How apoptosis and epithelial-to-mesenchymal transition are nested in EGFR inhibitors resistance in lung cancer
Editorial

How apoptosis and epithelial-to-mesenchymal transition are nested in EGFR inhibitors resistance in lung cancer

Simon Garinet1,2, Audrey Didelot1, Elena Garelli3, Karine Pallier4, Hélène Blons1,2, Antoine Legras1,5

1INSERM UMR-S1147, CNRS SNC 5014, Saints-Pères Research Center, 45 rue des Saints-Pères Paris-Descartes University, Sorbonne Paris Cité University, Paris 75006, France;2Molecular Biology Department, Georges Pompidou European Hospital, Assistance Publique - Hôpitaux de Paris, Paris 75015, France;3Thoracic Surgery Department, Cochin Hospital, Sorbonne Paris Cité University, Assistance Publique des Hôpitaux de Paris, Paris 75014, France;4Haute-Vienne Coordination Center in Oncology, Limoges University Hospital Center, Limoges 87000, France;5Thoracic Surgery Department, Trousseau Hospital, Tours University Hospital Center, Chambray-lès-Tours 37170, France

Correspondence to: Antoine Legras, MD, PhD. Thoracic Surgery Department, Trousseau Hospital, Tours University Hospital Center, Avenue de la République, Chambray-lès-Tours 37170, France. Email: antoine.legras@chu-tours.fr.

Provenance: This is an invited Editorial commissioned by Executive Editor-in-Chief Jianxing He (Director of the Thoracic Surgery Department, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China).

Comment on: Song KA, Niederst MJ, Lochmann TL, et al. Epithelial-to-Mesenchymal Transition Antagonizes Response to Targeted Therapies in Lung Cancer by Suppressing BIM. Clin Cancer Res 2018;24:197-208.


Submitted Nov 03, 2018. Accepted for publication Dec 21, 2018.

doi: 10.21037/jtd.2018.12.117


Non-small cell lung cancer (NSCLC) is today the first cancer worldwide and represents the first cause of cancer-related death. The identification of EGFR activating mutations and development of specific EGFR inhibitors (EGFRi) such as tyrosine kinase inhibitor (TKI) has changed the management of metastatic NSCLC. However, TKI resistance progressively appeared and EGFR secondary resistance mutation have been identified such as the most frequent T790M (1). Also, the epithelial-to-mesenchymal transition (EMT) was identified as a complex molecular and cellular process involved in tissue remodeling that plays essential roles in cell invasion, migration and drug resistance in many cancer types including NSCLC (2). During EMT, cells undergo conversion from epithelial to mesenchymal state (3). Epithelial differentiated characteristics are lost, including cell-cell adhesion, planar and apical-basal polarity and lack of mobility. On the contrary, mesenchymal features are briefly acquired, such as cell mobility, invasiveness, gain of stem cell properties and a reinforced resistance to apoptosis. After migration, cells may reverse their phenotype through the mesenchymal to epithelial transition (MET) (4).

In their manuscript entitled “Epithelial-to-mesenchymal transition antagonizes response to targeted therapies in lung cancer by suppressing BIM”, published at the beginning of 2018, in the well-recognized Clinical Cancer Research journal, Kyung-A Song and co-authors found that mesenchymal EGFR mutant lung cancers were resistant to EGFRi-induced apoptosis via insufficient expression of BIM, preventing cell death despite potent suppression of oncogenic signaling following EGFRi treatment (5). They initially studied available public datasets, from CCLE and ExpO, to describe an inverse link between Vimentin/ZEB1 and BIM. Their conclusions were drawn after several investigations using real-time quantitative polymerase chain reaction to quantify expression levels of Twist1, ZEB1, Bim and β-actin to characterize EMT, and investigated also different EMT-pathways, based on Twist1 plasmid insertion in NSCLC cell lines, and TGF-β induced EMT. Similarly, BIM was assessed at both the mRNA and protein level. Furthermore, different ways of EGFRi resistance were evaluated, combining different drugs and different models. Mechanistically, they observed that the EMT transcription factor ZEB1 inhibits BIM expression by binding directly to the BIM promoter and repressing transcription. Furthermore, they interestingly found that derepression of BIM expression by depletion of ZEB1 (using shRNA and siRNA methods) or treatment with the BH3 mimetic ABT-263 to enhance “free” cellular BIM levels both led to re-sensitization of mesenchymal EGFR mutant cancers to EGFR inhibitors. This re-sensitization was observed in cell cultures but also in a xenograft mouse model (SCID mice and subcutaneous tumor cells injection), and confirmed in a recent study, showing that BCL2-inhibitors could restore sensitivity to osimertinib specifically in TWIST1 overexpressed cells (6). Therefore, re-establishing an apoptotic response in EMT-mediated EGFR mutant NSCLC resistant cells is a promising pharmaceutical strategy, in the landscape of a clinical trial (clinical trial number NCT02520778) using ABT-263 (navitoclax) with EGFRi in EGFR mutant NSCLC. Finally, they also showed that this relationship between EMT and loss of BIM was not restricted to EGFR mutant NSCLC as it was also observed in KRAS mutant NSCLC cell lines and in public datasets (CCLE).

This work must be pointed out, considering the excellent scientific tracking and the evidence, in their results, of a significant improvement in the neutralization of EMT. Transposition of these results into reality seems to be the necessary next step, in a clinical trial. The strength also of this work is the correlation between results obtained in cell lines cultures and in in vivo tumors. Nevertheless, potential discrepancies might be observed when during the transfer of the strategy, between strictly contained cell lines models, or mouse models, due to tumor microenvironment, constantly not explored in such models.

However, apoptosis escape may also be only an issue of EMT among others, leading to imagine the combination of several therapies, targeting apoptosis, such as navitoclax, and targeting other aspects of EMT. As EMT is a dynamic and complex phenomenon, characterizing and targeting EMT remain difficult. Different models have been proposed to characterize transitions between epithelial, epithelial-mesenchymal state, and mesenchymal phenotypes, using various terms in literature: partial EMT (7), intermediate EMT (8), incomplete EMT (9), hybrid epithelial/mesenchymal (10). MiR-200/ZEB1 seems to behave as a regulator enabling transitions among the constantly evolving EMT phenotypes (11). Finally in vivo tumor classification remains a challenge not only because EMT is a dynamic phenomenon but also because EMT markers are heterogeneously expressed with intra-tumor heterogeneity. EMT characterization may probably be restricted to the tumor invasive front (7). Thus, EMT represents a challenging therapeutic target due to their heterogeneous expression and to the complexity of the EMT regulation network.

Inhibition of EMT could restore senescence and apoptosis capacity (2,12). In a recent review, we and others reviewed distinct strategies to target EMT through:

  • Extracellular inducers of EMT, like TGF-β blockade (fresolimumab), HIF-1α downregulation (13), MMP inhibitors (14)
  • EMT-transcription factors inhibition, targeting TWIST1 (15), PRMT1 (16), glycosylation (17), chromatin (18).
  • Downstream effectors of EMT inhibition (19).

MicroRNAs, as EMT regulators, have emerged as a class of therapeutics targets (2). However there are some difficulties because of multiple targets per miRNAs. This can either be harmful due to collateral adverse events but it may also be of interest as miRNA blockage could inhibit a pathway at different levels. Therapeutic strategy could be either direct administration of anti-miRNAs (antisense miRNAs) to block oncomiRs, or restoration of tumor suppressor miRNAs expression (2). In NSCLC, enforced expression of miR-145 inhibited EMT and metastatic ability (20). Sato et al. showed that the introduction of miR-200c using pre-miR-200c caused LIN28B suppression in cells with acquired EGFR-TKI resistance that harboured EMT features (HCC4006 after chronic exposure to gefitinib) (21).

Nevertheless, inhibiting or reversing EMT could also lead to a serious adverse event: favouring MET and thus colonisation of metastatic sites by circulating tumor cells (22,23). Furthermore, tumor heterogeneity and the dynamic of EMT encourages the development of multimodal strategy, not focusing only on a single EMT-related target. As examples, combination of anti-miRNA with chemotherapy strategies and also models of miRNA replacement therapy were tested in vitro and in mouse models (21,24). Van Roosbroeck et al. proposed the use of cisplatin and anti-miR-155 in a mouse model of athymic nude mice (intrapulmonary injections of A549 cells stably infected with lentivirus containing a miR-155-overexpressing lentiviral vector) with significant results in term of primary tumour size and mediastinal lymph nodes (24). Here, by stimulating pro-apoptosis effectors, down-stream to EMT, as investigated by Song and co-authors, might not enhance malignant MET.

Finally, this article should be emphasized based on the rigorous work and on the potential near-future application, in the complex and large field of EMT in cancer.


Acknowledgements

We thank the Journal of Thoracic Disease editorial board for its solicitation to write this editorial.


Footnote

Conflicts of Interest: The authors have no conflicts of interest to declare.


References

  1. Camidge DR, Pao W, Sequist LV. Acquired resistance to TKIs in solid tumours: learning from lung cancer. Nat Rev Clin Oncol 2014;11:473-81. [Crossref] [PubMed]
  2. Legras A, Pécuchet N, Imbeaud S, et al. Epithelial-to-Mesenchymal Transition and MicroRNAs in Lung Cancer. Cancers (Basel) 2017.9. [PubMed]
  3. Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest 2009;119:1420-8. [Crossref] [PubMed]
  4. Thompson EW, Haviv I. The social aspects of EMT-MET plasticity. Nat Med 2011;17:1048-9. [Crossref] [PubMed]
  5. Song KA, Niederst MJ, Lochmann TL, et al. Epithelial-to-Mesenchymal Transition Antagonizes Response to Targeted Therapies in Lung Cancer by Suppressing BIM. Clin Cancer Res 2018;24:197-208. [Crossref] [PubMed]
  6. Yochum ZA, Cades J, Wang H, et al. Targeting the EMT transcription factor TWIST1 overcomes resistance to EGFR inhibitors in EGFR-mutant non-small-cell lung cancer. Oncogene 2018. [Epub ahead of print]. [Crossref] [PubMed]
  7. Grigore AD, Jolly MK, Jia D, et al. Tumor Budding: The Name is EMT. Partial EMT. J Clin Med 2016.5. [PubMed]
  8. Jordan NV, Johnson GL, Abell AN. Tracking the intermediate stages of epithelial-mesenchymal transition in epithelial stem cells and cancer. Cell Cycle 2011;10:2865-73. [Crossref] [PubMed]
  9. Umbreit C, Flanjak J, Weiss C, et al. Incomplete epithelial-mesenchymal transition in p16-positive squamous cell carcinoma cells correlates with β-catenin expression. Anticancer Res 2014;34:7061-9. [PubMed]
  10. Schliekelman MJ, Taguchi A, Zhu J, et al. Molecular portraits of epithelial, mesenchymal, and hybrid States in lung adenocarcinoma and their relevance to survival. Cancer Res 2015;75:1789-800. [Crossref] [PubMed]
  11. Jia D, Jolly MK, Tripathi SC, et al. Distinguishing mechanisms underlying EMT tristability. Cancer Converg 2017;1:2. [Crossref] [PubMed]
  12. Weinberg RA. Twisted epithelial-mesenchymal transition blocks senescence. Nat Cell Biol 2008;10:1021-3. [Crossref] [PubMed]
  13. Goto Y, Zeng L, Yeom CJ, et al. UCHL1 provides diagnostic and antimetastatic strategies due to its deubiquitinating effect on HIF-1α. Nat Commun 2015;6:6153. [Crossref] [PubMed]
  14. Sparano JA, Bernardo P, Stephenson P, et al. Randomized phase III trial of marimastat versus placebo in patients with metastatic breast cancer who have responding or stable disease after first-line chemotherapy: Eastern Cooperative Oncology Group trial E2196. J Clin Oncol 2004;22:4683-90. [Crossref] [PubMed]
  15. Burns TF, Dobromilskaya I, Murphy SC, et al. Inhibition of TWIST1 leads to activation of oncogene-induced senescence in oncogene-driven non-small cell lung cancer. Mol Cancer Res 2013;11:329-38. [Crossref] [PubMed]
  16. Avasarala S, Van Scoyk M, Karuppusamy Rathinam MK, et al. PRMT1 Is a Novel Regulator of Epithelial-Mesenchymal-Transition in Non-small Cell Lung Cancer. J Biol Chem 2015;290:13479-89. [Crossref] [PubMed]
  17. Trapannone R, Rafie K, van Aalten DMF. O-GlcNAc transferase inhibitors: current tools and future challenges. Biochem Soc Trans 2016;44:88-93. [Crossref] [PubMed]
  18. Wagner T, Jung M. New lysine methyltransferase drug targets in cancer. Nat Biotechnol 2012;30:622-3. [Crossref] [PubMed]
  19. Malek R, Wang H, Taparra K, et al. Therapeutic Targeting of Epithelial Plasticity Programs: Focus on the Epithelial-Mesenchymal Transition. Cells Tissues Organs 2017;203:114-27. [Crossref] [PubMed]
  20. Garg M. Targeting microRNAs in epithelial-to-mesenchymal transition-induced cancer stem cells: therapeutic approaches in cancer. Expert Opin Ther Targets 2015;19:285-97. [Crossref] [PubMed]
  21. Sato H, Shien K, Tomida S, et al. Targeting the miR-200c/LIN28B axis in acquired EGFR-TKI resistance non-small cell lung cancer cells harboring EMT features. Sci Rep 2017;7:40847. [Crossref] [PubMed]
  22. Ocaña OH, Córcoles R, Fabra A, et al. Metastatic colonization requires the repression of the epithelial-mesenchymal transition inducer Prrx1. Cancer Cell 2012;22:709-24. [Crossref] [PubMed]
  23. Tsai JH, Donaher JL, Murphy DA, et al. Spatiotemporal regulation of epithelial-mesenchymal transition is essential for squamous cell carcinoma metastasis. Cancer Cell 2012;22:725-36. [Crossref] [PubMed]
  24. Van Roosbroeck K, Fanini F, Setoyama T, et al. Combining Anti-Mir-155 with Chemotherapy for the Treatment of Lung Cancers. Clin Cancer Res 2017;23:2891-904. [Crossref] [PubMed]
Cite this article as: Garinet S, Didelot A, Garelli E, Pallier K, Blons H, Legras A. How apoptosis and epithelial-to-mesenchymal transition are nested in EGFR inhibitors resistance in lung cancer. J Thorac Dis 2019;11(1):47-49. doi: 10.21037/jtd.2018.12.117

Download Citation