Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.331
Filtrer
1.
Front Immunol ; 15: 1401972, 2024.
Article de Anglais | MEDLINE | ID: mdl-38911858

RÉSUMÉ

Myasthenia gravis with positive MuSK antibody often involves the bulbar muscles and is usually refractory to acetylcholinesterase inhibitors. For MuSK-MG patients who experience acute exacerbations and do not respond to conventional treatments, there is an urgent need to find more suitable treatment options. With the advent of biologic agents, efgartigimod has shown promising results in the treatment of MG. We report a 65-year-old MuSK-MG patient who presented with impaired eye movements initially, and the symptoms rapidly worsened within a week, affecting the limbs and neck muscles, and had difficulties in chewing and swallowing. Lymphoplasmapheresis did not achieve satisfactory results, but after a cycle of efgartigimod treatment, the patient's symptoms gradually improved and remained in a good clinical state for several months.


Sujet(s)
Myasthénie , Récepteurs cholinergiques , Humains , Myasthénie/traitement médicamenteux , Sujet âgé , Récepteurs cholinergiques/immunologie , Résultat thérapeutique , Récepteurs à activité tyrosine kinase/immunologie , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Autoanticorps/immunologie , Autoanticorps/sang , Mâle , Femelle
3.
Biomed Pharmacother ; 176: 116892, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38876048

RÉSUMÉ

The lesson from many studies investigating the efficacy of targeted therapy in glioblastoma (GBM) showed that a future perspective should be focused on combining multiple target treatments. Our research aimed to assess the efficacy of drug combinations against glioblastoma stem cells (GSCs). Patient-derived cells U3042, U3009, and U3039 were obtained from the Human Glioblastoma Cell Culture resource. Additionally, the study was conducted on a GBM commercial U251 cell line. Gene expression analysis related to receptor tyrosine kinases (RTKs), stem cell markers and genes associated with significant molecular targets was performed, and selected proteins encoded by these genes were assessed using the immunofluorescence and flow cytometry methods. The cytotoxicity studies were preceded by analyzing the expression of specific proteins that serve as targets for selected drugs. The cytotoxicity study using the MTS assay was conducted to evaluate the effects of selected drugs/candidates in monotherapy and combinations. The most cytotoxic compounds for U3042 cells were Disulfiram combined with Copper gluconate (DSF/Cu), Dacomitinib, and Foretinib with IC50 values of 52.37 nM, 4.38 µM, and 4.54 µM after 24 h incubation, respectively. Interactions were assessed using SynergyFinder Plus software. The analysis enabled the identification of the most effective drug combinations against patient-derived GSCs. Our findings indicate that the most promising drug combinations are Dacomitinib and Foretinib, Dacomitinib and DSF/Cu, and Foretinib and AZD3759. Since most tested combinations have not been previously examined against glioblastoma stem-like cells, these results can shed new light on designing the therapeutic approach to target the GSC population.


Sujet(s)
Repositionnement des médicaments , Glioblastome , Cellules souches tumorales , Inhibiteurs de protéines kinases , Humains , Glioblastome/traitement médicamenteux , Glioblastome/anatomopathologie , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/anatomopathologie , Repositionnement des médicaments/méthodes , Inhibiteurs de protéines kinases/pharmacologie , Lignée cellulaire tumorale , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Récepteurs à activité tyrosine kinase/métabolisme , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Antinéoplasiques/pharmacologie , Survie cellulaire/effets des médicaments et des substances chimiques
4.
Nat Commun ; 15(1): 5292, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38906855

RÉSUMÉ

Ewing sarcoma is a pediatric bone and soft tissue tumor treated with chemotherapy, radiation, and surgery. Despite intensive multimodality therapy, ~50% patients eventually relapse and die of the disease due to chemoresistance. Here, using phospho-profiling, we find Ewing sarcoma cells treated with chemotherapeutic agents activate TAM (TYRO3, AXL, MERTK) kinases to augment Akt and ERK signaling facilitating chemoresistance. Mechanistically, chemotherapy-induced JAK1-SQ phosphorylation releases JAK1 pseudokinase domain inhibition allowing for JAK1 activation. This alternative JAK1 activation mechanism leads to STAT6 nuclear translocation triggering transcription and secretion of the TAM kinase ligand GAS6 with autocrine/paracrine consequences. Importantly, pharmacological inhibition of either JAK1 by filgotinib or TAM kinases by UNC2025 sensitizes Ewing sarcoma to chemotherapy in vitro and in vivo. Excitingly, the TAM kinase inhibitor MRX-2843 currently in human clinical trials to treat AML and advanced solid tumors, enhances chemotherapy efficacy to further suppress Ewing sarcoma tumor growth in vivo. Our findings reveal an Ewing sarcoma chemoresistance mechanism with an immediate translational value.


