Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 60
Filtrer
1.
Chem Sci ; 14(31): 8288-8294, 2023 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-37564419

RÉSUMÉ

Optimisation of the affinity of lead compounds is a critical challenge in the identification of drug candidates and chemical probes and is a process that takes many years. Fragment-based drug discovery has become established as one of the methods of choice for drug discovery starting with small, low affinity compounds. Due to their low affinity, the evolution of fragments to desirable levels of affinity is often a key challenge. The accepted best method for increasing the potency of fragments is by iterative fragment growing, which can be very time consuming and complex. Here, we introduce a paradigm for fragment hit optimisation using poised DNA-encoded chemical libraries (DELs). The synthesis of a poised DEL, a partially constructed library that retains a reactive handle, allows the coupling of any active fragment for a specific target protein, allowing rapid discovery of potent ligands. This is illustrated for bromodomain-containing protein 4 (BRD4), in which a weakly binding fragment was coupled to a 42-member poised DEL via Suzuki-Miyaura cross coupling resulting in the identification of an inhibitor with 51 nM affinity in a single step, representing an increase in potency of several orders of magnitude from an original fragment. The potency of the compound was shown to arise from the synergistic combination of substructures, which would have been very difficult to discover by any other method and was rationalised by X-ray crystallography. The compound showed attractive lead-like properties suitable for further optimisation and demonstrated BRD4-dependent cellular pharmacology. This work demonstrates the power of poised DELs to rapidly optimise fragments, representing an attractive generic approach to drug discovery.

2.
J Med Chem ; 66(17): 12324-12341, 2023 09 14.
Article de Anglais | MEDLINE | ID: mdl-37647129

RÉSUMÉ

A major drawback of cytotoxic chemotherapy is the lack of selectivity toward noncancerous cells. The targeted delivery of cytotoxic drugs to tumor cells is a longstanding goal in cancer research. We proposed that covalent inhibitors could be adapted to deliver cytotoxic agents, conjugated to the ß-position of the Michael acceptor, via an addition-elimination mechanism promoted by covalent binding. Studies on model systems showed that conjugated 5-fluorouracil (5FU) could be released upon thiol addition in relevant time scales. A series of covalent epidermal growth factor receptor (EGFR) inhibitors were synthesized as their 5FU derivatives. Achieving the desired release of 5FU was demonstrated to depend on the electronics and geometry of the compounds. Mass spectrometry and NMR studies demonstrated an anilinoquinazoline acrylate ester conjugate bound to EGFR with the release of 5FU. This work establishes that acrylates can be used to release conjugated molecules upon covalent binding to proteins and could be used to develop targeted therapeutics.


Sujet(s)
Cytotoxines , Fluorouracil , Fluorouracil/pharmacologie , Récepteurs ErbB , Esters , Spectrométrie de masse
3.
J Med Chem ; 66(7): 4491-4502, 2023 04 13.
Article de Anglais | MEDLINE | ID: mdl-37002872

RÉSUMÉ

The extracellular signal-regulated kinase 5 (ERK5) signaling pathway is one of four conventional mitogen-activated protein (MAP) kinase pathways. Genetic perturbation of ERK5 has suggested that modulation of ERK5 activity may have therapeutic potential in cancer chemotherapy. This Miniperspective examines the evidence for ERK5 as a drug target in cancer, the structure of ERK5, and the evolution of structurally distinct chemotypes of ERK5 kinase domain inhibitors. The emerging complexities of ERK5 pharmacology are discussed, including the confounding phenomenon of paradoxical ERK5 activation by small-molecule ERK5 inhibitors. The impact of the recent development and biological evaluation of potent and selective bifunctional degraders of ERK5 and future opportunities in ERK modulation are also explored.


Sujet(s)
Système de signalisation des MAP kinases , Transduction du signal , Transduction du signal/physiologie , Phosphorylation , Mitogen-Activated Protein Kinase 7 , Maturation post-traductionnelle des protéines
4.
Clin Cancer Res ; 29(8): 1429-1439, 2023 04 14.
Article de Anglais | MEDLINE | ID: mdl-36652553

RÉSUMÉ

PURPOSE: Inhibition of monocarboxylate transporter (MCT) 1-mediated lactate transport may have cytostatic and/or cytotoxic effects on tumor cells. We report results from the dose-escalation part of a first-in-human trial of AZD3965, a first-in-class MCT1 inhibitor, in advanced cancer. PATIENTS AND METHODS: This multicentre, phase I, dose-escalation and dose-expansion trial enrolled patients with advanced solid tumors or lymphoma and no standard therapy options. Exclusion criteria included history of retinal and/or cardiac disease, due to MCT1 expression in the eye and heart. Patients received daily oral AZD3965 according to a 3+3 then rolling six design. Primary objectives were to assess safety and determine the MTD and/or recommended phase II dose (RP2D). Secondary objectives for dose escalation included measurement of pharmacokinetic and pharmacodynamic activity. Exploratory biomarkers included tumor expression of MCT1 and MCT4, functional imaging of biological impact, and metabolomics. RESULTS: During dose escalation, 40 patients received AZD3965 at 5-30 mg once daily or 10 or 15 mg twice daily. Treatment-emergent adverse events were primarily grade 1 and/or 2, most commonly electroretinogram changes (retinopathy), fatigue, anorexia, and constipation. Seven patients receiving ≥20 mg daily experienced dose-limiting toxicities (DLT): grade 3 cardiac troponin rise (n = 1), asymptomatic ocular DLTs (n = 5), and grade 3 acidosis (n = 1). Plasma pharmacokinetics demonstrated attainment of target concentrations; pharmacodynamic measurements indicated on-target activity. CONCLUSIONS: AZD3965 is tolerated at doses that produce target engagement. DLTs were on-target and primarily dose-dependent, asymptomatic, reversible ocular changes. An RP2D of 10 mg twice daily was established for use in dose expansion in cancers that generally express high MCT1/low MCT4).


Sujet(s)
Antinéoplasiques , Tumeurs , Humains , Tumeurs/traitement médicamenteux , Tumeurs/induit chimiquement , Pyrimidinones/pharmacologie , Antinéoplasiques/effets indésirables , Thiophènes/pharmacologie , Dose maximale tolérée , Relation dose-effet des médicaments
5.
Br J Cancer ; 127(5): 937-947, 2022 09.
Article de Anglais | MEDLINE | ID: mdl-35618788

RÉSUMÉ

BACKGROUND: We evaluated the therapeutic potential of combining the monocarboxylate transporter 1 (MCT1) inhibitor AZD3965 with the mitochondrial respiratory Complex I inhibitor IACS-010759, for the treatment of diffuse large B-cell lymphoma (DLBCL), a potential clinically actionable strategy to target tumour metabolism. METHODS: AZD3965 and IACS-010759 sensitivity were determined in DLBCL cell lines and tumour xenograft models. Lactate concentrations, oxygen consumption rate and metabolomics were examined as mechanistic endpoints. In vivo plasma concentrations of IACS-010759 in mice were determined by LC-MS to select a dose that reflected clinically attainable concentrations. RESULTS: In vitro, the combination of AZD3965 and IACS-010759 is synergistic and induces DLBCL cell death, whereas monotherapy treatments induce a cytostatic response. Significant anti-tumour activity was evident in Toledo and Farage models when the two inhibitors were administered concurrently despite limited or no effect on the growth of DLBCL xenografts as monotherapies. CONCLUSIONS: This is the first study to examine a combination of two distinct approaches to targeting tumour metabolism in DLBCL xenografts. Whilst nanomolar concentrations of either AZD3965 or IACS-010759 monotherapy demonstrate anti-proliferative activity against DLBCL cell lines in vitro, appreciable clinical activity in DLBCL patients may only be realised through their combined use.


Sujet(s)
Lymphome B diffus à grandes cellules , Symporteurs , Animaux , Apoptose , Lignée cellulaire tumorale , Glycolyse , Humains , Lymphome B diffus à grandes cellules/anatomopathologie , Souris , Transporteurs d'acides monocarboxyliques , Phosphorylation oxydative , Symporteurs/métabolisme
6.
J Med Chem ; 65(9): 6513-6540, 2022 05 12.
Article de Anglais | MEDLINE | ID: mdl-35468293

RÉSUMÉ

The nonclassical extracellular signal-related kinase 5 (ERK5) mitogen-activated protein kinase pathway has been implicated in increased cellular proliferation, migration, survival, and angiogenesis; hence, ERK5 inhibition may be an attractive approach for cancer treatment. However, the development of selective ERK5 inhibitors has been challenging. Previously, we described the development of a pyrrole carboxamide high-throughput screening hit into a selective, submicromolar inhibitor of ERK5 kinase activity. Improvement in the ERK5 potency was necessary for the identification of a tool ERK5 inhibitor for target validation studies. Herein, we describe the optimization of this series to identify nanomolar pyrrole carboxamide inhibitors of ERK5 incorporating a basic center, which suffered from poor oral bioavailability. Parallel optimization of potency and in vitro pharmacokinetic parameters led to the identification of a nonbasic pyrazole analogue with an optimal balance of ERK5 inhibition and oral exposure.


Sujet(s)
Mitogen-Activated Protein Kinase 7 , Pyrroles , Prolifération cellulaire , Pyrroles/pharmacologie
7.
J Med Chem ; 64(14): 10001-10018, 2021 07 22.
Article de Anglais | MEDLINE | ID: mdl-34212719

RÉSUMÉ

NF-κB-inducing kinase (NIK) is a key enzyme in the noncanonical NF-κB pathway, of interest in the treatment of a variety of diseases including cancer. Validation of NIK as a drug target requires potent and selective inhibitors. The protein contains a cysteine residue at position 444 in the back pocket of the active site, unique within the kinome. Analysis of existing inhibitor scaffolds and early structure-activity relationships (SARs) led to the design of C444-targeting covalent inhibitors based on alkynyl heterocycle warheads. Mass spectrometry provided proof of the covalent mechanism, and the SAR was rationalized by computational modeling. Profiling of more potent analogues in tumor cell lines with constitutively activated NIK signaling induced a weak antiproliferative effect, suggesting that kinase inhibition may have limited impact on cancer cell growth. This study shows that alkynyl heterocycles are potential cysteine traps, which may be employed where common Michael acceptors, such as acrylamides, are not tolerated.


Sujet(s)
Alcynes/pharmacologie , Cystéine/pharmacologie , Inhibiteurs de protéines kinases/pharmacologie , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Pyrimidines/pharmacologie , Alcynes/synthèse chimique , Alcynes/composition chimique , Cystéine/composition chimique , Relation dose-effet des médicaments , Humains , Structure moléculaire , Inhibiteurs de protéines kinases/synthèse chimique , Inhibiteurs de protéines kinases/composition chimique , Protein-Serine-Threonine Kinases/métabolisme , Pyrimidines/synthèse chimique , Pyrimidines/composition chimique , Relation structure-activité ,
8.
Mol Cancer Ther ; 20(9): 1663-1671, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34158348

RÉSUMÉ

Radiation-induced DNA double-strand breaks (DSBs) can be repaired by homologous recombination (HR) and nonhomologous end joining (NHEJ). Recently, it has been found that chronic tumor hypoxia compromises HR repair of DNA DSBs but activates the NHEJ protein DNAPK. We therefore hypothesized that inhibition of DNAPK can preferentially potentiate the sensitivity of chronically hypoxic cancer cells to radiation through contextual synthetic lethality in vivo In this study, we investigated the impact of DNAPK inhibition by a novel selective DNAPK inhibitor, NU5455, on the repair of radiation-induced DNA DSBs in chronically hypoxic and nonhypoxic cells across a range of xenograft models. We found that NU5455 inhibited DSB repair following radiation in both chronically hypoxic and nonhypoxic tumor cells. Most importantly, the inhibitory effect was more pronounced in chronically hypoxic tumor cells than in nonhypoxic tumor cells. This is the first in vivo study to indicate that DNAPK inhibition may preferentially sensitize chronically hypoxic tumor cells to radiotherapy, suggesting a broader therapeutic window for transient DNAPK inhibition combined with radiotherapy.


Sujet(s)
Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Cassures double-brin de l'ADN , Réparation de l'ADN , DNA-activated protein kinase/antagonistes et inhibiteurs , Hypoxie/physiopathologie , Tumeurs du poumon/traitement médicamenteux , Rayonnement ionisant , Animaux , Apoptose , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Prolifération cellulaire , Femelle , Recombinaison homologue , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Souris , Souris de lignée BALB C , Souris nude , Inhibiteurs de protéines kinases/pharmacologie , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
9.
J Med Chem ; 64(7): 4071-4088, 2021 04 08.
Article de Anglais | MEDLINE | ID: mdl-33761253

RÉSUMÉ

Inhibition of murine double minute 2 (MDM2)-p53 protein-protein interaction with small molecules has been shown to reactivate p53 and inhibit tumor growth. Here, we describe rational, structure-guided, design of novel isoindolinone-based MDM2 inhibitors. MDM2 X-ray crystallography, quantum mechanics ligand-based design, and metabolite identification all contributed toward the discovery of potent in vitro and in vivo inhibitors of the MDM2-p53 interaction with representative compounds inducing cytostasis in an SJSA-1 osteosarcoma xenograft model following once-daily oral administration.


Sujet(s)
Antinéoplasiques/pharmacologie , Isoindoles/pharmacologie , Ostéosarcome/traitement médicamenteux , Multimérisation de protéines/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-mdm2/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Animaux , Antinéoplasiques/synthèse chimique , Antinéoplasiques/métabolisme , Tumeurs osseuses/traitement médicamenteux , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cristallographie aux rayons X , Stabilité de médicament , Femelle , Humains , Isoindoles/synthèse chimique , Isoindoles/métabolisme , Macaca fascicularis , Mâle , Souris de lignée BALB C , Souris nude , Microsomes du foie/métabolisme , Structure moléculaire , Liaison aux protéines , Relation structure-activité , Tests d'activité antitumorale sur modèle de xénogreffe
10.
J Clin Invest ; 130(1): 258-271, 2020 01 02.
Article de Anglais | MEDLINE | ID: mdl-31581151

