Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 9 de 9
Filtrer
Plus de filtres










Gamme d'année
1.
Acta Pharmacol Sin ; 45(6): 1252-1263, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38360931

RÉSUMÉ

Although ALK tyrosine kinase inhibitors (ALK-TKIs) have shown remarkable benefits in EML4-ALK positive NSCLC patients compared to conventional chemotherapy, the optimal sequence of ALK-TKIs treatment remains unclear due to the emergence of primary and acquired resistance and the lack of potential prognostic biomarkers. In this study, we systematically explored the validity of sequential ALK inhibitors (alectinib, lorlatinib, crizotinib, ceritinib and brigatinib) for a heavy-treated patient with EML4-ALK fusion via developing an in vitro and in vivo drug testing system based on patient-derived models. Based on the patient-derived models and clinical responses of the patient, we found that crizotinib might inhibit proliferation of EML4-ALK positive tumors resistant to alectinib and lorlatinib. In addition, NSCLC patients harboring the G1269A mutation, which was identified in alectinib, lorlatinib and crizotinib-resistant NSCLC, showed responsiveness to brigatinib and ceritinib. Transcriptomic analysis revealed that brigatinib suppressed the activation of multiple inflammatory signaling pathways, potentially contributing to its anti-tumor activity. Moreover, we constructed a prognostic model based on the expression of IL6, CXCL1, and CXCL5, providing novel perspectives for predicting prognosis in EML4-ALK positive NSCLC patients. In summary, our results delineate clinical responses of sequential ALK-TKIs treatments and provide insights into the mechanisms underlying the superior effects of brigatinib in patients harboring ALKG1269A mutation and resistant towards alectinib, lorlatinib and crizotinib. The molecular signatures model based on the combination of IL6, CXCL1 and CXCL5 has the potential to predict prognosis of EML4-ALK positive NSCLC patients.


Sujet(s)
Adénocarcinome pulmonaire , Antinéoplasiques , Tumeurs du poumon , Protéines de fusion oncogènes , Composés organiques du phosphore , Inhibiteurs de protéines kinases , Pyrimidines , Humains , Composés organiques du phosphore/usage thérapeutique , Composés organiques du phosphore/pharmacologie , Pyrimidines/usage thérapeutique , Pyrimidines/pharmacologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Protéines de fusion oncogènes/génétique , Protéines de fusion oncogènes/métabolisme , Animaux , Adénocarcinome pulmonaire/traitement médicamenteux , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Pronostic , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Résistance aux médicaments antinéoplasiques , Lactames/usage thérapeutique , Carbazoles/usage thérapeutique , Carbazoles/pharmacologie , Sulfones/usage thérapeutique , Sulfones/pharmacologie , Crizotinib/usage thérapeutique , Crizotinib/pharmacologie , Lignée cellulaire tumorale , Pipéridines/usage thérapeutique , Pipéridines/pharmacologie , Femelle , Souris , Inflammation/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Pyrazoles/usage thérapeutique , Pyrazoles/pharmacologie , Mâle , Kinase du lymphome anaplasique/génétique , Kinase du lymphome anaplasique/antagonistes et inhibiteurs , Kinase du lymphome anaplasique/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Mutation , Aminopyridines/usage thérapeutique , Aminopyridines/pharmacologie
2.
Cancer Res ; 78(14): 3995-4006, 2018 07 15.
Article de Anglais | MEDLINE | ID: mdl-29669759

RÉSUMÉ

Given that Yes-associated protein (YAP) signaling acts as a critical survival input for hypoxic cancer cells in hepatocellular carcinoma (HCC), disruption of YAP function and the maintenance of hypoxia is an attractive way to treat HCC. Utilizing a cell-based YAP-TEAD luciferase reporter assay and functional analyses, we identified CT-707, a China-FDA approved multi-kinase inhibitor under clinical trial with remarkable inhibitory activity against YAP function. CT-707 exhibited prominent cytotoxicity under hypoxia on HCC cells, which was attributable to the inhibition of YAP signaling. CT-707 arrested tumor growth in HepG2, Bel-7402, and HCC patient-derived xenografts. Mechanistically, the inhibitory activity of CT-707 on YAP signaling was due to the interruption of hypoxia-activated IGF1R. Overall, these findings not only identify CT-707 as a promising hypoxia-targeting agent against HCC, but they also unveil IGF1R as a new modulator specifically regulating hypoxia-activated YAP signaling.Significance: CT-707 may represent a novel clinical approach for patients with HCC suffering poor drug response due to intratumor hypoxia. Cancer Res; 78(14); 3995-4006. ©2018 AACR.


Sujet(s)
Hypoxie/traitement médicamenteux , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/métabolisme , Protéines nucléaires/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Récepteur IGF de type 1/métabolisme , Facteurs de transcription/métabolisme , Animaux , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/métabolisme , Protéines du cycle cellulaire , Lignée cellulaire tumorale , Chine , Cytotoxines/pharmacologie , Cellules HepG2 , Humains , Hypoxie/métabolisme , Souris , Souris nude , Transduction du signal/effets des médicaments et des substances chimiques
3.
Cancer Lett ; 412: 1-9, 2018 01 01.
Article de Anglais | MEDLINE | ID: mdl-29024815

RÉSUMÉ

Inhibition of autophagy is a promising strategy for non-small cell lung cancer (NSCLC) treatment, which is in the clinical trials. However, only chloroquine is used in clinic as an autophagic inhibitor and the inhibitory effect of chloroquine on autophagy is finite. Therefore, the development of an alternative autophagic inhibitor for NSCLC therapy becomes necessary. In the present study, cepharanthine (CEP), an alkaloid extracted from Stephania cepharantha Hayata, was identified as a novel autophagic inhibitor in NSCLC cells. The potential mechanism of the CEP-inhibited autophagy was by blockage of autophagosome-lysosome fusion and inhibition of lysosomal cathepsin B and cathepsin D maturation. Furthermore, we found for the first time that dacomitinib (DAC), a second-generation epidermal growth factor receptor inhibitor that in the phase III clinical trials for NSCLC treatment, induced a protective autophagy to decrease its anti-cancer effect. Combined treatment with CEP increased the anti-proliferative and apoptotic effects of DAC in vitro and enhanced the anti-cancer effect of DAC in NCI-H1975 xenograft mice. Collectively, CEP might be further developed as an autophagic inhibitor, and combined treatment of CEP and DAC could offer an effective strategy for NSCLC treatment.


Sujet(s)
Antinéoplasiques/pharmacologie , Autophagie/effets des médicaments et des substances chimiques , Benzylisoquinoléines/pharmacologie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Tumeurs du poumon/traitement médicamenteux , Quinazolinones/pharmacologie , Animaux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Cathepsines/métabolisme , Lignée cellulaire tumorale , Récepteurs ErbB/antagonistes et inhibiteurs , Humains , Concentration en ions d'hydrogène , Tumeurs du poumon/anatomopathologie , Souris
4.
Toxicol Appl Pharmacol ; 330: 65-73, 2017 09 01.
Article de Anglais | MEDLINE | ID: mdl-28711525

RÉSUMÉ

Heat shock protein 90 (Hsp90) is a critically conserved molecular chaperone protein and promising therapeutic target for cancer treatment. In this study, platycodin D (PD), a saponin isolated from traditional Chinese herb Platycodonis Radix, was identified as a novel Hsp90 inhibitor. We verified that PD did not affect the ATPase activity of Hsp90. However, PD disrupted the co-chaperone interaction of Hsp90/cell division cycle protein 37 (Cdc37) and subsequently degraded multiple Hsp90 client proteins without the feedback increase of Hsp70. In different genotypes of non-small cell lung cancer cells, co-treatment with the mTOR inhibitor Everolimus and PD enhanced antiproliferation activity and apoptotic effect. The feedback survival signal upon mTOR inhibition was fully terminated by the co-administration with PD through reduced epidermal growth factor receptor (EGFR) and insulin growth factor 1 receptor (IGF1R) expression, suppressed AKT activity, and reinforced 4E-BP1 inhibition. Our results not only identified PD as a novel Hsp90 inhibitor by disrupting the protein-protein interaction of Hsp90/Cdc37 complex, but also provided mechanistic insights into the ineffectiveness of mTOR inhibitors and identified therapeutic strategy for cancer treatment.


Sujet(s)
Antinéoplasiques/pharmacologie , Protéines du cycle cellulaire/effets des médicaments et des substances chimiques , Chaperonines/effets des médicaments et des substances chimiques , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Saponines/toxicité , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs , Triterpènes/toxicité , Apoptose/effets des médicaments et des substances chimiques , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Récepteurs ErbB/antagonistes et inhibiteurs , Évérolimus/pharmacologie , Humains , Immunosuppresseurs/pharmacologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Protéine oncogène v-akt/antagonistes et inhibiteurs , Récepteur IGF de type 1 , Récepteurs des somatomédines/antagonistes et inhibiteurs
5.
Article de Chinois | WPRIM (Pacifique Occidental) | ID: wpr-666539

RÉSUMÉ

OBJECTIVE Identification of novel autophagy inhibitors for the combinational treatment of non-small cell lung cancer (NSCLC). METHODS MTT assay and annexin V/PI staining assay were used to evaluate the cell proliferation and apoptosis, respectively. Immunofluorescence staining and cathepsin activity assay were used to detect autophagy. Small interfering RNA was performed to silence the genes and Western blot assay was used to evaluate the protein express levels. Xenograft experiments were applied for in vivo evaluation. RESULTS Cepharanthine, a natural compound, increased LC3-II expression and GFP-LC3 puncta formation in NSCLC NCI-H1975 cells. Numerous yellow puncta were observed in cepharanthine- treated cells with mRFP- EGFP- LC3 transfection. Co-staining of GFP-LC3 with LysoTracker red or LAMP1 antibody suggested that cepharanthine inhibits autophagosomes- lysosomes fusion. Moreover, cepharanthine attenuated the lysosomal cathepsins maturation. We also confirmed that dacomitinib induced cytoprotective autophagy. Combined treatment with cepharanthine increased the anti- cancer effects of dacomitinib in vitro and in vivo. Besides, cepharanthine could not enhance the anti-cancer effect of dacomitinib in autophagy deficient cells. CONCLUSION Cepharanthine might be further developed as a promising autophagic inhibitor, and combined treatment cepharanthine with dacomitinib could pose as an effective strategy for NSCLC treatment.

6.
Mol Cancer Ther ; 15(12): 2916-2925, 2016 12.
Article de Anglais | MEDLINE | ID: mdl-27638856

RÉSUMÉ

Hepatocellular carcinoma is among the leading causes of cancer-related deaths worldwide, and the development of new treatment regimens is urgently needed to improve therapeutic approach. In our study, we found that the combination of a Met inhibitor, cabozantinib, and a novel FAK inhibitor, CT-707, exerted synergistic antitumor effects against hepatocellular carcinoma in vitro and in vivo Interestingly, further studies showed that therapeutic concentrations of cabozantinib increased the phosphorylation of FAK, which might attenuate the antitumor activity of cabozantinib. The simultaneous exposure to CT-707 effectively inhibited the activation of FAK that was induced by cabozantinib, which contributes to the synergistic effect of the combination. Furthermore, cabozantinib increased the mRNA and protein levels of integrin α5, which is a canonical upstream of FAK, and the introduction of cilengitide to block integrin function could abrogate FAK activation by cabozantinib, indicating that cabozantinib upregulated the phosphorylation of FAK in an integrin-dependent manner. Similar synergy was also observed on PHA-665752, another selective MET inhibitor, indicating that this observation might be a common characteristic of MET-targeting strategies. Our findings not only favor the development of the novel FAK inhibitor CT-707 as a therapeutic agent against hepatocellular carcinoma but also provide a new strategy of combining MET and FAK inhibitors to potentiate the anticancer activities of these two types of agents for treating hepatocellular carcinoma patients. Mol Cancer Ther; 15(12); 2916-25. ©2016 AACR.


Sujet(s)
Anilides/pharmacologie , Carcinome hépatocellulaire/métabolisme , Focal adhesion kinase 1/antagonistes et inhibiteurs , Tumeurs du foie/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Pyridines/pharmacologie , Anilides/composition chimique , Animaux , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/anatomopathologie , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Synergie des médicaments , Activation enzymatique/effets des médicaments et des substances chimiques , Humains , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/anatomopathologie , Phosphorylation , Inhibiteurs de protéines kinases/composition chimique , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-met/antagonistes et inhibiteurs , Protéines proto-oncogènes c-met/métabolisme , Pyridines/composition chimique , Transduction du signal/effets des médicaments et des substances chimiques , Charge tumorale/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
7.
Oncotarget ; 7(6): 6933-47, 2016 Feb 09.
Article de Anglais | MEDLINE | ID: mdl-26771844

RÉSUMÉ

Although hypoxia is a prominent feature contributing to the therapeutic resistance of hepatocellular carcinoma cells (HCC) against chemotherapeutic agents, including the Topoisomerase I inhibitor SN38, the underlying mechanism is not fully understood and its understanding remains a major clinical challenge. In the present study, we found that hypoxia-induced nuclear translocation and accumulation of YAP acted as a survival input to promote resistance to SN38 in HCC. The induction of YAP by hypoxia was not mediated by HIF-1α because manipulating the abundance of HIF-1α with CoCl2, exogenous expression, and RNA interference had no effect on the phosphorylation or total levels of YAP. The mevalonate-HMG-CoA reductase (HMGCR) pathway may modulate the YAP activation under hypoxia. Combined YAP inhibition using either siRNA or the HMGCR inhibitor statins together with SN38 treatment produced improved anti-cancer effects in HCC cells. The increased anti-cancer effect of the combined treatment with statins and irinotecan (the prodrug of SN-38) was further validated in a human HepG2 xenograft model of HCC in nude mice. Taken together, our findings identify YAP as a novel mediator of hypoxic-resistance to SN38. These results suggest that the administration of SN28 together with the suppression of YAP using statins is a promising strategy for enhancing the treatment response in HCC patients, particularly in advanced stage HCC cases presenting hypoxic resistance.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Camptothécine/analogues et dérivés , Carcinome hépatocellulaire/anatomopathologie , Noyau de la cellule/métabolisme , Résistance aux médicaments antinéoplasiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Hypoxie/physiopathologie , Tumeurs du foie/anatomopathologie , Phosphoprotéines/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Animaux , Antinéoplasiques d'origine végétale/pharmacologie , Apoptose , Technique de Western , Camptothécine/pharmacologie , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/métabolisme , Prolifération cellulaire , Technique d'immunofluorescence , Humains , Hypoxie/complications , Techniques immunoenzymatiques , Irinotécan , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/métabolisme , Souris , Souris nude , Phosphoprotéines/génétique , Transport des protéines , ARN messager/génétique , Réaction de polymérisation en chaine en temps réel , RT-PCR , Transduction du signal/effets des médicaments et des substances chimiques , Facteurs de transcription , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe , Protéines de signalisation YAP
8.
Carbohydr Polym ; 89(1): 89-97, 2012 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-24750608

RÉSUMÉ

GA-13315 (13-chlorine-3,15-dioxy-gibberllic acid methyl ester) was semi-synthesized by GA3 (gibberellic acid) as a potential anticancer drug. To pursue its promising application, cyclodextrin was used for forming complexes to overcome its drawbacks such as poor water solubility and stability. So, GA-13315/CD complexes were prepared with native ß-cyclodextrin and its derivatives (hydroxypropyl-ß-cyclodextrin (HPßCD)) and their inclusion complexation behavior, characterization and binding ability in both solution and the solid state was studied by means of UV, XRD, DSC, SEM, (1)H and 2D NMR spectroscopy. Furthermore, preliminary in vitro cytotoxicity assay showed that the complexes still maintain antitumor activities, compared with GA-13315 or adriamycin (ADM, positive control) as the positive control. The results showed that the water solubility and stability of GA-13315 were obviously improved in the inclusion complex with cyclodextrins, suggesting the inclusion complexes as promising future therapeutic agents.


Sujet(s)
Antinéoplasiques , Systèmes de délivrance de médicaments , Gibbérellines , Antinéoplasiques/administration et posologie , Antinéoplasiques/composition chimique , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Cyclodextrines/composition chimique , Préparation de médicament , Stabilité de médicament , Gibbérellines/administration et posologie , Gibbérellines/composition chimique , Humains , Solubilité
9.
Eur J Med Chem ; 46(4): 1172-80, 2011 Apr.
Article de Anglais | MEDLINE | ID: mdl-21316818

RÉSUMÉ

A series of polyhalo 1,3-diazaheterocycle fused isoquinolin-1(2H)-imines were evaluated in vitro against human tumour cell lines including A431, K562, HL60, HepG2 and Skov-3. As a result, some of the target compounds such as 5b, 5c, 5i, 5o, 6c, 6h and 7f showed stronger cytotoxicity against K562, H562 and Skov-3 cells in comparison with cisplatin, and the others displayed moderate cytotoxicity to A431 and HepG2. Biological investigations using the representative compounds 5c, 6c and 6h were also performed in mice bearing S(180) and H(22) tumours. The results indicated that these three compounds inhibit S(180) and H(22) growth. In addition, compounds 6c and 6h have very low acute toxicities. The preliminary analysis of structure-activity relationships is also discussed.


Sujet(s)
Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Halogènes/composition chimique , Isoquinoléines/composition chimique , Isoquinoléines/pharmacologie , Animaux , Antinéoplasiques/synthèse chimique , Benzène/composition chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Humains , Concentration inhibitrice 50 , Isoquinoléines/synthèse chimique , Souris , Relation structure-activité , Tests d'activité antitumorale sur modèle de xénogreffe
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...