@article {Prokopie000832, author = {Anastasia Prokopi and Christoph H Tripp and Bart Tummers and Florian Hornsteiner and Sarah Spoeck and Jeremy Chase Crawford and Derek R Clements and Mirjana Efremova and Katharina Hutter and Lydia Bellmann and Giuseppe Cappellano and Bruno L Cadilha and Sebastian Kobold and Louis Boon and Daniela Ortner and Zlatko Trajanoski and Suzie Chen and Tanja D de Gruijl and Juliana Idoyaga and Douglas R Green and Patrizia Stoitzner}, title = {Skin dendritic cells in melanoma are key for successful checkpoint blockade therapy}, volume = {9}, number = {1}, elocation-id = {e000832}, year = {2021}, doi = {10.1136/jitc-2020-000832}, publisher = {BMJ Specialist Journals}, abstract = {Background Immunotherapy with checkpoint inhibitors has shown impressive results in patients with melanoma, but still many do not benefit from this line of treatment. A lack of tumor-infiltrating T cells is a common reason for therapy failure but also a loss of intratumoral dendritic cells (DCs) has been described.Methods We used the transgenic tg(Grm1)EPv melanoma mouse strain that develops spontaneous, slow-growing tumors to perform immunological analysis during tumor progression. With flow cytometry, the frequencies of DCs and T cells at different tumor stages and the expression of the inhibitory molecules programmed cell death protein-1 (PD-1) and T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) on T cells were analyzed. This was complemented with RNA-sequencing (RNA-seq) and real-time quantitative PCR (RT-qPCR) analysis to investigate the immune status of the tumors. To boost DC numbers and function, we administered Fms-related tyrosine 3 ligand (Flt3L) plus an adjuvant mix of polyI:C and anti-CD40. To enhance T cell function, we tested several checkpoint blockade antibodies. Immunological alterations were characterized in tumor and tumor-draining lymph nodes (LNs) by flow cytometry, CyTOF, microarray and RT-qPCR to understand how immune cells can control tumor growth. The specific role of migratory skin DCs was investigated by coculture of sorted DC subsets with melanoma-specific CD8+ T cells.Results Our study revealed that tumor progression is characterized by upregulation of checkpoint molecules and a gradual loss of the dermal conventional DC (cDC) 2 subset. Monotherapy with checkpoint blockade could not restore antitumor immunity, whereas boosting DC numbers and activation increased tumor immunogenicity. This was reflected by higher numbers of activated cDC1 and cDC2 as well as CD4+ and CD8+ T cells in treated tumors. At the same time, the DC boost approach reinforced migratory dermal DC subsets to prime gp100-specific CD8+ T cells in tumor-draining LNs that expressed PD-1/TIM-3 and produced interferon γ (IFNγ)/tumor necrosis factor α (TNFα). As a consequence, the combination of the DC boost with antibodies against PD-1 and TIM-3 released the brake from T cells, leading to improved function within the tumors and delayed tumor growth.Conclusions Our results set forth the importance of skin DC in cancer immunotherapy, and demonstrates that restoring DC function is key to enhancing tumor immunogenicity and subsequently responsiveness to checkpoint blockade therapy.}, URL = {https://jitc.bmj.com/content/9/1/e000832}, eprint = {https://jitc.bmj.com/content/9/1/e000832.full.pdf}, journal = {Journal for ImmunoTherapy of Cancer} }