bims-meluca Biomed News
on Metabolism of non-small cell lung carcinoma
Issue of 2024‒09‒22
three papers selected by
the Muñoz-Pinedo/Nadal (PReTT) lab, L’Institut d’Investigació Biomèdica de Bellvitge



  1. Heliyon. 2024 Sep 15. 10(17): e37414
      Background: The L-type amino acid transporter (LAT1) exhibits significantly increased expression within tumor cells across various neoplasms. However, the clinical significance of LAT1 expression in patients with pleural mesothelioma (PM) remains unclear.Methods: Eighty patients diagnosed with PM between June 2007 and August 2022, were eligible for this study. LAT1, alanine-serine-cysteine transporter 2 (ASCT2), Ki-67, and VEGFR2 were evaluated by immunohistochemistry. Inflammatory and nutritional indices were also correlated with different variables, including neutrophil to lymphocyte ratio (NLR), platelet to lymphocyte ratio (PLR), systemic immune-inflammation index (SII), prognostic nutritional index (PNI), advanced lung cancer inflammation index (ALI), and Glasgow prognostic score (GPS).
    Results: LAT1 was highly expressed in 57.5 % of patients with PM. Among the 80 patients included in this study, 65 (81.3 %) received chemotherapy, either alone or followed by surgical resection, while 15 (18.7 %) opted for best supportive care. The level of LAT1 significantly correlated with cell proliferation and ASCT2. Factors such as performance status, histology, LAT1 expression, PNI, ALI, and GPS were significant prognostic indicators for progression-free survival (PFS), while Ki-67, LAT1, NLR, SII, PNI, ALI, and GPS were identified as significant predictors for overall survival (OS). LAT1 expression emerged as an independent prognostic factor for predicting PFS and OS in all patients, as well as in the subgroup of 65 patients receiving chemotherapy. Notably, high LAT1 expression proved to be a significant predictor of outcome, particularly in the subgroup with high PLR and SII.
    Conclusion: LAT1 was a significant predictor of outcomes in patients with PM and was more predictive of worse outcomes in patients with high inflammatory and low nutritional status.
    Keywords:  Immunohistochemistry; LAT1; Malignant mesothelioma; Predictive marker; Prognosis
    DOI:  https://doi.org/10.1016/j.heliyon.2024.e37414
  2. Cell Signal. 2024 Sep 16. pii: S0898-6568(24)00383-8. [Epub ahead of print]124 111415
      The MAPK and PI3K/AKT/mTOR pathways are aberrantly activated in non-small cell lung cancer (NSCLC) patients, but therapeutic efficacy of NSCLC using trametinib (MEK inhibitor) or BEZ-235 (dual PI3K/mTOR inhibitor) alone is still unsatisfactory. Therefore, in this study, we aimed to determine whether the combination of trametinib with BEZ-235 exerted synergistic effects against NSCLC in both in vitro and in vivo models, and we preliminarily explored the effect of this combination therapy on glucose metabolism. Our results showed that trametinib combined with BEZ-235 could better inhibit cell proliferation and colony formation, induce G0/G1 phase arrest and apoptosis, and suppress cell invasion and migration compared with the single agent. The combination index demonstrated that trametinib and BEZ-235 exerted strong synergistic effects. Additionally, trametinib and BEZ-235 exhibited synergistic antitumor effects in vivo. Furthermore, trametinib and BEZ-235 synergistically downregulated the expression of related proteins in the MAPK and PI3K/AKT/mTOR pathways, and decreased glucose consumption and lactic acid production through suppressing the expressions of glucose transporter 1 (GLUT1) and lactate dehydrogenase A (LDHA). These data imply that simultaneous inhibition of the MAPK and PI3K/AKT/mTOR pathways using trametinib combined with BEZ-235 could synergistically impair glucose metabolism, resulting in an obvious synergistic therapeutic effect against NSCLC.
    Keywords:  BEZ-235; Non-small cell lung cancer; Synergy; Trametinib
    DOI:  https://doi.org/10.1016/j.cellsig.2024.111415
  3. JTCVS Open. 2024 Aug;20 194-201
      Objectives: Reprogramming of energy metabolism is a well-established hallmark of cancer, with aerobic glycolysis classically considered a prominent feature. We investigate the heterogeneity in glucose metabolism pathways within resectable primary lung adenocarcinoma and its clinical significance.Methods: Using The Cancer Genome Atlas data, RNA expressions were extracted from 489 primary lung adenocarcinoma samples. Prognostic influence of glycolytic, aerobic, and mitochondrial markers (monocarboxylate transporter [MCT]4, MCT1, and translocase of outer mitochondrial membrane 20, respectively) was assessed using Kaplan-Meier analysis. Clustering of 35 genes involved in glucose metabolism was performed using the k-means method. The clusters were then analyzed for associations with demographic, clinical, and pathologic variables. Overall survival was assessed using the Kaplan-Meier estimator. Multivariate analysis was performed to assess the independent prognostic value of cluster membership.
    Results: Classical statistical approach showed that higher expression of MCT4 was associated with a significantly worse prognosis. Increased expression of translocase of outer mitochondrial membrane 20 was associated with a nonsignificant trend toward better prognosis, and increased expression of MCT1 was associated with a better outcome. Clustering identified 3 major metabolic phenotypes, dominantly hypometabolic, dominantly oxidative, and dominantly mixed oxidative/glycolytic with significantly different pathologic stage distribution and prognosis; mixed oxidative/glycolytic was associated with worse survival. Cluster membership was independently associated with survival.
    Conclusions: This study demonstrates the existence of distinct glucose metabolism clusters in resectable lung adenocarcinoma, providing valuable prognostic information. The findings highlight the potential relevance of considering metabolic profiles when designing strategies for reprogramming energy metabolism. Further studies are warranted to validate these findings in different cancer types and populations.
    Keywords:  gene clustering; glucose metabolism; lung cancer; survival
    DOI:  https://doi.org/10.1016/j.xjon.2024.06.010