Your browser doesn't support javascript.
loading
In vivo nanoparticle-based T cell imaging can predict therapy response towards adoptive T cell therapy in experimental glioma.
Hunger, Jessica; Schregel, Katharina; Boztepe, Berin; Agardy, Dennis Alexander; Turco, Verena; Karimian-Jazi, Kianush; Weidenfeld, Ina; Streibel, Yannik; Fischer, Manuel; Sturm, Volker; Santarella-Mellwig, Rachel; Kilian, Michael; Jähne, Kristine; Sahm, Katharina; Wick, Wolfgang; Bunse, Lukas; Heiland, Sabine; Bunse, Theresa; Bendszus, Martin; Platten, Michael; Breckwoldt, Michael O.
Afiliação
  • Hunger J; Neuroradiology Department, University Hospital Heidelberg, Heidelberg, Germany.
  • Schregel K; Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DKTK) within the German Cancer Research Center (DKFZ), Heidelberg, Germany.
  • Boztepe B; Neuroradiology Department, University Hospital Heidelberg, Heidelberg, Germany.
  • Agardy DA; Neuroradiology Department, University Hospital Heidelberg, Heidelberg, Germany.
  • Turco V; Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DKTK) within the German Cancer Research Center (DKFZ), Heidelberg, Germany.
  • Karimian-Jazi K; Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DKTK) within the German Cancer Research Center (DKFZ), Heidelberg, Germany.
  • Weidenfeld I; Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences, Heidelberg University, Mannheim, Germany.
  • Streibel Y; Neuroradiology Department, University Hospital Heidelberg, Heidelberg, Germany.
  • Fischer M; Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DKTK) within the German Cancer Research Center (DKFZ), Heidelberg, Germany.
  • Sturm V; Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences, Heidelberg University, Mannheim, Germany.
  • Santarella-Mellwig R; Neuroradiology Department, University Hospital Heidelberg, Heidelberg, Germany.
  • Kilian M; Neuroradiology Department, University Hospital Heidelberg, Heidelberg, Germany.
  • Jähne K; Neuroradiology Department, University Hospital Heidelberg, Heidelberg, Germany.
  • Sahm K; Neuroradiology Department, University Hospital Heidelberg, Heidelberg, Germany.
  • Wick W; Neuroradiology Department, University Hospital Heidelberg, Heidelberg, Germany.
  • Bunse L; European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
  • Heiland S; Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DKTK) within the German Cancer Research Center (DKFZ), Heidelberg, Germany.
  • Bunse T; Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences, Heidelberg University, Mannheim, Germany.
  • Bendszus M; Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DKTK) within the German Cancer Research Center (DKFZ), Heidelberg, Germany.
  • Platten M; Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences, Heidelberg University, Mannheim, Germany.
  • Breckwoldt MO; Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DKTK) within the German Cancer Research Center (DKFZ), Heidelberg, Germany.
Theranostics ; 13(15): 5170-5182, 2023.
Article em En | MEDLINE | ID: mdl-37908732
ABSTRACT
Rationale Intrinsic brain tumors, such as gliomas are largely resistant to immunotherapies including immune checkpoint blockade. Adoptive cell therapies (ACT) including chimeric antigen receptor (CAR) or T cell receptor (TCR)-transgenic T cell therapy targeting glioma-associated antigens are an emerging field in glioma immunotherapy. However, imaging techniques for non-invasive monitoring of adoptively transferred T cells homing to the glioma microenvironment are currently lacking.

Methods:

Ultrasmall iron oxide nanoparticles (NP) can be visualized non-invasively by magnetic resonance imaging (MRI) and dedicated MRI sequences such as T2* mapping. Here, we develop a protocol for efficient ex vivo labeling of murine and human TCR-transgenic and CAR T cells with iron oxide NPs. We assess labeling efficiency and T cell functionality by flow cytometry and transmission electron microscopy (TEM). NP labeled T cells are visualized by MRI at 9.4 T in vivo after adoptive T cell transfer and correlated with 3D models of cleared brains obtained by light sheet microscopy (LSM).

Results:

NP are incorporated into T cells in subcellular cytoplasmic vesicles with high labeling efficiency without interfering with T cell viability, proliferation and effector function as assessed by cytokine secretion and antigen-specific killing assays in vitro. We further demonstrate that adoptively transferred T cells can be longitudinally monitored intratumorally by high field MRI at 9.4 Tesla in a murine glioma model with high sensitivity. We find that T cell influx and homogenous spatial distribution of T cells within the TME as assessed by T2* imaging predicts tumor response to ACT whereas incomplete T cell coverage results in treatment resistance.

Conclusion:

This study showcases a rational for monitoring adoptive T cell therapies non-invasively by iron oxide NP in gliomas to track intratumoral T cell influx and ultimately predict treatment outcome.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Linfócitos T / Glioma Limite: Animals / Humans Idioma: En Ano de publicação: 2023 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Linfócitos T / Glioma Limite: Animals / Humans Idioma: En Ano de publicação: 2023 Tipo de documento: Article