Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 116
Filtrer
1.
Sci Rep ; 14(1): 16803, 2024 07 22.
Article de Anglais | MEDLINE | ID: mdl-39039104

RÉSUMÉ

The success of chemotherapy regimens in patients with non-small cell lung cancer (NSCLC) could be restricted at least in part by cancer stem cells (CSC) niches within the tumor microenvironment (TME). CSC express CD133, CD44, CD47, and SOX2, among other markers and factors. Analysis of public data revealed that high expression of hyaluronan (HA), the main glycosaminoglycan of TME, correlated positively with CSC phenotype and decreased disease-free interval in NSCLC patients. We aimed to cross-validate these findings on human and murine lung cancer cells and observed that CD133 + CSC differentially expressed higher levels of HA, HAS3, ABCC5, SOX2, and CD47 (p < 0.01). We modulated HA expression with 4-methylumbelliferone (4Mu) and detected an increase in sensitivity to paclitaxel (Pa). We evaluated the effect of 4Mu + chemotherapy on survival, HA metabolism, and CSC profile. The combination of 4Mu with Pa reduced the clonogenic and tumor-forming ability of CSC. Pa-induced HAS3, ABCC5, SOX2, and CD47 expression was mitigated by 4Mu. Pa + 4Mu combination significantly reduced in vivo tumor growth, enhancing animal survival and restoring the CSC profile in the TME to basal levels. Our results suggest that HA is involved in lung CSC phenotype and chemosensitivity, and its modulation by 4Mu improves treatment efficacy to inhibit tumor progression.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Résistance aux médicaments antinéoplasiques , Acide hyaluronique , Hymécromone , Tumeurs du poumon , Cellules souches tumorales , Paclitaxel , Microenvironnement tumoral , Acide hyaluronique/métabolisme , Cellules souches tumorales/métabolisme , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/anatomopathologie , Humains , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Animaux , Souris , Paclitaxel/pharmacologie , Paclitaxel/usage thérapeutique , Hymécromone/pharmacologie , Lignée cellulaire tumorale , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie
2.
Eur J Clin Invest ; 54(11): e14288, 2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-39058257

RÉSUMÉ

BACKGROUND: Low physical performance is associated with higher mortality rate in multiple pathological conditions. Here, we aimed to determine whether body composition and physical performance could be prognostic factors in non-small cell lung cancer (NSCLC) patients. Moreover, we performed an exploratory approach to determine whether plasma samples from NSCLC patients could directly affect metabolic and structural phenotypes in primary muscle cells. METHODS: This prospective cohort study included 55 metastatic NSCLC patients and seven age-matched control subjects. Assessments included physical performance, body composition, quality of life and overall survival rate. Plasma samples from a sub cohort of 18 patients were collected for exploratory studies in cell culture and metabolomic analysis. RESULTS: We observed a higher survival rate in NSCLC patients with high performance in the timed up-and-go (+320%; p = .007), sit-to-stand (+256%; p = .01) and six-minute walking (+323%; p = .002) tests when compared to NSCLC patients with low physical performance. There was no significant association for similar analysis with body composition measurements (p > .05). Primary human myotubes incubated with plasma from NSCLC patients with low physical performance had impaired oxygen consumption rate (-54.2%; p < .0001) and cell proliferation (-44.9%; p = .007). An unbiased metabolomic analysis revealed a list of specific metabolites differentially expressed in the plasma of NSCLC patients with low physical performance. CONCLUSION: These novel findings indicate that physical performance is a prognostic factor for overall survival in NSCLC patients and provide novel insights into circulating factors that could impair skeletal muscle metabolism.


Sujet(s)
Composition corporelle , Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Performance fonctionnelle physique , Humains , Tumeurs du poumon/métabolisme , Tumeurs du poumon/sang , Tumeurs du poumon/anatomopathologie , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/sang , Carcinome pulmonaire non à petites cellules/anatomopathologie , Mâle , Femelle , Adulte d'âge moyen , Pronostic , Sujet âgé , Études prospectives , Métabolome/physiologie , Études cas-témoins , Consommation d'oxygène/physiologie , Taux de survie , Qualité de vie , Fibres musculaires squelettiques/métabolisme , Prolifération cellulaire , Test de marche
3.
Clinics (Sao Paulo) ; 79: 100407, 2024.
Article de Anglais | MEDLINE | ID: mdl-38889502

