Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 6 de 6
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Anticancer Res ; 44(6): 2393-2406, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38821585

RÉSUMÉ

BACKGROUND/AIM: Cholangiocarcinoma (CCA) is an aggressive tumor with limited treatment options especially in 2nd line or later treatments. Targeting fibroblast growth factor receptor (FGFR) 2 has recently emerged as a promising treatment option for patients with CCA harboring FGFR2-fusion. This study investigated the antitumor activities of tasurgratinib as an orally available FGFR1-3 inhibitor, in preclinical FGFR2-driven CCA models. MATERIALS AND METHODS: Antitumor activities of tasurgratinib were examined in vitro and in vivo using NIH/3T3 cells expressing FGFR2-fusion as FGFR2-driven CCA models, and in vivo using a CCA patient-derived xenograft model. The molecular mechanism of action of tasurgratinib was elucidated through co-crystal structure analysis with FGFR1, manual complex model analysis with FGFR2, and binding kinetics analysis with FGFR2. Furthermore, the cell-based inhibitory activities against acquired resistant FGFR2 mutations in patients with CCA treated with FGFR inhibitors were evaluated. RESULTS: Tasurgratinib showed antitumor activity in preclinical FGFR2-driven CCA models by inhibiting the FGFR signaling pathway in vitro and in vivo. Furthermore, cell-based target engagement assays indicated that tasurgratinib had potent inhibitory activities against FGFR2 mutations, such as N549H/K, which are the major acquired mutations in CCA. We also confirmed that tasurgratinib exhibited fast association and slow dissociation kinetics with FGFR2, binding to the ATP-binding site and the neighboring region, and adopting an Asp-Phe-Gly (DFG)-"in" conformation. CONCLUSION: These data demonstrate the therapeutic potential of tasurgratinib in FGFR2-driven CCA and provide molecular mechanistic insights into its unique inhibitory profile against secondary FGFR2 resistance mutations in patients with CCA treated with FGFR inhibitors.


Sujet(s)
Tumeurs des canaux biliaires , Cholangiocarcinome , Récepteur FGFR2 , Tests d'activité antitumorale sur modèle de xénogreffe , Cholangiocarcinome/traitement médicamenteux , Cholangiocarcinome/génétique , Cholangiocarcinome/anatomopathologie , Cholangiocarcinome/métabolisme , Animaux , Humains , Récepteur FGFR2/antagonistes et inhibiteurs , Récepteur FGFR2/génétique , Récepteur FGFR2/métabolisme , Souris , Tumeurs des canaux biliaires/traitement médicamenteux , Tumeurs des canaux biliaires/anatomopathologie , Tumeurs des canaux biliaires/génétique , Tumeurs des canaux biliaires/métabolisme , Administration par voie orale , Récepteur FGFR1/antagonistes et inhibiteurs , Récepteur FGFR1/génétique , Récepteur FGFR1/métabolisme , Cellules NIH 3T3 , Récepteur de type 3 des facteurs de croissance fibroblastique/antagonistes et inhibiteurs , Récepteur de type 3 des facteurs de croissance fibroblastique/génétique , Récepteur de type 3 des facteurs de croissance fibroblastique/métabolisme , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Antinéoplasiques/administration et posologie , Inhibiteurs de protéines kinases/pharmacologie , Pyrimidines/pharmacologie , Pyrimidines/administration et posologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Protéines de fusion oncogènes/génétique , Protéines de fusion oncogènes/métabolisme , Protéines de fusion oncogènes/antagonistes et inhibiteurs
2.
Mol Cancer Ther ; 22(1): 12-24, 2023 01 03.
Article de Anglais | MEDLINE | ID: mdl-36279567

RÉSUMÉ

Innate and adaptive resistance to cancer therapies, such as chemotherapies, molecularly targeted therapies, and immune-modulating therapies, is a major issue in clinical practice. Subpopulations of tumor cells expressing the receptor tyrosine kinase AXL become enriched after treatment with antimitotic drugs, causing tumor relapse. Elevated AXL expression is closely associated with drug resistance in clinical samples, suggesting that AXL plays a pivotal role in drug resistance. Although several molecules with AXL inhibitory activity have been developed, none have sufficient activity and selectivity to be clinically effective when administered in combination with a cancer therapy. Here, we report a novel small molecule, ER-851, which is a potent and highly selective AXL inhibitor. To investigate resistance mechanisms and identify driving molecules, we conducted a comprehensive gene expression analysis of chemoresistant tumor cells in mouse xenograft models of genetically engineered human lung cancer and human triple-negative breast cancer. Consistent with the effect of AXL knockdown, cotreatment of ER-851 and antimitotic drugs produced an antitumor effect and prolonged relapse-free survival in the mouse xenograft model of human triple-negative breast cancer. Importantly, when orally administered to BALB/c mice, this compound did not induce retinal toxicity, a known side effect of chronic MER inhibition. Together, these data strongly suggest that AXL is a therapeutic target for overcoming drug resistance and that ER-851 is a promising candidate therapeutic agent for use against AXL-expressing antimitotic-resistant tumors.


