TY - JOUR T1 - Immunogenomic determinants of tumor microenvironment correlate with superior survival in high-risk neuroblastoma JF - Journal for ImmunoTherapy of Cancer JO - J Immunother Cancer DO - 10.1136/jitc-2021-002417 VL - 9 IS - 7 SP - e002417 AU - Riyue Bao AU - Stefani Spranger AU - Kyle Hernandez AU - Yuanyuan Zha AU - Peter Pytel AU - Jason J Luke AU - Thomas F Gajewski AU - Samuel L Volchenboum AU - Susan L Cohn AU - Ami V Desai Y1 - 2021/07/01 UR - http://jitc.bmj.com/content/9/7/e002417.abstract N2 - Background Tumor-infiltrating CD8+ T cells and neoantigens are predictors of a favorable prognosis and response to immunotherapy with checkpoint inhibitors in many types of adult cancer, but little is known about their role in pediatric malignancies. Here, we analyzed the prognostic strength of T cell-inflamed gene expression and neoantigen load in high-risk neuroblastoma. We also compared transcriptional programs in T cell-inflamed and non-T cell-inflamed high-risk neuroblastomas to investigate possible mechanisms of immune exclusion.Methods A defined T cell-inflamed gene expression signature was used to categorize high-risk neuroblastomas in the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) program (n=123), and the Gabriella Miller Kids First (GMKF) program (n=48) into T cell-inflamed, non-T cell-inflamed, and intermediate groups. Associations between the T cell-inflamed and non-T cell-inflamed group, MYCN amplification, and survival were analyzed by Cox proportional hazards models. Additional survival analysis was conducted after integrating neoantigen load predicted from somatic mutations. Pathways activated in non-T cell-inflamed relative to T cell-inflamed tumors were analyzed using causal network analysis.Results Patients with T cell-inflamed high-risk tumors showed improved overall survival compared with those with non-T cell-inflamed tumors (p<0.05), independent of MYCN amplification status, in both TARGET and GMKF cohorts. Higher neoantigen load was also associated with better event-free and overall survival (p<0.005) and was independent of the T cell-inflamed signature. Activation of MYCN, ASCL1, SOX11, and KMT2A transcriptional programs was inversely correlated with the T cell-inflamed signature in both cohorts.Conclusions Our results indicate that tumors from children with high-risk neuroblastoma harboring a strong T cell-inflamed signature have a more favorable clinical outcome, and neoantigen load is a prognosis predictor, independent of T cell inflammation. Strategies to target SOX11 and other signaling pathways associated with non-T cell-inflamed tumors should be pursued as potential immune-potentiating interventions.All data relevant to the study are included in the article or uploaded as supplementary information. The gene expression, somatic mutations, and clinical data were downloaded from GDC data portal (https://portal.gdc.cancer.gov), and GMKF data portal (https://portal.kidsfirstdrc.org/dashboard). Patient-level clinical data of the GMKF cohort are under controlled access at INRG Data Commons and can be requested by contacting the INRG Review Committee (https://inrgdb.org). IHC image data files were deposited on a publicly accessible GitHub Repository (https://github.com/riyuebao/NBL-TME-Immunogenomics). Other data will be provided upon request from the corresponding author. ER -