Sujet(s)
Protéines et peptides de signalisation intercellulaire , Janus kinase 1 , Récepteurs à activité tyrosine kinase , Sarcome d'Ewing , Transduction du signal , Sarcome d'Ewing/traitement médicamenteux , Sarcome d'Ewing/métabolisme , Sarcome d'Ewing/anatomopathologie , Sarcome d'Ewing/génétique , Humains , Janus kinase 1/métabolisme , Janus kinase 1/antagonistes et inhibiteurs , Janus kinase 1/génétique , Lignée cellulaire tumorale , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Récepteurs à activité tyrosine kinase/métabolisme , Récepteurs à activité tyrosine kinase/génétique , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Souris , Protéines et peptides de signalisation intercellulaire/métabolisme , Axl Receptor Tyrosine Kinase , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/antagonistes et inhibiteurs , Tumeurs osseuses/traitement médicamenteux , Tumeurs osseuses/métabolisme , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/génétique , Tests d'activité antitumorale sur modèle de xénogreffe , c-Mer Tyrosine kinase/métabolisme , c-Mer Tyrosine kinase/génétique , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Phosphorylation/effets des médicaments et des substances chimiques , Femelle , Facteur de transcription STAT-6
5.
Anticancer Res ; 44(7): 2805-2813, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38925827

RÉSUMÉ

BACKGROUND/AIM: Randomized trials have shown the benefit of combining tyrosine kinase inhibitors (TKI) and chemotherapy in the treatment of epidermal growth factor receptor-mutant non-small-cell lung cancer (NSCLC). For anaplastic lymphoma kinase-rearranged (ALK+) NSCLC, prospective trial results of the combination are not available and have not even been thoroughly investigated in vitro. In this study, we investigated combinations of TKI and chemotherapy using in vitro models of ALK+ NSCLC. MATERIALS AND METHODS: ALK+ cell line models H3122, H2228, and DFCI032 with differing primary resistance to ALK receptor TKIs were used. We investigated short-(viability assay) and long-term (colony-formation assay) cytotoxicity, apoptosis, and cell signaling in response to the combinations of agents. We selected the most commonly used agents, alectinib, cisplatin, and pemetrexed, to investigate the combination effects. RESULTS: In the combination experiments with short-term exposure, synergism between TKI and pemetrexed was observed, while cisplatin had antagonistic effects. In the long-term experiments, the combination of cisplatin and TKI was synergistic in all lines, while no synergism was observed with pemetrexed. Among the chemotherapy and TKI sequences, cisplatin followed by TKI was more cytotoxic than the opposite in two out of the three models. In the TKI-sensitive H3122 cell line, the combination of chemotherapy and TKI combination increased apoptosis. Interestingly, pemetrexed treatment resulted in the activation of ALK, which was abolished with TKI. CONCLUSION: Combining TKI and chemotherapy in ALK+ models has some synergistic effects that overcome primary TKI resistance. However, the synergy varies depending on the chemotherapeutic agent, cytotoxic assay, and the cell line used. Prospective clinical trials are warranted to fully characterize the potential of combination chemotherapy with TKIs in ALK+ NSCLC.


Sujet(s)
Kinase du lymphome anaplasique , Protocoles de polychimiothérapie antinéoplasique , Carcinome pulmonaire non à petites cellules , Cisplatine , Tumeurs du poumon , Pémétrexed , Inhibiteurs de protéines kinases , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Kinase du lymphome anaplasique/antagonistes et inhibiteurs , Kinase du lymphome anaplasique/génétique , Kinase du lymphome anaplasique/métabolisme , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Inhibiteurs de protéines kinases/pharmacologie , Lignée cellulaire tumorale , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Cisplatine/pharmacologie , Cisplatine/administration et posologie , Pémétrexed/pharmacologie , Pémétrexed/administration et posologie , Apoptose/effets des médicaments et des substances chimiques , Synergie des médicaments , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Récepteurs à activité tyrosine kinase/génétique , Récepteurs à activité tyrosine kinase/métabolisme , Pipéridines/pharmacologie , Pipéridines/administration et posologie , Carbazoles/pharmacologie , Carbazoles/administration et posologie
6.
Bioconjug Chem ; 35(5): 674-681, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38695582

RÉSUMÉ

Aptamers are widely used molecular recognition tools in targeted therapy, but their ability to effectively penetrate deep into solid tumors remains a significant challenge, leading to suboptimal treatment efficacy. Here, we developed a polyfluoroalkyl (PFA) decoration strategy to enhance aptamer recognition, cell internalization, and solid tumor penetration. Our results indicate that PFA with around 11 fluorine atoms significantly improves aptamer internalization both in vitro and in vivo settings. However, we also observed that the use of PFA tags containing 19 and 23 fluorine atoms on aptamers resulted in nonspecific cell anchoring in control cell lines, affecting the specificity of aptamers. Overall, we found that using a chemical modification strategy could enhance the deep tumor penetration ability of aptamers and validate their effectiveness in vivo. This approach has significant practical applications in targeted drug delivery for cancer treatment.