Sujet(s)
Antimitotiques , Tumeurs du sein triple-négatives , Humains , Animaux , Souris , Axl Receptor Tyrosine Kinase , Antimitotiques/pharmacologie , Protéines proto-oncogènes/métabolisme , Résistance aux médicaments antinéoplasiques , Lignée cellulaire tumorale , Inhibiteurs de protéines kinases/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe
3.
Cancer Res ; 82(2): 292-306, 2022 01 15.
Article de Anglais | MEDLINE | ID: mdl-34753772

RÉSUMÉ

Combination therapies consisting of immune checkpoint inhibitors plus anti-VEGF therapy show enhanced antitumor activity and are approved treatments for patients with renal cell carcinoma (RCC). The immunosuppressive roles of VEGF in the tumor microenvironment are well studied, but those of FGF/FGFR signaling remain largely unknown. Lenvatinib is a receptor tyrosine kinase inhibitor that targets both VEGFR and FGFR. Here, we examine the antitumor activity of anti-PD-1 mAb combined with either lenvatinib or axitinib, a VEGFR-selective inhibitor, in RCC. Both combination treatments showed greater antitumor activity and longer survival in mouse models versus either single agent treatment, whereas anti-PD-1 mAb plus lenvatinib had enhanced antitumor activity compared with anti-PD-1 mAb plus axitinib. Flow cytometry analysis showed that lenvatinib decreased the population of tumor-associated macrophages and increased that of IFNγ-positive CD8+ T cells. Activation of FGFR signaling inhibited the IFNγ-stimulated JAK/STAT signaling pathway and decreased expression of its target genes, including B2M, CXCL10, and PD-L1. Furthermore, inhibition of FGFR signaling by lenvatinib restored the tumor response to IFNγ stimulation in mouse and human RCC cell lines. These preclinical results reveal novel roles of tumor FGFR signaling in the regulation of cancer immunity through inhibition of the IFNγ pathway, and the inhibitory activity of lenvatinib against FGFRs likely contributes to the enhanced antitumor activity of combination treatment comprising lenvatinib plus anti-PD-1 mAb. SIGNIFICANCE: FGFR pathway activation inhibits IFNγ signaling in tumor cells, and FGFR inhibition with lenvatinib enhances antitumor immunity and the activity of anti-PD-1 antibodies.


Sujet(s)
Anticorps monoclonaux/administration et posologie , Antinéoplasiques/administration et posologie , Néphrocarcinome/traitement médicamenteux , Néphrocarcinome/métabolisme , Interféron gamma/métabolisme , Tumeurs du rein/traitement médicamenteux , Tumeurs du rein/métabolisme , Phénylurées/administration et posologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/administration et posologie , Quinoléines/administration et posologie , Récepteur facteur croissance fibroblaste/antagonistes et inhibiteurs , Transduction du signal/effets des médicaments et des substances chimiques , Animaux , Anticorps monoclonaux/immunologie , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Néphrocarcinome/anatomopathologie , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Synergie des médicaments , Femelle , Humains , Tumeurs du rein/anatomopathologie , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Souris , Souris de lignée BALB C , Récepteur-1 de mort cellulaire programmée/immunologie , Résultat thérapeutique
4.
Bioorg Med Chem Lett ; 48: 128247, 2021 09 15.
Article de Anglais | MEDLINE | ID: mdl-34271070

RÉSUMÉ

Axl and Mer are members of the TAM (Tyro3-Axl-Mer) family of receptor tyrosine kinases. Previously, we reported that enzyme-mediated inhibition of Mer by an Axl/Mer dual inhibitor led to retinal toxicity in mice, whereas selective Axl inhibition by compound 1 did not. On the other hand, compound 1 showed low membrane permeability. Here, we designed and synthesized a novel series of 5,6,7,8-tetrahydropyrido[3,4-d]pyrimidine derivatives and evaluated their Axl and Mer inhibitory activities, leading to identification of ER-001259851-000 as a potent and selective Axl inhibitor with drug-likeness and a promising pharmacokinetic profile in mice.


