File Download

  • Find it @ UNIST can give you direct access to the published full text of this article. (UNISTARs only)
Related Researcher

박성호

Park, Sung Ho
Laboratory of Molecular Immunology
Read More

Views & Downloads

Detailed Information

Cited time in webofscience Cited time in scopus
Metadata Downloads

Lactate oxidase/catalase-displaying nanoparticles efficiently consume lactate in the tumor microenvironment to effectively suppress tumor growth

Author(s)
Choi, HyukjunYeo, MiraeKang, YujinKim, Hyo JeongPark, Seong GukJang, EunjungPark, Sung HoKim, EunheeKang, Sebyung
Issued Date
2023-01
DOI
10.1186/s12951-022-01762-6
URI
https://scholarworks.unist.ac.kr/handle/201301/61581
Citation
JOURNAL OF NANOBIOTECHNOLOGY, v.21, no.1, pp.5
Abstract
AbstractThe aggressive proliferation of tumor cells often requires increased glucose uptake and excessive anaerobic glycolysis, leading to the massive production and secretion of lactate to form a unique tumor microenvironment (TME). Therefore, regulating appropriate lactate levels in the TME would be a promising approach to control tumor cell proliferation and immune suppression. To effectively consume lactate in the TME, lactate oxidase (LOX) and catalase (CAT) were displayed onto Aquifex aeolicus lumazine synthase protein nanoparticles (AaLS) to form either AaLS/LOX or AaLS/LOX/CAT. These complexes successfully consumed lactate produced by CT26 murine colon carcinoma cells under both normoxic and hypoxic conditions. Specifically, AaLS/LOX generated a large amount of H2O2 with complete lactate consumption to induce drastic necrotic cell death regardless of culture condition. However, AaLS/LOX/CAT generated residual H2O2, leading to necrotic cell death only under hypoxic condition similar to the TME. While the local administration of AaLS/LOX to the tumor site resulted in mice death, that of AaLS/LOX/CAT significantly suppressed tumor growth without any severe side effects. AaLS/LOX/CAT effectively consumed lactate to produce adequate amounts of H2O2 which sufficiently suppress tumor growth and adequately modulate the TME, transforming environments that are favorable to tumor suppressive neutrophils but adverse to tumor-supportive tumor-associated macrophages. Collectively, these findings showed that the modular functionalization of protein nanoparticles with multiple metabolic enzymes may offer the opportunity to develop new enzyme complex-based therapeutic tools that can modulate the TME by controlling cancer metabolism.
Graphical Abstract
Publisher
BioMed Central
ISSN
1477-3155
Keyword (Author)
Lactate oxidaseCatalaseTumor microenvironmentLactate consumptionTumor suppression
Keyword
AEROCOCCUS-VIRIDANSLUNG-CANCERLACTIC-ACIDPOLARIZATIONMACROPHAGESMETABOLISMCATALASEREVEALSOXIDASEPROTEIN

qrcode

Items in Repository are protected by copyright, with all rights reserved, unless otherwise indicated.