Sujet(s)
Aptamères nucléotidiques , Récepteurs à activité tyrosine kinase , Aptamères nucléotidiques/composition chimique , Humains , Animaux , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Récepteurs à activité tyrosine kinase/métabolisme , Lignée cellulaire tumorale , Souris , Molécules d'adhérence cellulaire/métabolisme , Molécules d'adhérence cellulaire/antagonistes et inhibiteurs , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Systèmes de délivrance de médicaments/méthodes
7.
Oncogene ; 43(26): 1973-1984, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38773263

RÉSUMÉ

The generation of drugs counteracting deregulated protein kinases has been a major focus in cancer therapy development. Breakthroughs in this effort have produced many therapeutic agents to the benefit of patients, mostly through the development of chemical or antibody-based drugs targeting active kinases. These strategies are challenged when considering catalytically inactive protein kinases (or pseudokinases), which represent 10% of the human kinome with many of relevance in cancer. Among the so-called pseudotyrosine kinases, the PTK7 receptor tyrosine kinase (RTK) stands as a bona fide target overexpressed in several solid tumors and hematological malignancies and linked to metastasis, poor prognosis, and resistance to treatment. Despite the lack of catalytic activity, PTK7 has signaling capacities through heterodimerization with active RTKs and offers pharmacological targeting opportunities through its inactive kinase domain. Moreover, PTK7-targeting strategies based on antibody-drug conjugates, aptamers, and CAR-T cell-based therapies have demonstrated encouraging results in preclinical and clinical settings. We review the most recent data assigning to PTK7 a prominent role in cancer progression as well as current preclinical and clinical targeting strategies against RTK family pseudokinases including PTK7.


Sujet(s)
Molécules d'adhérence cellulaire , Thérapie moléculaire ciblée , Tumeurs , Récepteurs à activité tyrosine kinase , Humains , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Tumeurs/métabolisme , Récepteurs à activité tyrosine kinase/métabolisme , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Molécules d'adhérence cellulaire/métabolisme , Molécules d'adhérence cellulaire/antagonistes et inhibiteurs , Thérapie moléculaire ciblée/méthodes , Animaux , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques
8.
J Neurol Sci ; 461: 123047, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38759248

RÉSUMÉ

BACKGROUND: Myasthenia gravis (MG) with MuSK antibodies (MuSK-MG) represents a distinct subtype with different responses to treatments compared to patients with AChR antibodies, especially in terms of tolerance to acetylcholinesterase inhibitors (AChEI). However, AChEI are often used as first line symptomatic treatment in MuSK-MG, despite reports that they are poorly tolerated, seldom effective or even deleterious. METHODS: We analyzed demographic, clinical and therapeutic responses and side-effects in the large cohort of 202 MuSK-MG patients cared for at the MG Clinic of Azienda Ospedaliero-Universitaria Pisana. RESULTS: 165 patients had received AChEI at first evaluation. Only 7/165 patients (4.2%) reported an initial clinical benefit. Conversely, 76.9% of patients reported at least one side effect, most commonly neuromuscular hyperexcitability (68.4%), gastrointestinal (53.9%) and neurovegetative (35.8%) disturbances. 56 (33.9%) patients reported a concomitant worsening of muscle weakness and twelve patients (7.3%) suffered a cholinergic crisis. According to these patients, the severity of cholinergic side effects was greater at higher doses of AChEI, but side effects occurred regardless of the dose administered and ceased once the drug was discontinued. CONCLUSIONS: This is the largest population of MuSK-MG patients reported for perceived responsiveness and tolerance to AChEI treatment. Our obervations strongly suggest avoiding this treatment in MuSK-MG.


Sujet(s)
Autoanticorps , Anticholinestérasiques , Myasthénie , Récepteurs à activité tyrosine kinase , Récepteurs cholinergiques , Humains , Myasthénie/traitement médicamenteux , Myasthénie/immunologie , Anticholinestérasiques/usage thérapeutique , Mâle , Femelle , Adulte d'âge moyen , Récepteurs cholinergiques/immunologie , Adulte , Récepteurs à activité tyrosine kinase/immunologie , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Sujet âgé , Autoanticorps/sang , Jeune adulte , Adolescent , Sujet âgé de 80 ans ou plus , Résultat thérapeutique , Études de cohortes
9.
Eur J Med Chem ; 272: 116475, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38714043

RÉSUMÉ

AXL, a receptor tyrosine kinase (RTK), plays a pivotal role in various cellular functions. It is primarily involved in processes such as epithelial-mesenchymal transition (EMT) in tumor cells, angiogenesis, apoptosis, immune regulation, and chemotherapy resistance mechanisms. Therefore, targeting AXL is a promising therapeutic approach for the treatment of cancer. AXL inhibitors that have entered clinical trials, such as BGB324(1), have shown promising efficacy in the treatment of melanoma and non-small cell lung cancer. Additionally, novel AXL-targeted drugs, such as AXL degraders, offer a potential solution to overcome the limitations of traditional small-molecule AXL inhibitors targeting single pathways. We provide an overview of the structure and biological functions of AXL, discusses its correlation with various cancers, and critically analyzes the structure-activity relationship of AXL small-molecule inhibitors in cellular contexts. Additionally, we summarize multiple research and development strategies, offering insights for the future development of innovative AXL inhibitors.


