4.7 Article

Effect of neoadjuvant chemotherapy on the immune microenvironment in gastric cancer as determined by multiplex immunofluorescence and T cell receptor repertoire analysis

期刊

出版社

BMJ PUBLISHING GROUP
DOI: 10.1136/jitc-2021-003984

关键词

gastrointestinal neoplasms; tumor microenvironment; tumor biomarkers

资金

  1. third round of public welfare development and reform pilot projects of Beijing Municipal Medical Research Institutes (Beijing Medical Research Institute) [2019-1]
  2. National Natural Science Foundation of China [U20A20371, 81872502, 81802471, 81972758, 82073312, 32000468]
  3. National High Technology Research and Development Program of China (863 Program) [2014AA020603]
  4. 'Double First Class' disciplinary development Foundation of Peking University [BMU2019LCKXJ011]
  5. Beijing municipal administration of hospitals' youth programme [QML20181102]
  6. Beijing Municipal Administration of Hospitals Incubating Program [PX2019040]
  7. Research Fund for Young Scholars of Beijing [2018000021469G265]
  8. Clinical Medicine Plus X--Young Scholars Project
  9. Peking University (the Fundamental Research Funds for the Central Universities) [PKU2020LCXQ001, PKU2021LCXQ022]
  10. Science Foundation of Peking University Cancer Hospital [2020-6, 2020-22]
  11. Capital's funds for health improvement and research [2018-2-1023]

向作者/读者索取更多资源

The study provides insights into the immune modulation effect of chemotherapy in gastric cancer, suggesting that the immune profile of specimens after standard chemotherapy should be considered for personalized immunotherapy to improve clinical outcomes.
Background The combination of immune checkpoint blockade and chemotherapy has revolutionized the treatment of advanced gastric cancer (GC). It is crucial to unravel chemotherapy-induced tumor microenvironment (TME) modulation and identify which immunotherapy would improve antitumor effect. Methods In this study, tumor-associated immune cells (TAICs) infiltration in residual tumor after neoadjuvant chemotherapy (NAC) together with 1075 cases of treatment-naive GC patients was analyzed first. Then we performed multiplex fluorescence staining of a panel of immune markers (CD3, CD4, CD8, FOXP3 and PDL1) and T cell receptor beta-chain sequencing to phenotype and enumerate T cell subpopulations and clonal expansion in paired GC samples (prechemotherapy and postchemotherapy) from another cohort of 30 cases of stage II/III GC patients. Results Infiltration of CD68(+) macrophages in residual tumors after NAC was significantly decreased compared with treatment-naive GC patients, while no significant difference observed with respect to other immune markers. In residual tumors, post-NAC CD8 +T cells and CD68+ macrophages levels were significantly associated with chemotherapy response. Post-NAC CD8+ T cell levels remained as an independent predictor for favorable prognosis. Furthermore, when comparing the paired samples before and after NAC from 30 cases of stage II/III GC patients, we found FOXP3+ regulatory T cells proportion significantly decreased after chemotherapy. Pre-NAC FOXP3+ T reg cells level was much richer in the response group and decreased more significantly in the stromal compartment. CD8+ cytotoxic T lymphocytes levels were elevated after chemotherapy, which was more significant in the group treated with XELOX regimen and in patients with better response, consistent with the TCR diversity elevation. Conclusions These findings have deepened our understanding of the immune modulating effect of chemotherapy and suggest that the immune profile of specimens after standard chemotherapy should be considered for the personalized immunotherapy to ultimately improve clinical outcome in patients with GC.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据