72 results on '"Corey Strickland"'
Search Results
2. Structures of active-state orexin receptor 2 rationalize peptide and small-molecule agonist recognition and receptor activation
- Author
-
Chuan Hong, Noel J. Byrne, Beata Zamlynny, Srivanya Tummala, Li Xiao, Jennifer M. Shipman, Andrea T. Partridge, Christina Minnick, Michael J. Breslin, Michael T. Rudd, Shawn J. Stachel, Vanessa L. Rada, Jeffrey C. Kern, Kira A. Armacost, Scott A. Hollingsworth, Julie A. O’Brien, Dawn L. Hall, Terrence P. McDonald, Corey Strickland, Alexei Brooun, Stephen M. Soisson, and Kaspar Hollenstein
- Subjects
Science - Abstract
Agonists of the orexin receptor 2 (OX2R) show promise in the treatment of narcolepsy. Cryo-EM structures of active-state OX2R bound to an endogenous peptide agonist and a small-molecule agonist suggest a molecular mechanism that rationalizes both receptor activation and inhibition.
- Published
- 2021
- Full Text
- View/download PDF
3. Author Correction: Cryo-EM structures of inhibitory antibodies complexed with arginase 1 provide insight into mechanism of action
- Author
-
Rachel L. Palte, Veronica Juan, Yacob Gomez-Llorente, Marc Andre Bailly, Kalyan Chakravarthy, Xun Chen, Daniel Cipriano, Ghassan N. Fayad, Laurence Fayadat-Dilman, Symon Gathiaka, Heiko Greb, Brian Hall, Mas Handa, Mark Hsieh, Esther Kofman, Heping Lin, J. Richard Miller, Nhung Nguyen, Jennifer O’Neil, Hussam Shaheen, Eric Sterner, Corey Strickland, Angie Sun, Shane Taremi, and Giovanna Scapin
- Subjects
Biology (General) ,QH301-705.5 - Published
- 2021
- Full Text
- View/download PDF
4. Characterization and Modeling of Reversible Antibody Self-Association Provide Insights into Behavior, Prediction, and Correction
- Author
-
Carl Mieczkowski, Alan Cheng, Thierry Fischmann, Mark Hsieh, Jeanne Baker, Makiko Uchida, Gopalan Raghunathan, Corey Strickland, and Laurence Fayadat-Dilman
- Subjects
antibody ,protein ,self-association ,self-interaction ,developability ,in silico prediction ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Reversible antibody self-association, while having major developability and therapeutic implications, is not fully understood or readily predictable and correctable. For a strongly self-associating humanized mAb variant, resulting in unacceptable viscosity, the monovalent affinity of self-interaction was measured in the low μM range, typical of many specific and biologically relevant protein–protein interactions. A face-to-face interaction model extending across both the heavy-chain (HC) and light-chain (LC) Complementary Determining Regions (CDRs) was apparent from biochemical and mutagenesis approaches as well as computational modeling. Light scattering experiments involving individual mAb, Fc, Fab, and Fab’2 domains revealed that Fabs self-interact to form dimers, while bivalent mAb/Fab’2 forms lead to significant oligomerization. Site-directed mutagenesis of aromatic residues identified by homology model patch analysis and self-docking dramatically affected self-association, demonstrating the utility of these predictive approaches, while revealing a highly specific and tunable nature of self-binding modulated by single point mutations. Mutagenesis at these same key HC/LC CDR positions that affect self-interaction also typically abolished target binding with notable exceptions, clearly demonstrating the difficulties yet possibility of correcting self-association through engineering. Clear correlations were also observed between different methods used to assess self-interaction, such as Dynamic Light Scattering (DLS) and Affinity-Capture Self-Interaction Nanoparticle Spectroscopy (AC-SINS). Our findings advance our understanding of therapeutic protein and antibody self-association and offer insights into its prediction, evaluation and corrective mitigation to aid therapeutic development.
- Published
- 2021
- Full Text
- View/download PDF
5. Crystal Structure and Characterization of Human Heavy-Chain Only Antibodies Reveals a Novel, Stable Dimeric Structure Similar to Monoclonal Antibodies
- Author
-
Carl Mieczkowski, Soheila Bahmanjah, Yao Yu, Jeanne Baker, Gopalan Raghunathan, Daniela Tomazela, Mark Hsieh, Mark McCoy, Corey Strickland, and Laurence Fayadat-Dilman
- Subjects
antibody ,heavy-chain dimer ,heavy-chain antibody ,crystal structure ,Immunologic diseases. Allergy ,RC581-607 - Abstract
We report the novel crystal structure and characterization of symmetrical, homodimeric humanized heavy-chain-only antibodies or dimers (HC2s). HC2s were found to be significantly coexpressed and secreted along with mAbs from transient CHO HC/LC cotransfection, resulting in an unacceptable mAb developability attribute. Expression of full-length HC2s in the absence of LC followed by purification resulted in HC2s with high purity and thermal stability similar to conventional mAbs. The VH and CH1 portion of the heavy chain (or Fd) was also efficiently expressed and yielded a stable, covalent, and reducible dimer (Fd2). Mutagenesis of all heavy chain cysteines involved in disulfide bond formation revealed that Fd2 intermolecular disulfide formation was similar to Fabs and elucidated requirements for Fd2 folding and expression. For one HC2, we solved the crystal structure of the Fd2 domain to 2.9 Å, revealing a highly symmetrical homodimer that is structurally similar to Fabs and is mediated by conserved (CH1) and variable (VH) contacts with all CDRs positioned outward for target binding. Interfacial dimer contacts revealed by the crystal structure were mutated for two HC2s and were found to dramatically affect HC2 formation while maintaining mAb bioactivity, offering a potential means to modulate novel HC2 formation through engineering. These findings indicate that human heavy-chain dimers can be secreted efficiently in the absence of light chains, may show good physicochemical properties and stability, are structurally similar to Fabs, offer insights into their mechanism of formation, and may be amenable as a novel therapeutic modality.
- Published
- 2020
- Full Text
- View/download PDF
6. Discovery and Structure-Based Design of Macrocyclic Peptides Targeting STUB1
- Author
-
Simon Ng, Alexander C. Brueckner, Soheila Bahmanjah, Qiaolin Deng, Jennifer M. Johnston, Lan Ge, Ruchia Duggal, Bahanu Habulihaz, Benjamin Barlock, Sookhee Ha, Ahmad Sadruddin, Constance Yeo, Corey Strickland, Andrea Peier, Brian Henry, Edward C. Sherer, and Anthony W. Partridge
- Subjects
Drug Discovery ,Molecular Medicine - Abstract
STIP1 homology and U-Box containing protein 1 (STUB1) plays a key role in maintaining cell health during stress and aging. Recent evidence suggested STUB1 also helps regulate immunity with the potential of clearing malignant cells. Indeed, we and others have shown that STUB1 is a pivotal negative regulator of interferon gamma sensing – a process critical to the immunosurveillance of tumors and pathogens. Thus far, investigation of STUB1’s role relies mostly on genetic approaches as pharmacological inhibitors of this protein are lacking. Identification of a STUB1 tool compound is important as it would allow therapeutically relevant target validation in a broader sense. Accordingly, we leveraged phage display and computational modeling to identify and refine STUB1 binders. Screening of >10E9 macrocyclic peptides resulted in several conserved motifs as well as structurally diverse leads. Co-crystal structure of the peptide hit and STUB1 has enabled us to employ structure-based in silico design for further optimization. Of the modifications employed, replacing the hydrophilic solvent-exposed region of the macrocyclic peptides with a hydrophobic scaffold improved cellular permeability, while the binding conformation was maintained. Further substitution of the permeability-limiting terminal aspartic acid with a tetrazole bioisostere retained the binding to certain extent while improving permeability, suggesting a path forward. The current lead, although not optimal for cellular study, provides a valuable template for further development into selective tool compounds for STUB1 to enable target validation.
- Published
- 2022
- Full Text
- View/download PDF
7. Structures of active-state orexin receptor 2 rationalize peptide and small-molecule agonist recognition and receptor activation
- Author
-
Corey Strickland, Christina Minnick, Michael J. Breslin, Srivanya Tummala, Kaspar Hollenstein, Alexei Brooun, Vanessa L. Rada, Shawn J. Stachel, Beata Zamlynny, Kira A. Armacost, Dawn L. Hall, Li Xiao, Terrence P. McDonald, Chuan Hong, Kern Jeffrey, Scott A. Hollingsworth, Stephen M. Soisson, Julie A. O'Brien, Andrea T. Partridge, Jennifer M. Shipman, Michael T. Rudd, and Noel Byrne
- Subjects
Agonist ,Protein Conformation, alpha-Helical ,medicine.drug_class ,Science ,Genetic Vectors ,General Physics and Astronomy ,Aminopyridines ,Gene Expression ,Peptide ,Plasma protein binding ,Pharmacology ,Molecular Dynamics Simulation ,General Biochemistry, Genetics and Molecular Biology ,Article ,G protein-coupled receptors ,Cryoelectron microscopy ,Orexin Receptors ,medicine ,Escherichia coli ,Humans ,Protein Interaction Domains and Motifs ,Cloning, Molecular ,chemistry.chemical_classification ,Sulfonamides ,Multidisciplinary ,Binding Sites ,Chemistry ,HEK 293 cells ,digestive, oral, and skin physiology ,General Chemistry ,Azepines ,Triazoles ,medicine.disease ,Small molecule ,Orexin receptor ,Recombinant Proteins ,Orexin ,HEK293 Cells ,Sleep Aids, Pharmaceutical ,Orexin Receptor Antagonists ,Protein Conformation, beta-Strand ,Peptides ,Narcolepsy ,Protein Binding - Abstract
Narcolepsy type 1 (NT1) is a chronic neurological disorder that impairs the brain’s ability to control sleep-wake cycles. Current therapies are limited to the management of symptoms with modest effectiveness and substantial adverse effects. Agonists of the orexin receptor 2 (OX2R) have shown promise as novel therapeutics that directly target the pathophysiology of the disease. However, identification of drug-like OX2R agonists has proven difficult. Here we report cryo-electron microscopy structures of active-state OX2R bound to an endogenous peptide agonist and a small-molecule agonist. The extended carboxy-terminal segment of the peptide reaches into the core of OX2R to stabilize an active conformation, while the small-molecule agonist binds deep inside the orthosteric pocket, making similar key interactions. Comparison with antagonist-bound OX2R suggests a molecular mechanism that rationalizes both receptor activation and inhibition. Our results enable structure-based discovery of therapeutic orexin agonists for the treatment of NT1 and other hypersomnia disorders., Agonists of the orexin receptor 2 (OX2R) show promise in the treatment of narcolepsy. Cryo-EM structures of active-state OX2R bound to an endogenous peptide agonist and a small-molecule agonist suggest a molecular mechanism that rationalizes both receptor activation and inhibition.
