programmed cell death-ligand 1 (PD-L1)

程序性细胞死亡配体 1 (PD - L1)
  • 文章类型: Journal Article
    背景:男性原发性性腺外绒毛膜癌(PEGCC)很少见。它是高度恶性的,通常在诊断时出现远处转移,对治疗反应不佳。由于PD-L1的高水平,PD-1/PD-L1途径可能是治疗靶标。在这里,我们报告了我们在三级医院治疗6名男孩的儿科PEGCC的经验。
    方法:我们分析了2009年至2021年间经病理证实为PEGCC的六个男孩的数据。他们的临床人口统计学和组织病理学特征以及治疗和临床结果从他们的医学图表中检索。
    结果:患者的中位年龄为15岁(范围:12-17岁)。最常见的原发肿瘤部位是纵隔(67%,4/6),腹膜后(16.7%)和脑(16.7%)各1例。除了脑部PEGCC患者,均在诊断时出现转移。观察到以下转移部位:肺(100%,5/5),大脑(3/5,60%),肝脏(3/5,60%),肾脏(2/5,40%),和脾脏(1/5,20%)。大多数病人有干咳,呼吸困难,初次报告时咯血,可能是由于肺转移。所有患者的血清人绒毛膜促性腺激素(HCG)水平均高度升高。所有患者均接受基于铂的细胞毒性化疗。患有脑绒毛膜癌的患者接受了手术肿瘤切除术;所有其他人仅接受了手术活检。两名患者的PD-L1免疫组织化学染色呈强阳性。一名患者接受了PD-L1抑制剂派姆单抗,并获得了良好的反应。我们队列的1年生存率为33.3%,中位生存期为4.34个月。在随访期间,两名幸存者的血清HCG水平保持正常。
    结论:目前以铂类为基础的化疗反应不佳仍然是儿科PEGCC治疗的主要挑战。在常规化疗方案中加入派姆单抗可能会改善PEGCC男孩的预后。
    BACKGROUND: Primary extragonadal choriocarcinoma (PEGCC) in male is rare. It is highly malignant, typically presents with distant metastasis at the time of diagnosis, and responds poorly to treatment. Because of its associated high levels of PD-L1, the PD-1/PD-L1 pathway is a likely therapeutic target. Herein, we report our experience of treating pediatric PEGCC in six boys at a tertiary hospital.
    METHODS: We analyzed the data of six boys with pathologically confirmed PEGCC between 2009 and 2021. Their clinicodemographic and histopathological characteristics as well as treatments and clinical outcomes were retrieved from their medical charts.
    RESULTS: The patients\' median age was 15 (range: 12-17) years. The most common primary tumor site was the mediastinum (67%, 4/6), with one case each in the retroperitoneum (16.7%) and brain (16.7%). Except for the patient with brain PEGCC, all presented with metastasis at the time of diagnosis. The following metastatic sites were observed: the lungs (100%, 5/5), brain (3/5, 60%), liver (3/5, 60%), kidneys (2/5, 40%), and spleen (1/5, 20%). Most patients had dry cough, dyspnea, and hemoptysis at initial presentation, likely due to lung metastasis. Serum human chorionic gonadotropin (HCG) levels were highly elevated in all patients. All patients received platinum-based cytotoxic chemotherapy. The patient with brain choriocarcinoma underwent surgical tumor resection; all others underwent only surgical biopsy. Strong positive PD-L1 immunohistochemical staining was noted for two patients. One patient received the PD-L1 inhibitor pembrolizumab and achieved a good response. Our cohort\'s 1-year survival rate was 33.3%, with a median survival of 4.34 months. Serum HCG levels remained normal in the two survivors during follow-up visits.
    CONCLUSIONS: The poor response to current platinum-based chemotherapy remains a major challenge in the management of pediatric PEGCC. Adding pembrolizumab to a conventional chemotherapy regimen may improve the outcomes in boys with PEGCC.