RÉSUMÉ

BACKGROUND: NSCLC is one of the most common causes of death. The hypoxia microenvironment contributes to cancer progression. The purpose was to explore the effects and mechanism of melittin on NSCLC cells in the hypoxic microenvironment. METHODS: NSCLC cell lines (A549 and H1299) were cultured in normoxia or hypoxia conditions with or without melittin treatment. The viability of the cells was detected via MTT assay and the proliferation ability was evaluated by EdU assay. QRT-PCR was performed to evaluate GLUT1, LDHA, HK2, VEGF and LATS2 mRNA levels. Glucose transport was assessed by the 2-NBDG uptake assay. The angiogenesis was determined by the tubule formation assay. The protein expressions of GLUT1, LDHA, HK2, VEGF, LATS2, YAP, p-YAP and HIF-1α were detected via western blotting assay. The tumor formation assay was conducted to examine the roles of melittin and LATS2 in vivo. RESULTS: Melittin inhibited hypoxia-induced cell viability, proliferation, glycolysis and angiogenesis as well as suppressed YAP binding to HIF-1α in NSCLC. Melittin inactivated the YAP/HIF-1α pathway via up-regulation of LATS2, ultimately inhibiting cancer progression of NSCLC. Moreover, melittin suppressed tumor growth via up-regulation of LATS2 in vivo. CONCLUSION: Melittin inactivated the YAP/HIF-1α pathway via up-regulation of LATS2 to contribute to the development of NSCLC. Therefore, melittin is expected to become a potential prognostic drug for the therapy of NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Prolifération cellulaire , Glycolyse , Sous-unité alpha du facteur-1 induit par l'hypoxie , Tumeurs du poumon , Mélittine , Néovascularisation pathologique , Protein-Serine-Threonine Kinases , Protéines suppresseurs de tumeurs , Régulation positive , Protéines de signalisation YAP , Humains , Protein-Serine-Threonine Kinases/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/vascularisation , Carcinome pulmonaire non à petites cellules/anatomopathologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Régulation positive/effets des médicaments et des substances chimiques , Glycolyse/effets des médicaments et des substances chimiques , Protéines suppresseurs de tumeurs/métabolisme , Néovascularisation pathologique/traitement médicamenteux , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Protéines de signalisation YAP/métabolisme , Mélittine/pharmacologie , Mélittine/usage thérapeutique , Lignée cellulaire tumorale , Facteurs de transcription/métabolisme , Animaux , Protéines adaptatrices de la transduction du signal/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Phosphoprotéines/métabolisme , Angiogenesis
4.
Einstein (Sao Paulo) ; 22: eAO0575, 2024.
Article de Anglais | MEDLINE | ID: mdl-38922219

RÉSUMÉ

OBJECTIVE: Currently programmed cell death protein 1 (PD-1) inhibitors in combination with other therapies are being evaluated to determine their efficacy in cancer treatment. However, the effect of PD-ligand (L) 1 expression on disease outcomes in stage III (EC III) non-small cell lung cancer is not completely understood. Therefore, this study aimed to assess the influence of PD-L1 expression on the outcomes of EC III non-small cell lung cancer. METHODS: This study was conducted on patients diagnosed with EC III non-small cell lung cancer who underwent treatment at a tertiary care hospital. PD-L1 expression was determined using immunohistochemical staining, all patients expressed PD-L1. Survival was estimated using the Kaplan-Meier method. Relationships between variables were assessed using Cox proportional regression models. RESULTS: A total of 49 patients (median age=69 years) with EC III non-small cell lung cancer and PD-L1 expression were evaluated. More than half of the patients were men, and most were regular smokers. The patients were treated with neoadjuvant chemotherapy, surgery, or sequential or combined chemotherapy and radiotherapy. The median progression-free survival of the entire cohort was 14.2 months, and the median overall survival was 20 months. There was no significant association between PD-L1 expression and disease progression, clinical characteristics, or overall survival. CONCLUSIONS: PD-L1 expression was not correlated with EC III non-small cell lung cancer outcomes. Whether these findings differ from the association with immune checkpoint inhibitors remains to be addressed in future studies.


Sujet(s)
Antigène CD274 , Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Stadification tumorale , Humains , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/mortalité , Carcinome pulmonaire non à petites cellules/thérapie , Mâle , Femelle , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/mortalité , Antigène CD274/analyse , Antigène CD274/métabolisme , Études rétrospectives , Sujet âgé , Adulte d'âge moyen , Pronostic , Estimation de Kaplan-Meier , Immunohistochimie , Sujet âgé de 80 ans ou plus , Marqueurs biologiques tumoraux/analyse , Marqueurs biologiques tumoraux/métabolisme , Adulte
5.
Clin Transl Oncol ; 26(9): 2250-2261, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38554191

RÉSUMÉ

BACKGROUND: The objective of this research was to investigate how the combination of semen coicis extract and PD-1 inhibitors can potentially work together to enhance the anti-tumor effects, with a focus on understanding the underlying mechanism. METHODS: We obtained the active components and specific targets of semen coicis in the treatment of NSCLC from various databases, namely TCMSP, GeneCard, and OMIM. By utilizing the STRING database and Cytoscape software, we established a protein interaction network (PPI) for the active ingredient of semen coicis and the target genes related to NSCLC. To explore the potential pathways involved, we conducted gene ontology (GO) and biological pathway (KEGG) enrichment analyses, which were further supported by molecular docking technology. Additionally, we conducted cyto-inhibition experiments to verify the inhibitory effects of semen coicis alone or in combination with a PD-1 inhibitor on A549 cells, along with examining the associated pathways. Furthermore, we investigated the synergistic mechanism of these two drugs through cytokine release experiments and the PD-L1 expression study on A549 cells. RESULTS: Semen coicis contains two main active components, Omaine and (S)-4-Nonanolide. Its primary targets include PIK3R1, PIK3CD, PIK3CA, AKT2, and mTOR. Molecular docking experiments confirmed that these ingredients and targets form stable bonds. In vitro experiments showed that semen coicis demonstrates inhibitory effects against A549 cells, and this effect was further enhanced when combined with PD-1 inhibitors. PCR and WB analysis confirmed that the inhibition of the PI3K-AKT-mTOR pathway may contribute to this effect. Additionally, semen coicis was observed to decrease the levels of IFN-γ, IL-6, and TNF-α, promoting the recovery of the human anti-tumor immune response. And semen coicis could inhibit the induced expression of PD­L1 of A549 cells stimulated by IFN­Î³ as well. CONCLUSION: Semen coicis not only has the ability to kill tumor cells directly but also alleviates the immunosuppression found in the tumor microenvironment. Additionally, it collaboratively enhances the effectiveness of PD-1 inhibitors against tumors by blocking the activation of PI3K-AKT-mTOR.


