Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
ACS Med Chem Lett ; 14(12): 1631-1639, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38116426

ABSTRACT

Redirecting E3 ligases to neo-substrates, leading to their proteasomal disassembly, known as targeted protein degradation (TPD), has emerged as a promising alternative to traditional, occupancy-driven pharmacology. Although the field has expanded tremendously over the past years, the choice of E3 ligases remains limited, with an almost exclusive focus on CRBN and VHL. Here, we report the discovery of novel ligands to the PRY-SPRY domain of TRIM58, a RING ligase that is specifically expressed in erythroid precursor cells. A DSF screen, followed by validation using additional biophysical methods, led to the identification of TRIM58 ligand TRIM-473. A basic SAR around the chemotype was established by utilizing a competitive binding assay employing a short FP peptide probe derived from an endogenous TRIM58 substrate. The X-ray co-crystal structure of TRIM58 in complex with TRIM-473 gave insights into the binding mode and potential exit vectors for bifunctional degrader design.

2.
J Med Chem ; 63(15): 8088-8113, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32551603

ABSTRACT

The serine protease factor XI (FXI) is a prominent drug target as it holds promise to deliver efficacious anticoagulation without an enhanced risk of major bleeds. Several efforts have been described targeting the active form of the enzyme, FXIa. Herein, we disclose our efforts to identify potent, selective, and orally bioavailable inhibitors of FXIa. Compound 1, identified from a diverse library of internal serine protease inhibitors, was originally designed as a complement factor D inhibitor and exhibited submicromolar FXIa activity and an encouraging absorption, distribution, metabolism, and excretion (ADME) profile while being devoid of a peptidomimetic architecture. Optimization of interactions in the S1, S1ß, and S1' pockets of FXIa through a combination of structure-based drug design and traditional medicinal chemistry led to the discovery of compound 23 with subnanomolar potency on FXIa, enhanced selectivity over other coagulation proteases, and a preclinical pharmacokinetics (PK) profile consistent with bid dosing in patients.


Subject(s)
Factor XIa/antagonists & inhibitors , Factor XIa/genetics , Factor Xa Inhibitors/administration & dosage , Factor Xa Inhibitors/chemistry , Administration, Oral , Amino Acid Sequence , Animals , Biological Availability , Dogs , Drug Evaluation, Preclinical/methods , Humans , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
3.
Mol Cell ; 75(3): 483-497.e9, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31253574

ABSTRACT

In mammals, ∼100 deubiquitinases act on ∼20,000 intracellular ubiquitination sites. Deubiquitinases are commonly regarded as constitutively active, with limited regulatory and targeting capacity. The BRCA1-A and BRISC complexes serve in DNA double-strand break repair and immune signaling and contain the lysine-63 linkage-specific BRCC36 subunit that is functionalized by scaffold subunits ABRAXAS and ABRO1, respectively. The molecular basis underlying BRCA1-A and BRISC function is currently unknown. Here we show that in the BRCA1-A complex structure, ABRAXAS integrates the DNA repair protein RAP80 and provides a high-affinity binding site that sequesters the tumor suppressor BRCA1 away from the break site. In the BRISC structure, ABRO1 binds SHMT2α, a metabolic enzyme enabling cancer growth in hypoxic environments, which we find prevents BRCC36 from binding and cleaving ubiquitin chains. Our work explains modularity in the BRCC36 DUB family, with different adaptor subunits conferring diversified targeting and regulatory functions.


Subject(s)
BRCA1 Protein/genetics , DNA Repair/genetics , DNA-Binding Proteins/genetics , Deubiquitinating Enzymes/genetics , Histone Chaperones/genetics , Neoplasms/genetics , Binding Sites/genetics , Carrier Proteins/genetics , Cell Nucleus/genetics , Cell Nucleus/immunology , Cytoplasm/genetics , Cytoplasm/immunology , DNA Breaks, Double-Stranded , DNA Repair/immunology , Deubiquitinating Enzymes/immunology , HeLa Cells , Humans , Immunity, Cellular/genetics , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Neoplasms/immunology , Nuclear Matrix-Associated Proteins/genetics , Protein Binding/genetics , Ubiquitin/genetics , Ubiquitin-Specific Proteases/genetics , Ubiquitination/genetics
4.
Blood ; 133(13): 1507-1516, 2019 03 28.
Article in English | MEDLINE | ID: mdl-30692123

