21 research outputs found

    Tumor cell survival pathways activated by photodynamic therapy: a molecular basis for pharmacological inhibition strategies

    Get PDF

    Tranexamic Acid-Encapsulating Thermosensitive Liposomes for Site-Specific Pharmaco-Laser Therapy of Port Wine Stains

    No full text
    Site-specific pharmaco-laser therapy (SSPLT) is a developmental stage treatment modality designed to non-invasively remove superficial vascular pathologies such as port wine stains (PWS) by combining conventional laser therapy with the prior administration of a prothrombotic and/or antifibrinolytic pharmaceutical-containing drug delivery system. For the antifibrinolytic SSPLT component, six different PEGylated thermosensitive liposomal formulations encapsulating tranexamic acid (TA), a potent antifibrinolytic lysine analogue, were characterized for drug:lipid ratio, encapsulation efficiency, size, endovesicular TA concentration (CTA), phase transition temperature (Tm), and assayed for heat-induced TA release. Assays were developed for the quantification of liposomal TA and heat-induced TA release from two candidate formulations. The outcome parameters were then combined with a 3D histological reconstruction of a port wine stain biopsy to extrapolate in vivo posologies for SSPLT. The prime formulation, DPPC:DSPE-PEG2000 (96:4 molar ratio), had a drug:lipid molar ratio of 0.82, an encapsulation efficiency of 1.29%, a diameter of 155 nm, and a CTA of 214 mM. The peak TA release from this formulation (Tm = 42.3 °C) comprised 96% within 2.5 min, whereas this was 94% in 2 min for DPPC:MPPC:DSPE-PEG2000 (86:10:4) liposomes (Tm = 41.5 °C). Computational analysis revealed that <400 DPPC:DSPE-PEG2000 (96:4 molar ratio) liposomes are needed to treat a PWS of 40 cm(2), compared to a three-fold greater quantity of DPPC:MPPC:DSPE-PEG2000 (86:10:4) liposomes, indicating that, in light of the assayed parameters and endovascular laser-tissue interactions, the former formulation is most suitable for antifibrinolytic SSPLT. This was further confirmed with experiments involving ex vivo and in vivo liposome-platelet and liposome-red blood cell association as well as uptake and toxicity assays with cultured endothelial cells (HUVECs), macrophages (RAW 264.7), and hepatocytes (HepG2

    The impact of P-glycoprotein and breast cancer resistance protein on the brain pharmacokinetics and pharmacodynamics of a panel of MEK inhibitors

    No full text
    Mitogen/extracellular signal-regulated kinase (MEK) inhibitors have been tested in clinical trials for treatment of intracranial neoplasms, including glioblastoma (GBM), but efficacy of these drugs has not yet been demonstrated. The blood-brain barrier (BBB) is a major impediment to adequate delivery of drugs into the brain and may thereby also limit the successful implementation of MEK inhibitors against intracranial malignancies. The BBB is equipped with a range of ATP-dependent efflux transport proteins, of which P-gp (ABCB1) and BCRP (ABCG2) are the two most dominant for drug efflux from the brain. We investigated their impact on the pharmacokinetics and target engagement of a panel of clinically applied MEK inhibitors, in order to select the most promising candidate for brain cancers in the context of clinical pharmacokinetics and inhibitor characteristics. To this end, we used in vitro drug transport assays and conducted pharmacokinetic and pharmacodynamic studies in wildtype and ABC-transporter knockout mice. PD0325901 displayed more promising characteristics than trametinib (GSK1120212), binimetinib (MEK162), selumetinib (AZD6244), and pimasertib (AS703026): PD0325901 was the weakest substrate of P-gp and BCRP in vitro, its brain penetration was only marginally higher in Abcb1a/b;Abcg2-/-mice, and efficient target inhibition in the brain could be achieved at clinically relevant plasma levels. Notably, target inhibition could also be demonstrated for selumetinib, but only at plasma levels far above levels in patients receiving the maximum tolerated dose. In summary, our study recommends further development of PD0325901 for the treatment of intracranial neoplasms

    Inhibition of hypoxia-inducible factor 1 with acriflavine sensitizes hypoxic tumor cells to photodynamic therapy with zinc phthalocyanine-encapsulating cationic liposomes

    No full text
    Photodynamic therapy (PDT) is a tumor treatment modality in which a tumorlocalized photosensitizer is excited with light, which results in local production of reactive oxygen species, destruction of tumor vasculature, tumor hypoxia, tumor cell death, and induction of an anti-tumor immune response. However, pre-existing tumor hypoxia may desensitize tumors to PDT by activating the hypoxia-inducible factor 1 (HIF-1) survival pathway. Therefore, we hypothesized that inhibition of HIF-1 with acriflavine (ACF) would exacerbate cell death in human epidermoid carcinoma (A431) cells. PDT of A431 tumor cells was performed using newly developed and optimized PEGylated cationic liposomes containing the photosensitizer zinc phthalocyanine (ZnPC). Molecular docking revealed that ACF binds to the dimerization domain of HIF-1 alpha, and confocal microscopy confirmed translocation of ACF from the cytosol to the nucleus under hypoxia. HIF-1 was stabilized in hypoxic, but not normoxic, A431 cells following PDT. Inhibition of HIF-1 with ACF increased the extent of PDT-induced cell death under hypoxic conditions and reduced the expression of the HIF-1 target genes VEGF, PTGS2, and EDN1. Moreover, co-encapsulation of ACF in the aqueous core of ZnPC-containing liposomes yielded an adjuvant effect on PDT efficacy that was comparable to non-encapsulated ACF. In conclusion, HIF-1 contributes to A431 tumor cell survival following PDT with liposomal ZnPC. Inhibition of HIF-1 with free or liposomal ACF improves PDT efficac

    Additional file 3: Table S1. of Low-power photodynamic therapy induces survival signaling in perihilar cholangiocarcinoma cells

    No full text
    Overview of transcriptional targets of the five survival pathways. Official gene names are listed that were derived from the Hugo Gene Nomenclature Committee (HGNC) along with their Genbank accession numbers (Gene ID). (XLSX 16 kb

    Additional file 1: Figure S1. of Low-power photodynamic therapy induces survival signaling in perihilar cholangiocarcinoma cells

    No full text
    In vivo toxicity evaluation of ZnPC-ITLs. (A) Chicken embryos were intravenously injected on embryonic development day 12 with different concentrations of ZnPC-ITLs (n = 7 per group). The concentrations indicate the final lipid concentration in blood. Control embryos were intravenously injected with an equal volume of 0.75 % NaCl. (B) C57BL/6 mice were intravenously injected with ZnPC-ITLs (2.5 mM, final lipid concentration in blood, red line) or physiological buffer (black line). Mice were weighed every four days until day 28 post-injection. Data is presented as mean ± SD with n = 8 per group. (C) Biochemical and hematological parameters assessed in C57BL/6 mice 28 days after systemic administration of ZnPC-ITLs. Data is presented as mean ± SD with n = 8 per group. Statistical analysis was performed as described in section "Statistical analysis". Abbreviations: ALT, alanine transaminase; AST, aspartate transaminase; CK-MB, creatine kinase M and B; CPK, creatine phosphokinase; LDH, lactate dehydrogenase. (D) Histology of liver, spleen, and lung of C57BL/6 mice 28 days after systemic administration of ZnPC-ITLs or physiological buffer (control). Hematoxylin and eosin staining, magnification 20 × . (DOC 5788 kb
    corecore