity Relationship ,Diabetes mellitus ,Internal medicine ,Drug Discovery ,Humans ,Medicine ,Obesity ,Enzyme Inhibitors ,Molecular Biology ,Protein Tyrosine Phosphatase, Non-Receptor Type 1 ,Pharmacology ,business.industry ,Leptin ,Insulin ,Type 2 Diabetes Mellitus ,General Medicine ,medicine.disease ,Endocrinology ,Diabetes Mellitus, Type 2 ,Protein Tyrosine Phosphatases ,business ,hormones, hormone substitutes, and hormone antagonists - Abstract
Increased incidence of type 2 diabetes mellitus and obesity has elevated the medical need for new agents to treat these disease states. Resistance to the hormones insulin and leptin are hallmarks of both type 2 diabetes and obesity. Drugs that can ameliorate this resistance should be effective in treating type 2 diabetes and possibly obesity. Protein tyrosine phosphatase 1B (PTP1B) is thought to function as a negative regulator of insulin and leptin signal transduction. This article reviews PTP1B as a novel target for type 2 diabetes, and looks at the challenges in developing small-molecule inhibitors of this phosphatase.
- Published
- 2002
- Full Text
- View/download PDF
17. Structure-Based Design, Synthesis, and Biological Evaluation of Irreversible Human Rhinovirus 3C Protease Inhibitors. Part 7: Structure–Activity Studies of Bicyclic 2-Pyridone-Containing Peptidomimetics
- Author
-
Ru Zhou, Theodore O. Johnson, Shella A. Fuhrman, David A. Matthews, Stephen T. Worland, Fausto Maldonado, Amy K. Patick, Leora S. Zalman, Thomas J. Prins, Peter S. Dragovich, Rose Ann Ferre, James W. Meador, Edward L. Brown, and Xinjun Hou
- Subjects
Rhinovirus ,Picornain 3C ,Pyridones ,medicine.drug_class ,Stereochemistry ,Peptidomimetic ,Clinical Biochemistry ,Pharmaceutical Science ,Carboxamide ,Cysteine Proteinase Inhibitors ,Antiviral Agents ,Biochemistry ,Chemical synthesis ,Cell Line ,Structure-Activity Relationship ,Viral Proteins ,stomatognathic system ,Drug Discovery ,medicine ,Humans ,Structure–activity relationship ,Serotyping ,Molecular Biology ,chemistry.chemical_classification ,biology ,Bicyclic molecule ,Chemistry ,Molecular Mimicry ,Organic Chemistry ,3C Viral Proteases ,virus diseases ,Bridged Bicyclo Compounds, Heterocyclic ,Cysteine Endopeptidases ,Enzyme ,Enzyme inhibitor ,Drug Design ,biology.protein ,Molecular Medicine - Abstract
The structure-based design, chemical synthesis, and biological evaluation of bicyclic 2-pyridone-containing human rhinovirus (HRV) 3C protease (3CP) inhibitors are described. An optimized compound is shown to exhibit antiviral activity when tested against a variety of HRV serotypes (EC(50)'s ranging from 0.037 to 0.162 microM).
- Published
- 2002
- Full Text
- View/download PDF
18. Characterization, biomarkers, and reversibility of a monoclonal antibody-induced immune complex disease in cynomolgus monkeys (Macaca fascicularis)
- Author
-
Sangeetha Bollini, Mark G. Evans, Nasir K. Khan, Walter F. Bobrowski, Thomas P. Brown, Allison Vitsky, Niraj Tripathi, Bora Han, John C. Lin, Jennifer L. Rojko, Jonathan R. Heyen, Theodore O. Johnson, and Shana Dalton
- Subjects
Male ,Drug-Related Side Effects and Adverse Reactions ,medicine.drug_class ,Interleukin-1beta ,Drug Evaluation, Preclinical ,Complement Membrane Attack Complex ,Biology ,Urinalysis ,Toxicology ,Monoclonal antibody ,Pathology and Forensic Medicine ,Interferon-gamma ,Immune system ,Monocytosis ,Microscopy, Electron, Transmission ,medicine ,Animals ,Immune Complex Diseases ,Type III hypersensitivity ,Molecular Biology ,Dose-Response Relationship, Drug ,Interleukin-6 ,Tumor Necrosis Factor-alpha ,Antibodies, Monoclonal ,Cell Biology ,medicine.disease ,Immunohistochemistry ,Neutrophilia ,Macaca fascicularis ,C-Reactive Protein ,Immunology ,biology.protein ,Female ,medicine.symptom ,Antibody ,Immune complex disease ,Biomarkers - Abstract
Two 6-month repeat-dose toxicity studies in cynomolgus monkeys illustrated immune complex–mediated adverse findings in individual monkeys and identified parameters that potentially signal the onset of immune complex–mediated reactions following administration of RN6G, a monoclonal antibody (mAb). In the first study, 3 monkeys exhibited nondose-dependent severe clinical signs accompanied by decreased erythrocytes with increased reticulocytes, neutrophilia, monocytosis, thrombocytopenia, coagulopathy, decreased albumin, azotemia, and increased serum levels of activated complement products, prompting unscheduled euthanasia. Histologically, immunohistochemical localization of RN6G was associated with monkey immunoglobulin and complement components in glomeruli and other tissues, attributable to immune complex disease (ICD). All 3 animals also had anti-RN6G antibodies and decreased plasma levels of RN6G. Subsequently, an investigational study was designed and conducted with regulatory agency input to detect early onset of ICD and assess reversibility to support further clinical development. Dosing of individual animals ceased when biomarkers of ICD indicated adverse findings. Of the 12 monkeys, 1 developed anti-RN6G antibodies and decreased RN6G exposure that preceded elevations in complement products, interleukin-6, and coagulation parameters and decreases in albumin and fibrinogen. All findings in this monkey, except for antidrug antibody (ADA), reversed after cessation of dosing without progressing to adverse sequelae typically associated with ICD.
- Published
- 2014
19. Identification of potent inhibitors of Plasmodium falciparum plasmepsin II from an encoded statine combinatorial library
- Author
-
Anna Oksman, Daniel E. Goldberg, Roland E. Dolle, Theodore O. Johnson, Daniel Chelsky, Marc Orlowski, Joan Guo, Carolyn DiIanni Carroll, Ilya Y. Gluzman, Tao Guo, Zhen-Min He, Cullen L. Cavallaro, Hitesh K. Patel, and James A. Connelly
- Subjects
Plasmodium falciparum ,Clinical Biochemistry ,Protozoan Proteins ,Pharmaceutical Science ,Biochemistry ,Substrate Specificity ,Apicomplexa ,Antimalarials ,Hemoglobins ,Structure-Activity Relationship ,chemistry.chemical_compound ,Plasmepsin II ,Peptide Library ,parasitic diseases ,Drug Discovery ,Statine ,Animals ,Aspartic Acid Endopeptidases ,Humans ,Structure–activity relationship ,Protease Inhibitors ,Amino Acids ,Cloning, Molecular ,Peptide library ,Molecular Biology ,chemistry.chemical_classification ,Molecular Structure ,biology ,Organic Chemistry ,biology.organism_classification ,Recombinant Proteins ,Enzyme ,Databases as Topic ,chemistry ,Enzyme inhibitor ,Drug Design ,biology.protein ,Molecular Medicine ,Oligopeptides - Abstract
An encoded 13,020-member combinatorial library was synthesized containing a statine core. Evaluation of this library with plasmepsin II, an aspartyl protease required for hemoglobin metabolism in the malaria parasite, led to the identification of potent and selective inhibitors as well as novel structure-activity relationships.
