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

Molecular features and evolutionary trajectory of ASCL1+ and NEUROD1+ SCLC cells

Abstract

Background

Small cell lung cancer (SCLC) is the most aggressive subtype of lung cancer without recognised morphologic or genetic heterogeneity. Based on the expression of four transcription factors, ASCL1, NEUROD1, POU2F3, and YAP1, SCLCs are classified into four subtypes. However, biological functions of these different subtypes are largely uncharacterised.

Methods

We studied intratumoural heterogeneity of resected human primary SCLC tissues using single-cell RNA-Seq. In addition, we undertook a series of in vitro and in vivo functional studies to reveal the distinct features of SCLC subtypes.

Results

We identify the coexistence of ASCL1+ and NEUROD1+ SCLC cells within the same human primary SCLC tissue. Compared with ASCL1+ SCLC cells, NEUROD1+ SCLC cells show reduced epithelial features and lack EPCAM expression. Thus, EPCAM can be considered as a cell surface marker to distinguish ASCL1+ SCLC cells from NEUROD1+ SCLC cells. We further demonstrate that NEUROD1+ SCLC cells exhibit higher metastatic capability than ASCL1+ SCLC cells and can be derived from ASCL1+ SCLC cells.

Conclusions

Our studies unveil the biology and evolutionary trajectory of ASCL1+ and NEUROD1+ SCLC cells, shedding light on SCLC tumourigenesis and progression.

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: Single-cell RNA-Seq revealed the intratumoural heterogeneity of SCLC1.
Fig. 2: NEUROD1+ SCLC cells showed reduced epithelial features and lack EPCAM expression.
Fig. 3: Identification of EPCAM as a cell surface marker to distinguish ASCL1+ SCLC cells from NEUROD1+ SCLC cells.
Fig. 4: ASCL1+ cancer cells can transform into NEUROD1+ cancer cells.
Fig. 5: NEUROD1+ cells exhibited higher metastatic capability than ASCL1+ cells both in vivo and in vitro analyses.

Similar content being viewed by others

Data availability

The data generated in this study are publicly available in the Gene Expression Omnibus (GEO) database at GSE164145 and GSE164404. The data that supported the findings of this study are available in the Human Tumour Atlas Network (HTAN) database (RU426, RU1066, RU1145 and RU1229A) [12].

References

  1. Gazdar AF, Bunn PA, Minna JD. Small-cell lung cancer: what we know, what we need to know and the path forward. Nat Rev Cancer. 2017;17:725–37.

    Article  CAS  PubMed  Google Scholar 

  2. Rudin CM, Poirier JT, Byers LA, Dive C, Dowlati A, George J, et al. Molecular subtypes of small cell lung cancer: a synthesis of human and mouse model data. Nat Rev Cancer. 2019;19:289–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. George J, Lim JS, Jang SJ, Cun Y, Ozretic L, Kong G, et al. Comprehensive genomic profiles of small cell lung cancer. Nature. 2015;524:47–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Peifer M, Fernandez-Cuesta L, Sos ML, George J, Seidel D, Kasper LH, et al. Integrative genome analyses identify key somatic driver mutations of small-cell lung cancer. Nat Genet. 2012;44:1104–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Rudin CM, Durinck S, Stawiski EW, Poirier JT, Modrusan Z, Shames DS, et al. Comprehensive genomic analysis identifies SOX2 as a frequently amplified gene in small-cell lung cancer. Nat Genet. 2012;44:1111–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Calbo J, van Montfort E, Proost N, van Drunen E, Beverloo HB, Meuwissen R, et al. A functional role for tumor cell heterogeneity in a mouse model of small cell lung cancer. Cancer Cell. 2011;19:244–56.

    Article  CAS  PubMed  Google Scholar 

  7. Borromeo MD, Savage TK, Kollipara RK, He M, Augustyn A, Osborne JK, et al. ASCL1 and NEUROD1 reveal heterogeneity in pulmonary neuroendocrine tumors and regulate distinct genetic programs. Cell Rep. 2016;16:1259–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Bottger F, Semenova EA, Song JY, Ferone G, van der Vliet J, Cozijnsen M, et al. Tumor heterogeneity underlies differential cisplatin sensitivity in mouse models of small-cell lung cancer. Cell Rep. 2019;27:3345–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Rudin CM, Ismaila N, Hann CL, Malhotra N, Movsas B, Norris K, et al. Treatment of small-cell lung cancer: American Society of Clinical Oncology Endorsement of the American College of Chest Physicians Guideline. J Clin Oncol. 2015;33:4106–11.