Sujet(s)
Antinéoplasiques , Axl Receptor Tyrosine Kinase , Découverte de médicament , Inhibiteurs de protéines kinases , Protéines proto-oncogènes , Récepteurs à activité tyrosine kinase , Humains , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Récepteurs à activité tyrosine kinase/métabolisme , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Protéines proto-oncogènes/antagonistes et inhibiteurs , Protéines proto-oncogènes/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/synthèse chimique , Relation structure-activité , Structure moléculaire , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Animaux , Tests de criblage d'agents antitumoraux , Prolifération cellulaire/effets des médicaments et des substances chimiques , Développement de médicament
10.
Front Immunol ; 15: 1400553, 2024.
Article de Anglais | MEDLINE | ID: mdl-38817615

RÉSUMÉ

Background and aims: Metabolic dysfunction-associated steatohepatitis (MASH) is a significant health concern with limited treatment options. AXL, a receptor tyrosine kinase activated by the GAS6 ligand, promotes MASH through activation of hepatic stellate cells and inflammatory macrophages. This study identified cell subsets affected by MASH progression and the effect of AXL inhibition. Methods: Mice were fed chow or different fat-enriched diets to induce MASH, and small molecule AXL kinase inhibition with bemcentinib was evaluated. Gene expression was measured by qPCR. Time-of-flight mass cytometry (CyTOF) used single cells from dissociated livers, acquired on the Fluidigm Helios, and cell populations were studied using machine learning. Results: In mice fed different fat-enriched diets, liver steatosis alone was insufficient to elevate plasma soluble AXL (sAXL) levels. However, in conjunction with inflammation, sAXL increases, serving as an early indicator of steatohepatitis progression. Bemcentinib, an AXL inhibitor, effectively reduced proinflammatory responses in MASH models, even before fibrosis appearance. Utilizing CyTOF analysis, we detected a decreased population of Kupffer cells during MASH while promoting infiltration of monocytes/macrophages and CD8+ T cells. Bemcentinib partially restored Kupffer cells, reduced pDCs and GzmB- NK cells, and increased GzmB+CD8+ T cells and LSECs. Additionally, AXL inhibition enhanced a subtype of GzmB+CD8+ tissue-resident memory T cells characterized by CX3CR1 expression. Furthermore, bemcentinib altered the transcriptomic landscape associated with MASH progression, particularly in TLR signaling and inflammatory response, exhibiting differential cytokine expression in the plasma, consistent with liver repair and decreased inflammation. Conclusion: Our findings highlight sAXL as a biomarker for monitoring MASH progression and demonstrate that AXL targeting shifted liver macrophages and CD8+ T-cell subsets away from an inflammatory phenotype toward fibrotic resolution and organ healing, presenting a promising strategy for MASH treatment.


Sujet(s)
Axl Receptor Tyrosine Kinase , Cirrhose du foie , Protéines proto-oncogènes , Récepteurs à activité tyrosine kinase , Animaux , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Récepteurs à activité tyrosine kinase/métabolisme , Souris , Protéines proto-oncogènes/antagonistes et inhibiteurs , Protéines proto-oncogènes/métabolisme , Cirrhose du foie/immunologie , Cirrhose du foie/traitement médicamenteux , Cirrhose du foie/anatomopathologie , Cirrhose du foie/métabolisme , Mâle , Modèles animaux de maladie humaine , Souris de lignée C57BL , Benzocycloheptènes/pharmacologie , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Foie/anatomopathologie , Foie/immunologie , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Macrophages/immunologie , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/métabolisme , Triazoles
11.
Oncol Res ; 32(5): 849-875, 2024.
Article de Anglais | MEDLINE | ID: mdl-38686058

RÉSUMÉ

Glioblastoma, the most aggressive form of brain tumor, poses significant challenges in terms of treatment success and patient survival. Current treatment modalities for glioblastoma include radiation therapy, surgical intervention, and chemotherapy. Unfortunately, the median survival rate remains dishearteningly low at 12-15 months. One of the major obstacles in treating glioblastoma is the recurrence of tumors, making chemotherapy the primary approach for secondary glioma patients. However, the efficacy of drugs is hampered by the presence of the blood-brain barrier and multidrug resistance mechanisms. Consequently, considerable research efforts have been directed toward understanding the underlying signaling pathways involved in glioma and developing targeted drugs. To tackle glioma, numerous studies have examined kinase-downstream signaling pathways such as RAS-RAF-MEK-ERK-MPAK. By targeting specific signaling pathways, heterocyclic compounds have demonstrated efficacy in glioma therapeutics. Additionally, key kinases including phosphatidylinositol 3-kinase (PI3K), serine/threonine kinase, cytoplasmic tyrosine kinase (CTK), receptor tyrosine kinase (RTK) and lipid kinase (LK) have been considered for investigation. These pathways play crucial roles in drug effectiveness in glioma treatment. Heterocyclic compounds, encompassing pyrimidine, thiazole, quinazoline, imidazole, indole, acridone, triazine, and other derivatives, have shown promising results in targeting these pathways. As part of this review, we propose exploring novel structures with low toxicity and high potency for glioma treatment. The development of these compounds should strive to overcome multidrug resistance mechanisms and efficiently penetrate the blood-brain barrier. By optimizing the chemical properties and designing compounds with enhanced drug-like characteristics, we can maximize their therapeutic value and minimize adverse effects. Considering the complex nature of glioblastoma, these novel structures should be rigorously tested and evaluated for their efficacy and safety profiles.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Inhibiteurs de protéines kinases , Humains , Glioblastome/traitement médicamenteux , Glioblastome/anatomopathologie , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Récepteurs à activité tyrosine kinase/métabolisme , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Composés hétérocycliques/usage thérapeutique , Composés hétérocycliques/composition chimique , Composés hétérocycliques/pharmacologie , Thérapie moléculaire ciblée , Animaux
12.
Cancer Lett ; 592: 216905, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38677641

