Skip to main content

Advertisement

Log in

Establishment and characterization of NCC-MRT1-C1: a novel cell line of malignant rhabdoid tumor

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

A Correction to this article was published on 18 October 2022

This article has been updated

Abstract

Malignant rhabdoid tumor (MRT) is a sarcoma histologically characterized by rhabdoid cells and genetically characterized by loss of function of the chromatin remodeling complex SWI/SNF induced by SMARCB1 gene deficiency. MRT mainly occurs in children, may arise in various locations, but is predominantly in the central nervous system (CNS) and kidney. Although MRT exhibits poor prognosis, standard treatment has not yet been established due to its extreme rarity. Patient-derived cancer cell lines are critical tools for basic and pre-clinical research in the development of chemotherapy. However, none of the MRT cell lines was derived from adult patients, and only one cell line was derived from the MRT of a soft tissue, despite the clinical behavior of MRT varying according to patient age and anatomic site. Herein, we reported the first cell line of MRT isolated from the soft tissue of an adult patient and named it NCC-MRT1-C1. NCC-MRT1-C1 cells showed a biallelic loss of the SMARCB1 gene. NCC-MRT1-C1 cells demonstrated rapid proliferation, spheroid formation, invasion capability in vitro, and tumorigenesis in nude mice. Screening of antitumor agents in NCC-MRT1-C1 cells resulted in the identification of six effective drugs. In conclusion, we report the first MRT cell line from the soft tissue of an adult patient. We believe that NCC-MRT1-C1 is a useful tool for developing novel chemotherapies for MRT.

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.

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

Similar content being viewed by others

Change history

References

  1. Brennan B, Stiller C, Bourdeaut F. Extracranial rhabdoid tumours: what we have learned so far and future directions. Lancet Oncol. 2013;14:e329–36.

    Article  Google Scholar 

  2. Eaton KW, Tooke LS, Wainwright LM, Judkins AR, Biegel JA. Spectrum of SMARCB1/INI1 mutations in familial and sporadic rhabdoid tumors. Pediatr Blood Cancer. 2011;56:7–15.

    Article  Google Scholar 

  3. Kohashi K, Yamada Y, Hotokebuchi Y, et al. ERG and SALL4 expressions in SMARCB1/INI1-deficient tumors: a useful tool for distinguishing epithelioid sarcoma from malignant rhabdoid tumor. Hum Pathol. 2015;46:225–30.

    Article  CAS  Google Scholar 

  4. Kohashi K, Tanaka Y, Kishimoto H, et al. Reclassification of rhabdoid tumor and pediatric undifferentiated/unclassified sarcoma with complete loss of SMARCB1/INI1 protein expression: three subtypes of rhabdoid tumor according to their histological features. Mod Pathol. 2016;29:1232–42.

    Article  CAS  Google Scholar 

  5. Madigan CE, Armenian SH, Malogolowkin MH, Mascarenhas L. Extracranial malignant rhabdoid tumors in childhood: the Childrens Hospital Los Angeles experience. Cancer. 2007;110:2061–6.

    Article  Google Scholar 

  6. Oda Y, Tsuneyoshi M. Extrarenal rhabdoid tumors of soft tissue: clinicopathological and molecular genetic review and distinction from other soft-tissue sarcomas with rhabdoid features. Pathol Int. 2006;56:287–95.

    Article  Google Scholar 

  7. Cai W, Liu X, Ge W, et al. Factors affecting the outcomes of patients with malignant rhabdoid tumors: a population-based study. Int J Med Sci. 2021;18:911–20.

    Article  Google Scholar 

  8. Sultan I, Qaddoumi I, Rodríguez-Galindo C, Nassan AA, Ghandour K, Al-Hussaini M. Age, stage, and radiotherapy, but not primary tumor site, affects the outcome of patients with malignant rhabdoid tumors. Pediatr Blood Cancer. 2010;54:35–40.

    Article  Google Scholar 

  9. Frühwald MC, Hasselblatt M, Nemes K, et al. Age and DNA methylation subgroup as potential independent risk factors for treatment stratification in children with atypical teratoid/rhabdoid tumors. Neuro Oncol. 2020;22:1006–17.

    Article  Google Scholar 

  10. Torchia J, Golbourn B, Feng S, et al. Integrated (epi)-Genomic analyses identify subgroup-specific therapeutic targets in CNS rhabdoid tumors. Cancer Cell. 2016;30:891–908.

    Article  CAS  Google Scholar 

  11. Johann PD, Erkek S, Zapatka M, et al. Atypical teratoid/rhabdoid tumors are comprised of three epigenetic subgroups with distinct enhancer landscapes. Cancer Cell. 2016;29:379–93.

    Article  CAS  Google Scholar 

  12. Bourdeaut F, Chi SN, Frühwald MC. Rhabdoid tumors: integrating biological insights with clinical success: a report from the SMARCB1 and Rhabdoid Tumor Symposium, Paris, December 12–14, 2013. Cancer Genet. 2014;207:346–51.

    Article  Google Scholar 

  13. Brennan B, De Salvo GL, Orbach D, et al. Outcome of extracranial malignant rhabdoid tumours in children registered in the European Paediatric Soft Tissue Sarcoma Study Group Non-Rhabdomyosarcoma Soft Tissue Sarcoma 2005 Study-EpSSG NRSTS 2005. Eur J Cancer. 2016;60:69–82.

    Article  Google Scholar 

  14. Mittal P, Roberts CWM. The SWI/SNF complex in cancer—biology, biomarkers and therapy. Nat Rev Clin Oncol. 2020;17:435–48.

    Article  CAS  Google Scholar 

  15. Shain AH, Pollack JR. The spectrum of SWI/SNF mutations, ubiquitous in human cancers. PLoS ONE. 2013;8: e55119.

    Article  Google Scholar 

  16. Kadoch C, Hargreaves DC, Hodges C, et al. Proteomic and bioinformatic analysis of mammalian SWI/SNF complexes identifies extensive roles in human malignancy. Nat Genet. 2013;45:592–601.

    Article  CAS  Google Scholar 

  17. Orlando KA, Nguyen V, Raab JR, Walhart T, Weissman BE. Remodeling the cancer epigenome: mutations in the SWI/SNF complex offer new therapeutic opportunities. Expert Rev Anticancer Ther. 2019;19:375–91.

    Article  CAS  Google Scholar 

  18. Savas S, Skardasi G. The SWI/SNF complex subunit genes: their functions, variations, and links to risk and survival outcomes in human cancers. Crit Rev Oncol Hematol. 2018;123:114–31.

    Article  Google Scholar 

  19. Hadfield KD, Newman WG, Bowers NL, et al. Molecular characterisation of SMARCB1 and NF2 in familial and sporadic schwannomatosis. J Med Genet. 2008;45:332–9.

    Article  CAS  Google Scholar 

  20. Smith MJ, Walker JA, Shen Y, Stemmer-Rachamimov A, Gusella JF, Plotkin SR. Expression of SMARCB1 (INI1) mutations in familial schwannomatosis. Hum Mol Genet. 2012;21:5239–45.

    Article  CAS  Google Scholar 

  21. Smith MJ, Wallace AJ, Bowers NL, Eaton H, Evans DG. SMARCB1 mutations in schwannomatosis and genotype correlations with rhabdoid tumors. Cancer Genet. 2014;207:373–8.

    Article  CAS  Google Scholar 

  22. Wang X, Werneck MB, Wilson BG, et al. TCR-dependent transformation of mature memory phenotype T cells in mice. J Clin Invest. 2011;121:3834–45.

    Article  CAS  Google Scholar 

  23. Misawa A, Hosoi H, Imoto I, Iehara T, Sugimoto T, Inazawa J. Translocation (1;22)(p36;q11.2) with concurrent del(22)(q11.2) resulted in homozygous deletion of SNF5/INI1 in a newly established cell line derived from extrarenal rhabdoid tumor. J Hum Genet. 2004; 49:586–9.

  24. Hideyuki T, Yoko T, Daisuke M, Miharu K, Tohru M, Hiroshi M. Cell line individualization by STR multiplex system in the cell bank found cross-contamination between ECV304 AND EJ-1/T24. Tissue Cult Res Commun. 1999;18:329–38.

    Google Scholar 

  25. Masters JR, Thomson JA, Daly-Burns B, et al. Short tandem repeat profiling provides an international reference standard for human cell lines. Proc Natl Acad Sci USA. 2001;98:8012–7.

    Article  CAS  Google Scholar 

  26. Drexler HG, Dirks WG, MacLeod RA, Uphoff CC. False and mycoplasma-contaminated leukemia-lymphoma cell lines: time for a reappraisal. Int J Cancer. 2017;140:1209–14.

    Article  CAS  Google Scholar 

  27. Yoshimatsu Y, Noguchi R, Tsuchiya R, et al. Establishment and characterization of NCC-CDS2-C1: a novel patient-derived cell line of CIC-DUX4 sarcoma. Hum Cell. 2020;33:427–36.

    Article  Google Scholar 

  28. Tate JG, Bamford S, Jubb HC, et al. COSMIC: the Catalogue of somatic mutations in Cancer. Nucleic Acids Res. 2019;47:D941–7.

    Article  CAS  Google Scholar 

  29. 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 

  30. Cox C, Teknos TN, Barrios M, Brewer GJ, Dick RD, Merajver SD. The role of copper suppression as an antiangiogenic strategy in head and neck squamous cell carcinoma. Laryngoscope. 2001;111:696–701.

    Article  CAS  Google Scholar 

  31. Lee RS, Stewart C, Carter SL, et al. A remarkably simple genome underlies highly malignant pediatric rhabdoid cancers. J Clin Invest. 2012;122:2983–8.

    Article  CAS  Google Scholar 

  32. Kieran MW, Roberts CW, Chi SN, et al. Absence of oncogenic canonical pathway mutations in aggressive pediatric rhabdoid tumors. Pediatr Blood Cancer. 2012;59:1155–7.

    Article  Google Scholar 

  33. Watanabe M, Adachi S, Matsubara H, et al. Induction of autophagy in malignant rhabdoid tumor cells by the histone deacetylase inhibitor FK228 through AIF translocation. Int J Cancer. 2009;124:55–67.

    Article  CAS  Google Scholar 

  34. Sugimoto Y, Katsumi Y, Iehara T, et al. The novel histone deacetylase inhibitor, OBP-801, induces apoptosis in rhabdoid tumors by releasing the silencing of NOXA. Mol Cancer Ther. 2020;19:1992–2000.

    Article  CAS  Google Scholar 

  35. Tran HM, Wu KS, Sung SY, et al. Upregulation of protein synthesis and proteasome degradation confers sensitivity to proteasome inhibitor bortezomib in myc-atypical teratoid/rhabdoid tumors. Cancers (Basel). 2020; 12.

  36. Howard TP, Oberlick EM, Rees MG, et al. Rhabdoid tumors are sensitive to the protein-translation inhibitor homoharringtonine. Clin Cancer Res. 2020;26:4995–5006.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Drs. E. Kobayashi, F. Nakatani, S. Iwata, K. Ogura, K. Ozaki, S. Fukushima, K. Sato, S. Ishihara Y. Toda (Department of Musculoskeletal Oncology), K. Yokoyama (Department of Diagnostic Pathology), and the National Cancer Center Hospital for sampling tumor tissue specimens from surgically resected materials. We also appreciate the technical assistance provided by Mrs. Y. Kuwata (Division of Rare Cancer Research) and the technical support provided by Mrs. Y. Shiotani, Mr. N. Uchiya, and Dr. T. Imai (Central Animal Division, National Cancer Center Research Institute). We wish to thank Editage (www.editage.jp) for providing English language editing services, and constructive comments on this manuscript. Technical assistance was rendered by the Fundamental Innovative Oncology Core of the National Cancer Center.

Funding

This research was supported by the Japan Agency for Medical Research and Development (Grant No. 20ck0106537h0002).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Tadashi Kondo.

Ethics declarations

Conflict of interest

The authors have no relevant financial or non-financial interests to disclose.

Ethics approval

The ethical committee of the National Cancer Center approved the use of clinical materials for this study (approval number 2004–050). Animal experiments were conducted in compliance with the guidelines of the Institute for Laboratory Animal Research, National Cancer Center Research Institute.

Informed consent

Written informed consent 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

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

Akiyama, T., Yoshimatsu, Y., Noguchi, R. et al. Establishment and characterization of NCC-MRT1-C1: a novel cell line of malignant rhabdoid tumor. Human Cell 35, 2002–2010 (2022). https://doi.org/10.1007/s13577-022-00751-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13577-022-00751-z

Keywords

Navigation