Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Blood ; 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38905635

ABSTRACT

The interaction between menin and histone-lysine N-methyltransferase 2A (KMT2A) is a critical dependency for KMT2A- or nucleophosmin 1 (NPM1)-altered leukemias and an emerging opportunity for therapeutic development. JNJ-75276617 is a novel, orally bioavailable, potent, and selective protein-protein interaction inhibitor of the binding between menin and KMT2A. In KMT2A-rearranged (KMT2A-r) and NPM1-mutant (NPM1c) AML cells, JNJ-75276617 inhibited the association of the menin-KMT2A complex with chromatin at target gene promoters, resulting in reduced expression of several menin-KMT2A target genes, including MEIS1 and FLT3. JNJ-75276617 displayed potent anti-proliferative activity across several AML and ALL cell lines and patient samples harboring KMT2A- or NPM1-alterations in vitro. In xenograft models of AML and ALL, JNJ-75276617 reduced leukemic burden and provided a significant dose-dependent survival benefit accompanied by expression changes of menin-KMT2A target genes. JNJ-75276617 demonstrated synergistic effects with gilteritinib in vitro in AML cells harboring KMT2A-r. JNJ-75276617 further exhibited synergistic effects with venetoclax and azacitidine in AML cells bearing KMT2A-r in vitro, and significantly increased survival in mice. Interestingly, JNJ-75276617 showed potent anti-proliferative activity in cell lines engineered with recently discovered mutations (MEN1M327I or MEN1T349M) that developed in patients refractory to the menin-KMT2A inhibitor revumenib. A co-crystal structure of menin in complex with JNJ-75276617 indicates a unique binding mode distinct from other menin-KMT2A inhibitors, including revumenib. JNJ-75276617 is being clinically investigated for acute leukemias harboring KMT2A or NPM1 alterations, as a monotherapy for relapsed/refractory (R/R) acute leukemia (NCT04811560), or in combination with AML-directed therapies (NCT05453903).

2.
Proteins ; 92(7): 819-829, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38337153

ABSTRACT

Proteolysis Targeting Chimeras (PROTACs) are an emerging therapeutic modality and chemical biology tools for Targeted Protein Degradation (TPD). PROTACs contain a ligand targeting the protein of interest, a ligand recruiting an E3 ligase and a linker connecting these two ligands. There are over 600 E3 ligases known so far, but only a handful have been exploited for TPD applications. A key reason for this is the scarcity of ligands binding various E3 ligases and the paucity of structural data available, which complicates ligand design across the family. In this study, we aim to progress PROTAC discovery by proposing a shortlist of E3 ligases that can be prioritized for covalent targeting by performing systematic structural ligandability analysis on a chemoproteomic dataset of potentially reactive cysteines across hundreds of E3 ligases. One of the goals of this study is to apply AlphaFold (AF) models for ligandability evaluations, as for a vast majority of these ligases an experimental structure is not available in the protein data bank (PDB). Using a combination of pocket features, AF model quality and additional aspects, we propose a shortlist of E3 ligases and corresponding cysteines that can be prioritized to potentially discover covalent ligands and expand the PROTAC toolbox.


Subject(s)
Cysteine , Protein Binding , Proteolysis , Ubiquitin-Protein Ligases , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Ligands , Cysteine/chemistry , Cysteine/metabolism , Humans , Models, Molecular , Binding Sites , Databases, Protein
3.
J Org Chem ; 85(23): 14989-15005, 2020 12 04.
Article in English | MEDLINE | ID: mdl-33196210

ABSTRACT

A novel class of substituted spiro[3.4]octanes can be accessed via a [2 + 2]-cycloaddition of dichloroketene on a readily prepared exo-methylene cyclopentane building block. This reaction sequence was found to be robust on a multigram scale and afforded a central spirocyclobutanone scaffold for carbocyclic nucleosides. The reactivity of this constrained building block was evaluated and compared to the corresponding 4'-spirocyclic furanose analogues. Density functional theory calculations were performed to support the observed selectivity in the carbonyl reduction of spirocyclobutanone building blocks. Starting from novel spirocyclic intermediates, we exemplified the preparation of an undescribed class of carbocyclic nucleoside analogues and provided a proof of concept for application as inhibitors for the protein methyltransferase target PRMT5.