RÉSUMÉ

Craniopharyngiomas (CPs), particularly Adamantinomatous Craniopharyngiomas (ACPs), often exhibit a heightened risk of postoperative recurrence and severe complications of the endocrine and hypothalamic function. The primary objective of this study is to investigate potential novel targeted therapies within the microenvironment of ACP tumors. Cancer-Associated Fibroblasts (CAFs) were identified in the craniopharyngioma microenvironment, notably in regions characterized by cholesterol clefts, wet keratin, ghost cells, and fibrous stroma in ACPs. CAFs, alongside ghost cells, basaloid-like epithelium cells and calcifications, were found to secrete PROS1 and GAS6, which can activate AXL receptors on the surface of tumor epithelium cells, promoting immune suppression and tumor progression in ACPs. Additionally, the AXL inhibitor Bemcentinib effectively inhibited the proliferation organoids and enhanced the immunotherapeutic efficacy of Atezolizumab. Furthermore, neural crest-like cells were observed in the glial reactive tissue surrounding finger-like protrusions. Overall, our results revealed that the AXL might be a potentially effective therapeutic target for ACPs.


Sujet(s)
Axl Receptor Tyrosine Kinase , Craniopharyngiome , Tumeurs de l'hypophyse , Protéines proto-oncogènes , Récepteurs à activité tyrosine kinase , Microenvironnement tumoral , Humains , Craniopharyngiome/génétique , Craniopharyngiome/traitement médicamenteux , Craniopharyngiome/anatomopathologie , Craniopharyngiome/métabolisme , Récepteurs à activité tyrosine kinase/génétique , Récepteurs à activité tyrosine kinase/métabolisme , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , Tumeurs de l'hypophyse/génétique , Tumeurs de l'hypophyse/anatomopathologie , Tumeurs de l'hypophyse/traitement médicamenteux , Tumeurs de l'hypophyse/métabolisme , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Femelle , Mâle , Protéines et peptides de signalisation intercellulaire/génétique , Protéines et peptides de signalisation intercellulaire/métabolisme , Fibroblastes associés au cancer/effets des médicaments et des substances chimiques , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/anatomopathologie , Analyse de profil d'expression de gènes/méthodes , RNA-Seq , Benzocycloheptènes/pharmacologie , Animaux , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Souris , Prolifération cellulaire/effets des médicaments et des substances chimiques , Adulte , Thérapie moléculaire ciblée , Adulte d'âge moyen , Triazoles
13.
Eur J Pharmacol ; 973: 176600, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38643834

RÉSUMÉ

Multiple sclerosis is an autoimmune disease that causes inflammatory damage to the central nervous system. At present, the pathogenesis of the disease is unknown. There is a lack of few effective therapy medications available. Therefore, it is necessary to further explore the pathogenesis of this illness and develop potential therapeutic drugs. Dabrafenib is potential therapeutic medicine for nervous system disease. In this study, we preliminarily studied the possible mechanism of dabrafenib in the treatment of multiple sclerosis from the perspective of ferroptosis. First, we observed that dabrafenib significantly improved symptoms of gait abnormalities, limb weakness or paralysis, and down-regulated levels of spinal cord inflammation in an experimental autoimmune encephalitis (EAE) model. Meanwhile, we also observed that dabrafenib could inhibit the proteins of ferroptosis in spinal cord tissue of EAE mice by Western blot. The results of immunohistochemical analysis showed that the effect of dabrafenib on ferroptosis mainly occurred in microglia. Second, dabrafenib was demonstrated to be able to inhibit the S phase of the cell cycle, reduce ROS levels, and reinstate mitochondrial activity in the LPS-induced BV2 inflammatory cell model. Futhermore, we found that dabrafenib inhibits P-JAK2 and P-STAT3 activation by acting Axl receptor, which in turn prevents neurogenic inflammation in microglia. The co-stimulated BV2 cell model with LPS and Erastin also verified these findings. Ultimately, the Axl knockout mice used to construct the EAE model allowed for the confirmation that dabrafenib prevented ferroptosis in microglia by up-regulating Axl receptor, which reduced the inflammatory demyelination associated with EAE. In summary, our research demonstrates the advantages of dabrafenib in multiple sclerosis treatment, which can prevent ferroptosis in microglia in multiple sclerosis through up-regulating Axl receptor, thus halting the progression of multiple sclerosis.


