Show simple item record

dc.contributor.authorKuczynski, EA
dc.contributor.authorYin, M
dc.contributor.authorBar-Zion, A
dc.contributor.authorLee, CR
dc.contributor.authorButz, H
dc.contributor.authorMan, S
dc.contributor.authorDaley, F
dc.contributor.authorVermeulen, PB
dc.contributor.authorYousef, GM
dc.contributor.authorFoster, FS
dc.contributor.authorReynolds, AR
dc.contributor.authorKerbel, RS
dc.date.accessioned2016-10-19T16:33:22Z
dc.date.issued2016-04-08
dc.identifier.citationJournal of the National Cancer Institute, 2016, 108 (8)
dc.identifier.issn0027-8874
dc.identifier.urihttps://repository.icr.ac.uk/handle/internal/180
dc.identifier.eissn1460-2105
dc.identifier.doi10.1093/jnci/djw030
dc.description.abstractBACKGROUND: The anti-angiogenic Sorafenib is the only approved systemic therapy for advanced hepatocellular carcinoma (HCC). However, acquired resistance limits its efficacy. An emerging theory to explain intrinsic resistance to other anti-angiogenic drugs is 'vessel co-option,' ie, the ability of tumors to hijack the existing vasculature in organs such as the lungs or liver, thus limiting the need for sprouting angiogenesis. Vessel co-option has not been evaluated as a potential mechanism for acquired resistance to anti-angiogenic agents. METHODS: To study sorafenib resistance mechanisms, we used an orthotopic human HCC model (n = 4-11 per group), where tumor cells are tagged with a secreted protein biomarker to monitor disease burden and response to therapy. Histopathology, vessel perfusion assessed by contrast-enhanced ultrasound, and miRNA sequencing and quantitative real-time polymerase chain reaction were used to monitor changes in tumor biology. RESULTS: While sorafenib initially inhibited angiogenesis and stabilized tumor growth, no angiogenic 'rebound' effect was observed during development of resistance unless therapy was stopped. Instead, resistant tumors became more locally infiltrative, which facilitated extensive incorporation of liver parenchyma and the co-option of liver-associated vessels. Up to 75% (±10.9%) of total vessels were provided by vessel co-option in resistant tumors relative to 23.3% (±10.3%) in untreated controls. miRNA sequencing implicated pro-invasive signaling and epithelial-to-mesenchymal-like transition during resistance development while functional imaging further supported a shift from angiogenesis to vessel co-option. CONCLUSIONS: This is the first documentation of vessel co-option as a mechanism of acquired resistance to anti-angiogenic therapy and could have important implications including the potential therapeutic benefits of targeting vessel co-option in conjunction with vascular endothelial growth factor receptor signaling.
dc.formatPrint-Electronic
dc.languageeng
dc.language.isoeng
dc.publisherOXFORD UNIV PRESS INC
dc.rights.urihttps://creativecommons.org/licenses/by-nc/4.0
dc.subjectLiver
dc.subjectBlood Vessels
dc.subjectAnimals
dc.subjectHumans
dc.subjectMice
dc.subjectMice, SCID
dc.subjectCarcinoma, Hepatocellular
dc.subjectLiver Neoplasms
dc.subjectNeoplasm Invasiveness
dc.subjectDisease Models, Animal
dc.subjectNeovascularization, Pathologic
dc.subjectPhenylurea Compounds
dc.subjectNiacinamide
dc.subjectVimentin
dc.subjectActins
dc.subjectVascular Endothelial Growth Factor A
dc.subjectHomeodomain Proteins
dc.subjectRepressor Proteins
dc.subjectMicroRNAs
dc.subjectAntineoplastic Agents
dc.subjectAntigens, CD34
dc.subjectContrast Media
dc.subjectUltrasonography
dc.subjectSequence Analysis, RNA
dc.subjectNeoplasm Transplantation
dc.subjectSignal Transduction
dc.subjectUp-Regulation
dc.subjectDrug Resistance, Neoplasm
dc.subjectMale
dc.subjectOsteopontin
dc.subjectEpithelial-Mesenchymal Transition
dc.subjectZinc Finger E-box-Binding Homeobox 1
dc.subjectZinc Finger E-box Binding Homeobox 2
dc.subjectSorafenib
dc.titleCo-option of Liver Vessels and Not Sprouting Angiogenesis Drives Acquired Sorafenib Resistance in Hepatocellular Carcinoma.
dc.typeJournal Article
dcterms.dateAccepted2016-02-08
rioxxterms.versionofrecord10.1093/jnci/djw030
rioxxterms.licenseref.urihttps://creativecommons.org/licenses/by-nc/4.0
rioxxterms.licenseref.startdate2016-08
rioxxterms.typeJournal Article/Review
dc.relation.isPartOfJournal of the National Cancer Institute
pubs.issue8
pubs.notesNot known
pubs.organisational-group/ICR
pubs.organisational-group/ICR/Primary Group
pubs.organisational-group/ICR/Primary Group/ICR Divisions
pubs.organisational-group/ICR/Primary Group/ICR Divisions/Closed research teams
pubs.organisational-group/ICR/Primary Group/ICR Divisions/Closed research teams/Tumour Biology
pubs.organisational-group/ICR
pubs.organisational-group/ICR/Primary Group
pubs.organisational-group/ICR/Primary Group/ICR Divisions
pubs.organisational-group/ICR/Primary Group/ICR Divisions/Closed research teams
pubs.organisational-group/ICR/Primary Group/ICR Divisions/Closed research teams/Tumour Biology
pubs.publication-statusPublished
pubs.volume108
pubs.embargo.termsNot known
icr.researchteamTumour Biology
dc.contributor.icrauthorDaley, Frances
dc.contributor.icrauthorReynolds, Andrew


Files in this item

Thumbnail

This item appears in the following collection(s)

Show simple item record

https://creativecommons.org/licenses/by-nc/4.0
Except where otherwise noted, this item's license is described as https://creativecommons.org/licenses/by-nc/4.0