Mostrar el registro sencillo del ítem

dc.contributor.authorRackov, Gorjana
dc.contributor.authorIegiani, Giorgia
dc.contributor.authorUribe, Daniel
dc.contributor.authorQuezada, Claudia
dc.contributor.authorQuezada, Claudia
dc.contributor.authorBelda-Iniesta, Cristóbal
dc.contributor.authorEscobedo-Lucea, Carmen
dc.contributor.authorSilva, Augusto
dc.contributor.authorPuig, Pere
dc.contributor.authorGonzález-Rumayor, Víctor
dc.contributor.authorAyuso Sacido, Ángel 
dc.date.accessioned2021-10-14T13:38:05Z
dc.date.available2021-10-14T13:38:05Z
dc.date.issued2020
dc.identifier.issn2234-943Xspa
dc.identifier.urihttp://hdl.handle.net/10641/2493
dc.description.abstractGlioblastoma (GBM) is the most devastating and least treatable brain tumor with median survival <15 months and extremely high recurrence rates. Promising results of immune checkpoint blockade obtained from pre-clinical studies in mice did not translate to clinic, and new strategies are urgently needed, particularly those targeting GBM stem cells (GSCs) that are held responsible for drug resistance and tumor recurrence. Patient-derived GSC cultures are critical for finding effective brain tumor therapies. Here, we investigated the ability of the recently described monoclonal antibody Nilo1 to specifically recognize GSCs isolated from GBM surgical samples. We employed five patient-derived GSC cultures with different stemness marker expression and differentiation potential, able to recapitulate original tumors when xenotransplanted in vivo. To answer whether Nilo1 has any functional effects in patient-derived GSCs lines, we treated the cells with Nilo1 in vitro and analyzed cell proliferation, cell cycle, apoptosis, sphere formation, as well as the expression of stem vs. differentiation markers. All tested GSCs stained positively for Nilo1, and the ability of Nilo1 to recognize GSCs strongly relied on their stem-like phenotype. Our results showed that a subset of patient-derived GSCs were sensitive to Nilo1 treatment. In three GSC lines Nilo1 triggered differentiation accompanied by the induction of p21. Most strikingly, in one GSC line Nilo1 completely abrogated self-renewal and led to Bax-associated apoptosis. Our data suggest that Nilo1 targets a molecule functionally relevant for stemness maintenance and pinpoint Nilo1 as a novel antibody-based therapeutical strategy to be used either alone or in combination with cytotoxic drugs for GSC targeting. Further pre-clinical studies are needed to validate the effectiveness of GSC-specific Nilo1 targeting in vivo.spa
dc.language.isoengspa
dc.publisherFrontiers in Oncologyspa
dc.rightsAtribución-NoComercial-SinDerivadas 3.0 España*
dc.rights.urihttp://creativecommons.org/licenses/by-nc-nd/3.0/es/*
dc.subjectNilo1spa
dc.subjectAntibodyspa
dc.subjectGlioblastomaspa
dc.subjectGlioma stem cellsspa
dc.subjectNeural stem cellsspa
dc.subjectImmunotherapyspa
dc.titlePotential Therapeutic Effects of the Neural Stem Cell-Targeting Antibody Nilo1 in Patient-Derived Glioblastoma Stem Cells.spa
dc.typejournal articlespa
dc.type.hasVersionAMspa
dc.rights.accessRightsopen accessspa
dc.description.extent3,70 MBspa
dc.identifier.doi10.3389/fonc.2020.01665spa
dc.relation.publisherversionhttps://www.frontiersin.org/articles/10.3389/fonc.2020.01665/fullspa


Ficheros en el ítem

FicherosTamañoFormatoVer
fonc-10-01665.pdf3.706MbPDFVer/

Este ítem aparece en la(s) siguiente(s) colección(ones)

Mostrar el registro sencillo del ítem

Atribución-NoComercial-SinDerivadas 3.0 España
Excepto si se señala otra cosa, la licencia del ítem se describe como Atribución-NoComercial-SinDerivadas 3.0 España