RESUMEN
Moving towards high-grade glioma drug discovery, this study aimed to detect the mechanism of cellular death (apoptosis, necrosis and/or autophagy) induced by three carboranyl-based lead compounds. For that, we performed in U87 MG cells, flow cytometry experiments, as the gold standard technique, as well as confocal microscopy and 1 H-NMR experiments as non-invasive assays. We selected three hybrid leads (1-3) from the in-house-library and the corresponding parent compounds, and recognized tyrosine kinase inhibitors (lapatinib, sunitinib and erlotinib) to put to the test in these experiments. Flow cytometry with Annexin V-FITC/DAPI staining showed that leads 1 and 3 and lapatinib mainly induced necrosis in U87 MG upon a 24 h treatment at IC50 dose; meanwhile, hybrid 2, sunitinib and erlotinib seem to induce apoptosis in such cells. In general, confocal microscopy studies were in agreement with flow cytometry observing loss of cell membrane integrity in necrotic cells and features of apoptosis, that is, chromatin condensation, in apoptotic cells. Finally, NMR results showed that glioblastoma cells treated with hybrid 1, 3 or lapatinib displayed changes in CH2 /CH3 signal ratio and choline signals that could indicate necrotic cell death mechanism: meanwhile, 2-, sunitinib- or erlotinib-treated cells showed apoptotic characteristic behaviors. Additionally, carboranyl-hybrid 2 also produced autophagy in U87 MG cells.
Asunto(s)
Antineoplásicos , Glioblastoma , Humanos , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Lapatinib/farmacología , Lapatinib/uso terapéutico , Sunitinib , Clorhidrato de Erlotinib/farmacología , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis , Necrosis/tratamiento farmacológico , Proliferación CelularRESUMEN
The involvement of LncRNA SOX2-overlapping transcript (SOX2-OT), SOX2, and GLI-1 transcription factors in cancer has been well documented. Nonetheless, it is still unknown whether co-expressed SOX2-OT/SOX2 or SOX2-OT/SOX2/GLI-1 axes are epigenetically/transcriptionally involved in terms of resistance to oncology therapy and in poorer clinical outcomes for patients with lung cancer. We evaluated the role of SOX2-OT/SOX2 and SOX2-OT/SOX2/GLI-1 axes using RT-qPCR, western blot, immunofluorescence analyses, gene silencing, cellular cytotoxic, and ChIP-qPCR assays on human cell lines, solid lung malignant tumors, and normal lung tissue. We detected that the SOX2-OT/SOX2/GLI-1 axis promotes resistance to tyrosine kinase inhibitor (TKI)-erlotinib and cisplatin-based therapy. Evidence from this study show that SOX2-OT modulates the expression/activation of EGFR-pathway members AKT/ERK. Further, both SOX2-OT and GLI-1 genes are epigenetically regulated at their promoter sequences, in an LncRNA SOX2-OT-dependent manner, mainly through modifying the enrichment of the activation histone mark H3K4me3/H3K27Ac, versus the repressive histone mark H3K9me3/H3K27me3. In addition, we identified that inhibition of SOX2-OT and reduced expression of SOX2/GLI-1 sensitizes lung cancer cells to EGFR/TKI-erlotinib or cisplatin-based treatment. Finally, we show that high co-expression of SOX2-OT/SOX2 transcripts and SOX2/GLI-1 proteins appears to correlate with a poor clinical prognosis and lung malignant phenotype. Collectively, these results present evidence that LncRNA SOX2-OT modulates an orchestrated resistance mechanism, promoting poor prognosis and human lung malignancy through genetic, epigenetic, and post-translational mechanisms.
Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/genética , Neoplasias Pulmonares/genética , Proteínas Proto-Oncogénicas c-akt/genética , ARN Largo no Codificante/genética , Factores de Transcripción SOXB1/genética , Proteína con Dedos de Zinc GLI1/genética , Línea Celular Tumoral , Cisplatino/farmacología , Resistencia a Antineoplásicos , Epigénesis Genética , Clorhidrato de Erlotinib/farmacología , Histonas , Humanos , Neoplasias Pulmonares/diagnóstico , PronósticoRESUMEN
Lung cancer is the most frequent type of cancer and the leading cause of cancer-related mortality worldwide. This study aimed to develop erlotinib (ELB)-loaded poly(ε-caprolactone) nanocapsules (NCELB) and evaluated their in vitro cytotoxicity in A549 cells. The formulation was characterized in relation to hydrodynamic diameter (171 nm), polydispersity index (0.076), zeta potential (- 8 mV), drug content (0.5 mg.mL-1), encapsulation efficiency (99%), and pH (6.0). NCELB presented higher cytotoxicity than ELB in solution against A549 cells in the MTT and LIVE/DEAD cell viability assays after 24 h of treatment. The main mechanism of cytotoxicity of NCELB was the induction of apoptosis in A549 cells. Further, a significant decrease in A549 colony formation was verified after NCELB treatment in comparison with the unencapsulated drug treatment. The reduction in clonogenic capacity is very relevant as it can reduce the risk of tumor recurrence and metastasis. In conclusion, erlotinib-loaded PCL nanocapsules are promising nanoparticles carriers to increase the efficacy of ELB in lung cancer treatment.
Asunto(s)
Antineoplásicos/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Clorhidrato de Erlotinib/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Poliésteres/química , Células A549 , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Clorhidrato de Erlotinib/química , Clorhidrato de Erlotinib/farmacología , Humanos , Nanocápsulas/química , Nanopartículas/químicaRESUMEN
Nitric oxide-releasing aspirins (NO-aspirins) are aspirin derivatives that are safer than the parent drug in the gastrointestinal context and have shown superior cytotoxic effects in several cancer models. Despite the rationale for their design, the influence of nitric oxide (NOâ¢) on the effects of NO-aspirins has been queried. Moreover, different isomers exhibit varying antitumor activity, apparently related to their ability to release NOâ¢. Here, we investigated the effects and mode of action of NO-aspirins in non-small-cell lung cancer (NSCLC) cells, comparing two isomers, NCX4016 and NCX4040 (-meta and -para isomers, respectively). NCX4040 was more potent in decreasing NSCLC cell viability and migration and exhibited significant synergistic effects in combination with erlotinib (an epidermal growth factor receptor inhibitor) in erlotinib-resistant cells. We also studied the relationship among the effects of NO-aspirins, NO⢠release, and PGE2 levels. NCX4040 released more NO⢠and significantly decreased PGE2 synthesis relative to NCX4016; however, NO⢠scavenger treatment reversed the antiproliferative effects of NCX4016, but not those of NCX4040. By contrast, misoprostol (a PGE2 receptor agonist) significantly reversed the antiproliferative effect of NCX4040, but not those of NCX4016. Furthermore, misoprostol reversed the antimigratory effects of NCX4040. Overall, these results indicate that PGE2 inhibition is important in the mode of action of NO-aspirins.
Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Aspirina/farmacología , Inhibidores de la Ciclooxigenasa/farmacología , Óxido Nítrico/metabolismo , Antiinflamatorios no Esteroideos/química , Aspirina/análogos & derivados , Aspirina/química , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Inhibidores de la Ciclooxigenasa/química , Dinoprostona/biosíntesis , Sinergismo Farmacológico , Clorhidrato de Erlotinib/farmacología , Humanos , Neoplasias Pulmonares/metabolismo , Estructura Molecular , Nitrocompuestos/farmacologíaRESUMEN
BACKGROUND: Lung cancer harboring epidermal growth factor receptor (EGFR) mutations and treated with EGFR tyrosine kinase inhibitors (TKIs) all eventually develop acquired resistance to the treatment, with half of the patients developing EGFR T790M resistance mutations. OBJECTIVE: The purpose of this study was to assess histological and clinical characteristics and survival outcomes in Hispanic EGFR mutated lung cancer patients after disease progression. PATIENTS AND METHODS: EGFR mutation-positive lung cancer patients (n = 34) with acquired resistance to the EGFR-TKI erlotinib were identified from 2011 to 2015. Post-progression tumor specimens were collected for molecular analysis. Post-progression interventions, response to treatment, and survival were assessed and compared among all patients and those with and without T790M mutations. RESULTS: Mean age was 59.4 ± 13.9 years, 65% were never-smokers, and 53% had a performance status 0-1. All patients received erlotinib as first-line treatment. Identified mutations included: 60% DelE19 (Del746-750) and 40% L858R. First-line erlotinib overall response rate (ORR) was 61.8% and progression free survival (PFS) was 16.8 months (95% CI: 13.7-19.9). Acquired resistance mutations identified were T790M mutation (47.1%); PI3K mutations (14.7%); EGFR amplification (14.7%); KRAS mutation (5.9%); MET amplification (8.8%); HER2 alterations (5.9%, deletions/insertions in e20); and SCLC transformation (2.9%). Of patients, 79.4% received treatment after progression. ORR for post-erlotinib treatment was 47.1% (CR 2/PR 14) and median PFS was 8.3 months (95% CI: 2.2-36.6). Median overall survival (OS) from treatment initiation was 32.9 months (95% CI: 30.4-35.3), and only the use of post-progression therapy affected OS in a multivariate analysis (p = 0.05). CONCLUSIONS: Hispanic patients with acquired resistance to erlotinib continued to be sensitive to other treatments after progression. The proportion of T790M+ patients appears to be similar to that previously reported in Caucasians.
Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/etnología , Antineoplásicos/uso terapéutico , Receptores ErbB/genética , Clorhidrato de Erlotinib/uso terapéutico , Hispánicos o Latinos/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/etnología , Mutación , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Antineoplásicos/farmacología , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Clorhidrato de Erlotinib/farmacología , Femenino , Humanos , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana EdadRESUMEN
We investigated the effects of kinase insert domain receptor (KDR) gene silencing on the proliferation of A549 cells and their sensitivity to erlotinib. A KDR small interfering RNA (siRNA) sequence was designed and synthesized; then, it was transfected into A549 cells using Lipofectamine(TM) 2000. KDR mRNA and protein expression after KDR gene silencing was detected by reverse transcription polymerase chain reaction and western blotting; the A549 cell cycle was detected by flow cytometry. A 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and colony formation assay were performed to determine the sensitivity of A549 cells to erlotinib after KDR gene silencing. After 48h of KDR gene silencing, there was a significant decrease in KDR gene and protein expression (P < 0.05). The A549 cell cycle was arrested at the G0/G1 phase, and the number of cells in the S phase decreased; the difference was statistically significant (P < 0.05). In the KDR gene silencing group, the sensitivity of A549 cells to erlotinib was significantly enhanced (P < 0.05). KDR siRNA can significantly silence the KDR gene in A549 cells, inhibit the proliferation of A549 cells, and enhance their sensitivity to erlotinib.
Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Clorhidrato de Erlotinib/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Células A549 , Adenocarcinoma del Pulmón , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Silenciador del Gen , Humanos , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Sales de Tetrazolio , Tiazoles , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismoRESUMEN
We examined the effects and molecular mechanism of the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor erlotinib on NKG2D ligand expression in human lung adenocarcinoma A549 cells and the cytotoxicity of cytokine-induced killer cells. Flow cytometry was used to detect NKG2D ligand expression in A549 cells under effects of erlotinib and EGFR downstream molecules, including LY294002 (phosphoinositide 3-kinase inhibitor), SB203580 (mitogen-activated protein kinase inhibitor), and STAT21 (signal transduction and transcription 3 inhibitor) after 24 h. A lactate dehydrogenase release assay was used to detect, at different effector-to-target ratios, the A549 cell killing activity of cytokine-induced killer cells before and after treatment with 10 mM erlotinib. Erlotinib suppressed MICA expression in A549 cells and upregulated MICB and UL16 binding protein 1 expression. EGFR downstream molecules mitogen-activated protein kinase and signal transduction and transcription 3 inhibitor did not affect the expression of NKG2D ligands in A549 cells. The phosphoinositide 3-kinase inhibitor reduced MICA expression in A549 cells, while erlotinib enhanced the killing sensitivity of cytokine-induced killer cells in A549 cells. The anti-lung carcinoma effects of EGFR tyrosine kinase inhibitor were associated with the sensitivity of lung cancer cells to enhanced immune cell killing.