Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
PLoS One ; 15(3): e0229672, 2020.
Article in English | MEDLINE | ID: mdl-32214362

ABSTRACT

More than 170 types of human papilloma viruses (HPV) exist with many causing proliferative diseases linked to malignancy in indications such as cervical cancer and head and neck squamous cell carcinoma. Characterization of antibody levels toward HPV serology is challenging due to complex biology of oncoproteins, pre-existing titers to multiple HPV types, cross-reactivity, and low affinity, polyclonal responses. Using multiplex technology from MSD, we have developed an assay that simultaneously characterizes antibodies against E6 and E7 oncoproteins of HPV16 and 18, the primary drivers of HPV-associated oncogenesis. We fusion tagged our E6 and E7 proteins with MBP via two-step purification, spot-printed an optimized concentration of protein into wells of MSD 96-well plates, and assayed various cynomolgus monkey, human and HPV+ cervical cancer patient serum to validate the assay. The dynamic range of the assay covered 4-orders of magnitude and antibodies were detected in serum at a dilution up to 100,000-fold. The assay was very precise (n = 5 assay runs) with median CV of human serum samples ~ 5.3% and inter-run variability of 11.4%. The multiplex serology method has strong cross-reactivity between E6 oncoproteins from human serum samples as HPV18 E6 antigens neutralized 5 of 6 serum samples as strongly as HPV16 E6. Moderate concordance (Spearman's Rank = 0.775) was found between antibody responses against HPV16 E7 in the multiplex assay compared to standard ELISA serology methods. These results demonstrate the development of a high-throughput, multi-plex assay that requires lower sample quantity input with greater dynamic range to detect type-specific anti-HPV concentrations to E6 and E7 oncoproteins of HPV16 and 18.


Subject(s)
Antibodies, Viral/blood , Human papillomavirus 16/immunology , Human papillomavirus 18/immunology , Immunoassay/methods , Immunoglobulin G/blood , Animals , Antibody Specificity , Cross Reactions , DNA-Binding Proteins/immunology , Electrochemical Techniques , Enzyme-Linked Immunosorbent Assay , Female , High-Throughput Screening Assays/methods , High-Throughput Screening Assays/statistics & numerical data , Humans , Immunoassay/statistics & numerical data , Limit of Detection , Luminescent Measurements/methods , Luminescent Measurements/statistics & numerical data , Macaca fascicularis , Oncogene Proteins, Viral/immunology , Papillomavirus E7 Proteins/immunology , Repressor Proteins/immunology , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/virology
2.
Mol Pharm ; 17(2): 507-516, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31841002

ABSTRACT

Targeted strategies to deliver and retain drugs to kidneys are needed to improve drug accumulation and efficacy in a myriad of kidney diseases. These drug delivery systems show potential for improving the therapeutic windows of drugs acting in the kidney. Biodistribution of antibody-based therapeutics in vivo is governed by several factors including binding affinity, size, and valency. Investigations of how the biophysical and biochemical properties of biologics enable them to overcome biological barriers and reach kidneys are therefore of interest. Although renal accumulation of antibody fragments in cancer diagnostics and treatment has been observed, reports on effective delivery of antibody fragments to the kidneys remain scarce. Previously, we demonstrated that targeting plasmalemma vesicle-associated protein (PV1), a caveolae-associated protein, can promote accumulation of antibodies in both the lungs and the kidneys. Here, by fine-tuning the binding affinity of an antibody toward PV1, we observe that the anti-PV1 antibody with reduced binding affinity lost the capability for kidney targeting while retaining the lung targeting activity, suggesting that binding affinity is a critical factor for kidney targeting of the anti-PV1 antibody. We next use the antibody fragment F(ab')2 targeting PV1 to assess the dual effects of rapid kidney filtration and PV1 targeting on kidney-selective targeting. Ex vivo fluorescence imaging results demonstrated that after rapidly accumulating in kidneys at 4 h, PV1-targeted F(ab')2 was continually retained in the kidney at 24 h, whereas the isotype control F(ab')2 underwent urinary elimination with significantly reduced signaling in the kidney. Confocal imaging studies confirmed the localization of PV1-targeted F(ab')2 in the kidney. In addition, the monovalent antibody fragment (Fab-C4) lost the capability for kidney homing, indicating that the binding avidity of anti-PV1 F(ab')2 is important for kidney targeting. Our findings suggest that PV1-targeted F(ab')2 might be useful as a drug carrier for renal targeting and highlight the importance of affinity optimization for tissue targeting antibodies.


Subject(s)
Antibodies, Monoclonal/immunology , Caveolae/metabolism , Drug Carriers/pharmacokinetics , Immunoglobulin Fab Fragments/immunology , Kidney/drug effects , Membrane Proteins/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Antibody Affinity , Drug Carriers/administration & dosage , Female , HEK293 Cells , Humans , Immunoglobulin Fab Fragments/administration & dosage , Kidney/metabolism , Lung/drug effects , Lung/metabolism , Mice , Mice, Inbred BALB C , Tissue Distribution
3.
PLoS Comput Biol ; 15(5): e1006980, 2019 05.
Article in English | MEDLINE | ID: mdl-31042706

ABSTRACT

Antibodies are an important class of therapeutics that have significant clinical impact for the treatment of severe diseases. Computational tools to support antibody drug discovery have been developing at an increasing rate over the last decade and typically rely upon a predetermined co-crystal structure of the antibody bound to the antigen for structural predictions. Here, we show an example of successful in silico affinity maturation of a hybridoma derived antibody, AB1, using just a homology model of the antibody fragment variable region and a protein-protein docking model of the AB1 antibody bound to the antigen, murine CCL20 (muCCL20). In silico affinity maturation, together with alanine scanning, has allowed us to fine-tune the protein-protein docking model to subsequently enable the identification of two single-point mutations that increase the affinity of AB1 for muCCL20. To our knowledge, this is one of the first examples of the use of homology modelling and protein docking for affinity maturation and represents an approach that can be widely deployed.


Subject(s)
Antibody Affinity/physiology , Computational Biology/methods , Amino Acid Sequence , Animals , Antibodies/chemistry , Chemokine CCL20 , Computer Simulation , Drug Design , Immunoglobulin Variable Region , Mice , Models, Molecular , Protein Binding , Protein Conformation
4.
Bioconjug Chem ; 30(4): 1232-1243, 2019 04 17.
Article in English | MEDLINE | ID: mdl-30912649

ABSTRACT

Despite some clinical success with antibody-drug conjugates (ADCs) in patients with solid tumors and hematological malignancies, improvements in ADC design are still desirable due to the narrow therapeutic window of these compounds. Tumor-targeting antibody fragments have distinct advantages over monoclonal antibodies, including more rapid tumor accumulation and enhanced penetration, but are subject to rapid clearance. Half-life extension technologies such as PEGylation and albumin-binding domains (ABDs) have been widely used to improve the pharmacokinetics of many different types of biologics. PEGylation improves pharmacokinetics by increasing hydrodynamic size to reduce renal clearance, whereas ABDs extend half-life via FcRn-mediated recycling. In this study, we used an anti-oncofetal antigen 5T4 diabody conjugated with a highly potent cytotoxic pyrrolobenzodiazepine (PBD) warhead to assess and compare the effects of PEGylation and albumin binding on the in vivo efficacy of antibody fragment drug conjugates. Conjugation of 2× PEG20K to a diabody improved half-life from 40 min to 33 h, and an ABD-diabody fusion protein exhibited a half-life of 45 h in mice. In a xenograft model of breast cancer MDA-MB-436, the ABD-diabody-PBD showed greater tumor growth suppression and better tolerability than either PEG-diabody-PBD or diabody-PBD. These results suggest that the mechanism of half-life extension is an important consideration for designing cytotoxic antitumor agents.


Subject(s)
Antineoplastic Agents/therapeutic use , Immunoconjugates/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Binding, Competitive , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Enzyme-Linked Immunosorbent Assay , Female , Half-Life , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Mice , Mice, Nude , Polyethylene Glycols/chemistry , Xenograft Model Antitumor Assays
5.
Commun Biol ; 2: 92, 2019.
Article in English | MEDLINE | ID: mdl-30854484