Sujet(s)
Découverte de médicament , Inhibiteurs de protéines kinases/pharmacologie , Protéines proto-oncogènes/antagonistes et inhibiteurs , Pyridines/pharmacologie , Pyrimidines/pharmacologie , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , c-Mer Tyrosine kinase/antagonistes et inhibiteurs , Animaux , Relation dose-effet des médicaments , Humains , Souris , Microsomes du foie/composition chimique , Microsomes du foie/métabolisme , Structure moléculaire , Inhibiteurs de protéines kinases/synthèse chimique , Inhibiteurs de protéines kinases/composition chimique , Protéines proto-oncogènes/métabolisme , Pyridines/synthèse chimique , Pyridines/composition chimique , Pyrimidines/synthèse chimique , Pyrimidines/composition chimique , Récepteurs à activité tyrosine kinase/métabolisme , Relation structure-activité , c-Mer Tyrosine kinase/métabolisme , Axl Receptor Tyrosine Kinase
5.
Bioorg Med Chem ; 39: 116137, 2021 06 01.
Article de Anglais | MEDLINE | ID: mdl-33930844

RÉSUMÉ

Axl and Mer are a members of the TAM (Tyro3-Axl-Mer) family of receptor tyrosine kinases, which, when activated, can promote tumor cell survival, proliferation, migration, invasion, angiogenesis, and tumor-host interactions. Chronic inhibition of Mer leads to retinal toxicity in mice. Therefore, successful development of an Axl targeting agent requires ensuring that it is safe for prolonged treatment. Here, to clarify whether enzyme inhibition of Mer by a small molecule leads to retinal toxicity in mice, we designed and synthesized Axl/Mer inhibitors and Axl-selective inhibitors. We identified an Axl/Mer dual inhibitor 28a, which showed retinal toxicity at a dose of 100 mg/kg in mice. Subsequent derivatization of a pyridine derivative led to the discovery of a pyrimidine derivative, 33g, which selectively inhibited the activity of Axl over Mer without retinal toxicity at a dose of 100 mg/kg in mice. Additionally, the compound displayed in vivo anti-tumor effects without influencing body weight in a Ba/F3-Axl isogenic subcutaneous model.


Sujet(s)
Découverte de médicament , Inhibiteurs de protéines kinases/pharmacologie , Protéines proto-oncogènes/antagonistes et inhibiteurs , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Animaux , Souris , Modèles animaux , Inhibiteurs de protéines kinases/composition chimique , Rétine/effets des médicaments et des substances chimiques , Analyse spectrale/méthodes , Relation structure-activité , Axl Receptor Tyrosine Kinase
6.
Adv Neonatal Care ; 17(3): 184-191, 2017 Jun.
Article de Anglais | MEDLINE | ID: mdl-28114148

RÉSUMÉ

BACKGROUND: Neonatal pain management guidelines have been released; however, there is insufficient systematic institutional support for the adoption of evidence-based pain management in Japan. PURPOSE: To evaluate the impact of a collaborative quality improvement program on the implementation of pain management improvements in Japanese neonatal intensive care units (NICUs). METHODS: Seven Japanese level III NICUs participated in a neonatal pain management quality improvement program based on an Institute for Healthcare Improvement collaborative model. The NICUs developed evidence-based practice points for pain management and implemented these over a 12-month period. Changes were introduced through a series of Plan-Do-Study-Act cycles, and throughout the process, pain management quality indicators were tracked as performance measures. Jonckheere's trend test and the Cochran-Armitage test for trend were used to examine the changes in quality indicator implementations over time (baseline, 3 months, 6 months, and 12 months). FINDINGS: Baseline pain management data from the 7 sites revealed substantial opportunities for improvement of pain management, and testing changes in the NICU setting resulted in measurable improvements in pain management. During the intervention phase, all participating sites introduced new pain assessment tools, and all sites developed electronic medical record forms to capture pain score, interventions, and infant responses to interventions. IMPLICATIONS FOR PRACTICE: The use of collaborative quality improvement techniques played a key role in improving pain management in the NICUs. IMPLICATIONS FOR RESEARCH: Collaborative improvement programs provide an attractive strategy for solving evidence-practice gaps in the NICU setting.


Sujet(s)
Unités de soins intensifs néonatals , Gestion de la douleur/méthodes , Assurance de la qualité des soins de santé/méthodes , Amélioration de la qualité , Médecine factuelle , Humains , Nourrisson , Nouveau-né , Japon , Soins infirmiers en néonatalogie , Néonatologistes , Études prospectives , Amélioration de la qualité/organisation et administration
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE