Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 36
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
J Med Chem ; 67(6): 4541-4559, 2024 Mar 28.
Article de Anglais | MEDLINE | ID: mdl-38466661

RÉSUMÉ

The optimization of an allosteric fragment, discovered by differential scanning fluorimetry, to an in vivo MAT2a tool inhibitor is discussed. The structure-based drug discovery approach, aided by relative binding free energy calculations, resulted in AZ'9567 (21), a potent inhibitor in vitro with excellent preclinical pharmacokinetic properties. This tool showed a selective antiproliferative effect on methylthioadenosine phosphorylase (MTAP) KO cells, both in vitro and in vivo, providing further evidence to support the utility of MAT2a inhibitors as potential anticancer therapies for MTAP-deficient tumors.


Sujet(s)
Tumeurs , Humains , Entropie , Methionine adenosyltransferase/métabolisme
2.
J Med Chem ; 67(2): 1500-1512, 2024 Jan 25.
Article de Anglais | MEDLINE | ID: mdl-38227216

RÉSUMÉ

Casitas B-lymphoma proto-oncogene-b (Cbl-b), a member of the Cbl family of RING finger E3 ubiquitin ligases, has been demonstrated to play a central role in regulating effector T-cell function. Multiple studies using gene-targeting approaches have provided direct evidence that Cbl-b negatively regulates T, B, and NK cell activation via a ubiquitin-mediated protein modulation. Thus, inhibition of Cbl-b ligase activity can lead to immune activation and has therapeutic potential in immuno-oncology. Herein, we describe the discovery and optimization of an arylpyridone series as Cbl-b inhibitors by structure-based drug discovery to afford compound 31. This compound binds to Cbl-b with an IC50 value of 30 nM and induces IL-2 production in T-cells with an EC50 value of 230 nM. Compound 31 also shows robust intracellular target engagement demonstrated through inhibition of Cbl-b autoubiquitination, inhibition of ubiquitin transfer to ZAP70, and the cellular modulation of phosphorylation of a downstream signal within the TCR axis.


Sujet(s)
Protéines proto-oncogènes c-cbl , Ubiquitin-protein ligases , Protéines proto-oncogènes c-cbl/métabolisme , Ubiquitin-protein ligases/métabolisme , Lymphocytes T/métabolisme , Phosphorylation , Ubiquitine/métabolisme
3.
ACS Med Chem Lett ; 14(12): 1848-1856, 2023 Dec 14.
Article de Anglais | MEDLINE | ID: mdl-38116444

RÉSUMÉ

Casitas B-lineage lymphoma proto-oncogene-b (Cbl-b) is a RING finger E3 ligase that is responsible for repressing T-cell, natural killer (NK) cell, and B-cell activation. The robust antitumor activity observed in Cbl-b deficient mice arising from elevated T-cell and NK-cell activity justified our discovery effort toward Cbl-b inhibitors that might show therapeutic promise in immuno-oncology, where activation of the immune system can drive the recognition and killing of cancer cells. We undertook a high-throughput screening campaign followed by structure-enabled optimization to develop a novel benzodiazepine series of potent Cbl-b inhibitors. This series displayed nanomolar levels of biochemical potency, as well as potent T-cell activation. The functional activity of this class of Cbl-b inhibitors was further corroborated with ubiquitin-based cellular assays.

4.
Chem Sci ; 14(25): 7057-7067, 2023 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-37389247

RÉSUMÉ

Understanding allosteric regulation in biomolecules is of great interest to pharmaceutical research and computational methods emerged during the last decades to characterize allosteric coupling. However, the prediction of allosteric sites in a protein structure remains a challenging task. Here, we integrate local binding site information, coevolutionary information, and information on dynamic allostery into a structure-based three-parameter model to identify potentially hidden allosteric sites in ensembles of protein structures with orthosteric ligands. When tested on five allosteric proteins (LFA-1, p38-α, GR, MAT2A, and BCKDK), the model successfully ranked all known allosteric pockets in the top three positions. Finally, we identified a novel druggable site in MAT2A confirmed by X-ray crystallography and SPR and a hitherto unknown druggable allosteric site in BCKDK validated by biochemical and X-ray crystallography analyses. Our model can be applied in drug discovery to identify allosteric pockets.

