5 research outputs found

    Additional file 1 of Co-delivery of dimeric camptothecin and chlorin e6 via polypeptide-based micelles for chemo-photodynamic synergistic therapy

    No full text
    Additional file 1: Figure S1. Particle size and PDI of PKF-Ce6 blank micelles. Figure S2. Pharmacokinetics profiles of Ce6 after administration in mice (n=3). Figure S3. H&E staining of major organs (heart, liver, spleen, lung and kidneys) collected from one mouse after treatment, scale bar is 20 μm. Table S1. Particle size of PCD micelles with different ratio of Ce6 to DCPT. Table S2. Pharmacokinetic parameters of DCPT, PCD and PPCD after administration in mice (n=3). Table S3. Pharmacokinetic parameters of Ce6, PCD and PPCD after administration in mice (n=3)

    Table_1_Decitabine Enhances Vγ9Vδ2 T Cell-Mediated Cytotoxic Effects on Osteosarcoma Cells via the NKG2DL–NKG2D Axis.docx

    No full text
    <p>γδ T cell-based immunotherapy for osteosarcoma (OS) has shown limited success thus far. DNA-demethylating agents not only induce tumor cell death but also have an immunomodulatory function. In this study, we have assessed the potential benefit of combining decitabine (DAC, a DNA demethylation drug) and γδ T cells for OS immunotherapy. DAC increased the expression of natural killer group 2D (NKG2D) ligands (NKG2DLs), including major histocompatibility complex class I-related chains B (MICB) and UL16-binding protein 1 (ULBP1), on the OS cell surface, making the cells more sensitive to recognition and destruction by cytotoxic γδ T cells. The upregulation of MICB and ULBP1 was due to promoter DNA demethylation. Importantly, the killing of OS cells by γδ T cells was partially reversed by blocking the NKG2D receptor, suggesting that the γδ T cell-mediated cytolysis of DAC-pretreated OS cells was mainly dependent on the NKG2D–NKG2DL axis. The in vivo results were consistent with the in vitro results. In summary, DAC could upregulate MICB and ULBP1 expression in OS cells, and combination treatment involving γδ T cell immunotherapy and DAC could be used to enhance the cytotoxic killing of OS cells by γδ T cells.</p

    Image_1_Decitabine Enhances Vγ9Vδ2 T Cell-Mediated Cytotoxic Effects on Osteosarcoma Cells via the NKG2DL–NKG2D Axis.jpeg

    No full text
    <p>γδ T cell-based immunotherapy for osteosarcoma (OS) has shown limited success thus far. DNA-demethylating agents not only induce tumor cell death but also have an immunomodulatory function. In this study, we have assessed the potential benefit of combining decitabine (DAC, a DNA demethylation drug) and γδ T cells for OS immunotherapy. DAC increased the expression of natural killer group 2D (NKG2D) ligands (NKG2DLs), including major histocompatibility complex class I-related chains B (MICB) and UL16-binding protein 1 (ULBP1), on the OS cell surface, making the cells more sensitive to recognition and destruction by cytotoxic γδ T cells. The upregulation of MICB and ULBP1 was due to promoter DNA demethylation. Importantly, the killing of OS cells by γδ T cells was partially reversed by blocking the NKG2D receptor, suggesting that the γδ T cell-mediated cytolysis of DAC-pretreated OS cells was mainly dependent on the NKG2D–NKG2DL axis. The in vivo results were consistent with the in vitro results. In summary, DAC could upregulate MICB and ULBP1 expression in OS cells, and combination treatment involving γδ T cell immunotherapy and DAC could be used to enhance the cytotoxic killing of OS cells by γδ T cells.</p

    Image_1.tif

    No full text
    <p>The long-term survival of osteosarcoma has remained unchanged in the last several decades. Immunotherapy is proved to be a promising therapeutic strategy against osteosarcoma, especially for those with metastasis. Our previous study explored the sensibilization of zoledronate (ZOL) in γδ T cell-mediated cytotoxicity against osteosarcoma, but we have not yet elucidated the specific mechanism. Besides, high concentration is required to achieve these effects, whereas plasma ZOL concentration declines rapidly in the circulation. Valproic acid (VPA), a histone deacetylase inhibitor commonly used as the antiepileptic drug, has attracted much attention due to its synergistic antitumor efficacy with chemotherapy or immunotherapy. Here, we demonstrated that VPA combined with ZOL revealed the synergistic effect in enhancing antitumor efficacy of γδ T cells against osteosarcoma cells. This enhancement was mainly TCR-mediated and largely dependent on granule exocytose pathway. Of note, our findings indicated that ZOL sensitized osteosarcoma cells to γδ T cells by increasing the accumulation of the mevalonate pathway intermediates, which could be facilitated by VPA. We also found that this combination had similar effects on primary osteosarcoma cells. All the results suggested that VPA combined with ZOL could reduce the dose required to achieve a significant antitumor effect of γδ T cells, promoting it to be a novel therapy against osteosarcoma.</p

    Image_2.tif

    No full text
    <p>The long-term survival of osteosarcoma has remained unchanged in the last several decades. Immunotherapy is proved to be a promising therapeutic strategy against osteosarcoma, especially for those with metastasis. Our previous study explored the sensibilization of zoledronate (ZOL) in γδ T cell-mediated cytotoxicity against osteosarcoma, but we have not yet elucidated the specific mechanism. Besides, high concentration is required to achieve these effects, whereas plasma ZOL concentration declines rapidly in the circulation. Valproic acid (VPA), a histone deacetylase inhibitor commonly used as the antiepileptic drug, has attracted much attention due to its synergistic antitumor efficacy with chemotherapy or immunotherapy. Here, we demonstrated that VPA combined with ZOL revealed the synergistic effect in enhancing antitumor efficacy of γδ T cells against osteosarcoma cells. This enhancement was mainly TCR-mediated and largely dependent on granule exocytose pathway. Of note, our findings indicated that ZOL sensitized osteosarcoma cells to γδ T cells by increasing the accumulation of the mevalonate pathway intermediates, which could be facilitated by VPA. We also found that this combination had similar effects on primary osteosarcoma cells. All the results suggested that VPA combined with ZOL could reduce the dose required to achieve a significant antitumor effect of γδ T cells, promoting it to be a novel therapy against osteosarcoma.</p
    corecore