Skip to main content

Advertisement

Log in

Establishment and characterization of NCC-DDLPS1-C1: a novel patient-derived cell line of dedifferentiated liposarcoma

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

Abstract

Dedifferentiated liposarcoma (DDLPS) is one of the four subtypes of liposarcomas; it is characterized by the amplification of the 12q13-15 region, which includes MDM2 and CDK4 genes. DDLPS has an extremely high local recurrence rate and is refractory to chemotherapy and radiation, which leads to poor prognosis. Therefore, a novel therapeutic strategy should be urgently established for improving the prognosis of DDLPS. Although patient-derived cell lines are important tools for basic research, there are no DDLPS cell lines available from public cell banks. Here, we report the establishment of a novel DDLPS cell line. Using the surgically resected tumor tissue from a patient with DDLPS, we established a cell line and named it NCC-DDLPS1-C1. The NCC-DDLPS1-C1 cells contained 12q13-15, 1p32, and 1q23 amplicons and highly expressed MDM2 and CDK4 proteins. NCC-DDLPS-C1 cells exhibited constant growth, spheroid formation, aggressive invasion, and tumorigenesis in mice. By screening a drug library, we identified that the proteasome inhibitor, bortezomib, had inhibitory effects on the proliferation of NCC-DDLPS1-C1 cells. We concluded that the NCC-DDLPS1-C1 cell line may serve as a useful tool for basic and pre-clinical studies of DDLPS.

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
Fig. 6

Similar content being viewed by others

References

  1. The WHO Classification of Tumors Editorial Board. WHO classification of tumours of soft tissue and bone. 5th ed. Lyon: IARC; 2020.

    Google Scholar 

  2. Dei Tos AP. Liposarcomas: diagnostic pitfalls and new insights. Histopathology. 2014;64:38–52.

    Article  Google Scholar 

  3. Evans HL. Liposarcoma: a study of 55 cases with a reassessment of its classification. Am J Surg Pathol. 1979;3:507–23.

    Article  CAS  Google Scholar 

  4. Hirata M, Asano N, Katayama K, et al. Integrated exome and RNA sequencing of dedifferentiated liposarcoma. Nat Commun. 2019;10:5683.

    Article  CAS  Google Scholar 

  5. Kanojia D, Nagata Y, Garg M, et al. Genomic landscape of liposarcoma. Oncotarget. 2015;6:42429–44.

    Article  Google Scholar 

  6. Asano N, Yoshida A, Mitani S, et al. Frequent amplification of receptor tyrosine kinase genes in well differentiated/dedifferentiated liposarcoma. Oncotarget. 2017;8:12941–52.

    Article  Google Scholar 

  7. Tap WD, Eilber FC, Ginther C, et al. Evaluation of well-differentiated/de-differentiated liposarcomas by high-resolution oligonucleotide array-based comparative genomic hybridization. Genes Chromosom Cancer. 2011;50:95–112.

    Article  CAS  Google Scholar 

  8. Louis-Brennetot C, Coindre JM, Ferreira C, Perot G, Terrier P, Aurias A. The CDKN2A/CDKN2B/CDK4/CCND1 pathway is pivotal in well-differentiated and dedifferentiated liposarcoma oncogenesis: an analysis of 104 tumors. Genes Chromosom Cancer. 2011;50:896–907.

    Article  CAS  Google Scholar 

  9. Italiano A, Bianchini L, Gjernes E, et al. Clinical and biological significance of CDK4 amplification in well-differentiated and dedifferentiated liposarcomas. Clin Cancer Res. 2009;15:5696–703.

    Article  CAS  Google Scholar 

  10. Italiano A, Bianchini L, Keslair F, et al. HMGA2 is the partner of MDM2 in well-differentiated and dedifferentiated liposarcomas whereas CDK4 belongs to a distinct inconsistent amplicon. Int J Cancer. 2008;122:2233–41.

    Article  CAS  Google Scholar 

  11. Snyder EL, Sandstrom DJ, Law K, et al. c-Jun amplification and overexpression are oncogenic in liposarcoma but not always sufficient to inhibit the adipocytic differentiation programme. J Pathol. 2009;218:292–300.

    Article  CAS  Google Scholar 

  12. Mariani O, Brennetot C, Coindre JM, et al. JUN oncogene amplification and overexpression block adipocytic differentiation in highly aggressive sarcomas. Cancer Cell. 2007;11:361–74.

    Article  CAS  Google Scholar 

  13. Coindre JM, Mariani O, Chibon F, et al. Most malignant fibrous histiocytomas developed in the retroperitoneum are dedifferentiated liposarcomas: a review of 25 cases initially diagnosed as malignant fibrous histiocytoma. Mod Pathol. 2003;16:256–62.

    Article  Google Scholar 

  14. Coindre JM, Hostein I, Maire G, et al. Inflammatory malignant fibrous histiocytomas and dedifferentiated liposarcomas: histological review, genomic profile, and MDM2 and CDK4 status favour a single entity. J Pathol. 2004;203:822–30.