6.
ACS Med Chem Lett ; 13(8): 1295-1301, 2022 Aug 11.
Article de Anglais | MEDLINE | ID: mdl-35978693

RÉSUMÉ

The DNA-PK complex is activated by double-strand DNA breaks and regulates the non-homologous end-joining repair pathway; thus, targeting DNA-PK by inhibiting the DNA-PK catalytic subunit (DNA-PKcs) is potentially a useful therapeutic approach for oncology. A previously reported series of neutral DNA-PKcs inhibitors were modified to incorporate a basic group, with the rationale that increasing the volume of distribution while maintaining good metabolic stability should increase the half-life. However, adding a basic group introduced hERG activity, and basic compounds with modest hERG activity (IC50 = 10-15 µM) prolonged QTc (time from the start of the Q wave to the end of the T wave, corrected by heart rate) in an anaesthetized guinea pig cardiovascular model. Further optimization was necessary, including modulation of pK a, to identify compound 18, which combines low hERG activity (IC50 = 75 µM) with excellent kinome selectivity and favorable pharmacokinetic properties.

7.
J Med Chem ; 65(4): 3306-3331, 2022 02 24.
Article de Anglais | MEDLINE | ID: mdl-35133824

RÉSUMÉ

ATAD2 is an epigenetic bromodomain-containing target which is overexpressed in many cancers and has been suggested as a potential oncology target. While several small molecule inhibitors have been described in the literature, their cellular activity has proved to be underwhelming. In this work, we describe the identification of a novel series of ATAD2 inhibitors by high throughput screening, confirmation of the bromodomain region as the site of action, and the optimization campaign undertaken to improve the potency, selectivity, and permeability of the initial hit. The result is compound 5 (AZ13824374), a highly potent and selective ATAD2 inhibitor which shows cellular target engagement and antiproliferative activity in a range of breast cancer models.


Sujet(s)
ATPases associated with diverse cellular activities/antagonistes et inhibiteurs , Antinéoplasiques/synthèse chimique , Antinéoplasiques/pharmacologie , Tumeurs du sein/traitement médicamenteux , Protéines de liaison à l'ADN/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Cristallographie aux rayons X , Découverte de médicament , Tests de criblage d'agents antitumoraux , Femelle , Humains , Modèles moléculaires , Bibliothèques de petites molécules , Relation structure-activité , Spécificité du substrat , Test clonogénique de cellules souches tumorales
8.
J Med Chem ; 64(19): 14498-14512, 2021 10 14.
Article de Anglais | MEDLINE | ID: mdl-34570508

RÉSUMÉ

Poly-ADP-ribose-polymerase (PARP) inhibitors have achieved regulatory approval in oncology for homologous recombination repair deficient tumors including BRCA mutation. However, some have failed in combination with first-line chemotherapies, usually due to overlapping hematological toxicities. Currently approved PARP inhibitors lack selectivity for PARP1 over PARP2 and some other 16 PARP family members, and we hypothesized that this could contribute to toxicity. Recent literature has demonstrated that PARP1 inhibition and PARP1-DNA trapping are key for driving efficacy in a BRCA mutant background. Herein, we describe the structure- and property-based design of 25 (AZD5305), a potent and selective PARP1 inhibitor and PARP1-DNA trapper with excellent in vivo efficacy in a BRCA mutant HBCx-17 PDX model. Compound 25 is highly selective for PARP1 over other PARP family members, with good secondary pharmacology and physicochemical properties and excellent pharmacokinetics in preclinical species, with reduced effects on human bone marrow progenitor cells in vitro.


Sujet(s)
ADN , Poly (ADP-Ribose) polymerase-1 , Inhibiteurs de poly(ADP-ribose) polymérases , Poly(ADP-ribose) polymerases , Humains , Cristallographie aux rayons X , ADN/composition chimique , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Poly(ADP-ribose) polymerases/métabolisme , Spécificité du substrat
9.
J Med Chem ; 64(18): 13524-13539, 2021 09 23.
Article de Anglais | MEDLINE | ID: mdl-34478292

RÉSUMÉ

Inhibition of Mer and Axl kinases has been implicated as a potential way to improve the efficacy of current immuno-oncology therapeutics by restoring the innate immune response in the tumor microenvironment. Highly selective dual Mer/Axl kinase inhibitors are required to validate this hypothesis. Starting from hits from a DNA-encoded library screen, we optimized an imidazo[1,2-a]pyridine series using structure-based compound design to improve potency and reduce lipophilicity, resulting in a highly selective in vivo probe compound 32. We demonstrated dose-dependent in vivo efficacy and target engagement in Mer- and Axl-dependent efficacy models using two structurally differentiated and selective dual Mer/Axl inhibitors. Additionally, in vivo efficacy was observed in a preclinical MC38 immuno-oncology model in combination with anti-PD1 antibodies and ionizing radiation.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Imidazoles/usage thérapeutique , Tumeurs/traitement médicamenteux , Inhibiteurs de protéines kinases/usage thérapeutique , Pyridines/usage thérapeutique , Animaux , Antinéoplasiques/synthèse chimique , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tests de criblage d'agents antitumoraux , Femelle , Imidazoles/synthèse chimique , Mâle , Souris de lignée C57BL , Souris nude , Structure moléculaire , Inhibiteurs de protéines kinases/synthèse chimique , Protéines proto-oncogènes/métabolisme , Pyridines/synthèse chimique , Récepteurs à activité tyrosine kinase/métabolisme , Relation structure-activité , c-Mer Tyrosine kinase/métabolisme , Axl Receptor Tyrosine Kinase
10.
Curr Res Struct Biol ; 3: 19-29, 2021.
Article de Anglais | MEDLINE | ID: mdl-34235483

RÉSUMÉ

Helicobacter pylori (H. pylori) uses several outer membrane proteins for adhering to its host's gastric mucosa, an important step in establishing and preserving colonization. Several adhesins (SabA, BabA, HopQ) have been characterized in terms of their three-dimensional structure. A recent addition to the growing list of outer membrane porins is LabA (LacdiNAc-binding adhesin), which is thought to bind specifically to GalNAcß1-4GlcNAc, occurring in the gastric mucosa. LabA47-496 protein expressed as His-tagged protein in the periplasm of E. coli and purified via subtractive IMAC after TEV cleavage and subsequent size exclusion chromatography, resulted in bipyramidal crystals with good diffraction properties. Here, we describe the 2.06 â€‹Å resolution structure of the exodomain of LabA from H. pylori strain J99 (PDB ID: 6GMM). Strikingly, despite the relatively low levels of sequence identity with the other three structurally characterized adhesins (20-49%), LabA shares an L-shaped fold with SabA and BabA. The 'head' region contains a 4 â€‹+ â€‹3 α-helix bundle, with a small insertion domain consisting of a short antiparallel beta sheet and an unstructured region, not resolved in the crystal structure. Sequence alignment of LabA from different strains shows a high level of conservation in the N- and C-termini, and identifies two main types based on the length of the insertion domain ('crown' region), the 'J99-type' (insertion ~31 â€‹amino acids), and the H. pylori '26695 type' (insertion ~46 â€‹amino acids). Analysis of ligand binding using Native Electrospray Ionization Mass Spectrometry (ESI-MS) together with solid phase-bound, ELISA-type assays could not confirm the originally described binding of GalNAcß1-4GlcNAc-containing oligosaccharides, in line with other recent reports, which also failed to confirm LacdiNAc binding.

11.
Int J Mol Sci ; 22(10)2021 May 12.
Article de Anglais | MEDLINE | ID: mdl-34066057

RÉSUMÉ

Poly (ADP-ribose) polymerases (PARP) 1-3 are well-known multi-domain enzymes, catalysing the covalent modification of proteins, DNA, and themselves. They attach mono- or poly-ADP-ribose to targets using NAD+ as a substrate. Poly-ADP-ribosylation (PARylation) is central to the important functions of PARP enzymes in the DNA damage response and nucleosome remodelling. Activation of PARP happens through DNA binding via zinc fingers and/or the WGR domain. Modulation of their activity using PARP inhibitors occupying the NAD+ binding site has proven successful in cancer therapies. For decades, studies set out to elucidate their full-length molecular structure and activation mechanism. In the last five years, significant advances have progressed the structural and functional understanding of PARP1-3, such as understanding allosteric activation via inter-domain contacts, how PARP senses damaged DNA in the crowded nucleus, and the complementary role of histone PARylation factor 1 in modulating the active site of PARP. Here, we review these advances together with the versatility of PARP domains involved in DNA binding, the targets and shape of PARylation and the role of PARPs in nucleosome remodelling.


Sujet(s)
Protéines du cycle cellulaire/composition chimique , Nucléosomes/métabolisme , Poly (ADP-Ribose) polymerase-1/composition chimique , Poly(ADP-ribose) polymerases/composition chimique , Régulation allostérique/effets des médicaments et des substances chimiques , Protéines de transport/métabolisme , Protéines du cycle cellulaire/métabolisme , Réparation de l'ADN , Humains , Modèles moléculaires , Protéines nucléaires/métabolisme , Poly (ADP-Ribose) polymerase-1/métabolisme , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Poly(ADP-ribose) polymerases/métabolisme , Domaines protéiques/effets des médicaments et des substances chimiques
12.
J Med Chem ; 64(10): 6814-6826, 2021 05 27.
Article de Anglais | MEDLINE | ID: mdl-33900758

RÉSUMÉ

MAT2a is a methionine adenosyltransferase that synthesizes the essential metabolite S-adenosylmethionine (SAM) from methionine and ATP. Tumors bearing the co-deletion of p16 and MTAP genes have been shown to be sensitive to MAT2a inhibition, making it an attractive target for treatment of MTAP-deleted cancers. A fragment-based lead generation campaign identified weak but efficient hits binding in a known allosteric site. By use of structure-guided design and systematic SAR exploration, the hits were elaborated through a merging and growing strategy into an arylquinazolinone series of potent MAT2a inhibitors. The selected in vivo tool compound 28 reduced SAM-dependent methylation events in cells and inhibited proliferation of MTAP-null cells in vitro. In vivo studies showed that 28 was able to induce antitumor response in an MTAP knockout HCT116 xenograft model.


Sujet(s)
Conception de médicament , Antienzymes/composition chimique , Methionine adenosyltransferase/antagonistes et inhibiteurs , Site allostérique , Animaux , Prolifération cellulaire , Antienzymes/métabolisme , Antienzymes/pharmacologie , Antienzymes/usage thérapeutique , Techniques de knock-out de gènes , Cellules HCT116 , Période , Humains , Methionine adenosyltransferase/génétique , Methionine adenosyltransferase/métabolisme , Souris , Simulation de dynamique moléculaire , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Quinazolines/composition chimique , Quinazolines/métabolisme , Quinazolines/pharmacologie , Quinazolines/usage thérapeutique , Rats , Adémétionine/métabolisme , Relation structure-activité , Transplantation hétérologue
13.
J Med Chem ; 64(6): 3165-3184, 2021 03 25.
Article de Anglais | MEDLINE | ID: mdl-33683117

RÉSUMÉ

Mer is a member of the TAM (Tyro3, Axl, Mer) kinase family that has been associated with cancer progression, metastasis, and drug resistance. Their essential function in immune homeostasis has prompted an interest in their role as modulators of antitumor immune response in the tumor microenvironment. Here we illustrate the outcomes of an extensive lead-generation campaign for identification of Mer inhibitors, focusing on the results from concurrent, orthogonal high-throughput screening approaches. Data mining, HT (high-throughput), and DECL (DNA-encoded chemical library) screens offered means to evaluate large numbers of compounds. We discuss campaign strategy and screening outcomes, and exemplify series resulting from prioritization of hits that were identified. Concurrent execution of HT and DECL screening successfully yielded a large number of potent, selective, and novel starting points, covering a range of selectivity profiles across the TAM family members and modes of kinase binding, and offered excellent start points for lead development.


Sujet(s)
Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/pharmacologie , c-Mer Tyrosine kinase/antagonistes et inhibiteurs , Animaux , Cristallographie aux rayons X , Fouille de données , Découverte de médicament , Humains , Modèles moléculaires , c-Mer Tyrosine kinase/composition chimique , c-Mer Tyrosine kinase/métabolisme
14.
Nucleic Acids Res ; 49(4): 2266-2288, 2021 02 26.
Article de Anglais | MEDLINE | ID: mdl-33511412

RÉSUMÉ

PARP-1 is a key early responder to DNA damage in eukaryotic cells. An allosteric mechanism links initial sensing of DNA single-strand breaks by PARP-1's F1 and F2 domains via a process of further domain assembly to activation of the catalytic domain (CAT); synthesis and attachment of poly(ADP-ribose) (PAR) chains to protein sidechains then signals for assembly of DNA repair components. A key component in transmission of the allosteric signal is the HD subdomain of CAT, which alone bridges between the assembled DNA-binding domains and the active site in the ART subdomain of CAT. Here we present a study of isolated CAT domain from human PARP-1, using NMR-based dynamics experiments to analyse WT apo-protein as well as a set of inhibitor complexes (with veliparib, olaparib, talazoparib and EB-47) and point mutants (L713F, L765A and L765F), together with new crystal structures of the free CAT domain and inhibitor complexes. Variations in both dynamics and structures amongst these species point to a model for full-length PARP-1 activation where first DNA binding and then substrate interaction successively destabilise the folded structure of the HD subdomain to the point where its steric blockade of the active site is released and PAR synthesis can proceed.


Sujet(s)
Poly (ADP-Ribose) polymerase-1/composition chimique , Régulation allostérique , Amides/composition chimique , Domaine catalytique , Cristallographie aux rayons X , Altération de l'ADN , Activation enzymatique , Modèles moléculaires , Mutation , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/génétique , Poly (ADP-Ribose) polymerase-1/métabolisme , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Domaines protéiques
15.
Biochem J ; 477(22): 4443-4452, 2020 11 27.
Article de Anglais | MEDLINE | ID: mdl-33119085

RÉSUMÉ

The activation loop (A-loop) plays a key role in regulating the catalytic activity of protein kinases. Phosphorylation in this region enhances the phosphoryl transfer rate of the kinase domain and increases its affinity for ATP. Furthermore, the A-loop possesses autoinhibitory functions in some kinases, where it collapses onto the protein surface and blocks substrate binding when unphosphorylated. Due to its flexible nature, the A-loop is usually disordered and untraceable in kinase domain crystal structures. The resulting lack of structural information is regrettable as it impedes the design of drug A-loop contacts, which have proven favourable in multiple cases. Here, we characterize the binding with A-loop engagement between type 1.5 kinase inhibitor 'example 172' (EX172) and Mer tyrosine kinase (MerTK). With the help of crystal structures and binding kinetics, we portray how the recruitment of the A-loop elicits a two-step binding mechanism which results in a drug-target complex characterized by high affinity and long residence time. In addition, the type 1.5 compound possesses excellent kinome selectivity and a remarkable preference for the phosphorylated over the dephosphorylated form of MerTK. We discuss these unique characteristics in the context of known type 1 and type 2 inhibitors and highlight opportunities for future kinase inhibitor design.


Sujet(s)
Adénosine triphosphate/composition chimique , Inhibiteurs de protéines kinases/composition chimique , c-Mer Tyrosine kinase/antagonistes et inhibiteurs , c-Mer Tyrosine kinase/composition chimique , Humains , Structure secondaire des protéines
16.
J Am Chem Soc ; 142(23): 10358-10372, 2020 06 10.
Article de Anglais | MEDLINE | ID: mdl-32412754

RÉSUMÉ

With a resurgence in interest in covalent drugs, there is a need to identify new moieties capable of cysteine bond formation that are differentiated from commonly employed systems such as acrylamide. Herein, we report on the discovery of new alkynyl benzoxazine and dihydroquinazoline moieties capable of covalent reaction with cysteine. Their utility as alternative electrophilic warheads for chemical biological probes and drug molecules is demonstrated through site-selective protein modification and incorporation into kinase drug scaffolds. A potent covalent inhibitor of JAK3 kinase was identified with superior selectivity across the kinome and improvements in in vitro pharmacokinetic profile relative to the related acrylamide-based inhibitor. In addition, the use of a novel heterocycle as a cysteine reactive warhead is employed to target Cys788 in c-KIT, where acrylamide has previously failed to form covalent interactions. These new reactive and selective heterocyclic warheads supplement the current repertoire for cysteine covalent modification while avoiding some of the limitations generally associated with established moieties.


Sujet(s)
Benzoxazines/pharmacologie , Janus kinase 3/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/pharmacologie , Quinazolines/pharmacologie , Benzoxazines/synthèse chimique , Benzoxazines/composition chimique , Humains , Janus kinase 3/métabolisme , Modèles moléculaires , Structure moléculaire , Inhibiteurs de protéines kinases/synthèse chimique , Inhibiteurs de protéines kinases/composition chimique , Quinazolines/synthèse chimique , Quinazolines/composition chimique
17.
Mod Pathol ; 33(4): 518-530, 2020 04.
Article de Anglais | MEDLINE | ID: mdl-31558782

RÉSUMÉ

Programmed cell death ligand-1 (PD-L1) expression levels in patient tumor samples have proven clinical utility across various cancer types. Several independently developed PD-L1 immunohistochemical (IHC) predictive assays are commercially available. Published studies using the VENTANA PD-L1 (SP263) Assay, VENTANA PD-L1 (SP142) Assay, Dako PD-L1 IHC 22C3 pharmDx assay, Dako PD-L1 IHC 28-8 pharmDx assay, and laboratory-developed tests utilizing the E1L3N antibody (Cell Signaling Technology), have demonstrated differing levels of PD-L1 staining between assays, resulting in conjecture as to whether antibody-binding epitopes could be responsible for discordance between assays. Therefore, to understand the performance of different PD-L1 predictive immunohistochemistry assays, we aimed to distinguish the epitopes within the PD-L1 protein responsible for antibody binding. The sites at which antibody clones SP263, SP142, 22C3, 28-8, and E1L3N bind to recombinant PD-L1 were assessed using several methods, including conformational peptide array, surface plasmon resonance, and/or hydrogen/deuterium exchange mass spectrometry. Putative binding sites were confirmed by site-directed mutagenesis of PD-L1, followed by western blotting and immunohistochemical analysis of cell lines expressing mutant constructs. Our results demonstrate that clones SP263 and SP142 bind to an identical epitope in the cytoplasmic domain at the extreme C-terminus of PD-L1, distinct from 22C3 and 28-8. Using mutated PD-L1 constructs, an additional clone, E1L3N, was also found to bind to the cytoplasmic domain of PD-L1. The E1L3N binding epitope overlaps considerably with the SP263/SP142 binding site but is not identical. Clones 22C3 and 28-8 have binding profiles in the extracellular domain of PD-L1, which differ from one another. Despite identifying epitope binding variance among antibodies, evidence indicates that only the SP142 assay generates significantly discordant immunohistochemical staining, which can be resolved by altering the assay protocol. Therefore, inter-assay discordances are more likely attributable to tumor heterogeneity, assay, or platform variables rather than antibody epitope.


