1. Academic Validation
  2. Antimetabolite pemetrexed primes a favorable tumor microenvironment for immune checkpoint blockade therapy

Antimetabolite pemetrexed primes a favorable tumor microenvironment for immune checkpoint blockade therapy

  • J Immunother Cancer. 2020 Nov;8(2):e001392. doi: 10.1136/jitc-2020-001392.
Chia-Sing Lu  # 1 Ching-Wen Lin  # 2 Ya-Hsuan Chang 3 Hsuan-Yu Chen 3 Wei-Chia Chung 2 Wei-Yun Lai 2 Chao-Chi Ho 1 Tong-Hong Wang 4 Chi-Yuan Chen 4 Chen-Lin Yeh 1 Sean Wu 2 Shu-Ping Wang 5 Pan-Chyr Yang 6 2 7
Affiliations

Affiliations

  • 1 Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan.
  • 2 Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
  • 3 Institute of Statistical Science, Academia Sinica, Taipei, Taiwan.
  • 4 Tissue Bank, Chang Gung Memorial Hospital; Graduate Institute of Health Industry Technology and Research Center for Industry of Human Ecology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
  • 5 Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan [email protected] [email protected].
  • 6 Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan [email protected] [email protected].
  • 7 Institute of Biomedical Sciences and Genomics Research Center, Academia Sinica, Taipei, Taiwan.
  • # Contributed equally.
Abstract

Background: The immune checkpoint blockade (ICB) targeting programmed cell death-1 (PD-1) and its ligand (PD-L1) has been proved beneficial for numerous types of cancers, including non-small-cell lung Cancer (NSCLC). However, a significant number of patients with NSCLC still fail to respond to ICB due to unfavorable tumor microenvironment. To improve the efficacy, the immune-chemotherapy combination with pemetrexed, cis/carboplatin and pembrolizumab (anti-PD-1) has been recently approved as first-line treatment in advanced NSCLCs. While chemotherapeutic agents exert beneficial effects, the underlying antitumor mechanism(s) remains unclear.

Methods: Pemetrexed, cisplatin and other chemotherapeutic agents were tested for the potential to induce PD-L1 expression in NSCLC cells by immunoblotting and flow cytometry. The ability to prime the tumor immune microenvironment was then determined by NSCLC/T cell coculture systems and syngeneic mouse models. Subpopulations of NSCLC cells responding differently to pemetrexed were selected and subjected to RNA-sequencing analysis. The key signaling pathways were identified and validated in vitro and in vivo.

Results: Pemetrexed induced the transcriptional activation of PD-L1 (encoded by CD274) by inactivating Thymidylate Synthase (TS) in NSCLC cells and, in turn, activating T-lymphocytes when combined with the anti-PD-1/PD-L1 therapy. Nuclear factor κB (NF-κB) signaling was activated by intracellular Reactive Oxygen Species (ROSs) that were elevated by pemetrexed-mediated TS inactivation. The TS-ROS-NF-κB regulatory axis actively involves in pemetrexed-induced PD-L1 upregulation, whereas when pemetrexed fails to induce PD-L1 expression in NSCLC cells, NF-κB signaling is unregulated. In syngeneic mouse models, the combinatory treatment of pemetrexed with anti-PD-L1 antibody created a more favorable tumor microenvironment for the inhibition of tumor growth.

Conclusions: Our findings reveal novel mechanisms showing that pemetrexed upregulates PD-L1 expression and primes a favorable microenvironment for ICB, which provides a mechanistic basis for the combinatory chemoimmunotherapy in NSCLC treatment.

Keywords

combination; costimulatory and inhibitory t-cell receptors; drug therapy; immunotherapy; tumor escape; tumor microenvironment.

Figures
Products