Skip to content
2000
Volume 32, Issue 31
  • ISSN: 0929-8673
  • E-ISSN: 1875-533X

Abstract

Background

Entrectinib, a ROS1 inhibitor, is effective in patients with ROS1-positive non-small-cell lung cancer (NSCLC). However, entrectinib resistance remains a challenge worldwide. The biomarkers of entrectinib resistance and molecular mechanisms have not been clarified based on the Gene Expression Omnibus (GEO) database.

Objectives

The aim of this study is to identify key genes and signaling pathways involved in the development of entrectinib-resistant NSCLC through bioinformatics analysis and experimental validation.

Methods

Differentially expressed genes (DEGs) were screened between entrectinib resistant and parental human NSCLC cell lines of the GSE214715 dataset, lung adenocarcinoma (LUAD) and non-tumor adjacent tissues of the GSE75037 dataset, and NSCLC and non-tumor adjacent tissues of the GSE18842 dataset. Functional enrichment analyses were performed, including Gene Ontology (GO) term and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Overlapped DEGs among those three datasets were identified using the Venn diagram package. The transcriptional levels of key genes were investigated using the University of ALabama at Birmingham CANcer data analysis Portal (UALCAN). The association between transcriptional levels of key genes and survival was analyzed using Kaplan-Meier Plotter (https://www.kmplot.com/analysis/). The correlations between hub genes and immune cell infiltration were investigated using the Tumor Immune Estimation Resource (TIMER) database. Specific signaling pathway enrichment analysis was performed using Gene Set Enrichment Analysis (GSEA) of LinkedOmics. Competitive endogenous RNA (ceRNA) networks, Genome-wide Association Studies (GWAS), and drug sensitivity analyses of key genes were further investigated. The expression of ZEB2 was subsequently confirmed in both parental HCC78 cells and entrectinib-resistant HCC78 cells using real-time quantitative polymerase chain reaction (qRT-PCR).

Results

708 DEGs were identified between entrectinib-resistant CUTO28 (CUTO28-ER) and parental CUTO28 cell lines in the GSE214715 dataset. One thousand three hundred and ninety-five DEGs were identified between entrectinib resistant (CUTO37-ER) and parental CUTO37 cell lines in the GSE214715 dataset. Eight hundred and forty-nine DEGs were identified between LUAD and non-tumor adjacent tissues in the GSE75037 dataset. Seven hundred and sevety-three DEGs were identified between NSCLC and non-tumor adjacent tissues in the GSE18842 dataset. Among these three datasets, seven overlapped DEGs were identified, including ZBED2, CHI3L2, CELF2, SEMA5A, ZEB2, S100A12, and PDK4. Among these seven overlapped DEGs, the expression levels of CHI3L2, ZEB2, and S100A12 were downregulated in those three datasets. The results of analysis using the UALCAN database showed that these three genes were significantly downregulated in LUAD and LUSC patients compared with the normal population. However, only the lower transcriptional level of ZEB2 was linked to worse survival in patients with lung cancer. GSEA analysis revealed that ZEB2 was significantly negatively correlated with Nucleotide Excision Repair (NER) in LUAD, and Homologous Recombination (HR) and NER in LUSC, which were linked to drug resistance. A ceRNA network of THRB-AS1/ has-miR-1293/ ZEB2 in LUAD was established.

Conclusion

We have identified core genes associated with non-small cell resistance to entrectinib, including CHI3L2, ZEB2, and S100A12. ZEB2 is a core gene associated with acquired resistance to entetinib in NSCLC.

Loading

Article metrics loading...

/content/journals/cmc/10.2174/0109298673320448240801061941
2024-08-06
2025-10-23
Loading full text...

Full text loading...

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; & Jemal, A. Cancer statistics, 2022. CA: a cancer journal for clinicians, 72(1), 7-30.
  2. RizzoA. Identifying optimal first-line treatment for advanced non-small cell lung carcinoma with high PD-L1 expression: a matter of debate.Br. J. Cancer202212781381138210.1038/s41416‑022‑01929‑w36064585
    [Google Scholar]
  3. GuvenD.C. SahinT.K. ErulE. RizzoA. RicciA.D. AksoyS. YalcinS. The association between albumin levels and survival in patients treated with immune checkpoint inhibitors: A systematic review and meta-analysis.Front. Mol. Biosci.20229103912110.3389/fmolb.2022.103912136533070
    [Google Scholar]
  4. Dall’OlioF.G. RizzoA. MollicaV. MassucciM. MaggioI. MassariF. Immortal time bias in the association between toxicity and response for immune checkpoint inhibitors: a meta-analysis.Immunotherapy202113325727010.2217/imt‑2020‑017933225800
    [Google Scholar]
  5. RizzoA. MollicaV. TateoV. TassinariE. MarchettiA. RoselliniM. De LucaR. SantoniM. MassariF. Hypertransaminasemia in cancer patients receiving immunotherapy and immune-based combinations: the MOUSEION-05 study.Cancer Immunol. Immunother.20237261381139410.1007/s00262‑023‑03366‑x36695827
    [Google Scholar]
  6. DziadziuszkoR. KrebsM.G. De BraudF. SienaS. DrilonA. DoebeleR.C. PatelM.R. ChoB.C. LiuS.V. AhnM.J. ChiuC.H. FaragoA.F. LinC.C. KarapetisC.S. LiY.C. DayB. ChenD. WilsonT.R. BarlesiF. Updated integrated analysis of the efficacy and safety of entrectinib in locally advanced or metastatic ROS1 fusion–positive non–small-cell lung cancer.J. Clin. Oncol.202139111253126310.1200/JCO.20.0302533646820
    [Google Scholar]
  7. DrilonA. SienaS. DziadziuszkoR. BarlesiF. KrebsM.G. ShawA.T. de BraudF. RolfoC. AhnM.J. WolfJ. SetoT. ChoB.C. PatelM.R. ChiuC.H. JohnT. GotoK. KarapetisC.S. ArkenauH.T. KimS.W. OheY. LiY.C. ChaeY.K. ChungC.H. OttersonG.A. MurakamiH. LinC.C. TanD.S.W. PrenenH. RiehlT. Chow-ManevalE. SimmonsB. CuiN. JohnsonA. EngS. WilsonT.R. DoebeleR.C. Entrectinib in ROS1 fusion-positive non-small-cell lung cancer: integrated analysis of three phase 1–2 trials.Lancet Oncol.202021226127010.1016/S1470‑2045(19)30690‑431838015
    [Google Scholar]
  8. Sartore-BianchiA. PizzutiloE.G. MarrapeseG. TosiF. CereaG. SienaS. Entrectinib for the treatment of metastatic NSCLC: safety and efficacy.Expert Rev. Anticancer Ther.202020533334110.1080/14737140.2020.174743932223357
    [Google Scholar]
  9. KuB.M. BaeY.H. LeeK.Y. SunJ.M. LeeS.H. AhnJ.S. ParkK. AhnM.J. Entrectinib resistance mechanisms in ROS1-rearranged non-small cell lung cancer.Invest. New Drugs202038236036810.1007/s10637‑019‑00795‑331124056
    [Google Scholar]
  10. LiZ. FanY. MaY. MengN. LiD. WangD. LianJ. HuC. Identification of crucial genes and signaling pathways in alectinib-resistant lung adenocarcinoma using bioinformatic analysis.Mol. Biotechnol.202310.1007/s12033‑023‑00973‑y38142454
    [Google Scholar]
  11. GirardL. Rodriguez-CanalesJ. BehrensC. ThompsonD.M. BotrosI.W. TangH. XieY. RekhtmanN. TravisW.D. WistubaI.I. MinnaJ.D. GazdarA.F. An expression signature as an aid to the histologic classification of non–small cell lung cancer.Clin. Cancer Res.201622194880488910.1158/1078‑0432.CCR‑15‑290027354471
    [Google Scholar]
  12. Sanchez-PalenciaA. Gomez-MoralesM. Gomez-CapillaJ.A. PedrazaV. BoyeroL. RosellR. Fárez-VidalM.E. Gene expression profiling reveals novel biomarkers in nonsmall cell lung cancer.Int. J. Cancer2011129235536410.1002/ijc.2570420878980
    [Google Scholar]
  13. ChandrashekarD.S. KarthikeyanS.K. KorlaP.K. PatelH. ShovonA.R. AtharM. NettoG.J. QinZ.S. KumarS. ManneU. CreightonC.J. VaramballyS. UALCAN: An update to the integrated cancer data analysis platform.Neoplasia202225182710.1016/j.neo.2022.01.00135078134
    [Google Scholar]
  14. ChandrashekarD.S. BashelB. BalasubramanyaS.A.H. CreightonC.J. Ponce-RodriguezI. ChakravarthiB.V. S.K. VaramballyS. UALCAN: A Portal for Facilitating Tumor Subgroup Gene Expression and Survival Analyses.Neoplasia201719864965810.1016/j.neo.2017.05.00228732212
    [Google Scholar]
  15. GyorffyB. Transcriptome-level discovery of survival-associated biomarkers and therapy targets in non-small-cell lung cancer.Br. J. Pharmacol.2024181336237437783508
    [Google Scholar]
  16. LiT. FuJ. ZengZ. CohenD. LiJ. ChenQ. LiB. LiuX.S. TIMER2.0 for analysis of tumor-infiltrating immune cells.Nucleic Acids Res.202048W1W509W51410.1093/nar/gkaa40732442275
    [Google Scholar]
  17. LiT. FanJ. WangB. TraughN. ChenQ. LiuJ.S. LiB. LiuX.S. TIMER: a web server for comprehensive analysis of tumor-infiltrating immune cells.Cancer Res.20177721e108e11010.1158/0008‑5472.CAN‑17‑030729092952
    [Google Scholar]
  18. LiB. SeversonE. PignonJ.C. ZhaoH. LiT. NovakJ. JiangP. ShenH. AsterJ.C. RodigS. SignorettiS. LiuJ.S. LiuX.S. Comprehensive analyses of tumor immunity: implications for cancer immunotherapy.Genome Biol.201617117410.1186/s13059‑016‑1028‑727549193
    [Google Scholar]
  19. VasaikarS.V. StraubP. WangJ. ZhangB. Linked-omics: analyzing multi-omics data within and across 32 cancer types.Nucleic Acids Res.201846D1D956D96310.1093/nar/gkx109029136207
    [Google Scholar]
  20. ZhaoP. ZhenH. ZhaoH. HuangY. CaoB. Identification of hub genes and potential molecular mechanisms related to radiotherapy sensitivity in rectal cancer based on multiple datasets.J. Transl. Med.202321117610.1186/s12967‑023‑04029‑236879254
    [Google Scholar]
  21. AdamL. ZhongM. ChoiW. QiW. NicolosoM. AroraA. CalinG. WangH. Siefker-RadtkeA. McConkeyD. Bar-EliM. DinneyC. miR-200 expression regulates epithelial-to-mesenchymal transition in bladder cancer cells and reverses resistance to epidermal growth factor receptor therapy.Clin. Cancer Res.200915165060507210.1158/1078‑0432.CCR‑08‑224519671845
    [Google Scholar]
  22. FangS. ZengX. ZhuW. TangR. ChaoY. GuoL. Zinc finger E-box-binding homeobox 2 (ZEB2) regulated by miR-200b contributes to multi-drug resistance of small cell lung cancer.Exp. Mol. Pathol.201496343844410.1016/j.yexmp.2014.04.00824769353
    [Google Scholar]
  23. LeeE. WangJ. YumotoK. JungY. CackowskiF.C. DeckerA.M. LiY. FranceschiR.T. PientaK.J. TaichmanR.S. DNMT1 regulates epithelial-mesenchymal transition and cancer stem cells, which promotes prostate cancer metastasis.Neoplasia201618955356610.1016/j.neo.2016.07.00727659015
    [Google Scholar]
  24. ChuP.Y. HuF.W. YuC.C. TsaiL.L. YuC.H. WuB.C. ChenY.W. HuangP.I. LoW.L. Epithelial–mesenchymal transition transcription factor ZEB1/ZEB2 co-expression predicts poor prognosis and maintains tumor-initiating properties in head and neck cancer.Oral Oncol.2013491344110.1016/j.oraloncology.2012.07.01222892238
    [Google Scholar]
  25. RenD. WangM. GuoW. HuangS. WangZ. ZhaoX. DuH. SongL. PengX. Double-negative feedback loop between ZEB2 and miR-145 regulates epithelial-mesenchymal transition and stem cell properties in prostate cancer cells.Cell Tissue Res.2014358376377810.1007/s00441‑014‑2001‑y25296715
    [Google Scholar]
  26. LiM.Z. WangJ.J. YangS.B. LiW.F. XiaoL.B. HeY.L. SongX.M. ZEB2 promotes tumor metastasis and correlates with poor prognosis of human colorectal cancer.Am. J. Transl. Res.2017962838285128670373
    [Google Scholar]
  27. KarrethF.A. TayY. PernaD. AlaU. TanS.M. RustA.G. DeNicolaG. WebsterK.A. WeissD. Perez-ManceraP.A. KrauthammerM. HalabanR. ProveroP. AdamsD.J. TuvesonD.A. PandolfiP.P. In vivo identification of tumor- suppressive PTEN ceRNAs in an oncogenic BRAF-induced mouse model of melanoma.Cell2011147238239510.1016/j.cell.2011.09.03222000016
    [Google Scholar]
  28. JiangM. ZhongT. ZhangW. XiaoZ. HuG. ZhouH. KuangH. Reduced expression of miR-205-5p promotes apoptosis and inhibits proliferation and invasion in lung cancer A549 cells by upregulation of ZEB2 and downregulation of erbB3.Mol. Med. Rep.20171553231323810.3892/mmr.2017.639828350117
    [Google Scholar]
  29. LinglingJ. XiangaoJ. GuiqingH. JichanS. FeifeiS. HaiyanZ. SNHG20 knockdown suppresses proliferation, migration and invasion, and promotes apoptosis in non-small cell lung cancer through acting as a miR-154 sponge.Biomed. Pharmacother.201911210864810.1016/j.biopha.2019.10864830780105
    [Google Scholar]
  30. LiZ. ZhouB. ZhuX. YangF. JinK. DaiJ. ZhuY. SongX. JiangG. Differentiation-related genes in tumor-associated macrophages as potential prognostic biomarkers in non-small cell lung cancer.Front. Immunol.202314112384010.3389/fimmu.2023.112384036969247
    [Google Scholar]
  31. WangL.E. YinM. DongQ. StewartD.J. MerrimanK.W. AmosC.I. SpitzM.R. WeiQ. DNA repair capacity in peripheral lymphocytes predicts survival of patients with non-small-cell lung cancer treated with first-line platinum-based chemotherapy.J. Clin. Oncol.201129314121412810.1200/JCO.2010.34.361621947825
    [Google Scholar]
  32. OliverT.G. MercerK.L. SaylesL.C. BurkeJ.R. MendusD. LovejoyK.S. ChengM.H. SubramanianA. MuD. PowersS. CrowleyD. BronsonR.T. WhittakerC.A. BhutkarA. LippardS.J. GolubT. ThomaleJ. JacksT. Sweet-CorderoE.A. Chronic cisplatin treatment promotes enhanced damage repair and tumor progression in a mouse model of lung cancer.Genes Dev.201024883785210.1101/gad.189701020395368
    [Google Scholar]
  33. LeeY.S. DuttaA. MicroRNAs in Cancer.Annu. Rev. Pathol.20094119922710.1146/annurev.pathol.4.110807.09222218817506
    [Google Scholar]
  34. ChanJ.J. TayY. Noncoding RNA:RNA regulatory networks in cancer.Int. J. Mol. Sci.2018195131010.3390/ijms1905131029702599
    [Google Scholar]
/content/journals/cmc/10.2174/0109298673320448240801061941
Loading
/content/journals/cmc/10.2174/0109298673320448240801061941
Loading

Data & Media loading...

Supplements

Supplementary material is available on the publisher’s website along with the published article.


  • Article Type:
    Research Article
Keyword(s): Entrectinib; GEO; HCC78 cells; non-small-cell lung cancer (NSCLC); ROS1; ZEB2
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error
Please enter a valid_number test