- Published
- 2021
8. Cryo-EM structures of inhibitory antibodies complexed with arginase 1 provide insight into mechanism of action
- Author
-
Laurence Fayadat-Dilman, Jennifer O'Neil, Hussam Hisham Shaheen, Ghassan N. Fayad, Heping Lin, S. Shane Taremi, Giovanna Scapin, Corey Strickland, Heiko Greb, Nhung Nguyen, Symon Gathiaka, Mas Handa, Daniel Cipriano, Rachel L. Palte, Brian Hall, Eric Sterner, Veronica Juan, Esther Kofman, Xun Chen, Kalyan Chakravarthy, Yacob Gomez-Llorente, Mark Hsieh, J. Richard Miller, Angie Sun, and Marc Andre Bailly
- Subjects
Ornithine ,0301 basic medicine ,QH301-705.5 ,medicine.drug_class ,Immunology ,Allosteric regulation ,Medicine (miscellaneous) ,Arginine ,Monoclonal antibody ,Article ,General Biochemistry, Genetics and Molecular Biology ,Epitope ,Substrate Specificity ,03 medical and health sciences ,0302 clinical medicine ,Cryoelectron microscopy ,medicine ,Biology (General) ,Author Correction ,chemistry.chemical_classification ,Binding Sites ,Arginase ,biology ,Small molecule ,030104 developmental biology ,Enzyme ,chemistry ,Mechanism of action ,Biochemistry ,030220 oncology & carcinogenesis ,biology.protein ,medicine.symptom ,Antibody ,General Agricultural and Biological Sciences ,Protein Binding - Abstract
Human Arginase 1 (hArg1) is a metalloenzyme that catalyzes the hydrolysis of l-arginine to l-ornithine and urea, and modulates T-cell-mediated immune response. Arginase-targeted therapies have been pursued across several disease areas including immunology, oncology, nervous system dysfunction, and cardiovascular dysfunction and diseases. Currently, all published hArg1 inhibitors are small molecules usually less than 350 Da in size. Here we report the cryo-electron microscopy structures of potent and inhibitory anti-hArg antibodies bound to hArg1 which form distinct macromolecular complexes that are greater than 650 kDa. With local resolutions of 3.5 Å or better we unambiguously mapped epitopes and paratopes for all five antibodies and determined that the antibodies act through orthosteric and allosteric mechanisms. These hArg1:antibody complexes present an alternative mechanism to inhibit hArg1 activity and highlight the ability to utilize antibodies as probes in the discovery and development of peptide and small molecule inhibitors for enzymes in general., Palte et al provide cryo-EM structures of five potent and inhibitory monoclonal antibodies bound to human Arginase 1, a T-cell modulating metalloenzyme and a cancer drug target. They provide structural insights that will aid in the evaluation of these antibodies as therapeutic inhibitors of arginasemediated T-cell suppression.
- Published
- 2021
- Full Text
- View/download PDF
9. Author Correction: Cryo-EM structures of inhibitory antibodies complexed with arginase 1 provide insight into mechanism of action
- Author
-
Symon Gathiaka, Mark Hsieh, Marc Andre Bailly, Brian Hall, Angie Sun, S. Shane Taremi, Veronica Juan, Xun Chen, J. Richard Miller, Mas Handa, Ghassan N. Fayad, Esther Kofman, Laurence Fayadat-Dilman, Jennifer O'Neil, Eric Sterner, Hussam Hisham Shaheen, Corey Strickland, Heping Lin, Kalyan Chakravarthy, Yacob Gomez-Llorente, Rachel L. Palte, Daniel Cipriano, Heiko Greb, Giovanna Scapin, and Nhung Nguyen
- Subjects
Arginase ,Mechanism of action ,Biochemistry ,Chemistry ,Cryo-electron microscopy ,QH301-705.5 ,medicine ,Medicine (miscellaneous) ,medicine.symptom ,Biology (General) ,General Agricultural and Biological Sciences ,Inhibitory antibodies ,General Biochemistry, Genetics and Molecular Biology - Published
- 2021
10. Discovery of an Anion-Dependent Farnesyltransferase Inhibitor from a Phenotypic Screen
- Author
-
Ian M. Bell, Steve S. Carroll, Paula J. Hancock, Richard J. O. Barnard, M. Katharine Holloway, David M. Tellers, Alan Hruza, Gregory C. Adam, Christopher D. Cox, Peter S. Kutchukian, Matthew Tudor, Jing Li, B. Wesley Trotter, Anthony W. Shaw, Corey Strickland, Marina Bukhtiyarova, Scott E. Wolkenberg, David A. Powell, Erica M. Cook, Ivan Cornella-Taracido, and Philip M. McKenna
- Subjects
business.industry ,Chemistry ,medicine.drug_class ,Phenotypic screening ,Organic Chemistry ,Farnesyltransferase inhibitor ,Histone deacetylase inhibitor ,Human immunodeficiency virus (HIV) ,medicine.disease_cause ,Biochemistry ,Text mining ,Drug Discovery ,Proteome ,medicine ,Cancer research ,Latency (engineering) ,business ,Vorinostat ,medicine.drug - Abstract
[Image: see text] By employing a phenotypic screen, a set of compounds, exemplified by 1, were identified which potentiate the ability of histone deacetylase inhibitor vorinostat to reverse HIV latency. Proteome enrichment followed by quantitative mass spectrometric analysis employing a modified analogue of 1 as affinity bait identified farnesyl transferase (FTase) as the primary interacting protein in cell lysates. This ligand-FTase binding interaction was confirmed via X-ray crystallography and temperature dependent fluorescence studies, despite 1 lacking structural and binding similarity to known FTase inhibitors. Although multiple lines of evidence established the binding interaction, these ligands exhibited minimal inhibitory activity in a cell-free biochemical FTase inhibition assay. Subsequent modification of the biochemical assay by increasing anion concentration demonstrated FTase inhibitory activity in this novel class. We propose 1 binds together with the anion in the active site to inhibit farnesyl transferase. Implications for phenotypic screening deconvolution and HIV reactivation are discussed.
- Published
- 2020
11. Pembrolizumab microgravity crystallization experimentation
- Author
-
Denarra Simmons, Giovanna Scapin, Chakravarthy Nachu Narasimhan, Paul Reichert, Corey Strickland, Ray Polniak, Erika Walsh, Xiaoyu Yang, Winifred W. Prosise, Wendy Benjamin, Thierry O. Fischmann, Johnathan Welch, Daya Patel, and April Spinale
- Subjects
Materials science ,Physics and Astronomy (miscellaneous) ,Materials Science (miscellaneous) ,lcsh:Biotechnology ,Medicine (miscellaneous) ,02 engineering and technology ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Biochemistry ,lcsh:Physiology ,Article ,law.invention ,03 medical and health sciences ,Viscosity ,Rheology ,law ,lcsh:TP248.13-248.65 ,Biophysical chemistry ,Crystallization ,030304 developmental biology ,0303 health sciences ,lcsh:QP1-981 ,Sedimentation ,021001 nanoscience & nanotechnology ,Agricultural and Biological Sciences (miscellaneous) ,Chemical engineering ,Space and Planetary Science ,Homogeneous ,Drug delivery ,Particle-size distribution ,0210 nano-technology ,National laboratory - Abstract
Crystallization processes have been widely used in the pharmaceutical industry for the manufacture, storage, and delivery of small-molecule and small protein therapeutics. However, the identification of crystallization processes for biologics, particularly monoclonal antibodies, has been prohibitive due to the size and the flexibility of their overall structure. There remains a challenge and an opportunity to utilize the benefits of crystallization of biologics. The research laboratories of Merck Sharp & Dome Corp. (MSD) in collaboration with the International Space Station (ISS) National Laboratory performed crystallization experiments with pembrolizumab (Keytruda®) on the SpaceX-Commercial Resupply Services-10 mission to the ISS. By leveraging microgravity effects such as reduced sedimentation and minimal convection currents, conditions producing crystalline suspensions of homogeneous monomodal particle size distribution (39 μm) in high yield were identified. In contrast, the control ground experiments produced crystalline suspensions with a heterogeneous bimodal distribution of 13 and 102 μm particles. In addition, the flight crystalline suspensions were less viscous and sedimented more uniformly than the comparable ground-based crystalline suspensions. These results have been applied to the production of crystalline suspensions on earth, using rotational mixers to reduce sedimentation and temperature gradients to induce and control crystallization. Using these techniques, we have been able to produce uniform crystalline suspensions (1–5 μm) with acceptable viscosity (
- Published
- 2019
12. Structure of the insulin receptor–insulin complex by single-particle cryo-EM analysis
- Author
-
Corey Strickland, Bridget Carragher, Winifred W. Prosise, Theresa M. Kelly, Clinton S. Potter, Alan Hruza, Venkata P. Dandey, Todd Mayhood, Zhening Zhang, and Giovanna Scapin
- Subjects
Models, Molecular ,0301 basic medicine ,medicine.medical_treatment ,Crystallography, X-Ray ,Article ,03 medical and health sciences ,medicine ,Humans ,Insulin ,Glucose homeostasis ,Binding site ,Multidisciplinary ,biology ,Chemistry ,Cryoelectron Microscopy ,Receptor, Insulin ,Single Molecule Imaging ,Cell biology ,Insulin receptor ,Transmembrane domain ,030104 developmental biology ,Ectodomain ,biology.protein ,Protein Multimerization ,Signal transduction ,Apoproteins ,Tyrosine kinase ,Signal Transduction - Abstract
Cryo-electron microscopy structures of insulin in a complex with the insulin receptor define the S2 binding site on the receptor and suggest a mechanism for downstream propagation of insulin signalling. Insulin is a peptide hormone with important roles in metabolism. Recent crystal structures of insulin bound to a truncated form of the insulin receptor have provided insights into the binding mode of this complex. Giovanna Scapin and colleagues now describe the three-dimensional structure of the ectodomain of the insulin receptor in complex with an insulin dimer, solved to a resolution of 4.6 A using single-particle cryo-electron microscopy. The findings advance our structural and mechanistic knowledge about the formation of the insulin–receptor complex and receptor activation. The insulin receptor is a dimeric protein that has a crucial role in controlling glucose homeostasis, regulating lipid, protein and carbohydrate metabolism, and modulating brain neurotransmitter levels1,2. Insulin receptor dysfunction has been associated with many diseases, including diabetes, cancer and Alzheimer’s disease1,3,4. The primary sequence of the receptor has been known since the 1980s5, and is composed of an extracellular portion (the ectodomain, ECD), a single transmembrane helix and an intracellular tyrosine kinase domain. Binding of insulin to the dimeric ECD triggers auto-phosphorylation of the tyrosine kinase domain and subsequent activation of downstream signalling molecules. Biochemical and mutagenesis data have identified two putative insulin-binding sites, S1 and S26. The structures of insulin bound to an ECD fragment containing S1 and of the apo ectodomain have previously been reported7,8, but details of insulin binding to the full receptor and the signal propagation mechanism are still not understood. Here we report single-particle cryo-electron microscopy reconstructions of the 1:2 (4.3 A) and 1:1 (7.4 A) complexes of the insulin receptor ECD dimer with insulin. The symmetrical 4.3 A structure shows two insulin molecules per dimer, each bound between the leucine-rich subdomain L1 of one monomer and the first fibronectin-like domain (FnIII-1) of the other monomer, and making extensive interactions with the α-subunit C-terminal helix (α-CT helix). The 7.4 A structure has only one similarly bound insulin per receptor dimer. The structures confirm the binding interactions at S1 and define the full S2 binding site. These insulin receptor states suggest that recruitment of the α-CT helix upon binding of the first insulin changes the relative subdomain orientations and triggers downstream signal propagation.
