Skip to main content

Advertisement

Log in

Establishment and characterization of NCC-ssRMS2-C1: a novel patient-derived cell line of spindle cell/sclerosing rhabdomyosarcoma

  • Cell Line
  • Published:
Human Cell Aims and scope Submit manuscript

Abstract

Spindle cell/sclerosing rhabdomyosarcoma (ssRMS) is a rare subtype of rhabdomyosarcoma (RMS) that has fascicular spindle cell and/or sclerosing morphology. SsRMS has a diverse molecular background and is categorized into three groups: congenital/infantile ssRMS with a gene fusion involving the NCOA2 and VGLL2, ssRMS with the MYOD1 mutation, and ssRMS with no recurrent identifiable genetic alterations. Because ssRMS is a newly defined disease concept of RMS, the optimal treatment methods have not been determined. This results in unfavorable prognosis and consequently signals the urgent need for continuous research. Patient-derived cell lines are essential tools in basic and translational research. However, only two ssRMS cell lines with the MYOD1 mutation have been reported to date. Thus, we established a novel ssRMS cell line named NCC-ssRMS2-C1 using a surgically resected tumor tissue from an adult ssRMS patient. NCC-ssRMS2-C1 cells retained the copy number alterations corresponding to the original tumor and are categorized into the group with no recurrent identifiable genetic alterations. NCC-ssRMS2-C1 cells demonstrated constant proliferation, spheroid formation, and capability for invasion in vitro, reflecting the malignant features of the original tumor tissue. In a drug screening test, ssRMS demonstrated remarkable sensitivity to romidepsin, trabectedin, actinomycin D, and bortezomib. Hence, we conclude that the NCC-ssRMS2-C1 cell line is the first ssRMS cell line which belongs to the group with no recurrent identifiable genetic alterations, and it will be a useful resource in both basic and translational studies for ssRMS.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  1. WHO Classification of Tumours of Soft Tissue and Bone. 5th ed. Lyon: IARC; 2020.

  2. Dziuba I, Kurzawa P, Dopierala M, Larque AB, Januszkiewicz-Lewandowska D. Rhabdomyosarcoma in children - current pathologic and molecular classification. Pol J Pathol. 2018;69:20–32.

    Article  Google Scholar 

  3. Pappo AS, Dirksen U. Rhabdomyosarcoma, ewing sarcoma, and other round cell sarcomas. J Clin Oncol. 2018;36:168–79.

    Article  CAS  Google Scholar 

  4. Rekhi B, Singhvi T. Histopathological, immunohistochemical and molecular cytogenetic analysis of 21 spindle cell/sclerosing rhabdomyosarcomas. APMIS. 2014;122:1144–52.

    PubMed  Google Scholar 

  5. Alaggio R, Zhang L, Sung YS, et al. A molecular study of pediatric spindle and sclerosing rhabdomyosarcoma: identification of novel and recurrent VGLL2-related fusions in infantile cases. Am J Surg Pathol. 2016;40:224–35.

    Article  Google Scholar 

  6. Mosquera JM, Sboner A, Zhang L, et al. Recurrent NCOA2 gene rearrangements in congenital/infantile spindle cell rhabdomyosarcoma. Genes Chromosomes Cancer. 2013;52:538–50.

    Article  CAS  Google Scholar 

  7. Rekhi B, Upadhyay P, Ramteke MP, Dutt A. MYOD1 (L122R) mutations are associated with spindle cell and sclerosing rhabdomyosarcomas with aggressive clinical outcomes. Mod Pathol. 2016;29:1532–40.

    Article  CAS  Google Scholar 

  8. Szuhai K, de Jong D, Leung WY, Fletcher CD, Hogendoorn PC. Transactivating mutation of the MYOD1 gene is a frequent event in adult spindle cell rhabdomyosarcoma. J Pathol. 2014;232:300–7.

    Article  CAS  Google Scholar 

  9. Zhao Z, Yin Y, Zhang J, et al. Spindle cell/sclerosing rhabdomyosarcoma: case series from a single institution emphasizing morphology, immunohistochemistry and follow-up. Int J Clin Exp Pathol. 2015;8:13814–20.

    PubMed  PubMed Central  Google Scholar 

  10. Yasui N, Yoshida A, Kawamoto H, Yonemori K, Hosono A, Kawai A. Clinicopathologic analysis of spindle cell/sclerosing rhabdomyosarcoma. Pediatr Blood Cancer. 2015;62:1011–6.

    Article  CAS  Google Scholar 

  11. Tseng YY, Boehm JS. From cell lines to living biosensors: new opportunities to prioritize cancer dependencies using ex vivo tumor cultures. Curr Opin Genet Dev. 2019;54:33–40.

    Article  CAS  Google Scholar 

  12. Ben-David U, Siranosian B, Ha G, et al. Genetic and transcriptional evolution alters cancer cell line drug response. Nature. 2018;560:325–30.

    Article  CAS  Google Scholar 

  13. Ben-David U, Beroukhim R, Golub TR. Genomic evolution of cancer models: perils and opportunities. Nat Rev Cancer. 2019;19:97–109.

    Article  CAS  Google Scholar 

  14. Saito S, Morita K, Kohara A, et al. Use of BAC array CGH for evaluation of chromosomal stability of clinically used human mesenchymal stem cells and of cancer cell lines. Hum Cell. 2011;24:2–8.

    Article  Google Scholar 

  15. Shoemaker RH. The NCI60 human tumour cell line anticancer drug screen. Nat Rev Cancer. 2006;6:813–23.

    Article  CAS  Google Scholar 

  16. Barretina J, Caponigro G, Stransky N, et al. The cancer cell line encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature. 2012;483:603–7.

    Article  CAS  Google Scholar 

  17. Basu A, Bodycombe NE, Cheah JH, et al. An interactive resource to identify cancer genetic and lineage dependencies targeted by small molecules. Cell. 2013;154:1151–61.

    Article  CAS  Google Scholar 

  18. Yang W, Soares J, Greninger P, et al. Genomics of Drug Sensitivity in Cancer (GDSC): a resource for therapeutic biomarker discovery in cancer cells. Nucleic Acids Res. 2013;41:D955–61.

    Article  CAS  Google Scholar 

  19. Goodspeed A, Heiser LM, Gray JW, Costello JC. Tumor-derived cell lines as molecular models of cancer pharmacogenomics. Mol Cancer Res. 2016;14:3–13.

    Article  CAS  Google Scholar 

  20. Yoshimatsu Y, Noguchi R, Tsuchiya R, et al. Establishment and characterization of NCC-ssRMS1-C1: a novel patient-derived spindle-cell/sclerosing rhabdomyosarcoma cell line. Hum Cell. 2020;33:886–93.

    Article  CAS  Google Scholar 

  21. Schleicher S, Grote S, Malenke E, et al. Establishment and characterization of a sclerosing spindle cell rhabdomyosarcoma cell line with a complex genomic profile. Cells. 2020;9:2668.

    Article  CAS  Google Scholar 

  22. Bairoch A. The cellosaurus, a cell-line knowledge resource. J Biomol Tech. 2018;29:25–38.

    Article  Google Scholar 

  23. Sin Y, Yoshimatsu Y, Noguchi R, et al. Establishment and characterization of a novel alveolar rhabdomyosarcoma cell line, NCC-aRMS1-C1. Hum Cell. 2020;33:1311–20.

    Article  CAS  Google Scholar 

  24. Capes-Davis A, Reid YA, Kline MC, et al. Match criteria for human cell line authentication: where do we draw the line? Int J Cancer. 2013;132:2510–9.

    Article  CAS  Google Scholar 

  25. Tsuchiya R, Yoshimatsu Y, Noguchi R, et al. Establishment and characterization of NCC-DDLPS3-C1: a novel patient-derived cell line of dedifferentiated liposarcoma. Hum Cell. 2021;34:1008–18.

  26. Billiau A, Edy VG, Heremans H, et al. Human interferon: mass production in a newly established cell line, MG-63. Antimicrob Agents Chemother. 1977;12:11–5.

    Article  CAS  Google Scholar 

  27. Walther C, Mayrhofer M, Nilsson J, et al. Genetic heterogeneity in rhabdomyosarcoma revealed by SNP array analysis. Genes Chromosomes Cancer. 2016;55:3–15.

    Article  CAS  Google Scholar 

  28. Yang X, Yang S, Wang C, Kuang S. The hypoxia-inducible factors HIF1alpha and HIF2alpha are dispensable for embryonic muscle development but essential for postnatal muscle regeneration. J Biol Chem. 2017;292:5981–91.

    Article  CAS  Google Scholar 

  29. Demetri GD, von Mehren M, Jones RL, et al. Efficacy and safety of trabectedin or dacarbazine for metastatic liposarcoma or leiomyosarcoma after failure of conventional chemotherapy: results of a phase III randomized multicenter clinical trial. J Clin Oncol. 2016;34:786–93.

    Article  CAS  Google Scholar 

  30. Iyer SP, Foss FF. Romidepsin for the treatment of peripheral T-Cell lymphoma. Oncologist. 2015;20:1084–91.

    Article  CAS  Google Scholar 

  31. Bharathy N, Berlow NE, Wang E, et al. Preclinical rationale for entinostat in embryonal rhabdomyosarcoma. Skelet Muscle. 2019;9:12.

    Article  Google Scholar 

  32. Bharathy N, Berlow NE, Wang E, et al. The HDAC3-SMARCA4-miR-27a axis promotes expression of the PAX3:FOXO1 fusion oncogene in rhabdomyosarcoma. Sci Signal. 2018;11:eaau7632.

    Article  CAS  Google Scholar 

  33. Tomoyasu C, Kikuchi K, Kaneda D, et al. OBP801, a novel histone deacetylase inhibitor, induces Mphase arrest and apoptosis in rhabdomyosarcoma cells. Oncol Rep. 2019;41:643–9.

    CAS  PubMed  Google Scholar 

  34. Gandolfi S, Laubach JP, Hideshima T, Chauhan D, Anderson KC, Richardson PG. The proteasome and proteasome inhibitors in multiple myeloma. Cancer Metastasis Rev. 2017;36:561–84.

    Article  CAS  Google Scholar 

  35. Bersani F, Taulli R, Accornero P, et al. Bortezomib-mediated proteasome inhibition as a potential strategy for the treatment of rhabdomyosarcoma. Eur J Cancer. 2008;44:876–84.

    Article  CAS  Google Scholar 

  36. Peron M, Bonvini P, Rosolen A. Effect of inhibition of the ubiquitin-proteasome system and Hsp90 on growth and survival of rhabdomyosarcoma cells in vitro. BMC Cancer. 2012;12:233.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Drs. E. Kobayashi, S. Iwata, S. Fukushima, M. Nakagawa, T. Komatsubara, C. Sato (Department of Musculoskeletal Oncology), and Drs. N. Kojima, J. Kashima, M. Arakaki (Department of Diagnostic Pathology), National Cancer Center Hospital, for sampling tumor tissue specimens from surgically resected materials. We also appreciate the technical assistance provided by Ms. Y. Kuwata (Division of Rare Cancer Research). We appreciate the technical support provided by Ms. Y. Shiotani, Mr. N. Uchiya, and Dr. T. Imai (Central Animal Division, National Cancer Center Research Institute). We would also like to thank Editage (www.editage.jp) for their help with English language editing and their constructive comments on the manuscript. This research was technically assisted by the Fundamental Innovative Oncology Core in the National Cancer Center.

Funding

This research was supported by the Japan Agency for Medical Research and Development (Grant number 20ck0106537h0001).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Tadashi Kondo.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Ethical approval

The ethical committee of the National Cancer Center approved the use of clinical materials for this study (Approval number 2004–050).

Informed consent

Written informed consent for publication was provided by the patient.

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tsuchiya, R., Yoshimatsu, Y., Noguchi, R. et al. Establishment and characterization of NCC-ssRMS2-C1: a novel patient-derived cell line of spindle cell/sclerosing rhabdomyosarcoma. Human Cell 34, 1569–1578 (2021). https://doi.org/10.1007/s13577-021-00569-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13577-021-00569-1

Keywords

Navigation