ABSTRACT

A large unmet medical need exists for safer antithrombotic drugs because all currently approved anticoagulant agents interfere with hemostasis, leading to an increased risk of bleeding. Genetic and pharmacologic evidence in humans and animals suggests that reducing factor XI (FXI) levels has the potential to effectively prevent and treat thrombosis with a minimal risk of bleeding. We generated a fully human antibody (MAA868) that binds the catalytic domain of both FXI (zymogen) and activated FXI. Our structural studies show that MAA868 traps FXI and activated FXI in an inactive, zymogen-like conformation, explaining its equally high binding affinity for both forms of the enzyme. This binding mode allows the enzyme to be neutralized before entering the coagulation process, revealing a particularly attractive anticoagulant profile of the antibody. MAA868 exhibited favorable anticoagulant activity in mice with a dose-dependent protection from carotid occlusion in a ferric chloride-induced thrombosis model. MAA868 also caused robust and sustained anticoagulant activity in cynomolgus monkeys as assessed by activated partial thromboplastin time without any evidence of bleeding. Based on these preclinical findings, we conducted a first-in-human study in healthy subjects and showed that single subcutaneous doses of MAA868 were safe and well tolerated. MAA868 resulted in dose- and time-dependent robust and sustained prolongation of activated partial thromboplastin time and FXI suppression for up to 4 weeks or longer, supporting further clinical investigation as a potential once-monthly subcutaneous anticoagulant therapy.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Anticoagulants/therapeutic use , Blood Coagulation/drug effects , Factor XI/antagonists & inhibitors , Thrombosis/drug therapy , Adolescent , Adult , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Anticoagulants/pharmacology , Female , Humans , Immunoglobulin G/pharmacology , Immunoglobulin G/therapeutic use , Macaca fascicularis , Male , Mice, Inbred C57BL , Middle Aged , Molecular Docking Simulation , Thrombosis/blood , Young Adult
5.
J Nucl Med ; 60(3): 393-399, 2019 03.
Article in English | MEDLINE | ID: mdl-30002107

ABSTRACT

Patients with metastatic medullary thyroid cancer (MTC) have limited systemic treatment options. The use of radiolabeled gastrin analogs targeting the cholecystokinin-2 receptor (CCK2R) is an attractive approach. However, their therapeutic efficacy is presumably decreased by their enzymatic degradation in vivo. We aimed to investigate whether the chemically stabilized analog 177Lu-DOTA-PP-F11N (177Lu-DOTA-(dGlu)6-Ala-Tyr-Gly-Trp-Nle-Asp-Phe-NH2) performs better than reference analogs with varying in vivo stability, namely 177Lu-DOTA-MG11 (177Lu-DOTA-dGlu-Ala-Tyr-Gly-Trp-Met-Asp-Phe-NH2) and 177Lu-DOTA-PP-F11 (177Lu-DOTA-(dGlu)6-Ala-Tyr-Gly-Trp-Met-Asp-Phe-NH2), and whether the use of protease inhibitors further improves CCKR2 targeting. First human data on 177Lu-DOTA-PP-F11N are also reported. Methods: In vitro stability of all analogs was assessed against a panel of extra- and intracellular endoproteases, whereas their in vitro evaluation was performed using the human MTC MZ-CRC-1 and the transfected A431-CCK2R(+) cell lines. Biodistribution without and with the protease inhibitors phosphoramidon and thiorphan was assessed 4 h after injection in MZ-CRC-1 and A431-CCK2R(+) dual xenografts. Autoradiography of 177Lu-DOTA-PP-F11N (without and with phosphoramidon) and NanoSPECT/CT were performed. SPECT/CT images of 177Lu-DOTA-PP-F11N in a metastatic MTC patient were also acquired. Results:natLu-DOTA-PP-F11N is less of a substrate for neprilysins than the other analogs, whereas intracellular cysteine proteases, such as cathepsin-L, might be involved in the degradation of gastrin analogs. The uptake of all radiotracers was higher in MZ-CRC-1 tumors than in A431-CCK2R(+), apparently because of the higher number of binding sites on MZ-CRC-1 cells. 177Lu-DOTA-PP-F11N had the same biodistribution as 177Lu-DOTA-PP-F11; however, uptake in the MZ-CRC-1 tumors was almost double (20.7 ± 1.71 vs. 11.2 ± 2.94 %IA [percentage injected activity]/g, P = 0.0002). Coadministration of phosphoramidon or thiorphan increases 177Lu-DOTA-MG11 uptake significantly in the CCK2R(+) tumors and stomach. Less profound was the effect on 177Lu-DOTA-PP-F11, whereas no influence or even reduction was observed for 177Lu-DOTA-PP-F11N (20.7 ± 1.71 vs. 15.6 ± 3.80 [with phosphoramidon] %IA/g, P < 0.05 in MZ-CRC-1 tumors). The first clinical data show high 177Lu-DOTA-PP-F11N accumulation in tumors, stomach, kidneys, and colon. Conclusion: The performance of 177Lu-DOTA-PP-F11N without protease inhibitors is as good as the performance of 177Lu-DOTA-MG11 in the presence of inhibitors. The human application of single compounds without unessential additives is preferable. Preliminary clinical data spotlight the stomach as a potential dose-limiting organ besides the kidneys.


Subject(s)
Gastrins/chemistry , Gastrins/metabolism , Heterocyclic Compounds, 1-Ring/chemistry , Lutetium , Protease Inhibitors/pharmacology , Radioisotopes , Receptor, Cholecystokinin B/metabolism , Amino Acid Sequence , Animals , Biological Transport/drug effects , Cell Line, Tumor , Drug Stability , Female , Gastrins/pharmacokinetics , Humans , Mice , Positron Emission Tomography Computed Tomography , Single Photon Emission Computed Tomography Computed Tomography , Tissue Distribution/drug effects
6.
Article in English | MEDLINE | ID: mdl-28113517

ABSTRACT

Interleukin-8 (IL-8, CXCL8) is a neutrophil chemotactic factor belonging to the family of chemokines. IL-8 was shown to resist pepsin cleavage displaying its high resistance to this protease. However, the molecular mechanisms underlying this resistance are not fully understood. Using our in-house database containing the data on three-dimensional arrangements of secondary structure elements from the whole Protein Data Bank, we found a striking structural similarity between IL-8 and pepsin inhibitor-3. Such similarity could play a key role in understanding IL-8 resistance to the protease pepsin. To support this hypothesis, we applied pepsin assays confirming that intact IL-8 is not degraded by pepsin in comparison to IL-8 in a denaturated state. Applying 1H-15N Heteronuclear Single Quantum Coherence NMR measurements, we determined the putative regions at IL-8 that are potentially responsible for interactions with the pepsin. The results obtained in this work contribute to the understanding of the resistance of IL-8 to pepsin proteolysis in terms of its structural properties.


Subject(s)
Computational Biology/methods , Interleukin-8/chemistry , Interleukin-8/metabolism , Pepsin A/chemistry , Pepsin A/metabolism , Computer Simulation , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Structure, Secondary
7.
Methods Appl Fluoresc ; 5(3): 034002, 2017 09 22.
Article in English | MEDLINE | ID: mdl-28699919

ABSTRACT

Time-resolved fluorescence detection for robust sensing of biomolecular interactions is developed by implementing time-correlated single photon counting in high-throughput conditions. Droplet microfluidics is used as a promising platform for the very fast handling of low-volume samples. We illustrate the potential of this very sensitive and cost-effective technology in the context of an enzymatic activity assay based on fluorescently-labeled biomolecules. Fluorescence lifetime detection by time-correlated single photon counting is shown to enable reliable discrimination between positive and negative control samples at a throughput as high as several hundred samples per second.


Subject(s)
Biological Assay , Fluorescence , Microfluidic Analytical Techniques , Enzyme Assays , Fluorescent Dyes/chemistry , Peptides/chemistry , Trypsin/chemistry
8.
Angew Chem Int Ed Engl ; 56(5): 1294-1297, 2017 01 24.
Article in English | MEDLINE | ID: mdl-27981705

ABSTRACT

CSN5 is the zinc metalloprotease subunit of the COP9 signalosome (CSN), which is an important regulator of cullin-RING E3 ubiquitin ligases (CRLs). CSN5 is responsible for the cleavage of NEDD8 from CRLs, and blocking deconjugation of NEDD8 traps the CRLs in a hyperactive state, thereby leading to auto-ubiquitination and ultimately degradation of the substrate recognition subunits. Herein, we describe the discovery of azaindoles as a new class of CSN5 inhibitors, which interact with the active-site zinc ion of CSN5 through an unprecedented binding mode. The best compounds inhibited CSN5 with nanomolar potency, led to degradation of the substrate recognition subunit Skp2 in cells, and reduced the viability of HCT116 cells.


Subject(s)
COP9 Signalosome Complex/antagonists & inhibitors , Indoles/metabolism , Zinc/metabolism , Binding Sites , COP9 Signalosome Complex/genetics , COP9 Signalosome Complex/metabolism , Catalytic Domain , Cell Proliferation/drug effects , Crystallography, X-Ray , Fluorescence Resonance Energy Transfer , HCT116 Cells , Humans , Indoles/chemistry , Indoles/pharmacology , Molecular Docking Simulation , NEDD8 Protein/chemistry , NEDD8 Protein/metabolism , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , Protein Subunits/metabolism , RNA Interference , RNA, Small Interfering/metabolism , S-Phase Kinase-Associated Proteins/chemistry , S-Phase Kinase-Associated Proteins/metabolism , Zinc/chemistry
9.
Nat Commun ; 7: 13166, 2016 10 24.
Article in English | MEDLINE | ID: mdl-27774986

ABSTRACT

The COP9 signalosome (CSN) is a central component of the activation and remodelling cycle of cullin-RING E3 ubiquitin ligases (CRLs), the largest enzyme family of the ubiquitin-proteasome system in humans. CRLs are implicated in the regulation of numerous cellular processes, including cell cycle progression and apoptosis, and aberrant CRL activity is frequently associated with cancer. Remodelling of CRLs is initiated by CSN-catalysed cleavage of the ubiquitin-like activator NEDD8 from CRLs. Here we describe CSN5i-3, a potent, selective and orally available inhibitor of CSN5, the proteolytic subunit of CSN. The compound traps CRLs in the neddylated state, which leads to inactivation of a subset of CRLs by inducing degradation of their substrate recognition module. CSN5i-3 differentially affects the viability of tumour cell lines and suppresses growth of a human xenograft in mice. Our results provide insights into how CSN regulates CRLs and suggest that CSN5 inhibition has potential for anti-tumour therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Azepines/pharmacology , COP9 Signalosome Complex/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Lymphoma, Large-Cell, Anaplastic/drug therapy , Pyrazoles/pharmacology , Ubiquitin-Protein Ligases/genetics , Animals , Antineoplastic Agents/chemical synthesis , Azepines/chemical synthesis , COP9 Signalosome Complex/genetics , COP9 Signalosome Complex/metabolism , Female , HCT116 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Lymphoma, Large-Cell, Anaplastic/genetics , Lymphoma, Large-Cell, Anaplastic/metabolism , Lymphoma, Large-Cell, Anaplastic/pathology , Mice , Mice, SCID , Molecular Targeted Therapy , NEDD8 Protein/genetics , NEDD8 Protein/metabolism , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Protein Processing, Post-Translational , Proteolysis/drug effects , Pyrazoles/chemical synthesis , THP-1 Cells , Tumor Burden/drug effects , Ubiquitin-Protein Ligases/metabolism , Xenograft Model Antitumor Assays
10.
Nature ; 531(7596): 598-603, 2016 Mar 31.
Article in English | MEDLINE | ID: mdl-27029275

ABSTRACT

The cullin-RING ubiquitin E3 ligase (CRL) family comprises over 200 members in humans. The COP9 signalosome complex (CSN) regulates CRLs by removing their ubiquitin-like activator NEDD8. The CUL4A-RBX1-DDB1-DDB2 complex (CRL4A(DDB2)) monitors the genome for ultraviolet-light-induced DNA damage. CRL4A(DBB2) is inactive in the absence of damaged DNA and requires CSN to regulate the repair process. The structural basis of CSN binding to CRL4A(DDB2) and the principles of CSN activation are poorly understood. Here we present cryo-electron microscopy structures for CSN in complex with neddylated CRL4A ligases to 6.4 Å resolution. The CSN conformers defined by cryo-electron microscopy and a novel apo-CSN crystal structure indicate an induced-fit mechanism that drives CSN activation by neddylated CRLs. We find that CSN and a substrate cannot bind simultaneously to CRL4A, favouring a deneddylated, inactive state for substrate-free CRL4 complexes. These architectural and regulatory principles appear conserved across CRL families, allowing global regulation by CSN.


Subject(s)
Biocatalysis , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Peptide Hydrolases/metabolism , Peptide Hydrolases/ultrastructure , Allosteric Regulation , Apoproteins/chemistry , Apoproteins/metabolism , Apoproteins/ultrastructure , Binding Sites , COP9 Signalosome Complex , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Carrier Proteins/ultrastructure , Cryoelectron Microscopy , Crystallography, X-Ray , Cullin Proteins/chemistry , Cullin Proteins/metabolism , Cullin Proteins/ultrastructure , DNA Damage , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/ultrastructure , Humans , Kinetics , Models, Molecular , Multiprotein Complexes/chemistry , Peptide Hydrolases/chemistry , Protein Binding , Ubiquitination , Ubiquitins/metabolism
11.
J Allergy Clin Immunol ; 135(4): 1031-1043.e6, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25240785

ABSTRACT

BACKGROUND: Anaphylaxis is an acute, potentially lethal, multisystem syndrome resulting from the sudden release of mast cell-derived mediators into the circulation. OBJECTIVES AND METHODS: We report here that a plasma protease cascade, the factor XII-driven contact system, critically contributes to the pathogenesis of anaphylaxis in both murine models and human subjects. RESULTS: Deficiency in or pharmacologic inhibition of factor XII, plasma kallikrein, high-molecular-weight kininogen, or the bradykinin B2 receptor, but not the B1 receptor, largely attenuated allergen/IgE-mediated mast cell hyperresponsiveness in mice. Reconstitutions of factor XII null mice with human factor XII restored susceptibility for allergen/IgE-mediated hypotension. Activated mast cells systemically released heparin, which provided a negatively charged surface for factor XII autoactivation. Activated factor XII generates plasma kallikrein, which proteolyzes kininogen, leading to the liberation of bradykinin. We evaluated the contact system in patients with anaphylaxis. In all 10 plasma samples immunoblotting revealed activation of factor XII, plasma kallikrein, and kininogen during the acute phase of anaphylaxis but not at basal conditions or in healthy control subjects. The severity of anaphylaxis was associated with mast cell degranulation, increased plasma heparin levels, the intensity of contact system activation, and bradykinin formation. CONCLUSIONS: In summary, the data collectively show a role of the contact system in patients with anaphylaxis and support the hypothesis that targeting bradykinin generation and signaling provides a novel and alternative treatment strategy for anaphylactic attacks.


Subject(s)
Anaphylaxis/immunology , Anaphylaxis/metabolism , Factor XII/metabolism , Hypersensitivity/immunology , Hypersensitivity/metabolism , Mast Cells/immunology , Adult , Aged , Anaphylaxis/complications , Anaphylaxis/genetics , Animals , Biomarkers , Bradykinin/metabolism , Disease Models, Animal , Factor XII/antagonists & inhibitors , Factor XII/genetics , Female , Humans , Hypersensitivity/complications , Hypersensitivity/genetics , Hypotension/etiology , Kininogens/metabolism , Male , Mice, Knockout , Middle Aged , Receptor, Bradykinin B2/genetics , Receptor, Bradykinin B2/metabolism , Signal Transduction , Time Factors , Young Adult
12.
Nature ; 512(7513): 161-5, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25043011

ABSTRACT

Ubiquitination is a crucial cellular signalling process, and is controlled on multiple levels. Cullin-RING E3 ubiquitin ligases (CRLs) are regulated by the eight-subunit COP9 signalosome (CSN). CSN inactivates CRLs by removing their covalently attached activator, NEDD8. NEDD8 cleavage by CSN is catalysed by CSN5, a Zn(2+)-dependent isopeptidase that is inactive in isolation. Here we present the crystal structure of the entire ∼350-kDa human CSN holoenzyme at 3.8 Å resolution, detailing the molecular architecture of the complex. CSN has two organizational centres: a horseshoe-shaped ring created by its six proteasome lid-CSN-initiation factor 3 (PCI) domain proteins, and a large bundle formed by the carboxy-terminal α-helices of every subunit. CSN5 and its dimerization partner, CSN6, are intricately embedded at the core of the helical bundle. In the substrate-free holoenzyme, CSN5 is autoinhibited, which precludes access to the active site. We find that neddylated CRL binding to CSN is sensed by CSN4, and communicated to CSN5 with the assistance of CSN6, resulting in activation of the deneddylase.


Subject(s)
Models, Molecular , Multiprotein Complexes/chemistry , Peptide Hydrolases/chemistry , Adaptor Proteins, Signal Transducing , COP9 Signalosome Complex , Catalytic Domain , Crystallography, X-Ray , Enzyme Activation , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Peptide Hydrolases/metabolism , Protein Binding , Protein Structure, Tertiary , Transcription Factors/metabolism
13.
Nature ; 512(7512): 49-53, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25043012

ABSTRACT

In the 1950s, the drug thalidomide, administered as a sedative to pregnant women, led to the birth of thousands of children with multiple defects. Despite the teratogenicity of thalidomide and its derivatives lenalidomide and pomalidomide, these immunomodulatory drugs (IMiDs) recently emerged as effective treatments for multiple myeloma and 5q-deletion-associated dysplasia. IMiDs target the E3 ubiquitin ligase CUL4-RBX1-DDB1-CRBN (known as CRL4(CRBN)) and promote the ubiquitination of the IKAROS family transcription factors IKZF1 and IKZF3 by CRL4(CRBN). Here we present crystal structures of the DDB1-CRBN complex bound to thalidomide, lenalidomide and pomalidomide. The structure establishes that CRBN is a substrate receptor within CRL4(CRBN) and enantioselectively binds IMiDs. Using an unbiased screen, we identified the homeobox transcription factor MEIS2 as an endogenous substrate of CRL4(CRBN). Our studies suggest that IMiDs block endogenous substrates (MEIS2) from binding to CRL4(CRBN) while the ligase complex is recruiting IKZF1 or IKZF3 for degradation. This dual activity implies that small molecules can modulate an E3 ubiquitin ligase and thereby upregulate or downregulate the ubiquitination of proteins.


Subject(s)
Peptide Hydrolases/chemistry , Thalidomide/chemistry , Ubiquitin-Protein Ligases/chemistry , Adaptor Proteins, Signal Transducing , Crystallography, X-Ray , DNA-Binding Proteins/agonists , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Humans , Lenalidomide , Models, Molecular , Multiprotein Complexes/agonists , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Peptide Hydrolases/metabolism , Protein Binding , Structure-Activity Relationship , Substrate Specificity , Thalidomide/analogs & derivatives , Thalidomide/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/metabolism
14.
J Biomol Screen ; 19(6): 870-7, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24487225

ABSTRACT

Fluorescence lifetime (FLT)-based assays have developed to become highly attractive tools in drug discovery. All recently published examples of FLT-based assays essentially describe their use for monitoring enzyme-mediated peptide modifications, such as proteolytic cleavage or phosphorylation/dephosphorylation. Here we report the development of competitive binding assays as novel, inhibitor-centric assays, principally employing the FLT of the acridone dye Puretime 14 (PT14) as the readout parameter. Exemplified with two case studies on human serine proteases, the details of the rationale for both the design and synthesis of probes (i.e., active site-directed low-molecular-weight inhibitors conjugated to PT14) are provided. Data obtained from testing inhibitors with the novel assay format match those obtained with alternative formats such as FLT-based protease activity and time-resolved fluorescence resonance energy transfer-based competitive binding assays.


Subject(s)
Drug Discovery/methods , Protease Inhibitors/chemistry , Spectrometry, Fluorescence/methods , Acridones/chemistry , Binding, Competitive , Buffers , Catalytic Domain , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/chemistry , Humans , Hydrogen-Ion Concentration , Inhibitory Concentration 50 , Kinetics , Lung/enzymology , Molecular Conformation , Molecular Weight , Peptides/chemistry , Protein Binding , Recombinant Proteins/chemistry , Serine Proteases/chemistry , Tryptases/chemistry
15.
Bioorg Med Chem Lett ; 24(3): 731-6, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24439847

ABSTRACT

The successful launches of dipeptidyl peptidase IV (DPP IV) inhibitors as oral anti-diabetics warrant and spur the further quest for additional chemical entities in this promising class of therapeutics. Numerous pharmaceutical companies have pursued their proprietary candidates towards the clinic, resulting in a large body of published chemical structures associated with DPP IV. Herein, we report the discovery of a novel chemotype for DPP IV inhibition based on the C-(1-aryl-cyclohexyl)-methylamine scaffold and its optimization to compounds which selectively inhibit DPP IV at low-nM potency and exhibit an excellent oral pharmacokinetic profile in the rat.


