BiTE molecules are being investigated in solid tumor trials but the regulation of their activity is unclear. Kim et al. identified an effector memory subpopulation of CD8+ T cells from PBMCs and human tumors that expressed high levels of cytolytic molecules and was highly potent when combined with a BiTE. These CD45RA+CCR7- TEMRA cells expressed leukocyte Ig-like receptor B1 (LILRB1) that acts as a negative regulator of BiTE function. PD-1 and LILRB1 were independently expressed on different CD8+ populations and blocking both synergistically increased CD8+ T cell function. Blockade of LILRB1 may specifically enhance BiTE therapy.

Contributed by Katherine Turner

Elicitation of tumor cell killing by CD8(+) T cells is an effective therapeutic approach for cancer. In addition to using immune checkpoint blockade to reinvigorate existing but unresponsive tumor-specific T cells, alternative therapeutic approaches have been developed, including stimulation of polyclonal T cell cytolytic activity against tumors using bispecific T cell engager (BiTE) molecules that simultaneously engage the TCR complex and a tumor-associated Ag. BiTE molecules are efficacious against hematologic tumors and are currently being explored as an immunotherapy for solid tumors. To understand mechanisms regulating BiTE molecule--mediated CD8(+) T cell activity against solid tumors, we sought to define human CD8(+) T cell populations that efficiently respond to BiTE molecule stimulation and identify factors regulating their cytolytic activity. We find that human CD45RA(+)CCR7(-) CD8(+) T cells are highly responsive to BiTE molecule stimulation, are enriched in genes associated with cytolytic effector function, and express multiple unique inhibitory receptors, including leukocyte Ig-like receptor B1 (LILRB1). LILRB1 and programmed cell death protein 1 (PD1) were found to be expressed by distinct CD8(+) T cell populations, suggesting different roles in regulating the antitumor response. Engaging LILRB1 with its ligand HLA-G on tumor cells significantly inhibited BiTE molecule-induced CD8(+) T cell activation. Blockades of LILRB1 and PD1 induced greater CD8(+) T cell activation than either treatment alone. Together, our data suggest that LILRB1 functions as a negative regulator of human CD8(+) effector T cells and that blocking LILRB1 represents a unique strategy to enhance BiTE molecule therapeutic activity against solid tumors.

Author Info: (1) Department of Inflammation and Oncology, Amgen Research, Amgen Inc., South San Francisco, CA 94080. (2) Department of Inflammation and Oncology, Amgen Research, Amgen Inc., Sou

Author Info: (1) Department of Inflammation and Oncology, Amgen Research, Amgen Inc., South San Francisco, CA 94080. (2) Department of Inflammation and Oncology, Amgen Research, Amgen Inc., South San Francisco, CA 94080. (3) Genome Analysis Unit, Amgen Research, Amgen Inc., South San Francisco, CA 94080. (4) Research Informatics, Amgen Research, Amgen Inc., South San Francisco, CA 94080. (5) Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom. (6) Biomedical Pioneering Innovation Center, Peking University, Beijing 100871, China; and. Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing 100871, China. (7) Department of Inflammation and Oncology, Amgen Research, Amgen Inc., South San Francisco, CA 94080. (8) Department of Inflammation and Oncology, Amgen Research, Amgen Inc., South San Francisco, CA 94080. (9) Department of Inflammation and Oncology, Amgen Research, Amgen Inc., South San Francisco, CA 94080; xiyu@amgen.com.