- Published
- 1998
- Full Text
- View/download PDF
20. ChemInform Abstract: Ring-Enlarging Cyclopentene Annulations
- Author
-
Theodore O. Johnson and Larry E. Overman
- Subjects
chemistry.chemical_compound ,Annulation ,chemistry ,Cyclopentene ,General Medicine ,Ring (chemistry) ,Medicinal chemistry - Abstract
Ethynylation of 2-[(2,2-dialkoxy)ethyl]cycloalkanones followed by Lewis acid-promoted rearrangement occasions cyclopentene annulation with concomitant one-carbon ring expansion of the starting cycloalkanone.
- Published
- 2010
- Full Text
- View/download PDF
21. 4-methylpteridinones as orally active and selective PI3K/mTOR dual inhibitors
- Author
-
Zhengyu Liu, Haitao Wang, Bernadette Pascual, Jing Yuan, Dan Knighton, Shubha Bagrodia, Simon Bailey, Graham L. Smith, Caroline Rodgers, Lisa Gao, Yang Anle, Cheng Hengmiao, Kevin K.-C. Liu, Jacqui Elizabeth Hoffman, Kristina Rafidi, Shaoxian Sun, Samantha Elizabeth Greasley, Aihua Zou, Sacha Ninkovic, Hui Chen, Nambu Mitchell David, Matthew A. Marx, Qiyue Hu, and Theodore O. Johnson
- Subjects
Models, Molecular ,Molecular model ,Clinical Biochemistry ,Pharmaceutical Science ,Administration, Oral ,Antineoplastic Agents ,Pharmacology ,Biochemistry ,Mice ,Phosphatidylinositol 3-Kinases ,Structure-Activity Relationship ,In vivo ,Oral administration ,Cell Line, Tumor ,Neoplasms ,Drug Discovery ,Potency ,Animals ,Humans ,Molecular Biology ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors ,biology ,Kinase ,Chemistry ,Pteridines ,TOR Serine-Threonine Kinases ,Organic Chemistry ,Pteridinones ,Glioma ,Solubility ,Enzyme inhibitor ,biology.protein ,Molecular Medicine - Abstract
Pteridinones were designed based on a non-selective kinase template. Because of the uniqueness of the PI3K and mTOR binding pockets, a methyl group was introduced to C-4 position of the peteridinone core to give compounds with excellent selectivity for PI3K and mTOR. This series of compounds were further optimized to improve their potency against PI3Kα and mTOR. Finally, orally active compounds with improved solubility and robust in vivo efficacy in tumor growth inhibition were identified as well.
- Published
- 2010
22. A strategy for risk management of drug-induced phospholipidosis
- Author
-
Daniel Morton, Theodore O. Johnson, Linda A. Chatman, and Susan D. Anway
- Subjects
Drug ,Drug Industry ,Drug-Related Side Effects and Adverse Reactions ,media_common.quotation_subject ,Metabolite ,Drug Evaluation, Preclinical ,Risk management tools ,Pharmacology ,Toxicology ,Lipidoses ,Pathology and Forensic Medicine ,chemistry.chemical_compound ,In vivo ,Medicine ,Humans ,Computer Simulation ,Molecular Biology ,Drug induced phospholipidosis ,Lung ,Risk management ,Phospholipids ,media_common ,Phospholipidosis ,Risk Management ,business.industry ,Cell Biology ,chemistry ,Toxicity ,business ,Lysosomes - Abstract
Drug-induced phospholipidosis (PL) is an excessive accumulation of phospholipids and drug in lysosomes. Phospholipidosis signals a change in cell membrane integrity and accumulation of intracellular drug or metabolite in tissues. The sensitivity and susceptibility of preclinical models to detect PL vary with therapeutic agents, and PL is expected to be reversible after discontinuation of drug treatment. The prevailing scientific opinion is that PL by itself is not adverse; however, some regulatory authorities consider PL to be adverse because a small number of chemicals are able to cause PL and concurrent organ toxicity. Until a greater understanding of PL emerges, a well-thought-out risk management strategy for PL will increase confidence in safety and improve selection and development of new drugs. This paper provides a tiered approach to risk management of drug-induced PL. It begins with use of in silico and in vitro tools to design and select compounds with reduced potential to produce PL. Early in vivo studies in two species are used to better characterize potential for toxicity and PL. Finally, routine risk management tools (i.e., translational biomarkers, assessment of reversibility) are used to support confidence in safety of compounds that induce PL in animals.