Subject(s)
Cyclopentanes , Nucleosides , Cycloaddition Reaction
4.
Chemistry ; 25(67): 15419-15423, 2019 Dec 02.
Article in English | MEDLINE | ID: mdl-31609050

ABSTRACT

Despite the large variety of modified nucleosides that have been reported, the preparation of constrained 4'-spirocyclic adenosine analogues has received very little attention. We discovered that the [2+2]-cycloaddition of dichloroketene on readily available 4'-exo-methylene furanose sugars efficiently results in the diastereoselective formation of novel 4'-spirocyclobutanones. The reaction mechanism was investigated via density functional theory (DFT) and found to proceed either via a non-synchronous or stepwise reaction sequence, controlled by the stereochemistry at the 3'-position of the sugar substrate. The obtained dichlorocyclobutanones were converted into nucleoside analogues, providing access to a novel class of chiral 4'-spirocyclobutyl adenosine mimetics in eight steps from commercially available sugars. Assessment of the biological activity of designed 4'-spirocyclic adenosine analogues identified potent inhibitors for protein methyltransferase target PRMT5.


Subject(s)
Adenosine/chemistry , Nucleosides/analogs & derivatives , Nucleosides/chemical synthesis , Carbohydrates/chemistry , Cycloaddition Reaction , Density Functional Theory , Dichloroethylenes/chemistry , Glycosylation , Metals/chemistry , Molecular Structure , Oxidation-Reduction , Stereoisomerism , Thermodynamics
5.
Bioorg Med Chem Lett ; 28(19): 3216-3221, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30143425

ABSTRACT

In a continuing effort to discover novel TLR agonists, herein we report on the discovery and structure-activity relationship of novel tetrahydropyridopyrimidine TLR 7/8 agonists. Optimization of this series towards dual agonist activity and a high clearance profile resulted in the identification of compound 52a1. Evaluation in vivo revealed an interferon stimulated response (ISG) in mice with limited systemic exposure and demonstrated the potential in antiviral treatment or as a vaccine adjuvant.


Subject(s)
Pyrimidines/pharmacology , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists , Administration, Oral , Animals , Drug Design , Mice , Structure-Activity Relationship
6.
Proteins ; 83(7): 1316-26, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25974248

ABSTRACT

Protein families involved in chromatin-templated events are emerging as novel target classes in oncology and other disease areas. The ability to discover selective inhibitors against chromatin factors depends on the presence of structural features that are unique to the targeted sites. To evaluate challenges and opportunities toward the development of selective inhibitors, we calculated all pair wise structural distances between 575 structures from the protein databank representing 163 unique binding pockets found in protein domains that write, read or erase post-translational modifications on histones, DNA, and RNA. We find that the structural similarity of binding sites does not always follow the sequence similarity of protein domains. Our analysis reveals increased risks of activity across target-class for compounds competing with the cofactor of protein arginine methyltransferases, lysine acetyltransferases, and sirtuins, while exploiting the conformational plasticity of a protein target is a path toward selective inhibition. The structural diversity landscape of the epigenetics pocketome can be explored via an open-access graphic user interface at thesgc.org/epigenetics_pocketome.


Subject(s)
Antineoplastic Agents/chemistry , Epigenesis, Genetic , Histones/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Protein Processing, Post-Translational , Small Molecule Libraries/chemistry , Software , Acetyltransferases/antagonists & inhibitors , Acetyltransferases/chemistry , Acetyltransferases/genetics , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Binding Sites , Chromatin/chemistry , Chromatin/drug effects , DNA, Neoplasm/antagonists & inhibitors , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , Databases, Protein , Histones/chemistry , Histones/genetics , Humans , Internet , Ligands , Models, Molecular , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasms/chemistry , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Protein Binding , Protein Structure, Tertiary , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/chemistry , Protein-Arginine N-Methyltransferases/genetics , RNA, Neoplasm/antagonists & inhibitors , RNA, Neoplasm/chemistry , RNA, Neoplasm/genetics , Sirtuins/antagonists & inhibitors , Sirtuins/chemistry , Sirtuins/genetics , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacology
7.
J Phys Chem B ; 128(26): 6233-6245, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38904218

ABSTRACT

The characterization of cryptic pockets has been elusive, despite substantial efforts. Computational modeling approaches, such as molecular dynamics (MD) simulations, can provide atomic-level details of binding site motions and binding pathways. However, the time scale that MD can achieve at a reasonable cost often limits its application for cryptic pocket identification. Enhanced sampling techniques can improve the efficiency of MD simulations by focused sampling of important regions of the protein, but prior knowledge of the simulated system is required to define the appropriate coordinates. In the case of a novel, unknown cryptic pocket, such information is not available, limiting the application of enhanced sampling techniques for cryptic pocket identification. In this work, we explore the ability of SiteMap and Site Finder, widely used commercial packages for pocket identification, to detect focus points on the protein and further apply other advanced computational methods. The information gained from this analysis enables the use of computational modeling, including enhanced MD sampling techniques, to explore potential cryptic binding pockets suggested by SiteMap and Site Finder. Here, we examined SiteMap and Site Finder results on 136 known cryptic pockets from a combination of the PocketMiner dataset (a recently curated set of cryptic pockets), the Cryptosite Set (a classic set of cryptic pockets), and Natural killer group 2D (NKG2D, a protein target where a cryptic pocket is confirmed). Our findings demonstrate the application of existing, well-studied tools in efficiently mapping potential regions harboring cryptic pockets.


Subject(s)
Molecular Dynamics Simulation , Binding Sites , Proteins/chemistry
8.
J Chem Theory Comput ; 19(21): 7437-7458, 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-37902715

ABSTRACT

Membrane proteins have diverse functions within cells and are well-established drug targets. The advances in membrane protein structural biology have revealed drug and lipid binding sites on membrane proteins, while computational methods such as molecular simulations can resolve the thermodynamic basis of these interactions. Particularly, alchemical free energy calculations have shown promise in the calculation of reliable and reproducible binding free energies of protein-ligand and protein-lipid complexes in membrane-associated systems. In this review, we present an overview of representative alchemical free energy studies on G-protein-coupled receptors, ion channels, transporters as well as protein-lipid interactions, with emphasis on best practices and critical aspects of running these simulations. Additionally, we analyze challenges and successes when running alchemical free energy calculations on membrane-associated proteins. Finally, we highlight the value of alchemical free energy calculations calculations in drug discovery and their applicability in the pharmaceutical industry.


Subject(s)
Membrane Proteins , Molecular Dynamics Simulation , Entropy , Thermodynamics , Ligands , Lipids , Protein Binding
9.
SLAS Discov ; 27(5): 306-313, 2022 07.
Article in English | MEDLINE | ID: mdl-35513262

ABSTRACT

The dysregulation of the PRC1/2 complex plays a key role in lineage plasticity in prostate cancer and may be required to maintain neuroendocrine phenotype. [1] CBX2, a key component of the canonical PRC1 complex, is an epigenetic reader, recognizing trimethylated lysine on histone 3 (H3K27me3) [2] and is overexpressed in metastatic neuroendocrine prostate cancer. [3,4] We implemented a screening strategy using nucleosome substrates to identify inhibitors of CBX2 binding to chromatin. Construct design and phosphorylation state of CBX2 were critical for successful implementation and execution of an HTS library screen. A rigorous screening funnel including counter and selectivity assays allowed us to quickly focus on true positive hit matter. Two distinct non-peptide-like chemotypes were identified and confirmed in orthogonal biochemical and biophysical assays demonstrating disruption of CBX2 binding to nucleosomes and direct binding to purified CBX2, respectively.