ABSTRACT

Systemic administration of bio-therapeutics can result in only a fraction of drug reaching targeted tissues, with the majority of drug being distributed to tissues irrelevant to the drug's site of action. Targeted delivery to specific organs may allow for greater accumulation, better efficacy, and improved safety. We investigated how targeting plasmalemma vesicle-associated protein (PV1), a protein found in the endothelial caveolae of lungs and kidneys, can promote accumulation in these organs. Using ex vivo fluorescence imaging, we show that intravenously administered αPV1 antibodies localize to mouse lungs and kidneys. In a bleomycin-induced idiopathic pulmonary fibrosis (IPF) mouse model, αPV1 conjugated to Prostaglandin E2 (PGE2), a known anti-fibrotic agent, significantly reduced collagen content and fibrosis whereas a non-targeted PGE2 antibody conjugate failed to slow fibrosis progression. Our results demonstrate that PV1 targeting can be utilized to deliver therapeutics to lungs and this approach is potentially applicable for various lung diseases.


Subject(s)
Drug Carriers , Drug Delivery Systems , Idiopathic Pulmonary Fibrosis/drug therapy , Membrane Proteins/metabolism , Animals , Biomarkers , Bleomycin/adverse effects , Dinoprostone/metabolism , Disease Models, Animal , Gene Expression , Humans , Idiopathic Pulmonary Fibrosis/etiology , Idiopathic Pulmonary Fibrosis/pathology , Immunohistochemistry , Kidney/metabolism , Kidney/pathology , Lung/drug effects , Lung/metabolism , Lung/pathology , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice
6.
J Control Release ; 279: 126-135, 2018 06 10.
Article in English | MEDLINE | ID: mdl-29653224

ABSTRACT

The accumulation, dissemination and clearance of monoclonal antibody-based therapeutics or imaging reagents targeting tumor associated antigens is governed by several factors including affinity, size, charge, and valency. Tumor targeting antibody fragments have distinct advantages over intact monoclonal antibodies such as enhanced penetration within the tumor and rapid accumulation but are subject to rapid clearance. Polyethylene glycol (PEG)-modified antibody fragments can provide a way to balance tumor penetration and accumulation with improved serum persistence. In this study, we use a diabody, the dimeric antibody fragment, targeting the 5T4 antigen to assess the impact of PEGs of distinct size and shape on tumor accumulation and pharmacokinetics (PK). We show that PEG-modified diabodies improved the PK of the parental diabody from a half-life of 40 min to over 40 h for the higher molecular weight PEG conjugated diabodies. This improvement correlates with the increasing hydrodynamic size of pegylated diabodies, and can serve as a better predictor of the PK behavior of pegylated molecules than molecular weight alone. Tumor uptake profiles determined by quantitative PET imaging differed significantly based on PEG size and shape with diabody-PEG5K showing peak accumulation early on, but with the larger diabody-PEG20K showing better sustained tumor uptake at later time points. In addition, we demonstrate that a diabody-PEG20K-B with a hydrodynamic radius (Rh) of 6 nm had superior tumor uptake than the larger diabody-PEG40K-B with Rh of 12 nm, indicating that beyond 6 nm, larger pegylated diabodies have a slower tumor uptake rate while having comparable clearance kinetics. Our data demonstrate that pegylated diabodies with Rh of ~6 nm have an optimal size and PK profile for tumor uptake. Understanding the impact of pegylation on PK and tumor uptake could facilitate the development of pegylated diabodies as therapeutics.


Subject(s)
Drug Delivery Systems , Immunoglobulin Fragments/administration & dosage , Neoplasms/metabolism , Polyethylene Glycols/chemistry , Animals , Biological Transport , Cell Line, Tumor , Female , Half-Life , Humans , Hydrodynamics , Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/metabolism , Mice , Mice, Nude , Molecular Weight , Positron-Emission Tomography , Tissue Distribution
7.
MAbs ; 8(6): 1118-25, 2016.
Article in English | MEDLINE | ID: mdl-27210548

ABSTRACT

Fusion of proteins to the Fc region of IgG is widely used to express cellular receptors and other extracellular proteins, but cleavage of the fusion partner is sometimes required for downstream applications. Immunoglobulin G-degrading enzyme of Streptococcus pyogenes (IdeS) is a protease with exquisite specificity for human IgG, and it can also cleave Fc-fusion proteins at a single site in the N-terminal region of the CH2 domain. However, the site of IdeS cleavage results in the disulfide-linked hinge region partitioning with the released protein, complicating downstream usage of the cleaved product. To tailor the Fc fragment for release of partner proteins by IdeS treatment, we investigated the effect of deleting regions of IgG-derived sequence that are upstream of the cleavage site. Elimination of the IgG-derived hinge sequence along with several residues of the CH2 domain had negligible effects on expression and purity of the fusion protein, while retaining efficient processing by IdeS. An optimal Fc fragment comprising residues 235-447 of the human IgG1 heavy chain sufficed for efficient production of fusion proteins and minimized the amount of residual Ig-derived sequence on the cleavage product following IdeS treatment. Pairing of this truncated Fc fragment with IdeS cleavage enables highly specific cleavage of Fc-fusion proteins, thus eliminating the need to engineer extraneous cleavage sequences. This system should be helpful for producing Fc-fusion proteins requiring downstream cleavage, particularly those that are sensitive to internal miscleavage if treated with alternative proteases.


Subject(s)
Bacterial Proteins/chemistry , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin G/chemistry , Proteolysis , Recombinant Fusion Proteins/chemistry , Chromatography, Gel , Chromatography, Liquid , Hinge Exons , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin G/genetics , Mass Spectrometry , Protein Domains , Recombinant Fusion Proteins/genetics , Substrate Specificity
8.
J Biomol Screen ; 21(6): 608-19, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26969322

ABSTRACT

The primary objective of early drug discovery is to associate druggable target space with a desired phenotype. The inability to efficiently associate these often leads to failure early in the drug discovery process. In this proof-of-concept study, the most tractable starting points for drug discovery within the NF-κB pathway model system were identified by integrating affinity selection-mass spectrometry (AS-MS) with functional cellular assays. The AS-MS platform Automated Ligand Identification System (ALIS) was used to rapidly screen 15 NF-κB proteins in parallel against large-compound libraries. ALIS identified 382 target-selective compounds binding to 14 of the 15 proteins. Without any chemical optimization, 22 of the 382 target-selective compounds exhibited a cellular phenotype consistent with the respective target associated in ALIS. Further studies on structurally related compounds distinguished two chemical series that exhibited a preliminary structure-activity relationship and confirmed target-driven cellular activity to NF-κB1/p105 and TRAF5, respectively. These two series represent new drug discovery opportunities for chemical optimization. The results described herein demonstrate the power of combining ALIS with cell functional assays in a high-throughput, target-based approach to determine the most tractable drug discovery opportunities within a pathway.


Subject(s)
Drug Discovery , High-Throughput Screening Assays/methods , NF-kappa B/antagonists & inhibitors , Structure-Activity Relationship , Ligands , Mass Spectrometry/methods , NF-kappa B/chemistry , Protein Binding , Signal Transduction/drug effects , TNF Receptor-Associated Factor 5/antagonists & inhibitors , TNF Receptor-Associated Factor 5/chemistry , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelA/chemistry
9.
MAbs ; 8(3): 501-12, 2016.
Article in English | MEDLINE | ID: mdl-26852694

ABSTRACT

The enormous diversity created by gene recombination and somatic hypermutation makes de novo protein sequencing of monoclonal antibodies a uniquely challenging problem. Modern mass spectrometry-based sequencing will rarely, if ever, provide a single unambiguous sequence for the variable domains. A more likely outcome is computation of an ensemble of highly similar sequences that can satisfy the experimental data. This outcome can result in the need for empirical testing of many candidate sequences, sometimes iteratively, to identity one which can replicate the activity of the parental antibody. Here we describe an improved approach to antibody protein sequencing by using phage display technology to generate a combinatorial library of sequences that satisfy the mass spectrometry data, and selecting for functional candidates that bind antigen. This approach was used to reverse engineer 2 commercially-obtained monoclonal antibodies against murine CD137. Proteomic data enabled us to assign the majority of the variable domain sequences, with the exception of 3-5% of the sequence located within or adjacent to complementarity-determining regions. To efficiently resolve the sequence in these regions, small phage-displayed libraries were generated and subjected to antigen binding selection. Following enrichment of antigen-binding clones, 2 clones were selected for each antibody and recombinantly expressed as antigen-binding fragments (Fabs). In both cases, the reverse-engineered Fabs exhibited identical antigen binding affinity, within error, as Fabs produced from the commercial IgGs. This combination of proteomic and protein engineering techniques provides a useful approach to simplifying the technically challenging process of reverse engineering monoclonal antibodies from protein material.


Subject(s)
Peptide Library , Protein Engineering/methods , Sequence Analysis, Protein , Single-Chain Antibodies , Animals , Mice , Rats , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors , Tumor Necrosis Factor Receptor Superfamily, Member 9/chemistry
10.
J Biol Chem ; 290(33): 20360-73, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26134571

ABSTRACT

G-protein-coupled receptor (GPCR) kinases (GRKs) bind to and phosphorylate GPCRs, initiating the process of GPCR desensitization and internalization. GRK4 is implicated in the regulation of blood pressure, and three GRK4 polymorphisms (R65L, A142V, and A486V) are associated with hypertension. Here, we describe the 2.6 Å structure of human GRK4α A486V crystallized in the presence of 5'-adenylyl ß,γ-imidodiphosphate. The structure of GRK4α is similar to other GRKs, although slight differences exist within the RGS homology (RH) bundle subdomain, substrate-binding site, and kinase C-tail. The RH bundle subdomain and kinase C-terminal lobe form a strikingly acidic surface, whereas the kinase N-terminal lobe and RH terminal subdomain surfaces are much more basic. In this respect, GRK4α is more similar to GRK2 than GRK6. A fully ordered kinase C-tail reveals interactions linking the C-tail with important determinants of kinase activity, including the αB helix, αD helix, and the P-loop. Autophosphorylation of wild-type GRK4α is required for full kinase activity, as indicated by a lag in phosphorylation of a peptide from the dopamine D1 receptor without ATP preincubation. In contrast, this lag is not observed in GRK4α A486V. Phosphopeptide mapping by mass spectrometry indicates an increased rate of autophosphorylation of a number of residues in GRK4α A486V relative to wild-type GRK4α, including Ser-485 in the kinase C-tail.


Subject(s)
G-Protein-Coupled Receptor Kinase 4/chemistry , G-Protein-Coupled Receptor Kinase 4/metabolism , Hypertension/genetics , Amino Acid Sequence , Crystallography, X-Ray , G-Protein-Coupled Receptor Kinase 4/genetics , Humans , Models, Molecular , Molecular Sequence Data , Phosphorylation , Protein Conformation , Sequence Homology, Amino Acid , Substrate Specificity
11.
Bioorg Med Chem Lett ; 25(9): 1831-5, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25851938

ABSTRACT

Novel bacterial topoisomerase inhibitors (NBTIs) represent a new class of broad-spectrum antibacterial agents targeting bacterial Gyrase A and ParC and have potential utility in combating antibiotic resistance. A series of novel oxabicyclooctane-linked NBTIs with new tricyclic-1,5-naphthyridinone left hand side moieties have been described. Compounds with a (R)-hydroxy-1,5-naphthyridinone moiety (7) showed potent antibacterial activity (e.g., Staphylococcus aureus MIC 0.25 µg/mL), acceptable Gram-positive and Gram-negative spectrum with rapidly bactericidal activity. The compound 7 showed intravenous and oral efficacy (ED50) at 3.2 and 27 mg/kg doses, respectively, in a murine model of bacteremia. Most importantly they showed significant attenuation of functional hERG activity (IC50 >170 µM). In general, lower logD attenuated hERG activity but also reduced Gram-negative activity. The co-crystal structure of a hydroxy-tricyclic NBTI bound to a DNA-gyrase complex exhibited a binding mode that show enantiomeric preference for R isomer and explains the activity and SAR. The discovery, synthesis, SAR and X-ray crystal structure of the left-hand-side tricyclic 1,5-naphthyridinone based oxabicyclooctane linked NBTIs are described.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cyclooctanes/pharmacology , DNA Topoisomerases, Type II/metabolism , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Naphthyridines/pharmacology , Topoisomerase II Inhibitors/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Cyclooctanes/chemical synthesis , Cyclooctanes/chemistry , Dose-Response Relationship, Drug , Gram-Negative Bacteria/enzymology , Gram-Positive Bacteria/enzymology , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Naphthyridines/chemical synthesis , Naphthyridines/chemistry , Structure-Activity Relationship , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/chemistry
12.
ACS Med Chem Lett ; 5(5): 609-14, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24900889

ABSTRACT

Bacterial resistance is eroding the clinical utility of existing antibiotics necessitating the discovery of new agents. Bacterial type II topoisomerase is a clinically validated, highly effective, and proven drug target. This target is amenable to inhibition by diverse classes of inhibitors with alternative and distinct binding sites to quinolone antibiotics, thus enabling the development of agents that lack cross-resistance to quinolones. Described here are novel bacterial topoisomerase inhibitors (NBTIs), which are a new class of gyrase and topo IV inhibitors and consist of three distinct structural moieties. The substitution of the linker moiety led to discovery of potent broad-spectrum NBTIs with reduced off-target activity (hERG IC50 > 18 µM) and improved physical properties. AM8191 is bactericidal and selectively inhibits DNA synthesis and Staphylococcus aureus gyrase (IC50 = 1.02 µM) and topo IV (IC50 = 10.4 µM). AM8191 showed parenteral and oral efficacy (ED50) at less than 2.5 mg/kg doses in a S. aureus murine infection model. A cocrystal structure of AM8191 bound to S. aureus DNA-gyrase showed binding interactions similar to that reported for GSK299423, displaying a key contact of Asp83 with the basic amine at position-7 of the linker.

13.
J Biol Chem ; 288(47): 34073-34080, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24108127

ABSTRACT

The emergence of antibiotic-resistant strains of pathogenic bacteria is an increasing threat to global health that underscores an urgent need for an expanded antibacterial armamentarium. Gram-negative bacteria, such as Escherichia coli, have become increasingly important clinical pathogens with limited treatment options. This is due in part to their lipopolysaccharide (LPS) outer membrane components, which dually serve as endotoxins while also protecting Gram-negative bacteria from antibiotic entry. The LpxC enzyme catalyzes the committed step of LPS biosynthesis, making LpxC a promising target for new antibacterials. Here, we present the first structure of an LpxC enzyme in complex with the deacetylation reaction product, UDP-(3-O-(R-3-hydroxymyristoyl))-glucosamine. These studies provide valuable insight into recognition of substrates and products by LpxC and a platform for structure-guided drug discovery of broad spectrum Gram-negative antibiotics.


Subject(s)
Amidohydrolases/chemistry , Escherichia coli/enzymology , Myristic Acids/chemistry , Protons , Uridine Diphosphate N-Acetylglucosamine/analogs & derivatives , Amidohydrolases/metabolism , Crystallography, X-Ray , Lipopolysaccharides/biosynthesis , Lipopolysaccharides/chemistry , Myristic Acids/metabolism , Protein Structure, Tertiary , Uridine Diphosphate N-Acetylglucosamine/chemistry , Uridine Diphosphate N-Acetylglucosamine/metabolism
14.
J Med Chem ; 56(6): 2294-310, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23379595

ABSTRACT

This report documents the first example of a specific inhibitor of protein kinases with preferential binding to the activated kinase conformation: 5H-benzo[4,5]cyclohepta[1,2-b]pyridin-5-one 11r (MK-8033), a dual c-Met/Ron inhibitor under investigation as a treatment for cancer. The design of 11r was based on the desire to reduce time-dependent inhibition of CYP3A4 (TDI) by members of this structural class. A novel two-step protocol for the synthesis of benzylic sulfonamides was developed to access 11r and analogues. We provide a rationale for the observed selectivity based on X-ray crystallographic evidence and discuss selectivity trends with additional examples. Importantly, 11r provides full inhibition of tumor growth in a c-Met amplified (GTL-16) subcutaneous tumor xenograft model and may have an advantage over inactive form kinase inhibitors due to equal potency against a panel of oncogenic activating mutations of c-Met in contrast to c-Met inhibitors without preferential binding to the active kinase conformation.


Subject(s)
Benzocycloheptenes/metabolism , Benzocycloheptenes/pharmacology , Drug Discovery , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Sulfonamides/metabolism , Sulfonamides/pharmacology , Animals , Benzocycloheptenes/chemistry , Cell Line, Tumor , Dogs , Enzyme Activation/drug effects , Female , Humans , Mice , Models, Molecular , Protein Conformation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/chemistry , Rats , Substrate Specificity , Sulfonamides/chemistry , Xenograft Model Antitumor Assays
15.
J Med Chem ; 54(12): 4092-108, 2011 Jun 23.
Article in English | MEDLINE | ID: mdl-21608528

ABSTRACT

c-Met is a transmembrane tyrosine kinase that mediates activation of several signaling pathways implicated in aggressive cancer phenotypes. In recent years, research into this area has highlighted c-Met as an attractive cancer drug target, triggering a number of approaches to disrupt aberrant c-Met signaling. Screening efforts identified a unique class of 5H-benzo[4,5]cyclohepta[1,2-b]pyridin-5-one kinase inhibitors, exemplified by 1. Subsequent SAR studies led to the development of 81 (MK-2461), a potent inhibitor of c-Met that was efficacious in preclinical animal models of tumor suppression. In addition, biochemical studies and X-ray analysis have revealed that this unique class of kinase inhibitors binds preferentially to the activated (phosphorylated) form of the kinase. This report details the development of 81 and provides a description of its unique biochemical properties.


Subject(s)
Antineoplastic Agents/chemical synthesis , Benzocycloheptenes/chemical synthesis , Pyridines/chemical synthesis , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Benzocycloheptenes/pharmacokinetics , Benzocycloheptenes/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Dogs , Drug Screening Assays, Antitumor , Female , Haplorhini , Humans , Mice , Mice, Nude , Models, Molecular , Mutation , Neoplasm Transplantation , Phosphorylation , Protein Binding , Pyrazoles/chemical synthesis , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Pyridines/pharmacokinetics , Pyridines/pharmacology , Rats , Receptor Protein-Tyrosine Kinases/genetics , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology , Transplantation, Heterologous
16.
J Biol Chem ; 286(13): 11218-25, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21247903

ABSTRACT

The receptor tyrosine kinase c-Met is implicated in oncogenesis and is the target for several small molecule and biologic agents in clinical trials for the treatment of cancer. Binding of the hepatocyte growth factor to the cell surface receptor of c-Met induces activation via autophosphorylation of the kinase domain. Here we describe the structural basis of c-Met activation upon autophosphorylation and the selective small molecule inhibiton of autophosphorylated c-Met. MK-2461 is a potent c-Met inhibitor that is selective for the phosphorylated state of the enzyme. Compound 1 is an MK-2461 analog with a 20-fold enthalpy-driven preference for the autophosphorylated over unphosphorylated c-Met kinase domain. The crystal structure of the unbound kinase domain phosphorylated at Tyr-1234 and Tyr-1235 shows that activation loop phosphorylation leads to the ejection and disorder of the activation loop and rearrangement of helix αC and the G loop to generate a viable active site. Helix αC adopts a orientation different from that seen in activation loop mutants. The crystal structure of the complex formed by the autophosphorylated c-Met kinase domain and compound 1 reveals a significant induced fit conformational change of the G loop and ordering of the activation loop, explaining the selectivity of compound 1 for the autophosphorylated state. The results highlight the role of structural plasticity within the kinase domain in imparting the specificity of ligand binding and provide the framework for structure-guided design of activated c-Met inhibitors.


Subject(s)
Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/chemistry , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/chemistry , Animals , Cell Line , Crystallography, X-Ray , Drug Design , Humans , Phosphorylation , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Spodoptera , Structure-Activity Relationship , c-Mer Tyrosine Kinase
17.
BMC Struct Biol ; 10: 16, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20540760

ABSTRACT

BACKGROUND: The unique S28 family of proteases is comprised of the carboxypeptidase PRCP and the aminopeptidase DPP7. The structural basis of the different substrate specificities of the two enzymes is not understood nor has the structure of the S28 fold been described. RESULTS: The experimentally phased 2.8 A crystal structure is presented for human PRCP. PRCP contains an alpha/beta hydrolase domain harboring the catalytic Asp-His-Ser triad and a novel helical structural domain that caps the active site. Structural comparisons with prolylendopeptidase and DPP4 identify the S1 proline binding site of PRCP. A structure-based alignment with the previously undescribed structure of DPP7 illuminates the mechanism of orthogonal substrate specificity of PRCP and DPP7. PRCP has an extended active-site cleft that can accommodate proline substrates with multiple N-terminal residues. In contrast, the substrate binding groove of DPP7 is occluded by a short amino-acid insertion unique to DPP7 that creates a truncated active site selective for dipeptidyl proteolysis of N-terminal substrates. CONCLUSION: The results define the structure of the S28 family of proteases, provide the structural basis of PRCP and DPP7 substrate specificity and enable the rational design of selective PRCP modulators.