- Published
- 2009
23. Ring-enlarging cyclopentene annulations
- Author
-
Theodore O. Johnson and Larry E. Overman
- Subjects
Annulation ,Bicyclic molecule ,Stereochemistry ,Organic Chemistry ,Acetal ,Ring (chemistry) ,Biochemistry ,Medicinal chemistry ,chemistry.chemical_compound ,Acid catalysis ,chemistry ,Drug Discovery ,Cyclopentene ,Lewis acids and bases ,Enone - Abstract
Ethynylation of 2-[(2,2-dialkoxy)ethyl]cycloalkanones followed by Lewis acid-promoted rearrangement occasions cyclopentene annulation with concomitant one-carbon ring expansion of the starting cycloalkanone.
- Published
- 1991
- Full Text
- View/download PDF
24. Glucokinase Activators for the Treatment of Type 2 Diabetes
- Author
-
Paul S. Humphries and Theodore O. Johnson
- Subjects
chemistry.chemical_classification ,Regulation of gene expression ,Hexokinase ,Glycogen ,Glucokinase ,Insulin ,medicine.medical_treatment ,chemistry.chemical_compound ,Enzyme ,chemistry ,Biochemistry ,medicine ,Glucose homeostasis ,Adenosine triphosphate - Abstract
Publisher Summary This chapter discusses the mechanism of enzymatic catalysis by glucokinase (GK), its function in the liver and pancreatic β-cells, and key publications on small-molecule GK activators (GKAs) that report full characterization. Key compound disclosures from patents are also discussed. Studies have ascertained an essential function for GK, also known as hexokinase IV or hexokinase D, in regulating glucose homeostasis. GK is expressed mainly in the two tissues of high importance to glucose homeostasis and therapeutic strategies to treat type 2 diabetes (T2D), pancreatic β-cells, and hepatocytes. In the liver, GK functions as a high-capacity enzyme that removes glucose from the blood and helps it react with adenosine triphosphate (ATP) to form glucose-6-phosphate (G-6-P)—the first biosynthetic step in the conversion of glucose to its storage form, glycogen. In pancreatic β-cells, through its ability to sense glucose concentrations, GK operates as a glucose sensor to determine the threshold for glucose-stimulated insulin release (GSIR). To seek out small molecules that increase GK enzymatic activity, a library of 120,000 structurally diverse synthetic compounds are screened. A solitary hit was identified from this high-throughput screening (HTS) enzyme coupled assay, in the presence of GK regulatory protein (GKRP). Ensuing kinetic analysis, in the absence of GKRP, confirmed the capacity of the hit molecule to directly bind and activate GK, rather than activating by disrupting the interaction of GKRP with GK.