Subject(s)
Polycomb Repressive Complex 1 , Prostatic Neoplasms , Cell Nucleus/metabolism , Chromatin , Histones/metabolism , Humans , Male , Polycomb Repressive Complex 1/genetics , Prostatic Neoplasms/metabolism
10.
ACS Med Chem Lett ; 12(8): 1245-1252, 2021 Aug 12.
Article in English | MEDLINE | ID: mdl-34422225

ABSTRACT

Androgen receptor (AR) transcriptional reactivation plays a key role in the development and progression of lethal castration-resistant prostate cancer (CRPC). Recurrent alterations in the AR enable persistent AR pathway signaling and drive resistance to the treatment of second-generation antiandrogens. AR F877L, a point mutation in the ligand binding domain of the AR, was identified in patients who acquired resistance to enzalutamide or apalutamide. In parallel to our previous structure-activity relationship (SAR) studies of compound 4 (JNJ-pan-AR) and clinical stage compound 5 (JNJ-63576253), we discovered additional AR antagonists that provide opportunities for future development. Here we report a highly potent series of spirocyclic thiohydantoins as AR antagonists for the treatment of the F877L mutant and wild-type CRPC.

11.
J Med Chem ; 64(2): 909-924, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33470111

ABSTRACT

Persistent androgen receptor (AR) activation drives therapeutic resistance to second-generation AR pathway inhibitors and contributes to the progression of advanced prostate cancer. One resistance mechanism is point mutations in the ligand binding domain of AR that can transform antagonists into agonists. The AR F877L mutation, identified in patients treated with enzalutamide or apalutamide, confers resistance to both enzalutamide and apalutamide. Compound 4 (JNJ-pan-AR) was identified as a pan-AR antagonist with potent activity against wild-type and clinically relevant AR mutations including F877L. Metabolite identification studies revealed a latent bioactivation pathway associated with 4. Subsequent lead optimization of 4 led to amelioration of this pathway and nomination of 5 (JNJ-63576253) as a clinical stage, next-generation AR antagonist for the treatment of castration-resistant prostate cancer (CRPC).


Subject(s)
Androgen Receptor Antagonists/pharmacology , Nitriles/pharmacology , Picolines/pharmacology , Piperidines/pharmacology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms/drug therapy , Pyridines/pharmacology , Spiro Compounds/pharmacology , Androgen Receptor Antagonists/pharmacokinetics , Androgen Receptor Antagonists/therapeutic use , Animals , Biotransformation , Cell Line, Tumor , Dogs , Drug Discovery , Drug Resistance, Neoplasm/genetics , Hepatocytes/metabolism , Humans , Male , Models, Molecular , Mutation , Nitriles/pharmacokinetics , Nitriles/therapeutic use , Picolines/pharmacokinetics , Picolines/therapeutic use , Piperidines/pharmacokinetics , Piperidines/therapeutic use , Prostatic Neoplasms/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Pyridines/pharmacokinetics , Pyridines/therapeutic use , Rats , Spiro Compounds/pharmacokinetics , Spiro Compounds/therapeutic use , Structure-Activity Relationship
12.
Mol Cancer Ther ; 20(5): 763-774, 2021 05.
Article in English | MEDLINE | ID: mdl-33649102

ABSTRACT

Numerous mechanisms of resistance arise in response to treatment with second-generation androgen receptor (AR) pathway inhibitors in metastatic castration-resistant prostate cancer (mCRPC). Among these, point mutations in the ligand binding domain can transform antagonists into agonists, driving the disease through activation of AR signaling. To address this unmet need, we report the discovery of JNJ-63576253, a next-generation AR pathway inhibitor that potently abrogates AR signaling in models of human prostate adenocarcinoma. JNJ-63576253 is advancing as a clinical candidate with potential effectiveness in the subset of patients who do not respond to or are progressing while on second-generation AR-targeted therapeutics.


Subject(s)
Androgen Receptor Antagonists/therapeutic use , Prostatic Neoplasms, Castration-Resistant/drug therapy , Protein Domains/genetics , Androgen Receptor Antagonists/pharmacology , Animals , Cell Line, Tumor , Humans , Ligands , Male , Mice , Models, Molecular , Mutation , Rats , Xenograft Model Antitumor Assays
13.
J Med Chem ; 64(15): 11570-11596, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34279934

ABSTRACT

Selective cyclooxygenase (COX)-2 inhibitors have been extensively studied for colorectal cancer (CRC) chemoprevention. Celecoxib has been reported to reduce the incidence of colorectal adenomas and CRC but is also associated with an increased risk of cardiovascular events. Here, we report a series of gut-restricted, selective COX-2 inhibitors characterized by high colonic exposure and minimized systemic exposure. By establishing acute ex vivo 18F-FDG uptake attenuation as an efficacy proxy, we identified a subset of analogues that demonstrated statistically significant in vivo dose-dependent inhibition of adenoma progression and survival extension in an APCmin/+ mouse model. However, in vitro-in vivo correlation analysis showed their chemoprotective effects were driven by residual systemic COX-2 inhibition, rationalizing their less than expected efficacies and highlighting the challenges associated with COX-2-mediated CRC disease chemoprevention.


Subject(s)
Antineoplastic Agents/pharmacology , Celecoxib/pharmacology , Colorectal Neoplasms/drug therapy , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/metabolism , Etoricoxib/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Celecoxib/chemistry , Celecoxib/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cyclooxygenase 2 Inhibitors/chemistry , Cyclooxygenase 2 Inhibitors/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Etoricoxib/chemistry , Etoricoxib/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Structure-Activity Relationship
14.
Mol Cancer Ther ; 20(12): 2317-2328, 2021 12.
Article in English | MEDLINE | ID: mdl-34583982

ABSTRACT

The protein arginine methyltransferase 5 (PRMT5) methylates a variety of proteins involved in splicing, multiple signal transduction pathways, epigenetic control of gene expression, and mechanisms leading to protein expression required for cellular proliferation. Dysregulation of PRMT5 is associated with clinical features of several cancers, including lymphomas, lung cancer, and breast cancer. Here, we describe the characterization of JNJ-64619178, a novel, selective, and potent PRMT5 inhibitor, currently in clinical trials for patients with advanced solid tumors, non-Hodgkin's lymphoma, and lower-risk myelodysplastic syndrome. JNJ-64619178 demonstrated a prolonged inhibition of PRMT5 and potent antiproliferative activity in subsets of cancer cell lines derived from various histologies, including lung, breast, pancreatic, and hematological malignancies. In primary acute myelogenous leukemia samples, the presence of splicing factor mutations correlated with a higher ex vivo sensitivity to JNJ-64619178. Furthermore, the potent and unique mechanism of inhibition of JNJ-64619178, combined with highly optimized pharmacological properties, led to efficient tumor growth inhibition and regression in several xenograft models in vivo, with once-daily or intermittent oral-dosing schedules. An increase in splicing burden was observed upon JNJ-64619178 treatment. Overall, these observations support the continued clinical evaluation of JNJ-64619178 in patients with aberrant PRMT5 activity-driven tumors.


Subject(s)
Enzyme Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Protein-Arginine N-Methyltransferases/drug effects , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Animals , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Humans , Lung Neoplasms/pathology , Mice , Pyrimidines/pharmacology , Pyrroles/pharmacology
15.
Biochim Biophys Acta ; 1792(2): 132-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19084593

ABSTRACT

TWINKLE is a DNA helicase needed for mitochondrial DNA replication. In lower eukaryotes the protein also harbors a primase activity, which is lost from TWINKLE encoded by mammalian cells. Mutations in TWINKLE underlie autosomal dominant progressive external ophthalmoplegia (adPEO), a disorder associated with multiple deletions in the mtDNA. Four different adPEO-causing mutations (W315L, K319T, R334Q, and P335L) are located in the N-terminal domain of TWINKLE. The mutations cause a dramatic decrease in ATPase activity, which is partially overcome in the presence of single-stranded DNA. The mutated proteins have defects in DNA helicase activity and cannot support normal levels of DNA replication. To explain the phenotypes, we use a molecular model of TWINKLE based on sequence similarities with the phage T7 gene 4 protein. The four adPEO-causing mutations are located in a region required to bind single-stranded DNA. These mutations may therefore impair an essential element of the catalytic cycle in hexameric helicases, i.e. the interplay between single-stranded DNA binding and ATP hydrolysis.


Subject(s)
DNA Helicases/chemistry , DNA Helicases/metabolism , Ophthalmoplegia, Chronic Progressive External/enzymology , Amino Acid Sequence , DNA Helicases/genetics , DNA Helicases/isolation & purification , DNA Replication/genetics , DNA, Mitochondrial/genetics , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Ophthalmoplegia, Chronic Progressive External/genetics , Protein Structure, Quaternary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
16.
Nucleic Acids Res ; 36(5): 1508-16, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18203740

ABSTRACT

Locked nucleic acid (LNA) is a chemically modified nucleic acid with its sugar ring locked in an RNA-like (C3'-endo) conformation. LNAs show extraordinary thermal stabilities when hybridized with DNA, RNA or LNA itself. We performed molecular dynamics simulations on five isosequential duplexes (LNA-DNA, LNA-LNA, LNA-RNA, RNA-DNA and RNA-RNA) in order to characterize their structure, dynamics and hydration. Structurally, the LNA-DNA and LNA-RNA duplexes are found to be similar to regular RNA-DNA and RNA-RNA duplexes, whereas the LNA-LNA duplex is found to have its helix partly unwound and does not resemble RNA-RNA duplex in a number of properties. Duplexes with an LNA strand have on average longer interstrand phosphate distances compared to RNA-DNA and RNA-RNA duplexes. Furthermore, intrastrand phosphate distances in LNA strands are found to be shorter than in DNA and slightly shorter than in RNA. In case of induced sugar puckering, LNA is found to tune the sugar puckers in partner DNA strand toward C3'-endo conformations more efficiently than RNA. The LNA-LNA duplex has lesser backbone flexibility compared to the RNA-RNA duplex. Finally, LNA is less hydrated compared to DNA or RNA but is found to have a well-organized water structure.


Subject(s)
DNA/chemistry , Oligonucleotides/chemistry , RNA/chemistry , Base Pairing , Carbohydrate Conformation , Computational Biology , Computer Simulation , Models, Molecular , Nucleic Acid Conformation , Water/chemistry
17.
ACS Med Chem Lett ; 11(11): 2227-2231, 2020 Nov 12.
Article in English | MEDLINE | ID: mdl-33214833

ABSTRACT

Protein arginine methyltransferase 5 (PRMT5) is an enzyme that can symmetrically dimethylate arginine residues in histones and nonhistone proteins by using S-adenosyl methionine (SAM) as the methyl donating cofactor. We have designed a library of SAM analogues and discovered potent, cell-active, and selective spiro diamines as inhibitors of the enzymatic function of PRMT5. Crystallographic studies confirmed a very interesting binding mode, involving protein flexibility, where both the cofactor pocket and part of substrate binding site are occupied by these inhibitors.

18.
J Med Chem ; 61(14): 6236-6246, 2018 07 26.
Article in English | MEDLINE | ID: mdl-29965759

ABSTRACT

A novel series of 2,4-diaminoquinazolines was identified as potent dual Toll-like receptor (TLR) 7 and 8 agonists with reduced off-target activity. The stereochemistry of the amino alcohol was found to influence the TLR7/8 selectivity with the ( R) isomer resulting in selective TLR8 agonism. Lead optimization toward a dual agonist afforded ( S)-3-((2-amino-8-fluoroquinazolin-4-yl)amino)hexanol 31 as a potent analog, being structurally different from previously described dual agonists ( McGowan J. Med. Chem. 2016 , 59 , 7936 ). Pharmacokinetic and pharmacodynamic (PK/PD) studies revealed the desired high first pass profile aimed at limiting systemic cytokine activation. In vivo pharmacodynamic studies with lead compound 31 demonstrated production of cytokines consistent with TLR7/8 activation in mice and cynomolgus monkeys and ex vivo inhibition of hepatitis B virus (HBV).


Subject(s)
Antiviral Agents/pharmacology , Hepatitis B virus/drug effects , Quinazolines/pharmacology , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/metabolism , Animals , Antiviral Agents/chemistry , Antiviral Agents/metabolism , HEK293 Cells , Humans , Male , Mice , Molecular Docking Simulation , Protein Conformation , Quinazolines/chemistry , Quinazolines/metabolism , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Toll-Like Receptor 7/chemistry , Toll-Like Receptor 8/chemistry
19.
Elife ; 72018 04 20.
Article in English | MEDLINE | ID: mdl-29676732

ABSTRACT

Potent, selective and broadly characterized small molecule modulators of protein function (chemical probes) are powerful research reagents. The pharmaceutical industry has generated many high-quality chemical probes and several of these have been made available to academia. However, probe-associated data and control compounds, such as inactive structurally related molecules and their associated data, are generally not accessible. The lack of data and guidance makes it difficult for researchers to decide which chemical tools to choose. Several pharmaceutical companies (AbbVie, Bayer, Boehringer Ingelheim, Janssen, MSD, Pfizer, and Takeda) have therefore entered into a pre-competitive collaboration to make available a large number of innovative high-quality probes, including all probe-associated data, control compounds and recommendations on use (https://openscienceprobes.sgc-frankfurt.de/). Here we describe the chemical tools and target-related knowledge that have been made available, and encourage others to join the project.


Subject(s)
Molecular Probes/metabolism , Pharmacology/methods , Proteins/metabolism , Technology, Pharmaceutical/methods
20.
J Med Chem ; 49(12): 3595-601, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16759101

ABSTRACT

Previously, we have reported that aurintricarboxylic acid (ATA) is one of the most potent inhibitors of the DNA binding of transcription factor NF-kappaB. We now report the NF-kappaB-DNA binding inhibitory activity of ATA analogues. An electrophoretic mobility shift assay has shown that bromopyrogallol red (BPR) is the most effective inhibitor of NF-kappaB-DNA binding among the studied analogues. The molecular modeling studies showed that BPR makes a strong network of hydrogen bonds with the DNA-binding region of the p50 subunit of NF-kappaB and has electronegative potential on its peripheral surface. Because zinc has been reported to influence the DNA binding of NF-kappaB, the interaction of these analogues with zinc was studied. Chemical speciation and formation-constant studies showed that BPR forms the most stable 1:1 complex with zinc. BPR has also been found to be the most potent antioxidant among the studied analogues.


Subject(s)
Antiviral Agents/chemistry , Aurintricarboxylic Acid/analogs & derivatives , Aurintricarboxylic Acid/chemistry , Chelating Agents/chemistry , DNA/metabolism , Models, Molecular , NF-kappa B/metabolism , Organometallic Compounds/chemistry , Zinc , DNA/chemistry , Electricity , Electrophoretic Mobility Shift Assay , Hydrogen Bonding , NF-kappa B/chemistry , NF-kappa B p50 Subunit/chemistry , Protein Binding , Pyrogallol/chemistry , Structure-Activity Relationship , Zinc/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL