Vis enkel innførsel

dc.contributor.authorZhang, Xin
dc.contributor.authorDing, Kaikai
dc.contributor.authorJi, Jianxiong
dc.contributor.authorParajuli, Himalaya
dc.contributor.authorAasen, Synnøve Nymark
dc.contributor.authorEspedal, Heidi
dc.contributor.authorHuang, Bin
dc.contributor.authorChen, Anjing
dc.contributor.authorWang, Jian
dc.contributor.authorLi, Xingang
dc.contributor.authorThorsen, Frits
dc.date.accessioned2021-06-30T12:18:39Z
dc.date.available2021-06-30T12:18:39Z
dc.date.created2021-01-05T14:37:22Z
dc.date.issued2020
dc.identifier.issn2156-6976
dc.identifier.urihttps://hdl.handle.net/11250/2762572
dc.description.abstractBrain metastasis is a major cause of mortality in melanoma patients. The blood-brain barrier (BBB) prevents most anti-tumor compounds from entering the brain, which significantly limits their use in the treatment of brain metastasis. One strategy in the development of new treatments is to assess the anti-tumor potential of drugs currently used in the clinic. Here, we tested the anti-tumor effect of the BBB-penetrating antipsychotic trifluoperazine (TFP) on metastatic melanoma. H1 and Melmet1 human metastatic melanoma cell lines were used in vitro and in vivo. TFP effects on viability and toxicity were evaluated in proliferation and colony formation assays. Preclinical, therapeutic efficacy was evaluated in NOD/SCID mice, after intracardial injection of tumor cells. Molecular studies using immunohistochemistry, western blots, immunofluorescence and transmission electron microscopy were used to gain mechanistic insight into the biological activity of TFP. Our results showed that TFP decreased cell viability and proliferation, colony formation and spheroid growth in vitro. The drug also decreased tumor burden in mouse brains and prolonged animal survival after injection of tumor cells (53.0 days vs 44.5 days), TFP treated vs untreated animals, respectively (P < 0.01). At the molecular level, TFP treatment led to increased levels of LC3B and p62 in vitro and in vivo, suggesting an inhibition of autophagic flux. A decrease in LysoTracker Red uptake after treatment indicated impaired acidification of lysosomes. TFP caused accumulation of electron dense vesicles, an indication of damaged lysosomes, and reduced the expression of cathepsin B, a main lysosomal protease. Acridine orange and galectin-3 immunofluorescence staining were evidence of TFP induction of lysosomal membrane permeabilization. Finally, TFP was cytotoxic to melanoma brain metastases based on the increased release of lactate dehydrogenase into media. Through knockdown experiments, the processes of TFP-induced lysosomal membrane permeabilization and cell death appeared to be STAT3 dependent. In conclusion, our work provides a strong rationale for further clinical investigation of TFP as an adjuvant therapy for melanoma patients with metastases to the brain.en_US
dc.language.isoengen_US
dc.publishere-Century Publishingen_US
dc.rightsNavngivelse-Ikkekommersiell 4.0 Internasjonal*
dc.rights.urihttp://creativecommons.org/licenses/by-nc/4.0/deed.no*
dc.titleTrifluoperazine prolongs the survival of experimental brain metastases by STAT3-dependent lysosome membrane permeabilizationen_US
dc.typeJournal articleen_US
dc.typePeer revieweden_US
dc.description.versionpublishedVersionen_US
dc.rights.holderAJCR Copyright 2020en_US
cristin.ispublishedtrue
cristin.fulltextoriginal
cristin.qualitycode1
dc.identifier.cristin1865689
dc.source.journalAmerican Journal of Cancer Researchen_US
dc.source.pagenumber545-563en_US
dc.identifier.citationAmerican Journal of Cancer Research. 2020, 10 (2), 545-563.en_US
dc.source.volume10en_US
dc.source.issue2en_US


Tilhørende fil(er)

Thumbnail

Denne innførselen finnes i følgende samling(er)

Vis enkel innførsel

Navngivelse-Ikkekommersiell 4.0 Internasjonal
Med mindre annet er angitt, så er denne innførselen lisensiert som Navngivelse-Ikkekommersiell 4.0 Internasjonal