2012
DOI: 10.1186/1471-2407-12-508
|View full text |Cite
|
Sign up to set email alerts
|

Treatment of medulloblastoma using an oncolytic measles virus encoding the thyroidal sodium iodide symporter shows enhanced efficacy with radioiodine

Abstract: BackgroundMedulloblastoma is the most common malignant brain tumor of childhood. Although the clinical outcome for medulloblastoma patients has improved significantly, children afflicted with the disease frequently suffer from debilitating side effects related to the aggressive nature of currently available therapy. Alternative means for treating medulloblastoma are desperately needed. We have previously shown that oncolytic measles virus (MV) can selectively target and destroy medulloblastoma tumor cells in l… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
1
1
1
1

Citation Types

1
24
0

Year Published

2013
2013
2024
2024

Publication Types

Select...
6
2
1

Relationship

1
8

Authors

Journals

citations
Cited by 36 publications
(27 citation statements)
references
References 46 publications
1
24
0
Order By: Relevance
“…This may be due to the lower CD46 expression or a robust antiviral response to MV in HSCIL-1 cells as compared to the other MPNST lines; however, further study is needed to determine this difference. Similar to other preclinical cancer models (Dingli et al, 2004;Hasegawa et al, 2006;Li et al, 2012;Hutzen et al, 2012;Carlson et al, 2009;Penheiter et al, 2010), we observed that a single intratumoral administration of MV-NIS inhibited tumor xenograft growth in vivo and improved long-term survival in treated mice. Of the mice that survived to the end of the study, all but one had no appreciable tumor, indicating that one treatment of MV-NIS was capable of eradicating the MPNST-derived xenografts.…”
Section: Discussionsupporting
confidence: 81%
“…This may be due to the lower CD46 expression or a robust antiviral response to MV in HSCIL-1 cells as compared to the other MPNST lines; however, further study is needed to determine this difference. Similar to other preclinical cancer models (Dingli et al, 2004;Hasegawa et al, 2006;Li et al, 2012;Hutzen et al, 2012;Carlson et al, 2009;Penheiter et al, 2010), we observed that a single intratumoral administration of MV-NIS inhibited tumor xenograft growth in vivo and improved long-term survival in treated mice. Of the mice that survived to the end of the study, all but one had no appreciable tumor, indicating that one treatment of MV-NIS was capable of eradicating the MPNST-derived xenografts.…”
Section: Discussionsupporting
confidence: 81%
“…Besides tracking MV replication by MV-NIS, expression of NIS increases virotherapeutic efficacy by facilitating the intracellular entry of beta-emitting radioisotopes such as I131 and Re188, specifically inducing radiation damage within the tumour microenvironment [62,67,68]. Radiovirotherapy of in vivo multiple myeloma models with relatively small doses of MV-NIS followed by iodine-131 subsequently resulted in the eradication of infected tumour cells, which are otherwise resistant to MV-mediated lysis [62].…”
Section: Engineering Oncolytic Measles Virusmentioning
confidence: 99%
“…NIS expression in infected cells results in uptake of radioiodide isotopes or Tc99 m and thus facilitates in vitro and in vivo localization of infected cells [4852]. In addition to permitting anatomic localization of the infected tumor cells, NIS can also function as a therapeutic transgene, allowing uptake of therapeutic radioiodine isotopes, with resultant improved tumor regression, as well as local bystander effect [47,5357]. Attempts at maximizing therapy and monitoring with multiple transgenes have produced mixed results.…”
Section: Noninvasive Monitoringmentioning
confidence: 99%