Sujet(s)
Antinéoplasiques , Coix , Tumeurs du poumon , Récepteur-1 de mort cellulaire programmée , Transduction du signal , Humains , Cellules A549 , Antigène CD274/métabolisme , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Synergie des médicaments , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Simulation de docking moléculaire , Phosphatidylinositol 3-kinases/métabolisme , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Cartes d'interactions protéiques/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs , Coix/composition chimique , Antinéoplasiques/pharmacologie
6.
Clin Transl Oncol ; 26(4): 991-1000, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38030870

RÉSUMÉ

OBJECTIVES: The purpose of this meta-analysis was to investigate the relationship between serum carcinoembryonic antigen (CEA) expression and epidermal growth factor receptor (EGFR) mutation status in non-small cell lung cancer (NSCLC). METHODS: Databases such as PubMed, Cochrane, EMBASE and Google Scholar were systematically searched to identify studies assessing the association of serum CEA expression with EGFR mutations. Across 19 studies, 4168 patients were included between CEA expression and EGFR mutations odds ratio (OR) conjoint analysis of correlations. RESULTS: Compared with CEA-negative NSCLC, CEA-positive tumors had an increased EGFR mutation rate (OR = 1.85, 95% confidence interval: 1.48-2.32, P < 0.00001). This association was observed in both stage IIIB/IV patients (OR = 1.60, 95% CI: 1.18-2.15, P = 0.002) and stage I-IIIA (OR = 1.67, 95% CI: 1.01-2.77, P = 0.05) patients. In addition, CEA expression was associated with exon 19 (OR = 1.97, 95% CI: 1.25-3.11, P = 0.003) and exon 21 (OR = 1.51, 95% CI: 1.07-2.12, P = 0.02) EGFR mutations. In ADC pathological type had also showed the correlation (OR = 1.84, 95% CI: 1.31-2.57, P = 0.0004). CONCLUSIONS: This meta-analysis indicated that serum CEA expression was associated with EGFR mutations in NSCLC patients. The results of this study suggest that CEA level may play a predictive role in the EGFR mutation status of NSCLC patients. Detecting serum CEA expression levels can give a good suggestion to those patients who are confused about whether to undergo EGFR mutation tests. Moreover, it may help better plan of the follow-up treatment.


Sujet(s)
Antigène carcinoembryonnaire , Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Antigène carcinoembryonnaire/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Récepteurs ErbB/génétique , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Mutation , Inhibiteurs de protéines kinases
7.
Genes (Basel) ; 14(10)2023 10 04.
Article de Anglais | MEDLINE | ID: mdl-37895255

RÉSUMÉ

Lung cancer is a highly aggressive neoplasm and, despite the development of recent therapies, tumor progression and recurrence following the initial response remains unsolved. Several questions remain unanswered about non-small cell lung cancer (NSCLC): (1) Which patients will actually benefit from therapy? (2) What are the predictive factors of response to MAbs and TKIs? (3) What are the best combination strategies with conventional treatments or new antineoplastic drugs? To answer these questions, an integrative literature review was carried out, searching articles in PUBMED, NCBI-PMC, Google Academic, and others. Here, we will examine the molecular genetics of lung cancer, emphasizing NSCLC, and delineate the primary categories of inhibitors based on their molecular targets, alongside the main treatment alternatives depending on the type of acquired resistance. We highlighted new therapies based on epigenetic information and a single-cell approach as a potential source of new biomarkers. The current and future of NSCLC management hinges upon genotyping correct prognostic markers, as well as on the evolution of precision medicine, which guarantees a tailored drug combination with precise targeting.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Tumeurs du poumon/diagnostic , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Pronostic , Résistance aux médicaments antinéoplasiques/génétique , Récepteurs ErbB/génétique , Inhibiteurs de protéines kinases/pharmacologie , Mutation
8.
Int J Mol Sci ; 24(15)2023 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-37569262

RÉSUMÉ

Lung cancer has the highest mortality rate among all cancer types, resulting in over 1.8 million deaths annually. Immunotherapy utilizing immune checkpoint inhibitors (ICIs) has revolutionized the treatment of non-small cell lung cancer (NSCLC). ICIs, predominantly monoclonal antibodies, modulate co-stimulatory and co-inhibitory signals crucial for maintaining immune tolerance. Despite significant therapeutic advancements in NSCLC, patients still face challenges such as disease progression, recurrence, and high mortality rates. Therefore, there is a need for predictive biomarkers that can guide lung cancer treatment strategies. Currently, programmed death-ligand 1 (PD-L1) expression is the only established biomarker for predicting ICI response. However, its accuracy and robustness are not consistently reliable. This review provides an overview of potential biomarkers currently under development or in the validation stage that hold promise in improving the classification of responders and non-responders to ICI therapy in the near future.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Carcinome pulmonaire non à petites cellules/métabolisme , Tumeurs du poumon/métabolisme , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Récepteur-1 de mort cellulaire programmée , Marqueurs biologiques , Immunothérapie/méthodes , Antigène CD274 , Marqueurs biologiques tumoraux/métabolisme
9.
Clin Transl Oncol ; 24(9): 1661-1672, 2022 Sep.
Article de Anglais | MEDLINE | ID: mdl-35377059

RÉSUMÉ

The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway is recognized as a main mediator bridging innate and adaptive immunity, recent advances have expanded its roles to anti-tumor immunity and carcinogenesis. Loss of cGAS-STING signaling in non-small cell lung cancer (NSCLC) leads to enhanced tumorigenicity and decreased cytotoxic T lymphocyte infiltration. Apart from its anticancer response, persistent overreaction of cGAS-STING signaling promotes progression of certain inflammation-aggravated cancers. Activation of the pro-inflammatory nucleic acid sensing pathway can trigger Hippo pathway, which mediates the inactivation of Yes-associated protein 1 (YAP1) and its paralogue transcriptional co-regulators with PDZ-binding motif (TAZ, also known as WWTR1), and subsequent suppression of tumorigenesis. Active YAP acts as a transcriptional driver in bolstering immunosuppressive cytokines to evade immune surveillance and promote occurrence of preneoplasia. It is reasonable that aggressive tumors co-opt these regulators to generate few immunogenic antigens and drive tumorigenic behaviors via a highly cooperative manner. Given their multifaced roles, we profile the molecular biology characteristic and current status underpinning oncogenic YAP, review its crosstalk roles with cGAS/STING pathway in NSCLC, and summarize the major clinical investigations in NSCLC with TCGA database.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Protéines membranaires , Nucleotidyltransferases , Transduction du signal , Protéines de signalisation YAP , Carcinogenèse , Carcinome pulmonaire non à petites cellules/métabolisme , Humains , Tumeurs du poumon/métabolisme , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Nucleotidyltransferases/génétique , Nucleotidyltransferases/métabolisme , Protéines de signalisation YAP/génétique , Protéines de signalisation YAP/métabolisme
10.
J Cancer Res Ther ; 17(6): 1382-1388, 2021.
Article de Anglais | MEDLINE | ID: mdl-34916368

RÉSUMÉ

OBJECTIVES: The objective of the evaluate was to study and determine the usefulness of immunohistochemistry (IHC) staining in neoplastic lung lesions. MATERIALS AND METHODS: We evaluated seven IHC stains in fifty lung cancers that included adenocarcinoma (AC), squamous cell carcinoma (SCC), small cell carcinoma, and carcinoid tumors. RESULTS: P63 was expressed in all the cases of SCCs and thyroid transcription factor-1 (TTF-1) was expressed in all cases of ACs. CK 5/6 was expressed in 77.77% of SCCs and CK 7 was expressed in 92.59% of ACs. Synaptophysin and chromogranin-A were expressed in 100% of neuroendocrine (NE) carcinomas. CONCLUSION: P63 and TTF-1 are sensitive markers for SCCs and ACs. Synaptophysin and Chromogranin-A are sensitive markers for NE carcinomas.


Sujet(s)
Adénocarcinome pulmonaire/diagnostic , Marqueurs biologiques tumoraux/métabolisme , Carcinome pulmonaire non à petites cellules/diagnostic , Carcinome épidermoïde/diagnostic , Immunohistochimie/méthodes , Tumeurs du poumon/diagnostic , Carcinome pulmonaire à petites cellules/diagnostic , Adénocarcinome pulmonaire/métabolisme , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome épidermoïde/métabolisme , Diagnostic différentiel , Humains , Tumeurs du poumon/métabolisme , Pronostic , Carcinome pulmonaire à petites cellules/métabolisme
11.
Aging (Albany NY) ; 13(18): 21914-21940, 2021 09 16.
Article de Anglais | MEDLINE | ID: mdl-34528900

RÉSUMÉ

Metformin has been tested as an anti-cancer therapy with potential to improve conventional chemotherapy. However, in some cases, metformin fails to sensitize tumors to chemotherapy. Here we test if the presence of P53 could predict the activity of metformin as an adjuvant for cisplatin-based therapy in non-small cell lung cancer (NSCLC). A549, HCC 827 (TP53 WT), H1299, and H358 (TP53 null) cell lines were used in this study. A549 cells were pre-treated with a sub-lethal dose of cisplatin to induce chemoresistance. The effects of metformin were tested both in vitro and in vivo and related to the ability of cells to accumulate Jarid1b, a histone demethylase involved in cisplatin resistance in different cancers. Metformin sensitized A549 and HCC 827 cells (but not H1299 and H358 cells) to cisplatin in a P53-dependent manner, changing its subcellular localization to the mitochondria. Treatment with a sub-lethal dose of cisplatin increased Jarid1b expression, yet downregulated P53 levels, protecting A549Res cells from metformin-induced chemosensitization to cisplatin and favored a glycolytic phenotype. Treatment with FL3, a synthetic flavagline, sensitized A549Res cells to cisplatin. In conclusion, metformin could potentially be used as an adjuvant for cisplatin-based therapy in NSCLC cells if wild type P53 is present.


Sujet(s)
Antinéoplasiques/pharmacologie , Carcinome pulmonaire non à petites cellules/génétique , Cisplatine/pharmacologie , Jumonji Domain-Containing Histone Demethylases/génétique , Metformine/pharmacologie , Protéines nucléaires/génétique , Protéines de répression/génétique , Protéine p53 suppresseur de tumeur/génétique , Cellules A549 , Animaux , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/métabolisme , Synergie des médicaments , Régulation de l'expression des gènes tumoraux , Humains , Jumonji Domain-Containing Histone Demethylases/métabolisme , Mâle , Souris , Souris de lignée NOD , Protéines nucléaires/métabolisme , Protéines de répression/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme
12.
Front Immunol ; 12: 714230, 2021.
Article de Anglais | MEDLINE | ID: mdl-34484217