- Published
- 2006
- Full Text
- View/download PDF
25. Structure-based design, synthesis, and biological evaluation of irreversible human rhinovirus 3C protease inhibitors. 8. Pharmacological optimization of orally bioavailable 2-pyridone-containing peptidomimetics
- Author
-
Hiep T. Luu, Zhen Ping Wu, Theodore O. Johnson, Jia Liu, Bennett Chaplin Borer, Tove Tuntland, Junhua Tao, Caroline A. Lee, Fausto Maldonado, Amy K. Patick, Sylvie K. Sakata, Thomas J. Prins, Naresh K. Nayyar, Rose Ann Ferre, Michael Mohajeri, Peter W. Rose, Maha B. Kosa, David A. Matthews, Kevin Scott Mcgee, Leora S. Zalman, Steven Lee, Peter S. Dragovich, Ellen Y. Wu, Bo Liu, Shella A. Fuhrman, Ye Hua, Elena Z. Dovalsantos, Paul A. Rejto, Ru Zhou, Mark Christopher Guzman, Ming Guo, Edward L. Brown, Minerva R. Batugo, James W. Meador, Lijian Chen, Andreas Liese, Moran Terence Jarold, and Jean-Paul R. Gleeson
- Subjects
Male ,Magnetic Resonance Spectroscopy ,Picornain 3C ,Rhinovirus ,Peptidomimetic ,Stereochemistry ,Pyridones ,Biological Availability ,In Vitro Techniques ,Chemical synthesis ,Antiviral Agents ,Structure-Activity Relationship ,Viral Proteins ,Dogs ,Oral administration ,Drug Discovery ,Animals ,Humans ,Protease Inhibitors ,chemistry.chemical_classification ,biology ,3C Viral Proteases ,Biological activity ,Blood Proteins ,Blood proteins ,Cysteine Endopeptidases ,Macaca fascicularis ,Enzyme ,chemistry ,Solubility ,Enzyme inhibitor ,Drug Design ,biology.protein ,Hepatocytes ,Microsomes, Liver ,Molecular Medicine ,Indicators and Reagents ,Caco-2 Cells ,Half-Life ,Protein Binding - Abstract
The optimization of the pharmacokinetic performance of various 2-pyridone-containing human rhinovirus (HRV) 3C protease (3CP) inhibitors following oral administration to either beagle dogs or CM-monkeys is described. The molecules described in this work are composed of a 2-pyridone-containing peptidomimetic binding determinant and an alpha,beta-unsaturated ester Michael acceptor moiety which forms an irreversible covalent adduct with the active site cysteine residue of the 3C enzyme. Modification of the ester contained within these compounds is detailed along with alteration of the P(2) substituent present in the peptidomimetic portion of the inhibitors. The pharmacokinetics of several inhibitors in both dogs and monkeys are described (7 h plasma concentrations after oral administration) along with their human plasma stabilities, stabilities in incubations with human, dog, and monkey microsomes and hepatocytes, Caco-2 permeabilities, and aqueous solubilities. Compounds containing an alpha,beta-unsaturated ethyl ester fragment and either an ethyl or propargyl P(2) moiety displayed the most promising combination of 3C enzyme inhibition (k(obs)/[I] 170 000-223 000 M(-1) s(-1)), antiviral activity (EC(50) = 0.047-0.058 microM, mean vs seven HRV serotypes), and pharmacokinetics following oral administration (7 h dog plasma levels = 0.248-0.682 microM; 7 h CM-monkey plasma levels = 0.057-0.896 microM).
- Published
- 2003
26. Structure-based design of a parallel synthetic array directed toward the discovery of irreversible inhibitors of human rhinovirus 3C protease
- Author
-
Hiep T. Luu, Rose Ann Ferre, Fausto Maldonado, Peter S. Dragovich, Amy K. Patick, Skalitzky Donald James, Shella A. Fuhrman, Ye Hua, David A. Matthews, Shao Song Chu, Fora Chan, Edward L. Brown, Leora S. Zalman, Theodore O. Johnson, Eastman Brian Walter, Siegfried Reich, Michelle Yang, Stephen T. Worland, James W. Meador, and Thomas F. Hendrickson
- Subjects
Picornain 3C ,Rhinovirus ,Stereochemistry ,medicine.drug_class ,Carboxamide ,medicine.disease_cause ,Crystallography, X-Ray ,Chemical synthesis ,Antiviral Agents ,Structure-Activity Relationship ,Viral Proteins ,stomatognathic system ,Drug Discovery ,medicine ,Structure–activity relationship ,Combinatorial Chemistry Techniques ,Humans ,Enzyme Inhibitors ,Cytotoxicity ,chemistry.chemical_classification ,3C Viral Proteases ,virus diseases ,In vitro ,Cysteine Endopeptidases ,Enzyme ,Biochemistry ,chemistry ,Molecular Medicine ,HeLa Cells ,Protein Binding - Abstract
Utilizing the tools of parallel synthesis and structure-based design, a new class of Michael acceptor-containing, irreversible inhibitors of human rhinovirus 3C protease (HRV 3CP) was discovered. These inhibitors are shown to inhibit HRV-14 3CP with rates of inactivation ranging from 886 to 31 400 M(-1) sec(-1). These inhibitors exhibit antiviral activity when tested against HRV-14 infected H1-HeLa cells, with EC(50) values ranging from 1.94 to 0.15 microM. No cytotoxicity was observed at the limits of the assay concentration. A crystal structure of one of the more potent inhibitors covalently bound to HRV-2 3CP is detailed. These compounds were also tested against HRV serotypes other than type 14 and were found to have highly variable activities.
- Published
- 2002
27. Evaluation of a structure-based statine cyclic diamino amide encoded combinatorial library against plasmepsin II and cathepsin D
- Author
-
Roland E. Dolle, Marc Orlowski, Ilya Y. Gluzman, Giorgio Lauri, Carolyn DiIanni Carroll, Daniel E. Goldberg, Shewei Tao, and Theodore O. Johnson
- Subjects
Stereochemistry ,Clinical Biochemistry ,Protozoan Proteins ,Pharmaceutical Science ,Cathepsin D ,Biochemistry ,Chemical synthesis ,chemistry.chemical_compound ,Structure-Activity Relationship ,Plasmepsin II ,Amide ,Drug Discovery ,Statine ,Structure–activity relationship ,Aspartic Acid Endopeptidases ,Humans ,Protease Inhibitors ,Amino Acids ,Molecular Biology ,chemistry.chemical_classification ,biology ,Organic Chemistry ,Combinatorial chemistry ,Amides ,Enzyme ,chemistry ,Enzyme inhibitor ,biology.protein ,Molecular Medicine - Abstract
A structure-based 18,900-member combinatorial library was synthesized containing a statine template and three cyclic diamino acids as potential P1, P2-P4 surrogates. Evaluation of this encoded library against two aspartyl proteases, plasmepsin II and cathepsin D, led to the identification of selective inhibitors for each enzyme.
- Published
- 1999
28. Abstract 5779: The discovery of the potent and selective PI3K/mTOR dual inhibitor PF-04691502 through structure-based drug design
- Author
-
Shubha Bagrodia, Klaus Ruprecht Dress, Phuong Le, Robert Steven Kania, Andrew Pannifer, Dilip Bhumalkar, Kevin K.-C. Liu, Jacqui Elizabeth Hoffman, Hai Wang, Catherine Johnson, Peter Zhu, Henry Cheng, Jason Zbieg, Caroline Rodgers, Daniel R. Knighton, Pairish Mason Alan, Plewe Michael Bruno, Zhengyu Liu, Tran Khanh Tuan, Nowlin Dawn Marie, Xiaojun Huang, Simon Bailey, Sacha Ninkovic, Graham Smith, Li Haitao, Theodore O. Johnson, Matthew A. Marx, Martin Paul Edwards, Samuel Li, Mitch Nambu, Lisa Guo, Shaoxian Sun, Michael R. Gehring, and Qiyue Hu
- Subjects
Cancer Research ,biology ,Kinase ,chemistry.chemical_compound ,Oncology ,chemistry ,Biochemistry ,In vivo ,biology.protein ,Moiety ,PTEN ,Phosphatidylinositol ,Signal transduction ,Protein kinase B ,PI3K/AKT/mTOR pathway - Abstract
The phosphatidylinositol 3-kinase (PI3K) signaling pathway plays crucial roles in cell growth, proliferation and survival. Genomic aberrations in the PI3K pathway, such as mutational activation of PI3Kα or loss of function of tumor suppressor PTEN, have been closely linked to the development and progression of a wide range of cancers. Hence, inhibition of the key targets in the pathway, e.g. PI3K, AKT, mTOR, offers great potential for the treatment of cancer. In an effort to discover compounds that inhibit PI3Kα, a high throughput screen was carried out, and 4-methyl-pyrido-pyrimidine (MPP) derivatives were identified as potent and selective inhibitors of PI3Kα. For example, PF-00271897, 8-cyclopentyl-6-[3-(hydroxymethyl)phenyl]-4-methyl-2-(methylamino)pyrido[2,3-d]pyrimidin-7(8H)-one demonstrated PI3Kα Ki of 2.2 nM. Multiple crystal structures of inhibitors bound to PI3K gamma were determined to inform design and optimization of the ADMET properties of this lead series. Crystallographic studies with PI3K gamma protein indicated that the aminopyrimidine moiety forms two hydrogen bonds to the kinase backbone, and the aromatic moiety at the 6 position binds in a hydrophobic pocket. The X-ray structure suggested that the 4-methyl group on the MPP core structure conferred the excellent overall kinase selectivity to the series. The structure and SAR suggested optimization could come from keeping N-R group at 2 position very small and maintaining aromatic moiety at 6 position for hydrophobic interaction. Introduction of polar groups to the 8N side chains that are located in the ribose binding pocket increased both metabolic stability and solubility. Based on the overall properties, PF-04691502, 2-amino-8-[trans-4-(2-hydroxyethoxy)cyclohexyl]-6-(6-methoxypyridin-3-yl)-4-methylpyrido[2,3-d]pyrimidin-7(8H)-one, was selected as a clinical candidate. PF-04691502 demonstrated Ki's of 1.2-2.2 nM against PI3K α, β, γ and δ isoforms, and Ki of 9.1 nM against recombinant mTOR. PF-04691502 inhibited AKT phosphorylation at S473 in BT20 breast cancer line with IC50 of 12 nM. PF-04691502 is highly selective for inhibition of PI3K family kinases as shown by lack of activity against a panel of >75 protein kinases, including the class III PI3K hVps34. In the in vivo rat PK studies, PF-04691502 demonstrated the following properties: Clearance = 5.2 ml/min/kg, Vdss = 1.4 L/kg, T1/2 = 3.1 h, F% = 63%. The design, synthesis, in vitro potency SAR, selectivity, ADMET of the MPP derivatives will be discussed. The crystal structure of PF-04691502 in PI3K gamma will also be presented. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 5779.
- Published
- 2010
- Full Text
- View/download PDF
29. Azaindole-based HIV-1 Integrase Specific Inhibitors Display Potent Anti-Retroviral Activity
- Author
-
Plewe Michael Bruno, Hua Wu, Scott L. Butler, Dorothy Delisle, Qinghai Peng, Amy K. Patick, Theodore O. Johnson, Atsuo Kuki, and Joanne P. Graham
- Subjects
Pharmacology ,business.industry ,Virology ,Hiv 1 integrase ,Medicine ,Antiretroviral medication ,business - Published
- 2008
- Full Text
- View/download PDF
30. Dual Inhibition of Blood Coagulation Factors Xa and Iia Synergize To Reduce Thrombus Weight and Thrombin Generation in a Rabbit Arteriovenous Shunt Model of Thrombosis
- Author
-
Theodore O. Johnson, Weston R. Gould, Kathy Welch, Thomas B. McClanahan, Liguo Chi, Sangita M. Baxi, Tawny K. Dahring, Robert J. Leadley, and Gary Hicks
- Subjects
medicine.diagnostic_test ,Chemistry ,Immunology ,Cell Biology ,Hematology ,Pharmacology ,medicine.disease ,Biochemistry ,Argatroban ,Venous thrombosis ,Thrombin ,Bleeding time ,Direct thrombin inhibitor ,Antithrombotic ,medicine ,Thrombus ,Discovery and development of direct thrombin inhibitors ,medicine.drug - Abstract