Subject(s)
Carboxypeptidases/chemistry , Amino Acid Sequence , Binding Sites , Carboxypeptidases/genetics , Carboxypeptidases/metabolism , Catalytic Domain , Crystallography, X-Ray , Humans , Molecular Sequence Data , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity
18.
Article in English | MEDLINE | ID: mdl-20516604

ABSTRACT

Prolylcarboxypeptidase (PrCP) is a lysosomal serine carboxypeptidase that cleaves a variety of C-terminal amino acids adjacent to proline and has been implicated in diseases such as hypertension and obesity. Here, the robust production, purification and crystallization of glycosylated human PrCP from stably transformed CHO cells is described. Purified PrCP yielded crystals belonging to space group R32, with unit-cell parameters a = b = 181.14, c = 240.13 A, that diffracted to better than 2.8 A resolution.


Subject(s)
Carboxypeptidases/chemistry , Animals , CHO Cells , Carboxypeptidases/genetics , Carboxypeptidases/isolation & purification , Cricetinae , Cricetulus , Crystallization , Crystallography, X-Ray , Gene Expression , Glycosylation , Humans
19.
J Biol Chem ; 283(50): 34864-72, 2008 Dec 12.
Article in English | MEDLINE | ID: mdl-18922802

ABSTRACT

Prostasin (also called channel activating protease-1 (CAP1)) is an extracellular serine protease implicated in the modulation of fluid and electrolyte regulation via proteolysis of the epithelial sodium channel. Several disease states, particularly hypertension, can be affected by modulation of epithelial sodium channel activity. Thus, understanding the biochemical function of prostasin and developing specific agents to inhibit its activity could have a significant impact on a widespread disease. We report the expression of the prostasin proenzyme in Escherichia coli as insoluble inclusion bodies, refolding and activating via proteolytic removal of the N-terminal propeptide. The refolded and activated enzyme was shown to be pure and monomeric, with kinetic characteristics very similar to prostasin expressed from eukaryotic systems. Active prostasin was crystallized, and the structure was determined to 1.45 A resolution. These apoprotein crystals were soaked with nafamostat, allowing the structure of the inhibited acyl-enzyme intermediate structure to be determined to 2.0 A resolution. Comparison of the inhibited and apoprotein forms of prostasin suggest a mechanism of regulation through stabilization of a loop which interferes with substrate recognition.


Subject(s)
Hypertension/metabolism , Serine Endopeptidases/chemistry , Amino Acid Sequence , Apoproteins/chemistry , Benzamidines , Crystallography, X-Ray/methods , Escherichia coli/metabolism , Guanidines/chemistry , Humans , Molecular Conformation , Molecular Sequence Data , Protein Conformation , Protein Folding , Protein Renaturation , Sequence Homology, Amino Acid , Serine Endopeptidases/genetics , Substrate Specificity
20.
Arch Biochem Biophys ; 469(2): 220-31, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-17999913

ABSTRACT

The kinesin spindle protein (KSP, also known as Eg5) is essential for the proper separation of spindle poles during mitosis, and inhibition results in mitotic arrest and the formation of characteristic monoaster spindles. Several distinct classes of KSP inhibitors have been described previously in the public and patent literature. However, most appear to share a common induced-fit allosteric binding site, suggesting a common mechanism of inhibition. In a high-throughput screen for inhibitors of KSP, a novel class of thiazole-containing inhibitors was identified. Unlike the previously described allosteric KSP inhibitors, the thiazoles described here show ATP competitive kinetic behavior, consistent with binding within the nucleotide binding pocket. Although they bind to a pocket that is highly conserved across kinesins, these molecules exhibit significant selectivity for KSP over other kinesins and other ATP-utilizing enzymes. Several of these compounds are active in cells and produce a phenotype similar to that observed with previously published allosteric inhibitors of KSP.


Subject(s)
Adenosine Triphosphate/metabolism , Biochemistry/methods , Kinesins/antagonists & inhibitors , Kinesins/chemistry , Mitosis , Adenosine Triphosphate/chemistry , Allosteric Site , Binding, Competitive , Drug Design , Humans , Kinesins/metabolism , Models, Biological , Models, Chemical , Nucleotides/chemistry , Phenotype , Protein Binding , Thiazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...