Journal for ImmunoTherapy of Cancer (Feb 2021)

Therapeutic depletion of CCR8+ tumor-infiltrating regulatory T cells elicits antitumor immunity and synergizes with anti-PD-1 therapy

  • Bart Neyns,
  • Damya Laoui,
  • Julia Katharina Schwarze,
  • Helena Van Damme,
  • Bruno Dombrecht,
  • Máté Kiss,
  • Heleen Roose,
  • Eva Van Overmeire,
  • Daliya Kancheva,
  • Liesbet Martens,
  • Aleksandar Murgaski,
  • Pauline Madeleine Rachel Bardet,
  • Gillian Blancke,
  • Maude Jans,
  • Evangelia Bolli,
  • Maria Solange Martins,
  • Yvon Elkrim,
  • Kiavash Movahedi,
  • Niels Vandamme,
  • Sebahat Ocak,
  • Isabelle Scheyltjens,
  • Lars Vereecke,
  • Frank Aboubakar Nana,
  • Pascal Merchiers,
  • Jo Agnes Van Ginderachter

DOI
https://doi.org/10.1136/jitc-2020-001749
Journal volume & issue
Vol. 9, no. 2

Abstract

Read online

Background Modulation and depletion strategies of regulatory T cells (Tregs) constitute valid approaches in antitumor immunotherapy but suffer from severe adverse effects due to their lack of selectivity for the tumor-infiltrating (ti-)Treg population, indicating the need for a ti-Treg specific biomarker.Methods We employed single-cell RNA-sequencing in a mouse model of non-small cell lung carcinoma (NSCLC) to obtain a comprehensive overview of the tumor-infiltrating T-cell compartment, with a focus on ti-Treg subpopulations. These findings were validated by flow cytometric analysis of both mouse (LLC-OVA, MC38 and B16-OVA) and human (NSCLC and melanoma) tumor samples. We generated two CCR8-specific nanobodies (Nbs) that recognize distinct epitopes on the CCR8 extracellular domain. These Nbs were formulated as tetravalent Nb-Fc fusion proteins for optimal CCR8 binding and blocking, containing either an antibody-dependent cell-mediated cytotoxicity (ADCC)-deficient or an ADCC-prone Fc region. The therapeutic use of these Nb-Fc fusion proteins was evaluated, either as monotherapy or as combination therapy with anti-programmed cell death protein-1 (anti-PD-1), in both the LLC-OVA and MC38 mouse models.Results We were able to discern two ti-Treg populations, one of which is characterized by the unique expression of Ccr8 in conjunction with Treg activation markers. Ccr8 is also expressed by dysfunctional CD4+ and CD8+ T cells, but the CCR8 protein was only prominent on the highly activated and strongly T-cell suppressive ti-Treg subpopulation of mouse and human tumors, with no major CCR8-positivity found on peripheral Tregs. CCR8 expression resulted from TCR-mediated Treg triggering in an NF-κB-dependent fashion, but was not essential for the recruitment, activation nor suppressive capacity of these cells. While treatment of tumor-bearing mice with a blocking ADCC-deficient Nb-Fc did not influence tumor growth, ADCC-prone Nb-Fc elicited antitumor immunity and reduced tumor growth in synergy with anti-PD-1 therapy. Importantly, ADCC-prone Nb-Fc specifically depleted ti-Tregs in a natural killer (NK) cell-dependent fashion without affecting peripheral Tregs.Conclusions Collectively, our findings highlight the efficacy and safety of targeting CCR8 for the depletion of tumor-promoting ti-Tregs in combination with anti-PD-1 therapy.