Abstract

Abstract The response rate of 15-20% with anti-PD1 in head and neck squamous cell carcinoma (HNSCC) highlights the urgent need for strategies to overcome resistance. Our lab has previously developed a carcinogen-induced immunocompetent murine oral carcinoma (MOC) model to study HNSCC immunobiology. Specifically, MOC1 is an immunogenic cell line that, despite sensitivity to anti-PD-1, exhibits occasional development of escape tumors (MOC1esc). MOC1esc escape tumors display a resistance phenotype similar to those observed in HNSCC patients undergoing anti-PD1 therapy. When independent escape tumors are harvested and re-transplanted into naïve mice, they grow progressively and are resistant to anti-PD1 therapy. Intriguingly, whileMOC1esc is resistant to anti-PD1, it is completely rejected in tumor bearing mice treated with anti-CTLA4. Therefore, the anti-PD1 responsive MOC1 and resistant MOC1esc mouse model is an isogenic system that provides an excellent opportunity to study mechanism(s) in adaptive resistance to anti-PD1 therapy of HNSCC. To gain a comprehensive insight into the tumor microenvironment (TME) as a contributor to adaptive resistance, we analyzed tumor infiltrating lymphocytes (TIL) in naïve MOC1 and MOC1esc tumors using mass cytometry time-of-flight (CyTOF) with a 38-cell marker panel.MOC1esc tumors were highly infiltrated by CD103+ effector/memory regulatory T cells (Tregs) and M2-like tumor associated macrophages (TAMs), while MOC1 tumors have more M1-like TAMs and neutrophils. Furthermore, we observed that both anti-PD1 and anti-CTLA4 treatment dramatically expanded CD8+ T cells and decreased neutrophils in MOC1esc tumors. In responding MOC1esc tumors, anti-CTLA4 treatment resulted in depleted Tregs, decreased M2-like TAMs and neutrophils, as well as a striking increase in M1-like TAMs compared with isotype control treated tumors. In contrast, anti-PD1 treated resistant MOC1esc tumors showed decreased M1-like TAMs, while M2-like TAMs were increased compared with controls. Tregs were not affected by anti-PD1 treatment. Therefore, the comparison between the TME of anti-PD1 treated resistant tumors and anti-CTLA4 treated responding tumors suggeststhat Tregs, neutrophils, and TAMs may contribute to the sensitivity (or resistance) to checkpoint blockade therapy. Ongoing studies will test the functional contribution of these distinct TME components in antitumor immunity including cytokine production, proliferative capacity, and their roles in immunotherapy resistance. In summary, this study identified immune modulators within TME involved in adaptive immunotherapy resistance of HNSCC. Findings from this study have advanced our understanding of HNSCC immunotherapy resistance and will accelerate the discovery of new therapeutic targets and biomarkers for adaptive resistance. Citation Format: Liye Zhou, Yasutaka Nakahori, Fei Guo, Joshua Keegan, Rachel Riley, James A. Lederer, Ravindra Uppaluri. Identification of immunotherapy resistance mechanisms within tumor microenvironment in a mouse model of oral squamous cell carcinoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 1509.

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