@article {Mastermane000691, author = {Kelly-Anne Masterman and Oscar L Haigh and Kirsteen M Tullett and Ingrid M Leal-Rojas and Carina Walpole and Frances E Pearson and Jonathon Cebon and Christopher Schmidt and Liam O{\textquoteright}Brien and Nikita Rosendahl and Ghazal Daraj and Irina Caminschi and Eric H Gschweng and Roger P Hollis and Donald B Kohn and Mireille H Lahoud and Kristen J Radford}, title = {Human CLEC9A antibodies deliver NY-ESO-1 antigen to CD141+ dendritic cells to activate na{\"\i}ve and memory NY-ESO-1-specific CD8+ T cells}, volume = {8}, number = {2}, elocation-id = {e000691}, year = {2020}, doi = {10.1136/jitc-2020-000691}, publisher = {BMJ Specialist Journals}, abstract = {Background Dendritic cells (DCs) are crucial for the efficacy of cancer vaccines, but current vaccines do not harness the key cDC1 subtype required for effective CD8+ T-cell-mediated tumor immune responses. Vaccine immunogenicity could be enhanced by specific delivery of immunogenic tumor antigens to CD141+ DCs, the human cDC1 equivalent. CD141+ DCs exclusively express the C-type-lectin-like receptor CLEC9A, which is important for the regulation of CD8+ T cell responses. This study developed a new vaccine that harnesses a human anti-CLEC9A antibody to specifically deliver the immunogenic tumor antigen, NY-ESO-1 (New York esophageal squamous cell carcinoma 1), to human CD141+ DCs. The ability of the CLEC9A-NY-ESO-1 antibody to activate NY-ESO-1-specific na{\"\i}ve and memory CD8+ T cells was examined and compared with a vaccine comprised of a human DEC-205-NY-ESO-1 antibody that targets all human DCs.Methods Human anti-CLEC9A, anti-DEC-205 and isotype control IgG4 antibodies were genetically fused to NY-ESO-1 polypeptide. Cross-presentation to NY-ESO-1-epitope-specific CD8+ T cells and reactivity of T cell responses in patients with melanoma were assessed by interferon γ (IFNγ) production following incubation of CD141+ DCs and patient peripheral blood mononuclear cells with targeting antibodies. Humanized mice containing human DC subsets and a repertoire of na{\"\i}ve NY-ESO-1-specific CD8+ T cells were used to investigate na{\"\i}ve T cell priming. T cell effector function was measured by expression of IFNγ, MIP-1β, tumor necrosis factor and CD107a and by lysis of target tumor cells.Results CLEC9A-NY-ESO-1 antibodies (Abs) were effective at mediating delivery and cross-presentation of multiple NY-ESO-1 epitopes by CD141+ DCs for activation of NY-ESO-1-specific CD8+ T cells. When benchmarked to NY-ESO-1 conjugated to an untargeted control antibody or to anti-human DEC-205, CLEC9A-NY-ESO-1 was superior at ex vivo reactivation of NY-ESO-1-specific T cell responses in patients with melanoma. Moreover, CLEC9A-NY-ESO-1 induced priming of na{\"\i}ve NY-ESO-1-specific CD8+ T cells with polyclonal effector function and potent tumor killing capacity in vitro.Conclusions These data advocate human CLEC9A-NY-ESO-1 Ab as an attractive strategy for specific targeting of CD141+ DCs to enhance tumor immunogenicity in NY-ESO-1-expressing malignancies.}, URL = {https://jitc.bmj.com/content/8/2/e000691}, eprint = {https://jitc.bmj.com/content/8/2/e000691.full.pdf}, journal = {Journal for ImmunoTherapy of Cancer} }