10 research outputs found

    Low-dose mistletoe lectin-I reduces melanoma growth and spread in a scid mouse xenograft model

    Get PDF
    This study investigates the effects of mistletoe lectin-I (ML-I) on melanoma growth and spread in vivo. The human melanoma cell line MV3 was xenografted into severe combined immunodeficient mice and vehicle solution or purified ML-I was administered at 30, 150 and 500 ng per kg body weight (20 mice per group) daily. After 19 days, mice were killed, primary tumours (PTs) and lungs were dissected out, and tumour weights, number of lung metastases (LMs), number of tumour-infiltrating dendritic cells (DCs), and apoptosis rates in the melanoma cells and in the DCs were assessed. A 35% reduction of PT weight (P=0.03) and a 55% decrease in number of LMs (P=0.016) were evident for low-dose ML-I (30 ng kg−1) treatment but not for higher doses. Mistletoe lectin-I increased apoptosis rates in the melanoma cells of PTs at all doses, while no induction of apoptosis was noted in the LMs. Low-dose ML-I significantly increased the number of DCs infiltrating the PTs (P<0.0001) and protected DCs against apoptosis, while higher doses induced apoptosis in the DCs (P<0.01). Our results demonstrate that low-dose ML-I reduced melanoma growth and number of metastases in vivo, primarily due to immunomodulatory effects

    Differential responses of cord and adult blood-derived dendritic cells to dying cells

    No full text
    Normal turnover of body tissues yields apoptotic cells while infections cause tissue injuries and cell necrosis. The interaction of these dying cells with dendritic cells (DCs) may provide immunological instructions leading to either immune tolerance or activation. We hypothesize that neonatal and adult DCs differ in their responses to dying cells, thereby contributing to the observed differences in immune responses between neonates and adults. We compare the outcome of interaction of cord and adult blood-derived DCs with dying Epstein–Barr-virus-transformed lymphoblastoid cells (LCLs) and the responsiveness to lipopolysaccharide. While cord DCs were able to phagocytose both apoptotic and necrotic LCLs, the subsequent responses differed significantly from those of adult DCs. Interaction of adult DCs with necrotic but not early apoptotic LCLs resulted in high expression of DC costimulatory molecules (CD80/CD86) and activation markers (CD83), production of both proinflammatory and anti-inflammatory cytokines (tumour necrosis factor-α, interleukin-10), and strong T-cell-stimulating activities. In contrast, in response to either necrotic or apoptotic LCLs, cord DCs had minimal up-regulation of those DC functional markers, little cytokine production and poor stimulation on T-cell proliferation. In response to lipopolysaccharide, however, both adult and cord DCs produced comparable levels of tumour necrosis factor-α and interleukin-10, but only adult DCs produced interleukin-12(p70). Taken together, these results suggest that neonatal DCs generally favour immune tolerance with minimal activation and cytokine production, except in extremely dangerous situations, such as bacterial sepsis, when neonatal DCs may produce certain types of cytokines and stimulate T-cell proliferation

    Apoptotic cell death in conjunction with CD80 costimulation confers uveal melanoma cells with the ability to induce immune responses

    No full text
    Uveal melanoma is a rare malignancy with a poor prognosis despite current therapeutic intervention. The current investigation focuses on the immunogenicity of uveal melanoma cells genetically modified with recombinant adenovirus encoding CD80 (AdCD80) in contrast to their parental counterpart. We demonstrate that costimulation provided by uveal melanoma cells improved immune responses in vitro as determined by mixed lymphocyte tumour cell cultures and cytotoxic T-cell assays using lymphocytes from healthy donors and uveal melanoma patients. Flow cytometry revealed T-cell stimulation by activated CD4(+) and CD8(+) T cells. Additionally, autologous lymphocytes proliferated in response to CD80-expressing primary uveal melanomas, indicating that this patient group is suitable for immunotherapy. Moreover, this study utilized AdCD80 modified and parental apoptotic tumour cells, loaded onto immature dendritic cells, as a source of tumour antigen. The ability of live or apoptotic tumour cells to stimulate lymphocyte proliferation and activation was determined. Apoptotic uveal melanoma cells expressing CD80 were efficient at inducing an immune response and served as a potent immunogen. The use of apoptotic uveal melanoma cells in combination with expression of costimulatory molecules could prove a novel adjuvant therapy for the treatment of this disease
    corecore