4.8 Article

Glycolytic activation of peritumoral monocytes fosters immune privilege via the PFKFB3-PD-L1 axis in human hepatocellular carcinoma

Journal

JOURNAL OF HEPATOLOGY
Volume 71, Issue 2, Pages 333-343

Publisher

ELSEVIER SCIENCE BV
DOI: 10.1016/j.jhep.2019.04.007

Keywords

Glycolysis; PFKFB3; Tumor-associated monocytes; Immune checkpoint inhibitors; NF-kappa B; PD-L1; Cancer; HCC

Funding

  1. National 135 Major Project of China [2017YFA0505803, 2018ZX10302205]
  2. National Natural Science Foundation of China [81773054, 81730044, 91842308]
  3. National Postdoctoral Program for Innovative Talents [BX20180398]
  4. China Postdoctoral Science Foundation [2019M653169]
  5. Fundamental Research Funds for the Central Universities [171gjc32]

Ask authors/readers for more resources

Background & Aims: Programmed cell death 1 ligand 1 (PD-L1) expression on antigen-presenting cells is essential for T cell impairment, and PD-Ll -expressing macrophages may mechanistically shape and therapeutically predict the clinical efficacy of PD-Ll or programmed cell death 1 blockade. We aimed to elucidate the mechanisms underlying PD-Ll upregulation in human tumor microenvironments, which remain poorly understood despite the clinical success of immune checkpoint inhibitors. Methods: Monocytes/macrophages were purified from peripheral blood, non-tumor, or paired tumor tissues of patients with hepatocellular carcinoma (HCC), and their possible glycolytic switch was evaluated. The underlying regulatory mechanisms and clinical significance of metabolic switching were studied with both ex vivo analyses and in vitro experiments. Results: We found that monocytes significantly enhanced the levels of glycolysis at the peritumoral region of human HCC. The activation of glycolysis induced PD-Ll expression on these cells and subsequently attenuated cytotoxic T lymphocyte responses in tumor tissues. Mechanistically, tumor-derived soluble factors, including hyaluronan fragments, induced the upregulation of a key glycolytic enzyme, PFKFB3, in tumor-associated monocytes. This enzyme not only modulated the cellular metabolic switch but also mediated the increased expression of PD-Ll by activating the nuclear factor kappa B signaling pathway in these cells. Consistently, the levels of PFKFB3(+) CD68(+) cell infiltration in peritumoral tissues were negatively correlated with overall survival and could serve as an independent prognostic factor for survival in patients with HCC. Conclusions: Our results reveal a mechanism by which the cellular metabolic switch regulates the pro-tumor functions of monocytes in a specific human tumor microenvironment. PFKFB3 in both cancer cells and tumor-associated monocytes is a potential therapeutic target in human HCC. Lay summary: Programmed cell death 1 ligand 1 (PD-L1) expressed on antigen-presenting cells, rather than tumor cells, has been reported to play an essential role in checkpoint blockade therapy. A fundamental understanding of mechanisms that regulate the expression of PD-Ll on tumor-infiltrating monocytes/macrophages will undoubtedly lead to the possibility of developing novel PD-Ll blockade strategies with high specificity and efficiency. The current study unveils a novel mechanism by which metabolic switching links immune activation responses to immune tolerance in the tumor milieu, identifying potential targets for future immune-based anti-cancer therapies. (C) 2019 European Association for the Study of the Liver. Published by Elsevier B.V. All rights reserved.

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