    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
    肠道和口腔微生物群对口腔鳞状细胞癌(OSCC)患者临床结局的影响尚不清楚。我们比较了OSCC患者和健康对照(HCs)之间牙菌斑和粪便的细菌组成。在治疗开始前,从7例HC和18例OSCC患者收集粪便和牙菌斑样品。进行了16SrRNA基因的末端限制性片段长度多态性分析。检查了HC和OSCC组之间细菌多样性的差异。我们比较了取自OSCC和HC患者的样品中每种细菌物种的占有率,并分析了肿瘤标本中PD-L1表达与每种细菌物种占有率之间的相关性。OSCC患者的肠道和口腔微生物区比HC更多样化。OSCC患者的卟啉单胞菌和Prevotella明显高于HC。PD-L1阳性组肠道微生物群中梭菌亚簇XIVa的丰度显著高于PD-L1阴性组。OSCC患者的口腔和肠道微生物群处于菌群失调状态。我们的结果表明,使用益生菌和合生元针对这些疾病特异性微生物组的新癌症疗法的可能性。
    The impact of gut and oral microbiota on the clinical outcomes of patients with oral squamous cell carcinoma (OSCC) is unknown. We compared the bacterial composition of dental plaque and feces between patients with OSCC and healthy controls (HCs). Fecal and dental plaque samples were collected from 7 HCs and 18 patients with OSCC before treatment initiation. Terminal restriction fragment-length polymorphism analysis of 16S rRNA genes was performed. Differences in bacterial diversity between the HC and OSCC groups were examined. We compared the occupancy of each bacterial species in samples taken from patients with OSCC and HCs and analyzed the correlation between PD-L1 expression in the tumor specimens and the occupancy of each bacterial species. The gut and oral microbiota of patients with OSCC were more varied than those of HCs. Porphyromonas and Prevotella were significantly more abundant in patients with OSCC than in HCs. The abundance of Clostridium subcluster XIVa in the gut microbiota of the PD-L1-positive group was significantly greater than that in the PD-L1-negative group. The oral and gut microbiomes of patients with OSCC were in a state of dysbiosis. Our results suggest the possibility of new cancer therapies targeting these disease-specific microbiomes using probiotics and synbiotics.
    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
    免疫疗法治疗小细胞肺癌(SCLC)脑转移的疗效相对较低,SCLC脑转移瘤的肿瘤微环境尚不清楚。因此,我们研究了SCLC脑转移患者肿瘤浸润淋巴细胞(TIL)的分布和程序性细胞死亡配体1(PD-L1)的表达,以探讨SCLC脑转移的肿瘤微环境.
    回顾性分析2017年6月至2022年6月安徽医科大学第一附属医院神经外科收治的12例SCLC患者脑转移标本。本研究的纳入标准如下:(I)经病理证实诊断为SCLC脑转移瘤;(II)手术切除脑转移瘤;(III)年龄>18岁;(IV)临床资料完整。从住院病历系统检索患者相关数据,患者死亡日期的电话随访,总生存率(OS)。基于免疫荧光的组织微环境分析面板(MAP)用于TIL的检测,包括CD3,CD8,程序性细胞死亡1(PD-1),和PD-L1,在福尔马林固定和石蜡包埋的脑转移档案标本中。免疫组化法检测肿瘤细胞中PD-L1的表达水平。分析了12例患者的OS与上述标志物之间的相关性。
    12名患者被纳入研究。患者年龄为51-78岁,中位数为68岁,有1名女性和11名男性。在12例SCLC脑转移患者中:肿瘤实质中CD3TIL的阳性率与肿瘤间质为0.60%±0.94%vs.1.76%±2.72%(P=0.01),分别;肿瘤实质中CD8+TIL阳性率与肿瘤间质为0.80%±0.78%vs.2.46%±3.72%(P=0.02),分别。11例肿瘤实质中CD8+和PD-1+TILs均无共表达,仅1例,共表达CD3+和PD-1+TIL的浸润密度大于10/mm2。10例间质中无CD3+和PD-1+TIL共表达,2例CD8+和PD-1+TILs浸润密度均大于10/mm2。采用免疫组织化学方法检测12例SCLC转移灶中PD-L1的表达,阳性3例(25%)。生存分析显示,上皮内CD3+TILs阳性患者的OS显著延长[风险比3.383,95%置信区间(CI):0.959-11.940;P=0.04]。
    我们的研究进一步证明了SCLC脑转移的免疫微环境。TILs在SCLC脑转移中的分布较低,主要分布在间质中,这些肿瘤组织中PD-L1的表达较低。进一步探索SCLC脑转移的免疫微环境对潜在治疗具有重要意义。
    UNASSIGNED: The efficacy of immunotherapy for brain metastases from small cell lung cancer (SCLC) is relatively low, and the tumor microenvironment of SCLC brain metastases is still unknown. Therefore, we investigated the distribution of tumor-infiltrating lymphocytes (TILs) and the expression of programmed cell death-ligand 1 (PD-L1) in patients with brain metastases from SCLC to explore the tumor microenvironment of SCLC brain metastases.
    UNASSIGNED: A retrospective analysis was performed on 12 surgical specimens of brain metastases from patients with SCLC treated in the Department of Neurosurgery of The First Affiliated Hospital of Anhui Medical University from June 2017 to June 2022. The inclusion criteria for this study were the following: (I) a pathologically confirmed diagnosis of SCLC brain metastases; (II) surgical resection of brain metastases; (III) age >18 years; (IV) and complete clinical data. Patient-related data were retrieved from the inpatient medical record system, telephone follow-up of patients date of death, and overall survival (OS). The immunofluorescence-based tissue microenvironment analysis panel (MAP) was utilized for the detection of TILs, including CD3, CD8, programmed cell death 1 (PD-1), and PD-L1, in formalin-fixed and paraffin-embedded archival specimens of brain metastases. The expression levels of PD-L1 in tumor cells were detected by immunohistochemistry. The correlation between the OS and the above-mentioned markers was analyzed in the 12 patients.
    UNASSIGNED: Twelve patients were included in the study. The patients\' ages ranged from 51-78 years with a median of 68 years, with 1 female and 11 males. Among 12 patients with SCLC brain metastases: positive rates of CD3+ TILs in the tumor parenchyma vs. tumor stroma were 0.60%±0.94% vs. 1.76%±2.72% (P=0.01), respectively; positive rates of CD8+ TILs in the tumor parenchyma vs. tumor stroma were 0.80%±0.78% vs. 2.46%±3.72% (P=0.02), respectively. There was no co-expression of CD8+ and PD-1+ TILs in the tumor parenchyma of 11 cases, and the infiltration density of coexpressed CD3+ and PD-1+ TILs was more than 10/mm2 in only 1 case. There was no coexpression of CD3+ and PD-1+ TIL in the stroma of 10 cases, and the infiltration density of CD8+ and PD-1+ TILs was more than 10/mm2 in 2 cases. Immunohistochemistry was used to detect the expression of PD-L1 in 12 cases of SCLC metastatic lesions, and 3 cases (25%) were positive. Survival analysis showed that patients with positive intraepithelial CD3+ TILs had significantly longer OS [hazard ratio 3.383, 95% confidence interval (CI): 0.959-11.940; P=0.04].
    UNASSIGNED: Our study further demonstrated the immune microenvironment of SCLC brain metastases. The distribution of TILs in SCLC brain metastases is low and mainly distributed in the stroma, with the expression of PD-L1 in these tumor tissues being low. Further exploration of the immune microenvironment of SCLC brain metastases is of great significance for potential treatment.
    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
    肿瘤微环境(TME)在非小细胞肺癌(NSCLC)的肿瘤进展和免疫治疗反应中起重要作用。程序性细胞死亡1(PD-1)/程序性细胞死亡-配体1(PD-L1)检查点是TME中免疫抑制的中心介质。然而,目前仍需要鉴定能够反映NSCLC患者中TME和PD-L1表达差异的其他生物标志物.为此,我们关注G蛋白偶联受体C家族5组A型(GPRC5A)在非小细胞肺癌中的表达。GPRC5A,是一种视黄酸诱导基因,在非小细胞肺癌中发挥多种作用。然而,关于GPRC5A在调节TME和PD-L1中的作用知之甚少。我们的目的是描述GPRC5A在NSCLC中的表达在免疫细胞浸润中的关键作用。
    我们在复旦大学上海肿瘤防治中心(FUSCC)队列中确定GPRC5A表达与NSCLC患者临床病理特征之间的关系。此外,我们通过使用公共数据库验证GPRC5A作为预测生物标志物,以揭示GPRC5A表达与免疫细胞浸润之间的关系.为了将GPRC5A的表达与NSCLC样本中PD-L1的空间分布相关联,我们进行了多重免疫组织化学(mIHC)。
    低GPRC5A表达与早期病理阶段(pStage)相关。免疫细胞浸润分析表明,GPRC5A低表达与CD8+T细胞浸润增加之间存在关系,激活的CD4+T细胞,和TME内的M1巨噬细胞。此外,GPRC5A低表达与NSCLC中免疫表型评分(IPS)升高相关.此外,mIHC分析显示GPRC5A低表达与肿瘤PD-L1表达的空间分布之间存在相关性。
    我们的研究揭示了GPRC5A的低表达与NSCLC早期pStage之间的关系。此外,我们观察到GPRC5A的低表达与免疫细胞浸润增加有关,较高的IPS,PD-L1阳性肿瘤细胞的空间分布。因此,我们推测GPRC5A的低表达与免疫治疗有关,但仍需要进一步验证。
    UNASSIGNED: The tumor microenvironment (TME) plays an important role in tumor progression and immunotherapy responses in non-small cell lung cancer (NSCLC). The programmed cell death 1 (PD-1)/ programmed cell death-ligand 1 (PD-L1) checkpoint is a central mediator of immunosuppression in the TME. However, there is still a need to identify additional biomarkers that could reflect the difference in TME and PD-L1 expression in NSCLC patients. To this end, we focused on the expression of G-protein-coupled receptor family C group 5 type A (GPRC5A) in NSCLC. GPRC5A, is a retinoic acid-inducible gene that plays multiple roles in NSCLC. However, little is known about the role of GPRC5A in regulating the TME and PD-L1. Our objective was to describe the critical role of GPRC5A expression in NSCLC in the setting of immune cell infiltration.
