Skip to main content

Lund University Publications

LUND UNIVERSITY LIBRARIES

Inhibition of cyclooxygenase-2 enhances immunotherapy against experimental brain tumors.

Eberstål, Sofia LU ; Badn, Wiaam LU ; Fritzell, Sara LU ; Esbjörnsson, Magnus ; Darabi, Anna LU ; Visse, Edward LU and Siesjö, Peter LU orcid (2012) In Cancer immunology, immunotherapy 61(8). p.1191-1199
Abstract
Glioblastoma multiforme is the most common and aggressive malignant brain tumor in humans, and the prognosis is very poor despite conventional therapy. Immunotherapy represents a novel treatment approach, but the effect is often weakened by release of immune-suppressive molecules such as prostaglandins. In the current study, we investigated the effect of immunotherapy with irradiated interferon-γ (IFN-γ)-secreting tumor cells and administration of the selective cyclooxygease-2 (COX-2) inhibitor parecoxib as treatment of established rat brain tumors. COX-2 inhibition and immunotherapy significantly enhanced the long-term cure rate (81% survival) compared with immunotherapy alone (19% survival), and there was a significant increase in plasma... (More)
Glioblastoma multiforme is the most common and aggressive malignant brain tumor in humans, and the prognosis is very poor despite conventional therapy. Immunotherapy represents a novel treatment approach, but the effect is often weakened by release of immune-suppressive molecules such as prostaglandins. In the current study, we investigated the effect of immunotherapy with irradiated interferon-γ (IFN-γ)-secreting tumor cells and administration of the selective cyclooxygease-2 (COX-2) inhibitor parecoxib as treatment of established rat brain tumors. COX-2 inhibition and immunotherapy significantly enhanced the long-term cure rate (81% survival) compared with immunotherapy alone (19% survival), and there was a significant increase in plasma IFN-γ levels in animals treated with the combined therapy, suggesting a systemic T helper 1 immune response. COX-2 inhibition alone, however, did neither induce cure nor prolonged survival. The tumor cells were identified as the major source of COX-2 both in vivo and in vitro, and unmodified tumor cells produced prostaglandin E(2) in vitro, while the IFN-γ expressing tumor cells secreted significantly lower levels. In conclusion, we show that immunotherapy of experimental brain tumors is greatly potentiated when combined with COX-2 inhibition. Based on our results, the clinically available drug parecoxib may be added to immunotherapy against human brain tumors. Furthermore, the discovery that IFN-γ plasma levels can be used to determine the ongoing in vivo immune response has translational potential. (Less)
Please use this url to cite or link to this publication:
author
; ; ; ; ; and
organization
publishing date
type
Contribution to journal
publication status
published
subject
in
Cancer immunology, immunotherapy
volume
61
issue
8
pages
1191 - 1199
publisher
Springer
external identifiers
  • wos:000306730700004
  • pmid:22213142
  • scopus:84865744728
  • pmid:22213142
ISSN
1432-0851
DOI
10.1007/s00262-011-1196-y
language
English
LU publication?
yes
id
fd35b8aa-739e-4866-be45-5bcb37c493db (old id 2336771)
alternative location
http://www.ncbi.nlm.nih.gov/pubmed/22213142?dopt=Abstract
date added to LUP
2016-04-01 14:11:08
date last changed
2022-04-06 17:17:04
@article{fd35b8aa-739e-4866-be45-5bcb37c493db,
  abstract     = {{Glioblastoma multiforme is the most common and aggressive malignant brain tumor in humans, and the prognosis is very poor despite conventional therapy. Immunotherapy represents a novel treatment approach, but the effect is often weakened by release of immune-suppressive molecules such as prostaglandins. In the current study, we investigated the effect of immunotherapy with irradiated interferon-γ (IFN-γ)-secreting tumor cells and administration of the selective cyclooxygease-2 (COX-2) inhibitor parecoxib as treatment of established rat brain tumors. COX-2 inhibition and immunotherapy significantly enhanced the long-term cure rate (81% survival) compared with immunotherapy alone (19% survival), and there was a significant increase in plasma IFN-γ levels in animals treated with the combined therapy, suggesting a systemic T helper 1 immune response. COX-2 inhibition alone, however, did neither induce cure nor prolonged survival. The tumor cells were identified as the major source of COX-2 both in vivo and in vitro, and unmodified tumor cells produced prostaglandin E(2) in vitro, while the IFN-γ expressing tumor cells secreted significantly lower levels. In conclusion, we show that immunotherapy of experimental brain tumors is greatly potentiated when combined with COX-2 inhibition. Based on our results, the clinically available drug parecoxib may be added to immunotherapy against human brain tumors. Furthermore, the discovery that IFN-γ plasma levels can be used to determine the ongoing in vivo immune response has translational potential.}},
  author       = {{Eberstål, Sofia and Badn, Wiaam and Fritzell, Sara and Esbjörnsson, Magnus and Darabi, Anna and Visse, Edward and Siesjö, Peter}},
  issn         = {{1432-0851}},
  language     = {{eng}},
  number       = {{8}},
  pages        = {{1191--1199}},
  publisher    = {{Springer}},
  series       = {{Cancer immunology, immunotherapy}},
  title        = {{Inhibition of cyclooxygenase-2 enhances immunotherapy against experimental brain tumors.}},
  url          = {{https://lup.lub.lu.se/search/files/3830772/2370051.pdf}},
  doi          = {{10.1007/s00262-011-1196-y}},
  volume       = {{61}},
  year         = {{2012}},
}