Your browser doesn't support javascript.
loading
EP2/EP4 targeting prevents tumor-derived PGE2-mediated immunosuppression in cDC2s.
Cuenca-Escalona, Jorge; Bödder, Johanna; Subtil, Beatriz; Sánchez-Sánchez, Marta; Vidal-Manrique, Marcos; Sweep, Mark W D; Fauerbach, Jonathan A; Cambi, Alessandra; Flórez-Grau, Georgina; de Vries, I Jolanda M.
Affiliation
  • Cuenca-Escalona J; Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands.
  • Bödder J; Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands.
  • Subtil B; Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands.
  • Sánchez-Sánchez M; Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands.
  • Vidal-Manrique M; Laboratory of Hematology, Radboud University Medical Center, Nijmegen, the Netherlands.
  • Sweep MWD; Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands.
  • Fauerbach JA; Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands.
  • Cambi A; R&D Reagents, Chemical Biology Department, Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany.
  • Flórez-Grau G; Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands.
  • de Vries IJM; Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands.
J Leukoc Biol ; 2024 Jul 23.
Article in En | MEDLINE | ID: mdl-39041661
ABSTRACT
Tumor-derived prostaglandin E2 (PGE2) impairs anti-tumor immunity by priming suppressive functions on various immune cell types, including dendritic cells (DCs). In this way, tumors mediate DC dysfunction and hamper their anti-tumoral activity. PGE2 is known to modulate DC function via signaling through the E-prostanoid receptor type (EP) 2 and EP4. Preclinical studies have demonstrated the therapeutic value of targeting EP2/4 receptor signaling in DCs. Ongoing phase I clinical trials with EP antagonists have shown immunomodulation in cancer patients. However, the systemic drug administration leads to off-target events and subsequent side-effects. To limit the off-target effects of EP targeting, EP2 and EP4 antagonists were encapsulated in polymeric nanoparticles (NPs). In this study we evaluated the efficacy of EP2/4 specific antagonists encapsulated in NPs to protect cDC2s from suppressive effects of tumor-derived PGE2 in different tumor models. We show that tumor-derived PGE2 signals via EP2/4 to mediate the acquisition of a suppressive phenotype of cDC2s. EP2/4 antagonists encapsulated NPs impaired the conversion of cDC2s towards a suppressive state and inhibited the occurrence of suppressive features such as IL-10 production or the ability to expand Tregs. Importantly, the NPs abolished the transition towards this suppressive state in different tumor models Melanoma-conditioned media, ascites fluid derived from ovarian cancer patients (2D), and upon coculture with colorectal cancer patient-derived organoids (3D). We propose that targeting the PGE2-EP2/4 axis using NPs can achieve immunomodulation in the immune system of cancer patients, alleviate tumor-derived suppression, and thus facilitate the development of potent anti-tumor immunity in cancer patients.
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: J Leukoc Biol Year: 2024 Document type: Article Affiliation country: Países Bajos Country of publication: Reino Unido

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: J Leukoc Biol Year: 2024 Document type: Article Affiliation country: Países Bajos Country of publication: Reino Unido