bims-meluca Biomed News
on Metabolism of non-small cell lung carcinoma
Issue of 2019‒03‒10
six papers selected by
Cristina Muñoz Pinedo
L’Institut d’Investigació Biomèdica de Bellvitge


  1. Cancer Sci. 2019 Mar 07.
      Some driver gene mutations including EGFR have been reported to be involved in expression regulation of the immunosuppressive checkpoint protein programmed death ligand 1 (PD-L1), but the underlying mechanism remains obscure. Here, we investigated the potential role and precise mechanism of EGFR mutants in PD-L1 expression regulation in non-small cell lung cancer (NSCLC) cells. Examination of pivotal EGFR signaling effectors in eight NSCLC cells indicated apparent associations of PD-L1 overexpression with phosphorylation of AKT and ERK, especially with increased protein levels of phospho-IκBα (p-IκBα) and HIF-1α. Flow cytometry results showed stronger membrane co-expression of EGFR and PD-L1 in NSCLC cells with EGFR mutants compared with cells carrying wild-type EGFR. Additionally, ectopic expression or depletion of EGFR mutants and administration of EGFR pathway inhibitors targeting MEK/ERK, PI3K/AKT, mTOR/S6, IκBα, and HIF-1α indicated strong accordance among protein levels of PD-L1, p-IκBα, and HIF-1α in NSCLC cells. Further administration of pathway inhibitors significantly inhibited xenograft tumor growth and p-IκBα, HIF-1α, and PD-L1 expression of NSCLC cells carrying EGFR mutant in nude mice. Moreover, immunohistochemical analysis revealed obviously increased protein levels of p-IκBα, HIF-1α, and PD-L1 in NSCLC tissues with EGFR mutants compared with tissues carrying wild-type EGFR. NSCLC tissues with either p-IκBα or HIF-1α staining positive were more likely to possess elevated PD-L1 expression compared with tissues scored p-IκBα and HIF-1α both negative. Our findings demonstrated important roles of phosphorylation activation of AKT and ERK and potential interplay and cooperation between NF-κB and HIF-1α in PD-L1 expression regulation by EGFR mutants in NSCLC. This article is protected by copyright. All rights reserved.
    Keywords:  HIF-1α; NF-κB; PD-L1; epidermal growth factor receptor; non-small cell lung cancer
    DOI:  https://doi.org/10.1111/cas.13989
  2. J Thorac Oncol. 2019 Feb 27. pii: S1556-0864(19)30150-9. [Epub ahead of print]
      BACKGROUND: LKB1, also called STK11, is a tumor suppressor that functions as master regulator of cell growth, metabolism, survival, and polarity. Approximately 30-35% of patients with NSCLC possess inactivated LKB1, and these patients respond poorly to anti-PD-1/PD-L1 immunotherapy. Therefore, novel therapies targeting NSCLC with LKB1-loss are needed.METHODS: We used a new in silico signaling analysis method to identify the potential therapeutic targets and reposition drugs by integrating gene expression data with the Kyoto Encyclopedia of Genes and Genomes (KEGG) signaling pathways. LKB1 wild type and deficient NSCLC cell lines, including knock out clones generated by CRISPR-Cas9, were treated with inhibitors of mTOR and PI3K and a dual inhibitor.
    RESULTS: In silico experiment showed that inhibition of both mTOR and PI3K can be synergistically effective in LKB1 deficient NSCLC. In vitro and in vivo experiments showed the synergistic effect of mTOR inhibition and PI3K inhibition in LKB1 mutant NSCLC cell lines. The sensitivity to dual inhibition of mTOR and PI3K is higher in LKB1 mutant cell lines than wild-type cell lines. A higher compensatory increase of Akt phosphorylation after rapamycin treatment in LKB1 deficient cells compared to LKB1 wild type cells is responsible for the synergistic effect of mTOR and PI3K inhibition. Dual inhibition of mTOR and PI3K showed a greater decrease in protein expression of cell cycle regulating proteins in LKB1 knock out cells compared to LKB1 wild type cells.
    CONCLUSION: Dual molecular targeted therapy for mTOR and PI3K may be a promising therapeutic strategy in the specific population of lung cancer patients with LKB1 loss.
    Keywords:  LKB1; PI3K; lung cancer; mTOR; molecular targeted agent
    DOI:  https://doi.org/10.1016/j.jtho.2019.02.019
  3. Metabolomics. 2018 Aug 31. 14(9): 118
      INTRODUCTION: Histologically lung cancer is classified into four major types: adenocarcinoma (Ad), squamous cell carcinoma (SqCC), large cell carcinoma (LCC), and small cell lung cancer (SCLC). Presently, our understanding of cellular metabolism among them is still not clear.OBJECTIVES: The goal of this study was to assess the cellular metabolic profiles across these four types of lung cancer using an untargeted metabolomics approach.
    METHODS: Six lung cancer cell lines, viz., Ad (A549 and HCC827), SqCC (NCl-H226 and NCl-H520), LCC (NCl-H460), and SCLC (NCl-H526), were analyzed using liquid chromatography quadrupole time-of-flight mass spectrometry, with normal human small airway epithelial cells (SAEC) as the control group. The principal component analysis (PCA) was performed to identify the metabolic signatures that had characteristic alterations in each histological type. Further, a metabolite set enrichment analysis was performed for pathway analysis.
    RESULTS: Compared to the SAEC, 31, 27, 34, 34, 32, and 39 differential metabolites mainly in relation to nucleotides, amino acid, and fatty acid metabolism were identified in A549, HCC827, NCl-H226, NCl-H520, NCl-H460, and NCl-H526 cells, respectively. The metabolic signatures allowed the six cancerous cell lines to be clearly separated in a PCA score plot.