RÉSUMÉ

Potentiating radiotherapy and chemotherapy by inhibiting DNA damage repair is proposed as a therapeutic strategy to improve outcomes for patients with solid tumors. However, this approach risks enhancing normal tissue toxicity as much as tumor toxicity, thereby limiting its translational impact. Using NU5455, a newly identified highly selective oral inhibitor of DNA-dependent protein kinase catalytic subunit (DNA-PKcs) activity, we found that it was indeed possible to preferentially augment the effect of targeted radiotherapy on human orthotopic lung tumors without influencing acute DNA damage or a late radiation-induced toxicity (fibrosis) to normal mouse lung. Furthermore, while NU5455 administration increased both the efficacy and the toxicity of a parenterally administered topoisomerase inhibitor, it enhanced the activity of doxorubicin released locally in liver tumor xenografts without inducing any adverse effect. This strategy is particularly relevant to hepatocellular cancer, which is treated clinically with localized drug-eluting beads and for which DNA-PKcs activity is reported to confer resistance to treatment. We conclude that transient pharmacological inhibition of DNA-PKcs activity is effective and tolerable when combined with localized DNA-damaging therapies and thus has promising clinical potential.


Sujet(s)
Carcinome hépatocellulaire , DNA-activated protein kinase/antagonistes et inhibiteurs , Tumeurs expérimentales du foie , Protéines tumorales/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases , Animaux , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/enzymologie , Carcinome hépatocellulaire/anatomopathologie , DNA-activated protein kinase/métabolisme , Doxorubicine/pharmacologie , Humains , Tumeurs expérimentales du foie/traitement médicamenteux , Tumeurs expérimentales du foie/enzymologie , Tumeurs expérimentales du foie/anatomopathologie , Cellules MCF-7 , Souris , Protéines tumorales/métabolisme , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe
11.
Eur J Med Chem ; 178: 530-543, 2019 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-31212132

RÉSUMÉ

Extracellular regulated kinase 5 (ERK5) signalling has been implicated in driving a number of cellular phenotypes including endothelial cell angiogenesis and tumour cell motility. Novel ERK5 inhibitors were identified using high throughput screening, with a series of pyrrole-2-carboxamides substituted at the 4-position with an aroyl group being found to exhibit IC50 values in the micromolar range, but having no selectivity against p38α MAP kinase. Truncation of the N-substituent marginally enhanced potency (∼3-fold) against ERK5, but importantly attenuated inhibition of p38α. Systematic variation of the substituents on the aroyl group led to the selective inhibitor 4-(2-bromo-6-fluorobenzoyl)-N-(pyridin-3-yl)-1H-pyrrole-2-carboxamide (IC50 0.82 µM for ERK5; IC50 > 120 µM for p38α). The crystal structure (PDB 5O7I) of this compound in complex with ERK5 has been solved. This compound was orally bioavailable and inhibited bFGF-driven Matrigel plug angiogenesis and tumour xenograft growth. The selective ERK5 inhibitor described herein provides a lead for further development into a tool compound for more extensive studies seeking to examine the role of ERK5 signalling in cancer and other diseases.


Sujet(s)
Antinéoplasiques/pharmacologie , Mitogen-Activated Protein Kinase 14/antagonistes et inhibiteurs , Mitogen-Activated Protein Kinase 7/antagonistes et inhibiteurs , Protéines nucléaires/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/pharmacologie , Facteurs de transcription/antagonistes et inhibiteurs , Administration par voie orale , Animaux , Antinéoplasiques/administration et posologie , Antinéoplasiques/composition chimique , Biodisponibilité , Protéines du cycle cellulaire , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Tests de criblage d'agents antitumoraux , Femelle , Humains , Souris , Souris nude , Mitogen-Activated Protein Kinase 14/métabolisme , Mitogen-Activated Protein Kinase 7/métabolisme , Structure moléculaire , Tumeurs expérimentales/traitement médicamenteux , Tumeurs expérimentales/métabolisme , Tumeurs expérimentales/anatomopathologie , Protéines nucléaires/métabolisme , Inhibiteurs de protéines kinases/administration et posologie , Inhibiteurs de protéines kinases/composition chimique , Relation structure-activité , Facteurs de transcription/métabolisme
12.
Methods Appl Fluoresc ; 7(1): 015001, 2018 Oct 24.
Article de Anglais | MEDLINE | ID: mdl-30353887

RÉSUMÉ

Evasion of apoptosis is a hallmark of human cancer, and a desired endpoint of many anticancer agents is the induction of cell death. With the heterogeneity of cancer becoming increasingly apparent, to understand drug mechanisms of action and identify combination therapies in cell populations, the development of tools to assess drug effects at the single cell level is a necessity for future preclinical drug development. Herein we describe the development of pCasFSwitch, a genetically encoded reporter construct designed to identify cells undergoing caspase-3 mediated apoptosis, by a translocation of a GFP signal from the cell membrane into the nucleus. Anticipated cellular distribution was demonstrated by use of confocal microscopy and cleavage by caspase-3 was shown to be required for the translocation of the GFP signal seen in apoptotic cells. Quantification of apoptosis using the construct revealed similar levels to that obtained with a commercially available apoptosis imaging agent (22.6% versus 20.3%). Moreover, we demonstrated its capacity for use in a high-throughput setting making it a powerful tool for drug development pipelines.


Sujet(s)
Apoptose/physiologie , Caspase-3/métabolisme , Gènes rapporteurs/génétique , Protéines à fluorescence verte/métabolisme , Protéines de fusion recombinantes/métabolisme , Animaux , Séquence nucléotidique , Lignée cellulaire tumorale , Membrane cellulaire/métabolisme , Noyau de la cellule/métabolisme , Fluorescence , Protéines à fluorescence verte/génétique , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Souris , Microscopie confocale/méthodes , Microscopie de fluorescence/méthodes , Protéines nucléaires/génétique , Protéines nucléaires/métabolisme , Protéines de fusion recombinantes/génétique , Analyse sur cellule unique/méthodes
13.
Bioconjug Chem ; 29(6): 2100-2106, 2018 06 20.
Article de Anglais | MEDLINE | ID: mdl-29851469

RÉSUMÉ

MDM2 is a key negative regulator of the p53 tumor suppressor. Direct binding of MDM2 to p53 represses the protein's transcriptional activity and induces its polyubiquitination, targeting it for degradation by the proteasome. Consequently, small molecule inhibitors that antagonize MDM2-p53 binding, such as RG7388, have progressed into clinical development aiming to reactivate p53 function in TP53 wild-type tumors. Here, we describe the design, synthesis, and biological evaluation of a trans-cyclooctene tagged derivative of RG7388, RG7388-TCO, which showed high cellular potency and specificity for MDM2. The in-cell reaction of RG7388-TCO with a tetrazine-tagged BODIPY dye enabled fluorescence imaging of endogenous MDM2 in SJSA-1 and T778 tumor cells. RG7388-TCO was also used to pull down MDM2 by reaction with tetrazine-tagged agarose beads in SJSA-1 lysates. The data presented show that RG733-TCO enables precise imaging of MDM2 in cells and can permit a relative assessment of target engagement and MDM2-p53 antagonism in vitro.


Sujet(s)
Composés du bore/composition chimique , Colorants fluorescents/composition chimique , Protéines proto-oncogènes c-mdm2/analyse , Protéines proto-oncogènes c-mdm2/métabolisme , Pyrrolidines/composition chimique , Protéine p53 suppresseur de tumeur/métabolisme , para-Aminobenzoates/composition chimique , Lignée cellulaire tumorale , Chimie click , Cyclooctanes/analogues et dérivés , Cyclooctanes/pharmacologie , Humains , Simulation de docking moléculaire , Imagerie optique/méthodes , Liaison aux protéines/effets des médicaments et des substances chimiques , Cartes d'interactions protéiques/effets des médicaments et des substances chimiques , Pyrrolidines/pharmacologie , para-Aminobenzoates/pharmacologie
14.
Org Biomol Chem ; 16(11): 1843-1850, 2018 03 14.
Article de Anglais | MEDLINE | ID: mdl-29469144

RÉSUMÉ

ATAD2 is an ATPase that is overexpressed in a variety of cancers and associated with a poor patient prognosis. This protein has been suggested to function as a cofactor for a range of transcription factors, including the proto-oncogene MYC and the androgen receptor. ATAD2 comprises an ATPase domain, implicated in chromatin remodelling, and a bromodomain which allows it to interact with acetylated histone tails. Dissection of the functional roles of these two domains would benefit from the availability of selective, cell-permeable pharmacological probes. An in silico evaluation of the 3D structures of various bromodomains suggested that developing small molecule ligands for the bromodomain of ATAD2 is likely to be challenging, although recent reports have shown that ATAD2 bromodomain ligands can be identified. We report a structure-guided fragment-based approach to identify lead compounds for ATAD2 bromodomain inhibitor development. Our findings indicate that the ATAD2 bromodomain can accommodate fragment hits (Mr < 200) that yield productive structure-activity relationships, and structure-guided design enabled the introduction of selectivity over BRD4.


Sujet(s)
ATPases associated with diverse cellular activities/antagonistes et inhibiteurs , ATPases associated with diverse cellular activities/métabolisme , Protéines de liaison à l'ADN/antagonistes et inhibiteurs , Protéines de liaison à l'ADN/métabolisme , Conception de médicament , Protéines nucléaires/métabolisme , Bibliothèques de petites molécules/composition chimique , Bibliothèques de petites molécules/pharmacologie , Facteurs de transcription/métabolisme , ATPases associated with diverse cellular activities/composition chimique , Protéines du cycle cellulaire , Conception assistée par ordinateur , Protéines de liaison à l'ADN/composition chimique , Humains , Ligands , Simulation de docking moléculaire , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Protéines nucléaires/composition chimique , Liaison aux protéines , Domaines protéiques/effets des médicaments et des substances chimiques , Proto-oncogène Mas , Facteurs de transcription/composition chimique
15.
Cell Rep ; 21(5): 1386-1398, 2017 Oct 31.
Article de Anglais | MEDLINE | ID: mdl-29091774

RÉSUMÉ

Selective recruitment of protein kinases to the Hsp90 system is mediated by the adaptor co-chaperone Cdc37. We show that assembly of CDK4 and CDK6 into protein complexes is differentially regulated by the Cdc37-Hsp90 system. Like other Hsp90 kinase clients, binding of CDK4/6 to Cdc37 is blocked by ATP-competitive inhibitors. Cdc37-Hsp90 relinquishes CDK6 to D3- and virus-type cyclins and to INK family CDK inhibitors, whereas CDK4 is relinquished to INKs but less readily to cyclins. p21CIP1 and p27KIP1 CDK inhibitors are less potent than the INKs at displacing CDK4 and CDK6 from Cdc37. However, they cooperate with the D-type cyclins to generate CDK4/6-containing ternary complexes that are resistant to cyclin D displacement by Cdc37, suggesting a molecular mechanism to explain the assembly factor activity ascribed to CIP/KIP family members. Overall, our data reveal multiple mechanisms whereby the Hsp90 system may control formation of CDK4- and CDK6-cyclin complexes under different cellular conditions.


Sujet(s)
Protéines du cycle cellulaire/métabolisme , Chaperonines/métabolisme , Kinase-4 cycline-dépendante/métabolisme , Kinase-6 cycline-dépendante/métabolisme , Protéines du choc thermique HSP90/métabolisme , Adénosine triphosphate/composition chimique , Adénosine triphosphate/métabolisme , Aminopyridines/composition chimique , Aminopyridines/métabolisme , Benzimidazoles/métabolisme , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Protéines du cycle cellulaire/génétique , Chaperonines/antagonistes et inhibiteurs , Chaperonines/génétique , Cycline D/métabolisme , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Inhibiteur p16 de kinase cycline-dépendante/métabolisme , Inhibiteur p27 de kinase cycline-dépendante/métabolisme , Transfert d'énergie par résonance de fluorescence , Protéines du choc thermique HSP90/génétique , Humains , Concentration inhibitrice 50 , Cinétique , Pipérazines/composition chimique , Pipérazines/métabolisme , Liaison aux protéines , Purines/composition chimique , Purines/métabolisme , Pyridines/composition chimique , Pyridines/métabolisme , Résonance plasmonique de surface
16.
Sci Rep ; 7(1): 12020, 2017 09 20.
Article de Anglais | MEDLINE | ID: mdl-28931905

RÉSUMÉ

Although an array of new therapeutics has emerged for the treatment of colorectal cancer, their use is significantly impacted by variability in patient response. Better pre-clinical models could substantially improve efficacy as it may allow stratification of patients into the correct treatment regime. Here we explore acute, ex vivo treatment of fresh, surgically resected human colorectal tumour biopsies as a novel pre-clinical model for identifying patient response to specific therapeutics. The MEK1/2 inhibitor, Selumetinib (AZD6244, ARRY-142886) was used as a tool compound. Firstly, we established an acute treatment protocol and demonstrated this protocol could differentiate phenotypic and pharmacodynamic responses to Selumetinib (0-3uM). We then used the protocol to evaluate Selumetinib response in tumours from 23 colon cancer patients. These studies revealed that the agent inhibited pERK1/2 phosphorylation in all tumours, caused a significant decrease in proliferation in 5/23 (22%) tumours, and that KRAS/BRAF mutant tumours were particularly sensitive to the anti-proliferative effects of the agent. These data are consistent with data from clinical trials of Selumetinib, suggesting that acute treatment of small tumour biopsies is worthy of further exploration as a pre-clinical model to evaluate colorectal cancer response to novel therapies.


Sujet(s)
Benzimidazoles/usage thérapeutique , Côlon/effets des médicaments et des substances chimiques , Tumeurs colorectales/traitement médicamenteux , MAP Kinase Kinase 1/antagonistes et inhibiteurs , MAP Kinase Kinase 2/antagonistes et inhibiteurs , Rectum/effets des médicaments et des substances chimiques , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Benzimidazoles/pharmacologie , Biopsie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/génétique , Côlon/métabolisme , Côlon/anatomopathologie , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Femelle , Cellules HCT116 , Humains , MAP Kinase Kinase 1/métabolisme , MAP Kinase Kinase 2/métabolisme , Mâle , Adulte d'âge moyen , Mutation , Rectum/métabolisme , Rectum/anatomopathologie , Protéines G ras/génétique
17.
Haematologica ; 102(7): 1247-1257, 2017 07.
Article de Anglais | MEDLINE | ID: mdl-28385782

RÉSUMÉ

Inhibition of monocarboxylate transporter 1 has been proposed as a therapeutic approach to perturb lactate shuttling in tumor cells that lack monocarboxylate transporter 4. We examined the monocarboxylate transporter 1 inhibitor AZD3965, currently in phase I clinical studies, as a potential therapy for diffuse large B-cell lymphoma and Burkitt lymphoma. Whilst extensive monocarboxylate transporter 1 protein was found in 120 diffuse large B-cell lymphoma and 10 Burkitt lymphoma patients' tumors, monocarboxylate transporter 4 protein expression was undetectable in 73% of the diffuse large B-cell lymphoma samples and undetectable or negligible in each Burkitt lymphoma sample. AZD3965 treatment led to a rapid accumulation of intracellular lactate in a panel of lymphoma cell lines with low monocarboxylate transporter 4 protein expression and potently inhibited their proliferation. Metabolic changes induced by AZD3965 in lymphoma cells were consistent with a feedback inhibition of glycolysis. A profound cytostatic response was also observed in vivo: daily oral AZD3965 treatment for 24 days inhibited CA46 Burkitt lymphoma growth by 99%. Continuous exposure of CA46 cells to AZD3965 for 7 weeks in vitro resulted in a greater dependency upon oxidative phosphorylation. Combining AZD3965 with an inhibitor of mitochondrial complex I (central to oxidative phosphorylation) induced significant lymphoma cell death in vitro and reduced CA46 disease burden in vivo These data support clinical examination of AZD3965 in Burkitt lymphoma and diffuse large B-cell lymphoma patients with low tumor monocarboxylate transporter 4 expression and highlight the potential of combination strategies to optimally target the metabolic phenotype of tumors.


Sujet(s)
Antinéoplasiques/pharmacologie , Lymphome de Burkitt/métabolisme , Lymphome B diffus à grandes cellules/métabolisme , Transporteurs d'acides monocarboxyliques/antagonistes et inhibiteurs , Pyrimidinones/pharmacologie , Symporteurs/antagonistes et inhibiteurs , Thiophènes/pharmacologie , Antinéoplasiques/usage thérapeutique , Marqueurs biologiques , Lymphome de Burkitt/traitement médicamenteux , Lymphome de Burkitt/génétique , Lymphome de Burkitt/anatomopathologie , Mort cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques , Complexe I de la chaîne respiratoire/antagonistes et inhibiteurs , Métabolisme énergétique/effets des médicaments et des substances chimiques , Humains , Acide lactique/métabolisme , Lymphome B diffus à grandes cellules/traitement médicamenteux , Lymphome B diffus à grandes cellules/génétique , Lymphome B diffus à grandes cellules/anatomopathologie , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Transporteurs d'acides monocarboxyliques/génétique , Transporteurs d'acides monocarboxyliques/métabolisme , Protéines du muscle/génétique , Protéines du muscle/métabolisme , Phosphorylation oxydative/effets des médicaments et des substances chimiques , Pyrimidinones/usage thérapeutique , Symporteurs/génétique , Symporteurs/métabolisme , Thiophènes/usage thérapeutique
18.
J Med Chem ; 60(5): 1746-1767, 2017 03 09.
Article de Anglais | MEDLINE | ID: mdl-28005359

RÉSUMÉ

Purines and related heterocycles substituted at C-2 with 4'-sulfamoylanilino and at C-6 with a variety of groups have been synthesized with the aim of achieving selectivity of binding to CDK2 over CDK1. 6-Substituents that favor competitive inhibition at the ATP binding site of CDK2 were identified and typically exhibited 10-80-fold greater inhibition of CDK2 compared to CDK1. Most impressive was 4-((6-([1,1'-biphenyl]-3-yl)-9H-purin-2-yl)amino) benzenesulfonamide (73) that exhibited high potency toward CDK2 (IC50 0.044 µM) but was ∼2000-fold less active toward CDK1 (IC50 86 µM). This compound is therefore a useful tool for studies of cell cycle regulation. Crystal structures of inhibitor-kinase complexes showed that the inhibitor stabilizes a glycine-rich loop conformation that shapes the ATP ribose binding pocket and that is preferred in CDK2 but has not been observed in CDK1. This aspect of the active site may be exploited for the design of inhibitors that distinguish between CDK1 and CDK2.


Sujet(s)
Kinase-2 cycline-dépendante/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/pharmacologie , Purines/pharmacologie , Cristallographie aux rayons X , Inhibiteurs de protéines kinases/composition chimique , Analyse spectrale/méthodes , Relation structure-activité
19.
Nat Rev Clin Oncol ; 13(10): 627-42, 2016 10.
Article de Anglais | MEDLINE | ID: mdl-27245279

RÉSUMÉ

In countries with the best cancer outcomes, approximately 60% of patients receive radiotherapy as part of their treatment, which is one of the most cost-effective cancer treatments. Notably, around 40% of cancer cures include the use of radiotherapy, either as a single modality or combined with other treatments. Radiotherapy can provide enormous benefit to patients with cancer. In the past decade, significant technical advances, such as image-guided radiotherapy, intensity-modulated radiotherapy, stereotactic radiotherapy, and proton therapy enable higher doses of radiotherapy to be delivered to the tumour with significantly lower doses to normal surrounding tissues. However, apart from the combination of traditional cytotoxic chemotherapy with radiotherapy, little progress has been made in identifying and defining optimal targeted therapy and radiotherapy combinations to improve the efficacy of cancer treatment. The National Cancer Research Institute Clinical and Translational Radiotherapy Research Working Group (CTRad) formed a Joint Working Group with representatives from academia, industry, patient groups and regulatory bodies to address this lack of progress and to publish recommendations for future clinical research. Herein, we highlight the Working Group's consensus recommendations to increase the number of novel drugs being successfully registered in combination with radiotherapy to improve clinical outcomes for patients with cancer.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs/radiothérapie , Hypoxie cellulaire/effets des radiations , Essais cliniques comme sujet/méthodes , Association thérapeutique , Agrément de médicaments , Humains , Tumeurs/traitement médicamenteux , Éducation du patient comme sujet , Participation des patients , Assurance de la qualité des soins de santé , Qualité des soins de santé , Dose de rayonnement , Radiotolérance/effets des radiations , Résultat thérapeutique
20.
Cell Cycle ; 15(4): 506-18, 2016.
Article de Anglais | MEDLINE | ID: mdl-26959608

RÉSUMÉ

ERK5, encoded by MAPK7, has been proposed to play a role in cell proliferation, thus attracting interest as a cancer therapeutic target. While oncogenic RAS or BRAF cause sustained activation of the MEK1/2-ERK1/2 pathway, ERK5 is directly activated by MEK5. It has been proposed that RAS and RAF proteins can also promote ERK5 activation. Here we investigated the interplay between RAS-RAF-MEK-ERK and ERK5 signaling and studied the role of ERK5 in tumor cell proliferation in 2 disease-relevant cell models. We demonstrate that although an inducible form of CRAF (CRAF:ER*) can activate ERK5 in fibroblasts, the response is delayed and reflects feed-forward signaling. Additionally, oncogenic KRAS and BRAF do not activate ERK5 in epithelial cells. Although KRAS and BRAF do not couple directly to MEK5-ERK5, ERK5 signaling might still be permissive for proliferation. However, neither the selective MEK5 inhibitor BIX02189 or ERK5 siRNA inhibited proliferation of colorectal cancer cells harbouring KRAS(G12C/G13D) or BRAF(V600E). Furthermore, there was no additive or synergistic effect observed when BIX02189 was combined with the MEK1/2 inhibitor Selumetinib (AZD6244), suggesting that ERK5 was neither required for proliferation nor a driver of innate resistance to MEK1/2 inhibitors. Finally, even cancer cells with MAPK7 amplification were resistant to BIX02189 and ERK5 siRNA, showing that ERK5 amplification does not confer addiction to ERK5 for cell proliferation. Thus ERK5 signaling is unlikely to play a role in tumor cell proliferation downstream of KRAS or BRAF or in tumor cells with ERK5 amplification. These results have important implications for the role of ERK5 as an anti-cancer drug target.


Sujet(s)
Tumeurs colorectales/traitement médicamenteux , Mitogen-Activated Protein Kinase 7/génétique , Protéines proto-oncogènes B-raf/génétique , Protéines proto-oncogènes p21(ras)/génétique , Dérivés de l'aniline/administration et posologie , Benzimidazoles/administration et posologie , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Résistance aux médicaments antinéoplasiques/génétique , Humains , Indoles/administration et posologie , Mitogen-Activated Protein Kinase 7/métabolisme , Mutation , Inhibiteurs de protéines kinases/administration et posologie , Petit ARN interférent/génétique , Transduction du signal/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE