Human HLA-A*02:01/CHM1+ allo-restricted T cell receptor transgenic CD8+ T Cells specifically inhibit Ewing sarcoma growth in vitro and in vivo

2016 
// Franziska Blaeschke 1, 10, * , Uwe Thiel 1, * , Andreas Kirschner 1 , Melanie Thiede 1 , Rebeca Alba Rubio 2 , David Schirmer 1 , Thomas Kirchner 2, 3, 4 , Gunther H.S. Richter 1 , Sabine Mall 5 , Richard Klar 5 , Stanley Riddell 6 , Dirk H. Busch 7, 8 , Angela Krackhardt 5 , Thomas G.P. Grunewald 2, 3, 4 , Stefan Burdach 1, 9 1 Laboratory for Functional Genomics and Transplantation Biology, Department of Pediatrics and Children‘s Cancer Research Center, Klinikum rechts der Isar, Technische Universitaet Muenchen, Munich, Germany 2 Laboratory for Pediatric Sarcoma Biology, Institute of Pathology of the LMU Munich, Munich, Germany 3 German Cancer Consortium (DKTK), Heidelberg, Germany 4 German Cancer Research Center, Heidelberg, Germany 5 Medizinische Klinik III, Klinikum rechts der Isar, Technische Universitat Munchen, Munich, Germany 6 Department of Medicine, University of Washington, Seattle, WA, USA 7 Institute for Medical Microbiology, Immunology and Hygiene, Technische Universitaet Muenchen, Munchen, Germany 8 Focus Group “Clinical Cell Processing and Purification”, Institute for Advanced Study, Technische Universitat Munchen, Munich, Germany 9 Munich Comprehensive Cancer Center (CCC), Klinikum rechts der Isar, Technische Universitat Munchen, Munich, Germany 10 Present address: Laboratory for Immunotherapy, Dr. von Hauner Children’s Hospital, LMU Munchen, Munich, Germany * These authors share first authorship Correspondence to: Uwe Thiel, email: uwe.thiel@tum.de Keywords: Ewing sarcoma, immunotherapy, T cell receptor transgenic T cells, adoptive transfer, allogeneic stem cell transplantation Received: January 14, 2016      Accepted: April 24, 2016      Published: May 07, 2016 ABSTRACT The endochondral bone protein Chondromodulin-I (CHM1) provides oncogene addiction in Ewing sarcoma (ES). We pre-clinically tested the targetability of CHM1 by TCR transgenic, allo-restricted, peptide specific T cells to treat ES. We previously generated allo-restricted wildtype CD8 + T cells directed against the ES specific antigen CHM1 319 causing specific responses against ES. However, utilization of these cells in current therapy protocols is hampered due to high complexity in production, relatively low cell numbers, and rapid T cell exhaustion. In order to provide off-the-shelf products in the future, we successfully generated HLA-A*02:01-restricted T cell receptor (TCR) transgenic T cells directed against CHM1 319 by retroviral transduction. After short-term expansion a 100% purified CHM1 319 -TCR-transgenic T cell population expressed a CD62L + /CD45RO and CD62L + /CD45RA + phenotype. These cells displayed specific in vitro IFNg and granzyme B release in co-culture with HLA-A*02:01 + ES cell lines expressing CHM1. When co-injected with ES cells in Rag2 -/- ɣc -/- mice, CHM1-specific TCR-transgenic T cells significantly inhibited the formation of lung and liver metastases in contrast to control mice. Lungs and livers of representative mice displayed CD8 + T cell infiltration in the presence (control group treated with unspecific T cells) and in the absence (study group) of metastatic disease, respectively. Furthermore, mice receiving unspecific T cells showed signs of graft-versus-host-disease in contrast to all mice, receiving CHM1 319 -TCR-transgenic T cells. CHM1319 specific TCR-transgenic T cells were successfully generated causing anti-ES responses in vitro and in vivo . In the future, CHM1 319 -TCR-transgenic T cells may control minimal residual disease rendering donor lymphocyte infusions more efficacious and less toxic.
    • Correction
    • Source
    • Cite
    • Save
    • Machine Reading By IdeaReader
    30
    References
    15
    Citations
    NaN
    KQI
    []