English
 
Help Privacy Policy Disclaimer
  Advanced SearchBrowse

Item

ITEM ACTIONSEXPORT

Released

Journal Article

Human T cells loaded with superparamagnetic iron oxide nanoparticles retain antigen-specific TCR functionality

MPS-Authors
/persons/resource/persons255631

Kubánková,  Markéta
Guck Division, Max Planck Institute for the Science of Light, Max Planck Society;
Max-Planck-Zentrum für Physik und Medizin, Max Planck Institute for the Science of Light, Max Planck Society;

/persons/resource/persons241284

Guck,  Jochen
Guck Division, Max Planck Institute for the Science of Light, Max Planck Society;
Max-Planck-Zentrum für Physik und Medizin, Max Planck Institute for the Science of Light, Max Planck Society;
Friedrich-Alexander-Universität Erlangen-Nürnberg, Department Physik;

External Resource
No external resources are shared
Fulltext (restricted access)
There are currently no full texts shared for your IP range.
Fulltext (public)

Front Immunol 2023 Pfister.pdf
(Publisher version), 5MB

Supplementary Material (public)
There is no public supplementary material available
Citation

Pfister, F., Dörrie, J., Schaft, N., Buchele, V., Unterweger, H., Carnell, L. R., et al. (2023). Human T cells loaded with superparamagnetic iron oxide nanoparticles retain antigen-specific TCR functionality. Frontiers in Immunology, 14: 1223695. doi:10.3389/fimmu.2023.1223695.


Cite as: https://hdl.handle.net/21.11116/0000-000D-9993-0
Abstract
BACKGROUND: Immunotherapy of cancer is an emerging field with the potential to improve long-term survival. Thus far, adoptive transfer of tumor-specific T cells represents an effective treatment option for tumors of the hematological system such as lymphoma, leukemia or myeloma. However, in solid tumors, treatment efficacy is low owing to the immunosuppressive microenvironment, on-target/off-tumor toxicity, limited extravasation out of the blood vessel, or ineffective trafficking of T cells into the tumor region. Superparamagnetic iron oxide nanoparticles (SPIONs) can make cells magnetically controllable for the site-specific enrichment. METHODS: In this study, we investigated the influence of SPION-loading on primary human T cells for the magnetically targeted adoptive T cell therapy. For this, we analyzed cellular mechanics and the T cell response after stimulation via an exogenous T cell receptor (TCR) specific for the melanoma antigen MelanA or the endogenous TCR specific for the cytomegalovirus antigen pp65 and compared them to T cells that had not received SPIONs. RESULTS: SPION-loading of human T cells showed no influence on cellular mechanics, therefore retaining their ability to deform to external pressure. Additionally, SPION-loading did not impair the T cell proliferation, expression of activation markers, cytokine secretion, and tumor cell killing after antigen-specific activation mediated by the TCR. CONCLUSION: In summary, we demonstrated that SPION-loading of T cells did not affect cellular mechanics or the functionality of the endogenous or an exogenous TCR, which allows future approaches using SPIONs for the magnetically enrichment of T cells in solid tumors.