期刊论文详细信息
Biomarker Research
Integration of tumor inflammation, cell proliferation, and traditional biomarkers improves prediction of immunotherapy resistance and response
Vincent Giamo1  Sarabjot Pabla1  Shuang Gao1  Felicia L. Lenzo1  Paul DePietro1  Jonathan Andreas1  Erik Van Roey1  Shengle Zhang1  Blake Burgher1  R. J. Seager1  Carrie Hoefer1  Mary K. Nesline1  Yirong Wang1  Margot Schoenborn1  Roger Klein1  Jeffrey M. Conroy2  Sean T. Glenn2 
[1] OmniSeq, Inc, 700 Ellicott Street, 14203, Buffalo, NY, USA;OmniSeq, Inc, 700 Ellicott Street, 14203, Buffalo, NY, USA;Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, 14206, Buffalo, NY, USA;
关键词: Inflammation;    Cell proliferation;    Pembrolizumab;    Nivolumab;    Ipilimumab;    Algorithmic analysis;    Inflamed;    Borderline;    Non-inflamed;   
DOI  :  10.1186/s40364-021-00308-6
来源: Springer
PDF
【 摘 要 】

BackgroundContemporary to the rapidly evolving landscape of cancer immunotherapy is the equally changing understanding of immune tumor microenvironments (TMEs) which is crucial to the success of these therapies. Their reliance on a robust host immune response necessitates clinical grade measurements of immune TMEs at diagnosis. In this study, we describe a stable tumor immunogenic profile describing immune TMEs in multiple tumor types with ability to predict clinical benefit from immune checkpoint inhibitors (ICIs).MethodsA tumor immunogenic signature (TIGS) was derived from targeted RNA-sequencing (RNA-seq) and gene expression analysis of 1323 clinical solid tumor cases spanning 35 histologies using unsupervised analysis. TIGS correlation with ICI response and survival was assessed in a retrospective cohort of NSCLC, melanoma and RCC tumor blocks, alone and combined with TMB, PD-L1 IHC and cell proliferation biomarkers.ResultsUnsupervised clustering of RNA-seq profiles uncovered a 161 gene signature where T cell and B cell activation, IFNg, chemokine, cytokine and interleukin pathways are over-represented. Mean expression of these genes produced three distinct TIGS score categories: strong (n = 384/1323; 29.02%), moderate (n = 354/1323; 26.76%), and weak (n = 585/1323; 44.22%). Strong TIGS tumors presented an improved ICI response rate of 37% (30/81); with highest response rate advantage occurring in NSCLC (ORR = 36.6%; 16/44; p = 0.051). Similarly, overall survival for strong TIGS tumors trended upward (median = 25 months; p = 0.19). Integrating the TIGS score categories with neoplastic influence quantified via cell proliferation showed highly proliferative and strong TIGS tumors correlate with significantly higher ICI ORR than poorly proliferative and weak TIGS tumors [14.28%; p = 0.0006]. Importantly, we noted that strong TIGS and highly [median = not achieved; p = 0.025] or moderately [median = 16.2 months; p = 0.025] proliferative tumors had significantly better survival compared to weak TIGS, highly proliferative tumors [median = 7.03 months]. Importantly, TIGS discriminates subpopulations of potential ICI responders that were considered negative for response by TMB and PD-L1.ConclusionsTIGS is a comprehensive and informative measurement of immune TME that effectively characterizes host immune response to ICIs in multiple tumors. The results indicate that when combined with PD-L1, TMB and cell proliferation, TIGS provides greater context of both immune and neoplastic influences on the TME for implementation into clinical practice.

【 授权许可】

CC BY   

【 预 览 】
附件列表
Files Size Format View
RO202108115502836ZK.pdf 2225KB PDF download
  文献评价指标  
  下载次数:2次 浏览次数:2次