4.8 Article

An optimized ionizable cationic lipid for brain tumor-targeted siRNA delivery and glioblastoma immunotherapy

Journal

BIOMATERIALS
Volume 287, Issue -, Pages -

Publisher

ELSEVIER SCI LTD
DOI: 10.1016/j.biomaterials.2022.121645

Keywords

Blood -brain barrier; Cationic lipid nanoparticle; Glioblastoma multiforme; Tumor immunotherapy; Tumor microenvironment

Funding

  1. National Key Research and Development Program of China [2017YFA0208100]
  2. National Natural Science Foundation of China (NSFC) [81871478, 91642208, 21778056, 22077125, 81941008, 81422026]
  3. Jilin Scientific and Technological Development Program [20190201094JC, 20200301007RQ]
  4. Interdisciplinary Innovation Project of the First Hospital of Jilin University [JDYYJCHX001]
  5. Fundamental Research Funds for the Central Universities, JLU

Ask authors/readers for more resources

This study develops a novel cationic lipid nanoparticle (LNP) that efficiently delivers siRNA across the blood-brain barrier (BBB) and targets the tumor microenvironment in the brain of mice for immunotherapy of glioblastoma multiforme (GBM). The LNP is effective in delivering siRNA against CD47 and PD-L1 into the cranial GBM in mice, resulting in downregulation of target gene expression and activation of antitumor immunity.
Glioblastoma multiforme (GBM) is the most common and aggressive primary brain tumor with a high mortality rate. Immunotherapy has achieved promising clinical results in multiple cancers, but shows unsatisfactory outcome in GBM patients, and poor drug delivery across the blood-brain barrier (BBB) is believed to be one of the main limitations that hinder the therapeutic efficacy of drugs. Herein, a new cationic lipid nanoparticle (LNP) that can efficiently deliver siRNA across BBB and target mouse brain is prepared for modulating the tumor microenvironment for GBM immunotherapy. By designing and screening cationic LNPs with different ionizable amine headgroups, a lipid (named as BAMPA-O16B) is identified with an optimal acid dissociation constant (pKa) that significantly enhances the cellular uptake and endosomal escape of siRNA lipoplex in mouse GBM cells. Importantly, BAMPA-O16B/siRNA lipoplex is highly effective to deliver siRNA against CD47 and PD-L1 across the BBB into cranial GBM in mice, and downregulate target gene expression in the tumor, resulting in synergistically activating a T cell-dependent antitumor immunity in orthotopic GBM. Collectively, this study offers an effective strategy for brain targeted siRNA delivery and gene silencing by optimizing the physicochemical property of LNPs. The effectiveness of modulating immune environment of GBM could further be expanded for potential treatment of other brain tumors.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available