Subject(s)
Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/chemical synthesis , Dipeptidyl-Peptidase IV Inhibitors/pharmacokinetics , Drug Discovery , Methylamines/chemical synthesis , Methylamines/pharmacokinetics , Adamantane/analogs & derivatives , Adamantane/chemistry , Adamantane/pharmacology , Administration, Oral , Animals , Caco-2 Cells , Crystallography, X-Ray , Cyclization , Dipeptidyl-Peptidase IV Inhibitors/chemistry , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Enzyme Activation/drug effects , Humans , Inhibitory Concentration 50 , Methylamines/chemistry , Methylamines/pharmacology , Molecular Structure , Nitriles/chemistry , Nitriles/pharmacology , Pyrazines/chemistry , Pyrazines/pharmacology , Pyrrolidines/chemistry , Pyrrolidines/pharmacology , Rats , Sitagliptin Phosphate , Triazoles/chemistry , Triazoles/pharmacology , Vildagliptin
16.
J Med Chem ; 56(6): 2207-17, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23425156

ABSTRACT

The small-molecule trans-3,4-disubstituted pyrrolidine 6 was identified from in silico three-dimensional (3D) pharmacophore searches based on known X-ray structures of renin-inhibitor complexes and demonstrated to be a weakly active inhibitor of the human enzyme. The unexpected binding mode of the more potent enantiomer (3S,4S)-6a in an extended conformation spanning the nonprime and S1' pockets of the recombinant human (rh)-renin active site was elucidated by X-ray crystallography. Initial structure-activity relationship work focused on modifications of the hydrophobic diphenylamine portion positioned in S1 and extending toward the S2 pocket. Replacement with an optimized P3-P1 pharmacophore interacting to the nonsubstrate S3(sp) cavity eventually resulted in significantly improved in vitro potency and selectivity. The prototype analogue (3S,4S)-12a of this new class of direct renin inhibitors exerted blood pressure lowering effects in a hypertensive double-transgenic rat model after oral administration.


Subject(s)
Drug Discovery , Models, Molecular , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Pyrrolidines/chemistry , Pyrrolidines/pharmacology , Renin/antagonists & inhibitors , Administration, Oral , Animals , Biological Availability , Computational Biology , Humans , Protease Inhibitors/administration & dosage , Protease Inhibitors/pharmacokinetics , Protein Conformation , Pyrrolidines/administration & dosage , Pyrrolidines/pharmacokinetics , Rats , Renin/chemistry , Structure-Activity Relationship
17.
J Med Chem ; 56(6): 2196-206, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23360239

ABSTRACT

A small library of fragments comprising putative recognition motifs for the catalytic dyad of aspartic proteases was generated by in silico similarity searches within the corporate compound deck based on rh-renin active site docking and scoring filters. Subsequent screening by NMR identified the low-affinity hits 3 and 4 as competitive active site binders, which could be shown by X-ray crystallography to bind to the hydrophobic S3-S1 pocket of rh-renin. As part of a parallel multiple hit-finding approach, the 3,5-disubstituted piperidine (rac)-5 was discovered by HTS using a enzymatic assay. X-ray crystallography demonstrated the eutomer (3S,5R)-5 to be a peptidomimetic inhibitor binding to a nonsubstrate topography of the rh-renin prime site. The design of the potent and selective (3S,5R)-12 bearing a P3(sp)-tethered tricyclic P3-P1 pharmacophore derived from 3 is described. (3S,5R)-12 showed oral bioavailability in rats and demonstrated blood pressure lowering activity in the double-transgenic rat model.


