OncoImmunology (Dec 2022)

An Fc-inert PD-L1×4-1BB bispecific antibody mediates potent anti-tumor immunity in mice by combining checkpoint inhibition and conditional 4-1BB co-stimulation

  • Alexander Muik,
  • Isil Altintas,
  • Friederike Gieseke,
  • Kristina B Schoedel,
  • Saskia M Burm,
  • Aras Toker,
  • Theodora W. Salcedo,
  • Dennis Verzijl,
  • David Eisel,
  • Christian Grunwitz,
  • Lena M Kranz,
  • Mathias Vormehr,
  • David P.E. Satijn,
  • Mustafa Diken,
  • Sebastian Kreiter,
  • Kate Sasser,
  • Tahamtan Ahmadi,
  • Özlem Türeci,
  • Esther C.W. Breij,
  • Maria Jure-Kunkel,
  • Ugur Sahin

DOI
https://doi.org/10.1080/2162402X.2022.2030135
Journal volume & issue
Vol. 11, no. 1

Abstract

Read online

Immune checkpoint inhibitors (ICI) targeting the PD-1/PD-L1 axis have changed the treatment paradigm for advanced solid tumors; however, many patients experience treatment resistance. In preclinical models 4-1BB co-stimulation synergizes with ICI by activating cytotoxic T- and NK-cell-mediated anti-tumor immunity. Here we characterize the mechanism of action of a mouse-reactive Fc-inert PD-L1×4-1BB bispecific antibody (mbsAb-PD-L1×4-1BB) and provide proof-of-concept for enhanced anti-tumor activity. In reporter assays mbsAb-PD-L1×4-1BB exhibited conditional 4-1BB agonist activity that was dependent on simultaneous binding to PD-L1. mbsAb-PD-L1×4-1BB further blocked the PD-L1/PD-1 interaction independently of 4-1BB binding. By combining both mechanisms, mbsAb-PD-L1×4-1BB strongly enhanced T-cell proliferation, cytokine production and antigen-specific cytotoxicity using primary mouse cells in vitro. Furthermore, mbsAb-PD-L1×4-1BB exhibited potent anti-tumor activity in the CT26 and MC38 models in vivo, leading to the rejection of CT26 tumors that were unresponsive to PD-L1 blockade alone. Anti-tumor activity was associated with increased tumor-specific CD8+ T cells and reduced regulatory T cells within the tumor microenvironment and tumor-draining lymph nodes. In immunocompetent tumor-free mice, mbsAb-PD-L1×4-1BB treatment neither induced T-cell infiltration into the liver nor elevated liver enzymes in the blood. Dual targeting of PD-L1 and 4-1BB with a bispecific antibody may therefore address key limitations of first generation 4-1BB-agonistic antibodies, and may provide a novel approach to improve PD-1/PD-L1 checkpoint blockade.

Keywords