    Article  CAS  Google Scholar 

  15. Chibon F, Mariani O, Derré J, et al. A subgroup of malignant fibrous histiocytomas is associated with genetic changes similar to those of well-differentiated liposarcomas. Cancer Genet Cytogenet. 2002;139:24–9.

    Article  CAS  Google Scholar 

  16. Le Guellec S, Chibon F, Ouali M, et al. Are peripheral purely undifferentiated pleomorphic sarcomas with MDM2 amplification dedifferentiated liposarcomas? Am J Surg Pathol. 2014;38:293–304.

    Article  Google Scholar 

  17. Gahvari Z, Parkes A. Dedifferentiated liposarcoma: systemic therapy options. Curr Treat Options Oncol. 2020;21:15.

    Article  Google Scholar 

  18. Gootee J, Aurit S, Curtin C, Silberstein P. Primary anatomical site, adjuvant therapy, and other prognostic variables for dedifferentiated liposarcoma. J Cancer Res Clin Oncol. 2019;145:181–92.

    Article  CAS  Google Scholar 

  19. Keung EZ, Hornick JL, Bertagnolli MM, Baldini EH, Raut CP. Predictors of outcomes in patients with primary retroperitoneal dedifferentiated liposarcoma undergoing surgery. J Am Coll Surg. 2014;218:206–17.

    Article  Google Scholar 

  20. Crago AM, Singer S. Clinical and molecular approaches to well differentiated and dedifferentiated liposarcoma. Curr Opin Oncol. 2011;23:373–8.

    Article  CAS  Google Scholar 

  21. 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 

  22. Gisselsson D, Lichtenzstejn D, Kachko P, Karlsson J, Manor E, Mai S. Clonal evolution through genetic bottlenecks and telomere attrition: Potential threats to in vitro data reproducibility. Genes Chromosom Cancer. 2019;58:452–61.

    Article  CAS  Google Scholar 

  23. 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 

  24. 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 

  25. Hattori E, Oyama R, Kondo T. Systematic review of the current status of human sarcoma cell lines. Cells. 2019;8:157.

    Article  CAS  Google Scholar 

  26. 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 

  27. Crystal AS, Shaw AT, Sequist LV, et al. Patient-derived models of acquired resistance can identify effective drug combinations for cancer. Science. 2014;346:1480–6.

    Article  CAS  Google Scholar 

  28. Teicher BA, Polley E, Kunkel M, et al. Sarcoma cell line screen of oncology drugs and investigational agents identifies patterns associated with gene and microRNA expression. Mol Cancer Ther. 2015;14:2452–62.

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

  30. 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 

  31. 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 

  32. Deming DA, Ninan J, Bailey HH, et al. A Phase I study of intermittently dosed vorinostat in combination with bortezomib in patients with advanced solid tumors. Investig New Drugs. 2014;32:323–9.

    Article  CAS  Google Scholar 

  33. Xian M, Cao H, Cao J, et al. Bortezomib sensitizes human osteosarcoma cells to adriamycin-induced apoptosis through ROS-dependent activation of p-eIF2alpha/ATF4/CHOP axis. Int J Cancer. 2017;141:1029–41.

    Article  CAS  Google Scholar 

  34. Perez M, Peinado-Serrano J, Garcia-Heredia JM, et al. Efficacy of bortezomib in sarcomas with high levels of MAP17 (PDZK1IP1). Oncotarget. 2016;7:67033–46.

    Article  Google Scholar 

  35. Garcia-Heredia JM, Lucena-Cacace A, Verdugo-Sivianes EM, Perez M, Carnero A. The cargo protein MAP17 (PDZK1IP1) regulates the cancer stem cell pool activating the Notch pathway by abducting NUMB. Clin Cancer Res. 2017;23:3871–83.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Drs. F Nakatani, E Kobayashi, M Nakagawa, T Komatsubara, M Saito, C Sato (Department of Musculoskeletal Oncology), and Drs. T Shibayama, and H Tanaka (Department of Diagnostic Pathology), National Cancer Center Hospital, for sampling tumor tissue specimens from surgically resected materials. We thank Dr. T Kiyono (Division of Carcinogenesis and Prevention) for kindness in providing the HFF cell line. We also appreciate the technical assistance provided by Mr. K Tanoue, T Ono, and 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 (https://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.

Informed consent

Written informed consent for publication was provided by the patient.

Ethics approval

The ethical committee of the National Cancer Center approved the use of clinical materials for this study with the approval number 2004-050. The study using animal models was approved by the ethics committee of the National Cancer Center Research Institute (T13-016).

Additional information

Publisher's Note

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

Electronic supplementary material

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-DDLPS1-C1: a novel patient-derived cell line of dedifferentiated liposarcoma. Human Cell 34, 260–270 (2021). https://doi.org/10.1007/s13577-020-00436-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13577-020-00436-5

Keywords

Navigation