Wang Minyu, Qin Lei, Thia Kevin, Nguyen Thu, MacDonald Sean, Belobrov Simone, Kranz Sevastjan, Goode David, Trapani Joseph A, Wiesenfeld David, Neeson Paul Joseph
Cancer Immunology Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.
J Immunother Cancer. 2024 Oct 2;12(10):e009617. doi: 10.1136/jitc-2024-009617.
Locally advanced oral cavity squamous cell carcinoma (OCSCC) presents a significant clinical challenge despite being partially responsive to standard treatment modalities. This study investigates the prognostic implications of programmed death-ligand 1 (PD-L1) expression in these tumors, focusing on its association with treatment outcomes and the immune microenvironment.
We assessed tumor-infiltrating lymphocytes (TILs) in 132 patients with OCSCC to evaluate their impact on survival. Multiplex immunohistochemistry staining for CD3, CD68, CD11c, PD-L1, and P40 was used to explore correlations with clinical outcomes in patients with early-stage (n=22) and locally advanced (n=36) OCSCC. These initial findings were validated through differential gene expression analysis, gene set enrichment, and immune cell deconvolution in a The Cancer Genome Atlas cohort of 163 locally advanced OCSCC tumors. Additionally, single-cell RNA sequencing (scRNA-seq) on a smaller cohort (n=10) further characterized the PD-L1 or PD-L1 cancer cells in these tumors.
Elevated PD-L1 expression was associated with poor outcomes in patients with locally advanced OCSCC undergoing standard adjuvant therapy, irrespective of "hot" or "cold" classification based on TILs assessment. PD-L1 tumors exhibited an active immune response phenotype, enriched with M1 macrophages, CD8 T cells and T regulatory cells in the tumor microenvironment. Notably, the negative impact of PD-L1 expression on outcomes was primarily attributed to its expression by cancer cells, rather than immune cells. Furthermore, scRNA-seq revealed that immune interactions were not essential for PD-L1 upregulation in cancer cells, instead, complex regulatory networks were involved. Additionally, PD-L1 locally advanced tumors exhibited more complex pathway enrichment and diverse T-cell populations compared with those in the early-stage.
Our findings underscore the prognostic significance of PD-L1 expression in locally advanced OCSCC, and unveil the complex interplay between PD-L1 expression, immune responses, and molecular pathways in the tumor microenvironment. This study provides insights that may inform future therapeutic strategies, including the possibility of tailored immunotherapeutic approaches for patients with PD-L1 locally advanced OCSCC.
局部晚期口腔鳞状细胞癌(OCSCC)尽管对标准治疗方式有部分反应,但仍带来重大临床挑战。本研究调查程序性死亡配体1(PD-L1)在这些肿瘤中的表达对预后的影响,重点关注其与治疗结果及免疫微环境的关联。
我们评估了132例OCSCC患者的肿瘤浸润淋巴细胞(TILs),以评估它们对生存的影响。采用针对CD3、CD68、CD11c、PD-L1和P40的多重免疫组化染色,探讨早期(n = 22)和局部晚期(n = 36)OCSCC患者与临床结果的相关性。这些初步发现通过对163例局部晚期OCSCC肿瘤的癌症基因组图谱队列进行差异基因表达分析、基因集富集分析和免疫细胞反卷积分析得到验证。此外,对一个较小队列(n = 10)进行单细胞RNA测序(scRNA-seq),进一步表征了这些肿瘤中PD-L1或PD-L1癌细胞的特征。
在接受标准辅助治疗的局部晚期OCSCC患者中,无论基于TILs评估的“热”或“冷”分类如何,PD-L1表达升高均与不良预后相关。PD-L1肿瘤表现出活跃的免疫反应表型,在肿瘤微环境中富含M1巨噬细胞、CD8 T细胞和调节性T细胞。值得注意的是,PD-L1表达对结果的负面影响主要归因于癌细胞而非免疫细胞的表达。此外,scRNA-seq显示免疫相互作用对癌细胞中PD-L1上调并非必不可少,相反,涉及复杂的调控网络。此外,与早期肿瘤相比,PD-L1局部晚期肿瘤表现出更复杂的通路富集和多样的T细胞群体。
我们的研究结果强调了PD-L1表达在局部晚期OCSCC中的预后意义,并揭示了PD-L1表达、免疫反应和肿瘤微环境中分子通路之间的复杂相互作用。本研究提供的见解可能为未来的治疗策略提供参考,包括为PD-L1局部晚期OCSCC患者量身定制免疫治疗方法的可能性。