Effective induction of therapeutic antitumor immunity by dendritic cells coexpressing interleukin-18 and tumor antigen

2003 ◽  
Vol 81 (9) ◽  
pp. 585-596 ◽  
Author(s):  
Dajing Xia ◽  
Shu Zheng ◽  
Weiping Zhang ◽  
Long He ◽  
Qingqing Wang ◽  
...  
2002 ◽  
Vol 109 (11) ◽  
pp. 1463-1470 ◽  
Author(s):  
Helen Y. Wang ◽  
Tihui Fu ◽  
Gang Wang ◽  
Gang Zeng ◽  
Donna M. Perry-Lalley ◽  
...  

2003 ◽  
Vol 99 (4) ◽  
pp. 746-753 ◽  
Author(s):  
Ryuya Yamanaka ◽  
Naoto Tsuchiya ◽  
Naoki Yajima ◽  
Junpei Honma ◽  
Hitoshi Hasegawa ◽  
...  

Object. The aim of this study was to investigate further immunogene treatment of malignant brain tumor to improve its therapeutic efficacy. Methods. Intratumoral dendritic cells pulsed with Semliki Forest virus (SFV)—interleukin-18 (IL-18) and/or systemic IL-12 were injected into mice bearing the B16 brain tumor. To study the immune mechanisms involved in tumor regression, we monitored the growth of implanted B16 brain tumor cells in T cell—depleted mice and IFNγ-neutralized mice. To analyze the protective immunity created by tumor inoculation, B16 cells were injected into the left thighs of mice that had received an inoculation, and tumor growth was monitored. The local delivery of dendritic cells pulsed with IL-18 bound by SFV combined with the systemic administration of IL-12 enhanced the induction of the T helper type 1 response from tumor-specific CD4+ and CD8+ T cells and natural killer cells as well as antitumor immunity. Interferon-γ is partly responsible for this IL-18—mediated antitumor immunity. Furthermore, the protective immunity is mediated mainly by CD8+ T cells. Conclusions. Immunogene therapy that combines the local administration of dendritic cells pulsed with IL-18 bound by SFV and the systemic administration of IL-12 may be an excellent candidate for the development of a new treatment protocol. A self-replicating SFV system may therefore open a novel approach for the treatment of malignant brain tumor.


2012 ◽  
Vol 4 (4) ◽  
pp. 655-660 ◽  
Author(s):  
JING-YUE YANG ◽  
XIAO LI ◽  
LI GAO ◽  
ZENG-HUI TENG ◽  
WEN-CHAO LIU

2002 ◽  
Vol 25 (4) ◽  
pp. 289-303 ◽  
Author(s):  
Yaling Zhou ◽  
Marnix L. Bosch ◽  
Michael L. Salgaller

2014 ◽  
Vol 3 (9) ◽  
pp. e955684 ◽  
Author(s):  
Henri-Alexandre Michaud ◽  
Jean-François Eliaou ◽  
Virginie Lafont ◽  
Nathalie Bonnefoy ◽  
Laurent Gros

2021 ◽  
Vol 9 (5) ◽  
pp. e002054
Author(s):  
Francisco J Cueto ◽  
Carlos del Fresno ◽  
Paola Brandi ◽  
Alexis J. Combes ◽  
Elena Hernández-García ◽  
...  

BackgroundConventional type 1 dendritic cells (cDC1s) are central to antitumor immunity and their presence in the tumor microenvironment associates with improved outcomes in patients with cancer. DNGR-1 (CLEC9A) is a dead cell-sensing receptor highly restricted to cDC1s. DNGR-1 has been involved in both cross-presentation of dead cell-associated antigens and processes of disease tolerance, but its role in antitumor immunity has not been clarified yet.MethodsB16 and MC38 tumor cell lines were inoculated subcutaneously into wild-type (WT) and DNGR-1-deficient mice. To overexpress Flt3L systemically, we performed gene therapy through the hydrodynamic injection of an Flt3L-encoding plasmid. To characterize the immune response, we performed flow cytometry and RNA-Seq of tumor-infiltrating cDC1s.ResultsHere, we found that cross-presentation of tumor antigens in the steady state was DNGR-1-independent. However, on Flt3L systemic overexpression, tumor growth was delayed in DNGR-1-deficient mice compared with WT mice. Of note, this protection was recapitulated by anti-DNGR-1-blocking antibodies in mice following Flt3L gene therapy. This improved antitumor immunity was associated with Batf3-dependent enhanced accumulation of CD8+ T cells and cDC1s within tumors. Mechanistically, the deficiency in DNGR-1 boosted an Flt3L-induced specific inflammatory gene signature in cDC1s, including Ccl5 expression. Indeed, the increased infiltration of cDC1s within tumors and their protective effect rely on CCL5/CCR5 chemoattraction. Moreover, FLT3LG and CCL5 or CCR5 gene expression signatures correlate with an enhanced cDC1 signature and a favorable overall survival in patients with cancer. Notably, cyclophosphamide elevated serum Flt3L levels and, in combination with the absence of DNGR-1, synergized against tumor growth.ConclusionDNGR-1 limits the accumulation of tumor-infiltrating cDC1s promoted by Flt3L. Thus, DNGR-1 blockade may improve antitumor immunity in tumor therapy settings associated to high Flt3L expression.


2021 ◽  
Author(s):  
Jeong-Mi Lee ◽  
Kwang-Soo Shin ◽  
Choong-Hyun Koh ◽  
Boyeong Song ◽  
Insu Jeon ◽  
...  

Author(s):  
Leng Han ◽  
Lulu Bai ◽  
Chunjing Qu ◽  
Enyong Dai ◽  
Jiao Liu ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document