De Vries et al. demonstrated that γδ T cells contributed to the response to immune checkpoint blockade in patients with β2-microglobulin (B2M)-negative DNA mismatch repair-deficient (MMR-d) colon cancers. In MMR-d cancers in TCGA, B2M defects were associated with increased expression of Vδ1 and Vδ3 TCRs and KIRs, and single-cell sequencing of tumors showed these cells expressed PD-1 and cytotoxic molecules. PD-1+ Vδ1 and Vδ3 T cells isolated from MMR-d colon cancers showed preferential reactivity to HLA-class-I-negative cancer cell lines and organoids. In B2M-deficient colon cancers, dual PD-1 and CTLA-4 blockade substantially increased the frequency of γδ T cells.

Contributed by Shishir Pant

ABSTRACT: DNA mismatch repair-deficient (MMR-d) cancers present an abundance of neoantigens that is thought to explain their exceptional responsiveness to immune checkpoint blockade (ICB)1,2. Here, in contrast to other cancer types3-5, we observed that 20 out of 21 (95%) MMR-d cancers with genomic inactivation of β2-microglobulin (encoded by B2M) retained responsiveness to ICB, suggesting the involvement of immune effector cells other than CD8+ T cells in this context. We next identified a strong association between B2M inactivation and increased infiltration by γδ T cells in MMR-d cancers. These γδ T cells mainly comprised the Vδ1 and Vδ3 subsets, and expressed high levels of PD-1, other activation markers, including cytotoxic molecules, and a broad repertoire of killer-cell immunoglobulin-like receptors. In vitro, PD-1+ γδ T cells that were isolated from MMR-d colon cancers exhibited enhanced reactivity to human leukocyte antigen (HLA)-class-I-negative MMR-d colon cancer cell lines and B2M-knockout patient-derived tumour organoids compared with antigen-presentation-proficient cells. By comparing paired tumour samples from patients with MMR-d colon cancer that were obtained before and after dual PD-1 and CTLA-4 blockade, we found that immune checkpoint blockade substantially increased the frequency of γδ T cells in B2M-deficient cancers. Taken together, these data indicate that γδ T cells contribute to the response to immune checkpoint blockade in patients with HLA-class-I-negative MMR-d colon cancers, and underline the potential of γδ T cells in cancer immunotherapy.

Author Info: (1) Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands. Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands. (2) Dep

Author Info: (1) Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands. Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands. (2) Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands. Oncode Institute, Utrecht, The Netherlands. Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands. (3) Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands. Oncode Institute, Utrecht, The Netherlands. (4) Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands. Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands. Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands. (5) Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands. (6) Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands. (7) Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands. (8) Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands. (9) Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands. (10) Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands. Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands. (11) Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands. Oncode Institute, Utrecht, The Netherlands. (12) Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands. Oncode Institute, Utrecht, The Netherlands. (13) Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands. Oncode Institute, Utrecht, The Netherlands. (14) Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands. Oncode Institute, Utrecht, The Netherlands. (15) Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands. (16) Department of Medical Oncology, Erasmus MC, Rotterdam, The Netherlands. (17) Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands. Oncode Institute, Utrecht, The Netherlands. Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands. (18) Oncode Institute, Utrecht, The Netherlands. Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands. Faculty of EEMCS, Delft University of Technology, Delft, The Netherlands. (19) Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands. (20) Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands. n.f.de_miranda@lumc.nl. (21) Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands. e.voest@nki.nl. Oncode Institute, Utrecht, The Netherlands. e.voest@nki.nl.