Subject(s)
Drug Design , Piperidines/chemistry , Piperidines/pharmacology , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Renin/antagonists & inhibitors , Administration, Oral , Animals , Biological Availability , Inhibitory Concentration 50 , Models, Molecular , Piperidines/administration & dosage , Piperidines/pharmacokinetics , Protease Inhibitors/administration & dosage , Protease Inhibitors/pharmacokinetics , Protein Conformation , Rats , Renin/chemistry
18.
Bioorg Med Chem Lett ; 22(3): 1464-8, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22177783

ABSTRACT

Novel deazaxanthine-based DPP-4 inhibitors have been identified that are potent (IC(50) <10nM) and highly selective versus other dipeptidyl peptidases. Their synthesis and SAR are reported, along with initial efforts to improve the PK profile through decoration of the deazaxanthine core. Optimisation of compound 3a resulted in the identification of compound (S)-4i, which displayed an improved in vitro and ADME profile. Further enhancements to the PK profile were possible by changing from the deazahypoxanthine to the deazaxanthine template, culminating in compound 12g, which displayed good ex vivo DPP-4 inhibition and a superior PK profile in rat, suggestive of once daily dosing in man.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/chemistry , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Animals , Caco-2 Cells , Crystallography, X-Ray , Dipeptidyl-Peptidase IV Inhibitors/chemical synthesis , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Enzyme Activation/drug effects , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/therapeutic use , Humans , Inhibitory Concentration 50 , Male , Models, Molecular , Molecular Structure , Rats , Structure-Activity Relationship
19.
Proc Natl Acad Sci U S A ; 108(52): 21052-6, 2011 Dec 27.
Article in English | MEDLINE | ID: mdl-22160684

ABSTRACT

Hepatitis C virus (HCV) infection is a global health burden with over 170 million people infected worldwide. In a significant portion of patients chronic hepatitis C infection leads to serious liver diseases, including fibrosis, cirrhosis, and hepatocellular carcinoma. The HCV NS3 protein is essential for viral polyprotein processing and RNA replication and hence viral replication. It is composed of an N-terminal serine protease domain and a C-terminal helicase/NTPase domain. For full activity, the protease requires the NS4A protein as a cofactor. HCV NS3/4A protease is a prime target for developing direct-acting antiviral agents. First-generation NS3/4A protease inhibitors have recently been introduced into clinical practice, markedly changing HCV treatment options. To date, crystal structures of HCV NS3/4A protease inhibitors have only been reported in complex with the protease domain alone. Here, we present a unique structure of an inhibitor bound to the full-length, bifunctional protease-helicase NS3/4A and show that parts of the P4 capping and P2 moieties of the inhibitor interact with both protease and helicase residues. The structure sheds light on inhibitor binding to the more physiologically relevant form of the enzyme and supports exploring inhibitor-helicase interactions in the design of the next generation of HCV NS3/4A protease inhibitors. In addition, small angle X-ray scattering confirmed the observed protease-helicase domain assembly in solution.


Subject(s)
Carrier Proteins/chemistry , Hepatitis C/drug therapy , Models, Molecular , Protease Inhibitors/chemistry , Protein Conformation , Viral Nonstructural Proteins/chemistry , Carrier Proteins/metabolism , Chromatography, Gel , Crystallization , Crystallography, X-Ray , Escherichia coli , Inhibitory Concentration 50 , Intracellular Signaling Peptides and Proteins , Protease Inhibitors/metabolism , Protease Inhibitors/therapeutic use , Scattering, Small Angle , Viral Nonstructural Proteins/metabolism
20.
Expert Opin Drug Discov ; 6(6): 663-70, 2011 Jun.
Article in English | MEDLINE | ID: mdl-22646154

ABSTRACT

INTRODUCTION: Fluorescence lifetime assays complement the portfolio of established assay formats available in drug discovery, particularly with the recent advances in microplate readers and the commercial availability of novel fluorescent labels. Fluorescence lifetime assists in lowering complexity of compound screening assays, affording a modular, toolbox-like approach to assay development and yielding robust homogeneous assays. AREAS COVERED: To date, materials and procedures have been reported for biochemical assays on proteases, as well as on protein kinases and phosphatases. This article gives an overview of two assay families, distinguished by the origin of the fluorescence signal modulation. EXPERT OPINION: The pharmaceutical industry demands techniques with a robust, integrated compound profiling process and short turnaround times. Fluorescence lifetime assays have already helped the drug discovery field, in this sense, by enhancing productivity during the hit-to-lead and lead optimization phases. Future work will focus on covering other biochemical molecular modifications by investigating the detailed photo-physical mechanisms underlying the fluorescence signal.

SELECTION OF CITATIONS
SEARCH DETAIL
...