6 research outputs found

    Abstracts from the 8th International Conference on cGMP Generators, Effectors and Therapeutic Implications

    Get PDF
    This work was supported by a restricted research grant of Bayer AG

    High Expression Levels of the Genes cyclin-A2 and glucocorticoid receptor Are Associated with High-Quality Embryos in Gilthead Sea Bream (Sparus aurata L.)

    No full text
    Identifying early egg-quality predictors is a major challenge in finfish hatcheries, and relevant research is now focused on the development of molecular markers. In our study, we examined whether fertilization rates and early morphological abnormalities in sea bream egg batches of high (HQ) and low quality (LQ) are associated with mRNA levels of cathepsin D, cathepsin Z, cyclin-A2, and glucocorticoid receptor. Additionally, we examined whether these early quality descriptors were associated with the development of skeletal abnormalities during the larval period. HQ egg batches were characterized by significantly higher rates of normal embryos (95.8 ± 2.3%) and lower rates of unfertilized (2.8 ± 1.0%) and abnormal eggs (1.3 ± 1.4%), compared to LQ (84.2 ± 0.8% normal embryos, 12.3 ± 12.3 unfertilized eggs, and 3.5 ± 1.4% abnormal eggs) (p < 0.05, Mann–Whitney U test). Relative expression of cyclin-A2 and glucocorticoid receptor was found to be significantly higher in HQ embryos compared to those of LQ (respectively, p < 0.01 and p < 0.05, Mann–Whitney U test). No statistically significant differences were observed in the mRNA transcripts of cathepsin D and cathepsin Z (p > 0.05, Mann–Whitney U test). Differences in the rate of skeletal abnormalities between the two quality groups of larvae were not significant (p > 0.05, G-test), indicating that cyclin-A2 and glucocorticoid receptor may serve as reliable molecular markers for early prediction of fish egg quality but not for later larval stages

    Ex Vivo Generation of CAR Macrophages from Hematopoietic Stem and Progenitor Cells for Use in Cancer Therapy

    No full text
    Chimeric antigen receptor (CAR) T-cell therapies have shown impressive results in patients with hematological malignancies; however, little success has been achieved in the treatment of solid tumors. Recently, macrophages (M phi s) were identified as an additional candidate for the CAR approach, and initial proof of concept studies using peripheral blood-derived monocytes showed antigen-redirected activation of CAR M phi s. However, some patients may not be suitable for monocyte-apheresis, and prior cancer treatment regimens may negatively affect immune cell number and functionality. To address this problem, we here introduce primary human hematopoietic stem and progenitor cells (HSPCs) as a cell source to generate functional CAR M phi s ex vivo. Our data showed successful CAR expression in cord blood (CB)-derived HSPCs, with considerable cell expansion during differentiation to CAR M phi s. HSPC-derived M phi s showed typical M phi morphology, phenotype, and basic anti-bacterial functionality. CAR M phi s targeting the carcinoembryonic antigen (CEA) and containing either a DAP12- or a CD3 zeta-derived signaling domain showed antigen redirected activation as they secreted pro-inflammatory cytokines specifically upon contact with CEA(+) target cells. In addition, CD3 zeta-expressing CAR M phi s exhibited significantly enhanced phagocytosis of CEA(+) HT1080 cells. Our data establish human HSPCs as a suitable cell source to generate functional CAR M phi s and further support the use of CAR M phi s in the context of solid tumor therapy

    Generation of an NF kappa B-Driven Alpharetroviral All-in-One Vector Construct as a Potent Tool for CAR NK Cell Therapy

    Get PDF
    Immune cell therapeutics are increasingly applied in oncology. Especially chimeric antigen receptor (CAR) T cells are successfully used to treat several B cell malignancies. Efforts to engineer CAR T cells for improved activity against solid tumors include co-delivery of pro-inflammatory cytokines in addition to CARs, via either constitutive cytokine expression or inducible cytokine expression triggered by CAR recognition of its target antigen-so-called T cells redirected for universal cytokine-mediated killing (TRUCKs) or fourth-generation CARs. Here, we tested the hypothesis that TRUCK principles could be expanded to improve anticancer functions of NK cells. A comparison of the functionality of inducible promoters responsive to NFAT or NF kappa B in NK cells showed that, in contrast to T cells, the inclusion of NF kappa B-responsive elements within the inducible promoter construct was essential for CAR-inducible expression of the transgene. We demonstrated that GD2CAR-specific activation induced a tight NF kappa B-promoter-driven cytokine release in NK-92 and primary NK cells together with an enhanced cytotoxic capacity against GD2(+) target cells, also shown by increased secretion of cytolytic cytokines. The data demonstrate biologically relevant differences between T and NK cells that are important when clinically translating the TRUCK concept to NK cells for the treatment of solid malignancies

    GMP-Compliant Manufacturing of TRUCKs: CAR T Cells targeting GD2 and Releasing Inducible IL-18

    No full text
    Chimeric antigen receptor (CAR)-engineered T cells can be highly effective in the treatment of hematological malignancies, but mostly fail in the treatment of solid tumors. Thus, approaches using 4th advanced CAR T cells secreting immunomodulatory cytokines upon CAR signaling, known as TRUCKs (“T cells redirected for universal cytokine-mediated killing”), are currently under investigation. Based on our previous development and validation of automated and closed processing for GMP-compliant manufacturing of CAR T cells, we here present the proof of feasibility for translation of this method to TRUCKs. We generated IL-18-secreting TRUCKs targeting the tumor antigen GD2 using the CliniMACS Prodigy® system using a recently described “all-in-one” lentiviral vector combining constitutive anti-GD2 CAR expression and inducible IL-18. Starting with 0.84 x 108 and 0.91 x 108 T cells after enrichment of CD4+ and CD8+ we reached 68.3-fold and 71.4-fold T cell expansion rates, respectively, in two independent runs. Transduction efficiencies of 77.7% and 55.1% was obtained, and yields of 4.5 x 109 and 3.6 x 109 engineered T cells from the two donors, respectively, within 12 days. Preclinical characterization demonstrated antigen-specific GD2-CAR mediated activation after co-cultivation with GD2-expressing target cells. The functional capacities of the clinical-scale manufactured TRUCKs were similar to TRUCKs generated in laboratory-scale and were not impeded by cryopreservation. IL-18 TRUCKs were activated in an antigen-specific manner by co-cultivation with GD2-expressing target cells indicated by an increased expression of activation markers (e.g. CD25, CD69) on both CD4+ and CD8+ T cells and an enhanced release of pro-inflammatory cytokines and cytolytic mediators (e.g. IL-2, granzyme B, IFN-γ, perforin, TNF-α). Manufactured TRUCKs showed a specific cytotoxicity towards GD2-expressing target cells indicated by lactate dehydrogenase (LDH) release, a decrease of target cell numbers, microscopic detection of cytotoxic clusters and detachment of target cells in real-time impedance measurements (xCELLigence). Following antigen-specific CAR activation of TRUCKs, CAR-triggered release IL-18 was induced, and the cytokine was biologically active, as demonstrated in migration assays revealing specific attraction of monocytes and NK cells by supernatants of TRUCKs co-cultured with GD2-expressing target cells. In conclusion, GMP-compliant manufacturing of TRUCKs is feasible and delivers high quality T cell products
    corecore