Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Glycobiology ; 33(5): 364-368, 2023 06 03.
Article in English | MEDLINE | ID: mdl-36881660

ABSTRACT

Monoclonal antibodies comprise a major class of biologic therapeutics and are also extensively studied in immunology. Given the importance of glycans on antibodies, fluorescent labeling of enzymatically released glycans and their LC/MS analysis is routinely used for in-depth characterization of antibody glycosylation. In this technical note, we propose a method for facile characterization of glycans in the variable region of antibodies using sequential enzymatic digests with Endoglycosidase-S2 and RapidTM Peptide-N-Glycosidase-F followed by labeling with a fluorescent dye carrying an NHS-carbamate moiety. The results and proposed mechanism also suggest that the choice of glycosidases along with the labeling chemistry is critical for accurate glycan analysis for a desired application.


Subject(s)
Polysaccharides , Polysaccharides/immunology , Immunoglobulin G/immunology , Glycosylation
2.
Bioorg Med Chem Lett ; 24(6): 1484-8, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24582986

ABSTRACT

Apoptosis is regulated by the BCL-2 family of proteins, which is comprised of both pro-death and pro-survival members. Evasion of apoptosis is a hallmark of malignant cells. One way in which cancer cells achieve this evasion is thru overexpression of the pro-survival members of the BCL-2 family. Overexpression of MCL-1, a pro-survival protein, has been shown to be a resistance factor for Navitoclax, a potent inhibitor of BCL-2 and BCL-XL. Here we describe the use of fragment screening methods and structural biology to drive the discovery of novel MCL-1 inhibitors from two distinct structural classes. Specifically, cores derived from a biphenyl sulfonamide and salicylic acid were uncovered in an NMR-based fragment screen and elaborated using high throughput analog synthesis. This culminated in the discovery of selective and potent inhibitors of MCL-1 that may serve as promising leads for medicinal chemistry optimization efforts.


Subject(s)
Drug Design , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Binding Sites , Biphenyl Compounds/chemistry , Crystallography, X-Ray , Magnetic Resonance Spectroscopy , Molecular Dynamics Simulation , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Protein Binding , Protein Structure, Tertiary , Salicylic Acid/chemistry , Salicylic Acid/metabolism , Sulfonamides/chemistry , Sulfonamides/metabolism
3.
J Med Chem ; 64(1): 417-429, 2021 01 14.
Article in English | MEDLINE | ID: mdl-33378180

ABSTRACT

Tumor necrosis factor α (TNFα) is a soluble cytokine that is directly involved in systemic inflammation through the regulation of the intracellular NF-κB and MAPK signaling pathways. The development of biologic drugs that inhibit TNFα has led to improved clinical outcomes for patients with rheumatoid arthritis and other chronic autoimmune diseases; however, TNFα has proven to be difficult to drug with small molecules. Herein, we present a two-phase, fragment-based drug discovery (FBDD) effort in which we first identified isoquinoline fragments that disrupt TNFα ligand-receptor binding through an allosteric desymmetrization mechanism as observed in high-resolution crystal structures. The second phase of discovery focused on the de novo design and optimization of fragments with improved binding efficiency and drug-like properties. The 3-indolinone-based lead presented here displays oral, in vivo efficacy in a mouse glucose-6-phosphate isomerase (GPI)-induced paw swelling model comparable to that seen with a TNFα antibody.


Subject(s)
Biological Products/chemical synthesis , Drug Design , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Administration, Oral , Allosteric Regulation , Animals , Arthritis, Rheumatoid/drug therapy , Autoimmune Diseases/drug therapy , Biological Products/pharmacology , Biological Products/therapeutic use , Ligands , Mice , Tumor Necrosis Factor-alpha/metabolism
4.
Cancer Immunol Res ; 7(11): 1864-1875, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31462409

ABSTRACT

Agonistic CD40 monoclonal antibodies (mAb) have demonstrated some clinical activity, but with dose-limiting toxicity. To reduce systemic toxicity, we developed a bispecific molecule that was maximally active in the presence of a tumor antigen and had limited activity in the absence of the tumor antigen. LB-1 is a bispecific molecule containing single-chain Fv domains targeting mouse CD40 and the tumor antigen mesothelin. LB-1 exhibited enhanced activity upon binding to cell-surface mesothelin but was less potent in the absence of mesothelin binding. In a mouse model implanted with syngeneic 4T1 tumors expressing cell-surface mesothelin, LB-1 demonstrated comparable antitumor activity as an agonistic CD40 mAb but did not cause elevation of serum cytokines and liver enzymes, as was observed in anti-CD40-treated mice. The results from our study of LB-1 were used to develop a human cross-reactive bispecific molecule (ABBV-428) that targeted human CD40 and mesothelin. ABBV-428 demonstrated enhanced activation of antigen-presenting cells and T cells upon binding to cell-surface mesothelin, and inhibition of cultured or implanted PC3 tumor cell growth after immune activation. Although expression of cell-surface mesothelin is necessary, the bispecific molecules induced immune-mediated antitumor activity against both mesothelin+ and mesothelin- tumor cells. ABBV-428 represents a class of bispecific molecules with conditional activity dependent on the binding of a tumor-specific antigen, and such activity could potentially maximize antitumor potency while limiting systemic toxicity in clinical studies.


