Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Neuro-Oncology, Oxford University Press (OUP), Vol. 25, No. Supplement_2 ( 2023-09-08), p. ii4-ii4
    Abstract: Effective treatment options for meningioma patients beyond surgical resection and radiotherapy are lacking. Somatostatin receptor (SSTR) 2 is expressed by most meningiomas. Here, we report pre-clinical and translational evidence for the activity of an SSTR2-targeted adaptor CAR T cell system in meningiomas. MATERIAL AND METHODS Tissue microarrays of meningioma (N=384) patient samples stained for SSTR2. Fluorescein-linked, octameric cyclic peptide SSTR2 antagonist OCTO-FLUO (kindly provided by Philogen, Otelfingen, Switzerland). Adaptor FITC CAR T-cells generated by lentiviral transduction of healthy donor T-cells. Flow cytometry-based killing assays in vitro in SSTR2-expressing meningioma (Ben-Men-1 IOMM-Lee) cells, and ex vivo utilizing freshly dissociated surgical meningioma specimens, by co-incubation with OCTO-FLUO and adaptor FITC CAR T-cells. Orthotopic growth and mouse survival following subdural implantation of IOMM-Lee cells, genetically engineered to express luciferase, i.v. treated with OCTO-FLUO and intratumoral injection of CAR T-cells. RESULTS SSTR2 expression was overall heterogeneous and was high or intermediate in most World Health Organization (WHO) grade 2 (87.8 %) or grade 3 (87.5 %) meningiomas. In vitro, 10 nM was determined as the optimal concentration of OCTO-FLUO, yielding tumor cell killing within 72 hours at effector-target cell (ET) ratios as low as 1:100 in Ben-Men-1 (CAR vs untransduced T-cells [UTT] 47.9 % vs 4.7 %, p & lt; 0.001) and IOMM-Lee (CAR vs UTT 40.2 % vs 4.9 %, p & lt; 0.001) cells. Tumor growth inhibition by the adaptor FITC CAR T-cell system in vivo was confined to SSTR2 expressing cells and was paralleled by prolonged survival compared to UTT in IOMM-Lee (hazard ratio [95% confidence interval], 0.3 [0.1 - 0.9] , p=0.030). Preliminary ex vivo killing assays employing freshly dissociated patient samples yielded specific lysis-rates in the range of 15-20% (ET 1:1) at 12 hours. Updated ex vivo results employing optimized protocols will be presented at the conference. CONCLUSION Targeting SSTR2 positive meningioma cells with an adaptor FITC CAR T-cell system is feasible. Syngeneic murine models and the use of adaptor molecules directed against murine SSTR2 will be required to explore secondary immune reactions and off-tumor effects prior to proceeding to early phase clinical trials.
    Type of Medium: Online Resource
    ISSN: 1522-8517 , 1523-5866
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2023
    detail.hit.zdb_id: 2094060-9
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    Oxford University Press (OUP) ; 2023
    In:  Neuro-Oncology Vol. 25, No. Supplement_2 ( 2023-09-08), p. ii12-ii12
    In: Neuro-Oncology, Oxford University Press (OUP), Vol. 25, No. Supplement_2 ( 2023-09-08), p. ii12-ii12
    Abstract: Chimeric antigen receptor (CAR) T cell therapy has emerged as a potent immunotherapy against hematological malignancies. In glioblastoma, however, limited infiltration of the tumor by CAR T cells, tumor heterogeneity, antigen escape and an immunosuppressive tumor microenvironment remain significant obstacles. Vessel-targeting CAR T cells have been shown to infiltrate solid tumors more efficiently and have the additional benefit of co-targeting the tumor vasculature with genomic stability. Therefore, vascular endothelial growth factor receptor 2 (VEGFR2)-CAR T cells may represent a promising strategy by targeting the tumor vasculature as well as VEGFR2-expressing tumor cells. Here, we explored the efficacy of VEGFR2-specific CAR T cells against experimental gliomas as well as the contribution of anti-tumor and anti-vasculature-dependent effects of this strategy. METHODS Tissue microarrays of glioblastoma patients (n=113) were stained for VEGFR2 expression. Human CAR T cells were generated by lentiviral transduction to express a second generation CAR construct