    UNASSIGNED: We identified the relationship between GPRC5A expression and the clinicopathologic characteristics of NSCLC patients in the Fudan University Shanghai Cancer Center (FUSCC) cohort. Furthermore, we validated GPRC5A as a predictive biomarker by using public databases to reveal the relationship between GPRC5A expression and immune cell infiltration. To correlate the expression of GPRC5A with the spatial distribution of PD-L1 in NSCLC samples, we performed multiplex immunohistochemistry (mIHC).
    UNASSIGNED: Low GPRC5A expression is associated with earlier pathological stage (pStage). Analysis of immune cell infiltration indicates there is a relationship between low GPRC5A expression and increased infiltration of CD8+ T cells, activated CD4+ T cells, and M1 macrophages within the TME. Furthermore, low GPRC5A expression is associated with an increased immunophenotype score (IPS) in NSCLC. Additionally, analysis of mIHC reveals there is a correlation between low GPRC5A expression and spatial distribution of tumoral PD-L1 expression.
    UNASSIGNED: Our study revealed the relationship between low expression of GPRC5A and earlier pStage in NSCLC. Furthermore, we observed that low expression of GPRC5A is associated with increased infiltration of immune cells, higher IPS, and spatial distribution of PD-L1-positive tumor cells. Therefore, we speculate that low expression of GPRC5A is associated with immunotherapy, but further validation is still required.
    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
    三阴性乳腺癌(TNBC)代表了一种高度侵袭性的乳腺癌亚型,历史上使用主要涉及蒽环类和紫杉烷类的化疗方案进行管理,产生不利的预后。这篇综述致力于全面检查早期三阴性乳腺癌(eTNBC)的治疗策略的现状。特别强调免疫治疗方式,联合疗法,预测性生物标志物,和正在进行的临床试验。这篇综述的主要目的是仔细评估现有文献,确定重大发现,并参与讨论它们对未来研究工作的潜在影响,临床应用,和政策制定。
    这项审查是使用PubMed和GoogleScholar数据库进行的,最新更新于2023年3月进行。搜索策略旨在确保对文献进行全面分析,专注于最近的进步。
    我们严格评估当前的eTNBC治疗前景,涵盖单一疗法的疗效和局限性,联合疗法,和预测性生物标志物。我们强调了最近试验的有希望的结果,解决围绕化疗的争议,并探索辅助和新辅助治疗(NAT)的最佳方法。对个性化治疗策略的见解,正在进行的试验,并提供了未来的前景,推进我们对eTNBC治疗方案的理解。
    通过对文献的综合分析,这篇综述强调了免疫治疗的潜力,特别是联合化疗,作为治疗eTNBC的一种有希望的方法。然而,需要进一步的研究来优化治疗策略,完善患者选择标准,并确定可靠的生物标志物,以预测对免疫检查点抑制剂(ICI)的反应。这篇综述的发现对未来的研究具有重要意义,临床实践,和政策制定,提供对eTNBC治疗当前挑战和进步的宝贵见解。最终,这些知识有助于改善患者的预后,提高生活质量,以及开发更有效的eTNBC治疗方法。
    UNASSIGNED: Triple negative breast cancer (TNBC) represents a highly aggressive breast cancer subtype, historically managed with chemotherapy regimens predominantly involving anthracyclines and taxanes, yielding unfavorable prognoses. This review endeavors to offer a thorough examination of the present state of treatment strategies for early stage triple negative breast cancer (eTNBC), with a particular emphasis on immunotherapy modalities, combination therapies, predictive biomarkers, and ongoing clinical trials. The principal aim of this review is to meticulously assess the available literature, ascertain significant discoveries, and engage in discussions regarding their potential implications for future research endeavors, clinical applications, and policy formulation.
    UNASSIGNED: This review was conducted using PubMed and Google Scholar databases, with the latest update performed in March 2023. The search strategy was designed to ensure a comprehensive analysis of the literature, with a focus on recent advancements.
    UNASSIGNED: We critically assess the current eTNBC treatment landscape, covering efficacy and limitations of monotherapy, combination therapies, and predictive biomarkers. We highlight promising results from recent trials, address controversies surrounding chemotherapy, and explore optimal approaches for adjuvant and neoadjuvant therapy (NAT). Insights into personalized treatment strategies, ongoing trials, and future perspectives are provided, advancing our understanding of therapeutic options for eTNBC.
    UNASSIGNED: Through a comprehensive analysis of the literature, this review highlights the potential of immunotherapy, particularly in combination with chemotherapy, as a promising approach for treating eTNBC. However, further research is warranted to optimize treatment strategies, refine patient selection criteria, and identify reliable biomarkers for predicting response to immune checkpoint inhibitors (ICIs). The findings of this review hold significant implications for future research, clinical practice, and policy-making, offering valuable insights into the current challenges and advancements in eTNBC treatment. Ultimately, this knowledge can contribute to improved patient outcomes, enhanced quality of life, and the development of more effective therapeutic approaches for eTNBC.
    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
    背景:原发性心脏血管肉瘤(PCA)发病率低,预后差。目前,没有统一的临床治疗标准。
    方法:我们报告一例48岁男性胸闷,呼吸困难,和呼吸困难。影像学和术后组织病理学研究证实了PCA,并且肿瘤已侵入整个右心房。患者在术后放疗期间发展为进行性疾病(PD)。我们根据分子谱和三级淋巴结构(TLSs)和程序性细胞死亡配体1(PD-L1)的评估结果,将免疫治疗与靶向治疗相结合。治疗后,患者的转移性淋巴结在一定程度上减少,表明联合治疗是有效的。
    结论:据我们所知,这是放疗联合抗PD-1和酪氨酸激酶抑制剂(TKI)治疗PCA的首次报道.此外,这是关于基于新评估方法的PCA免疫治疗的第一份报告,包括TLS,PD-L1和基因组谱。
    BACKGROUND: Primary cardiac angiosarcoma(PCA) has a low incidence rate and poor prognosis. Currently, no unified clinical treatment standards are available.
    METHODS: We report the case of a 48-year-old man presenting chest tightness, breathlessness, and dyspnea. Imaging and postoperative histopathologic studies confirmed PCA and that the tumor had invaded the entire right atrium. The patient developed progressive disease (PD) during postoperative radiotherapy. We used immunotherapy combined with targeted therapy based on the results of molecular profile and evaluation of tertiary lymphoid structures (TLSs) and programmed cell death-ligand 1 (PD-L1). After treatment, the metastatic lymph nodes of the patient were reduced to a certain extent, indicating that combination therapy was effective.
    CONCLUSIONS: To the best of our knowledge, this is the first report of radiotherapy combined with anti-PD-1 and tyrosine kinase inhibitors(TKI) for PCA. In addition, this is the first report on immunotherapy for PCA based on new evaluation methods, including TLSs, PD-L1, and genomic profile.
    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
    Ramucirumab,抗血管内皮生长因子受体(VEGFR)单克隆抗体(mAb),抑制血管生成并降低肿瘤活性。程序性细胞死亡配体1(PD-L1)可能作用于VEGFR2以诱导癌细胞血管生成和转移。在这里,我们根据晚期胃癌(AGC)患者PD-L1表达状况,研究了雷莫西单抗和紫杉醇联合应用的疗效.
    该分析包括2018年12月1日至2022年2月28日在三星医疗中心接受雷莫西单抗联合紫杉醇作为二线治疗的AGC患者。所有患者数据分析包括使用组合阳性评分(CPS)评估PD-L1表达。我们根据患者的PD-L1表达分析其疗效和生存。
    我们在这项分析中纳入了117名患者,80例患者(68.4%)有一个或多个PD-L1CPS,37(31.6%)有5个或更多,19分(16.2%)有10分或以上。无进展生存期(PFS)和总生存期(OS)在PD-L1CPS小于1和1或以上的患者之间没有显著差异{PD-L1<1%vs.PD-L1≥1%;PFS:中位数3.6个月[95%置信区间(CI):2.4-4.8个月]vs.中位数4.1个月(95%CI:3.5-4.7个月),P=0.93;PD-L1<1%vs.PD-L1≥1%;OS:中位数7.0个月(95%CI:5.4-8.6个月)与中位数8.1个月(95%CI:6.4-9.8个月),P=0.32}。PD-L1CPS小于5和5或更高的患者之间的PFS和OS没有显着差异[PD-L1<5%vs.PD-L1≥5%;PFS:3.9个月(95%CI:3.3-4.5个月)与4.4个月(95%CI:3.0-5.8个月),P=0.57;OS:7.4个月(95%CI:6.5-8.3个月)与10.0个月(95%CI:1.1-18.9个月),P=0.07]。有趣的是,PD-L1CPS截止值为10时,PFS和OS确实存在显着差异[PD-L1<10%vs.PD-L1≥10%;PFS:3.8个月(95%CI:3.3-4.3个月)与5.7个月(95%CI:4.1-7.3个月),P=0.05;OS:7.2个月(95%CI:6.5-7.9个月)与18.9个月(95%CI:6.5-31.3个月),P=0.04]。
    尚未建立生物标志物来预测雷莫西单抗联合紫杉醇治疗后的生存时间。该分析表明,PD-L1CPS截止值为10可能是一种新的生物标志物,可以预测用雷莫西单抗和紫杉醇治疗的AGC患者的生存率。
    UNASSIGNED: Ramucirumab, an anti-vascular endothelial growth factor receptor (VEGFR) monoclonal antibody (mAb), inhibits angiogenesis and reduces tumor activity. Programmed cell death-ligand 1 (PD-L1) might act upon VEGFR2 to induce cancer cell angiogenesis and metastasis. Herein, we investigated the efficacy of combining ramucirumab and paclitaxel according to the status of PD-L1 expression in patients with advanced gastric cancer (AGC).
    UNASSIGNED: This analysis included AGC patients who received ramucirumab plus paclitaxel as 2nd line therapy between December 1, 2018, and February 28, 2022, at Samsung Medical Center. All patient data analyses included an evaluation of PD-L1 expression using the combined positive score (CPS). We analyzed the efficacy and the survival of patients according to their PD-L1 expression.
    UNASSIGNED: We included 117 patients in this analysis, and 80 patients (68.4%) had a PD-L1 CPS of one or more, 37 (31.6%) had five or more, and 19 (16.2%) had ten or more scores. Progression-free survival (PFS) and overall survival (OS) did not differ significantly between patients with a PD-L1 CPS of less than one and one or more {PD-L1 <1% vs. PD-L1 ≥1%; PFS: median 3.6 months [95% confidence interval (CI): 2.4-4.8 months] vs. median 4.1 months (95% CI: 3.5-4.7 months), P=0.93; PD-L1 <1% vs. PD-L1 ≥1%; OS: median 7.0 months (95% CI: 5.4-8.6 months) vs. median 8.1 months (95% CI: 6.4-9.8 months), P=0.32}. PFS and OS did not differ significantly between patients with a PD-L1 CPS of less than 5 and 5 or more [PD-L1 <5% vs. PD-L1 ≥5%; PFS: 3.9 months (95% CI: 3.3-4.5 months) vs. 4.4 months (95% CI: 3.0-5.8 months), P=0.57; OS: 7.4 months (95% CI: 6.5-8.3 months) vs. 10.0 months (95% CI: 1.1-18.9 months), P=0.07]. Interestingly, with a PD-L1 CPS cutoff of 10, PFS and OS did differ significantly [PD-L1 <10% vs. PD-L1 ≥10%; PFS: 3.8 months (95% CI: 3.3-4.3 months) vs. 5.7 months (95% CI: 4.1-7.3 months), P=0.05; OS: 7.2 months (95% CI: 6.5-7.9 months) vs. 18.9 months (95% CI: 6.5-31.3 months), P=0.04].
    UNASSIGNED: No biomarkers have been established to predict survival times after ramucirumab plus paclitaxel treatment. This analysis suggests that a PD-L1 CPS cutoff of 10 might be novel a biomarker to predict the survival of AGC patients treated with ramucirumab and paclitaxel.
    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
    最近的证据表明,肿瘤微环境(TME)中的多效性细胞因子干扰素-γ(IFN-γ)通过调节肝癌干细胞(LCSCs)在肝细胞癌(HCC)的进展中起着积极作用。本研究探讨了线粒体程序性细胞死亡配体1(PD-L1)及其对铁凋亡的调控在调节LCSCs癌干性中的作用和潜在机制。研究表明,模拟TMEIFN-γ暴露会增加HCC细胞中的LCSC比率和癌症干性表型。IFN-γ暴露通过增强谷胱甘肽过氧化物酶4(GPX4)表达以及LCSC中的活性氧(ROS)和脂质过氧化(LPO)生成来抑制索拉非尼(Sora)诱导的铁凋亡。此外,IFN-γ暴露上调PD-L1表达及其线粒体易位,在LCSCs中诱导动态蛋白相关蛋白1(Drp1)依赖性线粒体裂变并与糖酵解代谢重编程相关。PD-L1的遗传干预促进了Sora的铁凋亡依赖性抗肿瘤作用,减少糖酵解代谢重编程,并在体外和体内抑制HCC的癌症干细胞性。我们的研究结果揭示了一种新的机制,即IFN-γ暴露诱导的PD-L1线粒体易位增强糖酵解重编程,以介导LCSC中GPX4依赖性的铁凋亡抗性和癌症干性。这项研究为线粒体PD-L1-Drp1-GPX4信号轴在调节LCSCs中IFN-γ暴露相关癌症干性中的作用提供了新的见解,并验证了PD-L1靶向干预与Sora联合可能实现有希望的协同抗HCC作用。
    Evidence recently showed that pleiotropic cytokine interferon-gamma (IFN-γ) in the tumor microenvironment (TME) plays a positive role in hepatocellular carcinoma (HCC) progression through the regulation of liver cancer stem cells (LCSCs) in HCC. The present study explored the role and potential mechanism of mitochondrial programmed cell death-ligand 1 (PD-L1) and its regulation of ferroptosis in modulating the cancer stemness of LCSCs. It was shown that mimicking TME IFN-γ exposure increased the LCSCs ratio and cancer stemness phenotypes in HCC cells. IFN-γ exposure inhibited sorafenib (Sora)-induced ferroptosis by enhancing glutathione peroxidase 4 (GPX4) expression as well reactive oxygen species (ROS) and lipid peroxidation (LPO) generation in LCSCs. Furthermore, IFN-γ exposure upregulated PD-L1 expression and its mitochondrial translocation, inducing dynamin-related protein 1 (Drp1)-dependent mitochondrial fission and correlating with glycolytic metabolism reprogramming in LCSCs. The genetic intervention of PD-L1 promoted ferroptosis-dependent anti-tumor effects of Sora, reduced glycolytic metabolism reprogramming, and inhibited cancer stemness of HCC in vitro and in vivo. Our results revealed a novel mechanism that IFN-γ exposure-induced mitochondrial translocation of PD-L1 enhanced glycolytic reprogramming to mediate the GPX4-dependent ferroptosis resistance and cancer stemness in LCSCs. This study provided new insights into the role of mitochondrial PD-L1-Drp1-GPX4 signal axis in regulating IFN-γ exposure-associated cancer stemness in LCSCs and verified that PD-L1-targeted intervention in combination with Sora might achieve promising synergistic anti-HCC effects.
    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
    程序性细胞死亡配体1(PD-L1)是一种生物标志物,用于预测免疫检查点抑制剂在各种癌症类型中的临床疗效。细胞因子在调节肿瘤细胞PD-L1表达中的作用尚未得到充分表征,however.在这里,我们发现白细胞介素-1β(IL-1β)在非小细胞肺癌(NSCLC)中PD-L1表达的调节中起关键作用。
    我们使用可用的单细胞RNA序列数据对NSCLC组织中的细胞因子基因表达进行了综合筛选。然后,我们研究了IL-1β在体外的作用,以阐明其对PD-L1对NSCLC细胞的诱导作用。
    IL-1β基因在肿瘤微环境中高表达,特别是在巨噬细胞中。IL-1β和干扰素-γ(IFN-γ)的组合诱导NSCLC细胞系中PD-L1表达的协同增加。IL-1β和IFN-γ还协同激活丝裂原活化蛋白激酶(MAPK)信号,并促进下游转录因子与PD-L1基因启动子的结合。此外,MAPK信号抑制剂阻断IL-1β和IFN-γ对PD-L1的上调。
    我们的研究报告肿瘤微环境中高水平的IL-1β可能与IFN-γ协同作用,通过激活MAPK信号在肿瘤细胞中诱导最大的PD-L1表达,IL-1β-MAPK轴是减弱PD-L1介导的抗肿瘤免疫抑制的有希望的治疗靶标。
    Programmed cell death-ligand 1 (PD-L1) is a biomarker for prediction of the clinical efficacy of immune checkpoint inhibitors in various cancer types. The role of cytokines in regulation of PD-L1 expression in tumor cells has not been fully characterized, however. Here we show that interleukin-1β (IL-1β) plays a key role in regulation of PD-L1 expression in non-small cell lung cancer (NSCLC).
    We performed comprehensive screening of cytokine gene expression in NSCLC tissue using available single-cell RNA-Sequence data. Then we examined the role of IL-1β in vitro to elucidate its induction of PD-L1 on NSCLC cells.
    The IL-1β gene is highly expressed in the tumor microenvironment, particularly in macrophages. The combination of IL-1β and interferon-γ (IFN-γ) induced a synergistic increase in PD-L1 expression in NSCLC cell lines. IL-1β and IFN-γ also cooperatively activated mitogen-activated protein kinase (MAPK) signaling and promoted the binding of downstream transcription factors to the PD-L1 gene promoter. Furthermore, inhibitors of MAPK signaling blocked upregulation of PD-L1 by IL-1β and IFN-γ.
    Our study reports high levels of IL-1β in the tumor microenvironment may cooperate with IFN-γ to induce maximal PD-L1 expression in tumor cells via activation of MAPK signaling, with the IL-1β-MAPK axis being a promising therapeutic target for attenuation of PD-L1-mediated suppression of antitumor immunity.
    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
    免疫检查点抑制揭示了癌症治疗的新时代,和随机临床试验表明,转移性胃癌(GC)患者的整体人群中有一部分可能从免疫治疗中获得临床益处,这提高了识别预测性生物标志物的相关性。程序性细胞死亡配体1(PD-L1)表达已证明表达水平与GC中免疫检查点抑制产生的益处之间存在显着关联。然而,这种生物标志物显示了在将免疫检查点抑制作为GC护理标准的治疗决策中必须考虑的几个陷阱,如时空异质性,观察者间的可变性,免疫组织化学(IHC)测定,以及化疗或放疗的影响。
    在本次全面审查中,我们修订了关于GC中PD-L1评估的主要研究。
    在这里,我们描述了GC中肿瘤微环境的分子特征,解释PD-L1表达的障碍,并提供评估免疫检查点抑制的有效性和安全性以及与生物标志物表达的关联的临床试验数据,在一线和后期治疗中。
    从免疫检查点抑制的新兴预测生物标志物,PD-L1已证明肿瘤微环境中的表达水平与来自GC中免疫检查点抑制的益处之间有意义的关联。
    UNASSIGNED: Immune checkpoint inhibition has shed light on a new era in cancer therapy, and randomized clinical trials have demonstrated that a meaningful portion of the overall population of metastatic gastric cancer (GC) patients may derive clinical benefit from immunotherapy, which raises the relevance in identifying predictive biomarkers. Programmed cell death-ligand 1 (PD-L1) expression has demonstrated a significant association between level of expression and the magnitude of benefit derived from immune checkpoint inhibition in GC. Nevertheless, this biomarker shows several pitfalls that must be considered in the therapeutic decision to incorporate immune checkpoint inhibition as the standard of care of GC, such as spatial and temporal heterogeneity, interobserver variability, immunohistochemistry (IHC) assay, and influence by chemotherapy or radiation therapy.
    UNASSIGNED: In the present comprehensive review, we revised the main studies regarding PD-L1 evaluation in GC.
    UNASSIGNED: Here we describe the molecular characteristics of the tumor microenvironment in GC, the obstacles in the interpretation of PD-L1 expression and present the data of the clinical trials that have evaluated the efficacy and safety of immune checkpoint inhibition and the association with the biomarker expression, both in first-line and later lines of therapy.
    UNASSIGNED: From the emerging predictive biomarkers for immune checkpoint inhibition, PD-L1 has demonstrated a meaningful association between level of expression in tumor microenvironment and the magnitude of benefit derived from immune checkpoint inhibition in GC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号