Subject(s)
Antibodies, Bispecific/immunology , Antigens, Neoplasm/immunology , Antineoplastic Agents, Immunological/immunology , CD40 Antigens/immunology , GPI-Linked Proteins/immunology , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antigens, Neoplasm/metabolism , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , CD40 Antigens/agonists , Cell Line, Tumor , GPI-Linked Proteins/metabolism , Humans , Lymphocyte Activation/drug effects , Mesothelin , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
6.
ACS Chem Biol ; 6(3): 234-44, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21090814

ABSTRACT

Inhibition of protein kinases has validated therapeutic utility for cancer, with at least seven kinase inhibitor drugs on the market. Protein kinase inhibition also has significant potential for a variety of other diseases, including diabetes, pain, cognition, and chronic inflammatory and immunologic diseases. However, as the vast majority of current approaches to kinase inhibition target the highly conserved ATP-binding site, the use of kinase inhibitors in treating nononcology diseases may require great selectivity for the target kinase. As protein kinases are signal transducers that are involved in binding to a variety of other proteins, targeting alternative, less conserved sites on the protein may provide an avenue for greater selectivity. Here we report an affinity-based, high-throughput screening technique that allows nonbiased interrogation of small molecule libraries for binding to all exposed sites on a protein surface. This approach was used to screen both the c-Jun N-terminal protein kinase Jnk-1 (involved in insulin signaling) and p38α (involved in the formation of TNFα and other cytokines). In addition to canonical ATP-site ligands, compounds were identified that bind to novel allosteric sites. The nature, biological relevance, and mode of binding of these ligands were extensively characterized using two-dimensional (1)H/(13)C NMR spectroscopy, protein X-ray crystallography, surface plasmon resonance, and direct enzymatic activity and activation cascade assays. Jnk-1 and p38α both belong to the MAP kinase family, and the allosteric ligands for both targets bind similarly on a ledge of the protein surface exposed by the MAP insertion present in the CMGC family of protein kinases and distant from the active site. Medicinal chemistry studies resulted in an improved Jnk-1 ligand able to increase adiponectin secretion in human adipocytes and increase insulin-induced protein kinase PKB phosphorylation in human hepatocytes, in similar fashion to Jnk-1 siRNA and to rosiglitazone treatment. Together, the data suggest that these new ligand series bind to a novel, allosteric, and physiologically relevant site and therefore represent a unique approach to identify kinase inhibitors.


Subject(s)
Drug Discovery , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Binding Sites/drug effects , Crystallography, X-Ray , High-Throughput Screening Assays , Humans , Mitogen-Activated Protein Kinase 8/chemistry , Mitogen-Activated Protein Kinase 8/metabolism , Models, Molecular , Molecular Sequence Data , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Small Molecule Libraries , Stereoisomerism , Structure-Activity Relationship , p38 Mitogen-Activated Protein Kinases/chemistry , p38 Mitogen-Activated Protein Kinases/metabolism
7.
ACS Med Chem Lett ; 1(6): 295-9, 2010 Sep 09.
Article in English | MEDLINE | ID: mdl-24900211

ABSTRACT

NMR spectroscopy has enjoyed widespread success as a method for screening protein targets, especially in the area of fragment-based drug discovery. However, current methods for NMR-based screening all suffer certain limitations. Two-dimensional methods like "SAR by NMR" require isotopically labeled protein and are limited to proteins less than about 50 kDa. For one-dimensional, ligand-based methods, results can be confounded by nonspecific compound binding, resonance overlap, or the need for a special NMR probe. We present here a ligand-based method that relies on the exchange broadening observed for a (13)C-labeled molecule upon binding to a protein target (labeled ligand displacement). This method can be used to screen both individual compounds and mixtures and is free of the artifacts inherent in other ligand-based methods.

8.
Proc Natl Acad Sci U S A ; 104(19): 7875-80, 2007 May 08.
Article in English | MEDLINE | ID: mdl-17470806

ABSTRACT

The pituitary adenylate cyclase-activating polypeptide (PACAP) receptor is a class II G protein-coupled receptor that contributes to many different cellular functions including neurotransmission, neuronal survival, and synaptic plasticity. The solution structure of the potent antagonist PACAP (residues 6'-38') complexed to the N-terminal extracellular (EC) domain of the human splice variant hPAC1-R-short (hPAC1-R(S)) was determined by NMR. The PACAP peptide adopts a helical conformation when bound to hPAC1-R(S) with a bend at residue A18' and makes extensive hydrophobic and electrostatic interactions along the exposed beta-sheet and interconnecting loops of the N-terminal EC domain. Mutagenesis data on both the peptide and the receptor delineate the critical interactions between the C terminus of the peptide and the C terminus of the EC domain that define the high affinity and specificity of hormone binding to hPAC1-R(S). These results present a structural basis for hPAC1-R(S) selectivity for PACAP versus the vasoactive intestinal peptide and also differentiate PACAP residues involved in binding to the N-terminal extracellular domain versus other parts of the full-length hPAC1-R(S) receptor. The structural, mutational, and binding data are consistent with a model for peptide binding in which the C terminus of the peptide hormone interacts almost exclusively with the N-terminal EC domain, whereas the central region makes contacts to both the N-terminal and other extracellular parts of the receptor, ultimately positioning the N terminus of the peptide to contact the transmembrane region and result in receptor activation.


Subject(s)
Pituitary Adenylate Cyclase-Activating Polypeptide/chemistry , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/chemistry , Amino Acid Sequence , Animals , Humans , Mice , Molecular Sequence Data , Mutation , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Corticotropin-Releasing Hormone/chemistry , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Solutions
9.
Chem Res Toxicol ; 20(12): 1752-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18001056

ABSTRACT

We have recently reported on the development of a La assay to detect reactive molecules by nuclear magnetic resonance (ALARM NMR) to detect reactive false positive hits from high-throughput screening, in which we observed a surprisingly large number of compounds that can oxidize or form covalent adducts with protein thiols groups. In the vast majority of these cases, the covalent interactions are largely nonspecific (e.g., affect many protein targets) and therefore unsuitable for drug development. However, certain thiol-reactive species do appear to inhibit the target of interest in a specific manner. The question then arises as to the potential toxicology risks of developing a drug that can react with protein thiol groups. Here, we report on the evaluation of a large set of ALARM-reactive and -nonreactive compounds against a panel of additional proteins (aldehyde dehydrogenase, superoxide dismutase, and three cytochrome P450 enzymes). It was observed that ALARM-reactive compounds have significantly increased risks of interacting with one or more of these enzymes in vitro. Thus, ALARM NMR seems to be a sensitive tool to rapidly identify compounds with an enhanced risk of producing side effects in humans, including alcohol intolerance, the formation of reactive oxygen species, and drug-drug interactions. In conjunction with other toxicology assays, ALARM NMR should be a valuable tool for prioritizing compounds for lead optimization and animal testing.


Subject(s)
Aldehyde Dehydrogenase/chemistry , Autoantigens/chemistry , Cytochrome P-450 Enzyme Inhibitors , Pharmaceutical Preparations , Ribonucleoproteins/chemistry , Sulfhydryl Compounds/chemistry , Superoxide Dismutase/chemistry , Aldehyde Dehydrogenase/metabolism , Drug Design , Drug-Related Side Effects and Adverse Reactions , Humans , Magnetic Resonance Spectroscopy , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Molecular Structure , Pharmaceutical Preparations/analysis , Protein Binding , Sulfhydryl Compounds/metabolism , Superoxide Dismutase/metabolism , SS-B Antigen
10.
Protein Expr Purif ; 48(1): 56-60, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16442310

ABSTRACT

Silent information regulator 2 (Sir2) proteins are a class of protein deacetylase enzymes that play key roles in transcriptional gene silencing, DNA repair, and aging. Here, we describe the high-level bacterial expression and purification of a human SirT2 construct that yields high resolution NMR spectra. By removing the N-terminal helix alpha0 and using Thioredoxin as a fusion partner, greater than 10 mg/L of purified protein can be obtained from minimal media. The protein is fully functional and enables NMR-based screening and structural studies of this important protein.


Subject(s)
Escherichia coli/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification , Sirtuins/biosynthesis , Sirtuins/isolation & purification , Escherichia coli/metabolism , Humans , Nuclear Magnetic Resonance, Biomolecular , Recombinant Fusion Proteins/chemistry , Sirtuin 2 , Sirtuins/genetics , Thioredoxins/metabolism
12.
Biochemistry ; 44(48): 15834-41, 2005 Dec 06.
Article in English | MEDLINE | ID: mdl-16313186

ABSTRACT

Ion channels play critical roles in signaling processes and are attractive targets for treating various diseases. Here we describe an NMR-based strategy for structural analyses of potassium channel-ligand complexes using KcsA (residues 1-132, with six mutations to impart toxin binding and to mimic the eukaryotic hERG channel). Using this approach, we determined the solution structure of KcsA in complex with the high-affinity peptide antagonist charybdotoxin. The structural data reveal how charybdotoxin binds to the closed form of KcsA and makes specific contacts with the extracellular surface of the ion channel, resulting in pore blockage. This represents the first direct structural information about an ion channel complexed to a peptide antagonist and provides an experimental framework for understanding and interpreting earlier mutational analyses. The strategy presented here overcomes many of the limitations of conventional NMR approaches to helical membrane protein structure determination and can be applied in the study of the binding of druglike molecules to this important class of proteins.


Subject(s)
Bacterial Proteins/chemistry , Charybdotoxin/chemistry , Potassium Channels/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Crystallization , DNA Mutational Analysis , Electron Spin Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Peptides/chemistry , Potassium Channels/genetics , Scorpion Venoms/chemistry , Sequence Alignment , Shaker Superfamily of Potassium Channels
SELECTION OF CITATIONS
SEARCH DETAIL