    CONCLUSION: The metabolic signatures are unique to each histological type, and appeared to be related to their cell-of-origin and mutation status. The changes are useful for assessing the metabolic characteristics of lung cancer, and offer potential for the establishment of novel diagnostic tools for different origin and oncogenic mutation of lung cancer.
    Keywords:  Large cell carcinoma; Lung adenocarcinoma; Metabolic signatures; Pathway analysis; Small cell lung cancer; Squamous cell carcinoma
    DOI:  https://doi.org/10.1007/s11306-018-1417-x
  4. Cancer Med. 2019 Mar 07.
      The main aim of this study is to investigate whether baseline lactate dehydrogenase (LDH) is associated with the clinical outcome of non small-cell lung cancer (NSCLC) patients treated with immune checkpoint inhibitors (ICIs). We searched Pubmed, the Cochrane Central library and Embase for peripheral blood biomarker of LDH in advanced NSCLC patients treated with ICIs. We extracted the hazard ratio (HR) with 95% confidence interval (CI) for the progression free survival (PFS) and overall survival (OS) and performed meta-analysis of HR. Pooled estimates of treatment outcomes were calculated by stata 15.1. Six studies with 1136 patients were included in this study. The pooled results of univariate analysis suggested that an elevated pretreatment LDH level was correlated with significant shorter PFS (HR = 1.53, 95% CI 1.27-1.83, P < 0.001) and OS (HR = 2.11, 95% CI 1.43-3.11, P < 0.001). The association remained significant in the multivariate analysis that elevated pretreatment LDH level was associated with poor PFS (HR = 1.62, 95% CI 1.26-2.08, P < 0.001) and OS (HR = 2.38, 95% CI 1.37-4.12, P = 0.002). A high pretreatment LDH level was significantly correlated with shorter PFS and OS. Pretreatment LDH may serve as a predictive biomarker for advanced NSCLC patients treated with ICIs.
    Keywords:  Immunotherapy; Neutrophil-to-lymphocyte ratio; Non-small cell lung cancer; Peripheral blood biomarker
    DOI:  https://doi.org/10.1002/cam4.2024
  5. J Cell Physiol. 2019 Mar 09.
      Identification of to what extent tumor burden influences muscle mass independently of specific treatments for cancer-cachexia remains to be elucidated. We hypothesized that reduced tumor burden by selective treatment of tumor with immunomodulators may exert beneficial effects on muscle wasting and function in mice. Body and muscle weight, grip strength, physical activity, muscle morphometry, apoptotic nuclei, troponin-I systemic levels, interleukin-6, proteolytic markers, and tyrosine release, and apoptosis markers were determined in diaphragm and gastrocnemius muscles of lung cancer (LP07 adenocarcinoma cells) mice (BALB/c) treated with monoclonal antibodies (mAbs), against immune check-points and pathways (CD-137, cytotoxic T-lymphocyte associated protein-4, programed cell death-1, and CD-19; N = 10/group). Nontreated lung cancer cachectic mice were the controls. T and B cell numbers and macrophages were counted in tumors of both mouse groups. Compared to nontreated cachectic mice, in the mAbs-treated animals, T cells increased, no differences in B cells or macrophages, the variables final body weight, body weight and grip strength gains significantly improved. In diaphragm and gastrocnemius of mAbs-treated cachectic mice, number of apoptotic nuclei, tyrosine release, proteolysis, and apoptosis markers significantly decreased compared to nontreated cachectic mice. Systemic levels of troponin-I significantly decreased in treated cachectic mice compared to nontreated animals. We conclude that reduced tumor burden as a result of selective treatment of the lung cancer cells with immunomodulators elicits per se beneficial effects on muscle mass loss through attenuation of several biological mechanisms that lead to increased protein breakdown and apoptosis, which translated into significant improvements in limb muscle strength but not in physical activity parameters.
    Keywords:  cancer-induced cachexia; immunotherapy; lung tumor cells; muscle function and physical activity; muscle proteolysis and apoptosis
    DOI:  https://doi.org/10.1002/jcp.28437
  6. Cell Physiol Biochem. 2019 ;52(3): 368-381
      BACKGROUND/AIMS: The present study aimed to explore the function of NEAT1 on non-small cell lung cancer (NSCLC), as well as its underlying mechanisms.METHODS: Quantitative realtime PCR (qRT-PCR) was used to measure NEAT1 expression in NSCLC tissues and cells. MTT assay and transwell assay were performed to detect cell proliferation, migration and invasion. Potential target genes were identified via luciferase reporter assay. Protein analysis was performed through western blotting.
    RESULTS: The expressions of NEAT1 were significantly higher in both of NSCLC tissues and cells than in normal controls. High expression of NEAT1 was significantly associated with TNM stage (P=0.000) and metastasis (P=0.000). NEAT1 knockdown inhibited the proliferation, migration and invasion of NSCLC cells. Hypoxia induction mediated by HIF-2α promoted EMT and NEAT1 expressions. Moreover, miR-101-3p was a target of NEAT1. We also found that SOX9 was a target of miR-101-3p. Oncogenic function of NEAT1 on NSCLC progression was mediated by miR-101-3p/SOX9/Wnt/β-catenin signaling pathway.
    CONCLUSION: NEAT1 up-regulation induced by HIF-2α over-expression could promote the progression of NSCLC under hypoxic condition. Moreover, NEAT1 also takes part in NSCLC progression via miR-101-3p/SOX9/Wnt/β-catenin axis.
    Keywords:  Hypoxia; NEAT1; NSCLC; SOX9; Wnt/β-catenin; miR-101-3p
    DOI:  https://doi.org/10.33594/000000026