RÉSUMÉ

Non-small cell lung carcinoma (NSCLC) is a complex cancer biome composed of malignant cells embedded in a sophisticated tumor microenvironment (TME) combined with different initiating cell types, including immune cells and cancer-associated fibroblasts (CAFs), and extracellular matrix (ECM) proteins. However, little is known about these tumors' immune-matricellular relationship as functional and mechanical barriers. This study investigated 120 patients with NSCLC to describe the immune-matricellular phenotypes of their TME and their relationship with malignant cells. Immunohistochemistry (IHC) was performed to characterize immune checkpoints (PD-L1, LAG-3, CTLA-4+, VISTA 1), T cells (CD3+), cytotoxic T cells (CD8+, Granzyme B), macrophages (CD68+), regulatory T cells (FOXP3+, CD4+), natural killer cells (CD57+), and B lymphocytes (CD20+), whereas CAFs and collagen types I, III, and V were characterized by immunofluorescence (IF). We observed two distinct functional immune-cellular barriers-the first of which showed proximity between malignant cells and cytotoxic T cells, and the second of which showed distant proximity between non-cohesive nests of malignant cells and regulatory T cells. We also identified three tumor-associated matricellular barriers: the first, with a localized increase in CAFs and a low deposition of Col V, the second with increased CAFs, Col III and Col I fibers, and the third with a high amount of Col fibers and CAFs bundled and aligned perpendicularly to the tumor border. The Cox regression analysis was designed in two steps. First, we investigated the relationship between the immune-matricellular components and tumor pathological stage (I, II, and IIIA), and better survival rates were seen in patients whose tumors expressed collagen type III > 24.89 fibers/mm². Then, we included patients who had progressed to pathological stage IV and found an association between poor survival and tumor VISTA 1 expression > 52.86 cells/mm² and CD3+ ≤ 278.5 cells/mm². We thus concluded that differential patterns in the distribution of immune-matricellular phenotypes in the TME of NSCLC patients could be used in translational studies to predict new treatment strategies and improve patient outcome. These data raise the possibility that proteins with mechanical barrier function in NSCLC may be used by cancer cells to protect them from immune cell infiltration and immune-mediated destruction, which can otherwise be targeted effectively with immunotherapy or collagen therapy.


Sujet(s)
Marqueurs biologiques tumoraux , Carcinome pulmonaire non à petites cellules/étiologie , Carcinome pulmonaire non à petites cellules/métabolisme , Prédisposition aux maladies/immunologie , Tumeurs du poumon/étiologie , Tumeurs du poumon/métabolisme , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Carcinome pulmonaire non à petites cellules/mortalité , Carcinome pulmonaire non à petites cellules/anatomopathologie , Évolution de la maladie , Femelle , Régulation de l'expression des gènes , Humains , Immunohistochimie , Estimation de Kaplan-Meier , Tumeurs du poumon/mortalité , Tumeurs du poumon/anatomopathologie , Mâle , Adulte d'âge moyen , Grading des tumeurs , Stadification tumorale , Pronostic
13.
Clin Transl Oncol ; 23(12): 2548-2559, 2021 Dec.
Article de Anglais | MEDLINE | ID: mdl-34224057

RÉSUMÉ

PURPOSE: Persistent abnormal proliferation and long distant metastasis of tumors contribute to high mortality rate in non-small cell lung cancer (NSCLC) patients. Strategies that prevent NSCLC proliferation and/or metastasis have been studied but still need to be further explored. Numerous studies have proved the diversity functions of long noncoding RNAs (lncRNAs) exerted in cancer, including NSCLC. In this study, we aim to identify and investigate the role of novel lncRNAs in NSCLC progression. METHODS: RNA sequence data were retrieved from the Cancer Genome Atlas (TCGA), differentially expressed lncRNAs (DElncRNAs) were screened out based on the R language, then real-time PCR experiment was introduced to detect the DElncRNA expression levels. A series of experiments including MTT, cell cycle, transwell, and wound healing assays were employed to explore the effect of DElncRNA MGC27382 on cell proliferation and invasion ability. RESULTS: We detected that DElncRNA MGC27382 is down-regulated in NSCLC tissues and cells. Overexpression of MGC27382 prevented NSCLC cell proliferation via down-regulating cyclin D1 and cyclin E. Moreover, wound healing and transwell assays indicated that the ability of cell invasion and migration could be impaired when cells were treated with MGC27382 overexpression. Further studies demonstrated that MGC27382-mediated inhibition on NSCLC progression can be impaired by LY294002, which is a frequently used inhibitor of AKT/GSK3ß pathway. CONCLUSION: MGC27382 is down-regulated in NSCLC. It exerts an inhibitory role in NSCLC development through suppressing the AKT/GSK3ß pathway. Our results indicate that the lncRNA MGC27382 might be a tumor-suppressor gene in NSCLC. Overexpression of MGC27382 is thought to be a potential strategy for overcoming NSCLC progression.


Sujet(s)
Carcinome pulmonaire non à petites cellules/secondaire , Cycline E/métabolisme , Régulation de l'expression des gènes tumoraux , Glycogen synthase kinase 3 beta/métabolisme , Tumeurs du poumon/anatomopathologie , Protéines oncogènes/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , ARN long non codant/génétique , Apoptose , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Cycle cellulaire , Mouvement cellulaire , Prolifération cellulaire , Cycline E/génétique , Glycogen synthase kinase 3 beta/génétique , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Métastase tumorale , Protéines oncogènes/génétique , Pronostic , Protéines proto-oncogènes c-akt/génétique , Taux de survie , Cellules cancéreuses en culture
14.
Pathol Oncol Res ; 27: 597499, 2021.
Article de Anglais | MEDLINE | ID: mdl-34257548

RÉSUMÉ

Background: Programmed cell death-ligand 1 (PD-L1) protein expression is one of the most extensively studied biomarkers in patients with non-small cell lung cancer (NSCLC). However, there is scarce information regarding its association with distinct adenocarcinoma subtypes. This study evaluated the frequency of PD-L1 expression according to the IASLC/ATS/ERS classification and other relevant histological and clinical features. Patients and Methods: PD-L1 expression was assessed by immunohistochemistry (IHC). According to its positivity in tumor cells membrane, we stratified patients in three different tumor proportions score (TPS) cut-off points: a) <1% (negative), b) between 1 and 49%, and c) ≥50%; afterward, we analyzed the association among PD-L1 expression and lung adenocarcinoma (LADC) predominant subtypes, as well as other clinical features. As an exploratory outcome we evaluated if a PD-L1 TPS score ≥15% was useful as a biomarker for determining survival. Results: A total of 240 patients were included to our final analysis. Median age at diagnosis was 65 years (range 23-94 years). A PD-L1 TPS ≥1% was observed in 52.5% of the entire cohort; regarding specific predominant histological patterns, a PD-L1 TPS ≥1 was documented in 31.2% of patients with predominant-lepidic pattern, 46.2% of patients with predominant-acinar pattern, 42.8% of patients with a predominant-papillary pattern, and 68.7% of patients with predominant-solid pattern (p = 0.002). On the other hand, proportion of tumors with PD-L1 TPS ≥50% was not significantly different among adenocarcinoma subtypes. At the univariate survival analysis, a PD-L1 TPS cut-off value of ≥15% was associated with a worse PFS and OS. Conclusion: According to IASLC/ATS/ERS lung adenocarcinoma classification, the predominant-solid pattern is associated with a higher proportion of PD-L1 positive samples, no subtype was identified to be associated with a high (≥50%) TPS PD-L1.


Sujet(s)
Adénocarcinome pulmonaire/anatomopathologie , Antigène CD274/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Tumeurs du poumon/anatomopathologie , Mutation , Adénocarcinome pulmonaire/classification , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/métabolisme , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Antigène CD274/génétique , Marqueurs biologiques tumoraux/génétique , Carcinome pulmonaire non à petites cellules/classification , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Femelle , Études de suivi , Humains , Immunohistochimie , Tumeurs du poumon/classification , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Mâle , Adulte d'âge moyen , Pronostic , Études rétrospectives , Taux de survie , Jeune adulte
15.
Cancer Res ; 81(13): 3717-3726, 2021 07 01.
Article de Anglais | MEDLINE | ID: mdl-34006524

RÉSUMÉ

Non-small cell lung cancer (NSCLC) is the most common cancer worldwide. With overall 5-year survival estimated at <17%, it is critical to identify factors that regulate NSCLC disease prognosis. NSCLC is commonly driven by mutations in KRAS and TP53, with activation of additional kinases such as SRC promoting tumor invasion. In this study, we investigated the role of NEDD9, a SRC activator and scaffolding protein, in NSCLC tumorigenesis. In an inducible model of NSCLC dependent on Kras mutation and Trp53 loss (KP mice), deletion of Nedd9 (KPN mice) led to the emergence of larger tumors characterized by accelerated rates of tumor growth and elevated proliferation. Orthotopic injection of KP and KPN tumors into the lungs of Nedd9-wild-type and -null mice indicated the effect of Nedd9 loss was cell-autonomous. Tumors in KPN mice displayed reduced activation of SRC and AKT, indicating that activation of these pathways did not mediate enhanced growth of KPN tumors. NSCLC tumor growth has been shown to require active autophagy, a process dependent on activation of the kinases LKB1 and AMPK. KPN tumors contained high levels of active LKB1 and AMPK and increased autophagy compared with KP tumors. Treatment with the autophagy inhibitor chloroquine completely eliminated the growth advantage of KPN tumors. These data for the first time identify NEDD9 as a negative regulator of LKB1/AMPK-dependent autophagy during early NSCLC tumor growth. SIGNIFICANCE: This study demonstrates a novel role for the scaffolding protein NEDD9 in regulating LKB1-AMPK signaling in early stage non-small cell lung cancer, suppressing autophagy and tumor growth.


Sujet(s)
Protéines adaptatrices de la transduction du signal/physiologie , Autophagie , Carcinome pulmonaire non à petites cellules/anatomopathologie , Régulation de l'expression des gènes tumoraux , Tumeurs du poumon/anatomopathologie , Protéines proto-oncogènes p21(ras)/physiologie , Protéine p53 suppresseur de tumeur/physiologie , AMP-Activated Protein Kinases/génétique , AMP-Activated Protein Kinases/métabolisme , Animaux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Prolifération cellulaire , Modèles animaux de maladie humaine , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Souris , Souris de lignée C57BL , Souris knockout , Pronostic , Taux de survie , Cellules cancéreuses en culture
16.
Mol Pharmacol ; 99(6): 435-447, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-33795352

RÉSUMÉ

Rearranged during transfection (RET) rearrangements occur in 1% to 2% of lung adenocarcinomas as well as other malignancies and are now established targets for tyrosine kinase inhibitors. We developed three novel RET fusion-positive (RET+) patient-derived cancer cell lines, CUTO22 [kinesin 5B (KIF5B)-RET fusion], CUTO32 (KIF5B-RET fusion), and CUTO42 (echinoderm microtubule-associated protein-like 4-RET fusion), to study RET signaling and response to therapy. We confirmed each of our cell lines expresses the RET fusion protein and assessed their sensitivity to RET inhibitors. We found that the CUTO22 and CUTO42 cell lines were sensitive to multiple RET inhibitors, whereas the CUTO32 cell line was >10-fold more resistant to three RET inhibitors. We discovered that our RET+ cell lines had differential regulation of the mitogen-activated protein kinase and phosphoinositide 3-kinase/protein kinase B (AKT) pathways. After inhibition of RET, the CUTO42 cells had robust inhibition of phosphorylated AKT (pAKT), whereas CUTO22 and CUTO32 cells had sustained AKT activation. Next, we performed a drug screen, which revealed that the CUTO32 cells were sensitive (<1 nM IC50) to inhibition of two cell cycle-regulating proteins, polo-like kinase 1 and Aurora kinase A. Finally, we show that two of these cell lines, CUTO32 and CUTO42, successfully establish xenografted tumors in nude mice. We demonstrated that the RET inhibitor BLU-667 was effective at inhibiting tumor growth in CUTO42 tumors but had a much less profound effect in CUTO32 tumors, consistent with our in vitro experiments. These data highlight the utility of new RET+ models to elucidate differences in response to tyrosine kinase inhibitors and downstream signaling regulation. Our RET+ cell lines effectively recapitulate the interpatient heterogeneity observed in response to RET inhibitors and reveal opportunities for alternative or combination therapies. SIGNIFICANCE STATEMENT: We have derived and characterized three novel rearranged during transfection (RET) fusion non-small cell lung cancer cell lines and demonstrated that they have differential responses to RET inhibition as well as regulation of downstream signaling, an area that has previously been limited by a lack of diverse cell line modes with endogenous RET fusions. These data offer important insight into regulation of response to RET tyrosine kinase inhibitors and other potential therapeutic targets.


Sujet(s)
Antinéoplasiques/pharmacologie , Carcinome pulmonaire non à petites cellules/génétique , Tumeurs du poumon/génétique , Inhibiteurs de protéines kinases/pharmacologie , Protéines proto-oncogènes c-ret/antagonistes et inhibiteurs , Transduction du signal , Animaux , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Femelle , Humains , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Souris , Souris nude , Protéines proto-oncogènes c-ret/génétique , Protéines de fusion recombinantes/effets des médicaments et des substances chimiques , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
17.
Clin Transl Oncol ; 23(9): 1782-1793, 2021 Sep.
Article de Anglais | MEDLINE | ID: mdl-33847923

RÉSUMÉ

PURPOSE: This project aimed to survey the clinical characteristics and survivals of hyperprogressive disease (HPD) mediated by immune checkpoint inhibitors (ICIs) in an attempt to explore the potential predictors. METHODS: After searching PubMed, MEDLINE, Google Scholar and Cochrane Library databases, 12 studies incorporating 1766 individuals were enrolled. The data were analyzed with Review manager 5.3 software. RESULTS: The results revealed HPD correlated with previous metastatic sites > 2 (OR = 1.86, 95% CI 1.33-2.59, P = 0.0003), liver metastasis (OR = 3.35, 95% CI 2.09-5.35, P < 0.00001), Royal Marsden Hospital (RMH) score ≥ 2 (OR = 2.80, 95% CI 1.85-4.23, P < 0.00001), higher ECOG PS (OR = 1.60, 95% CI 1.13-2.27, P = 0.008) and LDH > upper limits of normal (ULN) (OR = 2.32, 95% CI 1.51-3.58, P = 0.0001). Instead, HPD was unrelated to gender, age, smoking status, PD-L1 expression, therapy, neutrophil-to-lymphocyte ratio, the histology, the status of EGFR, ALK and KRAS in non-small cell lung cancer and HER-2 expression in advanced gastric cancer. Moreover, HPD was evidently correlated with a shorter OS (HR = 2.92, 95% CI 1.79-4.76, P < 0.0001) and PFS (HR = 3.62, 95% CI 2.79-4.68, P < 0.00001). The same phenomena existed in stratified studies based on study regions and tumor types. CONCLUSIONS: This study demonstrated that HPD was related to the number of prior metastatic sites > 2, liver metastasis, RMH score ≥ 2, higher ECOG PS score and LDH > ULN. Moreover, HPD was correlated with a poor OS and PFS in patients following ICI therapy.


Sujet(s)
Marqueurs biologiques tumoraux/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Évolution de la maladie , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Tumeurs du poumon/anatomopathologie , Tumeurs de l'estomac/anatomopathologie , Facteurs âges , Kinase du lymphome anaplasique/métabolisme , Antigène CD274/métabolisme , Antigène CTLA-4/antagonistes et inhibiteurs , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/mortalité , Intervalles de confiance , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Femelle , Humains , Inhibiteurs de points de contrôle immunitaires/effets indésirables , L-Lactate dehydrogenase/génétique , L-Lactate dehydrogenase/métabolisme , Numération des leucocytes , Tumeurs du foie/mortalité , Tumeurs du foie/secondaire , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Tumeurs du poumon/mortalité , Mâle , Mutation , Odds ratio , Survie sans progression , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes p21(ras)/métabolisme , Facteurs sexuels , Tumeurs de l'estomac/traitement médicamenteux , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/mortalité , Échec thérapeutique , Charge tumorale
18.
Sci Rep ; 11(1): 4835, 2021 03 01.
Article de Anglais | MEDLINE | ID: mdl-33649335

RÉSUMÉ

The squamous cell carcinoma of the lung (SCLC) is one of the most common types of lung cancer. As GLOBOCAN reported in 2018, lung cancer was the first cause of death and new cases by cancer worldwide. Typically, diagnosis is made in the later stages of the disease with few treatment options available. The goal of this work was to find some key components underlying each stage of the disease, to help in the classification of tumor samples, and to increase the available options for experimental assays and molecular targets that could be used in treatment development. We employed two approaches. The first was based in the classic method of differential gene expression analysis, network analysis, and a novel concept known as network gatekeepers. The second approach was using machine learning algorithms. From our combined approach, we identified two sets of genes that could function as a signature to identify each stage of the cancer pathology. We also arrived at a network of 55 nodes, which according to their biological functions, they can be regarded as drivers in this cancer. Although biological experiments are necessary for their validation, we proposed that all these genes could be used for cancer development treatments.


Sujet(s)
Marqueurs biologiques tumoraux , Carcinome pulmonaire non à petites cellules , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux , Tumeurs du poumon , Apprentissage machine , Marqueurs biologiques tumoraux/biosynthèse , Marqueurs biologiques tumoraux/génétique , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Bases de données d'acides nucléiques , Femelle , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Mâle
19.
Clin Transl Oncol ; 23(9): 1818-1826, 2021 Sep.
Article de Anglais | MEDLINE | ID: mdl-33728869

RÉSUMÉ

INTRODUCTION: The efficacy of immune checkpoint inhibitors in patients with brain metastases (BMs) from non-oncogene addicted non-small cell lung cancer (NSCLC) is under investigation. Here, we sought to determine the optimal management of NSCLCs with PD-L1 ≥ 50% and asymptomatic BMs who were treated with first-line pembrolizumab. METHODS: Thirty patients from 15 institutions with PD-L1 ≥ 50% NSCLC had asymptomatic BMs, and met inclusion criteria. Patients were classified based on whether they had undergone upfront local radiotherapy for BMs as well as on the type of brain radiotherapy received. RESULTS: Nine patients were treated with upfront pembrolizumab alone, 8 patients with whole-brain radiotherapy (WBRT) followed by pembrolizumab and 13 patients with stereotactic radiosurgery (SRS) followed by pembrolizumab. Patients' characteristics were similar among the three groups of patients except for a higher number of BMs ≥ 3 in the WBRT group. One complete and 4 partial intracranial responses were observed with upfront pembrolizumab alone. The median survival was not reached for the pembrolizumab and WBRT (n = 8) groups, and it was 7.6 months for the SRS (n = 13) group (P = 0.09), with 12-month survival rates being 55.5%, 62.5%, and 23.0%, respectively. Salvage WBRT was delivered in 1 patient in the upfront pembrolizumab group and in 4 patients in the SRS group. CONCLUSIONS: Upfront pembrolizumab showed efficacy in selected patients with PD-L1 ≥ 50% non-oncogene addicted NSCLC and asymptomatic BMs. Prospective studies should address whether pembrolizumab alone, and deferral of radiotherapy, could be pursued in this patient population.


Sujet(s)
Anticorps monoclonaux humanisés/usage thérapeutique , Antinéoplasiques immunologiques/usage thérapeutique , Maladies asymptomatiques , Antigène CD274/métabolisme , Tumeurs du cerveau/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Tumeurs du poumon , Sujet âgé , Sujet âgé de 80 ans ou plus , Anticorps monoclonaux humanisés/effets indésirables , Antinéoplasiques immunologiques/effets indésirables , Tumeurs du cerveau/mortalité , Tumeurs du cerveau/radiothérapie , Tumeurs du cerveau/secondaire , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/radiothérapie , Carcinome pulmonaire non à petites cellules/secondaire , Irradiation crânienne/méthodes , Femelle , Humains , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Mâle , Adulte d'âge moyen , Radiochirurgie/statistiques et données numériques , Études rétrospectives , Thérapie de rattrapage/méthodes , Résultat thérapeutique
20.
Methods Mol Biol ; 2279: 59-73, 2021.
Article de Anglais | MEDLINE | ID: mdl-33683686

RÉSUMÉ

In an era of precision medicine important treatment decisions are dictated by expression of clinically informative tumor protein biomarkers. These biomarkers can be detected by immunohistochemistry (IHC) performed in tumor tissue sections obtained from biopsies or resections. Like all experimental procedures, IHC needs optimization for several of its steps. However, the investigator must avoid optimizing the IHC procedure using valuable human biopsy samples which may be difficult to obtain. Ideally, valuable biopsy samples should only be subjected to IHC once the IHC protocol has been optimized. In this chapter, we describe a procedure for IHC optimization using tri-dimensional (3D) cellular spheroids created from cultured cells. In this approach, cultured cells are pelleted into 3D spheroids, which are then processed just like a tissue sample, namely, fixed, embedded, sectioned, mounted on slides, and stained with IHC just like a human tissue sample. These 3D cellular spheroids have a tissue-like architecture and cellularity resembling a tumor section, and both cellular and antigen structure are preserved. This method is therefore acceptable for IHC optimization before proceeding to the IHC staining of human tumor samples.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Immunohistochimie , Tumeurs du poumon , Inclusion en paraffine , Sphéroïdes de cellules , Carcinome pulmonaire non à petites cellules/diagnostic , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Lignée cellulaire tumorale , Formaldéhyde/composition chimique , Humains , Tumeurs du poumon/diagnostic , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE