Abstract

Abstract Anti-PD-1 therapy is an important new treatment option for many different types of malignancies, but overall response rates are less than 40%. Limited understanding of how anti-PD-1 treatment changes the tumor immune microenvironment is a barrier to identifying rational combination therapies and understanding mechanisms of immunotherapy resistance. To overcome this barrier, we set out to understand the mechanisms by which anti-PD-1 therapy augments the anti-tumor immune response using single-cell genomics. We have developed a massively parallel single-cell RNA-sequencing platform (“Seq-Well”) that uses a fabricated chip with nearly 100,000 nanowells into which barcoded beads and individual cells are distributed prior to lysis and RNA capture. We used this platform to comprehensively define the global expression profile of all major immune lineages in the tumor microenvironment in a mouse tumor model of immunotherapy. Mice were implanted with the B16F10 melanoma transplantable model of cancer and treated with anti-PD-1 or control antibodies. Tumors were harvested and CD45+ tumor-infiltrating leukocytes isolated by FACS. In a single experiment we were able to sequence the transcriptomes of over 600 individual cells, allowing us to clearly distinguish different immune lineages within the tumor microenvironment. We detect two transcriptionally distinct populations of CD8+ T cells, one that is highly proliferative (as marked by Ki-67), and one that has higher expression of cytotoxic markers (i.e. perforin). The Ki-67hi population is enriched for a gene expression signature from terminally exhausted CD8+ T cells in chronic viral infection, suggesting that this is a more exhausted subset. Treatment with anti-PD-1 globally alters the tumor microenvironment, including enriching for CD8+ T cells in the Prfhi subpopulation compared with the Ki-67hi more terminally exhausted population. Studies to understand changes in the immune infiltrate of immunotherapy resistant tumors are currently ongoing. In conclusion, massively parallel single-cell RNA-sequencing allows us to dissect the mechanisms by which checkpoint blockade controls tumor growth, revealing shifts in the differentiation state of exhausted CD8+ T cells induced by checkpoint blockade. Citation Format: Brian C. Miller, Marc H. Wadsworth, Kevin Bi, Travis K. Hughes, Robert Manguso, Arlene H. Sharpe, Alex K. Shalek, Nicholas Haining. Dissecting mechanisms of anti-PD-1 therapy with massively parallel single-cell RNA-sequencing [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3027. doi:10.1158/1538-7445.AM2017-3027

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call