Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Cellular and Molecular Biology

MET exon 14 skipping mutation drives cancer progression and recurrence via activation of SMAD2 signalling

Abstract

Background

c-Met encoded by the proto-oncogene MET, also known as hepatocyte growth factor (HGF) receptor, plays a crucial role in cellular processes. MET exon 14 skipping alteration (METΔ14EX) is a newly discovered MET mutation. SMAD2 is an important downstream transcription factor in TGF-β pathway. Unfortunately, the mechanisms by which METΔ14EX leads to oncogenic transformation are scarcely understood. The relationship between METΔ14EX and SMAD2 has not been studied yet.

Methods

We generate METΔ14EX models by CRISPR-Cas9. In vitro transwell, wound-healing, soft-agar assay, in vivo metastasis and subcutaneous recurrence assay were used to study the role of METΔ14EX in tumour progression. RNA-seq, Western blotting, co-immunoprecipitation (CO-IP) and immunofluorescent were performed to explore the interaction between c-Met and SMAD2.

Results

Our results demonstrated that METΔ14EX, independent of HGF, can prolong the constitutive activation of c-Met downstream signalling pathways by impeding c-Met degradation and facilitating tumour metastasis and recurrence. Meanwhile, METΔ14EX strengthens the interaction between c-Met and SMAD2, promoting SMAD2 phosphorylation. Therapeutically, MET inhibitor crizotinib impedes METΔ14EX-mediated tumour metastasis by decreasing SMAD2 phosphorylation.

Conclusions

These data elucidated the previously unrecognised role of METΔ14EX in cancer progression via activation of SMAD2 independent of TGF-β, which helps to develop more effective therapies for such patients.

METΔ14EX alteration significantly triggers tumour progression via activation of SMAD2 signalling that are involved in activating tumour invasion, metastasis and recurrence. On the left, in the MET wild-type (METWT), the juxtamembrane (JM) domain is involved in the regulation of tyrosine kinase activity, receptor degradation, and caspase cleavage. On the right, the METΔ14EX mutation leads to the loss of the juxtamembrane domain, resulting in an abnormal MET protein lacking a CBL-binding site. This causes the accumulation of truncated MET receptors followed by constitutive activation of the MET signalling pathway. Thus, the METΔ14EX-mutated protein has strong binding and phosphorylation to SMAD2, which results in the phosphorylation of a large number of SMAD2/3 proteins that combine with SMAD4 to form a complex in the nucleus, activating downstream signalling pathways, such as EMT and ECM remodelling, resulting in tumour progression and recurrence. TF transcription factor.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: METΔ14EX alteration promotes cellular migration and invasion in vitro, independent of HGF.
Fig. 2: METΔ14EX alteration enhances tumour progression, metastasis and recurrence in vivo.
Fig. 3: RNA-seq analysis reveals that METΔ14EX alteration remarkably upregulated dominant cell movement-associated gene signatures.
Fig. 4: METΔ14EX alteration activates SMAD2 signalling via promoting phosphorylation of SMAD2.
Fig. 5: METΔ14EX-mediated tumour metastasis through activation of SMAD2 signalling.
Fig. 6: Crizotinib dramatically represses METΔ14EX-mediated tumour growth as well as metastasis.

Similar content being viewed by others

Data availability

All data generated or analysed during this study are included in this published article and its supplementary information files.

References

  1. Cooper CS, Park M, Blair DG, Tainsky MA, Huebner K, Croce CM, et al. Molecular cloning of a new transforming gene from a chemically transformed human cell line. Nature. 1984;311:29–33.

    Article  ADS  CAS  PubMed  Google Scholar 

  2. Drilon A, Cappuzzo F, Ou SI, Camidge DR. Targeting MET in lung cancer: will expectations finally be MET? J Thorac Oncol. 2017;12:15–26.

    Article  PubMed  Google Scholar 

  3. Gherardi E, Birchmeier W, Birchmeier C, Vande Woude G Targeting MET in cancer: rationale and progress. Nat Rev Cancer. 2012;12:89–103.

    Article  CAS  PubMed  Google Scholar 

  4. Peng S, Wang R, Zhang X, Ma Y, Zhong L, Li K, et al. EGFR-TKI resistance promotes immune escape in lung cancer via increased PD-L1 expression. Mol Cancer. 2019;18:165.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Hughes VS, Siemann DW. Have clinical trials properly assessed c-Met inhibitors? Trends Cancer. 2018;4:94–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Westover D, Zugazagoitia J, Cho BC, Lovly CM, Paz-Ares L. Mechanisms of acquired resistance to first- and second-generation EGFR tyrosine kinase inhibitors. Ann Oncol. 2018;29:i10–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Cortot AB, Kherrouche Z, Descarpentries C, Wislez M, Baldacci S, Furlan A, et al. Exon 14 deleted MET receptor as a new biomarker and target in cancers. J Natl Cancer Inst. 2017;109.

  8. Lee CC, Yamada KM. Identification of a novel type of alternative splicing of a tyrosine kinase receptor. Juxtamembrane deletion of the c-met protein kinase C serine phosphorylation regulatory site. J Biol Chem. 1994;269:19457–61.

    Article  CAS  PubMed  Google Scholar 

  9. Lee JH, Gao CF, Lee CC, Kim MD, Vande Woude GF. An alternatively spliced form of Met receptor is tumorigenic. Exp Mol Med. 2006;38:565–73.

    Article  CAS  PubMed  Google Scholar 

  10. Lee JM, Kim B, Lee SB, Jeong Y, Oh YM, Song YJ, et al. Cbl-independent degradation of Met: ways to avoid agonism of bivalent Met-targeting antibody. Oncogene. 2014;33:34–43.

    Article  CAS  PubMed  Google Scholar 

  11. Socinski MA, Pennell NA, Davies KD. MET exon 14 skipping mutations in non-small-cell lung cancer: an overview of biology, clinical outcomes, and testing considerations. JCO Precis Oncol. 2021;5:PO.20.00516.

  12. Frampton GM, Ali SM, Rosenzweig M, Chmielecki J, Lu X, Bauer TM, et al. Activation of MET via diverse exon 14 splicing alterations occurs in multiple tumor types and confers clinical sensitivity to MET inhibitors. Cancer Discov. 2015;5:850–9.

    Article  CAS  PubMed  Google Scholar 

  13. Ma PC, Kijima T, Maulik G, Fox EA, Sattler M, Griffin JD, et al. c-MET mutational analysis in small cell lung cancer: novel juxtamembrane domain mutations regulating cytoskeletal functions. Cancer Res. 2003;63:6272–81.

    CAS  PubMed  Google Scholar 

  14. Heist RS, Sequist LV, Borger D, Gainor JF, Arellano RS, Le LP, et al. Acquired resistance to crizotinib in NSCLC with MET exon 14 skipping. J Thorac Oncol. 2016;11:1242–5.

    Article  PubMed  Google Scholar 

  15. Cancer Genome Atlas Research Network. Author Correction: Comprehensive molecular profiling of lung adenocarcinoma. Nature. 2018;559:E12.

    Article  ADS  Google Scholar 

  16. Tong JH, Yeung SF, Chan AW, Chung LY, Chau SL, Lung RW, et al. MET amplification and exon 14 splice site mutation define unique molecular subgroups of non-small cell lung carcinoma with poor prognosis. Clin Cancer Res. 2016;22:3048–56.

    Article  CAS  PubMed  Google Scholar 

  17. Rotow JK, Gui P, Wu W, Raymond VM, Lanman RB, Kaye FJ, et al. Co-occurring alterations in the RAS-MAPK pathway limit response to MET inhibitor treatment in MET exon 14 skipping mutation-positive lung cancer. Clin Cancer Res. 2020;26:439–49.

    Article  CAS  PubMed  Google Scholar 

  18. Suzawa K, Offin M, Lu D, Kurzatkowski C, Vojnic M, Smith RS, et al. Activation of KRAS mediates resistance to targeted therapy in MET exon 14-mutant non-small cell lung cancer. Clin Cancer Res. 2019;25:1248–60.

    Article  CAS  PubMed  Google Scholar 

  19. Bahcall M, Awad MM, Sholl LM, Wilson FH, Xu M, Wang S, et al. Amplification of wild-type KRAS imparts resistance to crizotinib in MET exon 14 mutant non-small cell lung cancer. Clin Cancer Res. 2018;24:5963–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Drilon A, Clark JW, Weiss J, Ou SI, Camidge DR, Solomon BJ, et al. Antitumor activity of crizotinib in lung cancers harboring a MET exon 14 alteration. Nat Med. 2020;26:47–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Lu S, Fang J, Li X, Cao L, Zhou J, Guo Q, et al. Phase II study of savolitinib in patients (pts) with pulmonary sarcomatoid carcinoma (PSC) and other types of non-small cell lung cancer (NSCLC) harboring MET exon 14 skipping mutations (METex14+). J Clin Oncol. 2020;38:9519.

    Article  Google Scholar 

  22. French R, Feng Y, Pauklin S. Targeting TGFbeta signalling in cancer: toward context-specific strategies. Trends Cancer. 2020;6:538–40.

    Article  CAS  PubMed  Google Scholar 

  23. Shi Y, Massague J. Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell. 2003;113:685–700.

    Article  CAS  PubMed  Google Scholar 

  24. Loomans HA, Andl CD. Intertwining of activin A and TGFbeta signaling: dual roles in cancer progression and cancer cell invasion. Cancers (Basel). 2014;7:70–91.

    Article  PubMed  Google Scholar 

  25. Zhong G, Zhao Q, Chen Z, Yao T. TGF-beta signaling promotes cervical cancer metastasis via CDR1as. Mol Cancer. 2023;22:66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Zhang J, van Dinther M, Thorikay M, Gourabi BM, Kruithof BPT, Ten Dijke P. Opposing USP19 splice variants in TGF-beta signaling and TGF-beta-induced epithelial-mesenchymal transition of breast cancer cells. Cell Mol Life Sci. 2023;80:43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Vignjevic D, Montagnac G. Reorganisation of the dendritic actin network during cancer cell migration and invasion. Semin Cancer Biol. 2008;18:12–22.

    Article  CAS  PubMed  Google Scholar 

  28. Izdebska M, Zielinska W, Grzanka D, Gagat M. The role of actin dynamics and actin-binding proteins expression in epithelial-to-mesenchymal transition and its association with cancer progression and evaluation of possible therapeutic targets. Biomed Res Int. 2018;2018:4578373.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Mukai M, Sato S, Kimura T, Komatsu N, Ninomiya H, Nakasaki H, et al. Predicting the recurrence/metastasis of stage II and III breast cancer with lymph node metastasis. Oncol Rep. 2004;12:303–6.

    PubMed  Google Scholar 

  30. Wang F, Liu Y, Qiu W, Shum E, Feng M, Zhao D, et al. Functional analysis of MET exon 14 skipping alteration in cancer invasion and metastatic dissemination. Cancer Res. 2022;82:1365–79.

    Article  CAS  PubMed  Google Scholar 

  31. Hao Y, Baker D, Ten Dijke P. TGF-beta-mediated epithelial-mesenchymal transition and cancer metastasis. Int J Mol Sci. 2019;20:2767.

  32. Buwaneka P, Ralko A, Gorai S, Pham H, Cho W. Phosphoinositide-binding activity of Smad2 is essential for its function in TGF-beta signaling. J Biol Chem. 2021;297:101303.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Collie GW, Koh CM, O’Neill DJ, Stubbs CJ, Khurana P, Eddershaw A, et al. Structural and molecular insight into resistance mechanisms of first generation cMET inhibitors. ACS Med Chem Lett. 2019;10:1322–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Desai A, Cuellar S. The current landscape for METex14 skipping mutations in non-small cell lung cancer. J Adv Pract Oncol. 2022;13:539–44.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Peschard P, Park M. Escape from Cbl-mediated downregulation: a recurrent theme for oncogenic deregulation of receptor tyrosine kinases. Cancer Cell. 2003;3:519–23.

    Article  CAS  PubMed  Google Scholar 

  36. Hong L, Zhang J, Heymach J V, Le X. Current and future treatment options for MET exon 14 skipping alterations in non-small cell lung cancer. Ther Adv Med Oncol. 2021;13:1758835921992976.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Cortot A, Le X, Smit E, Viteri S, Kato T, Sakai H, et al. Safety of MET tyrosine kinase inhibitors in patients with MET exon 14 skipping non-small cell lung cancer: a clinical review. Clin Lung Cancer. 2022;23:195–207.

    Article  CAS  PubMed  Google Scholar 

  38. Wolf J, Garon EB, Groen HJM, Tan DSW, Gilloteau I, Le Mouhaer S, et al. Patient-reported outcomes in capmatinib-treated patients with METex14-mutated advanced NSCLC: results from the GEOMETRY mono-1 study. Eur J Cancer. 2022;183:98–108.

    Article  PubMed  Google Scholar 

  39. Babey H, Jamme P, Curcio H, Assie JB, Veillon R, Doubre H, et al. Real-world treatment outcomes of MET exon14 skipping in non-small cell lung cancer: GFPC 03-18 study. Target Oncol. 2023;18:585–91.

    Article  PubMed  Google Scholar 

  40. Yan SB, Um SL, Peek VL, Stephens JR, Zeng W, Konicek BW, et al. MET-targeting antibody (emibetuzumab) and kinase inhibitor (merestinib) as single agent or in combination in a cancer model bearing MET exon 14 skipping. Invest New Drugs. 2018;36:536–44.

    Article  CAS  PubMed  Google Scholar 

  41. Knowles LM, Stabile LP, Egloff AM, Rothstein ME, Thomas SM, Gubish CT, et al. HGF and c-Met participate in paracrine tumorigenic pathways in head and neck squamous cell cancer. Clin Cancer Res. 2009;15:3740–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Lengyel E, Prechtel D, Resau JH, Gauger K, Welk A, Lindemann K, et al. C-Met overexpression in node-positive breast cancer identifies patients with poor clinical outcome independent of Her2/neu. Int J Cancer. 2005;113:678–82.

    Article  CAS  PubMed  Google Scholar 

  43. Joffre C, Barrow R, Menard L, Calleja V, Hart IR, Kermorgant S. A direct role for Met endocytosis in tumorigenesis. Nat Cell Biol. 2011;13:827–37.

    Article  CAS  PubMed  Google Scholar 

  44. Li X, Wu Y, Tian T. TGF-beta signaling in metastatic colorectal cancer (mCRC): from underlying mechanism to potential applications in clinical development. Int J Mol Sci. 2022;23:14436.

  45. Wang Y, Shi J, Chai K, Ying X, Zhou BP. The role of snail in EMT and tumorigenesis. Curr Cancer Drug Targets. 2013;13:963–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Bakiri L, Macho-Maschler S, Custic I, Niemiec J, Guio-Carrion A, Hasenfuss SC, et al. Fra-1/AP-1 induces EMT in mammary epithelial cells by modulating Zeb1/2 and TGFbeta expression. Cell Death Differ. 2015;22:336–50.

    Article  CAS  PubMed  Google Scholar 

  47. Zhou H, Li L, Xie W, Wu L, Lin Y, He X. TAGLN and High-mobility Group AT-Hook 2 (HMGA2) complex regulates TGF-beta-induced colorectal cancer metastasis. Onco Targets Ther. 2020;13:10489–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Zhu S, Wang W, Clarke DC, Liu X. Activation of Mps1 promotes transforming growth factor-beta-independent Smad signaling. J Biol Chem. 2007;282:18327–38.

    Article  CAS  PubMed  Google Scholar 

  49. Pilotto S, Gkountakos A, Carbognin L, Scarpa A, Tortora G, Bria E. MET exon 14 juxtamembrane splicing mutations: clinical and therapeutical perspectives for cancer therapy. Ann Transl Med. 2017;5:2.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Salgia R, Sattler M, Scheele J, Stroh C, Felip E. The promise of selective MET inhibitors in non-small cell lung cancer with MET exon 14 skipping. Cancer Treat Rev. 2020;87:102022.

    Article  CAS  PubMed  Google Scholar 

  51. Kang J, Deng QM, Feng W, Chen ZH, Su JW, Chen HJ, et al. Response and acquired resistance to MET inhibitors in de novo MET fusion-positive advanced non-small cell lung cancer. Lung Cancer. 2023;178:66–74.

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

This study was supported by the National Natural Science Foundation of China (NSFC 82273293) and Shanghai Municipal Health Commission Health Industry Clinical Research Project (20224Y0120).

Author information

Authors and Affiliations

Authors

Contributions

BZ, MY, HW and QL performed all of the experiments. YH and QY participated in the research. QL, BZ and MY designed experiments, analysed data, and wrote the manuscript. HW provided c-Met inhibitors. QL and BZ are the guarantors of this work and, as such, had full access to all the data in the study and took responsibility for the integrity of the data and the accuracy of the data analysis. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Min Yu, Huijie Wang or Bing Zhao.

Ethics declarations

Competing interests

The authors declare no competing interests.

Ethics approval and consent to participate

All procedures performed in studies involving animals were in accordance with the ethical standards of the Department of Laboratory Animal Science of Fudan University.

Consent for publication

All authors read and approved the final manuscript.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liang, Q., Hu, Y., Yuan, Q. et al. MET exon 14 skipping mutation drives cancer progression and recurrence via activation of SMAD2 signalling. Br J Cancer 130, 380–393 (2024). https://doi.org/10.1038/s41416-023-02495-5

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41416-023-02495-5

Search

Quick links