scholarly journals Targeting dendritic cells in situ for breast cancer immunotherapy

2012 ◽  
Vol 1 (8) ◽  
pp. 1398-1400 ◽  
Author(s):  
Bei Wang
2020 ◽  
Vol 69 (6) ◽  
pp. 951-967 ◽  
Author(s):  
Meng Zhang ◽  
Yanmei Shi ◽  
Yujuan Zhang ◽  
Yifan Wang ◽  
Faizah Alotaibi ◽  
...  

2021 ◽  
Vol 9 (5) ◽  
pp. e002432
Author(s):  
Takaaki Oba ◽  
Kenichi Makino ◽  
Ryutaro Kajihara ◽  
Toshihiro Yokoi ◽  
Ryoko Araki ◽  
...  

BackgroundDendritic cells (DCs) are a promising therapeutic target in cancer immunotherapy given their ability to prime antigen-specific T cells, and initiate antitumor immune response. A major obstacle for DC-based immunotherapy is the difficulty to obtain a sufficient number of functional DCs. Theoretically, this limitation can be overcome by using induced pluripotent stem cells (iPSCs); however, therapeutic strategies to engage iPSC-derived DCs (iPSC-DCs) into cancer immunotherapy remain to be elucidated. Accumulating evidence showing that induction of tumor-residing DCs enhances immunomodulatory effect of radiotherapy (RT) prompted us to investigate antitumor efficacy of combining intratumoral administration of iPSC-DCs with local RT.MethodsMouse iPSCs were differentiated to iPSC-DCs on OP9 stromal cells expressing the notch ligand delta-like 1 in the presence of granulocyte macrophage colony-stimulating factor. Phenotype and the capacities of iPSC-DCs to traffic tumor-draining lymph nodes (TdLNs) and prime antigen-specific T cells were evaluated by flow cytometry and imaging flow cytometry. Antitumor efficacy of intratumoral injection of iPSC-DCs and RT was tested in syngeneic orthotopic mouse tumor models resistant to anti-PD-1 ligand 1 (PD-L1) therapy.ResultsMouse iPSC-DCs phenotypically resembled conventional type 2 DCs, and had a capacity to promote activation, proliferation and effector differentiation of antigen-specific CD8+ T cells in the presence of the cognate antigen in vitro. Combination of in situ administration of iPSC-DCs and RT facilitated the priming of tumor-specific CD8+ T cells, and synergistically delayed the growth of not only the treated tumor but also the distant non-irradiated tumors. Mechanistically, RT enhanced trafficking of intratumorally injected iPSC-DCs to the TdLN, upregulated CD40 expression, and increased the frequency of DC/CD8+ T cell aggregates. Phenotypic analysis of tumor-infiltrating CD8+ T cells and myeloid cells revealed an increase of stem-like Slamf6+ TIM3− CD8+ T cells and PD-L1 expression in tumor-associated macrophages and DCs. Consequently, combined therapy rendered poorly immunogenic tumors responsive to anti-PD-L1 therapy along with the development of tumor-specific immunological memory.ConclusionsOur findings illustrate the translational potential of iPSC-DCs, and identify the therapeutic efficacy of a combinatorial platform to engage them for overcoming resistance to anti-PD-L1 therapy in poorly immunogenic tumors.


ACS Nano ◽  
2019 ◽  
Vol 13 (7) ◽  
pp. 7568-7577 ◽  
Author(s):  
Yi Wang ◽  
Yao-Xin Lin ◽  
Jie Wang ◽  
Sheng-Lin Qiao ◽  
Yu-Ying Liu ◽  
...  

2021 ◽  
Author(s):  
Shin‐ichiro Fujii ◽  
Satoru Yamasaki ◽  
Kenichi Hanada ◽  
Shogo Ueda ◽  
Masami Kawamura ◽  
...  

Vaccine ◽  
2010 ◽  
Vol 28 (17) ◽  
pp. 2965-2972 ◽  
Author(s):  
Alice Banz ◽  
Magali Cremel ◽  
Audrey Rembert ◽  
Yann Godfrin

2012 ◽  
Vol 132 (4) ◽  
pp. 967-977 ◽  
Author(s):  
Xiufen Zheng ◽  
James Koropatnick ◽  
Di Chen ◽  
Thomas Velenosi ◽  
Hong Ling ◽  
...  

2001 ◽  
Vol 120 (5) ◽  
pp. A183-A183
Author(s):  
H KOBAYASHI ◽  
H NAGATA ◽  
S MIURA ◽  
T AZUMA ◽  
H SUZUKI ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document