4.8 Article

B Cells Are Required to Generate Optimal Anti-Melanoma Immunity in Response to Checkpoint Blockade

期刊

FRONTIERS IN IMMUNOLOGY
卷 13, 期 -, 页码 -

出版社

FRONTIERS MEDIA SA
DOI: 10.3389/fimmu.2022.794684

关键词

knockout (KO); programmed death-ligand 1 (PD-L1); cytotoxic T-lymphocyte-associated protein 4 (CTLA-4); the Cancer Genome Atlas melanoma (TCGA); immunotherapy; Blymphocytes; chemokineCXCL13; toll-like receptor 7; 8

资金

  1. Department of Defense Idea Award [CA160521]
  2. University of Texas MD Anderson Cancer Center Specialized Programs of Research Excellence in Melanoma [P50CA093459]
  3. National Institutes of Health/National Cancer Institute [P30CA016672]

向作者/读者索取更多资源

In this study, the role of B cells in anti-melanoma immunity was investigated. The researchers found that B cells were required for the effectiveness of PD-L1 antibody monotherapy and combination therapy with PD-L1 antibody and TLR-7/8 agonist. However, B cells were not essential for anti-CTLA-4 antibody activity. The presence of B cell chemokine CXCL13 and B cells together with CD8(+) T cells in tumors were strongly associated with improved overall survival in melanoma patients.
Immunotherapies such as checkpoint blockade therapies are known to enhance anti-melanoma CD8(+) T cell immunity, but only a fraction of patients treated with these therapies achieve durable immune response and disease control. It may be that CD8(+) T cells need help from other immune cells to generate effective and long-lasting anti-tumor immunity or that CD8(+) T cells alone are insufficient for complete tumor regression and cure. Melanoma contains significant numbers of B cells; however, the role of B cells in anti-melanoma immunity is controversial. In this study, B16 melanoma mouse models were used to determine the role of B cells in anti-melanoma immunity. C57BL/6 mice, B cell knockout (KO) C57BL/6 mice, anti-CD19, and anti-CXCL13 antibody-treated C57BL/6 mice were used to determine treatment efficacy and generation of tumor-specific CD8(+) T cells in response to PD-L1 blockade alone or combination with TLR-7/8 activation. Whole transcriptome analysis was performed on the tumors from B cell depleted and WT mice, untreated or treated with anti-PD-L1. Both CD40-positive and CD40-negative B cells were isolated from tumors of TLR-7/8 agonist-treated wild-type mice and adoptively transferred into tumor-bearing B cell KO mice, which were treated with anti-PD-L1 and TLR-7/8 agonist. Therapeutic efficacy was determined in the presence of activated or inactivated B cells. Microarray analysis was performed on TLR-7/8-treated tumors to look for the B cell signatures. We found B cells were required to enhance the therapeutic efficacy of monotherapy with anti-PD-L1 antibody and combination therapy with anti-PD-L1 antibody plus TLR-7/8 agonist. However, B cells were not essential for anti-CTLA-4 antibody activity. Interestingly, CD40-positive but not CD40-negative B cells contributed to anti-melanoma immunity. In addition, melanoma patients' TCGA data showed that the presence of B cell chemokine CXCL13 and B cells together with CD8(+) T cells in tumors were strongly associated with improved overall survival. Our transcriptome data suggest that the absence of B cells enhances immune checkpoints expression in the tumors microenvironment. These results revealed the importance of B cells in the generation of effective anti-melanoma immunity in response to PD-1-PD-L1 blockade immunotherapy. Our findings may facilitate the design of more effective anti-melanoma immunotherapy.

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

4.8
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