Programmed death ligand 1 (PD-L1)

程序性死亡配体 1 (pd - l1)
  • 文章类型: Editorial
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    程序性死亡配体1(PD-L1)的过度表达与头颈部鳞状细胞癌(HNSCC)患者对抗癌疗法的抗性和不良预后有关。尼妥珠单抗,人源化抗表皮生长因子受体(EGFR)单克隆抗体,临床上已广泛用于治疗多种实体瘤。然而,其抗癌作用是否涉及PD-L1表达的降低尚不清楚.本研究旨在研究尼妥珠单抗在体外和体内对HNSCC中PD-L1表达的调节作用和潜在机制。体外,尼妥珠单抗在HNSCC细胞系中在转录和蛋白质水平上抑制IFN-γ诱导的PD-L1上调。随后的机制研究表明,尼妥珠单抗主要通过抑制EGFR/MEK/ERK通路的磷酸化来抑制IFN-γ刺激的PD-L1的上调,MEK和ERK抑制剂进一步验证。在HNSCC荷瘤模型中,尼妥珠单抗在肿瘤进展或化疗期间显著降低PD-L1表达,这种减少伴随着肿瘤对多西他赛和阿替珠单抗的敏感性增加.此外,尼妥珠单抗与紫杉醇+顺铂(TP)诱导化疗方案联合治疗后可逆转PD-L1上调,并改善HNSCC患者的CD4+和CD8+T细胞浸润。这些发现为尼妥珠单抗在HNSCC中的抗癌机制提供了新的见解。
    The overexpression of programmed death ligand 1 (PD-L1) is associated with resistance to anticancer therapies and poor prognosis in patients with head and neck squamous cell carcinoma (HNSCC). Nimotuzumab, a humanized anti-epidermal growth factor receptor (EGFR) mAb, has been widely used clinically for treating several solid tumors. However, whether its anticancer effect involves a reduction in PD-L1 expression remains unclear. The current study aimed to investigate the regulatory effects and underlying mechanism of nimotuzumab on PD-L1 expression in HNSCC both in vitro and in vivo. In vitro, nimotuzumab inhibited IFN-γ-induced PD-L1 upregulation at both the transcriptional and protein levels in the HNSCC cell lines. Subsequent mechanism research revealed that nimotuzumab suppressed IFN-γ-stimulated PD-L1 upregulation mainly by inhibiting phosphorylation of EGFR/MEK/ERK pathway, which was further validated by MEK and ERK inhibitors. In a HNSCC tumor-bearing model, nimotuzumab significantly decreased PD-L1 expression during tumor progression or chemotherapy, and this reduction was accompanied by increased sensitivity of the tumor to docetaxel and atezolizumab. Additionally, nimotuzumab reversed PD-L1 upregulation when combined with Taxol + Cisplatin (TP) induction chemotherapy regimens and improved the CD4+ and CD8+ T cells infiltration in HNSCC patients. These findings provide new insights into the anticancer mechanisms of nimotuzumab in HNSCC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    肝细胞癌(HCC)是一种常见的恶性肿瘤,其特征是术后复发率极高,严重损害HCC患者的预后。然而,目前仍缺乏标准的术后治疗方案.尽管在一项新发表的针对血管内皮生长因子(VEGF)和程序性死亡配体1(抗PD-L1)的术后治疗的临床试验中获得了令人鼓舞的结果,其功效仍然受到限制。将止血水凝胶与基于纳米颗粒的药物递送系统组合提供了优化抗肿瘤效果的机会。在这里,我们开发了一个纳米平台,称为HMSN@Sor/aP@凝胶,包含止血纤维蛋白水凝胶和负载索拉非尼和抗PD-L1的官能化中空介孔二氧化硅纳米颗粒(HMSN),用于局部施用靶向免疫疗法以预防HCC的术后复发和转移。抗肿瘤机制基于Ras/Raf/MEK/ERK(MAPK)和磷脂酰肌醇-3-激酶(PI3K)/蛋白激酶B(AKT)途径的双重抑制,协同补充PD-L1阻断。HMSN@Sor/aP@Gel促进树突状细胞成熟,增强细胞毒性T淋巴细胞浸润,促进肿瘤相关巨噬细胞向M1表型的极化,诱导肿瘤免疫原性细胞死亡,逆转免疫抑制,建立免疫记忆以对抗术后复发。动物研究证实,HMSN@Sor/aP@Gel介导的靶向免疫疗法显着阻碍原发性和转移性肿瘤的生长,并建立免疫记忆以防止术后复发。这项研究为术后治疗提供了一种有希望的策略,具有相当大的临床转化潜力。
    Hepatocellular carcinoma (HCC) is a prevalent malignancy characterized by an exceedingly high recurrence rate post-surgery, significantly impairing the prognosis of HCC patients. However, a standard in-care strategy for postoperative therapy is still lacking. Although encouraging results have been obtained in a newly published clinical trial for postoperative therapy by targeting the vascular endothelial growth factor (VEGF) and programmed death ligand 1 (anti-PD-L1), its efficacy remains constrained. Combining a hemostatic hydrogel with a nanoparticle-based drug delivery system presents an opportunity to optimize the antitumor effect. Herein, we developed a nanoplatform, termed HMSN@Sor/aP@Gel, comprising a hemostatic fibrin hydrogel and functionalized hollow mesoporous silica nanoparticles (HMSNs) loaded with sorafenib and anti-PD-L1 for locally administered targeted-immunotherapy to prevent the postoperative recurrence and metastasis of HCC. The antitumor mechanism is grounded in dual inhibition of Ras/Raf/MEK/ERK (MAPK) and phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT) pathways, synergistically complemented by PD-L1 blockade. HMSN@Sor/aP@Gel facilitates dendritic cell maturation, enhances cytotoxic T-lymphocyte infiltration, promotes the polarization of tumor-associated macrophages to M1 phenotype, induces tumor immunogenic cell death, reverses immunosuppression, and establishes immune memory to counter postoperative recurrence. Animal studies corroborate that HMSN@Sor/aP@Gel-mediated targeted immunotherapy significantly impedes primary and metastatic tumor growth and establishes immune memory to prevent recurrence post-surgery. This investigation presents a promising strategy for postoperative therapy with considerable potential for clinical translation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    程序性死亡配体1(PD-L1)评分是预测免疫疗法反应的重要伴随诊断。免疫组织化学可以准确评估常规石蜡包埋组织中PD-L1的表达。然而,脱钙或脱色素组织是否仍然准确,可以作为伴随诊断存在争议。本研究试图通过分析不同时间脱钙和脱色对PD-L1表达的影响来解决这一争议。
    选择胎盘组织进行组织微阵列,根据6、12、24、36和48小时的时间梯度进行脱钙,根据1、5、15、30和60分钟的时间梯度进行脱色。观察并定量不同时间点的PD-L1表达强度。选择10例PD-L1阳性食管鳞癌标本进行脱钙治疗,还有PD-L1.综合积极评分(CPS),比较脱钙前后肿瘤比例评分(TPS)和免疫细胞比例评分(IPS)及阳性率。
    胎盘脱钙后,PD-L1阳性强度减弱,平均光密度(AOD)值随脱钙时间的延长而降低,24h时与对照组相比显著降低(P<0.05),36、48h与对照组比较差异有统计学意义(P<0.01)。高锰酸钾脱色后,PD-L1阳性强度明显减弱。此外,脱色时间达到5min后AOD值较对照组明显降低(P<0.01)。10例PD-L1阳性食管鳞癌24h脱钙治疗,虽然PD-L1评分有一定程度的下降(P>0.05),阳性率可达90%。治疗36小时后,PD-L1评分下降,CPS和IPS评分显著降低(P<0.05),阳性率仅为50%。
    高锰酸钾脱色显著降低PD-L1表达,即使在更短的时间内,影响结果的准确性。PD-L1在24h脱钙内保持较高的准确性。以上结果对临床选择免疫治疗具有一定的参考价值。
    UNASSIGNED: Programmed death ligand 1 (PD-L1) score is an important companion diagnosis to predict the response to immunotherapy. Immunohistochemistry can accurately assess the expression of PD-L1 in routine paraffin-embedded tissue. However, whether decalcified or depigmented tissue is still accurate and can be used as a companion diagnosis is controversial. This study attempts to resolve this controversy by analyzing the effects of decalcification and depigmentation at different times on PD-L1 expression.
    UNASSIGNED: Placental tissues were selected for tissue microarray, decalcification was performed according to time gradients of 6, 12, 24, 36, and 48 h, and depigmentation was performed according to time gradients of 1, 5, 15, 30, and 60 min. The intensity of PD-L1 expression at different time points was observed and quantified. Ten PD-L1-positive esophageal squamous carcinoma samples were selected for decalcification treatment, and the PD-L1. Combined Positive Score (CPS), Tumor Proportion Score (TPS) and Immunocyte Proportion Score (IPS) and the positivity rates were compared before and after decalcification.
    UNASSIGNED: After the placenta was decalcified, the intensity of PD-L1 positivity diminished, and the average optical density (AOD) value decreased with the prolongation of decalcification time and decreased significantly (P<0.05) at 24 h compared with the control group, and significantly (P<0.01) at 36 and 48 h compared with the control group. The intensity of PD-L1 positivity was weakened considerably after the treatment with potassium permanganate depigmentation. In addition, the AOD value decreased significantly (P<0.01) after the depigmentation time reached 5 min compared with the control group. Ten cases of PD-L1 positive esophageal squamous carcinoma were treated with 24 h decalcification, although the PD-L1 score decreased to a certain degree (P>0.05), and the positivity rate could reach 90%. After 36 h treatment, PD-L1 scores decreased, the CPS and IPS scores decreased significantly (P<0.05), and the positive rate was only 50%.
    UNASSIGNED: Potassium permanganate depigmentation significantly reduces PD-L1 expression, even for a shorter time, affecting the accuracy of the results. The accuracy of PD-L1 remained high within 24 h decalcification. The above results have certain reference value for clinical selection of immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    本研究探讨了脂肪间充质干细胞(AdMSCs)中程序性死亡配体1(PD-L1)和蛋白激酶B(Akt)过表达对改善心肌梗死(MI)后心功能障碍的协同作用。MI后成年Wistar大鼠分为四组:假,MI,AMSC治疗,和用PD-L1和Akt(AdMSC-PDL1-Akt)处理过表达的ADMSCs。MI是通过左冠状动脉前降支结扎诱发的,然后在心肌内注射AdMSC。四个多星期,使用压力-容积分析评估心脏功能和结构完整性,梗死面积测量,和免疫组织化学。AdMSC-PDL1-Akt在体外表现出对活性氧(ROS)的抵抗力增强,并通过改善收缩压末期-容积关系和预负荷可招募的卒中工作来改善体内MI诱导的收缩功能障碍,连同衰减梗死面积。分子分析显示AdMSC-PDL1-Akt组中MI心脏中caspase3和核因子-κB上调的实质性缓解。机械上,AdMSC-PDL1-Akt促进正常T细胞分化为CD25+调节性T细胞,与AdMSC-PDL1-Akt处理的大鼠中CD25的体内上调对齐。总的来说,PD-L1和Akt在AdMSCs中的过表达增强了体外对ROS介导的凋亡的抵抗,并增强了对MI诱导的功能障碍的心肌保护功效,可能通过T细胞调制,强调心肌缺血损伤的有希望的治疗策略。
    This study explores the synergistic impact of Programmed Death Ligand 1 (PD-L1) and Protein Kinase B (Akt) overexpression in adipose-derived mesenchymal stem cells (AdMSCs) for ameliorating cardiac dysfunction after myocardial infarction (MI). Post-MI adult Wistar rats were allocated into four groups: sham, MI, ADMSC treatment, and ADMSCs overexpressed with PD-L1 and Akt (AdMSC-PDL1-Akt) treatment. MI was induced via left anterior descending coronary artery ligation, followed by intramyocardial AdMSC injections. Over four weeks, cardiac functionality and structural integrity were assessed using pressure-volume analysis, infarct size measurement, and immunohistochemistry. AdMSC-PDL1-Akt exhibited enhanced resistance to reactive oxygen species (ROS) in vitro and ameliorated MI-induced contractile dysfunction in vivo by improving the end-systolic pressure-volume relationship and preload-recruitable stroke work, together with attenuating infarct size. Molecular analyses revealed substantial mitigation in caspase3 and nuclear factor-κB upregulation in MI hearts within the AdMSC-PDL1-Akt group. Mechanistically, AdMSC-PDL1-Akt fostered the differentiation of normal T cells into CD25+ regulatory T cells in vitro, aligning with in vivo upregulation of CD25 in AdMSC-PDL1-Akt-treated rats. Collectively, PD-L1 and Akt overexpression in AdMSCs bolsters resistance to ROS-mediated apoptosis in vitro and enhances myocardial protective efficacy against MI-induced dysfunction, potentially via T-cell modulation, underscoring a promising therapeutic strategy for myocardial ischemic injuries.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:在2015年世界卫生组织分类中采用了小活检中肺癌诊断的术语。如果非小细胞肺癌(NSCLC)没有明确的腺癌(AD)或鳞状细胞癌形态,根据粘蛋白或免疫组织化学染色将肿瘤进一步分类为NSCLC偏爱AD(NFAD),NSCLC有利于鳞状细胞癌,或NSCLC未另作说明。自从这个新术语被定义以来,AD和NFAD之间的差异尚未得到充分探索。这项研究旨在检查临床背景的差异,基因改变频率,和程序性死亡配体1(PD-L1)表达。
    方法:我们纳入了诊断为AD或NFAD的小样本患者,并且在2019年8月至2023年4月期间在神奈川癌症中心接受了OncomineDx目标测试。
    结果:本研究包含268名患者。共有96例患者在诊断为AD或NFAD后接受手术治疗。NFAD的临床分期比AD更晚期,病理N0更低。手术标本的病理学显示,NFAD中实性占优势的AD明显高于AD(p<0.001)。在AD和NFAD中,EGFR突变是最常见的基因改变,其次是KRAS突变。AD中EGFR突变的频率明显高于NFAD。NFAD中PD-L1表达显著高于AD(p<0.001)。
    结论:这项研究表明,就癌症进展而言,AD和NFAD之间存在明显差异,主要肿瘤的病理特征,遗传特征,和PD-L1表达。
    BACKGROUND: The terminology for lung cancer diagnosis in small biopsies was adopted in the 2015 World Health Organization classification. If non-small cell lung cancer (NSCLC) has no clear adenocarcinoma (AD) or squamous cell carcinoma morphology, the tumor is further classified based on mucin or immunohistochemical staining as NSCLC favor AD (NFAD), NSCLC favor squamous cell carcinoma, or NSCLC not otherwise specified. Since this new term was defined, the difference between AD and NFAD has not yet been fully explored. This study aimed to examine the differences in clinical background, gene alteration frequency, and programmed death ligand 1 (PD-L1) expression.
    METHODS: We included patients diagnosed with AD or NFAD with small samples, and who underwent testing with the Oncomine Dx target test between August 2019 and April 2023 in Kanagawa Cancer Center.
    RESULTS: This study comprised 268 patients. A total of 96 patients underwent surgery after AD or NFAD diagnosis. The clinical stage was more advanced and pathological N0 was lower in NFAD than in AD. The pathology of the surgical specimens revealed that solid predominant AD was significantly more common in NFAD than in AD (p < 0.001). In both AD and NFAD, EGFR mutation was the most frequent gene alteration, followed by KRAS mutation. The frequency of EGFR mutations was significantly higher in AD than in NFAD. PD-L1 expression was significantly higher in NFAD than in AD (p < 0.001).
    CONCLUSIONS: This study shows a clear difference between AD and NFAD in terms of cancer progression, pathological features of the main tumor, genetic characteristics, and PD-L1 expression.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    在过去的十年中,免疫疗法已成为一种有前途的癌症治疗方法,IHC是最常用的PDL-1/PD1评价方法。总的来说,可以对FFPE标本和细胞学样品进行PD-L1测定。然而,它们在涂片上的使用尚未得到证实或验证。如今,数字图像和先进的算法可以帮助解释细胞学样本中的PD-L1。了解非小细胞肺癌(NSCLC)的免疫环境对于开发成功的抗癌免疫疗法至关重要。对通过微创方法获得的细胞学样品使用多重免疫荧光(mIF)测定似乎是研究NSCLC免疫环境的可行选择。这篇综述旨在简要总结细胞病理学在通过免疫细胞化学(ICC)分析PD-L1中的作用以及细胞病理学在免疫治疗环境中的未来方向的知识。
    Immunotherapy has become a promising cancer treatment in the past decade, and IHC is the most commonly used testing method for PDL-1/PD1 evaluation. In general, PD-L1 assays can be performed on both FFPE specimens and cytological samples. However, their use on smears is not yet well-established or validated. Nowadays, digital images and advanced algorithms can aid in interpreting PD-L1 in cytological samples. Understanding the immune environment of non-small cell lung cancer (NSCLC) is critical in developing successful anticancer immunotherapies. The use of a multiplexed immunofluorescence (mIF) assay on cytological samples obtained through minimally invasive methods appears to be a viable option for investigating the immune environment of NSCLC. This review aims to briefly summarize the knowledge of the role of cytopathology in the analysis of PD-L1 by immunocytochemistry (ICC) and future directions of cytopathology in the immunotherapy setting.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:本研究旨在开发一种新的基于联合免疫评分(CIS)的模型来评估三阴性乳腺癌(TNBC)的预后。
    方法:8种免疫标志物(PD-1、PD-L1、PD-L2、IDO、TIM3,OX40,OX40L,和H7-H2)通过免疫组织化学对227例TNBC病例的肿瘤细胞(TC)和免疫细胞(IC)进行了评估,分别,随后与选定的临床病理参数和生存率相关。进一步检查从癌症基因组图谱(TCGA)检索的数据以验证我们的发现。
    结果:所有免疫标记通常在TC和IC中表达,除了PD-1在TC中不表达。在IC中,所有免疫标志物的表达彼此之间呈正相关,除了PD-L1和OX40之间,TIM3和OX40之间也是如此。在IC中,PD-1,PD-L1和OX40L阳性表达与更长的无进展生存期相关(PFS;分别为p=0.040,p=0.020和p=0.020)。在TC中,OX40阳性表达与较短的PFS相关(p=0.025)。随后,将TNBC患者分为高和低联合免疫评分组(CIS-H和CIS-L),基于在TC(TCIS-H或TCIS-L)和IC(ICIS-H或ICIS-L)中选择的生物标志物的表达水平。TCIS-H组与较长的PFS显著相关(p<0.001)。此外,ICIS-H组还与较长的PFS(p<0.001)和总生存期(OS;p=0.001)相关,在显著的水平。在多变量分析中,TCIS-H组和ICIS-H组均被确定为有利PFS的独立预测因子(分别为p=0.012和p=0.001).ICIS-H也被证明是有利OS的独立预测因子(p=0.003)。对来自TCGA的mRNA表达数据的分析也验证了我们关于TNBC的发现。
    结论:我们的新型TCIS和ICIS在TNBC中显示出显著的预后价值。需要更多的研究来加强我们的发现,并确定TNBC患者最有效的预后和预测性生物标志物。
    This study aimed to develop a novel combined immune score (CIS)-based model assessing prognosis in triple-negative breast cancer (TNBC).
    The expression of eight immune markers (PD-1, PD-L1, PD-L2, IDO, TIM3, OX40, OX40L, and H7-H2) was assessed with immunohistochemistry on the tumor cells (TCs) and immune cells (ICs) of 227 TNBC cases, respectively, and subsequently associated with selected clinicopathological parameters and survival. Data retrieved from The Cancer Genome Atlas (TCGA) were further examined to validate our findings.
    All immune markers were often expressed in TCs and ICs, except for PD-1 which was not expressed in TCs. In ICs, the expression of all immune markers was positively correlated between one another, except between PD-L1 and OX40, also TIM3 and OX40. In ICs, PD-1, PD-L1, and OX40L positive expression was associated with a longer progression-free survival (PFS; p = 0.040, p = 0.020, and p = 0.020, respectively). In TCs, OX40 positive expression was associated with a shorter PFS (p = 0.025). Subsequently, the TNBC patients were classified into high and low combined immune score groups (CIS-H and CIS-L), based on the expression levels of a selection of biomarkers in TCs (TCIS-H or TCIS-L) and ICs (ICIS-H or ICIS-L). The TCIS-H group was significantly associated with a longer PFS (p < 0.001). Furthermore, the ICIS-H group was additionally associated with a longer PFS (p < 0.001) and overall survival (OS; p = 0.001), at significant levels. In the multivariate analysis, both TCIS-H and ICIS-H groups were identified as independent predictors of favorable PFS (p = 0.012 and p = 0.001, respectively). ICIS-H was also shown to be an independent predictor of favorable OS (p = 0.003). The analysis of the mRNA expression data from TCGA also validated our findings regarding TNBC.
    Our novel TCIS and ICIS exhibited a significant prognostic value in TNBC. Additional research would be needed to strengthen our findings and identify the most efficient prognostic and predictive biomarkers for TNBC patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    Pembrolizumab是一种单克隆抗体,靶向T细胞上的程序性细胞死亡蛋白1(PD-1)受体,从而增强抗肿瘤免疫反应。Pembrolizumab已被证明可以改善各种癌症的生存率,但它也可能导致免疫相关的不良事件(irAE),可以影响任何器官系统。我们报告了两例由pembrolizumab引起的罕见但严重的irAE:重症肌无力(MG)和周围神经病变。两名患者在接受pembrolizumab治疗晚期癌症后均出现神经肌肉症状。根据他们的临床特征,他们被诊断为MG和周围神经病变,实验室测试,和不起眼的成像。治疗包括停用pembrolizumab并开始免疫抑制和支持治疗。一旦永久停用pembrolizumab并实施支持疗法,两名患者的症状和生活质量均得到改善。这些病例突出了识别和管理pembrolizumab罕见irAE的重要性,如MG和周围神经病变。早期诊断和治疗可以改善预后并降低发病率。此外,这些病例强调需要持续的上市后监测,以准确评估使用pembrolizumab的患者出现频率较低的药物不良反应的风险.在与患者讨论这种新疗法的利弊时,了解这些不良反应很重要。
    Pembrolizumab is a monoclonal antibody that targets the programmed cell death protein 1 (PD-1) receptor on T-cells, thereby enhancing the antitumor immune response. Pembrolizumab has been shown to improve survival in various cancers, but it can also cause immune-related adverse events (irAEs), which can affect any organ system. We report two cases of rare but serious irAEs caused by pembrolizumab: myasthenia gravis (MG) and peripheral neuropathy. Both patients presented with neuromuscular symptoms after receiving pembrolizumab for their advanced cancers. They were diagnosed with MG and peripheral neuropathy based on their clinical features, laboratory tests, and unremarkable imaging. Treatment involved discontinuing pembrolizumab and initiating immunosuppressive and supportive therapies. Both patients experienced improvement in their symptoms and quality of life once pembrolizumab was permanently discontinued and supportive therapies were in place. These cases highlight the importance of recognizing and managing rare irAEs of pembrolizumab, such as MG and peripheral neuropathy. Early diagnosis and treatment can improve outcomes and reduce morbidity. Furthermore, these cases emphasize the need for continued post-marketing surveillance to accurately assess the risk of less frequent adverse drug reactions seen in patients on pembrolizumab. Knowledge of these adverse reactions is important when discussing the pros and cons of this novel therapy with patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    微波(MW)热疗法已被开发为一种有效的临床策略,可以实现显着的抗肿瘤活性,也有可能引发抗肿瘤免疫。然而,患者通常在接受MW治疗后面临较高的肿瘤复发率,限制这种治疗的长期益处。MW治疗和免疫调节策略的组合可能代表了以有利于降低复发率的方式重新编程免疫抑制肿瘤微环境(TME)的有希望的手段。在这项研究中,Lenvatinib负载的Gd/Fe金属有机框架(Gd/FeMOF)被设计为增强此类抗肿瘤免疫力的有希望的方法。MW增强的动态Gd/FeMOF敏化可以促进MW辐照下高水平的活性氧产生,导致更强的免疫原性肿瘤细胞死亡。并行,从Gd/FeMOF制剂中释放的Lenvatinib可作为一种免疫佐剂,抑制程序性死亡配体1(PD-L1)的表达并驱动免疫抑制性TME的重编程.该MOF制剂中存在的Gd和Fe也赋予其磁共振成像能力。重要的是,体内动物模型实验证实了GdFeMOF治疗在防止复发的同时显着增强抗肿瘤免疫力的能力。因此,这项研究为旨在综合诊断和持久治疗癌症的有希望的策略奠定了基础.重要性声明:MW热治疗后的高肿瘤复发率限制了这种治疗的长期益处。我们发现Lenvatinib负载的Gd/FeMOF纳米颗粒的施用显著降低了MW热治疗后的肿瘤复发。在MW辐照下,发现Gd/FeMOF纳米颗粒由于促进免疫原性细胞死亡的过程而增强了免疫应答。此外,释放的Lenvatinib可以作为免疫佐剂下调PD-L1的表达,并重新编程肿瘤微环境的免疫抑制状态,从而进一步增强免疫反应。这是重要的,因为MW诱导的免疫应答相对较弱,并且通常不能有效地预防肿瘤复发。MW治疗与免疫调节策略的组合可以解决该问题。
    Microwave (MW) thermal therapy has been developed as an effective clinical strategy that can achieve pronounced antitumor activity and also has the potential to trigger antitumor immunity. However, patients generally face high rates of tumor recurrence following MW treatment, limiting the long-term benefits of such treatment. The combination of MW treatment and immunomodulatory strategies may represent a promising means of reprogramming the immunosuppressive tumor microenvironment (TME) in a manner conducive to lower recurrence rates. In this study, a Lenvatinib-loaded Gd/Fe metal-organic framework (Gd/FeMOF) was designed as a promising approach to enhancing such antitumor immunity. MW-enhanced dynamic Gd/FeMOF sensitization can facilitate high levels of reactive oxygen species production under MW irradiation, resulting in stronger immunogenic tumor cell death. In parallel, the Lenvatinib released from Gd/FeMOF preparations can serve as an immune adjuvant that suppresses programmed death ligand 1 (PD-L1) expression and drives the reprogramming of the immunosuppressive TME. The Gd and Fe present within this MOF preparation also imbue it with magnetic resonance imaging capabilities. Importantly, in vivo animal model experiments confirmed the ability of GdFeMOF treatment to significantly enhance antitumor immunity while protecting against recurrence. Accordingly, this study offers a foundation for promising strategies aimed at the integrated diagnosis and durable treatment of cancer. STATEMENT OF SIGNIFICANCE: High rates of tumor recurrence following MW thermal therapy limit the long-term benefits of such treatment. We found that the administration of Lenvatinib-loaded Gd/FeMOF nanoparticles significantly reduced tumor recurrence after MW thermal therapy. Under MW irradiation, the Gd/FeMOF nanoparticles were found to augment the immune response due to facilitation of the process of immunogenic cell death. In addition, the released Lenvatinib could act as an immune adjuvant to downregulate the expression of PD-L1 and reprogram the immunosuppressive state of the tumor microenvironment, thus further enhancing the immune response. This is significant because MW-induced immune responses are relatively weak and usually fail to effectively prevent tumor recurrence. The combination of MW treatment with an immunomodulatory strategy may solve this problem.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号