Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 4 de 4
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Cell Chem Biol ; 30(3): 235-247.e12, 2023 03 16.
Article de Anglais | MEDLINE | ID: mdl-36863346

RÉSUMÉ

Malignant tumors can evade destruction by the immune system by attracting immune-suppressive regulatory T cells (Treg) cells. The IKZF2 (Helios) transcription factor plays a crucial role in maintaining function and stability of Treg cells, and IKZF2 deficiency reduces tumor growth in mice. Here we report the discovery of NVP-DKY709, a selective molecular glue degrader of IKZF2 that spares IKZF1/3. We describe the recruitment-guided medicinal chemistry campaign leading to NVP-DKY709 that redirected the degradation selectivity of cereblon (CRBN) binders from IKZF1 toward IKZF2. Selectivity of NVP-DKY709 for IKZF2 was rationalized by analyzing the DDB1:CRBN:NVP-DKY709:IKZF2(ZF2 or ZF2-3) ternary complex X-ray structures. Exposure to NVP-DKY709 reduced the suppressive activity of human Treg cells and rescued cytokine production in exhausted T-effector cells. In vivo, treatment with NVP-DKY709 delayed tumor growth in mice with a humanized immune system and enhanced immunization responses in cynomolgus monkeys. NVP-DKY709 is being investigated in the clinic as an immune-enhancing agent for cancer immunotherapy.


Sujet(s)
Tumeurs , Facteurs de transcription , Animaux , Humains , Souris , Facteur de transcription Ikaros , Immunothérapie , Tumeurs/thérapie , Tumeurs/métabolisme , Lymphocytes T régulateurs/métabolisme , Facteurs de transcription/métabolisme
2.
Nature ; 535(7610): 148-52, 2016 07 07.
Article de Anglais | MEDLINE | ID: mdl-27362227

RÉSUMÉ

The non-receptor protein tyrosine phosphatase SHP2, encoded by PTPN11, has an important role in signal transduction downstream of growth factor receptor signalling and was the first reported oncogenic tyrosine phosphatase. Activating mutations of SHP2 have been associated with developmental pathologies such as Noonan syndrome and are found in multiple cancer types, including leukaemia, lung and breast cancer and neuroblastoma. SHP2 is ubiquitously expressed and regulates cell survival and proliferation primarily through activation of the RAS­ERK signalling pathway. It is also a key mediator of the programmed cell death 1 (PD-1) and B- and T-lymphocyte attenuator (BTLA) immune checkpoint pathways. Reduction of SHP2 activity suppresses tumour cell growth and is a potential target of cancer therapy. Here we report the discovery of a highly potent (IC50 = 0.071 µM), selective and orally bioavailable small-molecule SHP2 inhibitor, SHP099, that stabilizes SHP2 in an auto-inhibited conformation. SHP099 concurrently binds to the interface of the N-terminal SH2, C-terminal SH2, and protein tyrosine phosphatase domains, thus inhibiting SHP2 activity through an allosteric mechanism. SHP099 suppresses RAS­ERK signalling to inhibit the proliferation of receptor-tyrosine-kinase-driven human cancer cells in vitro and is efficacious in mouse tumour xenograft models. Together, these data demonstrate that pharmacological inhibition of SHP2 is a valid therapeutic approach for the treatment of cancers.


Sujet(s)
Tumeurs/traitement médicamenteux , Tumeurs/enzymologie , Pipéridines/pharmacologie , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonistes et inhibiteurs , Pyrimidines/pharmacologie , Récepteurs à activité tyrosine kinase/métabolisme , Régulation allostérique/effets des médicaments et des substances chimiques , Animaux , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Extracellular Signal-Regulated MAP Kinases/métabolisme , Femelle , Humains , Concentration inhibitrice 50 , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Souris , Souris nude , Modèles moléculaires , Tumeurs/anatomopathologie , Protéine oncogène p21(ras)/métabolisme , Pipéridines/composition chimique , Pipéridines/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Stabilité protéique/effets des médicaments et des substances chimiques , Structure tertiaire des protéines/effets des médicaments et des substances chimiques , Protein Tyrosine Phosphatase, Non-Receptor Type 11/composition chimique , Protein Tyrosine Phosphatase, Non-Receptor Type 11/génétique , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Pyrimidines/composition chimique , Pyrimidines/usage thérapeutique , Reproductibilité des résultats , Tests d'activité antitumorale sur modèle de xénogreffe
3.
Mol Cancer Ther ; 14(10): 2249-59, 2015 Oct.
Article de Anglais | MEDLINE | ID: mdl-26206331

RÉSUMÉ

The tumor suppressor p53 is a key regulator of apoptosis and functions upstream in the apoptotic cascade by both indirectly and directly regulating Bcl-2 family proteins. In cells expressing wild-type (WT) p53, the HDM2 protein binds to p53 and blocks its activity. Inhibition of HDM2:p53 interaction activates p53 and causes apoptosis or cell-cycle arrest. Here, we investigated the ability of the novel HDM2 inhibitor CGM097 to potently and selectively kill WT p53-expressing AML cells. The antileukemic effects of CGM097 were studied using cell-based proliferation assays (human AML cell lines, primary AML patient cells, and normal bone marrow samples), apoptosis, and cell-cycle assays, ELISA, immunoblotting, and an AML patient-derived in vivo mouse model. CGM097 potently and selectively inhibited the proliferation of human AML cell lines and the majority of primary AML cells expressing WT p53, but not mutant p53, in a target-specific manner. Several patient samples that harbored mutant p53 were comparatively unresponsive to CGM097. Synergy was observed when CGM097 was combined with FLT3 inhibition against oncogenic FLT3-expressing cells cultured both in the absence as well as the presence of cytoprotective stromal-secreted cytokines, as well as when combined with MEK inhibition in cells with activated MAPK signaling. Finally, CGM097 was effective in reducing leukemia burden in vivo. These data suggest that CGM097 is a promising treatment for AML characterized as harboring WT p53 as a single agent, as well as in combination with other therapies targeting oncogene-activated pathways that drive AML.


Sujet(s)
Antinéoplasiques/pharmacologie , Isoquinoléines/pharmacologie , Leucémie aigüe myéloïde/traitement médicamenteux , Pipérazines/pharmacologie , Protéines proto-oncogènes c-mdm2/antagonistes et inhibiteurs , Protéine p53 suppresseur de tumeur/métabolisme , Animaux , Apoptose/effets des médicaments et des substances chimiques , Benzothiazoles/pharmacologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Synergie des médicaments , Femelle , Expression des gènes , Humains , Concentration inhibitrice 50 , Souris de lignée NOD , Souris SCID , Phénylurées/pharmacologie , Staurosporine/analogues et dérivés , Staurosporine/pharmacologie , Charge tumorale/effets des médicaments et des substances chimiques , Cellules cancéreuses en culture , Protéine p53 suppresseur de tumeur/génétique , Tests d'activité antitumorale sur modèle de xénogreffe
4.
Cancer Res ; 74(12): 3294-305, 2014 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-24747911

RÉSUMÉ

Tankyrases (TNKS) play roles in Wnt signaling, telomere homeostasis, and mitosis, offering attractive targets for anticancer treatment. Using unbiased combination screening in a large panel of cancer cell lines, we have identified a strong synergy between TNKS and MEK inhibitors (MEKi) in KRAS-mutant cancer cells. Our study uncovers a novel function of TNKS in the relief of a feedback loop induced by MEK inhibition on FGFR2 signaling pathway. Moreover, dual inhibition of TNKS and MEK leads to more robust apoptosis and antitumor activity both in vitro and in vivo than effects observed by previously reported MEKi combinations. Altogether, our results show how a novel combination of TNKS and MEK inhibitors can be highly effective in targeting KRAS-mutant cancers by suppressing a newly discovered resistance mechanism.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protéines proto-oncogènes/génétique , Récepteur FGFR2/métabolisme , Tankyrases/métabolisme , Protéines G ras/génétique , Acétamides/administration et posologie , Aminopyridines/administration et posologie , Dérivés de l'aniline/administration et posologie , Animaux , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Lignée cellulaire tumorale , Synergie des médicaments , Chlorhydrate d'erlotinib , Rétrocontrôle physiologique , Femelle , Humains , MAP Kinase Kinase Kinases/antagonistes et inhibiteurs , MAP Kinase Kinase Kinases/métabolisme , Souris , Souris nude , Morpholines/administration et posologie , Mutation , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Protéines proto-oncogènes p21(ras) , Pyrimidinones/administration et posologie , Quinazolines/administration et posologie , Récepteur FGFR2/antagonistes et inhibiteurs , Transduction du signal , Sulfonamides/administration et posologie , Tankyrases/antagonistes et inhibiteurs , Thiazoles/administration et posologie , Tests d'activité antitumorale sur modèle de xénogreffe
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...