PD-L1, programmed death ligand1

  • 文章类型: Journal Article
    由于无法治愈的去势抵抗性前列腺癌(CRPC)在雄激素剥夺疗法(ADT)治疗后最终发展,制定新的治疗策略来治疗CRPC至关重要。靶向程序性细胞死亡蛋白1(PD-1)和程序性死亡配体-1(PD-L1)的治疗已被批准用于具有临床益处的人类癌症。然而,许多患者,尤其是前列腺癌,抗PD-1/PD-L1治疗无效,因此,迫切需要寻求一种支持策略来改进传统的PD-1/PD-L1靶向免疫治疗。在本研究中,分析前列腺癌组织微阵列的数据,我们发现PD-L1的表达与异质核核糖核蛋白L(HnRNPL)的表达呈正相关。因此,我们进一步研究了HnRNPL对PD-L1表达的潜在作用,在CRPC中,癌细胞对T细胞杀伤的敏感性以及与抗PD-1治疗的协同作用。的确,HnRNPL敲低可有效降低PD-L1的表达,并在体外和体内恢复癌细胞对T细胞杀伤的敏感性,相反,HnRNPL过表达在CRPC细胞中导致相反的作用。此外,与以前的研究一致,我们发现铁性凋亡在T细胞诱导的癌细胞死亡中起关键作用,和HnRNPL通过靶向YY1/PD-L1轴和抑制CRPC细胞的铁凋亡部分促进了癌症的免疫逃逸。此外,HnRNPL敲低通过招募浸润性CD8+T细胞增强抗肿瘤免疫力,并在CRPC肿瘤中与抗PD-1治疗协同作用。这项研究提供了生物学证据,表明HnRNPL敲低可能是PD-L1/PD-1阻断策略中增强CRPC抗肿瘤免疫应答的新型治疗剂。
    Owing to incurable castration-resistant prostate cancer (CRPC) ultimately developing after treating with androgen deprivation therapy (ADT), it is vital to devise new therapeutic strategies to treat CRPC. Treatments that target programmed cell death protein 1 (PD-1) and programmed death ligand-1 (PD-L1) have been approved for human cancers with clinical benefit. However, many patients, especially prostate cancer, fail to respond to anti-PD-1/PD-L1 treatment, so it is an urgent need to seek a support strategy for improving the traditional PD-1/PD-L1 targeting immunotherapy. In the present study, analyzing the data from our prostate cancer tissue microarray, we found that PD-L1 expression was positively correlated with the expression of heterogeneous nuclear ribonucleoprotein L (HnRNP L). Hence, we further investigated the potential role of HnRNP L on the PD-L1 expression, the sensitivity of cancer cells to T-cell killing and the synergistic effect with anti-PD-1 therapy in CRPC. Indeed, HnRNP L knockdown effectively decreased PD-L1 expression and recovered the sensitivity of cancer cells to T-cell killing in vitro and in vivo, on the contrary, HnRNP L overexpression led to the opposite effect in CRPC cells. In addition, consistent with the previous study, we revealed that ferroptosis played a critical role in T-cell-induced cancer cell death, and HnRNP L promoted the cancer immune escape partly through targeting YY1/PD-L1 axis and inhibiting ferroptosis in CRPC cells. Furthermore, HnRNP L knockdown enhanced antitumor immunity by recruiting infiltrating CD8+ T cells and synergized with anti-PD-1 therapy in CRPC tumors. This study provided biological evidence that HnRNP L knockdown might be a novel therapeutic agent in PD-L1/PD-1 blockade strategy that enhanced anti-tumor immune response in CRPC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号