Sujet(s)
Axl Receptor Tyrosine Kinase , Encéphalomyélite auto-immune expérimentale , Ferroptose , Imidazoles , Oximes , Protéines proto-oncogènes , Récepteurs à activité tyrosine kinase , Régulation positive , Animaux , Imidazoles/pharmacologie , Imidazoles/usage thérapeutique , Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Encéphalomyélite auto-immune expérimentale/anatomopathologie , Encéphalomyélite auto-immune expérimentale/métabolisme , Ferroptose/effets des médicaments et des substances chimiques , Protéines proto-oncogènes/métabolisme , Souris , Oximes/pharmacologie , Oximes/usage thérapeutique , Récepteurs à activité tyrosine kinase/métabolisme , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Régulation positive/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Femelle , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Microglie/anatomopathologie , Facteur de transcription STAT-3/métabolisme , Lignée cellulaire , Moelle spinale/effets des médicaments et des substances chimiques , Moelle spinale/anatomopathologie , Moelle spinale/métabolisme , Maladies neuro-inflammatoires/traitement médicamenteux , Maladies neuro-inflammatoires/anatomopathologie , Maladies neuro-inflammatoires/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
14.
Drug Discov Today ; 29(6): 104005, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38685399

RÉSUMÉ

AXL receptor tyrosine kinase (AXL) is a receptor tyrosine kinase whose aberrant expression has recently been associated with colorectal cancer (CRC), contributing to tumor growth, epithelial-mesenchymal transition (EMT), increased invasiveness, metastatic spreading, and the development of drug resistance. In this review we summarize preclinical data, the majority of which are limited to recent years, convincingly linking the AXL receptor to CRC. These findings support the value of targeting AXL with molecules in drug discovery, offering novel and advanced therapeutic or diagnostic tools for CRC management.


Sujet(s)
Antinéoplasiques , Axl Receptor Tyrosine Kinase , Tumeurs colorectales , Thérapie moléculaire ciblée , Protéines proto-oncogènes , Récepteurs à activité tyrosine kinase , Humains , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/antagonistes et inhibiteurs , Récepteurs à activité tyrosine kinase/métabolisme , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Animaux , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Transition épithélio-mésenchymateuse , Résistance aux médicaments antinéoplasiques
15.
Comput Biol Med ; 174: 108433, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38642491

RÉSUMÉ

Breast cancer, a highly formidable and diverse malignancy predominantly affecting women globally, poses a significant threat due to its intricate genetic variability, rendering it challenging to diagnose accurately. Various therapies such as immunotherapy, radiotherapy, and diverse chemotherapy approaches like drug repurposing and combination therapy are widely used depending on cancer subtype and metastasis severity. Our study revolves around an innovative drug discovery strategy targeting potential drug candidates specific to RTK signalling, a prominently targeted receptor class in cancer. To accomplish this, we have developed a multimodal deep neural network (MM-DNN) based QSAR model integrating omics datasets to elucidate genomic, proteomic expression data, and drug responses, validated rigorously. The results showcase an R2 value of 0.917 and an RMSE value of 0.312, affirming the model's commendable predictive capabilities. Structural analogs of drug molecules specific to RTK signalling were sourced from the PubChem database, followed by meticulous screening to eliminate dissimilar compounds. Leveraging the MM-DNN-based QSAR model, we predicted the biological activity of these molecules, subsequently clustering them into three distinct groups. Feature importance analysis was performed. Consequently, we successfully identified prime drug candidates tailored for each potential downstream regulatory protein within the RTK signalling pathway. This method makes the early stages of drug development faster by removing inactive compounds, providing a hopeful path in combating breast cancer.


Sujet(s)
Antinéoplasiques , Tumeurs du sein , Apprentissage profond , Découverte de médicament , Transduction du signal , Humains , Femelle , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Antinéoplasiques/composition chimique , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Récepteurs à activité tyrosine kinase/métabolisme , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Récepteurs à activité tyrosine kinase/génétique , Relation quantitative structure-activité , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie
16.
Adv Mater ; 36(21): e2308504, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38546279

RÉSUMÉ

Anexelekto (AXL) is an attractive molecular target for ovarian cancer therapy because of its important role in ovarian cancer initiation and progression. To date, several AXL inhibitors have entered clinical trials for the treatment of ovarian cancer. However, the disadvantages of low AXL affinity and severe off-target toxicity of these inhibitors limit their further clinical applications. Herein, by rational design of a nonapeptide derivative Nap-Phe-Phe-Glu-Ile-Arg-Leu-Arg-Phe-Lys (Nap-IR), a strategy of in situ nanofiber formation is proposed to suppress ovarian cancer growth. After administration, Nap-IR specifically targets overexpressed AXL on ovarian cancer cell membranes and undergoes a receptor-instructed nanoparticle-to-nanofiber transition. In vivo and in vitro experiments demonstrate that in situ formed Nap-IR nanofibers efficiently induce apoptosis of ovarian cancer cells by blocking AXL activation and disrupting subsequent downstream signaling events. Remarkably, Nap-IR can synergistically enhance the anticancer effect of cisplatin against HO8910 ovarian tumors. It is anticipated that the Nap-IR can be applied in clinical ovarian cancer therapy in the near future.


