1. Academic Validation
  2. Combination of EP4 antagonist MF-766 and anti-PD-1 promotes anti-tumor efficacy by modulating both lymphocytes and myeloid cells

Combination of EP4 antagonist MF-766 and anti-PD-1 promotes anti-tumor efficacy by modulating both lymphocytes and myeloid cells

  • Oncoimmunology. 2021 Mar 18;10(1):1896643. doi: 10.1080/2162402X.2021.1896643.
Yun Wang 1 Long Cui 2 Peter Georgiev 1 Latika Singh 1 Yanyan Zheng 1 Ying Yu 1 Jeff Grein 3 Chunsheng Zhang 3 Eric S Muise 3 David L Sloman 4 Heidi Ferguson 5 Hongshi Yu 5 Cristina St Pierre 1 Pranal J Dakle 1 Vincenzo Pucci 6 James Baker 6 Andrey Loboda 3 Doug Linn 2 Christopher Brynczka 7 Doug Wilson 3 Brian B Haines 2 Brian Long 2 Richard Wnek 8 Svetlana Sadekova 1 Michael Rosenzweig 1 Andrew Haidle 4 Yongxin Han 4 Sheila H Ranganath 1
Affiliations

Affiliations

  • 1 Department of Oncology Early Discovery, Merck & Co., Inc., Boston, Massachusetts, USA.
  • 2 Department of Quantitative Biosciences, Merck & Co., Inc., Boston, Massachusetts, USA.
  • 3 Department of Genetics and Pharmacogenomics, Merck & Co., Inc., Boston, Massachusetts, USA.
  • 4 Department of Discovery Chemistry, Merck & Co., Inc., Boston, Massachusetts, USA.
  • 5 Department of Pharmaceutical Science, Merck & Co., Inc., Boston, Massachusetts, USA.
  • 6 Department of Pharmacokinetics, Pharmacodynamics & Drug Metabolism, Merck & Co., Inc., Boston, Massachusetts, USA.
  • 7 Dept. Safety and Exploratory Pharmacology, Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., Boston, Massachusetts, USA.
  • 8 Department of Translational Biomarkers, Merck & Co., Inc., Boston, Massachusetts, USA.
Abstract

Prostaglandin E2 (PGE2), an arachidonic acid pathway metabolite produced by cyclooxygenase (COX)-1/2, has been shown to impair anti-tumor immunity through engagement with one or more E-type prostanoid receptors (EP1-4). Specific targeting of EP receptors, as opposed to COX-1/2 inhibition, has been proposed to achieve preferential antagonism of PGE2-mediated immune suppression. Here we describe the anti-tumor activity of MF-766, a potent and highly selective small-molecule inhibitor of the EP4 receptor. EP4 inhibition by MF-766 synergistically improved the efficacy of anti-programmed cell death protein 1 (PD-1) therapy in CT26 and EMT6 syngeneic tumor mouse models. Multiparameter flow cytometry analysis revealed that treatment with MF-766 promoted the infiltration of CD8+ T cells, natural killer (NK) cells and conventional dendritic cells (cDCs), induced M1-like macrophage reprogramming, and reduced granulocytic myeloid-derived suppressor cells (MDSC) in the tumor microenvironment (TME). In vitro experiments demonstrated that MF-766 restored PGE2-mediated inhibition of lipopolysaccharide (LPS)-induced tumor necrosis factor (TNF)-α production in THP-1 cells and human blood, and PGE2-mediated inhibition of interleukin (IL)-2-induced interferon (IFN)-γ production in human NK cells. MF-766 reversed the inhibition of IFN-γ in CD8+ T-cells by PGE2 and impaired suppression of CD8+ T-cells induced by myeloid-derived suppressor cells (MDSC)/PGE2. In translational studies using primary human tumors, MF-766 enhanced anti-CD3-stimulated IFN-γ, IL-2, and TNF-α production in primary histoculture and synergized with pembrolizumab in a PGE2 high TME. Our studies demonstrate that the combination of EP4 blockade with anti-PD-1 therapy enhances antitumor activity by differentially modulating myeloid cell, NK cell, CDC and T-cell infiltration profiles.

Keywords

Immunotherapy; ep4 antagonism; lymphocytes; myeloid cells; pge2.

Figures
Products