- Published
- 2018
- Full Text
- View/download PDF
13. From concept to reality: cryoEM as an integral part of drug discovery and development
- Author
-
Corey Strickland
- Subjects
Engineering ,business.industry ,Drug discovery ,business ,Instrumentation ,Data science - Published
- 2021
- Full Text
- View/download PDF
14. Characterization and Modeling of Reversible Antibody Self-Association Provide Insights into Behavior, Prediction, and Correction
- Author
-
Laurence Fayadat-Dilman, Thierry O. Fischmann, Jeanne Baker, Corey Strickland, Alan C. Cheng, Mark Hsieh, Makiko Uchida, Gopalan Raghunathan, and Carl Mieczkowski
- Subjects
lcsh:Immunologic diseases. Allergy ,computational modeling ,0301 basic medicine ,self-interaction ,in silico prediction ,medicine.drug_class ,Self association ,Immunology ,Monoclonal antibody ,030226 pharmacology & pharmacy ,Article ,03 medical and health sciences ,0302 clinical medicine ,Dynamic light scattering ,antibody ,developability ,Drug Discovery ,medicine ,Immunology and Allergy ,Homology modeling ,biology ,Chemistry ,Mutagenesis ,dynamic light scattering ,Interaction model ,Characterization (materials science) ,030104 developmental biology ,viscosity ,biology.protein ,Biophysics ,Antibody ,lcsh:RC581-607 ,protein ,self-association - Abstract
Reversible antibody self-association, while having major developability and therapeutic implications, is not fully understood or readily predictable and correctable. For a strongly self-associating humanized mAb variant, resulting in unacceptable viscosity, the monovalent affinity of self-interaction was measured in the low μM range, typical of many specific and biologically relevant protein–protein interactions. A face-to-face interaction model extending across both the heavy-chain (HC) and light-chain (LC) Complementary Determining Regions (CDRs) was apparent from biochemical and mutagenesis approaches as well as computational modeling. Light scattering experiments involving individual mAb, Fc, Fab, and Fab’2 domains revealed that Fabs self-interact to form dimers, while bivalent mAb/Fab’2 forms lead to significant oligomerization. Site-directed mutagenesis of aromatic residues identified by homology model patch analysis and self-docking dramatically affected self-association, demonstrating the utility of these predictive approaches, while revealing a highly specific and tunable nature of self-binding modulated by single point mutations. Mutagenesis at these same key HC/LC CDR positions that affect self-interaction also typically abolished target binding with notable exceptions, clearly demonstrating the difficulties yet possibility of correcting self-association through engineering. Clear correlations were also observed between different methods used to assess self-interaction, such as Dynamic Light Scattering (DLS) and Affinity-Capture Self-Interaction Nanoparticle Spectroscopy (AC-SINS). Our findings advance our understanding of therapeutic protein and antibody self-association and offer insights into its prediction, evaluation and corrective mitigation to aid therapeutic development.
- Published
- 2021
- Full Text
- View/download PDF
15. Structure–activity relationship study of 4-substituted piperidines at Leu26 moiety of novel p53–hDM2 inhibitors
- Author
-
Craig R. Gibeau, Yao Ma, Gerald W. Shipps, Tian Yuan, Brian R. Lahue, Corey Strickland, and Stephane L. Bogen
- Subjects
Models, Molecular ,0301 basic medicine ,Stereochemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Biochemistry ,Structure-Activity Relationship ,03 medical and health sciences ,0302 clinical medicine ,Mdm2 Protein ,Piperidines ,Leucine ,Drug Discovery ,Humans ,Moiety ,Structure–activity relationship ,Molecular Biology ,Structure modification ,Dose-Response Relationship, Drug ,Molecular Structure ,Chemistry ,Organic Chemistry ,Proto-Oncogene Proteins c-mdm2 ,Combinatorial chemistry ,030104 developmental biology ,030220 oncology & carcinogenesis ,Molecular Medicine ,Tumor Suppressor Protein p53 - Abstract
Led by the structural information of the screening hit with mDM2 protein, a structure modification of Leu26 moiety of the novel p53–hDM2 inhibitors was conducted. A structure–activity relationship study of 4-substituted piperidines revealed compound 20t with good potencies and excellent CYP450 profiles.
- Published
- 2016
- Full Text
- View/download PDF
16. Structure-Based Design of an Iminoheterocyclic β-Site Amyloid Precursor Protein Cleaving Enzyme (BACE) Inhibitor that Lowers Central Aβ in Nonhuman Primates
- Author
-
Presscott T. Leach, Jared N. Cumming, Peter Orth, Yusheng Wu, Zhaoning Zhu, Xiaoxiang Liu, Guoqing Li, Kaushik Mitra, Leonard Favreau, Misiaszek Jeffrey A, Corey Strickland, Hongwu Wang, Brad E. Smith, Robert Mazzola, Diane Grotz, Xia Chen, William J. Greenlee, Mihirbaran Mandal, Michael Grzelak, Matthew E. Kennedy, Eric M. Parker, Johannes H. Voigt, John P. Caldwell, Andrew Stamford, Irina Kazakevich, Kathleen Cox, Reshma Kuvelkar, Lili Zhang, Giuseppe Terracina, Qi Zhang, Maria S. Michener, Lynn A. Hyde, Alexei V. Buevich, and Mckittrick Brian A
- Subjects
0301 basic medicine ,Pyrimidine ,Stereochemistry ,Substituent ,Pyrrolidine ,Amyloid beta-Protein Precursor ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Heterocyclic Compounds ,In vivo ,mental disorders ,Drug Discovery ,Amyloid precursor protein ,Animals ,Structure–activity relationship ,Enzyme Inhibitors ,Cerebral Cortex ,chemistry.chemical_classification ,Amyloid beta-Peptides ,Molecular Structure ,CYP3A4 ,biology ,Rats ,Macaca fascicularis ,030104 developmental biology ,Enzyme ,chemistry ,Biochemistry ,Drug Design ,biology.protein ,Molecular Medicine ,Imines ,Amyloid Precursor Protein Secretases ,030217 neurology & neurosurgery - Abstract
We describe successful efforts to optimize the in vivo profile and address off-target liabilities of a series of BACE1 inhibitors represented by 6 that embodies the recently validated fused pyrrolidine iminopyrimidinone scaffold. Employing structure-based design, truncation of the cyanophenyl group of 6 that binds in the S3 pocket of BACE1 followed by modification of the thienyl group in S1 was pursued. Optimization of the pyrimidine substituent that binds in the S2'-S2″ pocket of BACE1 remediated time-dependent CYP3A4 inhibition of earlier analogues in this series and imparted high BACE1 affinity. These efforts resulted in the discovery of difluorophenyl analogue 9 (MBi-4), which robustly lowered CSF and cortex Aβ40 in both rats and cynomolgus monkeys following a single oral dose. Compound 9 represents a unique molecular shape among BACE inhibitors reported to potently lower central Aβ in nonrodent preclinical species.
- Published
- 2016
- Full Text
- View/download PDF
17. A novel storage system for cryoEM samples
- Author
-
Winifred W. Prosise, Michael K. Wismer, Corey Strickland, and Giovanna Scapin
- Subjects
Automation, Laboratory ,0301 basic medicine ,business.industry ,Computer science ,Cryoelectron Microscopy ,Preservation, Biological ,Grid ,Tracking (particle physics) ,Automation ,Sample (graphics) ,Article ,Specimen Handling ,Computational science ,03 medical and health sciences ,030104 developmental biology ,Research Design ,Structural Biology ,Computer data storage ,Grid energy storage ,business ,Simulation - Abstract
We present here a new CryoEM grid boxes storage system designed to simplify sample labeling, tracking and retrieval. The system is based on the crystal pucks widely used by the X-ray crystallographic community for storage and shipping of crystals. This system is suitable for any cryoEM laboratory, but especially for large facilities that will need accurate tracking of large numbers of samples coming from different sources.
- Published
- 2017
- Full Text
- View/download PDF
18. Discovery of Selective RNA-Binding Small Molecules by Affinity-Selection Mass Spectrometry
- Author
-
John A. Howe, Julja Burchard, Gabriel Mercado, Graham Smith, Daniel J. Klein, Margaret T. Butko, Scott S. Walker, Thierry O. Fischmann, Corey Strickland, Hai-Young Kim, Elliott B. Nickbarg, Alan Hruza, Ali Nahvi, Noreen F. Rizvi, Matthew Richards, Mark A. McCoy, Peter J. Dandliker, Chad Chamberlin, and Peter Saradjian
- Subjects
0301 basic medicine ,Riboswitch ,animal structures ,Flavin Mononucleotide ,Flavin mononucleotide ,Computational biology ,Crystallography, X-Ray ,Ligands ,Biochemistry ,Mass Spectrometry ,Small Molecule Libraries ,03 medical and health sciences ,chemistry.chemical_compound ,Drug Discovery ,Molecular Structure ,Drug discovery ,Ligand ,RNA ,General Medicine ,Non-coding RNA ,Small molecule ,030104 developmental biology ,Pyrimidines ,chemistry ,Molecular Medicine - Abstract
Recent advances in understanding the relevance of noncoding RNA (ncRNA) to disease have increased interest in drugging ncRNA with small molecules. The recent discovery of ribocil, a structurally distinct synthetic mimic of the natural ligand of the flavin mononucleotide (FMN) riboswitch, has revealed the potential chemical diversity of small molecules that target ncRNA. Affinity-selection mass spectrometry (AS-MS) is theoretically applicable to high-throughput screening (HTS) of small molecules binding to ncRNA. Here, we report the first application of the Automated Ligand Detection System (ALIS), an indirect AS-MS technique, for the selective detection of small molecule-ncRNA interactions, high-throughput screening against large unbiased small-molecule libraries, and identification and characterization of novel compounds (structurally distinct from both FMN and ribocil) that target the FMN riboswitch. Crystal structures reveal that different compounds induce various conformations of the FMN riboswitch, leading to different activity profiles. Our findings validate the ALIS platform for HTS screening for RNA-binding small molecules and further demonstrate that ncRNA can be broadly targeted by chemically diverse yet selective small molecules as therapeutics.
- Published
- 2018
19. Discovery of potent iminoheterocycle BACE1 inhibitors
- Author
-
Lili Zhang, Qi Zhang, Andrew Stamford, Liyang Wang, Peter Orth, Reshma Kuvelkar, Corey Strickland, James Durkin, Mckittrick Brian A, Zhaoning Zhu, Johannes H. Voigt, Julie Lee, Robert Mazzola, Nansie A. McHugh, Leonard Favreau, Xia Chen, Matthew E. Kennedy, Joseph Chen, and John P. Caldwell
- Subjects
Models, Molecular ,Molecular Structure ,Chemistry ,Stereochemistry ,Organic Chemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Biochemistry ,Affinities ,Rats ,Structure-Activity Relationship ,Aspartate protease ,Alzheimer Disease ,Oral administration ,Drug Design ,Drug Discovery ,β secretase ,Animals ,Aspartic Acid Endopeptidases ,Molecular Medicine ,Potency ,Amyloid Precursor Protein Secretases ,Binding site ,Molecular Biology - Abstract
The synthesis of a series of iminoheterocycles and their structure–activity relationships (SAR) as inhibitors of the aspartyl protease BACE1 will be detailed. An effort to access the S3 subsite directly from the S1 subsite initially yielded compounds with sub-micromolar potency. A subset of compounds from this effort unexpectedly occupied a different binding site and displayed excellent BACE1 affinities. Select compounds from this subset acutely lowered Aβ40 levels upon subcutaneous and oral administration to rats.
- Published
- 2014
- Full Text
- View/download PDF
20. Mechanism of Action and Epitopes of Clostridium difficile Toxin B-neutralizing Antibody Bezlotoxumab Revealed by X-ray Crystallography
- Author
-
Payal R. Sheth, Maribel Beaumont, Edward DiNunzio, Fred Racine, Li Xiao, Paul Reichert, Xiaoyu Yang, Jerzy Karczewski, Corey Strickland, Nicholas Murgolo, Susan Secore, Richard N. Ingram, Peter Orth, Todd Mayhood, Lorraine D. Hernandez, Grigori Ermakov, and Alex G. Therien
- Subjects
Models, Molecular ,Bacterial Toxins ,Molecular Sequence Data ,Clostridium difficile toxin B ,Crystallography, X-Ray ,Microbiology ,Biochemistry ,Epitope ,Epitopes ,Bacterial Proteins ,Amino Acid Sequence ,Binding site ,Neutralizing antibody ,Molecular Biology ,Peptide sequence ,Oligopeptide ,Binding Sites ,biology ,Clostridioides difficile ,Antibodies, Monoclonal ,Cell Biology ,Antibodies, Bacterial ,Antibodies, Neutralizing ,Protein Structure, Tertiary ,Epitope mapping ,Bezlotoxumab ,biology.protein ,Broadly Neutralizing Antibodies ,Epitope Mapping - Abstract
The symptoms of Clostridium difficile infections are caused by two exotoxins, TcdA and TcdB, which target host colonocytes by binding to unknown cell surface receptors, at least in part via their combined repetitive oligopeptide (CROP) domains. A combination of the anti-TcdA antibody actoxumab and the anti-TcdB antibody bezlotoxumab is currently under development for the prevention of recurrent C. difficile infections. We demonstrate here through various biophysical approaches that bezlotoxumab binds to specific regions within the N-terminal half of the TcdB CROP domain. Based on this information, we solved the x-ray structure of the N-terminal half of the TcdB CROP domain bound to Fab fragments of bezlotoxumab. The structure reveals that the TcdB CROP domain adopts a β-solenoid fold consisting of long and short repeats and that bezlotoxumab binds to two homologous sites within the CROP domain, partially occluding two of the four putative carbohydrate binding pockets located in TcdB. We also show that bezlotoxumab neutralizes TcdB by blocking binding of TcdB to mammalian cells. Overall, our data are consistent with a model wherein a single molecule of bezlotoxumab neutralizes TcdB by binding via its two Fab regions to two epitopes within the N-terminal half of the TcdB CROP domain, partially blocking the carbohydrate binding pockets of the toxin and preventing toxin binding to host cells.
- Published
- 2014
- Full Text
- View/download PDF
21. New class of azaheptapyridine FPT inhibitors as potential cancer therapy agents
- Author
-
Wang James J-S, Paul Kirschmeier, Ming Liu, Corey Strickland, Desai Jagdish A, Amin A. Nomeir, Alan B. Cooper, Viyyoor M. Girijavallabhan, Dinananth F. Rane, and Hugh Y. Zhu
- Subjects
Models, Molecular ,Pyridines ,Stereochemistry ,Clinical Biochemistry ,Dose dependence ,Cancer therapy ,Pharmaceutical Science ,Antineoplastic Agents ,Mice, Transgenic ,Pharmacology ,Crystallography, X-Ray ,Biochemistry ,Mice ,Structure-Activity Relationship ,Dogs ,Pharmacokinetics ,In vivo ,Cell Line, Tumor ,Soft agar ,Drug Discovery ,Animals ,Humans ,Enzyme Inhibitors ,Molecular Biology ,Cell Proliferation ,chemistry.chemical_classification ,Aza Compounds ,Alkyl and Aryl Transferases ,Dose-Response Relationship, Drug ,Molecular Structure ,Organic Chemistry ,Haplorhini ,Neoplasms, Experimental ,Xenograft Model Antitumor Assays ,Rats ,Enzyme ,chemistry ,Molecular Medicine ,Tumor growth inhibition - Abstract
Tertiary hydroxyl class of C-imidazole bridgehead azaheptapyridine FPT inhibitors were prepared in an attempt to block in vivo oxidation of secondary hydroxyl series. One representative compound 5a exhibited potent enzyme (IC50 = 1.4 nM) and cellular activities (soft agar IC50 = 1.3 nM) with excellent oral pharmacokinetic profiles in rats, mice, monkeys and dogs. The in vivo study in wap-ras TG mouse models showed dose dependent tumor growth inhibition and regression.
- Published
- 2014
- Full Text
- View/download PDF
22. Structure of the Insulin Receptor in Complex with Insulin using Single Particle CryoEM Analysis
- Author
-
Alan Hruza, Winifred W. Prosise, Corey Strickland, Giovanna Scapin, Bridget Carragher, Venkata P. Dandey, Zhening Zhang, and Clinton S. Potter
- Subjects
0301 basic medicine ,biology ,Chemistry ,Insulin ,medicine.medical_treatment ,03 medical and health sciences ,Crystallography ,Insulin receptor ,030104 developmental biology ,0302 clinical medicine ,Biophysics ,medicine ,biology.protein ,Particle ,Instrumentation ,030217 neurology & neurosurgery - Published
- 2017
- Full Text
- View/download PDF
23. Design, Synthesis, and X-ray Crystallographic Analysis of a Novel Class of HIV-1 Protease Inhibitors
- Author
-
Andrew T. McPhail, Chih-Hung Wang, Christine Burlein, A. K. Ganguly, Yong Zhang, Steven S. Carroll, Corey Strickland, Dipshikha Biswas, Danielle Caroccia, Peter Orth, Vandna Munshi, S. Alluri, and Eunhee Kang
- Subjects
Models, Molecular ,Protein Conformation ,Thiazepines ,Stereochemistry ,medicine.medical_treatment ,Crystallography, X-Ray ,Radical cyclization ,Structure-Activity Relationship ,HIV Protease ,HIV-1 protease ,Drug Discovery ,medicine ,Protease ,Molecular Structure ,biology ,Chemistry ,Kazal-type serine protease inhibitor domain ,Stereoisomerism ,HIV Protease Inhibitors ,Crystallography ,Design synthesis ,Drug Design ,biology.protein ,Molecular Medicine ,Carbamates ,Protein Binding - Abstract
In the present paper, design, synthesis, X-ray crystallographic analysis, and HIV-1 protease inhibitory activities of a novel class of compounds are disclosed. Compounds 28-30, 32, 35, and 40 were synthesized and found to be inhibitors of the HIV-1 protease. The crucial step in their synthesis involved an unusual endo radical cyclization process. Absolute stereochemistry of the three asymmetric centers in the above compounds have been established to be (4S,2'R,3'S) for optimal potency. X-ray crystallographic analysis has been used to determine the binding mode of the inhibitors to the HIV-1 protease.
- Published
- 2011
- Full Text
- View/download PDF
24. Structure of the insulin receptor in complex with insulin using single-particle cryo-EM analysis
- Author
-
Corey Strickland, Theresa M. Kelly, Venkata P. Dandey, Zhening Zhang, Bridget Carragher, Alan Hruza, Winifred W. Prosise, Clinton S. Potter, Todd Mayhood, and Giovanna Scapin
- Subjects
biology ,Cryo-electron microscopy ,Chemistry ,Insulin ,medicine.medical_treatment ,Condensed Matter Physics ,Biochemistry ,Inorganic Chemistry ,Insulin receptor ,Structural Biology ,biology.protein ,Biophysics ,medicine ,Particle ,General Materials Science ,Physical and Theoretical Chemistry - Published
- 2018
- Full Text
- View/download PDF
25. Synthesis, Properties, and Applications of Diazotrifluropropanoyl-Containing Photoactive Analogs of Farnesyl Diphosphate Containing Modified Linkages for Enhanced Stability
- Author
-
Marisa L. Hovlid, Corey Strickland, Joshua D. Ochocki, Alan Hruza, Fernando López-Gallego, Nicholas P. Labello, Mark D. Distefano, Claudia Schmidt-Dannert, Olivier Henry, Victor G. Young, Rebecca L. Edelstein, Stepan Lenevich, Amanda J. DeGraw, and Trista Talbot
- Subjects
Saccharomyces cerevisiae Proteins ,Stereochemistry ,Farnesyltransferase ,Prenyltransferase ,Photoaffinity Labels ,Biochemistry ,Article ,Substrate Specificity ,Structure-Activity Relationship ,Polyisoprenyl Phosphates ,Drug Discovery ,Farnesyltranstransferase ,Structure–activity relationship ,Pharmacology ,Binding Sites ,Photoaffinity labeling ,ATP synthase ,biology ,Chemistry ,Organic Chemistry ,Substrate (chemistry) ,Kinetics ,biology.protein ,Molecular Medicine ,Protein prenylation ,Sesquiterpenes - Abstract
Photoactive analogs of farnesyl diphosphate (FPP) are useful probes in studies of enzymes that employ this molecule as a substrate. Here, we describe the preparation and properties of two new FPP analogs that contain diazotrifluoropropanoyl photophores linked to geranyl diphosphate via amide or ester linkages. The amide-linked analog (3) was synthesized in 32P-labeled form from geraniol in seven steps. Experiments with Saccharomyces cerevisiae protein farnesyltransferase (ScPFTase) showed that 3 is an alternative substrate for the enzyme. Photolysis experiments with [(32)P]3 demonstrate that this compound labels the beta-subunits of both farnesyltransferase and geranylgeranyltransferase (types 1 and 2). However, the amide-linked probe 3 undergoes a rearrangement to a photochemically unreactive isomeric triazolone upon long term storage making it inconvenient to use. To address this stability issue, the ester-linked analog 4 was prepared in six steps from geraniol. Computational analysis and X-ray crystallographic studies suggest that 4 binds to protein farnesyl transferase (PFTase) in a similar fashion as FPP. Compound 4 is also an alternative substrate for PFTase, and a 32P-labeled form selectively photocrosslinks the beta-subunit of ScPFTase as well as E. coli farnesyldiphosphate synthase and a germacrene-producing sesquiterpene synthase from Nostoc sp. strain PCC7120 (a cyanobacterial source). Finally, nearly exclusive labeling of ScPFTase in crude E. coli extract was observed, suggesting that [32P]4 manifests significant selectivity and should hence be useful for identifying novel FPP-utilizing enzymes in crude protein preparations.
- Published
- 2010
- Full Text
- View/download PDF
26. Application of Fragment-Based NMR Screening, X-ray Crystallography, Structure-Based Design, and Focused Chemical Library Design to Identify Novel μM Leads for the Development of nM BACE-1 (β-Site APP Cleaving Enzyme 1) Inhibitors
- Author
-
Brian M. Beyer, Vincent Madison, Eric M. Parker, Mckittrick Brian A, Daniel F. Wyss, William J. Greenlee, Corey Strickland, T. Nechuta, Johannes H. Voigt, John C. Hunter, Mary M. Senior, Andrew Stamford, Matthew E. Kennedy, Michael Czarniecki, Yu-Sen Wang, and Elizabeth M. Smith
- Subjects
Models, Molecular ,Spectrometry, Mass, Electrospray Ionization ,Magnetic Resonance Spectroscopy ,Stereochemistry ,Aminopyridines ,Crystallography, X-Ray ,Chemical synthesis ,Chemical library ,Small Molecule Libraries ,Structure-Activity Relationship ,chemistry.chemical_compound ,Drug Discovery ,Hydrolase ,Aspartic Acid Endopeptidases ,Humans ,Structure–activity relationship ,Enzyme Inhibitors ,Molecular Structure ,biology ,Chemistry ,Active site ,Nuclear magnetic resonance spectroscopy ,Small molecule ,Drug Design ,biology.protein ,Molecular Medicine ,Chemical stability ,Amyloid Precursor Protein Secretases - Abstract
Fragment-based NMR screening, X-ray crystallography, structure-based design, and focused chemical library design were used to identify novel inhibitors for BACE-1. A rapid optimization of an initial NMR hit was achieved by a combination of NMR and a functional assay, resulting in the identification of an isothiourea hit with a K(d) of 15 microM for BACE-1. NMR data and the crystal structure revealed that this hit makes H-bond interactions with the two catalytic aspartates, occupies the nonprime side region of the active site of BACE-1, and extends toward the S3 subpocket (S3sp). A focused NMR-based search for heterocyclic isothiourea isosteres resulted in several distinct classes of BACE-1 active site directed compounds with improved chemical stability and physicochemical properties. The strategy for optimization of the 2-aminopyridine lead series to potent inhibitors of BACE-1 was demonstrated. The structure-based design of a cyclic acylguanidine lead series and its optimization into nanomolar BACE-1 inhibitors are the subject of the companion paper
- Published
- 2009
- Full Text
- View/download PDF
27. Rational design of novel, potent piperazinone and imidazolidinone BACE1 inhibitors
- Author
-
Babu Suresh D, R. Kuvelkar, Yuhua Huang, A.W. Stamford, Qi Zhang, Lili Zhang, P. Gaspari, J. Pan, Wang Lingyan, L. Ozgur, Ge Li, Yusheng Wu, Johannes H. Voigt, Xiao Chen, Kurt W. Saionz, J. Lowrie, Tao Guo, Ulrich Iserloh, N.A. McHugh, D. Tadesse, Le Thuy X H, Matthew E. Kennedy, Doug W. Hobbs, Leonard Favreau, Carolyn DiIanni Carroll, Corey Strickland, Jared N. Cumming, and Eric M. Parker
- Subjects
Models, Molecular ,Imidazolidinone ,medicine.drug_class ,Stereochemistry ,Peptidomimetic ,Memapsin-2 ,Clinical Biochemistry ,Molecular Conformation ,Pharmaceutical Science ,Mice, Transgenic ,Carboxamide ,Crystallography, X-Ray ,Imidazolidines ,Biochemistry ,Piperazines ,Mice ,Structure-Activity Relationship ,Drug Discovery ,medicine ,Animals ,Aspartic Acid Endopeptidases ,Humans ,Molecular Biology ,Molecular Structure ,Chemistry ,Organic Chemistry ,Rational design ,Disease Models, Animal ,Drug Design ,β secretase ,Molecular Medicine ,Amyloid Precursor Protein Secretases ,Aspartic Endopeptidases - Abstract
Guided by structure-based design, we synthesized two novel series of potent inhibitors of BACE1 and generated extensive SAR around both the prime and non-prime side binding pockets. The key feature of both series is a cyclic amine motif specifically crafted to achieve interactions with both the flap and with the S2′ pocket.
- Published
- 2008
- Full Text
- View/download PDF
28. Key steps in the structure-based optimization of the hepatitis C virus NS3/4A protease inhibitor SCH503034
- Author
-
Liu Yi-Tsung, Frank Bennett, Viyyoor M. Girijavallabhan, F. George Njoroge, Lata Ramanathan, John Pichardo, Kevin X. Chen, S. Shane Taremi, Andrew Prongay, Brian M. Beyer, Corey Strickland, Rumin Zhang, Vincent Madison, Edwin Jao, Zhuyan Guo, Zhi Hong, Patricia C. Weber, Anil K. Saksena, Stephane L. Bogen, Srikanth Venkatraman, Rong-Sheng Yang, Nanhua Yao, Joseph E. Myers, Bruce A. Malcolm, Taisa Yarosh-Tomaine, Raymond G. Lovey, Richard N. Ingram, Ashok Arasappan, Thierry O. Fischmann, Mary M. Senior, and Winifred W. Prosise
- Subjects
HCV protease ,Models, Molecular ,Nuclear and High Energy Physics ,Diffraction Structural Biology ,Proline ,medicine.medical_treatment ,Tripeptide ,Hepacivirus ,Viral Nonstructural Proteins ,hydrophobic binding ,Hydrophobic effect ,ketoamides ,medicine ,Potency ,Protease Inhibitors ,Instrumentation ,NS3 ,Radiation ,Protease ,Kunitz STI protease inhibitor ,biology ,Molecular Structure ,Chemistry ,Active site ,structure-based design ,NS2-3 protease ,Biochemistry ,biology.protein ,sense organs - Abstract
Crystal structures of protease/inhibitor complexes guided optimization of the buried nonpolar surface area thereby maximizing hydrophobic binding. The resulting potent tripeptide inhibitor is in clinical trials., The structures of both native and S139A holo-HCV NS3/4A protease domain were solved to high resolution. Subsequently, structures were determined for a series of ketoamide inhibitors in complex with the protease. The changes in the inhibitor potency were correlated with changes in the buried surface area upon binding the inhibitor to the active site. The largest contributions to the binding energy arise from the hydrophobic interactions of the P1 and P2 groups as they bind to the S1 and S2 pockets. This correlation of the changes in potency with increased buried surface area contributed directly to the design of a potent tripeptide inhibitor of the HCV NS3/4A protease, which is currently in clinical trials.
- Published
- 2008
29. Efficacy, Pharmacokinetics, and Metabolism of Tetrahydroquinoline Inhibitors of Plasmodium falciparum Protein Farnesyltransferase
- Author
-
Andrew D. Hamilton, Oliver Hucke, Pravin Bendale, Debopam Chakrabarti, Lynn K. Barrett, Kevin D. Bauer, David K. Williams, David M. Floyd, Laxman Nallan, Brian P. Smart, Frederick S. Buckner, Carolyn P. Hornéy, Sudha Ankala, Christophe L. M. J. Verlinde, Kohei Yokoyama, Kasey Rivas, Louis J. Lombardo, Corey Strickland, Wesley C. Van Voorhis, and Michael H. Gelb
- Subjects
Male ,Models, Molecular ,Cell Membrane Permeability ,Plasmodium berghei ,Metabolite ,Farnesyltransferase ,Plasmodium falciparum ,Crystallography, X-Ray ,Intestinal absorption ,Rats, Sprague-Dawley ,Antimalarials ,Mice ,chemistry.chemical_compound ,Parasitic Sensitivity Tests ,Pharmacokinetics ,In vivo ,Animals ,Farnesyltranstransferase ,Humans ,Experimental Therapeutics ,Pharmacology (medical) ,Enzyme Inhibitors ,Pharmacology ,Mice, Inbred BALB C ,Sulfonamides ,biology ,Mutagenicity Tests ,biology.organism_classification ,Malaria ,Rats ,Bioavailability ,Infectious Diseases ,Biochemistry ,chemistry ,Dealkylation ,Microsomes, Liver ,Quinolines ,Microsome ,biology.protein ,Female ,Bile Ducts ,Caco-2 Cells - Abstract
New antimalarials are urgently needed. We have shown that tetrahydroquinoline (THQ) protein farnesyltransferase (PFT) inhibitors (PFTIs) are effective against the Plasmodium falciparum PFT and are effective at killing P. falciparum in vitro. Previously described THQ PFTIs had limitations of poor oral bioavailability and rapid clearance from the circulation of rodents. In this paper, we validate both the Caco-2 cell permeability model for predicting THQ intestinal absorption and the in vitro liver microsome model for predicting THQ clearance in vivo. Incremental improvements in efficacy, oral absorption, and clearance rate were monitored by in vitro tests; and these tests were followed up with in vivo absorption, distribution, metabolism, and excretion studies. One compound, PB-93, achieved cure when it was given orally to P. berghei- infected rats every 8 h for a total of 72 h. However, PB-93 was rapidly cleared, and dosing every 12 h failed to cure the rats. Thus, the in vivo results corroborate the in vitro pharmacodynamics and demonstrate that 72 h of continuous high-level exposure to PFTIs is necessary to kill plasmodia. The metabolism of PB-93 was demonstrated by a novel technique that relied on double labeling with a radiolabel and heavy isotopes combined with radiometric liquid chromatography and mass spectrometry. The major liver microsome metabolite of PB-93 has the PFT Zn-binding N -methyl-imidazole removed; this metabolite is inactive in blocking PFT function. By solving the X-ray crystal structure of PB-93 bound to rat PFT, a model of PB-93 bound to malarial PFT was constructed. This model suggests areas of the THQ PFTIs that can be modified to retain efficacy and protect the Zn-binding N -methyl-imidazole from dealkylation.
- Published
- 2007
- Full Text
- View/download PDF
30. Discovery of the HCV NS3/4A Protease Inhibitor (1R,5S)-N-[3-Amino-1-(cyclobutylmethyl)-2,3-dioxopropyl]-3- [2(S)-[[[(1,1-dimethylethyl)amino]carbonyl]amino]-3,3-dimethyl-1-oxobutyl]- 6,6-dimethyl-3-azabicyclo[3.1.0]hexan-2(S)-carboxamide (Sch 503034) II. Key Steps in Structure-Based Optimization
- Author
-
Nanhua Yao, Lata Ramanathan, Kevin X. Chen, Thierry O. Fischmann, Andrew Prongay, Corey Strickland, Mary M. Senior, Raymond G. Lovey, Richard N. Ingram, Bruce A. Malcolm, Viyyoor M. Girijavallabhan, Frank Bennett, Patricia C. Weber, Taisa Yarosh-Tomaine, John Pichardo, Anil K. Saksena, F. George Njoroge, Rong-Sheng Yang, Zhuyan Guo, Zhi Hong, Stephane L. Bogen, Srikanth Venkatraman, Ashok Arasappan, S. Shane Taremi, Brian M. Beyer, Rumin Zhang, Vincent Madison, Edwin Jao, Winifred W. Prosise, Yi-Tsung Liu, and Joseph E. Myers
- Subjects
Alanine ,chemistry.chemical_classification ,NS3 ,Dipeptide ,Protease ,Stereochemistry ,medicine.drug_class ,medicine.medical_treatment ,virus diseases ,Peptide ,Carboxamide ,macromolecular substances ,digestive system diseases ,Protease inhibitor (biology) ,Serine ,chemistry.chemical_compound ,chemistry ,Drug Discovery ,medicine ,Molecular Medicine ,medicine.drug - Abstract
The structures of both the native holo-HCV NS3/4A protease domain and the protease domain with a serine 139 to alanine (S139A) mutation were solved to high resolution. Subsequently, structures were...
- Published
- 2007
- Full Text
- View/download PDF
31. Iminopyrimidinones: a novel pharmacophore for the development of orally active renin inhibitors
- Author
-
William J. Greenlee, Rumin Zhang, Linda B. Fleming, Ulrich Iserloh, Murali Rajagopalan, Tanweer A. Khan, Rachel E. Giessert, Madhu Chintala, Jesse K. Wong, Brandy Courneya, Peter Orth, Johannes H. Voigt, Liwu Hong, Charles R. Heap, Hongwu Wang, Robert Mazzola, Andrew J. Zych, Thomas Bara, Walter A. Korfmacher, Kristina Moore, Samuel A. Sakwa, Henry M. Vaccaro, Sudipta Roy, John W. Clader, John P. Caldwell, Michael Czarniecki, Ruth A. Duffy, Mckittrick Brian A, Corey Strickland, George Boykow, Ying Huang, Soumya Mitra, Xian Liang, and Josien Hubert B
- Subjects
Models, Molecular ,medicine.drug_class ,Clinical Biochemistry ,Pharmaceutical Science ,Administration, Oral ,Pyrimidinones ,Pharmacology ,Biochemistry ,Renin inhibitor ,Structure-Activity Relationship ,Aspartate protease ,Pharmacokinetics ,Drug Discovery ,Renin–angiotensin system ,Renin ,medicine ,Enzyme Inhibitors ,Molecular Biology ,Dose-Response Relationship, Drug ,Molecular Structure ,Chemistry ,Organic Chemistry ,Orally active ,Molecular Medicine ,Imines ,Pharmacophore - Abstract
The development of renin inhibitors with favorable oral pharmacokinetic profiles has been a longstanding challenge for the pharmaceutical industry. As part of our work to identify inhibitors of BACE1, we have previously developed iminopyrimidinones as a novel pharmacophore for aspartyl protease inhibition. In this letter we describe how we modified substitution around this pharmacophore to develop a potent, selective and orally active renin inhibitor.
- Published
- 2015
32. Guiding farnesyltransferase inhibitors from an ECLiPS® library to the catalytic zinc
- Author
-
Baldwin John J, Ashit K. Ganguly, He Huang, Laura L. Rokosz, Alan B. Cooper, Doll Ronald J, Chia-Yu Huang, John C. Reader, Ge Li, Corey Strickland, and Tara M. Stauffer
- Subjects
Molecular model ,Stereochemistry ,Farnesyltransferase ,Clinical Biochemistry ,Pharmaceutical Science ,Drug design ,chemistry.chemical_element ,Zinc ,Crystallography, X-Ray ,Biochemistry ,Chemical synthesis ,Catalysis ,Inhibitory Concentration 50 ,Mice ,Structure-Activity Relationship ,Peptide Library ,Drug Discovery ,Animals ,Farnesyltranstransferase ,Enzyme Inhibitors ,Molecular Biology ,chemistry.chemical_classification ,Farnesyl-diphosphate farnesyltransferase ,Binding Sites ,biology ,Chemistry ,Organic Chemistry ,Active site ,Enzyme ,NIH 3T3 Cells ,biology.protein ,Molecular Medicine - Abstract
Farnesyltransferase inhibitors identified from an ECLiPS® library were optimized using solution-phase synthesis. X-ray crystallography of inhibited complexes was used to identify substructures that coordinate to the active site zinc. The X-ray structures were ultimately used to guide the design of second-generation analogs with FTase IC50s of less than 1.0 nM.
- Published
- 2006
- Full Text
- View/download PDF
33. Bridgehead modification of trihalocycloheptabenzopyridine lead to a potent farnesyl protein transferase inhibitor with improved oral metabolic stability
- Author
-
Corey Strickland, Bancha Vibulbhan, Robert Bishop, Xiongwei Shi, Paul Kirschmeier, F. George Njoroge, Amin A. Nomeir, and Viyyoor M. Girijavallabhan
- Subjects
Farnesyl Protein Transferase ,Pyridines ,Stereochemistry ,Clinical Biochemistry ,Molecular Conformation ,Administration, Oral ,Pharmaceutical Science ,Biochemistry ,Chemical synthesis ,chemistry.chemical_compound ,Drug Stability ,Amide ,Drug Discovery ,Moiety ,Enzyme Inhibitors ,Molecular Biology ,Antitumor activity ,chemistry.chemical_classification ,Farnesyl-diphosphate farnesyltransferase ,Alkyl and Aryl Transferases ,biology ,Organic Chemistry ,Biological activity ,General Medicine ,Metabolic stability ,Sulfonamide ,chemistry ,Enzyme inhibitor ,Urea ,biology.protein ,Molecular Medicine ,Piperidine ,Protein Binding - Abstract
Modification of the ethano bridge of the core structure of the antitumor agent, SARASAR® (SCH66336) with concomitant introduction of a sulfonamide moiety off the distal piperidine afforded inhibitor 9-(S-), a compound with greatly improved PK profile. Other compounds with enhanced FPTase inhibitory activity were obtained as exemplified by amide 10-(S-) and urea 11-(S-): these compounds demonstrated activity in picomolar range.
- Published
- 2004
- Full Text
- View/download PDF
34. Crystallization of glycosylated human BACE protease domain expressed in Trichoplusia ni
- Author
-
Brian M. Beyer, Wenyan Wang, Julie Lee, Lili Zhang, S. Shane Taremi, Hung V. Le, Paul Reichert, Corey Strickland, JianJun Liu, and Yan-Hui Liu
- Subjects
PNGase F ,Glycosylation ,medicine.medical_treatment ,Biophysics ,Moths ,Biochemistry ,Analytical Chemistry ,chemistry.chemical_compound ,Protein structure ,Zymogen ,Endopeptidases ,medicine ,Trichoplusia ,Animals ,Aspartic Acid Endopeptidases ,Humans ,Molecular replacement ,Cloning, Molecular ,Molecular Biology ,Cells, Cultured ,Protease ,biology ,biology.organism_classification ,Protein Structure, Tertiary ,Transmembrane domain ,chemistry ,Amyloid Precursor Protein Secretases ,Crystallization ,Peptides - Abstract
Human beta-amyloid precursor protein cleaving enzyme (beta-secretase, or BACE) belongs to the aspartyl protease family, and is responsible for generating the N-terminus of beta-amyloid peptide (Abeta). BACE is a type I transmembrane glycoprotein with pre-, pro- and catalytic domains, a short transmembrane helix and a cytoplasmic region. In this study, a truncated form was engineered to produce the authentic catalytic domain of BACE in Trichoplusia ni (High 5) cells. The glycosylated BACE zymogen (proBACE) was secreted into the conditioned medium for facile purification by metal chelate and gel filtration chromatographies. The mature catalytic domain was obtained by a trans cleavage event under acidic conditions and crystallized in the absence of a bound inhibitor. A complete 3.4 A data set was collected on a single orthorhombic crystal with unit cell parameters a=74 A, b=130 A, c=134A. Successful molecular replacement shows two BACE molecules in the asymmetric unit.
- Published
- 2004
- Full Text
- View/download PDF
35. Crystal Structure of the Catalytic Domain of Human ADAM33
- Author
-
Jun Zou, Winifred W. Prosise, Urooj A. Mirza, Peter Orth, Hung V. Le, S. Shane Taremi, Li Xiao, Paul Reichert, Taisa Yarosh-Tomaine, Vincent Madison, Richard N. Ingram, Hammond Gerald S, Corey Strickland, and Wenyan Wang
- Subjects
Models, Molecular ,Cell signaling ,Biology ,Crystallography, X-Ray ,Hydroxamic Acids ,Structural Biology ,Catalytic Domain ,Hydrolase ,Disintegrin ,Humans ,Amino Acid Sequence ,Enzyme Inhibitors ,Molecular Biology ,chemistry.chemical_classification ,Metalloproteinase ,Cell fusion ,Molecular Structure ,Metalloendopeptidases ,Active site ,Protein Structure, Tertiary ,respiratory tract diseases ,carbohydrates (lipids) ,ADAM Proteins ,Enzyme ,chemistry ,Biochemistry ,Mutation ,biology.protein ,Glycoprotein ,Sequence Alignment ,Protein Binding - Abstract
Adam33 is a putative asthma susceptibility gene encoding for a membrane-anchored metalloprotease belonging to the ADAM family. The ADAMs (a disintegrin and metalloprotease) are a family of glycoproteins implicated in cell-cell interactions, cell fusion, and cell signaling. We have determined the crystal structure of the Adam33 catalytic domain in complex with the inhibitor marimastat and the inhibitor-free form. The structures reveal the polypeptide fold and active site environment resembling that of other metalloproteases. The substrate-binding site contains unique features that allow the structure-based design of specific inhibitors of this enzyme.
- Published
- 2004
- Full Text
- View/download PDF
36. Biochemical and Structural Studies with Prenyl Diphosphate Analogues Provide Insights into Isoprenoid Recognition by Protein Farnesyl Transferase
- Author
-
Corey Strickland, Mark D. Distefano, and Tammy C. Turek-Etienne
- Subjects
Models, Molecular ,Stereochemistry ,Protein Prenylation ,Peptide ,Photoaffinity Labels ,Saccharomyces cerevisiae ,Crystallography, X-Ray ,Biochemistry ,Substrate Specificity ,Structure-Activity Relationship ,Polyisoprenyl Phosphates ,Prenylation ,Humans ,Transferase ,Farnesyl Transferase ,chemistry.chemical_classification ,Alkyl and Aryl Transferases ,Binding Sites ,Chemistry ,organic chemicals ,A protein ,Terpenoid ,Yeast ,Enzyme ,lipids (amino acids, peptides, and proteins) ,Diterpenes ,Sesquiterpenes ,Protein Binding - Abstract
Protein farnesyl transferase (PFTase) catalyzes the reaction between farnesyl diphosphate and a protein substrate to form a thioether-linked prenylated protein. The fact that many prenylated proteins are involved in signaling processes has generated considerable interest in protein prenyl transferases as possible anticancer targets. While considerable progress has been made in understanding how prenyl transferases distinguish between related target proteins, the rules for isoprenoid discrimination by these enzymes are less well understood. To clarify how PFTase discriminates between FPP and larger prenyl diphosphates, we have examined the interactions between the enzyme and several isoprenoid analogues, GGPP, and the farnesylated peptide product using a combination of biochemical and structural methods. Two photoactive isoprenoid analogues were shown to inhibit yeast PFTase with K(I) values as low as 45 nM. Crystallographic analysis of one of these analogues bound to PFTase reveals that the diphosphate moiety and the two isoprene units bind in the same positions occupied by the corresponding atoms in FPP when bound to PFTase. However, the benzophenone group protrudes into the acceptor protein binding site and prevents the binding of the second (protein) substrate. Crystallographic analysis of geranylgeranyl diphosphate bound to PFTase shows that the terminal two isoprene units and diphosphate group of the molecule map to the corresponding atoms in FPP; however, the first and second isoprene units bulge away from the acceptor protein binding site. Comparison of the GGPP binding mode with the binding of the farnesylated peptide product suggests that the bulkier isoprenoid cannot rearrange to convert to product without unfavorable steric interactions with the acceptor protein. Taken together, these data do not support the "molecular ruler hypotheses". Instead, we propose a "second site exclusion model" in which PFTase binds larger isoprenoids in a fashion that prevents the subsequent productive binding of the acceptor protein or its conversion to product.
- Published
- 2003
- Full Text
- View/download PDF
37. Trihalobenzocycloheptapyridine analogues of Sch 66336 as potent inhibitors of farnesyl protein transferase
- Author
-
V. Girijavallabhan, Paul Kirschmeir, Patrick Pinto, Corey Strickland, F. George Njoroge, Bancha Vibulbhan, W. Robert Bishop, and Ashit K. Ganguly
- Subjects
Models, Molecular ,Farnesyl Protein Transferase ,Pyridines ,medicine.drug_class ,Stereochemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Antineoplastic Agents ,Carboxamide ,Kidney ,Tritium ,Biochemistry ,Chemical synthesis ,Structure-Activity Relationship ,chemistry.chemical_compound ,Piperidines ,Drug Discovery ,medicine ,Animals ,Enzyme Inhibitors ,Molecular Biology ,chemistry.chemical_classification ,Farnesyl-diphosphate farnesyltransferase ,Alkyl and Aryl Transferases ,biology ,Organic Chemistry ,Enzyme ,Solubility ,chemistry ,Enzyme inhibitor ,COS Cells ,biology.protein ,Molecular Medicine ,Piperidine ,Tricyclic - Abstract
SCH 66336 is a trihalo tricyclic compound that is currently undergoing Phase II clinical trials for the treatment of solid tumors. Modifications of SCH 66336 by incorporating such groups as amides, acids, esters, ureas and lactams off the first or the distal piperidine (from the tricycle) provided potent FPT inhibitors some of which exhibited good cellular activity. A number of these compounds incorporate properties that might improve pharmacokinetic stability of these inhibitors by virtue of their increased solubility or by their change in log P.
- Published
- 2003
- Full Text
- View/download PDF
38. Exploring the Role of Bromine at C(10) of (+)-4-[2-[4-(8-Chloro-3,10-dibromo- 6,11-dihydro-5H-benzo[5,6]cyclohepta[1,2-b]pyridin-11(R)-yl)-1-piperidinyl]-2- oxoethyl]-1-piperidinecarboxamide (Sch-66336): The Discovery of Indolocycloheptapyridine Inhibitors of Farnesyl Protein Transferase
- Author
-
Lalwani Tarik, Patricia C. Weber, Paul Kirschmeier, Mark E. Snow, Ming Liu, Amin A. Nomeir, Linda James, Viyyoor M. Girijavallabhan, William T. Windsor, Robert Patton, Aki Cynthia J, Corey Strickland, Frank Hollinger, Doll Ronald J, Chao Jianping, and Taveras Arthur G
- Subjects
Models, Molecular ,Indoles ,Farnesyl Protein Transferase ,Pyridines ,Stereochemistry ,medicine.drug_class ,Substituent ,Mice, Nude ,Antineoplastic Agents ,Carboxamide ,Crystallography, X-Ray ,Chemical synthesis ,Mice ,Structure-Activity Relationship ,chemistry.chemical_compound ,Piperidines ,Drug Discovery ,medicine ,Animals ,Moiety ,Enzyme Inhibitors ,Indole test ,Farnesyl-diphosphate farnesyltransferase ,Alkyl and Aryl Transferases ,Chemistry ,Farnesyl Transferase Inhibitor ,Bromine ,Thermodynamics ,Molecular Medicine ,Half-Life - Abstract
The 10-bromobenzocycloheptapyridyl farnesyl transferase inhibitor (FTI) Sch-66336 (1) is currently under clinical evaluation for the treatment of human cancers. During structure-activity relationship development leading to 1, 10-bromobenzocycloheptapyridyl FTIs were found to be more potent than analogous compounds lacking the 10-Br substituent. This potency enhancement was believed to be due, in part, to an increase in conformational rigidity as the 10-bromo substituent could restrict the conformation of the appended C(11) piperidyl substituent in an axial orientation. A novel and potent class of FTIs, represented by indolocycloheptapyridine Sch-207758 [(+)-10a], have been designed based on this principle. Although structural and thermodynamic results suggest that entropy plays a crucial role in the increased potency observed with (+)-10a through conformational constraints and solvation effects, the results also indicate that the indolocycloheptapyridine moiety in (+)-10a provides increased hydrophobic interactions with the protein through the addition of the indole group. This report details the X-ray structure and the thermodynamic and pharmacokinetic profiles of (+)-10a, as well as the synthesis of indolocycloheptapyridine FTIs and their potencies in biochemical and biological assays.
- Published
- 2002
- Full Text
- View/download PDF
39. Synthesis of 5,6-Dihydro-11H-benzo[5,6]-cyclohepta[1,2-b]pyridin-11-ylidene)-1-piperidine-N-cyanoguanidine Derivatives as Inhibitors of Ras Farnesyl Protein Transferase
- Author
-
Patricia C. Weber, Alan B. Cooper, Robert Patton, Paul Kirschmeier, W. Robert Bishop, V. Girijavallabhan, Corey Strickland, Jagdish A. Desai, and James Wang
- Subjects
Farnesyl Protein Transferase ,Stereochemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Crystallography, X-Ray ,Guanidines ,Heterocyclic Compounds, 4 or More Rings ,Biochemistry ,Chemical synthesis ,Inhibitory Concentration 50 ,Structure-Activity Relationship ,chemistry.chemical_compound ,Piperidines ,Amide ,Drug Discovery ,Humans ,Enzyme Inhibitors ,Molecular Biology ,chemistry.chemical_classification ,Farnesyl-diphosphate farnesyltransferase ,Alkyl and Aryl Transferases ,Molecular Structure ,biology ,Organic Chemistry ,Enzyme ,chemistry ,Enzyme inhibitor ,biology.protein ,Molecular Medicine ,Piperidine ,Tricyclic - Abstract
A series of novel N-cyanoguanidine tricyclic farnesyl protein transferase (FPT) inhibitors was prepared. Replacement of a piperidine amide-group with a N-cyanoguanidine functionality increased FPT activity. X-ray crystal structure determination of 42 complexed with FPT revealed differences in the interactions of the amide and N-cyanoguanidine groups with the protein.
- Published
- 2002
- Full Text
- View/download PDF
40. Synthesis of Farnesyl Diphosphate Analogues Containing Ether-Linked Photoactive Benzophenones and Their Application in Studies of Protein Prenyltransferases
- Author
-
Igor Gaon, Tammy C. Turek, Mark D. Distefano, and Corey Strickland
- Subjects
Models, Molecular ,Photochemistry ,Stereochemistry ,Farnesyltransferase ,Antineoplastic Agents ,Peptide ,Crystallography, X-Ray ,Benzophenones ,Inhibitory Concentration 50 ,Structure-Activity Relationship ,Polyisoprenyl Phosphates ,Prenylation ,Yeasts ,Animals ,Humans ,Enzyme Inhibitors ,chemistry.chemical_classification ,biology ,Photoaffinity labeling ,Chemistry ,Organic Chemistry ,Dimethylallyltranstransferase ,Rats ,Enzyme ,Biochemistry ,biology.protein ,Protein prenylation ,Lipid modification ,Phosphorus Radioisotopes ,Sesquiterpenes ,Ethers ,Cysteine - Abstract
Protein prenylation is a posttranslational lipid modification in which C(15) and C(20) isoprenoid units are linked to specific protein-derived cysteine residues through a thioether linkage. This process is catalyzed by a class of enzymes called prenyltransferases that are being intensively studied due to the finding that Ras protein is farnesylated coupled with the observation that mutant forms of Ras are implicated in a variety of human cancers. Inhibition of this posttranslational modification may serve as a possible cancer chemotherapy. Here, the syntheses of two new farnesyl diphosphate (FPP) analogues containing photoactive benzophenone groups are described. Each of these compounds was prepared in six steps from dimethylallyl alcohol. Substrate studies, inhibition kinetics, photoinactivation studies, and photolabeling experiments are also included; these experiments were performed with a number of protein prenyltransferases from different sources. A X-ray crystal structure of one of these analogues bound to rat farnesyltransferase illustrates that they are good substrate mimics. Of particular importance, these new analogues can be enzymatically incorporated into Ras-based peptide substrates allowing the preparation of molecules with photoactive isoprenoids that may serve as valuable probes for the study of prenylation function. Photoaffinity labeling of human protein geranylgeranyltransferase with (32)P-labeled forms of these analogues suggests that the C-10 locus of bound geranylgeranyl diphosphate (GGPP) is in close proximity to residues from the beta-subunit of this enzyme. These results clearly demonstrate the utility of these compounds as photoaffinity labeling analogues for the study of a variety of protein prenyltransferases and other enzymes that employ FPP or GGPP as their substrates.
- Published
- 2001
- Full Text
- View/download PDF
41. Discovery of C-imidazole azaheptapyridine FPT inhibitors
- Author
-
Alan Hruza, Doll Ronald J, Corey Strickland, George F. Njoroge, Viyyoor M. Girijavallabhan, Paul Kirschmeier, Hugh Y. Zhu, Alan B. Cooper, W. Robert Bishop, and Jagdish A. Desai
- Subjects
Pyridines ,Stereochemistry ,Clinical Biochemistry ,hERG ,Pharmaceutical Science ,Crystallography, X-Ray ,Biochemistry ,Chemical synthesis ,chemistry.chemical_compound ,Cell Line, Tumor ,Drug Discovery ,Farnesyltranstransferase ,Humans ,Transferase ,Imidazole ,Molecular Biology ,chemistry.chemical_classification ,biology ,Chemistry ,Organic Chemistry ,Imidazoles ,Active site ,Biological activity ,Enzyme ,Enzyme inhibitor ,biology.protein ,Molecular Medicine - Abstract
The discovery of C-linked imidazole azaheptapyridine bridgehead FPT inhibitors is described. This novel class of compounds are sub nM FPT enzyme inhibitors with potent cellular inhibitory activities. This series also has reduced hERG activity versus previous N-linked imidazole series. X-ray of compound 10a bound to FTase revealed strong interaction between bridgehead imidazole 3N with catalytic zinc atom.
- Published
- 2010
- Full Text
- View/download PDF
42. Crystal Structure of Farnesyl Protein Transferase Complexed with a CaaX Peptide and Farnesyl Diphosphate Analogue
- Author
-
Jeffrey Schwartz, Patricia C. Weber, Rosalinda Syto, Zhen Wu, Lorena S. Beese, William T. Windsor, Corey Strickland, Hung V. Le, Lynn Wang, and Richard Bond
- Subjects
Models, Molecular ,chemistry.chemical_classification ,Alkyl and Aryl Transferases ,Binding Sites ,Farnesyl Protein Transferase ,Macromolecular Substances ,Chemistry ,Organophosphonates ,Peptide ,Crystal structure ,Crystallography, X-Ray ,Farnesol ,Biochemistry ,Protein Structure, Secondary ,Protein Structure, Tertiary ,Rats ,Substrate Specificity ,Polyisoprenyl Phosphates ,Animals ,Humans ,Transferase ,Crystallization ,Oligopeptides ,Sesquiterpenes - Abstract
The crystallographic structure of acetyl-Cys-Val-Ile-selenoMet-COOH and alpha-hydroxyfarnesylphosphonic acid (alphaHFP) complexed with rat farnesyl protein transferase (FPT) (space group P61, a = b = 174. 13 A, c = 69.71 A, alpha = beta = 90 degrees, gamma = 120 degrees, Rfactor = 21.8%, Rfree = 29.2%, 2.5 A resolution) is reported. In the ternary complex, the bound substrates are within van der Waals contact of each other and the FPT enzyme. alphaHFP binds in an extended conformation in the active-site cavity where positively charged side chains and solvent molecules interact with the phosphate moiety and aromatic side chains pack adjacent to the isoprenoid chain. The backbone of the bound CaaX peptide adopts an extended conformation, and the side chains interact with both FPT and alphaHFP. The cysteine sulfur of the bound peptide coordinates the active-site zinc. Overall, peptide binding and recognition appear to be dominated by side-chain interactions. Comparison of the structures of the ternary complex and unliganded FPT [Park, H., Boduluri, S., Moomaw, J., Casey, P., and Beese, L. (1997) Science 275, 1800-1804] shows that major rearrangements of several active site side chains occur upon substrate binding.
- Published
- 1998
- Full Text
- View/download PDF
43. Discovery of an Orally Available, Brain Penetrant BACE1 Inhibitor that Affords Robust CNS Aβ Reduction
- Author
-
Peter Orth, Kathleen Cox, Corey Strickland, Rachael C. Hunter, Reshma Kuvelkar, Eric J. Gilbert, Lynn A. Hyde, Jared N. Cumming, Sarah W. Li, Chunli Huang, Eric M. Parker, Johannes H. Voigt, Yusheng Wu, Matthew E. Kennedy, Andrew Stamford, William J. Greenlee, Zhaoning Zhu, Dawit Tadesse, Liwu Hong, Babu Suresh D, Misiaszek Jeffrey A, Xia Chen, Tao Guo, Robert A. Hodgson, Jack D. Scott, Mckittrick Brian A, and Leonard Favreau
- Subjects
chemistry.chemical_compound ,Chemistry ,Drug discovery ,mental disorders ,Organic Chemistry ,Drug Discovery ,Aβ peptide ,Systemic administration ,Pharmacology ,Penetrant (biochemical) ,Biochemistry - Abstract
Inhibition of BACE1 to prevent brain Aβ peptide formation is a potential disease-modifying approach to the treatment of Alzheimer’s disease. Despite over a decade of drug discovery efforts, the identification of brain-penetrant BACE1 inhibitors that substantially lower CNS Aβ levels following systemic administration remains challenging. In this report we describe structure-based optimization of a series of brain-penetrant BACE1 inhibitors derived from an iminopyrimidinone scaffold. Application of structure-based design in tandem with control of physicochemical properties culminated in the discovery of compound 16, which potently reduced cortex and CSF Aβ40 levels when administered orally to rats.
- Published
- 2013
44. Inhibitors of BACE for treating Alzheimer's disease: a fragment-based drug discovery story
- Author
-
Corey Strickland and Andrew Stamford
- Subjects
Ligand efficiency ,Stereochemistry ,Chemistry ,Fragment-based lead discovery ,Patent literature ,Biochemistry ,Analytical Chemistry ,Amidine ,chemistry.chemical_compound ,Alzheimer Disease ,Drug Design ,Drug Discovery ,Animals ,Aspartic Acid Endopeptidases ,Humans ,Pharmacophore ,Amyloid Precursor Protein Secretases ,Enzyme Inhibitors - Abstract
Several fragment-based methods have been applied to the discovery of new lead sources for inhibitors of BACE1, an important therapeutic target for Alzheimer's disease. Among the most common fragment hits were various amidine-containing molecules in which the amidine engaged in discrete H-bond donor-acceptor interaction with the BACE1 catalytic dyad. Structure and medicinal chemistry knowledge-based optimization with emphasis on ligand efficiency resulted in identification of a key pharmacophore comprising a non-planar cyclic amidine scaffold directly attached to a phenyl group projecting into S1. This key pharmacophore is a common feature of known clinical candidates and has dominated the recent patent literature. A structural comparison of the non-planar cyclic amidine motif with other BACE1 pharmacophores highlights its uniqueness and distinct advantages.
- Published
- 2013
45. Design and validation of bicyclic iminopyrimidinones as beta amyloid cleaving enzyme-1 (BACE1) inhibitors: conformational constraint to favor a bioactive conformation
- Author
-
William J. Greenlee, Lili Zhang, Leonard Favreau, Qi Zhang, John P. Caldwell, Reshma Kuvelkar, Giuseppe Terracina, Johannes H. Voigt, Irina Kazakevich, Robert Mazzola, Eric M. Parker, Xia Chen, Lynn A. Hyde, Mckittrick Brian A, Kathleen Cox, Michael Grzelak, Prescott T. Leach, Peter Orth, Hongwu Wang, Zhaoning Zhu, Alexei V. Buevich, Xiaoxiang Liu, Matthew E. Kennedy, Corey Strickland, Mihirbaran Mandal, Jared N. Cumming, and Andrew Stamford
- Subjects
Models, Molecular ,Stereochemistry ,Substituent ,Molecular Conformation ,Administration, Oral ,Stereoisomerism ,Pyrimidinones ,Thiophenes ,Ring (chemistry) ,Crystallography, X-Ray ,Pyrrolidine ,Permeability ,chemistry.chemical_compound ,Structure-Activity Relationship ,Drug Discovery ,Nitriles ,Structure–activity relationship ,Molecule ,Animals ,Aspartic Acid Endopeptidases ,Humans ,chemistry.chemical_classification ,Cerebral Cortex ,Amyloid beta-Peptides ,Bicyclic molecule ,Bridged Bicyclo Compounds, Heterocyclic ,Peptide Fragments ,Rats ,Macaca fascicularis ,Enzyme ,HEK293 Cells ,Pyrimidines ,chemistry ,Molecular Medicine ,Quantum Theory ,Thermodynamics ,Amyloid Precursor Protein Secretases - Abstract
On the basis of our observation that the biaryl substituent of iminopyrimidinone 7 must be in a pseudoaxial conformation to occupy the contiguous S1-S3 subsites of BACE1, we have designed a novel fused bicyclic iminopyrimidinone scaffold intended to favor this bioactive conformation. Strategic incorporation of a nitrogen atom in the new constrained ring allowed us to develop SAR around the S2' binding pocket and ultimately resulted in analogues with low nanomolar potency for BACE1. In particular, optimization of the prime side substituent led to major improvements in potency by displacement of two conserved water molecules from a region near S2'. Further optimization of the pharmacokinetic properties of this fused pyrrolidine series, in conjunction with facile access to a rat pharmacodynamic model, led to identification of compound 43, which is an orally active, brain penetrant inhibitor that reduces Aβ(40) in the plasma, CSF, and cortex of rats in a dose-dependent manner.
- Published
- 2012
46. Structure based design of iminohydantoin BACE1 inhibitors: identification of an orally available, centrally active BACE1 inhibitor
- Author
-
Jianping Pan, Yusheng Wu, Matthew E. Kennedy, Eric M. Parker, James Durkin, Misiaszek Jeffrey A, William J. Greenlee, Jared N. Cumming, Corey Strickland, Xia Chen, Elizabeth M. Smith, Leonard Favreau, Johannes H. Voigt, Zhaoning Zhu, Kathleen Cox, Michael Czarniecki, Andrew Stamford, Wang Lingyan, Mckittrick Brian A, Reshma Kuvelkar, Lynn A. Hyde, and Ulrich Iserloh
- Subjects
Models, Molecular ,Clinical Biochemistry ,Pharmaceutical Science ,Administration, Oral ,Pharmacology ,Crystallography, X-Ray ,Biochemistry ,Rats, Sprague-Dawley ,Oral administration ,In vivo ,Alzheimer Disease ,mental disorders ,Drug Discovery ,Animals ,Aspartic Acid Endopeptidases ,Humans ,Computer Simulation ,Enzyme Inhibitors ,Molecular Biology ,Amyloid beta-Peptides ,Binding Sites ,Chemistry ,Hydantoins ,Organic Chemistry ,Rats ,Sprague dawley ,Disease Models, Animal ,Drug Design ,β secretase ,Molecular Medicine ,Structure based ,Anticonvulsants ,Amyloid Precursor Protein Secretases ,Protein Binding - Abstract
From an initial lead 1, a structure-based design approach led to identification of a novel, high-affinity iminohydantoin BACE1 inhibitor that lowers CNS-derived Aβ following oral administration to rats. Herein we report SAR development in the S3 and F' subsites of BACE1 for this series, the synthetic approaches employed in this effort, and in vivo data for the optimized compound.
- Published
- 2012
47. Combining NMR and X-ray crystallography in fragment-based drug discovery: discovery of highly potent and selective BACE-1 inhibitors
- Author
-
Daniel F, Wyss, Yu-Sen, Wang, Hugh L, Eaton, Corey, Strickland, Johannes H, Voigt, Zhaoning, Zhu, and Andrew W, Stamford
- Subjects
Models, Molecular ,Small Molecule Libraries ,Structure-Activity Relationship ,Drug Discovery ,Amyloid Precursor Protein Secretases ,Enzyme Inhibitors ,Crystallography, X-Ray ,Nuclear Magnetic Resonance, Biomolecular ,High-Throughput Screening Assays - Abstract
Fragment-based drug discovery (FBDD) has become increasingly popular over the last decade. We review here how we have used highly structure-driven fragment-based approaches to complement more traditional lead discovery to tackle high priority targets and those struggling for leads. Combining biomolecular nuclear magnetic resonance (NMR), X-ray crystallography, and molecular modeling with structure-assisted chemistry and innovative biology as an integrated approach for FBDD can solve very difficult problems, as illustrated in this chapter. Here, a successful FBDD campaign is described that has allowed the development of a clinical candidate for BACE-1, a challenging CNS drug target. Crucial to this achievement were the initial identification of a ligand-efficient isothiourea fragment through target-based NMR screening and the determination of its X-ray crystal structure in complex with BACE-1, which revealed an extensive H-bond network with the two active site aspartate residues. This detailed 3D structural information then enabled the design and validation of novel, chemically stable and accessible heterocyclic acylguanidines as aspartic acid protease inhibitor cores. Structure-assisted fragment hit-to-lead optimization yielded iminoheterocyclic BACE-1 inhibitors that possess desirable molecular properties as potential therapeutic agents to test the amyloid hypothesis of Alzheimer's disease in a clinical setting.
- Published
- 2011
48. Combining NMR and X-ray Crystallography in Fragment-Based Drug Discovery: Discovery of Highly Potent and Selective BACE-1 Inhibitors
- Author
-
Yu-Sen Wang, Daniel F. Wyss, Andrew Stamford, Corey Strickland, Zhaoning Zhu, Hugh L. Eaton, and Johannes H. Voigt
- Subjects
biology ,Molecular model ,Chemistry ,Drug discovery ,Fragment-based lead discovery ,Active site ,Computational biology ,Integrated approach ,Protease inhibitor (biology) ,Fragment (logic) ,biology.protein ,medicine ,Structure–activity relationship ,medicine.drug - Abstract
Fragment-based drug discovery (FBDD) has become increasingly popular over the last decade. We review here how we have used highly structure-driven fragment-based approaches to complement more traditional lead discovery to tackle high priority targets and those struggling for leads. Combining biomolecular nuclear magnetic resonance (NMR), X-ray crystallography, and molecular modeling with structure-assisted chemistry and innovative biology as an integrated approach for FBDD can solve very difficult problems, as illustrated in this chapter. Here, a successful FBDD campaign is described that has allowed the development of a clinical candidate for BACE-1, a challenging CNS drug target. Crucial to this achievement were the initial identification of a ligand-efficient isothiourea fragment through target-based NMR screening and the determination of its X-ray crystal structure in complex with BACE-1, which revealed an extensive H-bond network with the two active site aspartate residues. This detailed 3D structural information then enabled the design and validation of novel, chemically stable and accessible heterocyclic acylguanidines as aspartic acid protease inhibitor cores. Structure-assisted fragment hit-to-lead optimization yielded iminoheterocyclic BACE-1 inhibitors that possess desirable molecular properties as potential therapeutic agents to test the amyloid hypothesis of Alzheimer’s disease in a clinical setting.
- Published
- 2011
- Full Text
- View/download PDF
49. Crystallization of an apo form of human arginase: using all the tools in the toolbox simultaneously
- Author
-
Janet Newman, Lesley A. Pearce, Corey Strickland, Thomas S. Peat, and Charles A. Lesburg
- Subjects
inorganic chemicals ,Stereochemistry ,Molecular Sequence Data ,Biophysics ,macromolecular substances ,Biology ,Biochemistry ,Isozyme ,law.invention ,chemistry.chemical_compound ,Structural Biology ,law ,Genetics ,Animals ,Humans ,Crystallization ,Aminohydrolase ,chemistry.chemical_classification ,Arginase ,organic chemicals ,technology, industry, and agriculture ,Ornithine ,Condensed Matter Physics ,Isoenzymes ,Enzyme ,chemistry ,Crystallization Communications ,embryonic structures ,Urea - Abstract
Arginase (EC 3.5.3.1) is an aminohydrolase that acts on l-arginine to produce urea and ornithine. Two isotypes of the enzyme are found in humans. Type I is predominantly produced in the liver and is a homotrimer of 35 kDa subunits. Human arginase (hArginase) I is seen to be up-regulated in many diseases and is a potential therapeutic target for many diverse indications. Previous reports of crystallization and structure determination of hArginase have always included inhibitors of the enzyme: here, the first case of a true apo crystal form of the enzyme which is suitable for small-molecule soaking is reported. The crystals belonged to space group P212121 and have approximate unit-cell parameters a = 53, b = 67.5, c = 250 A. The crystals showed slightly anisotropic diffraction to beyond 2.0 A resolution.
- Published
- 2010
50. P3‐272: Pharmacological characterization of novel iminoheterocyclic BACE1 inhibitors for the treatment of Alzheimer's disease
- Author
-
Reshma Kuvelkar, Brian Beyer, Ulrich Iserloh, Zhaoning Zhu, Yu-Sen Wang, Janping Pan, Elizabeth Smith, Jared N. Cumming, Mckittrick Brian A, Corey Strickland, Andrew Stamford, Eric M. Parker, Xia Chen, Johannes H. Voigt, Zhong-Yue Sun, Daniel F. Wyss, and Matthew E. Kennedy
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,business.industry ,Health Policy ,Medicine ,Neurology (clinical) ,Disease ,Geriatrics and Gerontology ,Pharmacology ,business - Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.