Sujet(s)
Axl Receptor Tyrosine Kinase , Protéines et peptides de signalisation intercellulaire , Nanofibres , Tumeurs de l'ovaire , Protéines proto-oncogènes , Récepteurs à activité tyrosine kinase , Femelle , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/métabolisme , Tumeurs de l'ovaire/anatomopathologie , Humains , Récepteurs à activité tyrosine kinase/métabolisme , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Nanofibres/composition chimique , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Animaux , Protéines et peptides de signalisation intercellulaire/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Oligopeptides/composition chimique , Oligopeptides/pharmacologie , Souris , Liaison aux protéines , Cisplatine/pharmacologie , Cisplatine/composition chimique
17.
Mol Cancer Ther ; 23(6): 864-876, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38471796

RÉSUMÉ

Rhabdomyosarcoma (RMS) is a highly aggressive pediatric cancer with features of skeletal muscle differentiation. More than 80% of the high-risk patients ultimately fail to respond to chemotherapy treatment, leading to limited therapeutic options and dismal prognostic rates. The lack of response and subsequent tumor recurrence is driven in part by stem cell-like cells, the tumor subpopulation that is enriched after treatment, and characterized by expression of the AXL receptor tyrosine kinase (AXL). AXL mediates survival, migration, and therapy resistance in several cancer types; however, its function in RMS remains unclear. In this study, we investigated the role of AXL in RMS tumorigenesis, migration, and chemotherapy response, and whether targeting of AXL with small-molecule inhibitors could potentiate the efficacy of chemotherapy. We show that AXL is expressed in a heterogeneous manner in patient-derived xenografts (PDX), primary cultures and cell line models of RMS, consistent with its stem cell-state selectivity. By generating a CRISPR/Cas9 AXL knock-out and overexpressing models, we show that AXL contributes to the migratory phenotype of RMS, but not to chemotherapy resistance. Instead, pharmacologic blockade with the AXL inhibitors bemcentinib (BGB324), cabozantinib and NPS-1034 rapidly killed RMS cells in an AXL-independent manner and augmented the efficacy of the chemotherapeutics vincristine and cyclophosphamide. In vivo administration of the combination of bemcentinib and vincristine exerted strong antitumoral activity in a rapidly progressing PDX mouse model, significantly reducing tumor burden compared with single-agent treatment. Collectively, our data identify bemcentinib as a promising drug to improve chemotherapy efficacy in patients with RMS.


Sujet(s)
Axl Receptor Tyrosine Kinase , Benzocycloheptènes , Protéines proto-oncogènes , Récepteurs à activité tyrosine kinase , Rhabdomyosarcome , Humains , Rhabdomyosarcome/traitement médicamenteux , Rhabdomyosarcome/anatomopathologie , Rhabdomyosarcome/génétique , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Récepteurs à activité tyrosine kinase/métabolisme , Animaux , Souris , Protéines proto-oncogènes/antagonistes et inhibiteurs , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Benzocycloheptènes/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe , Lignée cellulaire tumorale , Enfant , Prolifération cellulaire/effets des médicaments et des substances chimiques , Protéines de fusion oncogènes/génétique , Protéines de fusion oncogènes/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Inhibiteurs de protéines kinases/pharmacologie , Triazoles
18.
Acta Pharmacol Sin ; 45(6): 1264-1275, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38438582

RÉSUMÉ

In addition to the classical resistance mechanisms, receptor tyrosine-protein kinase AXL is a main mechanism of resistance to third-generation epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) osimertinib in EGFR-mutated non-small cell lung cancer (NSCLC). Developing an effective AXL inhibitor is important to sensitize osimertinib in clinical application. In this study we assessed the efficacy of brigatinib, a second-generation of anaplastic lymphoma kinase (ALK)-TKI, as a novel AXL inhibitor, in overcoming acquired resistance to osimertinib induced by AXL activation. We established an AXL-overexpression NSCLC cell line and conducted high-throughput screening of a small molecule chemical library containing 510 anti-tumor drugs. We found that brigatinib potently inhibited AXL expression, and that brigatinib (0.5 µM) significantly enhanced the anti-tumor efficacy of osimertinib (1 µM) in AXL-mediated osimertinib-resistant NSCLC cell lines in vitro. We demonstrated that brigatinib had a potential ability to bind AXL kinase protein and further inhibit its downstream pathways in NSCLC cell lines. Furthermore, we revealed that brigatinib might decrease AXL expression through increasing K48-linked ubiquitination of AXL and promoting AXL degradation in HCC827OR cells and PC-9OR cells. In AXL-high expression osimertinib-resistant PC-9OR and HCC827OR cells derived xenograft mouse models, administration of osimertinib (10 mg·kg-1·d-1) alone for 3 weeks had no effect, and administration of brigatinib (25 mg·kg-1·d-1) alone caused a minor inhibition on the tumor growth; whereas combination of osimertinib and brigatinib caused marked tumor shrinkages. We concluded that brigatinib may be a promising clinical strategy for enhancing osimertinib efficacy in AXL-mediated osimertinib-resistant NSCLC patients.


Sujet(s)
Acrylamides , Dérivés de l'aniline , Antinéoplasiques , Axl Receptor Tyrosine Kinase , Carcinome pulmonaire non à petites cellules , Résistance aux médicaments antinéoplasiques , Récepteurs ErbB , Tumeurs du poumon , Souris nude , Composés organiques du phosphore , Inhibiteurs de protéines kinases , Protéines proto-oncogènes , Pyrimidines , Récepteurs à activité tyrosine kinase , Animaux , Femelle , Souris , Acrylamides/pharmacologie , Acrylamides/usage thérapeutique , Dérivés de l'aniline/pharmacologie , Dérivés de l'aniline/usage thérapeutique , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/métabolisme , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Récepteurs ErbB/antagonistes et inhibiteurs , Récepteurs ErbB/métabolisme , Indoles , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Souris de lignée BALB C , Mutation , Composés organiques du phosphore/pharmacologie , Composés organiques du phosphore/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/antagonistes et inhibiteurs , Pyrimidines/pharmacologie , Pyrimidines/usage thérapeutique , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Récepteurs à activité tyrosine kinase/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
19.
Eur J Med Chem ; 245(Pt 1): 114884, 2023 Jan 05.
Article de Anglais | MEDLINE | ID: mdl-36335744

RÉSUMÉ

Colony-Stimulating Factor-1 Receptor (CSF1R) is a receptor tyrosine kinase that controls the differentiation and maintenance of most tissue-resident macrophages and bone-resorbing osteoclasts. Mutations of CSF1R have been implicated in neurodegeneration, skeletal anomalies, and cancers. Activation of CSF1R by endogenous cytokine ligation to the ectodomain triggers the autophosphorylation of the intracellular tyrosine kinase domain, and thereafter, activation of several downstream pro-survival kinase cascades, including PI3K, ERK1/2, and JNK. The immunological role of CSF1R in regulating tumor-associate macrophages (TAMs) have been well-documented. TAMs harboring activated CSF1R release tumorigenic cytokines, which further deconditioning tumor microenvironment to a protumoral phenotype. Pharmacological inhibition of CSF1R has emerged as a promising antitumor strategy, with PLX3397 (pexidartinib) been approved by the FDA for the treatment of tenosynovial giant cell tumor in 2019. Research around developing novel small-molecule CSF1R inhibitors, as well as expanding their potential indications, have drawn numerous attentions thenceforward. Herein, we've comprehensively reviewed the latest progression of CSF1R inhibitors under clinical and preclinical studies. Key findings of CSF1R targeted therapies either as monotherapy or combinatorial therapy have also been discussed.


Sujet(s)
Antinéoplasiques , Immunothérapie , Tumeurs , Récepteurs à activité tyrosine kinase , Récepteur de facteur de croissance granulocyte-macrophage , Humains , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Cytokines/métabolisme , Immunothérapie/méthodes , Tumeurs/thérapie , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Récepteur de facteur de croissance granulocyte-macrophage/antagonistes et inhibiteurs , Microenvironnement tumoral/effets des médicaments et des substances chimiques , /pharmacologie , /usage thérapeutique
20.
Bioorg Chem ; 130: 106215, 2023 01.
Article de Anglais | MEDLINE | ID: mdl-36384067

RÉSUMÉ

Discoidin domain receptors (DDRs) are one of the less explored targets for the treatment of cancer which belong to receptor tyrosine kinases family. Discoidin domain receptors (DDRs) are a collagen-activated receptor tyrosine kinase and essential for controlling cellular functions like proliferation, morphogenesis, adhesion, differentiation, invasion, matrix remodeling, and migration. Although there are many targets and their inhibitors are reported which treat cancer. But most of drugs were amalgamated with moderate to severe side effects. This results in untreated cancerous cells. One of the reasons that cancer is considered challenging to treat because the targets were mutating rapidly and the inhibitor become less potent. The target identification is a tedious task for the researchers from the early 1990 s till date. When it comes to cancer, there has not been any magical stick to treat it undisputedly. Therefore, need for discovery of new receptor may helpful to overcome these difficulties. The development of DDR inhibitors has received a lot of attention ever since the target was discovered. In this review we have reported the development of most promising DDR1 and DDR2 small molecule inhibitors from the perspective of medicinal chemistry. We have also discussed about the clinical trials, recent patents, selectivity biological activity, and structure-activity relationship (SAR) of DDR1 and DDR2 inhibitors.


Sujet(s)
Antinéoplasiques , Récepteurs à domaine discoïdine , Humains , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Récepteurs à domaine discoïdine/antagonistes et inhibiteurs , Tumeurs/traitement médicamenteux , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Récepteurs à activité tyrosine kinase/métabolisme , Récepteur mitogène/composition chimique , Relation structure-activité
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...