Rawat et al. demonstrated that natural Abs (Nabs) are critical for the early-stage recognition of and immune cell activation against adoptively transferred immunogenic and nonimmunogenic neoantigen-expressing cells and transplanted and spontaneous tumors. Adoptively transferred neoantigen-expressing cells persisted in NAb-deficient mice, whereas they were eliminated in wild-type (WT) mice. Restoration of the NAb repertoire in NAb-deficient mice with either WT B cells or naive serum elicited the recognition and elimination of neoantigen-expressing cells in a T- and B cell-dependent manner. The antitumor immune response was dependent on NAbs and not on the Ag-presenting property of B cells.

Contributed by Shishir Pant

ABSTRACT: Recent studies have revealed a critical role for natural Abs (NAbs) in antitumor immune responses. However, the role of NAbs in cancer immunosurveillance remains unexplored, mainly because of the lack of in vivo models that mimic the early recognition and elimination of transforming cells. In this article, we propose a role for NAbs in alerting the immune system against precancerous neoantigen-expressing cells immediately after they escape intrinsic tumor suppression mechanisms. We identify four distinct reproducible, trackable, MHC-matched neoantigen-expressing cell models that do not form tumors as the end point. This amplified readout in the critical window prior to tumor formation allows investigation of new mediators of cancer immunosurveillance. We found that neoantigen-expressing cells adoptively transferred in NAb-deficient mice persisted, whereas they were eliminated in wild-type mice, indicating that the circulating NAb repertoire alerts the immune system to the presence of transformed cells. Moreover, immunity is mounted against immunogenic and nonimmunogenic neoantigens contained in the NAb-tagged cells, regardless of whether the NAb directly recognizes the neoantigens. Beyond these neoantigen-expressing model systems, we observed a significantly greater tumor burden in chemically and virally induced tumor models in NAb-deficient mice compared with wild-type mice. Restoration of the NAb repertoire in NAb-deficient mice elicited the recognition and elimination of neoantigen-expressing cells and cancer. These data show that NAbs are required and sufficient for elimination of transformed cells early in tumorigenesis. These models can now be used to investigate how NAbs stimulate immunity via recognition receptors to eliminate precancerous cells.

Author Info: (1) Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, NH; and kavita.rawat@dartmouth.edu claudia.jakubzick@dartmouth.edu. (2) Department of B

Author Info: (1) Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, NH; and kavita.rawat@dartmouth.edu claudia.jakubzick@dartmouth.edu. (2) Department of Biomedical Data Science, Dartmouth Geisel School of Medicine, Hanover, NH. (3) Department of Biomedical Data Science, Dartmouth Geisel School of Medicine, Hanover, NH. (4) Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, NH; and. (5) Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, NH; and. (6) Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, NH; and. (7) Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, NH; and kavita.rawat@dartmouth.edu claudia.jakubzick@dartmouth.edu.