Several compounds currently in development for the treatment of thrombotic disorders demonstrate high levels of specificity for single targets of the blood coagulation cascade such as factor Xa and thrombin. However, development of a single molecule dual inhibitor against factor Xa and thrombin may expand the efficacy to safety ratio of treatment options for arterial and venous thrombosis. The objective of this study was to determine if simultaneous administration of PD 0313052, a selective Xa inhibitor and argatroban, a direct thrombin inhibitor, would lead to a synergistic antithrombotic effect in a rabbit AV shunt model of thrombosis. Intravenous administration of PD 0313052 alone at doses of 0.1, 0.3, and 1.0 mg/kg/min resulted in thrombus weight (TW) reductions of 11±3, 25±10 and 67±7 % compared to the vehicle group. Argatroban at 1, 3 and 10 mg/kg/min reduced TW 16±13, 47±10 and 75±6 %. When PD 0313052 was administered at 0.1 mg/kg/min in combination with argatroban at 1, 3 or 10 mg/kg/min TW was reduced 50±7, 60±7 and 82±9 %. Likewise, argatroban at 1 mg/kg/min combined with 0.1, 0.3 or 1mg/kg/min of PD 0313052 resulted in TW reductions of 56±9, 60±9 and 84±5 %, respectively. At the lowest combined doses of PD 0313052 and argatroban there was no change in bleeding time relative to the additive fold-increases from each drug alone. The EC50 of intravenously administered PD 0313052 and argatroban was 67±23 and 178±58 ng/ml, respectively. When the drugs were combined the EC50 was reduced to 12±6 ng/ml with the PD 0313052/argatroban combination and to 83±29 ng/ml with the argatroban/PD 0313052 combination. A synergistic effect was also observed in an ex vivo assay of thrombin generation (TG). Predicted additive inhibition of TG based on the individual effects of each compound was −9±7, 9±2 and 29±7 % compared to 10±5, 32±5 and 55±3 % with the 313052/argatroban combination. The predicted effects of the argatroban/PD 0313052 combination was −9±7, 1±7 and 16±9 % compared to the actual inhibition of 5±3, 14±5 and 31±7 %. These results demonstrate a significant synergistic antithrombotic effect by combining low doses of a factor Xa and a thrombin inhibitor and support the hypothesis that development of a single molecule inhibitor against different hemostatic targets may offer greater efficacy in the prevention and treatment of venous and arterial thrombosis.
- Published
- 2005
- Full Text
- View/download PDF
31. First total synthesis of Lycopodium alkaloids of the magellanane group. Enantioselective total syntheses of (-)-magellanine and (+)-magellaninone
- Author
-
Larry E. Overman, Gavin C. Hirst, and Theodore O. Johnson
- Subjects
Lycopodium ,biology ,Stereochemistry ,Alkaloid ,Enantioselective synthesis ,Total synthesis ,General Chemistry ,biology.organism_classification ,Biochemistry ,Catalysis ,chemistry.chemical_compound ,Colloid and Surface Chemistry ,chemistry ,Group (periodic table) ,Magellaninone ,Organic chemistry ,Enone ,Amination - Published
- 1993
- Full Text
- View/download PDF
32. Evaluation of a Published in Silico Model and Construction of a Novel Bayesian Model for Predicting Phospholipidosis Inducing Potential.
- Author
-
Dennis J. Pelletier, Daniel Gehlhaar, Anne Tilloy-Ellul, Theodore O. Johnson, and Nigel Greene
- Published
- 2007
- Full Text
- View/download PDF
33. An Analytical Survey of the Fifteen Two-Part Inventions by J.S. Bach
- Author
-
Theodore O. Johnson and Theodore O. Johnson
- Abstract
To find out more information about Rowman & Littlefield titles please visit us at www.rowmanlittlefield.com.
- Published
- 1982
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.