Vis enkel innførsel

dc.contributor.authorSarowar, Shahin
dc.contributor.authorCirillo, Davide
dc.contributor.authorJátiva, Pablo
dc.contributor.authorNilsen, Mette Hartmark
dc.contributor.authorOtragane, Sarah-Muheha Anni
dc.contributor.authorHeggdal, Jan Ingemann
dc.contributor.authorSelheim, Frode
dc.contributor.authorCeña, Valentín
dc.contributor.authorBjørsvik, Hans-René
dc.contributor.authorEnger, Per Øyvind
dc.date.accessioned2022-06-08T08:30:13Z
dc.date.available2022-06-08T08:30:13Z
dc.date.created2022-05-19T15:50:09Z
dc.date.issued2022
dc.identifier.issn2234-943X
dc.identifier.urihttps://hdl.handle.net/11250/2997827
dc.description.abstractMetastatic tumors with moderate radiosensitivity account for most cancer-related deaths, highlighting the limitations of current radiotherapy regimens. The xCT-inhibitor sulfasalazine (SAS) sensitizes cancer cells to radiotherapy by blocking cystine uptake via the xCT membrane antiporter, and thereby glutathione (GSH) synthesis protecting against radiation-induced oxidative stress. The expression of xCT in multiple tumor types implies it as a target generic to cancer rather than confined to few subtypes. However, SAS has limited clinical potential as a radiosensitizer due to side effects and low bioavailability. Using SAS as a starting point, we previously developed synthetic xCT-inhibitors through scaffold hopping and structure optimization aided by structure-activity relationship analysis (SAR). Notably, the compound DC10 exhibited inhibition of GSH synthesis. In this study, we validated DC10 as a radiosensitizer in the xCT-expressing cancer cell lines A172, A375 and MCF7, and mice harboring melanoma xenografts. After DC10 treatment, we measured 14C-cystine uptake in the cancer cells using liquid scintillation counting, and intracellular GSH levels and reactive oxygen species (ROS) using luminescence assays. We performed immunoblotting of H2AX and ATM to assess DNA damage after treatment with DC10 and radiotherapy. We then assessed the effect of adding DC10 to radiation upon cancer cell colony formation. Blood samples from mice treated with DC10 underwent biochemical analysis to assess toxicity. Finally, mice with A375 melanomas in the flank, received DC10 and radiotherapy in combination, as monotherapies or no treatment. Notably, DC10 reduced cystine uptake and GSH synthesis and increased ROS levels in a dose-dependent manner. Furthermore, DC10 interacted synergistically with radiation to increase DNA damage and reduce tumor cell colony formation. Mice receiving DC10 were clinically unaffected, whereas blood samples analysis to assess bone marrow suppression, liver or kidney toxicity revealed no significant differences between treated mice and untreated controls. Importantly, DC10 potentiated the anti-tumor efficacy of radiation in mice with melanoma xenografts. We conclude that DC10 is well tolerated and acts as a radiosensitizer by inhibiting cystine uptake, leading to GSH depletion and increased oxidative stress. Our findings demonstrate the feasibility of using synthetic xCT-inhibitors to overcome radioresistance.en_US
dc.language.isoengen_US
dc.publisherFrontiersen_US
dc.rightsNavngivelse 4.0 Internasjonal*
dc.rights.urihttp://creativecommons.org/licenses/by/4.0/deed.no*
dc.titleThe styryl benzoic acid derivative DC10 potentiates radiotherapy by targeting the xCT-glutathione axisen_US
dc.typeJournal articleen_US
dc.typePeer revieweden_US
dc.description.versionpublishedVersionen_US
dc.rights.holderCopyright 2022 The Author(s)en_US
dc.source.articlenumber786739en_US
cristin.ispublishedtrue
cristin.fulltextoriginal
cristin.qualitycode1
dc.identifier.doi10.3389/fonc.2022.786739
dc.identifier.cristin2025749
dc.source.journalFrontiers in Oncologyen_US
dc.identifier.citationFrontiers in Oncology. 2022, 12, 786739.en_US
dc.source.volume12en_US


Tilhørende fil(er)

Thumbnail

Denne innførselen finnes i følgende samling(er)

Vis enkel innførsel

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