Dual TLR9 and PD-L1 targeting unleashes dendritic cells to induce durable antitumor immunity.

Details

Ressource 1Download: 37208130_BIB_334B6B7FC1F4.pdf (7000.63 [Ko])
State: Public
Version: Final published version
License: CC BY-NC 4.0
Serval ID
serval:BIB_334B6B7FC1F4
Type
Article: article from journal or magazin.
Collection
Publications
Institution
Title
Dual TLR9 and PD-L1 targeting unleashes dendritic cells to induce durable antitumor immunity.
Journal
Journal for immunotherapy of cancer
Author(s)
Fernandez-Rodriguez L., Cianciaruso C., Bill R., Trefny M.P., Klar R., Kirchhammer N., Buchi M., Festag J., Michel S., Kohler R.H., Jones E., Maaske A., Kashyap A.S., Jaschinski F., Dixon K.O., Pittet M.J., Zippelius A.
ISSN
2051-1426 (Electronic)
ISSN-L
2051-1426
Publication state
Published
Issued date
05/2023
Peer-reviewed
Oui
Volume
11
Number
5
Pages
e006714
Language
english
Notes
Publication types: Journal Article
Publication Status: ppublish
Abstract
Although immune checkpoint inhibitors have been a breakthrough in clinical oncology, these therapies fail to produce durable responses in a significant fraction of patients. This lack of long-term efficacy may be due to a poor pre-existing network linking innate and adaptive immunity. Here, we present an antisense oligonucleotide (ASO)-based strategy that dually targets toll-like receptor 9 (TLR9) and programmed cell death ligand 1 (PD-L1), aiming to overcome resistance to anti-PD-L1 monoclonal therapy.
We designed a high-affinity immunomodulatory IM-TLR9:PD-L1-ASO antisense oligonucleotide (hereafter, IM-T9P1-ASO) targeting mouse PD-L1 messenger RNA and activating TLR9. Then, we performed in vitro and in vivo studies to validate the IM-T9P1-ASO activity, efficacy, and biological effects in tumors and draining lymph nodes. We also performed intravital imaging to study IM-T9P1-ASO pharmacokinetics in the tumor.
IM-T9P1-ASO therapy, unlike PD-L1 antibody therapy, results in durable antitumor responses in multiple mouse cancer models. Mechanistically, IM-T9P1-ASO activates a state of tumor-associated dendritic cells (DCs), referred to here as DC3s, which have potent antitumor potential but express the PD-L1 checkpoint. IM-T9P1-ASO has two roles: it triggers the expansion of DC3s by engaging with TLR9 and downregulates PD-L1, thereby unleashing the antitumor functions of DC3s. This dual action leads to tumor rejection by T cells. The antitumor efficacy of IM-T9P1-ASO depends on the antitumor cytokine interleukin-12 (IL-12), produced by DC3s, and Batf3, a transcription factor required for DC development.
By simultaneously targeting TLR9 and PD-L1, IM-T9P1-ASO amplifies antitumor responses via DC activation, leading to sustained therapeutic efficacy in mice. By highlighting differences and similarities between mouse and human DCs, this study could serve to develop similar therapeutic strategies for patients with cancer.
Keywords
Humans, Mice, Animals, Toll-Like Receptor 9/metabolism, Immunotherapy/methods, Neoplasms/drug therapy, Oligonucleotides, Antisense, Dendritic Cells, combined modality therapy, dendritic cells, immune checkpoint inhibitors, immunotherapy
Pubmed
Web of science
Open Access
Yes
Create date
30/05/2023 11:44
Last modification date
25/01/2024 8:33
Usage data