Haematologica (Jun 2024)

Blockade of the CD47/SIRPα checkpoint axis potentiates the macrophage-mediated anti-tumor efficacy of tafasitamab

  • Alexander Biedermann,
  • Maria Patra-Kneuer,
  • Dimitrios Mougiakakos,
  • Maike Büttner-Herold,
  • Doris Mangelberger-Eberl,
  • Johannes Berges,
  • Christian Kellner,
  • Sarah Altmeyer,
  • Jörg Thomas Bittenbring,
  • Christian Augsberger,
  • Kristina Ilieva-Babinsky,
  • Stefan Haskamp,
  • Fabian Beier,
  • Christopher Lischer,
  • Julio Vera,
  • Anja Lührmann,
  • Simone Bertz,
  • Simon Völkl,
  • Benedikt Jacobs,
  • Stefan Steidl,
  • Andreas Mackensen,
  • Heiko Bruns

DOI
https://doi.org/10.3324/haematol.2023.284795
Journal volume & issue
Vol. 999, no. 1

Abstract

Read online

Macrophages are one of the key mediators of the therapeutic effects exerted by monoclonal antibodies, such as the anti-CD19 antibody tafasitamab, approved in combination with lenalidomide for the treatment of relapsed or refractory (r/r) diffuse large B cell lymphoma (DLBCL). However, antibody-dependent cellular phagocytosis (ADCP) in the tumor microenvironment can be counteracted by increased expression of the inhibitory receptor SIRPα on macrophages and its ligand, the immune checkpoint molecule CD47 on tumor cells. The aim of this study was to investigate the impact of the CD47-SIRPα axis on tafasitamabmediated phagocytosis and explore the potential of anti-CD47 blockade to enhance its antitumor activity. Elevated expression of both SIRPα and CD47 was observed in DLBCL patient-derived lymph node biopsies compared to healthy controls. CRISPR-mediated CD47 overexpression impacted tafasitamab-mediated ADCP in vitro and increased expression of SIRPα on macrophages correlated with decreased ADCP activity of tafasitamab against DLBCL cell lines. Combination of tafasitamab and an anti-CD47 blocking antibody enhanced ADCP activity of in vitro generated macrophages. Importantly, tafasitamab-mediated phagocytosis was elevated in combination with CD47 blockade using primary DLBCL cells and patient-derived lymphoma-associated macrophages (LAMs) in an autologous setting. Furthermore, lymphoma cells with low CD19 expression were efficiently eliminated by the combination treatment. Finally, combined treatment of tafasitamab and an anti-CD47 antibody resulted in enhanced tumor volume reduction and survival benefit in lymphoma xenograft mouse models. These findings provide evidence that CD47 blockade can enhance the phagocytic potential of tumor targeting immunotherapies such as tafasitamab and suggest there is value in exploring the combination in the clinic.