Skip to main content

Advertisement

Log in

TCR extracellular domain genetically linked to CD28, 2B4/41BB and DAP10/CD3ζ -engineered NK cells mediates antitumor effects

  • Research Report
  • Published:
Cancer Immunology, Immunotherapy Aims and scope Submit manuscript

Abstract

NK cells, especially FDA-approved NK-92 cells, could be used for TCR engineering owing to their specialized cytotoxicity against tumors, safety profile and potential use as an off-the-shelf cellular therapy. The TCR complex requires assembly of TCR- α/ β chains with CD3 molecules (CD3δ, CD3γ, CD3ε, CD3ζ) to be correctly expressed at the cell membrane, and yet NK cells lack expression of these CD3 subunits besides CD3ζ. Since transmembrane regions of TCR α and β chains are involved in TCR complex assembly, transmembrane regions of TCR replaced by CD28 transmembrane domain could result in the expression of TCR independent of its companion CD3 subunits. However, since the absence of CD3 signaling components can influence the transmission of TCR signals to NK cells, it is necessary to add the signaling molecules of NK cells followed by CD28 transmembrane domain. Both CD3ζ and DAP10 play an important role in the activation and cytotoxicity of NK cells; moreover, 2B4 and 4-1BB are the main costimulatory molecules in NK cells. Therefore, we designed a chimeric TCR that consisted of the extracellular domains of the TCR α and β chains specific for NYESO-1 fused to the CD28 transmembrane domain followed by the 41BB and CD3ζ signaling domains as well as the 2B4 and DAP10 signaling domain, respectively. The chimeric TCR genetically engineered NK-92 cells exhibit antigen-specific recognition and lysis of tumor cells both in vitro and in vivo. In addition, TCR-28-2B10/BBζ can be feasibly expressed in primary NK cells and exhibit antigen-reactive recognition and effect function. The overall encouraging data highlight the value of NK-92 cells and primary NK cells engineered to express therapeutic chimeric TCR for adoptive immunotherapies.

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

Similar content being viewed by others

Data availability

The datasets used or analyzed during the current study are available from the corresponding author on reasonable request.

References

  1. Sadelain M, Riviere I, Riddell S (2017) Therapeutic T cell engineering. Nature 545(7655):423–431

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Daher M, Rezvani K (2020) Outlook for new CAR-based therapies with a focus on CAR NK cells: what lies beyond CAR-engineered t cells in the race against cancer. Cancer discov 1:45–58

    Google Scholar 

  3. Parlar A, Sayitoglu EC, Ozkazanc D, Georgoudaki AM, Pamukcu C, Aras M et al (2019) Engineering antigen-specific NK cell lines against the melanoma-associated antigen tyrosinase via TCR gene transfer. Eur J Immunol 49(8):1278–1290

    CAS  PubMed  Google Scholar 

  4. Mensali N, Dillard P, Hebeisen M, Lorenz S, Theodossiou T, Myhre MR et al (2019) NK cells specifically TCR-dressed to kill cancer cells. EBioMedicine 40:106–117

    Article  PubMed  PubMed Central  Google Scholar 

  5. Liu E, Marin D, Banerjee P, Macapinlac HA, Thompson P, Basar R et al (2020) Use of CAR-Transduced Natural Killer Cells in CD19-Positive Lymphoid Tumors. N Engl J Med 382(6):545–553

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Morton LT, Wachsmann TL, Meeuwsen MH, Wouters AK, Remst DF, van Loenen MM, Falkenburg JF, Heemskerk MH (2022) T cell receptor engineering of primary NK cells to therapeutically target tumors and tumor immune evasion. J immunother cancer 10(3):e003715

    Article  PubMed  PubMed Central  Google Scholar 

  7. Shimasaki N, Jain A, Campana D (2020) NK cells for cancer immunotherapy. Nat Rev Drug Discovery 19(3):200–218

    Article  CAS  PubMed  Google Scholar 

  8. Milone MC, Xu J, Chen SJ, Collins MA, Zhou J, Powell DJ Jr et al (2021) Engineering enhanced CAR T-cells for improved cancer therapy. Nat Cancer 2(8):780–793

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Imai C, Iwamoto S, Campana D (2005) Genetic modification of primary natural killer cells overcomes inhibitory signals and induces specific killing of leukemic cells. Blood 106(1):376–383

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Xu Y, Liu Q, Zhong M, Wang Z, Chen Z, Zhang Y et al (2019) 2B4 costimulatory domain enhancing cytotoxic ability of anti-CD5 chimeric antigen receptor engineered natural killer cells against T cell malignancies. J Hematol Oncol 12(1):49

    Article  PubMed  PubMed Central  Google Scholar 

  11. Tan Q, Zhang C, Yang W, Liu Y, Heyilimu P, Feng D et al (2019) Isolation of T cell receptor specifically reactive with autologous tumour cells from tumour-infiltrating lymphocytes and construction of T cell receptor engineered T cells for esophageal squamous cell carcinoma. J Immunother Cancer 7(1):232

    Article  PubMed  PubMed Central  Google Scholar 

  12. Zhang C, Tan Q, Li S, Shen L, Zhang J, Liu Y et al (2021) Induction of EBV latent membrane protein-2A (LMP2A)-specific T cells and construction of individualized TCR-engineered T cells for EBV-associated malignancies. J Immunother Cancer. 9(7):e002516

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by Natural Science Foundation of China [Grant No 81972880–ZL, Grant No 82003246–CZ]; Capital’s Funds for Health Improvement and Research (Grant No 2022-1-1022–ZL, 2020-4-1028–CZ); Open Project funded by Key laboratory of Carcinogenesis and Translational Research, Ministry of Education/Beijing (2022 Open Project-1); Cooperation Fund of Beijing Cancer Hospital and Beijing Institute for Cancer Research; Clinical Medicine Plus X—Young Scholars Project (PKU2022LCXQ036), Peking University, the Fundamental Research Funds for the Central Universities.

Author information

Authors and Affiliations

Authors

Contributions

ZML, BTY and CTZ designed the research; SCL, CTZ, LYS, XT and YFX conducted experiments; SCL, CTZ, and ZML analyzed data; and CTZ, ZML and SCL wrote the paper.

Corresponding authors

Correspondence to Bentong Yu or Zheming Lu.

Ethics declarations

Conflict of interest

The authors declare that they have no competing interests.

Ethics approval

This project was approved by the Institutional Review Board of the Peking University School of Oncology, China. The patient gave written informed consent for the use of their tumor tissues for research purposes.

Consent for publication

All authors have approved for publication.

Additional information

Publisher's Note

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

Rights and permissions

Springer Nature or its licensor 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

Li, S., Zhang, C., Shen, L. et al. TCR extracellular domain genetically linked to CD28, 2B4/41BB and DAP10/CD3ζ -engineered NK cells mediates antitumor effects. Cancer Immunol Immunother 72, 769–774 (2023). https://doi.org/10.1007/s00262-022-03275-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00262-022-03275-5

Keywords

Navigation