Egyszerű nézet

dc.contributor.author Torok S
dc.contributor.author Rezeli M
dc.contributor.author Kelemen O
dc.contributor.author Vegvari A
dc.contributor.author Watanabe K
dc.contributor.author Sugihara Y
dc.contributor.author Tisza A
dc.contributor.author Marton T
dc.contributor.author Kovacs I
dc.contributor.author Tovari J
dc.contributor.author Laszlo V
dc.contributor.author Helbich TH
dc.contributor.author Hegedűs, Balázs
dc.contributor.author Klikovits T
dc.contributor.author Hoda MA
dc.contributor.author Klepetko W
dc.contributor.author Paku S
dc.contributor.author Marko-Varga G
dc.contributor.author Dome B
dc.date.accessioned 2017-01-05T14:30:25Z
dc.date.available 2017-01-05T14:30:25Z
dc.date.issued 2017
dc.identifier.citation pagination=400-412; journalVolume=7; journalIssueNumber=2; journalTitle=Theranostics;
dc.identifier.uri http://repo.lib.semmelweis.hu//handle/123456789/3998
dc.identifier.uri doi:10.7150/thno.16767
dc.description.abstract Resistance mechanisms against antiangiogenic drugs are unclear. Here, we correlated the antitumor and antivascular properties of five different antiangiogenic receptor tyrosine kinase inhibitors (RTKIs) (motesanib, pazopanib, sorafenib, sunitinib, vatalanib) with their intratumoral distribution data obtained by matrix-assisted laser desorption ionization mass spectrometry imaging (MALDI-MSI). In the first mouse model, only sunitinib exhibited broad-spectrum antivascular and antitumor activities by simultaneously suppressing vascular endothelial growth factor receptor-2 (VEGFR2) and desmin expression, and by increasing intratumoral hypoxia and inhibiting both tumor growth and vascularisation significantly. Importantly, the highest and most homogeneous intratumoral drug concentrations have been found in sunitinib-treated animals. In another animal model, where - in contrast to the first model - vatalanib was detectable at homogeneously high intratumoral concentrations, the drug significantly reduced tumor growth and angiogenesis. In conclusion, the tumor tissue penetration and thus the antiangiogenic and antitumor potential of antiangiogenic RTKIs vary among the tumor models and our study demonstrates the potential of MALDI-MSI to predict the efficacy of unlabelled small molecule antiangiogenic drugs in malignant tissue. Our approach is thus a major technical and preclinical advance demonstrating that primary resistance to angiogenesis inhibitors involves limited tumor tissue drug penetration. We also conclude that MALDI-MSI may significantly contribute to the improvement of antivascular cancer therapies.
dc.relation.ispartof urn:issn:1838-7640
dc.title Limited Tumor Tissue Drug Penetration Contributes to Primary Resistance against Angiogenesis Inhibitors
dc.type Journal Article
dc.date.updated 2017-01-05T13:29:11Z
dc.language.rfc3066 en
dc.identifier.mtmt 3161443
dc.identifier.pubmed 28042343
dc.contributor.department SE/AOK/I/IISZPI/MTA-SE Molekuláris Onkológia Kutatócsoport
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