against either mouse or human VEGFR2 (mVEGFR2 or hVEGFR2). Their activity was assessed in co-culture assays in vitro against murine endothelial and human glioma cells, respectively. Several orthotopic xenograft mouse glioma models were used to test the in vivo activity of the newly generated CAR T cells. RESULTS We confirmed high VEGFR2 expression on endothelial cells in glioblastoma tissue of stained tissue microarrays as well as in 20.3% of tissue samples also on tumor cells. In co-culture assays, hVEGFR2-CAR T cells were exclusively active against human glioma cells and mVEGFR2-CAR T cells against mouse endothelial cells, respectively. The specificity of hVEGFR2-CAR T cells was confirmed by a CRISPR/Cas9-mediated knockout of hVEGFR2 in a human glioma cell line. In all three in vivo glioma models, intratumoral treatment of hVEGFR2-CAR T cells significantly prolonged the survival of glioma-bearing mice and cured a substantial fraction of these animals in one model. Additionally, we found that survival was prolonged after mVEGFR2-CAR T cell treatment in one glioma model, which correlated with high vascularization of these tumors. CONCLUSION Our dataset demonstrates that VEGFR2-CAR T cells prolong the survival of glioma-bearing mice through anti-glioma and anti-glioma vasculature activity. The results suggest that the magnitude of the vasculature-targeting activity depends on vessel-density within the tumor. VEGFR2 might be a relevant target that could be exploited for a novel CAR T cell-based immunotherapeutic approach against glioblastoma.
    Type of Medium: Online Resource
    ISSN: 1522-8517 , 1523-5866
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2023
    detail.hit.zdb_id: 2094060-9
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    Oxford University Press (OUP) ; 2018
    In:  Neuro-Oncology Vol. 20, No. suppl_3 ( 2018-09-19), p. iii325-iii325
    In: Neuro-Oncology, Oxford University Press (OUP), Vol. 20, No. suppl_3 ( 2018-09-19), p. iii325-iii325
    Type of Medium: Online Resource
    ISSN: 1522-8517 , 1523-5866
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2018
    detail.hit.zdb_id: 2094060-9
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 25, No. 6 ( 2019-03-15), p. 1913-1922
    Abstract: Glioblastoma (GBM) is the most common primary malignant tumor in the central nervous system. Our recent preclinical work has suggested that PD-1/PD-L1 plays an important immunoregulatory role to limit effective antitumor T-cell responses induced by active immunotherapy. However, little is known about the functional role that PD-1 plays on human T lymphocytes in patients with malignant glioma. Experimental Design: In this study, we examined the immune landscape and function of PD-1 expression by T cells from tumor and peripheral blood in patients with malignant glioma. Results: We found several differences between PD-1+ tumor-infiltrating lymphocytes (TIL) and patient-matched PD-1+ peripheral blood T lymphocytes. Phenotypically, PD-1+ TILs exhibited higher expression of markers of activation and exhaustion than peripheral blood PD-1+ T cells, which instead had increased markers of memory. A comparison of the T-cell receptor variable chain populations revealed decreased diversity in T cells that expressed PD-1, regardless of the location obtained. Functionally, peripheral blood PD-1+ T cells had a significantly increased proliferative capacity upon activation compared with PD-1− T cells. Conclusions: Our evidence suggests that PD-1 expression in patients with glioma reflects chronically activated effector T cells that display hallmarks of memory and exhaustion depending on its anatomic location. The decreased diversity in PD-1+ T cells suggests that the PD-1–expressing population has a narrower range of cognate antigen targets compared with the PD-1 nonexpression population. This information can be used to inform how we interpret immune responses to PD-1–blocking therapies or other immunotherapies.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages