Egyszerű nézet

dc.contributor.author Merino VF
dc.contributor.author Cho S
dc.contributor.author Nguyen N
dc.contributor.author Sadik H
dc.contributor.author Narayan A
dc.contributor.author Talbot C Jr
dc.contributor.author Cope L
dc.contributor.author Zhou XC
dc.contributor.author Zhang Z
dc.contributor.author Győrffy, Balázs
dc.contributor.author Sukumar S
dc.date.accessioned 2019-04-15T07:03:21Z
dc.date.available 2019-04-15T07:03:21Z
dc.date.issued 2018
dc.identifier 85057559110
dc.identifier.citation journalVolume=20;journalIssueNumber=1;journalTitle=BREAST CANCER RESEARCH;pagination=145, pages: 14;journalAbbreviatedTitle=BREAST CANCER RES;
dc.identifier.uri http://repo.lib.semmelweis.hu//handle/123456789/6828
dc.identifier.uri doi:10.1186/s13058-018-1068-x
dc.description.abstract Background: A combination of entinostat, all-trans retinoic acid, and doxorubicin (EAD) induces cell death and differentiation and causes significant regression of xenografts of triple-negative breast cancer (TNBC). Methods: We investigated the mechanisms underlying the antitumor effects of each component of the EAD combination therapy by high-throughput gene expression profiling of drug-treated cells. Results: Microarray analysis showed that entinostat and doxorubicin (ED) altered expression of genes related to growth arrest, inflammation, and differentiation. ED downregulated MYC, E2F, and G2M cell cycle genes. Accordingly, entinostat sensitized the cells to doxorubicin-induced growth arrest at G2. ED induced interferon genes, which correlated with breast tumors containing a higher proportion of tumor-infiltrating lymphocytes. ED also increased the expression of immune checkpoint agonists and cancer testis antigens. Analysis of TNBC xenografts showed that EAD enhanced the inflammation score in nude mice. Among the genes differentially regulated between the EAD and ED groups, an all-trans retinoic acid (ATRA)-regulated gene, DHRS3, was induced in EAD-treated xenografts. DHRS3 was expressed at lower levels in human TNBC metastases compared to normal breast or primary tumors. High expression of ED-induced growth arrest and inflammatory genes was associated with better prognosis in TNBC patients. Conclusions: Entinostat potentiated doxorubicin-mediated cell death and the combination induced inflammatory signatures. The ED-induced immunomodulation may improve immunotherapy. Addition of ATRA to ED may potentiate inflammation and contribute to TNBC regression. © 2018 The Author(s).
dc.relation.ispartof urn:issn:1465-542X
dc.title Induction of cell cycle arrest and inflammatory genes by combined treatment with epigenetic, differentiating, and chemotherapeutic agents in triple-negative breast cancer
dc.type Journal Article
dc.date.updated 2019-03-05T11:16:07Z
dc.language.rfc3066 en
dc.rights.holder NULL
dc.identifier.mtmt 30347700
dc.identifier.wos 000451558200002
dc.identifier.pubmed 30486871
dc.contributor.department SE/AOK/K/II. Sz. Gyermekgyógyászati Klinika
dc.contributor.institution Semmelweis Egyetem


Kapcsolódó fájlok:

A fájl jelenleg csak egyetemi IP címről érhető el.

Megtekintés/Megnyitás

Ez a rekord az alábbi gyűjteményekben szerepel:

Egyszerű nézet