    Article  CAS  PubMed  Google Scholar 

  10. Semenova EA, Kwon MC, Monkhorst K, Song JY, Bhaskaran R, Krijgsman O, et al. Transcription factor NFIB is a driver of small cell lung cancer progression in mice and marks metastatic disease in patients. Cell Rep. 2016;16:631–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Zhang W, Girard L, Zhang YA, Haruki T, Papari-Zareei M, Stastny V, et al. Small cell lung cancer tumors and preclinical models display heterogeneity of neuroendocrine phenotypes. Transl Lung Cancer Res. 2018;7:32–49.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Chan JM, Quintanal-Villalonga A, Gao VR, Xie YB, Allaj V, Chaudhary O, et al. Signatures of plasticity, metastasis, and immunosuppression in an atlas of human small cell lung cancer. Cancer Cell. 2021;39:1479–96. e18

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Drapkin BJ, Rudin CM. Advances in small-cell lung cancer (SCLC) translational research. Cold Spring Harb Perspect Med. 2021;11:a038240.

    Article  CAS  PubMed  Google Scholar 

  14. Huang YH, Klingbeil O, He XY, Wu XLS, Arun G, Lu B, et al. POU2F3 is a master regulator of a tuft cell-like variant of small cell lung cancer. Gene Dev. 2018;32:915–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Ireland AS, Micinski AM, Kastner DW, Guo B, Wait SJ, Spainhower KB, et al. MYC drives temporal evolution of small cell lung cancer subtypes by reprogramming neuroendocrine fate. Cancer Cell. 2020;38:60.e12–78.e12.

    Article  Google Scholar 

  16. Stewart CA, Gay CM, Xi YX, Sivajothi S, Sivakamasundari V, Fujimoto J, et al. Single-cell analyses reveal increased intratumoral heterogeneity after the onset of therapy resistance in small-cell lung cancer. Nat Cancer. 2020;1:423–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Reyfman PA, Walter JM, Joshi N, Anekalla KR, McQuattie-Pimentel AC, Chiu S, et al. Single-cell transcriptomic analysis of human lung provides insights into the pathobiology of pulmonary fibrosis. Am J Respir Crit Care. 2019;199:1517–36.

    Article  CAS  Google Scholar 

  18. Cao JY, Spielmann M, Qiu XJ, Huang XF, Ibrahim DM, Hill AJ, et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature. 2019;566:496–502.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Bergen V, Lange M, Peidli S, Wolf FA, Theis FJ. Generalizing RNA velocity to transient cell states through dynamical modeling. Nat Biotechnol. 2020;38:1408–14.

    Article  CAS  PubMed  Google Scholar 

  20. Patel AP, Tirosh I, Trombetta JJ, Shalek AK, Gillespie SM, Wakimoto H, et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science. 2014;344:1396–401.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Tirosh I, Venteicher AS, Hebert C, Escalante LE, Patel AP, Yizhak K, et al. Single-cell RNA-seq supports a developmental hierarchy in human oligodendroglioma. Nature. 2016;539:309–13.

    Article  PubMed  PubMed Central  Google Scholar 

  22. George J, Walter V, Peifer M, Alexandrov LB, Seidel D, Leenders F, et al. Integrative genomic profiling of large-cell neuroendocrine carcinomas reveals distinct subtypes of high-grade neuroendocrine lung tumors. Nat Commun. 2018;9:1048.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Gay CM, Stewart CA, Park EM, Diao LX, Groves SM, Heeke S, et al. Patterns of transcription factor programs and immune pathway activation define four major subtypes of SCLC with distinct therapeutic vulnerabilities. Cancer Cell. 2021;39:346.e7–60.e7.

    Article  Google Scholar 

  24. Wang YZ, Bian TF, Song LN, Jiang YH, Huo ZG, Salloum RG, et al. Reducing chemotherapy-induced DNA damage via nAChR-mediated redox reprograming-a new mechanism for SCLC chemoresistance boosted by nicotine. Cancers. 2022;14:2272.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Wu XLS, He XY, Ipsaro JJ, Huang YH, Preall JB, Ng D, et al. OCA-T1 and OCA-T2 are coactivators of POU2F3 in the tuft cell lineage. Nature. 2022;607:169–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Augustyn A, Borromeo M, Wang T, Fujimoto J, Shao C, Dospoy PD, et al. ASCL1 is a lineage oncogene providing therapeutic targets for high-grade neuroendocrine lung cancers. Proc Natl Acad Sci USA. 2014;111:14788–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Osborne JK, Larsen JE, Shields MD, Gonzales JX, Shames DS, Sato MS, et al. NeuroD1 regulates survival and migration of neuroendocrine lung carcinomas via signaling molecules TrkB and NCAM. Proc Natl Acad Sci USA. 2013;110:6524–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Gazdar AF, Carney DN, Nau MM, Minna JD. Characterization of variant subclasses of cell-lines derived from small cell lung-cancer having distinctive biochemical, morphological, and growth-properties. Cancer Res. 1985;45:2924–30.

    CAS  PubMed  Google Scholar 

  29. Guillemot F, Hassan BA. Beyond proneural: emerging functions and regulations of proneural proteins. Curr Opin Neurobiol. 2017;42:93–101.

    Article  CAS  PubMed  Google Scholar 

  30. Ito T, Udaka N, Yazawa T, Okudela K, Hayashi H, Sudo T, et al. Basic helix-loop-helix transcription factors regulate the neuroendocrine differentiation of fetal mouse pulmonary epithelium. Development. 2000;127:3913–21.

    Article  CAS  PubMed  Google Scholar 

  31. Mollaoglu G, Guthrie MR, Bohm S, Bragelmann J, Can I, Ballieu PM, et al. MYC drives progression of small cell lung cancer to a variant neuroendocrine subtype with vulnerability to Aurora kinase inhibition. Cancer Cell. 2017;31:270–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Gardner EE, Lok BH, Schneeberger VE, Desmeules P, Miles LA, Arnold PK, et al. Chemosensitive relapse in small cell lung cancer proceeds through an EZH2-SLFN11 axis. Cancer Cell. 2017;31:286–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Bevilacqua S, Gallo M, Franco R, Rossi A, De Luca A, Rocco G, et al. A “live” biopsy in a small-cell lung cancer patient by detection of circulating tumor cells. Lung Cancer. 2009;65:123–5.

    Article  PubMed  Google Scholar 

  34. Messaritakis I, Stoltidis D, Kotsakis A, Dermitzaki EK, Koinis F, Lagoudaki E, et al. TTF-1-and/or CD56-positive circulating tumor cells in patients with small cell lung cancer (SCLC). Sci Rep. 2017;7:45351.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Obermayr E, Agreiter C, Schuster E, Fabikan H, Weinlinger C, Baluchova K, et al. Molecular characterization of circulating tumor cells enriched by a microfluidic platform in patients with small-cell lung cancer. Cells. 2019;8:880.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Andersen J, Urban N, Achimastou A, Ito A, Simic M, Ullom K, et al. A transcriptional mechanism integrating inputs from extracellular signals to activate hippocampal stem cells. Neuron. 2014;83:1085–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Castro DS, Martynoga B, Parras C, Ramesh V, Pacary E, Johnston C, et al. A novel function of the proneural factor Ascl1 in progenitor proliferation identified by genome-wide characterization of its targets. Gene Dev. 2011;25:930–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank National Human Genetic Resources Sharing Service Platform (2005DKA21300), the National Key Research and Development programme of China: The Net construction of human genetic resource Biobank in North China (2016YFC1201703) and Cancer Biobank of Tianjin Medical University Cancer Institute and Hospital for providing paraffin sections of human SCLC samples.

Funding

This work was supported by the National Natural Science Foundation of China (grants 81825017, 81773034 to ZL, 8217113342, 81872350 to ZM, 82273119 to ZZ), the Ministry of Science and Technology of China (grant 2018YFC1313002 to ZL), the Tianjin Municipal Science and Technology Commission (20JCZDJC00110 to ZL), and the Haihe Laboratory of Cell Ecosystem Innovation Fund (HH22KYZX0025 to ZL).

Author information

Authors and Affiliations

Authors

Contributions

ZL, XZ, and ZM designed the study and wrote the paper. XZ, HW, and WL performed experiments and analysed the results. XZ and WL performed bioinformatics. ZX, JC, WG and ZZ provided lung cancer specimens and resources. GW analysed pathological features of SCLCs. All authors reviewed the manuscript.

Corresponding author

Correspondence to Zhe Liu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Ethics approval and consent to participate

The use of all human lung cancer tissues was approved by the Institutional Review Board of Tianjin Medical University. Informed consents were obtained from all patients, and samples were deidentified prior to analysis. All animal procedures were approved by the Animal Care and Use Committee at Tianjin Medical University and confirmed to the legal mandates and national guidelines for the care and maintenance of laboratory animals.

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

Zhang, X., Wang, H., Liu, W. et al. Molecular features and evolutionary trajectory of ASCL1+ and NEUROD1+ SCLC cells. Br J Cancer 128, 748–759 (2023). https://doi.org/10.1038/s41416-022-02103-y

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41416-022-02103-y

Search

Quick links