Sujet(s)
Anticorps/immunologie , Spécificité des anticorps , Antigène CD274/immunologie , Sites de fixation des anticorps , Cartographie épitopique , Immunohistochimie , Tumeurs/immunologie , Anticorps/métabolisme , Antinéoplasiques immunologiques/usage thérapeutique , Antigène CD274/génétique , Antigène CD274/métabolisme , Glycosylation , Humains , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Mutation , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Valeur prédictive des tests , Liaison aux protéines , Reproductibilité des résultats
18.
J Med Chem ; 63(7): 3461-3471, 2020 04 09.
Article de Anglais | MEDLINE | ID: mdl-31851518

RÉSUMÉ

DNA-PK is a key component within the DNA damage response, as it is responsible for recognizing and repairing double-strand DNA breaks (DSBs) via non-homologous end joining. Historically it has been challenging to identify inhibitors of the DNA-PK catalytic subunit (DNA-PKcs) with good selectivity versus the structurally related PI3 (lipid) and PI3K-related protein kinases. We screened our corporate collection for DNA-PKcs inhibitors with good PI3 kinase selectivity, identifying compound 1. Optimization focused on further improving selectivity while improving physical and pharmacokinetic properties, notably co-optimization of permeability and metabolic stability, to identify compound 16 (AZD7648). Compound 16 had no significant off-target activity in the protein kinome and only weak activity versus PI3Kα/γ lipid kinases. Monotherapy activity in murine xenograft models was observed, and regressions were observed when combined with inducers of DSBs (doxorubicin or irradiation) or PARP inhibition (olaparib). These data support progression into clinical studies (NCT03907969).


Sujet(s)
DNA-activated protein kinase/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/usage thérapeutique , Purines/usage thérapeutique , Pyrannes/usage thérapeutique , Triazoles/usage thérapeutique , Animaux , Antinéoplasiques/synthèse chimique , Antinéoplasiques/pharmacocinétique , Antinéoplasiques/usage thérapeutique , Lignée cellulaire tumorale , Phosphatidylinositol 3-kinases de classe Ib/métabolisme , Chiens , Découverte de médicament , Humains , Souris , Structure moléculaire , Tumeurs/traitement médicamenteux , Inhibiteurs de protéines kinases/synthèse chimique , Inhibiteurs de protéines kinases/pharmacocinétique , Purines/synthèse chimique , Purines/pharmacocinétique , Pyrannes/synthèse chimique , Pyrannes/pharmacocinétique , Rats , Récepteur de facteur de croissance granulocyte-macrophage/antagonistes et inhibiteurs , Relation structure-activité , Triazoles/synthèse chimique , Triazoles/pharmacocinétique , Tests d'activité antitumorale sur modèle de xénogreffe
20.
Open Biol ; 9(11): 190192, 2019 11 29.
Article de Anglais | MEDLINE | ID: mdl-31771416

RÉSUMÉ

O-GlcNAcylation is an abundant post-translational modification in the nervous system, linked to both neurodevelopmental and neurodegenerative disease. However, the mechanistic links between these phenotypes and site-specific O-GlcNAcylation remain largely unexplored. Here, we show that Ser517 O-GlcNAcylation of the microtubule-binding protein Collapsin Response Mediator Protein-2 (CRMP2) increases with age. By generating and characterizing a Crmp2S517A knock-in mouse model, we demonstrate that loss of O-GlcNAcylation leads to a small decrease in body weight and mild memory impairment, suggesting that Ser517 O-GlcNAcylation has a small but detectable impact on mouse physiology and cognitive function.


Sujet(s)
Acétyl-glucosamine/métabolisme , Cognition , Protéines et peptides de signalisation intercellulaire/métabolisme , Mémoire à court terme , Protéines de tissu nerveux/métabolisme , Acétyl-glucosamine/analyse , Vieillissement , Séquence d'acides aminés , Animaux , Lignée cellulaire , Comportement d'exploration , Femelle , Techniques de knock-in de gènes , Humains , Protéines et peptides de signalisation intercellulaire/composition chimique , Protéines et peptides de signalisation intercellulaire/génétique , Mâle , Troubles de la mémoire/génétique , Troubles de la mémoire/métabolisme , Souris , Souris de lignée C57BL , Protéines de tissu nerveux/composition chimique , Protéines de tissu nerveux/génétique , Mutation ponctuelle , Maturation post-traductionnelle des protéines
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...