Skip to main content

Advertisement

Log in

Drug screening with a novel tumor-derived cell line identified alternative therapeutic options for patients with atypical teratoid/rhabdoid tumor

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

Abstract

Atypical teratoid/rhabdoid tumor (AT/RT) is a rare intracranial tumor occurring predominantly in young children. The prognosis is poor, and no effective treatment is currently available. To develop novel effective therapies, there is a need for experimental models for AT/RT. In this research, we established a cell line from a patient’s AT/RT tissue (designated ATRT_OCGH) and performed drug screening using 164 FDA-approved anti-cancer agents, to identify candidates for therapeutic options. We found that bortezomib, a proteasome inhibitor, was among the agents for which the cell line showed high sensitivity, along with tyrosine kinase inhibitors, topoisomerase inhibitors, and histone deacetylase inhibitors, which are known to exert anti-AT/RT effects. Concomitant use of panobinostat potentiated the inhibitory effect of bortezomib on AT/RT cell proliferation. Our findings may provide a rationale for considering combination therapy of panobinostat and bortezomib for treatment of AT/RT.

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

Similar content being viewed by others

References

  1. Fruhwald MC, Biegel JA, Bourdeaut F, Roberts CW, Chi SN. Atypical teratoid/rhabdoid tumors-current concepts, advances in biology, and potential future therapies. Neuro-oncology. 2016;18:764–78.

    Article  Google Scholar 

  2. Richardson EA, Ho B, Huang A. Atypical teratoid rhabdoid tumour: from tumours to therapies. J Korean Neurosurg Soc. 2018;61:302–11.

    Article  CAS  Google Scholar 

  3. Yamasaki K, Kiyotani C, Terashima K, et al. Clinical characteristics, treatment, and survival outcome in pediatric patients with atypical teratoid/rhabdoid tumors: a retrospective study by the Japan Children’s Cancer Group. J Neurosurg Pediatr. 2019;25:1–10.

    Google Scholar 

  4. 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 

  5. 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 

  6. Narendran A, Coppes L, Jayanthan A, et al. Establishment of atypical-teratoid/rhabdoid tumor (AT/RT) cell cultures from disseminated CSF cells: a model to elucidate biology and potential targeted therapeutics. J Neuro-Oncol. 2008;90:171–80.

    Article  CAS  Google Scholar 

  7. Yoshimatsu Y, Noguchi R, Tsuchiya R, et al. Establishment and characterization of NCC-DFSP3-C1: a novel patient-derived dermatofibrosarcoma protuberans cell line. Hum Cell. 2020;33:877.

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

  9. Moreau P. How I treat myeloma with new agents. Blood. 2017;130:1507–13.

    Article  CAS  Google Scholar 

  10. San-Miguel JF, Hungria VT, Yoon SS, et al. Panobinostat plus bortezomib and dexamethasone versus placebo plus bortezomib and dexamethasone in patients with relapsed or relapsed and refractory multiple myeloma: a multicentre, randomised, double-blind phase 3 trial. Lancet Oncol. 2014;15:1195–206.

    Article  CAS  Google Scholar 

  11. Hideshima T, Richardson P, Chauhan D, et al. The proteasome inhibitor PS-341 inhibits growth, induces apoptosis, and overcomes drug resistance in human multiple myeloma cells. Cancer Res. 2001;61:3071–6.

    CAS  PubMed  Google Scholar 

  12. Adams J. The proteasome: a suitable antineoplastic target. Nat Rev Cancer. 2004;4:349–60.

    Article  CAS  Google Scholar 

  13. Cheah CY, Seymour JF, Wang ML. Mantle cell lymphoma. J Clin Oncol. 2016;34:1256–69.

    Article  CAS  Google Scholar 

  14. Carugo A, Minelli R, Sapio L, et al. p53 is a aster regulator of proteostasis in SMARCB1-efficient malignant rhabdoid tumors. Cancer Cell. 2019;35:204–20.

    Article  CAS  Google Scholar 

  15. Wu DY, Tkachuck DC, Roberson RS, Schubach WH. The human SNF5/INI1 protein facilitates the function of the growth arrest and DNA damage-inducible protein (GADD34) and modulates GADD34-bound protein phosphatase-1 activity. J Biol Chem. 2002;277:27706–15.

    Article  CAS  Google Scholar 

  16. Hertwig F, Meyer K, Braun S, et al. Definition of genetic events directing the development of distinct types of brain tumors from postnatal neural stem/progenitor cells. Cancer Res. 2012;72:3381–92.

    Article  CAS  Google Scholar 

  17. Pakos-Zebrucka K, Koryga I, Mnich K, et al. The integrated stress response. EMBO Rep. 2016;17:1374–95.

    Article  CAS  Google Scholar 

  18. Schewe DM, Aguirre-Ghiso JA. Inhibition of eIF2alpha dephosphorylation maximizes bortezomib efficiency and eliminates quiescent multiple myeloma cells surviving proteasome inhibitor therapy. Cancer Res. 2009;69:1545–52.

    Article  CAS  Google Scholar 

  19. Hollmann TJ, Hornick JL. INI1-deficient tumors: diagnostic features and molecular genetics. Am J Surg Pathol. 2011;35:e47–e63.

    Article  Google Scholar 

  20. Carden MA, Smith S, Meany H, et al. Platinum plus bortezomib for the treatment of pediatric renal medullary carcinoma: two cases. Pediatr Blood Cancer. 2017;64:e26402.

    Article  Google Scholar 

  21. Ronnen EA, Kondagunta GV, Motzer RJ. Medullary renal cell carcinoma and response to therapy with bortezomib. J Clin Oncol. 2006;24:e14.

    Article  Google Scholar 

  22. Raizer JJ, Chandler JP, Ferrarese R, et al. A phase II trial evaluating the effects and intra-tumoral penetration of bortezomib in patients with recurrent malignant gliomas. J Neuro-Oncol. 2016;129:139–46.

    Article  CAS  Google Scholar 

  23. Hideshima T, Richardson PG, Anderson KC. Mechanism of action of proteasome inhibitors and deacetylase inhibitors and the biological basis of synergy in multiple myeloma. Mol Cancer Ther. 2011;10:2034–42.

    Article  CAS  Google Scholar 

  24. Kawaguchi Y, Kovacs JJ, McLaurin A, et al. The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress. Cell. 2003;115:727–38.

    Article  CAS  Google Scholar 

  25. Moreno N, Kerl K. Preclinical evaluation of combined targeted approaches in malignant rhabdoid tumors. Anticancer Res. 2016;36:3883–7.

    CAS  PubMed  Google Scholar 

  26. Jochems J, Boulden J, Lee BG, et al. Antidepressant-like properties of novel HDAC6-selective inhibitors with improved brain bioavailability. Neuropsychopharmacology. 2014;39:389–400.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank the patient and their family for participating in this research. We thank Y. Matsushita and K. Hatake for their technical support.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Yoshiko Nakano.

Ethics declarations

Conflict of interest

The authors declare that they have no conflicts of interest.

Ethics approval

The study was approved by the Institutional Review Boards at all participating institutions.

Informed consent

Informed, written consent was obtained from the patient’s parent.

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

Nakano, Y., Takadera, M., Miyazaki, M. et al. Drug screening with a novel tumor-derived cell line identified alternative therapeutic options for patients with atypical teratoid/rhabdoid tumor. Human Cell 34, 271–278 (2021). https://doi.org/10.1007/s13577-020-00438-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13577-020-00438-3

Keywords

Navigation